Next Article in Journal
Adjunct Culture of Non-Starter Lactic Acid Bacteria for the Production of Provola Dei Nebrodi PDO Cheese: In Vitro Screening and Pilot-Scale Cheese-Making
Next Article in Special Issue
No Change, No Life? What We Know about Phase Variation in Staphylococcus aureus
Previous Article in Journal
Elicitation of Antimicrobial Active Compounds by Streptomyces-Fungus Co-Cultures
Previous Article in Special Issue
Molecular Characterization of Staphylococcus aureus Isolated from Chronic Infected Wounds in Rural Ghana
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Performance of a Four-Antigen Staphylococcus aureus Vaccine in Preclinical Models of Invasive S. aureus Disease

Pfizer Vaccine Research & Development, Pearl River, NY 10965, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2021, 9(1), 177; https://doi.org/10.3390/microorganisms9010177
Submission received: 8 December 2020 / Revised: 7 January 2021 / Accepted: 7 January 2021 / Published: 15 January 2021
(This article belongs to the Special Issue Staphylococcal Infections (Host and Pathogenic Factors))

Abstract

:
A Staphylococcus aureus four-antigen vaccine (SA4Ag) was designed for the prevention of invasive disease in surgical patients. The vaccine is composed of capsular polysaccharide type 5 and type 8 CRM197 conjugates, a clumping factor A mutant (Y338A-ClfA) and manganese transporter subunit C (MntC). S. aureus pathogenicity is characterized by an ability to rapidly adapt to the host environment during infection, which can progress from a local infection to sepsis and invasion of distant organs. To test the protective capacity of the SA4Ag vaccine against progressive disease stages of an invasive S. aureus infection, a deep tissue infection mouse model, a bacteremia mouse model, a pyelonephritis model, and a rat model of infectious endocarditis were utilized. SA4Ag vaccination significantly reduced the bacterial burden in deep tissue infection, in bacteremia, and in the pyelonephritis model. Complete prevention of infection was demonstrated in a clinically relevant endocarditis model. Unfortunately, these positive preclinical findings with SA4Ag did not prove the clinical utility of SA4Ag in the prevention of surgery-associated invasive S. aureus infection.

1. Introduction

Staphylococcus aureus is a Gram-positive bacterium that is carried asymptomatically in the nares of 20–50% of the general population [1]. However, upon a breach of skin or mucosal barriers, it can cause a wide spectrum of diseases, ranging from relatively mild skin infections, such as carbuncles, to life-threatening wound and bloodstream infections [2]. S. aureus infections following surgery carry particularly high mortality rates, and survivors require an additional 13–17 days in the hospital, significantly increasing healthcare costs [3]. The burden of S. aureus disease is exacerbated by the prevalence of antibiotic-resistant S. aureus isolates [4], highlighting the need for an effective prophylactic vaccine.
A consideration in both development and preclinical evaluation of a S. aureus vaccine is the organism’s ability to rapidly adapt to the host microenvironment [5]. S. aureus can enter normally sterile sites through lesions such as those created during surgery or traumatic injury and rapidly deploy an array of pathogenesis mechanisms, rendering S. aureus a challenging vaccine target. Consequently, a licensed vaccine against S. aureus disease is not yet available, and a clinically validated correlate of protection has not yet been established. Prevention strategies for patients at high risk for invasive S. aureus disease, such as surgical patients, include decolonization and antibiotic prophylaxis [6]. These procedures are limited by the variable effectiveness of decolonization and by the development of antibiotic resistance [7,8,9]. Thus, alternative preventative strategies, such as prophylactic vaccines, would be a welcome addition to the clinician’s armamentarium.
With that aim, a four-antigen S. aureus vaccine (SA4Ag) was designed for the prevention of invasive S. aureus infections following elective surgery. Each vaccine component was carefully selected so that, when combined, the vaccine would contravene major S. aureus pathogenesis mechanisms, namely initial adhesion events (ClfA), nutrient acquisition sustaining growth (MntC), and evasion of neutrophil-mediated killing (MntC and capsular polysaccharide (CP) type 5 and type 8 conjugates). The adhesin ClfA enables the attachment of S. aureus to human fibrinogen, and antibodies directed against this protein inhibit ClfA-mediated fibrinogen binding [10]. MntC is a highly conserved component of the manganese transporter MntABC that is quickly upregulated in vivo [11] and is expressed during biofilm formation in animal models [12]. Manganese acquisition by S. aureus is important for both growth and evasion of neutrophil killing through detoxifying oxygen radicals [11,13]. Finally, the vaccine contains capsular polysaccharide type 5 and type 8 conjugated to cross-reactive material 197 (CRM197), which induce functional antibodies that kill the bacteria following opsonophagocytosis [14,15], the process whereby antibodies coat the bacterium, fix complement, and induce uptake and killing by phagocytes, such as neutrophils. It has become clear that antibodies that bind to a bacterial antigen, such as those measured by an enzyme-linked immunosorbent assay, are not always functional, especially in adults with pre-existing exposure to a pathogen. Thus, demonstration of functional antibody activity, such as through an opsonophagocytic killing assay or fibrinogen binding assay is important as another step in the evaluation of a vaccine candidate, along with in vivo proof of concept studies.
The correlate of protection for S. aureus is not yet known, and a vaccine for the prevention of S. aureus disease has yet to be commercially licensed. The two candidates previously advanced to the clinic were supported by preclinical serology [16] and/or murine sepsis model studies [17,18]. These studies demonstrated that the vaccine candidates were immunogenic in preclinical models, an important first step in selecting candidates to advance to the clinic. In the case of iron-regulated surface determinant B (IsdB), a single challenge model, the murine sepsis model, was used to support preclinical efficacy [17,18]. The murine sepsis model mimics hematogenous spread through the body, one important aspect of S. aureus infection. However, S. aureus has notoriously complex pathogenesis mechanisms, which can only be modeled using multiple preclinical models. The development of relevant preclinical models for S. aureus is challenging, as S. aureus is adapted to the human host environment, and preclinical models have failed to predict clinical efficacy [19,20]. Here, the ability of SA4Ag to protect against invasive disease is demonstrated in three preclinical rodent models of S. aureus infection, each mimicking a distinct phase of S. aureus infection, namely deep tissue invasion, bacteremia, and distal infection. In the absence of a defined correlate of protection, and due to the limited ability of any single preclinical model to predict S. aureus vaccine clinical success, demonstrating efficacy in multiple models of S. aureus invasive disease is relevant for advancing a vaccine into clinical trials.

2. Materials and Methods

2.1. Bacterial Strains

The S. aureus clinical isolates from Pfizer’s internal collection that were used for in vivo analyses are listed in Table 1. These strains represent a diverse set of clinical isolates.

2.2. Animal Studies

All animal work was performed in strict accordance with approved Institutional Animal Care and Use Committee protocols at an Association for Assessment and Accreditation of Laboratory Animal Care, International-accredited facility, under the following Animal Use Protocols: PRL-2011-00105 (approved in 2007), PRL-2011-00249 (approved in 2008), PRL-2011-00102 (approved in 2002) and PRL-2011-00338 (approved in 2002) for mouse studies, and PRL-2011-00440 (approved in 2002) for rat studies. For all animal studies, statistical significance was determined via Student’s t-test using Welch’s correction, and a p value of ≤ 0.05 was considered significant (GraphPad Prism). For each model, bacterial challenge dose and immunization schedule were optimized.

2.2.1. Surgical Site Infection Mouse Model

CD-1 female mice (6–8–week–old, 10–20/group; Charles River Laboratories) were vaccinated subcutaneously at weeks 0, 3, and 6 with 100 µL volume containing 1 µg CP8-CRM197 + 1 µg CP5-CRM197 + 10 µg Y338A ClfA + 10 µg MntC (SA4Ag, Pfizer, described in [11,14,21]) in QS-21 (Pfizer) or QS-21 alone. Two weeks after the final vaccination, animals underwent surgery, where a small incision was made in the thigh muscle parallel to the femur [22]. A stitch was placed in the deep tissue, then ~300 colony forming units (CFU) of S. aureus PFESA0158 in 10 µL phosphate-buffered saline (PBS) was instilled into the surgical site, which was then closed. Two days post-challenge, the mice were euthanized, quadriceps muscles were collected and homogenized, and serial dilutions of tissue homogenate were plated on tryptic soy agar (TSA; Becton Dickinson, Franklin Lakes, NJ, USA) plates (Becton Dickinson) to enumerate bacterial burden.

2.2.2. Murine Bacteremia Model

Groups of 10 female (6–8–week–old) CD-1 mice (Charles River Laboratories, Wilmington, MA, USA) were vaccinated by subcutaneous injection at weeks 0, 3, and 6 with either vehicle or SA4Ag (Pfizer) in 23 µg AlPO4 as adjuvant. On week 8, the animals were challenged by intraperitoneal injection with ~4 × 108 CFU of S. aureus CDC3 or PFESA0241. Three hours after challenge animals were exsanguinated by cardiac puncture and serial dilutions of blood plated on TSA to enumerate CFU.

2.2.3. Murine Pyelonephritis Model

Groups of 5 female (6–8 week–old) CD-1 mice (Charles River Laboratories) were vaccinated by subcutaneous injection at weeks 0, 3, and 6 with either vehicle or SA4Ag (Pfizer, New York, NY, USA) in 23 µg AlPO4 as adjuvant. On week 8, the animals were challenged by intraperitoneal injection with ~2 × 108 CFU of S. aureus Reynolds. Two days after challenge, kidneys were harvested and homogenized, and serial dilutions of homogenate plated on TSA to enumerate CFU.

2.2.4. Rat Endocarditis Model

The vaccine was tested in two rat endocarditis models, a standard model [23] and a refined model that more accurately represents clinical disease.

Standard Model

Groups of female Sprague Dawley rats (5–6–week–old, 20/group, Charles River Laboratories) were vaccinated at weeks 0, 3, and 6, with SA4Ag in AlPO4 or with placebo. At week 8, two weeks after the final vaccination, catheter placement surgery was performed to generate sterile valvular vegetations [23]. Due to the complex nature of the model and surgery required, a proportion (<10%) of animals succumbed before infectious challenge due to the surgery and were thus not included in the analysis. Two days after surgery, animals were challenged intravenously with ~4 × 106 CFU of S. aureus PFESA0158 in 0.1 mL PBS. Two days post-challenge, the rats were euthanized and hearts collected. Bacteria in homogenized heart tissue were enumerated (CFU/mL), and the arithmetic mean and 95% confidence interval (CI) were calculated for each treatment group.

Refined Model

In order to reflect the low challenge inoculum that has been reported in human infections [24], the endocarditis model was refined. Groups of female Sprague Dawley rats (5–6–week–old, 14/group; Charles River Laboratories) were vaccinated at weeks 0, 3, and 6, with SA4Ag in AlPO4 or with AlPO4 alone, and catheter placement surgery was conducted on week 8 as described above. Two days after surgery, animals were challenged intravenously with ~4 × 103 CFU of S. aureus PFESA0186. Two days post-challenge, the rats were euthanized and hearts and kidneys collected. Tissues were plated and the presence or absence of bacterial CFU scored.

2.3. Assessment of Immune Responses to Vaccination

2.3.1. Opsonophagocytic Activity Assay

Serologic responses to capsular polysaccharides CP5 and CP8 were measured by an opsonophagocytic activity (OPA) assay, as previously described [14]. Briefly, serial dilutions of heat-inactivated immune sera were mixed with either a CP5-expressing or CP8-expressing clinical isolate of S. aureus and allowed to opsonize the bacteria. The reaction mixtures were then mixed with baby rabbit complement (Pel-Freez, Rogers, AR, USA) and neutrophil-like HL-60 cells (ATCC® CCL-240™, Manassas, VA, USA). An OPA antibody titer was defined as the reciprocal of the highest serum dilution resulting in 50% reduction of the number of bacterial colonies after incubation for 60 min at 37 °C when compared to the background control from which serum was omitted. Samples without detectable activity at the lowest serum dilution of 100 were assigned OPA titer values of 50.

2.3.2. Competitive Luminex Immunoassay

Competitive Luminex-based immunoassays (cLIA) were used to quantify antigen-specific binding antibodies elicited by the investigational vaccine. The assays monitor the ability of each vaccine component to elicit antibodies that can compete with the binding of antigen-specific monoclonal antibodies (mAbs) that have shown functional activity either in vitro or in vivo. The ClfA mAb prevents binding of live S. aureus to fibrinogen [10], while the MntC mAb inhibits manganese uptake [25,26]. The mAbs used for the CP antigens facilitated killing of S. aureus as measured by the OPA assay. Spectrally distinct Luminex microspheres were coated individually with each of the antigens and incubated overnight with appropriately diluted serum samples. A mixture of phycoerythrin (PE)-labeled CP5-, CP8-, rmClfA-, and rP305A-specific mouse mAbs is then added to the microsphere/serum mixture, and after incubation, the unbound components are washed off. After reading in a Bio-Plex reader (Bio-Rad, Hercules, CA, USA), signals are converted to Units/mL.

3. Results

3.1. SA4Ag Is Immunogenic in Mice, Rats and Non-Human Primates

SA4Ag is comprised of capsular polysaccharides type 5 and type 8 CRM197-conjugates, ClfA, and MntC. SA4Ag was also shown to be able to elicit functional antibody responses in mice, rats and non-human primates, as measured by the OPA assay, which monitors the ability of serum samples to opsonize and induce uptake and killing of target bacteria by a neutrophil-like cell line, or by cLIA, which monitors the ability of serum to compete with mAbs for antigen binding. Importantly, the mAbs used in these assays inhibit the pathological function of the antigens they bind—e.g., fibrinogen binding in the case of ClfA [10] and manganese uptake in the case of MntC [26] (Figure 1). As Figure 1 shows, humans and non-human primates mount responses to the antigens in SA4Ag after a single dose (PD1), even in the absence of adjuvant, while rodents (mice and rats) require multiple immunizations with an adjuvant (e.g., AlPO4) to generate similar magnitude responses. This is likely due to the induction of an anamnestic response to the antigens in SA4Ag in non-human primates and humans, which is absent in the rodent species.

3.2. Immunization with SA4Ag Reduces Bacterial Burden in a Murine Model of Surgical Site Infection

Immunization with SA4Ag was evaluated in a murine model of surgical site infection. Analogous to human deep tissue surgical site infection, S. aureus was not introduced systemically but instead a low number of bacterial cells were inoculated into the surgical wound. As shown in Figure 1 and observed by others [28], mice respond relatively poorly to ClfA immunization, in comparison to both rats and humans, even in the presence of AlPO4 adjuvant. To enhance the ClfA response in a model where initial adhesion events are likely important, we considered the addition of an alternate, non–alum-based adjuvant. We limited our selection to adjuvants that are usable in human clinical trials, as some highly reactogenic adjuvants, such as Freund’s, overinflate immunogenicity. QS-21, a derivative of the bark of the Quillaja saponaria tree, has been used in human clinical trials and is purported to induce a more balanced IgG1/IgG2a response than alum-containing adjuvants in mice [29,30]. Addition of QS-21 to SA4Ag resulted in enhanced ClfA responses (geometric mean titer: 516.4; 95% CI: 222.4–1199.0; unpaired t-test with Welch’s correction: p = 0.048) as compared with AlPO4 (geometric mean titer: 42.8; 95% CI: 17.9–102.1). In the murine surgical site infection model, immunization with SA4Ag adjuvanted with QS-21 reduced bacterial burden (Table 2), illustrating an impact of SA4Ag in this difficult local infection model.

3.3. Immunization with SA4Ag Protects against MRSA Challenge in a Murine Bacteremia Model

S. aureus can disseminate from a local infection via the bloodstream. Therefore, SA4Ag was evaluated for its ability to reduce the bacterial burden in a murine bacteremia model, which mimics very early stages of hematogenous spread. Mice were immunized three times with SA4Ag with AlPO4 and then challenged with either S. aureus CDC3 or PFESA0241, both USA300 MRSA isolates. Blood was collected three hours post-challenge and bacteria were enumerated. Immunization with SA4Ag significantly reduced the number of recovered CFU with both USA300 MRSA isolates (Table 3).

3.4. Immunization with SA4Ag Reduces Bacterial Burden in a Murine Pyelonephritis Model

S. aureus can cause infection at sites distant from the initial site of infection, and the kidney is a common end organ for S. aureus infection. A murine pyelonephritis model was used to evaluate the ability of SA4Ag to reduce the bacterial burden in the kidney. Mice were immunized with SA4Ag and then challenged with S. aureus Reynolds. Two days after challenge, kidneys were harvested and homogenized to enumerate bacterial burden. Immunization with SA4Ag significantly reduced the number of recovered CFU from kidneys (Figure 2).

3.5. Immunization with SA4Ag Protects against Both CP5- and CP8-Expressing S. aureus in a Rat Endocarditis Model

S. aureus can cause infection at sites distant from the initial site of infection. SA4Ag was evaluated for its ability to reduce the bacterial burden in a model of foreign body–like infection, i.e., rat endocarditis following catheterization, considered a very stringent preclinical model. A catheter was inserted into the heart via the jugular vein across the aortic valve to create sterile valvular vegetations. Two days after catheter placement, animals were challenged intravenously with S. aureus. In two out of three studies, immunization with SA4Ag reduced the bacterial burden of a CP5-expressing S. aureus clinical isolate from infected heart tissue (Table 4). Meta-analysis of these three studies demonstrated a significant reduction in recovered CFU (Table 5, p = 0.0126).
The endocarditis model was next refined to improve clinical relevance by reducing the challenge inoculum. It is believed that S. aureus-induced foreign body infections and endocarditis are caused by the seeding of very low numbers of bacteria introduced to the bloodstream either during surgery or as a result of minor tissue injury, such as resulting from a scratch or brushing of teeth [24]. To model human bacterial exposure more closely, the infectious inoculum was reduced to a level just above the threshold for achieving an infection. In this model, S. aureus preferentially seeds the damaged valvular tissue and the kidneys; no bacterial burden was detected in the liver, spleen or lungs. In the refined model, the complete absence of detectable infection in both hearts and kidneys following immunization with SA4Ag was evaluated. Rats were immunized subcutaneously three times on weeks 0, 3, and 6 with SA4Ag, and then underwent catheter placement surgery. Two days after catheter placement, rats were challenged intravenously with a CP8-expressing clinical S. aureus isolate, PFESA0186. Immunization with SA4Ag reduced the number of animals with detectable S. aureus infection in the hearts and kidneys in two separate experiments. Meta-analysis of the two experiments showed a significant reduction in the number of infected animals upon immunization, representing a vaccine efficacy of 88.7% (Table 5).

4. Discussion

A four-antigen S. aureus vaccine (SA4Ag) was designed as a candidate to prevent invasive S. aureus disease in postsurgical populations. Therefore, SA4Ag was evaluated for the ability to protect against invasive S. aureus disease in a series of preclinical models that mimic aspects of human postsurgical infection. Individual components of SA4Ag have previously been shown to elicit robust antibody responses and efficacy in rodents [11,14,21,31], but efficacy of the combined SA4Ag vaccine in multiple preclinical models has not previously been reported. SA4Ag was found to significantly reduce or abrogate infection in murine models of surgical site infection, bacteremia, and in two iterations of a rat endocarditis model. These models mimic the progression of S. aureus postsurgical invasive disease from a local deep tissue infection, through hematogenous spread, and dissemination to distant tissue sites, such as the heart valves and kidneys. Together, the responses seen in these models demonstrated that vaccination with SA4Ag elicits an immune response that restrains multiple stages of invasive preclinical S. aureus infection and suggested that similar positive outcomes could be achieved in the clinic. Importantly, in the refined rat endocarditis model, a complete absence of infection was observed in 96% of vaccinated animals, reflecting a vaccine efficacy of 88.7%. This is the first time to our knowledge that a sterilizing immune response has been demonstrated after administration of an S. aureus vaccine in a preclinical model, and supported moving the SA4Ag into clinical development.
S. aureus disease is challenging to effectively model in animals due to the organism’s adaptation to the human host environment and the lack of a defined correlate of protection for invasive S. aureus disease. As demonstrated in Figure 1, rodents, which are commonly used in preclinical models of S. aureus pathogenesis, required three doses of SA4Ag with an adjuvant to achieve immune responses similar to those seen in non-human primates and humans after a single unadjuvanted dose of SA4Ag. It is possible, therefore, that the response of repeatedly immunized naïve animals does not fully recapitulate the maturation of an immune response originally elicited through natural exposure. The response to ClfA may serve as a case in point. Naïve mice respond poorly to ClfA, even after multiple immunizations in the presence of adjuvant, while humans and non-human primates respond well after a single dose. Interestingly, even though humans respond well to a single dose of ClfA-containing SA4Ag, implying ClfA is eliciting an anamnestic response, a functional anti-ClfA response, which can block the binding of ClfA to fibrinogen, is only observed after immunization [10]. This highlights the need to match the right vaccine with antigen presentation in the correct format.
Invasive S. aureus infection can proceed from a local inoculation site, through dissemination in the bloodstream, to distant tissues. A given vaccine candidate may show efficacy in one or more models, but it is the combination of vaccine candidates and evaluation in relevant models of infection that improves the robustness of the conclusions drawn from the preclinical program. Based in part on the preclinical data included here, the S. aureus vaccine was advanced into clinical testing. Prior to the clinical assessment of SA4Ag, two other S. aureus vaccines also failed to demonstrate efficacy in the clinic, despite having supportive preclinical data. Both vaccines had a single antigenic target, either capsular polysaccharide or IsdB. The StaphVax vaccine, comprised of capsular polysaccharides type 5 and type 8 conjugated to Pseudomonas exotoxin A, did not meet its Phase III clinical endpoint. It has been suggested that this may have resulted from quality variations of the conjugate used in the trial, as opposed to a failure of the mechanism of action [32]. Thus, S. aureus capsular polysaccharides had remained an attractive vaccine candidate but potentially not sufficient to protect against invasive S. aureus disease. A second single-antigen S. aureus vaccine, V710, contained the iron uptake component, IsdB, and also did not meet its primary efficacy endpoint (prevention of serious postoperative S. aureus infections following cardiothoracic surgery) in a randomized Phase 2b/3 trial. Additionally, V710 was associated with increased mortality among patients who developed S. aureus infections [33]. IsdB did protect mice in preclinical lethal challenge models [17,18]. However, S. aureus has multiple redundant iron acquisition systems and prefers human hemoglobin as an iron source, which may have limited the predictive value of the models used to evaluate this antigen [34]. In preclinical models, the protection seen with IsdB was improved with incorporation of additional S. aureus antigens [35]. The recognition that a single antigen approach may not be sufficient to protect against S. aureus-mediated disease spurred the development of several multi-antigen approaches in addition to SA4Ag [36,37,38].
In an early Phase 1/2 clinical study (NCT01364571), SA4Ag elicited robust functional immune responses and showed an acceptable safety profile [27]. However, a large randomized, placebo-controlled Phase 2b study (STRIVE; NCT02388165) in adult patients undergoing elective open posterior spinal fusion surgery [39], the results of a pre-planned interim analysis indicated low statistical probability to meet the pre-defined primary efficacy objective—prevention of invasive S. aureus disease (bloodstream and surgical site infections), leading to the decision to terminate the study and discontinue the clinical development [40]. This failure indicates that despite protection seen in the current preclinical studies, the animal models that were used had limitations that did not enable prediction of clinical efficacy of the vaccine. One such potential shortcoming is the measure of protection. For example, the absence of detectable infection was used as the criterion of induced protection for the refined endocarditis model. A second shortcoming is that the infection models all evaluated early time points before any animals could have succumbed to infection. Examination at later time points might have determined any impact of the vaccine on survival, and changes in pathology could be identified (e.g., wound size and induration, paralysis or some other signs of morbidity, or histological changes). Examination of the immune responses elicited by the vaccine in more detail (innate vs. adaptive) might also have provided more insight into the mechanism(s) of protection.
Since there is no human correlate of protection against S. aureus infection, establishing reliable animal models for preclinical evaluation of S. aureus vaccine candidates remains very challenging [19,20]. This is further supported by the observation that while non-human primates required a single dose of SA4Ag without adjuvant to generate functional immune responses, rodents required multiple doses of SA4Ag with adjuvant to achieve similar levels of immune response. Therefore, current preclinical efficacy models play an important yet circumscribed role in illustrating the protective potential of a S. aureus vaccine candidate. Animal models are useful in this context for demonstrating proof of principle or proof of mechanism, but ultimately clinical efficacy data, obtained using carefully selected vaccine formulations and target populations, is required until a reliable correlate of protection is identified [19,20]. Identification of a relevant correlate of protection for invasive S. aureus disease and development of preclinical models that accurately reflect these human correlates is needed to improve effective preclinical evaluation of S. aureus vaccine candidates.

5. Conclusions

Although a S. aureus vaccine candidate, SA4Ag, showed promise in the various preclinical models presented, it ultimately did not show clinical efficacy. SA4Ag is the latest in a series of S. aureus vaccines that showed protection in preclinical models, which did not translate into positive clinical outcomes. This highlights the need to identify relevant correlates of protection for invasive S. aureus infection, and to develop appropriate preclinical models that better predict clinical efficacy.

Author Contributions

Conceptualization, K.U.J., A.S.A., P.A.L., I.L.S.; methodology, Y.T., P.L., A.I.; formal analysis, I.L.S., P.A.L., A.S.A.; writing—original draft preparation, I.L.S., A.S.A.; writing—review and editing, I.L.S., Y.T., A.I., P.L., P.A.L., K.U.J., A.S.A. All authors have read and agreed to the published version of the manuscript.

Funding

The research was funded by Pfizer Inc.

Institutional Review Board Statement

All animal studies were performed in an AAALAC-accredited facility under IACUC-approved protocols.

Informed Consent Statement

Not applicable. This study did not involve humans; human data is reproduced from a cited publication.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors thank the following Pfizer colleagues: excellent and dedicated comparative medicine colleagues who assisted in the procedures reported in this manuscript; Danka Pavliakova and Hong Chen for the provision of cLIA data; and Nataliya Kushnir for editorial assistance.

Conflicts of Interest

I.L.S., Y.T., A.I., P.A.L., K.U.J. and A.S.A. are employees of Pfizer Inc. and may hold stock or stock options. P.L. was an employee of Pfizer at the time the work was done.

References

  1. Kluytmans, J.; van Belkum, A.; Verbrughn, H. Nasal carriage of Staphylococcus aureus: Epidemiology, underlying mechanisms, and associated risks. Clin. Microbiol. Rev. 1997, 10, 505–520. [Google Scholar] [CrossRef]
  2. Lowy, F.D. Staphylococcus aureus infections. N. Engl. J. Med. 1998, 339, 520–532. [Google Scholar] [CrossRef] [PubMed]
  3. Noskin, G.A.; Rubin, R.J.; Schentag, J.J.; Kluytmans, J.; Hedblom, E.C.; Jacobson, C.; Smulders, M.; Gemmen, E.; Bharmal, M. National trends in Staphylococcus aureus infection rates: Impact on economic burden and mortality over a 6-year period (1998–2003). Clin. Infect. Dis. 2007, 45, 1132–1140. [Google Scholar] [CrossRef] [PubMed]
  4. Carrel, M.; Perencevich, E.N.; David, M.Z. USA300 Methicillin-Resistant Staphylococcus aureus, United States, 2000–2013. Emerg. Infect. Dis. 2015, 21, 1973–1980. [Google Scholar] [CrossRef] [PubMed]
  5. Malachowa, N.; Whitney, A.R.; Kobayashi, S.D.; Sturdevant, D.E.; Kennedy, A.D.; Braughton, K.R.; Shabb, D.W.; Diep, B.A.; Chambers, H.F.; Otto, M.; et al. Global changes in Staphylococcus aureus gene expression in human blood. PLoS ONE 2011, 6, e18617. [Google Scholar] [CrossRef] [Green Version]
  6. Bode, L.; Kluytmans, J.; Wertheim, H.; Bogaers, D.; Vandenbroucke-Grauls, C.; Roosendaal, R.; Troelstra, A.; Box, A.; Voss, A.; Van Der Tweel, I.; et al. Preventing surgical-site infections in nasal carriers of Staphylococcus aureus. N. Engl. J. Med. 2010, 362, 9–17. [Google Scholar] [CrossRef] [Green Version]
  7. Kalmeijer, M.D.; Coertjens, H.; Van Nieuwland-Bollen, P.M.; Bogaers-Hofman, D.; De Baere, G.A.J.; Stuurman, A.; Van Belkum, A.; Kluytmans, J.A.J.W. Surgical site infections in orthopedic surgery: The effect of mupirocin nasal ointment in a double-blind, randomized, placebo-controlled study. Clin. Infect. Dis. 2002, 35, 353–358. [Google Scholar] [CrossRef]
  8. Simor, A.E.; Phillips, E.; McGeer, A.; Konvalinka, A.; Loeb, M.; Devlin, H.R.; Kiss, A. Randomized controlled trial of chlorhexidine gluconate for washing, intranasal mupirocin, and rifampin and doxycycline versus no treatment for the eradication of methicillin-resistant Staphylococcus aureus colonization. Clin. Infect. Dis. 2007, 44, 178–185. [Google Scholar] [CrossRef]
  9. Simor, A.E.; Stuart, T.L.; Louie, L.; Watt, C.; Ofner-Agostini, M.; Gravel, D.; Mulvey, M.; Loeb, M.; McGeer, A.; Bryce, E.; et al. Mupirocin-resistant, methicillin-resistant Staphylococcus aureus strains in Canadian hospitals. Antimicrob. Agents Chemother. 2007, 51, 3880–3886. [Google Scholar] [CrossRef] [Green Version]
  10. Hawkins, J.; Kodali, S.; Matsuka, Y.V.; McNeil, L.K.; Mininni, T.; Scully, I.L.; Vernachio, J.H.; Severina, E.; Girgenti, D.; Jansen, K.U.; et al. A recombinant clumping factor A-containing vaccine induces functional antibodies to Staphylococcus aureus that are not observed after natural exposure. Clin. Vaccine Immunol. 2012, 19, 1641–1650. [Google Scholar] [CrossRef] [Green Version]
  11. Anderson, A.S.; Scully, I.L.; Timofeyeva, Y.; Murphy, E.; McNeil, L.K.; Mininni, T.; Nuñez, L.; Carriere, M.; Singer, C.; Dilts, D.A.; et al. Staphylococcus aureus manganese transport protein C is a highly conserved cell surface protein that elicits protective immunity against S. aureus and Staphylococcus epidermidis. J. Infect. Dis. 2012, 205, 1688–1696. [Google Scholar] [CrossRef] [PubMed]
  12. Brady, R.A.; Leid, J.G.; Camper, A.K.; Costerton, J.W.; Shirtliff, M.E. Identification of Staphylococcus aureus proteins recognized by the antibody-mediated immune response to a biofilm infection. Infect. Immun. 2006, 74, 3415–3426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Handke, L.D.; Hawkins, J.C.; Miller, A.A.; Jansen, K.U.; Anderson, A.S. Regulation of Staphylococcus aureus MntC Expression and Its Role in Response to Oxidative Stress. PLoS ONE 2013, 8, e77874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Nanra, J.S.; Buitrago, S.M.; Crawford, S.; Ng, J.; Fink, P.S.; Hawkins, J.; Scully, I.L.; McNeil, L.K.; Aste-Amézaga, J.M.; Cooper, D.; et al. Capsular polysaccharides are an important immune evasion mechanism for Staphylococcus aureus. Hum. Vaccines Immunother. 2012, 9, 480–487. [Google Scholar] [CrossRef] [Green Version]
  15. Thakker, M.; Park, J.S.; Carey, V.; Lee, J.C. Staphylococcus aureus serotype 5 capsular polysaccharide is antiphagocytic and enhances bacterial virulence in a murine bacteremia model. Infect. Immun. 1998, 66, 5183–5189. [Google Scholar] [CrossRef] [Green Version]
  16. Fattom, A.; Schneerson, R.; Szu, S.C.; Vann, W.F.; Shiloach, J.; Karakawa, W.W.; Robbins, J.B. Synthesis and immunologic properties in mice of vaccines composed of Staphylococcus aureus type 5 and type 8 capsular polysaccharides conjugated to Pseudomonas aeruginosa exotoxin A. Infect Immun. 1990, 58, 2367–2374. [Google Scholar] [CrossRef] [Green Version]
  17. Kuklin, N.A.; Clark, D.J.; Secore, S.; Cook, J.; Cope, L.D.; McNeely, T.; Noble, L.; Brown, M.J.; Zorman, J.K.; Wang, X.M.; et al. A novel Staphylococcus aureus vaccine: Iron surface determinant B induces rapid antibody responses in rhesus macaques and specific increased survival in a murine S. aureus sepsis model. Infect. Immun. 2006, 74, 2215–2223. [Google Scholar] [CrossRef] [Green Version]
  18. Joshi, A.; Pancari, G.; Cope, L.; Bowman, E.P.; Cua, D.; Proctor, R.A.; McNeely, T. Immunization with Staphylococcus aureus iron regulated surface determinant B (IsdB) confers protection via Th17/IL17 pathway in a murine sepsis model. Hum. Vaccines Immunother. 2012, 8, 336–346. [Google Scholar] [CrossRef] [Green Version]
  19. Pozzi, C.; Olaniyi, R.; Liljeroos, L.; Galgani, I.; Rappuoli, R.; Bagnoli, F. Vaccines for Staphylococcus aureus and Target Populations. Curr. Top. Microbiol. Immunol. 2017, 409, 491–528. [Google Scholar] [CrossRef]
  20. Jansen, K.U.; Girgenti, D.Q.; Scully, I.L.; Anderson, A.S. Vaccine review: “Staphyloccocus aureus vaccines: Problems and prospects”. Vaccine 2013, 31, 2723–2730. [Google Scholar] [CrossRef]
  21. Scully, I.L.; Timofeyeva, Y.; Keeney, D.; Matsuka, Y.V.; Severina, E.; McNeil, L.K.; Nanra, J.; Hu, G.; Liberator, P.A.; Jansen, K.U.; et al. Demonstration of the preclinical correlate of protection for Staphylococcus aureus clumping factor A in a murine model of infection. Vaccine 2015, 33, 5452–5457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, L.; Lee, J.C. Murine Models of Bacteremia and Surgical Wound Infection for the Evaluation of Staphylococcus aureus Vaccine Candidates. Methods Mol. Biol. 2016, 1403, 409–418. [Google Scholar] [CrossRef] [PubMed]
  23. Schennings, T.; Heimdahl, A.; Coster, K.; Flock, J.I. Immunization with fibronectin binding protein from Staphylococcus aureus protects against experimental endocarditis in rats. Microb. Pathog. 1993, 15, 227–236. [Google Scholar] [CrossRef] [PubMed]
  24. Chambers, H.F.; Korzeniowski, O.M.; Sande, M.A. Staphylococcus aureus endocarditis: Clinical manifestations in addicts and nonaddicts. Medicine 1983, 62, 170–177. [Google Scholar] [CrossRef] [PubMed]
  25. Gribenko, A.V.; Mosyak, L.; Ghosh, S.; Parris, K.; Svenson, K.; Moran, J.; Chu, L.; Li, S.; Liu, T.; Woods, V.L.; et al. Three-dimensional structure and biophysical characterization of Staphylococcus aureus cell surface antigen-manganese transporter MntC. J. Mol. Biol. 2013, 425, 3429–3445. [Google Scholar] [CrossRef]
  26. Gribenko, A.V.; Parris, K.; Mosyak, L.; Lidia, M.; Handke, L.; Hawkins, J.C.; Severina, E.; Matsuka, Y.V.; Anderson, A.S. High Resolution Mapping of Bactericidal Monoclonal Antibody Binding Epitopes on Staphylococcus aureus Antigen MntC. PLoS Pathog. 2016, 12, e1005908. [Google Scholar] [CrossRef] [Green Version]
  27. Frenck, R.W., Jr.; Creech, C.B.; Sheldon, E.; Seiden, D.J.; Kankam, M.K.; Baber, J.; Zito, E.; Hubler, R.; Eiden, J.J.; Severs, J.M.; et al. Safety, tolerability, and immunogenicity of a 4-antigen Staphylococcus aureus vaccine (SA4Ag): Results from a first-in-human randomised, placebo-controlled phase 1/2 study. Vaccine 2017, 35, 375–384. [Google Scholar] [CrossRef]
  28. Li, X.; Wang, X.; Thompson, C.D.; Park, S.; Park, W.B.; Lee, J.C. Preclinical efficacy of clumping factor A in prevention of Staphylococcus aureus infection. mBio 2016, 7, e02232-15. [Google Scholar] [CrossRef] [Green Version]
  29. Zhu, D.; Tuo, W. QS-21: A Potent Vaccine Adjuvant. Nat. Prod. Chem. Res. 2016, 3, e113. [Google Scholar] [CrossRef] [Green Version]
  30. Bigaeva, E.; Doorn, E.; Liu, H.; Hak, E. Meta-Analysis on Randomized Controlled Trials of Vaccines with QS-21 or ISCOMATRIX Adjuvant: Safety and Tolerability. PLoS ONE 2016, 11, e0154757. [Google Scholar] [CrossRef] [Green Version]
  31. Nissen, M.D.; Marshall, H.S.; Richmond, P.; Shakib, S.; Jiang, Q.; Cooper, D.; Rill, D.; Baber, J.; Eiden, J.J.; Gruber, W.; et al. A randomized phase I study of the safety and immunogenicity of three ascending dose levels of a 3-antigen Staphylococcus aureus vaccine (SA3Ag) in healthy adults. Vaccine 2015, 33, 1846–1854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Fattom, A.; Matalon, A.; Buerkert, J.; Taylor, K.; Damaso, S.; Boutriau, D. Efficacy profile of a bivalent Staphylococcus aureus glycoconjugated vaccine in adults on hemodialysis: Phase III randomized study. Hum. Vaccines Immunother. 2015, 11, 632–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Fowler, V.G.; Allen, K.B.; Moreira, E.D.; Moustafa, M.; Isgro, F.; Boucher, H.W.; Corey, G.R.; Carmeli, Y.; Betts, R.; Hartzel, J.S.; et al. Effect of an investigational vaccine for preventing Staphylococcus aureus infections after cardiothoracic surgery: A randomized trial. JAMA 2013, 309, 1368–1378. [Google Scholar] [CrossRef] [PubMed]
  34. Haley, K.P.; Skaar, E.P. A battle for iron: Host sequestration and Staphylococcus aureus acquisition. Microbes Infect. 2012, 14, 217–227. [Google Scholar] [CrossRef] [Green Version]
  35. Stranger-Jones, Y.K.; Bae, T.; Schneewind, O. Vaccine assembly from surface proteins of Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2006, 103, 16942–16947. [Google Scholar] [CrossRef] [Green Version]
  36. Bagnoli, F.; Fontana, M.R.; Soldaini, E.; Mishra, R.P.N.; Fiaschi, L.; Cartocci, E.; Nardi-Dei, V.; Ruggiero, P.; Nosari, S.; De Falco, M.G.; et al. Vaccine composition formulated with a novel TLR7-dependent adjuvant induces high and broad protection against Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2015, 112, 3680–3685. [Google Scholar] [CrossRef] [Green Version]
  37. Torre, A.; Bacconi, M.; Sammicheli, C.; Galletti, B.; Laera, D.; Fontana, M.R.; Grandi, G.; De Gregorio, E.; Bagnoli, F.; Nuti, S.; et al. Four-Component Staphylococcus aureus Vaccine 4C-Staph Enhances Fcgamma Receptor Expression in Neutrophils and Monocytes and Mitigates S. aureus Infection in Neutropenic Mice. Infect. Immun. 2015, 83, 3157–3163. [Google Scholar] [CrossRef] [Green Version]
  38. Levy, J.; Licini, L.; Haelterman, E.; Moris, P.; Lestrate, P.; Damaso, S.; Van Belle, P.; Boutriau, D. Safety and immunogenicity of an investigational 4-component Staphylococcus aureus vaccine with or without AS03 adjuvant: Results of a randomized phase I trial. Hum. Vaccines Immunother. 2015, 11, 620–631. [Google Scholar] [CrossRef] [Green Version]
  39. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02388165?term=NCT02388165&draw=2&rank=1 (accessed on 25 April 2020).
  40. Pfizer Inc. Available online: https://investors.pfizer.com/investor-news/press-release-details/2018/Independent-Data-Monitoring-Committee-Recommends-Discontinuation-of-the-Phase-2b-STRIVE-Clinical-Trial-of-Staphylococcus-aureus-Vaccine-Following-Planned-Interim-Analysis/default.aspx (accessed on 25 April 2020).
Figure 1. SA4Ag antigens are immunogenic in preclinical species. Immune responses against SA4Ag antigens CP5, CP8, ClfA, and MntC were measured before (pre) and after (PD) immunization. Rodents were immunized three times subcutaneously with SA4Ag + AlPO4 prior to sample collection post-dose 3 (PD3). Non-human primates (NHP) were immunized a single time with SA4Ag without adjuvant. Anti-capsular immune responses were measured by the OPA assay for CP5 (A) and CP8 (B). Anti-protein immune responses were measured by cLIA for ClfA (C) and MntC (D). Human responses to a single unadjuvanted dose of SA4Ag are included as a comparator, adapted from [27].
Figure 1. SA4Ag antigens are immunogenic in preclinical species. Immune responses against SA4Ag antigens CP5, CP8, ClfA, and MntC were measured before (pre) and after (PD) immunization. Rodents were immunized three times subcutaneously with SA4Ag + AlPO4 prior to sample collection post-dose 3 (PD3). Non-human primates (NHP) were immunized a single time with SA4Ag without adjuvant. Anti-capsular immune responses were measured by the OPA assay for CP5 (A) and CP8 (B). Anti-protein immune responses were measured by cLIA for ClfA (C) and MntC (D). Human responses to a single unadjuvanted dose of SA4Ag are included as a comparator, adapted from [27].
Microorganisms 09 00177 g001aMicroorganisms 09 00177 g001b
Figure 2. Immunization with SA4Ag reduces bacterial burden in a S. aureus pyelonephritis model. Female CD-1 mice (n = 5) were vaccinated at weeks 0, 3, and 6, with SA4Ag or with vehicle alone. Two weeks after the final vaccination animals were challenged with ~2 × 108 S. aureus Reynolds. Two days post-challenge, the mice were euthanized and kidneys collected. Bacteria in kidneys were enumerated (colony-forming unit [CFU]/kidney). p value was calculated by Student’s t-test.
Figure 2. Immunization with SA4Ag reduces bacterial burden in a S. aureus pyelonephritis model. Female CD-1 mice (n = 5) were vaccinated at weeks 0, 3, and 6, with SA4Ag or with vehicle alone. Two weeks after the final vaccination animals were challenged with ~2 × 108 S. aureus Reynolds. Two days post-challenge, the mice were euthanized and kidneys collected. Bacteria in kidneys were enumerated (colony-forming unit [CFU]/kidney). p value was calculated by Student’s t-test.
Microorganisms 09 00177 g002
Table 1. S. aureus Strains Used in These Studies.
Table 1. S. aureus Strains Used in These Studies.
Strain NameCapsule TypeSourceMRSA/MSSAST/CCComments
CDC3 (USA300)5IDMRSAST8/CC8PVL+
PFESA0241 (USA300)5IDMRSAST8/CC8Mec IV, PVL+ TSST-neg
PFESA01585IDMSSAST28/CC25
PFESA01868CarriageMSSAST57/CC30
Reynolds5IDMSSAST25/CC25
ID, Invasive Disease; MRSA, Methicillin-resistant Staphylococcus aureus; MSSA, Methicillin-sensitive Staphylococcus aureus; PVL+, Panton-Valentine leukocidin-positive; ST/CC, sequence type/clonal complex; TSST-neg, Toxic shock syndrome toxin-negative.
Table 2. Immunization with SA4Ag Reduces S. aureus Burden in a Murine Surgical Site Infection Model.
Table 2. Immunization with SA4Ag Reduces S. aureus Burden in a Murine Surgical Site Infection Model.
TrialVaccineNumber of AnimalsMean log CFU/mL
(95% CI)
p Value vs. Control
1SA4Ag204.65 (3.90–5.40)0.0393
Vehicle205.72 (4.99–6.45)
2SA4Ag156.42 (5.07–7.77)0.0330
Vehicle137.89 (6.95–8.83)
3SA4Ag175.95 (4.60–7.31)0.2899
Vehicle166.45 (5.11–7.81)
Meta-analysisSA4Ag486.29 (5.57–7.01)0.0126
Vehicle437.36 (6.75–7.97)
Female CD1 mice (n = 10–20) were immunized on weeks 0, 3, and 6 with SA4Ag + QS-21 or QS-21 alone. On week 8, animals underwent surgery by placing an incision and stitch into the quadriceps muscle. Approximately 300 colony-forming units (CFU) of S. aureus PFESA0158 were instilled into the surgical site. Two days after surgery, tissue was harvested and bacterial burden was enumerated. p values were determined by Student’s t-test.
Table 3. Immunization with SA4Ag Reduces Bacterial Burden in a S. aureus Bacteremia Model.
Table 3. Immunization with SA4Ag Reduces Bacterial Burden in a S. aureus Bacteremia Model.
ChallengeTrialVaccineNumber of AnimalsMean log CFU/mL
(95% CI)
p Value vs. Control
S. aureus CDC3 (USA300)1SA4Ag104.65 (4.33–4.97)0.0056
Vehicle105.35 (4.96–5.73)
2SA4Ag104.49 (4.07–4.90)0.0147
Vehicle105.24 (4.76–5.72)
3SA4Ag103.63 (3.25–4.01)0.0056
Vehicle104.47 (4.00–4.94)
Meta-analysisSA4Ag304.25 (4.00–4.51)<0.0001
Vehicle305.02 (4.75–5.29)
S. aureus PFESA0241 (USA300)1SA4Ag104.17 (3.83–4.51)0.0491
Vehicle104.68 (4.25–5.12)
2SA4Ag104.26 (3.93–4.59)0.0157
Vehicle104.82 (4.48–5.15)
3SA4Ag104.38 (3.98–4.78)0.0455
Vehicle104.88 (4.54–5.22)
Meta-analysisSA4Ag304.27 (4.09–4.45)0.0002
vehicle304.79 (4.60–4.98)
Female CD-1 mice (n = 10) were vaccinated at weeks 0, 3, and 6, with SA4Ag or with vehicle alone. Two weeks after the final vaccination animals were challenged with ~2 × 108 S. aureus CDC3 or PFESA0241. Three hours post-challenge, the mice were euthanized and blood collected. Bacteria in blood were enumerated (colony-forming unit [CFU]/mL), and the log CFU reduction with 95% confidence interval (CI) was calculated compared to the vehicle-treated control. p values were determined by Student’s t-test.
Table 4. Immunization with SA4Ag Reduces Recovered CFU of a CP5-expressing S. aureus Clinical Isolate in a Rat Endocarditis Model.
Table 4. Immunization with SA4Ag Reduces Recovered CFU of a CP5-expressing S. aureus Clinical Isolate in a Rat Endocarditis Model.
TrialVaccineNumber of AnimalsMean Log CFU/mL
(95% CI)
p Value vs. Control
1SA4Ag166.52 (5.20–7.84)0.0319
Vehicle147.88 (7.19–8.58)
2SA4Ag156.42 (5.07–7.77)0.0330
Vehicle137.89 (6.95–8.83)
3SA4Ag175.95 (4.60–7.31)0.2899
Vehicle166.45 (5.11–7.81)
Meta-analysisSA4Ag486.29 (5.57–7.01)0.0126
Vehicle437.36 (6.75–7.97)
Groups of female Sprague Dawley rats (20/group) were vaccinated at weeks 0, 3, and 6, with SA4Ag in AlPO4 or with AlPO4 alone. Two weeks after the final vaccination, catheter placement surgery was performed to generate sterile valvular vegetations. A certain number of animals succumbed due to the surgery and were not included in the analysis. Two days after surgery, animals were challenged intravenously with S. aureus PFESA0158. Two days post-challenge, the rats were euthanized and hearts collected. Bacterial burden in heart tissue was enumerated (colony-forming unit [CFU]/mL) and the arithmetic mean and 95% confidence interval (CI) were calculated for each treatment group.
Table 5. Immunization with SA4Ag Reduces the Number of S. aureus Infections in a Rat Endocarditis Model of Infection.
Table 5. Immunization with SA4Ag Reduces the Number of S. aureus Infections in a Rat Endocarditis Model of Infection.
ExperimentVaccineNumber of Infected AnimalsNumber of Uninfected Animalsp Value vs. Control
1SA4Ag0110.0983
Vehicle49
2SA4Ag1110.0730
Vehicle67
Meta-analysisSA4Ag122<0.0001
Vehicle1016
Female Sprague Dawley rats (n = 11–14) were vaccinated at weeks 0, 3, and 6, with SA4Ag in AlPO4 or with AlPO4 alone. Two weeks after the final vaccination, catheter placement surgery was performed to generate sterile valvular vegetations. A certain number of animals succumb due to the surgery and were not included in the analysis. Two days after surgery, animals were challenged intravenously with ~4 × 103 colony-forming unit (CFU) S. aureus PFESA0186, a CP8-expressing clinical isolate. Two days post-challenge, the rats were euthanized, and tissue bacterial burden was quantitated in hearts and kidneys.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Scully, I.L.; Timofeyeva, Y.; Illenberger, A.; Lu, P.; Liberator, P.A.; Jansen, K.U.; Anderson, A.S. Performance of a Four-Antigen Staphylococcus aureus Vaccine in Preclinical Models of Invasive S. aureus Disease. Microorganisms 2021, 9, 177. https://doi.org/10.3390/microorganisms9010177

AMA Style

Scully IL, Timofeyeva Y, Illenberger A, Lu P, Liberator PA, Jansen KU, Anderson AS. Performance of a Four-Antigen Staphylococcus aureus Vaccine in Preclinical Models of Invasive S. aureus Disease. Microorganisms. 2021; 9(1):177. https://doi.org/10.3390/microorganisms9010177

Chicago/Turabian Style

Scully, Ingrid L., Yekaterina Timofeyeva, Arthur Illenberger, Peimin Lu, Paul A. Liberator, Kathrin U. Jansen, and Annaliesa S. Anderson. 2021. "Performance of a Four-Antigen Staphylococcus aureus Vaccine in Preclinical Models of Invasive S. aureus Disease" Microorganisms 9, no. 1: 177. https://doi.org/10.3390/microorganisms9010177

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop