Sunteți pe pagina 1din 7

SURGICAL INFECTIONS

Volume 19, Number 8, 2018


ª Mary Ann Liebert, Inc.
DOI: 10.1089/sur.2018.263

Immunization Against Staphylococcus aureus Infections

Philip S. Barie,1,2 Mayur Narayan,1 and Robert G. Sawyer3

Abstract
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

Background: Infections caused by Staphylococcus aureus continue to plague surgical patients, whether as
surgical site infections or other nosocomial infections that complicate surgical care. The only meaningful
methods available to decrease the risk of developing such infections are topical skin antisepsis (pre-operative
skin preparation) and peri-operative antibiotic prophylaxis, neither of which offer a panacea. Alternatives to the
latter are sought so as to minimize antibiotic selection pressure as a factor in the increasing problem of
antimicrobial drug resistance. This review considers the possibility that immunization against S. aureus may
offer a viable alternative for prophylaxis.
Methods: Review and synthesis of pertinent English-language medical literature.
Results: Vaccination against viral pathogens has been in successful clinical use for more than two centuries and
was instrumental in the eradication of smallpox and the near-elimination of diseases such as poliomyelitis.
Vaccinations against a limited number of bacterial pathogens (e.g., Bordetella pertussis, Clostridium tetanii,
Corynebacterium diphtheriae, Haemophilus influenzae type b, Neisseria meningiditis, Streptococcus pneumo-
niae) have also been introduced with success, whereas others against bacteria are in development (C. difficile,
Pseudomonas aeruginosa, S. aureus). Vaccination against S. aureus infection is in current veterinary use (e.g.,
to prevent mastitis among dairy cattle) but has not been successful to date in human beings despite multiple
attempts, although development continues.
Conclusions: Because of its complex microbiology, including multiple virulence factors and the ability to
evade host immune surveillance, S. aureus presents numerous antigenic targets for vaccine development.
Failure of two prior single-antigen vaccines in clinical trials has led to the consensus that future vaccine
candidates must be directed against multiple antigens. Two distinct four-antigen vaccines are in clinical trials,
but efficacy is yet to be determined.

Keywords: immunization; infection prevention; Staphylococcus aureus; surgical prophylaxis; vaccination

V accination is the administration of antigenic material


(the vaccine) to stimulate an individual’s immune sys-
tem to develop adaptive immunity to a pathogen, so as to
pathogens that, as established infections, carry substantial
morbidity and some risk of death. By contrast, inoculation
refers to the induction of active adaptive immunity, which
prevent or ameliorate infectious disease(s). Historically and uses un-attenuated live pathogens, whereas passive immunity
to the present day, vaccination has been directed clinically is the hoped-for result of administration of immunoglobulin
against viral pathogens with great success, but more recently or, experimentally, a monoclonal antibody or antibiotic–
vaccines against bacterial pathogens have become suc- antibody conjugates.
cessful (e.g., Bordetella pertussis, Clostridium tetanii, Cor- Vaccination is the most effective method of preventing
ynebacterium diphtheriae, Haemophilus influenzae type b, viral diseases, and in some circumstances the prevention may
Neisseria meningiditis, Streptococcus pneumoniae), many of extend to diseases consequent to chronic infection (e.g.,
which are utilized regularly in acute care surgery practice. hepatitis B vaccination and reduced incidence of hepatocel-
Interest in immunization (used herein synonymously with lular carcinoma [1,2]; human papilloma virus vaccination
vaccination) against bacterial pathogens would be stimulated uptake is not yet sufficiently prevalent to assess the effect on
if a vaccine were to be developed against hospital-acquired cervical or oropharyngeal carcinoma incidence [3,4]. When a

Departments of 1Surgery and 2Medicine, Weill Cornell Medicine, New York, New York.
3
Department of Surgery, Western Michigan University, Kalamazoo, Michigan.

750
IMMUNIZATION AGAINST STAPHYLOCOCCUS AUREUS 751

sufficiently large percentage of a population has been vac- nogenicity (i.e., drug allergy). Some such disadvantages re-
cinated, herd immunity results. Widespread immunity as a lated to highly purified or genetically engineered vaccines
result of vaccination is largely responsible for the worldwide can be overcome by innovative technologies, such as live
eradication of smallpox and the marked reduction of diseases vector vaccines, and DNA or RNA vaccines. Moreover, re-
such as poliomyelitis, measles, and tetanus (arguably the cent years have witnessed the development of novel adjuvant
most successful example of vaccination against a bacterial (in immunology, a substance used to enhance antigenicity,
pathogen-C. tetanii) from much of the world. e.g., a suspension of minerals [alum, aluminum hydroxide, or
phosphate] on which antigen is adsorbed) formulations that
Historic Perspective specifically focus on the augmentation or regulation of the
interplay between innate and adaptive immune systems and
Smallpox was likely the first disease subjected to prevention
the function of antigen-presenting cells. Finally, vaccine
by inoculation [5] and was the first disease for which a vaccine
design has become more tailored, which in turn enhances the
was produced. The smallpox vaccine was ‘‘invented’’ in 1796
potential of extending its application to complex microbial
by the British physician Edward Jenner and although at least
pathogens hitherto not accessible. This review provides an
six people had used the same principles years earlier, he was
overview of the key considerations and processes involved in
the first to publish evidence that it was effective and to provide
vaccine development against S. aureus.
advice on its production [6]. The terms ‘‘vaccine’’ and ‘‘vac-
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

cination’’ are derived from Variolae vaccinae (smallpox of the


Staphylococcus aureus as a Human Pathogen
cow), the term devised by Jenner to denote cowpox. The im-
munization was called vaccination because it was derived from Staphylococcus aureus is ubiquitous and a virulent human
a virus affecting cows (Latin: vacca ‘cow’) [5,6]. Smallpox and animal pathogen that may afflict healthy individuals or
was a contagious and deadly disease, causing the deaths of those with compromised immunity, outpatients, or those
20%–60% of infected adults and more than 80% of infected hospitalized. More than two million people become infected
children [7]. When smallpox was finally eradicated in 1979, it with S. aureus each year in the United States, with more than
had already killed an estimated 300–500 million people [8] in 100,000 deaths annually [10]. A multiplicity of infections has
the twentieth century. been observed, because the pathogen can infect almost any
In common parlance, vaccination and immunization have a tissue imaginable. Surgical infections of interest caused by
similar meaning. This distinguishes them from inoculation, S. aureus include community-acquired infections, especially
which uses un-attenuated live pathogens, although in com- of skin and soft tissue; surgical site infections (in which it is
mon usage any of the terms can refer to an immunization. the most common pathogen); and post-operative nosocomial
Vaccination efforts have been met with some controversy on infections [11].
scientific, ethical, political, medical safety, and religious Identification of the patient at risk is a crucial first step in
grounds, all largely unfounded. Early success of vaccination prevention. Population-based studies have identified male
brought widespread acceptance, and mass vaccination cam- gender and both extremes of age as risk factors for the ac-
paigns have reduced greatly the incidence of many diseases quisition of staphylococci. Dialysis dependence, diabetes
in numerous geographic regions. mellitus, cancer, human immunodeficiency virus infection,
intravenous drug use, alcohol abuse, recent surgery, critical
Principles of Vaccination illness, and staphylococcal nasal carriage have been identi-
fied as independent risk factors [12,13]. Individuals who are
Until relatively recently, vaccine development has been
persistent nasal carriers (the primary locus, although other
empirical, with limited understanding of how the immune
sites are possible) of S. aureus may represent as much as 30%
system is activated to elicit adaptive immunity, but, advances
of healthy people [14]. Most persistent carriers (as many
in immunology, microbiology, genetics, and molecular bi-
as 90%) harbor a single strain of methicillin-susceptible
ology have led to burgeoning understanding of the interac-
S. aureus (MSSA), although methicillin-resistant S. aureus
tions of microorganisms with the human immune system [9].
(MRSA) nasal carriage is recognized now in up to 10% of
Modern vaccine development builds on precise knowledge of
carriers). Patients who are persistent carriers are at higher risk
the microbiology of pathogens, their interaction with the
of acquiring S. aureus infection. In addition to decolonization
immune system, and their capacity to counteract and evade
of staphylococcal carriers [15,16], other tactics being investi-
innate and adaptive immune mechanisms, using technologies
gated include phage therapy, molecules aimed at blocking
to produce highly purified antigens that provide a lower re-
regulation of virulence determinants and surface adhesins,
actogenicity (the propensity of a vaccine to produce common,
and vaccination.
expected adverse reactions (e.g., fever, soreness at injection
site, bruising, erythema, induration, and edema). However, it
Mechanisms of virulence and immune evasion
is unclear whether a higher degree of reactogenicity to a
vaccine correlates with more severe adverse events; the U.S. The high prevalence and threat of S. aureus infections in
Food and Drug Administration (FDA) has been unable to surgical patients has led to vigorous efforts to develop spe-
make such an association. cific preventive and therapeutic measures directed against
Attempts to improve vaccine antigen purity may result in this pathogen, by enhanced infection control practices, use of
impaired vaccine immunogenicity (the ability of a particular topical antiseptics, and narrow-spectrum systemic antibac-
antigen or epitope [or antigenic determinant]; the part of an terial agents [17,18]. Prevention remains elusive despite
antigen molecule to which an antibody attaches itself to these efforts.
provoke an immune response). Distinction is made between Staphylococcus aureus manifests a broad array of viru-
wanted (i.e., successful vaccination) and unwanted immu- lence factors to account for its pathogenicity, and new
752 BARIE ET AL.

virulence characteristics continue to be acquired. Staphylo- brane pore formation and cytolysis. These virulence factors
coccus aureus is able to evade immune surveillance mecha- include bi-component leukocidins [29], heptameric beta-
nisms as well. The genome of S. aureus consists of 75% core barrel–forming alpha-toxin (Hla), and phenol-soluble mod-
genes that appear in all species and 25% accessory genes that ulins (PSMs) [33]. An array of cell wall-anchored (CWA)
vary among strains [19]. Within the accessory component of proteins, which number more than 25 and have substantial
the genome are mobile genetic elements that are transferable functional redundancy, bind to cell wall peptidoglycans [34].
among S. aureus strains. Accessory components may consist Clumping factor A (ClfA) is the major fibrinogen-binding
of pathogenicity islands, prophages (from bacteriophages), protein of S. aureus. Clumping factor A causes platelet ag-
staphylococcal cassette chromosomes (SCC), genetic is- gregation and clumping of bacteria on plasma, and is a major
lands, and plasmids [20]. Structural elements, metabolic factor relating to pathogen survival and dissemination in
regulators, and virulence factors of S. aureus may be common blood. By contrast, S. aureus attachment to nasal epithelial
to all species (e.g., coagulases), or they may be highly spe- cells is facilitated by the staphylococcal surface adhesin
cific for each strain (especially in the case of virulence fac- clumping factor B (ClfB) [34]. Fibronectin-binding proteins
tors). The exchange of mobile genetic units among strains of (FnBPs) A and B enable S. aureus to adhere to and invade a
S. aureus and the acquisition of genetic material from other host of cell types, including epithelia, fibroblasts, and oste-
bacterial species (e.g., Enterococcus), results in fluid patterns oblasts [35]. Protein A (SpA) is a conserved, multi-functional
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

of virulence and resistance. surface protein that has a number of immunosuppressive


Microbial virulence factors of S. aureus generally have been traits, including induced apoptosis of B-lymphocytes thereby
grouped into bacterial structural factors, secreted bacterial suppressing the host antibody response, and is arguably the
products and enzymes, and resistance mechanisms to antimi- most important mechanism of immune evasion in S. aureus
crobial agents [21]. Structural elements of the bacterial cell [34] (see below). Iron-regulated surface proteins (Isd) A and
include a polysaccharide capsule that resists phagocytosis and B are part of the heme uptake system (iron is a necessary
the peptidoglycan cell wall that anchors a number of surface nutrient for bacteria) [34]. The Isd proteins play a role in
adhesion proteins, allowing microbial binding to the extra- bacterial adherence to epithelial cells and IsdA on the bac-
cellular matrix of targeted host tissues [22]. The surface ad- terial surface causes the bacteria to become more hydrophilic
hesion proteins of the cell wall are referred to collectively as and negatively charged, facilitating cellular attachment.
microbial surface components recognizing adhesive matrix
molecules (MSCRAMMs) [11,22–24]. The MSCRAMMs are
Immune evasion
identified in many species of gram-positive organisms, of
which several are unique to S. aureus. When innate immune mechanisms are insufficient to clear
Products secreted by S. aureus include those that enhance a bacterial infection, adaptive immune mechanisms are ac-
microbial adherence to host tissues, exotoxins that are di- tivated [33–35]. The adaptive immune response to S. aureus
rectly toxic to host cells, and enzymes that degrade and digest requires an integrated response involving T-helper type 1
the extracellular matrix. Secretable expanded repertoire ad- (TH1), TH17, and humoral antibody (IgG) responses, gener-
hesive molecules (SERAMs) are a group of secreted proteins ating the production of interleukin (IL)-1 and IL-17A to
that have duplicative functions of the MSCRAMMs identi- promote abscess formation, which in turn is required for the
fied above, except that they are secreted proteins, not part of clearance of bacteria via phagocytosis and oxidative burst.
the staphylococcal cell wall [25]. The SERAMs of considerable Staphylococcal infections in human beings are known to
interest for S. aureus are coagulase (of which there are actually elicit only modest, transitory antibody responses [36,37], two
two, staphylocoagulase and von Willebrand factor binding tangible manifestations of which are the nasal carrier state
protein), which target prothrombin and fibrinogen. The result of (see above) and recurrent acute infections but also begging
the action of coagulases is generation of a staphylococcal– the question as to whether active immunization against
fibrin matrix and creation of a local environment that facilitates staphylococcal infections will be of benefit. Staphylococci
microbial adherence to tissue and a protected environment can exist as human commensal flora because several mech-
against host defense mechanisms and antimicrobial therapy anisms to ‘‘evade’’ detection by immune surveillance mech-
[26]. Coagulases have effects that are analogous to clumping anisms (e.g., complement deposition, neutrophil killing)
factor of the MSCRAMMs, but they are freely secreted and have evolved [36]. Elaborated enterotoxins act as potent
unique proteins [27]. T-lymphocyte superantigens, which bind major histocom-
Three exotoxin families predominate in the pathogenesis of patibility complex class II (MHC-II) determinants on the
S. aureus infections and allergic responses, namely pore- surface of antigen-presenting cells, but the multiplicity of
forming toxins, exfoliative toxins, and superantigens. Exotoxins these many toxins, both in number and functional redundancy
are those secreted products that are cytotoxic to host cells [28– of staphylococcal enterotoxins and toxic shock syndrome
30], whereas superantigens are secreted products that provoke toxins (TSSTs), has prevented development of a superantigen
an exuberant inflammatory response by the host [31,32]. In- vaccine (see below).
dividual cytotoxins have different cell targets but generally Also during infection, S. aureus produces adenosine
function similarly by damaging the cell membrane, creating (via adenosine synthase A-AdsA) from high-energy phos-
pores, and leading to extravasation of cellular contents. phates [38], which may be the most potent immunosup-
pressive signaling molecule. Adenosine receptor-mediated
signaling impedes phagocytosis by neutrophils (staphylo-
Expression of virulence
cocci that express wild-type AdsA are capable of surviving
Staphylococcus aureus secretes numerous virulence fac- neutrophil phagocytosis) and may degrade adaptive host
tors, including the aforementioned toxins capable of mem- immune responses by impairing antigen presentation to
IMMUNIZATION AGAINST STAPHYLOCOCCUS AUREUS 753

macrophages and dendritic cells. The B-cell superantigens nate anti-staphylococcal antibodies are directed) [47],
staphylococcal protein A (SpA) [39] and S. aureus binder of whereas the antibiotic chosen was the rifamycin derivative
immunoglobulin (Sbi) [40] (both of which harbor multiple rifalogue, which is bactericidal in the acidic milieu of the
immunoglobin-binding domains) inhibit opsonophagocytic phagolysosome and active against non-replicating bacteria
clearance of the pathogen, and may inhibit normal B- and antibiotic-tolerant persister cells. The antibody recog-
lymphocyte functioning, possibly by induction of clonal ex- nizes cell-free S. aureus, leading to opsonization and
pansion leading to B-call apoptosis, or by inhibiting receptor phagocytosis. Once intracellular, rifalogue is cleaved by ca-
interaction of complement factor C3. The SpA protein me- thepsins and exerts its effect. Preliminary work showed that
diates binding of IgG in an incorrect orientation on the bac- the conjugate is ineffective against planktonic S. aureus and
terial surface, leading to decreased neutrophil binding and that the conjugate does not penetrate mammalian cells be-
consequent evasion of phagocytosis. cause of the size of the antibody.

Staphylococcus aureus as a Target for Passive Staphylococcus aureus as a Target for Active
Immunization Immunization
Passive immunization against S. aureus has been unsuc- Prevention of S. aureus infection by an effective vaccine
cessful to date [41]. No efficacy was seen from infusion of would save lives and cost, be antibiotic sparing, and hinder the
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

enriched human serum (IgG infusion) from unvaccinated development of antibiotic resistance [49,50]. If an effective
subjects [42, cited in Janssen et al. (41)]. Neither was efficacy vaccine could be targeted for prevention when the risk of be-
demonstrated from either of two trials of immune serum col- coming infected is highest (e.g., in the short-term aftermath of
lected from subjects who received a capsular polysaccharide surgery or some other invasive procedure), success might be
(CP) CP5/CP8/Pseudomonas exotoxin conjugate vaccine achieved, because the need to provide immunity would exist
[43,44 (cited in Janssen et al. (41),45]. More recently, a phase 2 for just a limited time. An ideal vaccine would have three main
trial failed to prevent S. aureus ventilator-associated pneumonia attributes: Induction of antibodies to neutralize toxins causing
after pre-treatment of patients with heavy airway colonization inflammation and tissue necrosis; induction of antibodies
by S. aureus with an anti-staphylococcal monoclonal antibody against CWA proteins believed to be important in the initial
(ASN100) [46]. ASN100 is a combination of two monoclonal attachment of bacteria to host ligands; and the ability to induce
antibodies (ASN-1 and ASN-2) that together neutralize six S. a robust cytokine-mediated response to promote neutrophil
aureus cytotoxins. ASN-1 neutralizes a-hemolysin and four of recruitment and effective bacterial clearance. A successful anti-
the five leukocidins (HlgAB, HlgCB, LukED, and LukSF, the staphylococcal vaccine would not have to prevent infection by
latter also known as Panton-Valentine leukocidin or PVL). MRSA specifically, because putative vaccine targets could be
ASN-2 neutralizes the fifth leukocidin, LukGH (also known as independent of elements responsible for antibiotic resistance.
LukAB). LukGH, a potent immune evasion factor of S. aureus, Development of a vaccine directed against several strains
is expressed by the majority of all clinical isolates and con- will require biologic finesse. Although anti-staphylococcal
tributes substantially to S. aureus-mediated phagocyte killing. vaccines have been raised against a variety of targets, mul-
Attempts continue to develop effective passive anti- tiple efforts thus far have failed to demonstrate efficacy, in-
staphylococcal immunity. Although S. aureus is an extra- cluding in phase 3 trials. In a double-blind trial involving
cellular bacterium, it can evade host defenses by adapting as patients with end-stage renal disease who were undergoing
an intracellular organism, where it can evade the action of hemodialysis, Shinefield et al. [51] evaluated the safety,
antibodies but also antibacterial agents and in so doing, serve immunogenicity, and efficacy of a vaccine with S. aureus CP5
as a nidus for dissemination. A novel antibody–antibiotic and CP8 capsular polysaccharides conjugated to nontoxic
conjugate is being developed, designed to be activated spe- recombinant Pseudomonas aeruginosa exotoxin A. A total of
cifically inside phagocytes that have engulfed S. aureus 1,804 adult patients (73 hemodialysis centers) were assigned
[47,48]. The antibody is directed against cell wall component randomly to receive a single intramuscular injection of either
teichoic acid (against which approximately one-third of in- vaccine or saline. Immunoglobulin G (IgG) antibodies to both

Table 1. Anti-Staphylococcal Vaccines in Clinical Development


Vaccine designation Developer Antigenic targets Trial phase
SA4Ag Pfizer CP5, CP8, ClfA, MntC 2b (ongoing)
Unknown GSK CP5, CP8, AT, ClfA 1 (completed)
NOV-3A NovaDigm Als3p 1 (completed)
4C-Staph GSK (Novartis) FhuD2m Csa1A, EsxAB 1 (completed)
STEBVax IBT/NIAID SEB 1 (completed)
Unknown Nabi AT/PVL 2 (completed)
Unknown GSK CP5, CP8, AT Pre-clinical
Data derived from Mohamed et al. [53] and Bagnoli et al. [54].
Als = Candida albicans agglutinin-like sequence protein (has three dimensional structural similarity to clumping factor [Clf] A);
AT = alpha-toxin; CP = capsular polysaccharide; Csa = conserved staphylococcal antigen; EsxAB = fusion protein of secreted proteins ess
extracellular A and B; FhuD2 = ferric hydoxamate uptake lipoprotein D2, involved in iron-hydroxamate uptake; GSK = GlaxoSmithKline,
Middlesex, United Kingdom; Mnt = manganese transporter protein; NIAID = National Institute for Arthritis and Infectious Diseases;
PVL = Panton-Valentine leukocidin; SEB = staphylococcal enterotoxin B.
754 BARIE ET AL.

capsular polysaccharides were measured, and episodes of rapid, robust immune responses to each of the antigens, sus-
S. aureus bacteremia (primary end point, assessed at one year) tained for at least 12 mos. Meta-analysis of these trials [58]
were recorded. The capsular polysaccharides elicited an anti- confirmed the robust safety profile.
body response of at least 80 mcg/mL (the estimated minimal Arguably the most promising approach for anti-
level conferring protection) in 80% of patients for CP5 and in staphylococcal vaccination would be to vaccinate prior to
75% of patients for CP8. Between weeks 3 and 40 after vac- the at-risk period (e.g., prior to elective surgery). Not only
cination, S. aureus bacteremia developed in 11 of 892 evalu- would soft tissue infections be targeted (e.g., surgical site
able patients in the vaccine group, compared with 26 of 906 infection), but a vaccine-induced antibody response would
patients in the control group (vaccine efficacy [VE] estimate provide antibodies as well as human matrix proteins access
57%; 95% confidence interval (CI), 10%–81%; p = 0.02). to surgical implants (e.g., prosthetic joints, surgical mesh),
However, the efficacy during weeks 3 to 54 was only 26% and so potentially would, in theory, preclude bacterial ad-
(p = 0.23). Reactions to the vaccine were generally mild-to- hesion to the implant, biofilm formation, and subsequent
moderate, and most resolved within two days. The authors dissemination. Eventually, any type of elective operation
concluded that the vaccine conferred partial immunity against that is plagued by staphylococcal infection could be a can-
S. aureus bacteremia for approximately 40 wks, after which didate for pre-operative vaccination, but an ongoing trial
protection waned as antibody concentrations decreased. (Staphylococcus aureus Surgical Inpatient Vaccine Efficacy
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

In view of the failure of that trial, a second study in he- [STRIVE]) is targeting the vaccination of patients under-
modialysis patients evaluated the VE of two vaccine doses going spine surgery with multi-level instrumentation from
(rather than the single dose in the prior study) [52]. In a 10 to 60 d prior to surgery [59]. Future studies might target
double-blind trial, 3,359 patients were randomly assigned infra-inguinal vascular surgery with insertion of a prosthetic
(1:1) to receive vaccine or placebo at week 0 and 35. The VE graft, open incisional hernia repair with insertion of mesh,
in preventing S. aureus bacteremia was assessed between 3 or other high-risk (for staphylococcal infection) elective
and 35 wks and 3 and 60 wks post-dose 1. Serious adverse operations.
events (SAEs) were recorded for 42 d post-vaccination and
deaths until study end. No substantial difference in the inci- Author Disclosure Statement
dence of S. aureus bacteremia was observed between weeks
3–35 post-dose 1 (VE -23%, 95% CI: -98%–23%, p = 0.39) Doctor Barie is a consultant for and has received honoraria
or at 3–60 wks post-dose 1 (VE -8%, 95% CI: -57%–26%, from Arsanis, Inc. and Pfizer, Inc. Doctor Narayan has
p = 0.70). Serious adverse events were reported by 24% and nothing to disclose. Doctor Sawyer is a consultant for and has
25% of vaccine and placebo recipients, respectively. These received honoraria from Pfizer, Inc.
data did not show a protective effect of either one or two
vaccine doses against S. aureus bacteremia in hemodialysis References
patients despite a robust immune response and an acceptable 1. Kao JH. Hepatitis B vaccination and prevention of hepa-
safety profile. The authors suggested a need to expand the tocellular carcinoma. Best Pract Res Clin Gastroenterol
antigen composition of the vaccine among possible explana- 2015;29:907–917.
tions for the failure of the trial. 2. Adrian M, Di Bisceglie MD. Hepatitis B and hepatocellular
Several additional anti-staphylococcal vaccines have carcinoma. Hepatology 2009;49(5 Suppl):S56–S60.
reached clinical development, using various antigenic tar- 3. Signorelli C, Odone A, Ciorba V, et al. Human papillo-
gets (Table 1) [53,54]. Immunization against several anti- mavirus 9-valent vaccine for cancer prevention: A sys-
genic targets has heretofore been unsuccessful (e.g., tematic review of the available evidence. Epidemiol Infect
lipotechoic acid, alpha-toxin), as have single-antigen im- 2017;145:1962–1982.
munization approaches against plausible biologic (protein) 4. Newman PA, Logie CA, Lacombe-Duncan A, et al. Par-
targets. Pfizer (Pearl River, NY) is developing a four-antigen ents’ uptake of human papillomavirus vaccines for their
(SA4Ag) vaccine that targets CP5 and CP8 (both conjugated children: A systematic review and meta-analysis of obser-
to a mutant non-toxic diphtheria toxin carrier protein vational studies. BMJ Open 2018;8:e019206.
[CRM197]), a mutated recombinant ClfA protein (rmClfA), 5. Behbehani AM. The smallpox story: Life and death of an
and a recombinant form of the manganese transporter A old disease. Microbiol Rev 1983;47:457–509.
protein (MntC), an essential metabolic regulator (manga- 6. Plett PC. Peter Plett and other discoverers of cowpox
nese is essential for detoxification of the neutrophil respi- vaccination before Edward Jenner [in German]. Sudhoffs
ratory burst) (Table 1). These targeted virulence factors have Archiv 2006;90:219–232.
7. Riedel S. Edward Jenner and the history of smallpox and
the shared attribute that they are deployed early (experi-
vaccination. Baylor U Med Ctr Proc 2005;18:21–25.
mentally, within one to six hours) during infection and elicit 8. Koplow DA. Smallpox: The Fight to Eradicate a Global
robust functional antibody responses. In a preliminary safety Scourge. Berkeley, CA: University of California Press, 2003.
and efficacy trial [55], 100 participants were vaccinated with a 9. Zepp F. Principles of vaccination. Methods Mol Biol 2016;
single dose of SA4Ag. SA4Ag was well tolerated, with a 1403:57–84.
satisfactory safety profile. On day 29, antibody titers had in- 10. Klein EY, Sun L, Smith DL, Laxminarayan R. The changing
creased 69.2-fold (for CP5) and 28.9-fold (for CP8) and 19.6- epidemiology of methicillin-resistant Staphylococcus aureus
fold (for MntC) and 12.3-fold (for ClfA), respectively. The in the United States: A national observational study. Am J
majority of participants achieved the pre-defined biologically Epidemiol 2013;177:666–674.
relevant thresholds: CP5, 100%; CP8, 97.9%, ClfA, 87.8%; 11. Fry DE, Barie PS. The changing face of Staphylococcus
and MntC, 96.9%. In phase 2/2a studies conducted in healthy aureus: A continuing surgical challenge. Surg Infect 2011;
adults aged 18–64 yrs [56] and 65–85 yrs [57], SA4Ag elicited 12:191–203.
IMMUNIZATION AGAINST STAPHYLOCOCCUS AUREUS 755

12. Laupland KB, Church DL, Mucenski M, et al. Population- and host-expressed proteases. Front Cell Infect Microbiol
based study of the epidemiology of and risk factors for 2017;7:166.
invasive Staphylococcus aureus infections. J Infect Dis 31. Grumann D, Nübel U, Bröker BM. Staphylococcus aureus
2003;187:1452–1459. toxins—Their functions and genetics. Infect Genet Evol
13. Jacobsson G, Dashti S, Whalberg T, et al. The epidemi- 2014;21:583–592.
ology of and risk factors for invasive Staphylococcus au- 32. Stach CS, Herrera A, Schlievert PM. Staphylococcal su-
reus infections in western Sweden. Scand J Infect Dis 2007; perantigens interact with multiple host receptors to cause
39:6–15. serious diseases. Immunol Res 2014;59:177–181.
14. Mehraj J, Witte W, Akmatov MK, et al. Epidemiology of 33. Li Z, Peres AG, Damian AC, Madrenas J. Im-
Staphylococcus aureus nasal carriage patterns in the com- munomodulation and disease tolerance to Staphylococcus
munity. Curr Top Microbiol Immunol 2016;398:55–87. aureus. Pathogens 2015;4:793–815.
15. Wertheim HF, Melles DC, Vos MC, et al. The role of nasal 34. Lacey KA, Geoghegan JA, McLoughlin RM. The role of
carriage in Staphylococcus aureus infections. Lancet Infect Staphylococcus aureus virulence factors in skin infection
Dis 2005;5:751–762. and their potential as vaccine agents. Pathogens 2016;5:22.
16. Ammerlaan HS, Kluytmans JA, Wertheim HF, et al. Era- 35. Foster TJ. The remarkably multifunctional fibronectin
dication of methicillin-resistant Staphylococcus aureus binding proteins of Staphylococcus aureus. Eur J Clin
carriage: A systematic review. Clin Infect Dis 2009;48: Microbiol Infect Dis 2016;35:1923–1931.
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

922–930. 36. Kim HK, Thammavongsa V, Schneewind O, Missiakas DM.


17. Edmiston CE Jr, Ledeboer NA, Buchan BW, et al. Is Recurrent infections and immune evasion strategies of Sta-
staphylococcal screening and suppression an effective in- phylococcus aureus. Curr Opin Microbiol 2012;15:92–99.
terventional strategy for reduction of surgical site infec- 37. Jacobsson G, Colque-Navarro P, Gustafsson E, et al. An-
tion? Surg Infect 2016;17:158–166. tibody responses in patients with invasive Staphylococcus
18. Bhattacharya M, Wozniak DJ, Stoodley P, Hall-Stoodley L. aureus infections. Eur J Clin [39]Microbiol Infect Dis
Prevention and treatment of Staphylococcus aureus bio- 2010;29:715–725.
films. Expert Rev Anti Infect Ther 2015;13:1499–1516. 38. Thammavongsa V, Kern JW, Missiakas DM, Schneewind
19. Giulieri SG, Holmes NE, Stinear TP, Howden BP. Use of O. Staphylococcus aureus synthesizes adenosine to escape
bacterial whole-genome sequencing to understand and im- host immune responses. J Exp Med 2009;206:2417–2427.
prove the management of invasive Staphylococcus aureus 39. Goodyear CS, Silverman GJ, Staphylococcal toxin induced
infections. Expert Rev Anti Infect Ther 2016;14:1023– preferential and prolonged in vivo deletion of innate-like B
1036. lymphocytes. Proc Nat Acad Sci USA 2004;101:11392–
20. Planet PJ, Narechania A, Chen L, et al. Architecture of a 11397.
species: Phylogenomics of Staphylococcus aureus. Trends 40. Smith EJ, Visai L, Kerrigan SW, et al. The sbi protein is a
Microbiol 2017;25:153–166. multifunctional immune evasion factor of Staphylococcus
21. Lacey KA, Geoghegan JA, McLoughlin RM. The role of aureus. Infect Immun 2011;79:3801–3809.
Staphylococcus aureus virulence factors in skin infection and 41. Jansen KU, Girgenti DQ, Scully IL, Anderson AS. Vaccine
their potential as vaccine antigens. Pathogens 2016;5:22. review: Staphylococcus aureus vaccines: Problems and
22. Patti JM, Allen BL, McGavin MJ, et al. MCSRAAM- prospects. Vaccine 2013;31:2723–2730.
mediated adherence of microorganisms to host tissues. 42. Bristol-Myers Squibb. Safety and efficacy of Veronate
Annu Rev Microbiol 1994;48:585–617. versus placebo in preventing nosocomial staphylococcal
23. Lindsay JA, Holden MTG. Staphylococcus aureus: Su- sepsis in premature infants. ClinicalTrials.gov. Bethesda,
perbug, super genome? Trends Microbiol 2004;12:378–385. MD: National Library of Medicine, 2006.
24. Plata K, Rosat AE, Wegrzyn G. Staphylococcus aureus as 43. NabiBiopharmaceuticals. Safety study of an intravenous
an infectious agent: Overview of biochemistry and molec- iStaphylococcus aureus immune globulin (human), [altas-
ular genetics of its pathogenicity. Acta Biochim Polon taph] in low-birth-weight neonates. ClinicalTrials.gov.
2009;56:597–617. Bethesda, MD: National Library of Medicine, 2012.
25. Chavakis T, Wiechmann K, Preissner KT, Herrmann M. 44. NabiBiopharmaceuticals. Safety and behavior of S. aureus
Staphylococcus aureus interactions with the endothelium: immune globulin (human), [altastaph] in patients with S.
The role of bacterial ‘‘secretable expanded repertoire ad- aureus bacteremia and continuing fever. ClinicalTrials.gov.
hesive molecules’’ (SERAM) in disturbing host defense Bethesda, MD: National Library of Medicine, 2012.
systems. Thromb Haemost 2005;94:278–285. 45. Rupp ME, Holley Jr HP, Lutz J et al. Phase II, randomized,
26. Peetermans M, Verhamme P, Vanassche T. Coagulase ac- multicenter, double-blind, placebo-controlled trial of a
tivity by Staphylococcus aureus: A potential target for polyclonal anti-Staphylococcus aureus capsular polysac-
therapy? Semin Thromb Hemost 2015;41:433–444. charide immune globulin in treatment of Staphylococcus
27. McDevitt D, Vaudaux PE, Foster TJ, et al. Genetic evi- aureus bacteremia. Antimicrob Agents Chemother 2007;
dence that bound coagulase of Staphylococcus aureus is not 51:4249–4254.
clumping factor. Infect Immun 1992;60:1514–1523. 46. Arsanis provides update following completion of planned
28. Dinges MM, Orwin PM, Schlievert PM. Exotoxins of Sta- interim analysis of phase 2 clinical trial of ASN100. http://
phylococcus aureus. Clin Microbiol Rev 2000;13:16–34, investors.arsanis.com/news-releases/news-release-details/
29. Alonzo F 3rd, Torres VJ. The bicomponent pore-forming arsanis-provides-update-following-completion-planned-
leucocidins of Staphylococcus aureus. Microbiol Mol Biol interim. (Last accessed November 5, 2018).
Rev 2014;78:199–230. 47. Lehar SM, Pillow T, Xu, M. Novel antibody-antibiotic
30. Pietrocola G, Nobile G, Rindi S, Speziale P. Staphylo- conjugate eliminates intracellular S. aureus. Nature 2015;
coccus aureus manipulates innate immunity through own 527:323–328.
756 BARIE ET AL.

48. Shopsin B, Kaveri SV, Bayry J. Tackling difficult Staphy- aureus vaccine (SA4Ag): Results from a first-in-human
lococcus aureus infections: Antibodies show the way. Cell randomised, placebo-controlled phase 1/2 study. Vaccine
Host Microbe 2016;20:555–557. 2017;35:375–384.
49. Jansen KU, Anderson AS. The role of vaccines in fighting 57. Creech CB, Frenck RW Jr, Sheldon EA, et al. Safety, tol-
antimicrobial resistance (AMR). Hum Vaccine Immunother erability, and immunogenicity of a single dose 4-antigen or
2018;14:2142–2149. 3-antigen Staphylococcus aureus vaccine in healthy older
50. Jansen KU, Knirsch C, Anderson AS. The role of vaccines adults: Results of a randomised trial. Vaccine 2017;35:385–
in preventing bacterial antimicrobial resistance. Nat Med 394.
2018;24:10–19. 58. Xu X, Zhu H, Lv H. Safety of Staphylococcus aureus four-
51. Shinefield H, Black S, Fattom A, et al. Use of a Staphy- antigen and three-antigen vaccines in healthy adults: A
lococcus aureus conjugate vaccine in patients undergoing meta-analysis of randomized controlled trials. Hum Vaccin
hemodialysis. N Engl J Med 2002;346:491–496. Immunother 2018;14:314–321.
52. Fattom A, Matalon A, Buerkert J, et al. Efficacy profile of a 59. Safety and Efficacy of SA4Ag vaccine in adults having
Staphylococcus aureus glycoconjugated vaccine in adults elective open posterior spinal fusion procedures with mul-
on hemodialysis: Phase III randomized study. Hum Vaccin tilevel instrumentation (STRIVE). https://clinicaltrials.gov/
Immunother 2015;11:632–641. ct2/show/NCT02388165?cond=Staphylococcus+Aureus+
53. Mohamed N, Wang MY, Le Huec J-C, et al. Vaccine de- vaccination&rank=10 (Last accessed November 5, 2018).
Downloaded by Republic of Moldova Hinari Band 1 from www.liebertpub.com at 08/29/19. For personal use only.

velopment to prevent Staphylococcus aureus surgical-site


infections. Br J Surg 2017;104:e41–e54.
54. Bagnoli F, Fontana MR, Soldaini E, et al. Vaccine com- Address correspondence to:
position formulated with a novel TLR7-dependent adjuvant Dr. Philip S. Barie
indices high and broad protection against Staphylococcus Department of Surgery, P713A
aureus. Proc Nat Acad Sci USA 2015;112:3680–3685. New York-Presbyterian Hospital/
55. Begier E, Seiden DJ, Patton M, et al. SA4Ag, a 4-antigen Weill Cornell Medical Center
Staphylococcus aureus vaccine, rapidly induces high levels of 525 East 68th Street
bacteria-killing antibodies. Vaccine 2017;22;35:1132–1139. New York, NY 10065
56. Frenck RW Jr, Creech CB, Sheldon EA, et al. Safety, tol-
erability, and immunogenicity of a 4-antigen Staphylococcus E-mail: pbarie@med.cornell.edu

S-ar putea să vă placă și