Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Hum Vaccin Immunother ; 17(9): 2873-2893, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34033528

ABSTRACT

The emergence of the novel SARS-CoV-2 and COVID-19 has brought into sharp focus the need for a vaccine to prevent this disease. Vaccines have saved millions of lives since their introduction to the public over 200 years ago. The potential for vaccination reached new heights in the mid-20th century with the development of technologies that expanded the ability to create novel vaccines. Since then, there has been continued technological advancement in vaccine development. The resulting platforms provide the promise for solutions for many infectious diseases, including those that have been with us for decades as well as those just now emerging. Each vaccine platform represents a different technology with a unique set of advantages and challenges, especially when considering manufacturing. Therefore, it is essential to understand each platform as a separate product and process with its specific quality considerations. This review outlines the relevant platforms for developing a vaccine for SARS-CoV-2 and discusses the advantages and disadvantages of each.


Subject(s)
COVID-19 , Vaccines , COVID-19 Vaccines , Humans , SARS-CoV-2 , Vaccination
2.
Infect Drug Resist ; 13: 2521-2535, 2020.
Article in English | MEDLINE | ID: mdl-32801790

ABSTRACT

INTRODUCTION: Intravenous (IV) ß-lactam antibiotics, excreted through bile into the gastrointestinal (GI) tract, may disrupt the gut microbiome by eliminating the colonization resistance from beneficial bacteria. This increases the risk for Clostridium difficile infection (CDI) and can promote antimicrobial resistance by selecting resistant organisms and eliminating competition by non-resistant organisms. Ribaxamase is an orally administered ß-lactamase for use with IV ß-lactam antibiotics (penicillins and cephalosporins) and is intended to degrade excess antibiotics in the upper GI before they can disrupt the gut microbiome and alter the resistome. METHODS: Longitudinal fecal samples (349) were collected from patients who participated in a previous Phase 2b clinical study with ribaxamase for prevention of CDI. In that previous study, patients were treated with ceftriaxone for a lower respiratory tract infection and received concurrent ribaxamase or placebo. Extracted fecal DNA from the samples was subjected to whole-genome shotgun sequencing and analyzed for the presence of antimicrobial resistance (AMR) genes by alignment of sequences against the Comprehensive Antibiotic Resistance Database. A qPCR assay was also used to confirm some of the results. RESULTS: Database alignment identified ~1300 acquired AMR genes and gene variants, including those encoding ß-lactamases and vancomycin resistance which were significantly increased in placebo vs ribaxamase-treated patients following antibiotic exposure. qPCR corroborated the presence of these genes and supported both new acquisition and expansion of existing gene pools based on no detectable copy number or a low copy number in pre-antibiotic samples which increased post-antibiotics. Additional statistical analyses demonstrated significant correlations between changes in the gut resistome and clinical study parameters including study drug assignment and ß-lactamase and vancomycin resistance gene frequency. DISCUSSION: These findings demonstrated that ribaxamase reduced changes to the gut resistome subsequent to ceftriaxone administration and may help limit the emergence of AMR.

3.
Lancet Infect Dis ; 19(5): 487-496, 2019 05.
Article in English | MEDLINE | ID: mdl-30885591

ABSTRACT

BACKGROUND: Infections with Clostridium difficile are a health threat, yet no products are currently licensed for prevention of primary C difficile infections. Intravenous ß-lactam antibiotics are considered to confer a high risk of C difficile infection because of their biliary excretion into the gastrointestinal tract and disruption of the gut microbiome. ribaxamase (SYN-004) is an orally administered ß-lactamase that was designed to be given with intravenous ß-lactam antibiotics to degrade excess antibiotics in the upper gastrointestinal tract before they disrupt the gut microbiome and lead to C difficile infection. We therefore aimed to determine whether administration of ribaxamase could prevent C difficile infection in patients being treated with intravenous ceftriaxone for a lower respiratory tract infection, thereby supporting continued clinical development. METHODS: In this parallel-group, double-blind, multicentre, phase 2b, randomised placebo-controlled trial, we recruited patients who had been admitted to a hospital with a lower respiratory tract infection with a pneumonia index score of 90-130 and who were expected to be treated with ceftriaxone for at least 5 days. Patients were recruited from 54 clinical sites in the USA, Canada, Bulgaria, Hungary, Poland, Romania, and Serbia. We randomly assigned patients older than 50 years to groups (1:1) in blocks of four by use of an interactive web portal; these groups were assigned to receive either 150 mg ribaxamase or placebo four times per day during, and for 72 h after, treatment with ceftriaxone. All patients, clinical investigators, study staff, and sponsor personnel were masked to the study drug assignments. The primary endpoint was the incidence of C difficile infection, as diagnosed by the local laboratory, in patients who received at least one treatment dose, and this outcome was assessed during treatment and for 4 weeks after treatment. This study is registered with ClinicalTrials.gov, number NCT02563106. FINDINGS: Between Nov 16, 2015, and Nov 10, 2016, we screened 433 patients for inclusion in the study. Of these patients, 20 (5%) patients were excluded from the study (16 [4%] patients did not meet inclusion criteria; four [1%] patients because of dosing restrictions). We enrolled and randomly assigned 413 patients to groups, of whom 207 patients were assigned to receive ceftriaxone plus ribaxamase and 206 patients were assigned to receive ceftriaxone plus placebo. However, one (<1%) patient in the ribaxamase group withdrew consent and was not treated with ribaxamase. During the study and within the 4 weeks after antibiotic treatment, two (1·0%) patients in the ribaxamase group and seven (3·4%) patients in the placebo group were diagnosed with an infection with C difficile (risk reduction 2·4%, 95% CI -0·6 to 5·9; one-sided p=0·045). Adverse events were similar between groups but more deaths were reported in the ribaxamase group (11 deaths vs five deaths in the placebo group). This disparity was due to the higher incidence of deaths attributed to cardiac-associated causes in the ribaxamase group (six deaths vs one death in the placebo group). INTERPRETATION: In patients treated with intravenous ceftriaxone for lower respiratory tract infections, oral ribaxamase reduced the incidence of C difficile infections compared with placebo. The imbalance in deaths between the groups appeared to be related to the underlying health of the patients. Ribaxamase has the potential to prevent C difficile infection in patients treated with intravenous ß-lactam antibiotics, and our findings support continued clinical development of ribaxamase to prevent C difficile infection. FUNDING: Synthetic Biologics.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Clostridium Infections/prevention & control , Recombinant Proteins/administration & dosage , beta-Lactamases/administration & dosage , beta-Lactams/administration & dosage , Administration, Intravenous , Administration, Oral , Aged , Aged, 80 and over , Clostridioides difficile/drug effects , Clostridium Infections/epidemiology , Double-Blind Method , Female , Humans , Incidence , Male , Middle Aged , Respiratory Tract Infections/drug therapy , Treatment Outcome
4.
Anaerobe ; 53: 30-33, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29981838

ABSTRACT

During a Phase 2b study with SYN-004 (ribaxamase) for prevention of Clostridium difficile infection (CDI) conducted in North America and Eastern Europe, 45 C. difficile isolates from subjects with laboratory-confirmed CDI and or colonized with C. difficile were collected and characterized. Several C. difficile PCR ribotypes, including 027 and 198, were identified.


Subject(s)
Clostridioides difficile/classification , Clostridioides difficile/isolation & purification , Clostridium Infections/microbiology , Ribotyping , Clinical Trials, Phase II as Topic , Clostridioides difficile/genetics , Clostridium Infections/prevention & control , Europe, Eastern , Humans , North America , Recombinant Proteins/administration & dosage , beta-Lactamases/administration & dosage
5.
Article in English | MEDLINE | ID: mdl-28052855

ABSTRACT

SYN-004 (ribaxamase) is a ß-lactamase designed to be orally administered concurrently with intravenous ß-lactam antibiotics, including most penicillins and cephalosporins. Ribaxamase's anticipated mechanism of action is to degrade excess ß-lactam antibiotic that is excreted into the small intestine. This enzymatic inactivation of excreted antibiotic is expected to protect the gut microbiome from disruption and thus prevent undesirable side effects, including secondary infections such as Clostridium difficile infections, as well as other antibiotic-associated diarrheas. In phase 1 clinical studies, ribaxamase was well tolerated compared to a placebo group and displayed negligible systemic absorption. The two phase 2a clinical studies described here were performed to confirm the mechanism of action of ribaxamase, degradation of ß-lactam antibiotics in the human intestine, and were therefore conducted in subjects with functioning ileostomies to allow serial sampling of their intestinal chyme. Ribaxamase fully degraded ceftriaxone to below the level of quantitation in the intestines of all subjects in both studies. Coadministration of oral ribaxamase with intravenous ceftriaxone was also well tolerated, and the plasma pharmacokinetics of ceftriaxone were unchanged by ribaxamase administration. Since ribaxamase is formulated as a pH-dependent, delayed-release formulation, the activity of ribaxamase in the presence of the proton pump inhibitor esomeprazole was examined in the second study; coadministration of these drugs did not adversely affect ribaxamase's ability to degrade ceftriaxone excreted into the intestine. These studies have confirmed the in vivo mechanism of action of ribaxamase, degradation of ß-lactam antibiotics in the human intestine (registered at ClinicalTrials.gov under NCT02419001 and NCT02473640).


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Ceftriaxone/pharmacokinetics , Dysbiosis/prevention & control , Inactivation, Metabolic , Protective Agents/pharmacokinetics , Recombinant Proteins/pharmacokinetics , beta-Lactamases/pharmacokinetics , Administration, Oral , Drug Administration Schedule , Humans , Ileostomy , Infusions, Intravenous , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestines/drug effects
6.
Clin Drug Investig ; 36(9): 725-734, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27283946

ABSTRACT

BACKGROUND: SYN-004 is an orally administered ß-lactamase enzyme, designed to be given concurrently with certain intravenous ß-lactam antibiotics like cephalosporins. SYN-004 is intended to degrade residual antibiotics excreted into the intestine as a result of hepatobiliary excretion and to prevent the disruption of the gut microbiome by these excess antibiotics. Preserving the gut microbiome is expected to prevent secondary infections by pathogens like Clostridium difficile and protect against other antibiotic-associated diarrheas. METHODS: Two, randomized, double blind, placebo-controlled Phase 1 clinical studies were conducted in normal healthy adult volunteers to assess the tolerability and systemic absorption of single and multiple doses of SYN-004. A single-ascending dose study investigated single oral doses of 75-750 mg SYN-004 and was conducted in 40 subjects (five cohorts of six active and two placebo subjects). A multiple-ascending dose study investigated doses of 75-300 mg SYN-004, administered every 6 h for 7 days and was conducted in 24 subjects (three cohorts of six active and two placebo subjects). The safety and tolerability of SYN-004 was assessed and serial plasma and serum samples were collected to assess the pharmacokinetics and potential immunogenicity of SYN-004. RESULTS: Minimal and mild adverse events were reported in ~30 % of the subjects who received active drug and placebo and no antidrug antibodies were detected in any subject. Analysis of serial plasma samples demonstrated negligible systemic bioavailability of SYN-004 with most plasma concentrations being below the lower limit of quantitation (0.8 ng/mL) for the assay. SYN-004 was well tolerated in the 48 subjects who received active drug, and adverse events in those subjects were comparable to the 16 subjects who received placebo, up to the maximum doses administered in each study. CONCLUSION: SYN-004 was well tolerated up to a single oral dose of 750 mg and multiple doses of 300 mg every 6 h for 7 days. The pharmacokinetic results support that SYN-004 remained localized in the intestine.


Subject(s)
Clostridioides difficile , Clostridium Infections/drug therapy , Diarrhea/prevention & control , Recombinant Proteins/therapeutic use , beta-Lactamases/therapeutic use , Adolescent , Adult , Aged , Biological Availability , Clostridium Infections/complications , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Young Adult , beta-Lactamases/adverse effects , beta-Lactamases/pharmacokinetics
7.
Int J Toxicol ; 35(3): 309-16, 2016 05.
Article in English | MEDLINE | ID: mdl-26700136

ABSTRACT

SYN-004 is a first in class, recombinant ß-lactamase that degrades ß-lactam antibiotics and has been formulated to be administered orally to patients receiving intravenous ß-lactam antibiotics including cephalosporins. SYN-004 is intended to degrade unmetabolized antibiotics excreted into the intestines and thus has the potential to protect the gut microbiome from disruption by these antibiotics. Protection of the gut microbiome is expected to protect against opportunistic enteric infections such as Clostridium difficile infection as well as antibiotic-associated diarrhea. In order to demonstrate that oral SYN-004 is safe for human clinical trials, 2 Good Laboratory Practice-compliant toxicity studies were conducted in Beagle dogs. In both studies, SYN-004 was administered orally 3 times per day up to the maximum tolerated dose of the formulation. In the first study, doses of SYN-004 administered over 28 days were safe and well tolerated in dogs with the no-observed-adverse-effect level at the high dose of 57 mg/kg/day. Systemic absorption of SYN-004 was minimal and sporadic and showed no accumulation during the study. In the second study, doses up to 57 mg/kg/day were administered to dogs in combination with an intravenous dose of ceftriaxone (300 mg/kg) given once per day for 14 days. Coadministration of oral SYN-004 with intravenous ceftriaxone was safe and well tolerated, with SYN-004 having no noticeable effect on the plasma pharmacokinetics of ceftriaxone. These preclinical studies demonstrate that SYN-004 is well tolerated and, when coadministered with ceftriaxone, does not interfere with its systemic pharmacokinetics. These data supported advancing SYN-004 into human clinical trials.


Subject(s)
Anti-Bacterial Agents , Ceftriaxone , Protective Agents , Recombinant Proteins , beta-Lactamases , Administration, Intravenous , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacokinetics , Bile Ducts/metabolism , Ceftriaxone/administration & dosage , Ceftriaxone/blood , Ceftriaxone/pharmacokinetics , Dogs , Drug Interactions , Female , Gastrointestinal Microbiome , Male , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , Protective Agents/toxicity , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , Tablets, Enteric-Coated , Toxicity Tests, Subacute , beta-Lactamases/administration & dosage , beta-Lactamases/pharmacokinetics , beta-Lactamases/pharmacology , beta-Lactamases/toxicity
8.
Pediatrics ; 128(2): 271-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21788224

ABSTRACT

BACKGROUND: Pagibaximab, a human chimeric monoclonal antibody developed against lipoteichoic acid, was effective against staphylococci preclinically and seemed safe and well tolerated in phase 1 studies. OBJECTIVE: To evaluate the clinical activity, pharmacokinetics, safety, and tolerability of weekly pagibaximab versus placebo infusions in very low birth weight neonates. PATIENTS AND METHODS: A phase 2, randomized, double-blind, placebo-controlled study was conducted at 10 NICUs. Patients with a birth weight of 700 to 1300 g and 2 to 5 days old were randomly assigned to receive 3 once-a-week pagibaximab (90 or 60 mg/kg) or placebo infusions. Blood was collected for pharmacokinetics, bacterial killing, and safety analyses. Adverse event and clinical outcome data were collected. RESULTS: Eighty-eight patients received pagibaximab at 90 (n = 22) or 60 (n = 20) mg/kg or placebo (n = 46). Groups were not different in demography, mortality, or morbidity. Pagibaximab demonstrated linear pharmacokinetics, a 14.5-day half-life, and nonimmunogenicity. Definite staphylococcal sepsis occurred in 0%, 20%, and 13% (P < .11) and nonstaphylococcal sepsis occurred in 0%, 10%, and 15% (P < .15) of patients in the 90 mg/kg, 60 mg/kg, and placebo groups, respectively. In all patients with staphylococcal sepsis, estimated or observed pagibaximab levels were <500 µg/mL (target level) at infection. CONCLUSIONS: Three once-a-week 90 or 60 mg/kg pagibaximab infusions, in high-risk neonates, seemed safe and well tolerated. No staphylococcal sepsis occurred in infants who received 90 mg/kg. Target levels were only consistently achieved after 2 to 3 doses. Dose optimization should enhance protection.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Infant, Very Low Birth Weight , Sepsis/prevention & control , Staphylococcal Infections/prevention & control , Antibodies, Monoclonal/blood , Double-Blind Method , Drug Administration Schedule , Female , Humans , Infant, Newborn , Infant, Very Low Birth Weight/blood , Infusions, Intravenous , Male , Risk Factors , Sepsis/blood , Sepsis/etiology , Staphylococcal Infections/blood , Staphylococcal Infections/etiology
9.
Antimicrob Agents Chemother ; 53(7): 2879-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19380597

ABSTRACT

Staphylococcal sepsis is a major cause of morbidity and mortality in very-low-birth-weight (VLBW) infants. A human chimeric monoclonal antibody, pagibaximab, was developed against staphylococcal lipoteichoic acid. We evaluated the safety, tolerability, and pharmacokinetics of pagibaximab in VLBW neonates. A phase 1/2, randomized, double-blind, placebo-controlled, dose escalation study was conducted in VLBW infants (700 to 1,300 g) 3 to 7 days old. Patients received two doses 14 days apart of intravenous pagibaximab (10, 30, 60, or 90 mg/kg of body weight) or placebo in a 2:1 ratio. Blood and urine samples were obtained pre- and postinfusion for analysis of safety and pharmacokinetics, and data on adverse events were gathered. Staphylococcal organisms causing sepsis were collected and evaluated. Fifty-three patients received at least one dose of pagibaximab or placebo. The average gestational age was 27.6 weeks; the average birth weight was 1,003 g. All serious adverse events were deemed unrelated or probably not drug related. Morbidity and mortality were similar across treatment groups. No evidence of immunogenicity of pagibaximab was detected. Pagibaximab pharmacokinetics was linear. The mean clearance (CL), volume of distribution, and elimination half-life of pagibaximab were independent of dose. The serum half-life was 20.5 +/- 6.8 days. Pagibaximab enhanced serum opsonophagocytic activity. All staphylococci causing sepsis were opsonizable by pagibaximab. Two infusions of pagibaximab, administered 2 weeks apart to high-risk neonates appeared safe and tolerable, and pharmacokinetics were linear. Evaluation of more frequent doses, at the highest doses tested, in neonates at high-risk of staphylococcal sepsis, is warranted.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Infant, Very Low Birth Weight , Staphylococcal Infections/prevention & control , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Double-Blind Method , Humans , Infant, Newborn
10.
J Antimicrob Chemother ; 64(1): 94-100, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398455

ABSTRACT

OBJECTIVES: Staphylococcus aureus infections associated with indwelling devices can be very difficult to treat due to the recalcitrant nature of bacterial biofilms to conventional antibiotics. Lysostaphin has been shown to clear S. aureus biofilms in vitro, and in this study we determined whether lysostaphin could also eradicate established S. aureus biofilms on implanted jugular vein catheters in mice. METHODS: Jugular vein catheterized mice (four to six per group) challenged with S. aureus developed multiorgan infection and biofilm infections on the catheters. The infected mice with established biofilms received various doses of recombinant lysostaphin through the catheters, administered up to three times daily for up to 4 days. Some mice also received lysostaphin combined with nafcillin. Following treatment, mice were sacrificed and cfu on the catheter and in the liver and heart were determined. In another set of experiments, implanted jugular vein catheters in mice were pre-instilled with lysostaphin to determine whether this pre-treatment would protect the mice from biofilm infection. RESULTS AND CONCLUSIONS: Lysostaphin administered at 15 mg/kg in combination with 50 mg/kg nafcillin three times per day for 4 days eradicated established S. aureus, including methicillin-resistant S. aureus, biofilms from implanted catheters and sterilized heart and liver infections of S. aureus-infected mice. Furthermore, a single pre-instillation of 10 mg/kg lysostaphin in catheters completely protected catheterized mice from a subsequent biofilm infection. These results demonstrate that lysostaphin is an effective treatment as well as prophylaxis for S. aureus biofilms on indwelling catheters.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Lysostaphin/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Catheterization, Central Venous , Colony Count, Microbial , Equipment and Supplies/microbiology , Heart/microbiology , Humans , Jugular Veins , Liver/microbiology , Lysostaphin/administration & dosage , Mice , Nafcillin/therapeutic use
11.
Methods Mol Biol ; 431: 241-54, 2008.
Article in English | MEDLINE | ID: mdl-18287761

ABSTRACT

Staphylococcus aureus nasal colonization is a well-known risk factor for development of S.aureus infections in humans, but despite this established association, we are only beginning to understand the factors, both host and pathogen, that play a role in the colonization of the nares by S. aureus. The cotton rat is a model for many human respiratory pathogens and has proved its utility as a robust model for S. aureus nasal colonization. In this animal model, S. aureus is instilled in the nostrils of adult cotton rats, the bacteria rapidly colonize, and 7 days later S. aureus nasal colonization is enumerated by surgical removal of the nose and recovery of the colonizing S. aureus. This model is an excellent animal model to allow for the evaluation of the efficacy of various therapies, including semi-solid formulations, for determination of their ability to eradicate S. aureus nasal colonization. Further, the cotton rat model allows for assessment of the ability of defined genetic mutants of S. aureus to colonize mucosal surfaces. Finally, this model has demonstrated its utility for the assessment of various antigens as vaccine candidates to protect against S. aureus nasal colonization. This chapter will discuss in detail the method to establish nasal colonization, treatment and eradication of colonization, and recovery of the colonizing bacteria from the nose.


Subject(s)
Nose/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Animals , Disease Models, Animal , Humans , Rats , Time Factors
12.
Int J Med Microbiol ; 298(5-6): 505-13, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18221914

ABSTRACT

Most of the severe bacterial infections originate from the endogenous microflora of human body surfaces. However, the molecular basis of colonization, e.g. of the human nose by Staphylococcus aureus, has remained incompletely understood. Several surface-exposed proteins and wall teichoic acid (WTA) polymers have previously been implicated in S. aureus attachment to nasal epithelial cells. Here we dissect the role of these molecules in colonization using S. aureus sortase A (srtA) and tagO mutants deficient in surface protein and WTA display, respectively. Although the two mutants were similarly affected in attachment to nasal cells they were abrogated in binding to different types of epithelial ligands. Surface protein sorting, but not WTA, were required for keratin- or fibronectin-mediated interactions while only WTA-mediated binding to nasal cells was effectively inhibited by polyinosinic acid, indicating a possible role of scavenger receptor-like molecules in WTA-dependent epithelial interactions. Both mutants exhibited profound colonization defects in a cotton rat nasal colonization model, albeit at different stages of colonization (>90% reduced bacterial counts at 24h or several days after inoculation with the tagO or srtA mutant, respectively). These data indicate that S. aureus nasal colonization is a multifactorial process with various ligands affecting initial colonization and prolonged persistence in different ways. Our studies should be useful in the development of new preventive and therapeutic strategies.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Adhesion , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Membrane Proteins/metabolism , Staphylococcus aureus/physiology , Teichoic Acids/metabolism , Aminoacyltransferases/genetics , Animals , Bacterial Proteins/genetics , Cells, Cultured , Cysteine Endopeptidases/genetics , Epithelial Cells/microbiology , Fibronectins/metabolism , Gene Deletion , Humans , Keratins/metabolism , Nasal Cavity/microbiology , Rats , Receptors, Scavenger/metabolism , Sigmodontinae , Teichoic Acids/genetics
13.
Cell Host Microbe ; 1(3): 199-212, 2007 May 17.
Article in English | MEDLINE | ID: mdl-18005699

ABSTRACT

Resistance to human skin innate defenses is crucial for survival and carriage of Staphylococcus aureus, a common cutaneous pathogen and nasal colonizer. Free fatty acids extracted from human skin sebum possess potent antimicrobial activity against S. aureus. The mechanisms by which S. aureus overcomes this host defense during colonization remain unknown. Here, we show that S. aureus IsdA, a surface protein produced in response to the host, decreases bacterial cellular hydrophobicity rendering them resistant to bactericidal human skin fatty acids and peptides. IsdA is required for survival of S. aureus on live human skin. Reciprocally, skin fatty acids prevent the production of virulence determinants and the induction of antibiotic resistance in S. aureus and other Gram-positive pathogens. A purified human skin fatty acid was effective in treating systemic and topical infections of S. aureus suggesting that our natural defense mechanisms can be exploited to combat drug-resistant pathogens.


Subject(s)
Antigens, Bacterial/physiology , Skin/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , Adult , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/chemistry , Antimicrobial Cationic Peptides/pharmacology , Colony Count, Microbial , Fatty Acids/isolation & purification , Fatty Acids/pharmacology , Hemolysin Proteins/biosynthesis , Humans , Hydrophobic and Hydrophilic Interactions , Microbial Viability , Sebum/immunology , Sebum/microbiology , Skin/chemistry , Skin/microbiology , Staphylococcal Skin Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Virulence Factors/biosynthesis
14.
J Antimicrob Chemother ; 60(5): 1051-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17848374

ABSTRACT

OBJECTIVES: With the isolation of clinical strains of Staphylococcus aureus carrying the gene that confers vancomycin resistance, the need for novel antistaphylococcals has become more urgent. Lysostaphin, an example of such a novel therapeutic, is an endopeptidase that rapidly lyses S. aureus through proteolysis of the staphylococcal cell wall. We evaluated its efficacy as a therapeutic agent for treatment of systemic S. aureus infection in a mouse model. METHODS: Mice (5-10 per group) challenged with methicillin-susceptible S. aureus developed bacteraemia and organ infections while mice challenged with methicillin-resistant S. aureus (MRSA) developed organ infections. The challenged mice received various intravenous doses of recombinant lysostaphin, administered once a day for 1-3 days when compared with treatment with oxacillin or vancomycin. Some mice also received treatment of lysostaphin combined with oxacillin or vancomycin. Following treatment, bacteraemia was determined, and mice were sacrificed and organ infection was determined. RESULTS AND CONCLUSIONS: Lysostaphin administered at 5 mg/kg once a day for 3 days consistently cleared S. aureus from the blood and the organs of infected mice. Furthermore, the combination of lysostaphin and oxacillin or vancomycin demonstrated increased efficacy against MRSA over lysostaphin alone allowing the therapeutic dose of lysostaphin to be reduced to 1 mg/kg. These results demonstrate that lysostaphin is an effective treatment for eradicating S. aureus from the blood and from the organs of infected mice.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Lysostaphin/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus , Animals , Bacteremia/drug therapy , Bacteremia/microbiology , Dose-Response Relationship, Drug , Female , Kidney/microbiology , Liver/microbiology , Methicillin Resistance , Mice , Neutropenia , Neutrophils/microbiology , Spleen/microbiology , Staphylococcus aureus/drug effects
15.
Microb Pathog ; 43(5-6): 208-16, 2007.
Article in English | MEDLINE | ID: mdl-17689046

ABSTRACT

Bacterial super-infection of influenza patients is the primary cause of excess mortality during influenza pandemics, with Staphylococcus aureus (S. aureus) having the highest fatality rate. The cotton rat (Sigmodon hispidus) is an excellent model for both influenza and S. aureus pathogenesis, and therefore a potential tool to model co-infection. We compared physiologic and pathologic changes in cotton rats infected with both S. aureus and influenza A/Wuhan/359/95 (H3N2), with animals infected with each pathogen alone. Co-infected cotton rats demonstrated significantly higher mortality, lower temperatures on 2 and 3 days post-inoculation (p.i.), higher levels of bacteremia and pulmonary bacterial load 4 days p.i., and worse pathology 7 days p.i. Early indicators of exacerbated disease coincided with higher pulmonary mRNA levels for IL-1beta, IL-6, IL-10 and IFNy, supporting the idea that these may contribute to disease severity. Our results demonstrate that the cotton rat is a good model of influenza and S. aureus co-infection, with increased mortality and hypothermia as well as prolonged bacterial duration indicative of synergistic disease that may be the result of increased induction of both pro- and anti-inflammatory cytokines.


Subject(s)
Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections/complications , Staphylococcal Infections/complications , Staphylococcus aureus/isolation & purification , Animals , Antibodies, Viral/blood , Disease Models, Animal , Influenza A Virus, H3N2 Subtype/isolation & purification , Lung/pathology , Lung/virology , Sigmodontinae
16.
J Bacteriol ; 189(20): 7316-25, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17675373

ABSTRACT

Bacterial cell wall peptidoglycan is a dynamic structure requiring hydrolysis to allow cell wall growth and division. Staphylococcus aureus has many known and putative peptidoglycan hydrolases, including two likely lytic transglycosylases. These two proteins, IsaA and SceD, were both found to have autolytic activity. Regulatory studies showed that the isaA and sceD genes are partially mutually compensatory and that the production of SceD is upregulated in an isaA mutant. The expression of sceD is also greatly upregulated by the presence of NaCl. Several regulators of isaA and sceD expression were identified. Inactivation of sceD resulted in impaired cell separation, as shown by light microscopy, and "clumping" of bacterial cultures. An isaA sceD mutant is attenuated for virulence, while SceD is essential for nasal colonization in cotton rats, thus demonstrating the importance of cell wall dynamics in host-pathogen interactions.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Glycosyltransferases/physiology , Peptidoglycan Glycosyltransferase/physiology , Staphylococcus aureus/enzymology , Animals , Antigens, Bacterial/genetics , Arthritis, Infectious/microbiology , Bacterial Proteins/genetics , Bacteriolysis , Carrier State/microbiology , Gene Deletion , Gene Expression Regulation, Bacterial , Glycosyltransferases/genetics , Mice , Microbial Viability , Mutagenesis, Insertional , Peptidoglycan Glycosyltransferase/genetics , Sigmodontinae , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Virulence Factors/genetics , Virulence Factors/physiology
17.
Antimicrob Agents Chemother ; 51(2): 475-82, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17101683

ABSTRACT

Lysostaphin is under development as a therapy for serious staphylococcal infections. During preclinical development, lysostaphin-resistant Staphylococcus aureus variants have occasionally been reported in vitro and in vivo. The acquisition of resistance to this drug, however, leads to a significant increase in beta-lactam antibiotic susceptibility, rendering methicillin-resistant S. aureus (MRSA) strains functionally methicillin susceptible. In this study, we have demonstrated that the development of lysostaphin resistance by two strains of MRSA also led to a loss of fitness in the variants. Consistent with the mutations found in previously reported lysostaphin-resistant S. aureus variants, these two variants had mutations in their femA genes, resulting in nonfunctional FemA proteins and, thus, monoglycine cross bridges in the peptidoglycan. The diminished fitness of the lysostaphin-resistant variants was reflected by (i) a reduced logarithmic growth rate, with the variants being outcompeted in cocultures by their wild-type parental strains; (ii) increased susceptibility to elevated temperatures; and (iii) at least fivefold less virulence of the lysostaphin-resistant variants than their wild-type strains in a mouse kidney infection model, with the lysostaphin-resistant variants being outcompeted in coinfections with their wild-type parental strains. During a 14-day serial passage without selective pressure, the lysostaphin-resistant variants failed to develop compensatory mutations which restored their fitness. These results suggest that should lysostaphin resistance due to an alteration in the FemA function emerge in S. aureus during therapy with lysostaphin, the resistant variants would be less fit and less virulent, and, in addition, infections with these strains would be easily treatable with beta-lactam antibiotics.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Microbial , Lysostaphin , Staphylococcus aureus , Animals , Bacterial Proteins/metabolism , Drug Resistance, Microbial/genetics , Mice , Mutation , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development
18.
J Bacteriol ; 189(3): 1025-35, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17114262

ABSTRACT

Oxidative-stress resistance in Staphylococcus aureus is linked to metal ion homeostasis via several interacting regulators. In particular, PerR controls the expression of a regulon of genes, many of which encode antioxidants. Two PerR regulon members, ahpC (alkylhydroperoxide reductase) and katA (catalase), show compensatory regulation, with independent and linked functions. An ahpC mutation leads to increased H2O2 resistance due to greater katA expression via relief of PerR repression. Moreover, AhpC provides residual catalase activity present in a katA mutant. Mutation of both katA and ahpC leads to a severe growth defect under aerobic conditions in defined media (attributable to lack of catalase activity). This results in the inability to scavenge exogenous or endogenously produced H2O2, resulting in accumulation of H2O2 in the medium. This leads to DNA damage, the likely cause of the growth defect. Surprisingly, the katA ahpC mutant is not attenuated in two independent models of infection, which implies reduced oxygen availability during infection. In contrast, both AhpC and KatA are required for environmental persistence (desiccation) and nasal colonization. Thus, oxidative-stress resistance is an important factor in the ability of S. aureus to persist in the hospital environment and so contribute to the spread of human disease.


Subject(s)
Bacterial Proteins/metabolism , Catalase/metabolism , Nasal Cavity/microbiology , Peroxidases/metabolism , Staphylococcus aureus/metabolism , Animals , Antioxidants/metabolism , Antioxidants/physiology , Bacterial Proteins/genetics , Catalase/genetics , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial , Genetic Complementation Test , Glucose/deficiency , Glucose/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Male , Microbial Viability/genetics , Mutation , Oxidative Stress , Peroxidases/genetics , Rats , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence/genetics
19.
J Infect Dis ; 193(8): 1098-108, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16544250

ABSTRACT

A spectrum of in vivo-expressed Staphylococcus aureus antigens was identified by probing bacteriophage expression libraries of S. aureus with serum samples from infected and uninfected individuals. Eleven recombinant antigenic proteins were produced, and specific antibody titers in a large collection of human serum samples were determined. Significantly increased concentrations of reactive immunoglobulin G (IgG) to 7 antigens were found in serum samples from ill individuals, compared with those in healthy individuals. Significantly higher concentrations of reactive IgG to 4 antigens, including iron-responsive surface determinant (Isd) A and IsdH, were found in serum samples from healthy individuals who were not nasal carriers of S. aureus, compared with those in healthy carriers. Vaccination of cotton rats with IsdA or IsdH protected against nasal carriage. Also, IsdA is involved in adherence of S. aureus to human desquamated nasal epithelial cells and is required for nasal colonization in the cotton rat model. Thus, vaccination with these antigens may prevent S. aureus carriage and reduce the prevalence of human disease.


Subject(s)
Antigens, Bacterial/isolation & purification , Bacterial Vaccines , Carrier State/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/blood , Antigens, Bacterial/classification , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Carrier State/immunology , Cation Transport Proteins/blood , Cation Transport Proteins/immunology , Cells, Cultured , DNA Primers/chemistry , Epithelial Cells/microbiology , Female , Humans , Immunoglobulin G/blood , Nasal Cavity/microbiology , Receptors, Cell Surface , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Sigmodontinae , Staphylococcal Infections/immunology
20.
Antimicrob Agents Chemother ; 49(8): 3256-63, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16048934

ABSTRACT

Lysostaphin is an endopeptidase that cleaves the pentaglycine cross-bridges of the staphylococcal cell wall rapidly lysing the bacteria. Recently, lysostaphin has been examined for its potential to treat infections and to clear Staphylococcus aureus nasal colonization, requiring a reliable method for determining the lysostaphin susceptibility of strains of S. aureus. We compared four methods for determining the lysostaphin susceptibility of 57 strains of methicillin-sensitive S. aureus, methicillin-resistant S. aureus, vancomycin intermediately susceptible S. aureus (VISA), mupirocin-resistant S. aureus, and various defined genetic mutants of S. aureus. Three reference lysostaphin-resistant S. aureus variants were also included in the assays as negative controls. The assays examined included turbidity, MIC, minimum bactericidal concentration (MBC), and disk diffusion assays. All of the strains of S. aureus tested, including a VISA strain which had previously been reported to be lysostaphin resistant, were susceptible to lysostaphin by all four methods. The three reference lysostaphin-resistant variants were resistant by all four methods. The disk diffusion assay was the simplest method to differentiate lysostaphin-susceptible S. aureus strains from lysostaphin-resistant variants, while the MBC assay could be used as a follow-up assay if required. In the disk diffusion assay, all strains of S. aureus tested revealed zones of inhibition of >/=11 mm using a 50-microg lysostaphin disk, while the three reference lysostaphin-resistant S. aureus variants had no zones of inhibition. In MBC assays, concentrations of lysostaphin ranging from 0.16 microg/ml to 2.5 microg/ml were found to cause a 3 log or greater drop from the initial CFU of S. aureus within 30 min for all strains tested.


Subject(s)
Anti-Bacterial Agents/pharmacology , Lysostaphin/pharmacology , Microbial Sensitivity Tests/methods , Staphylococcus aureus/drug effects , Culture Media , Humans , Methicillin/pharmacology , Methicillin Resistance , Staphylococcus aureus/classification , Staphylococcus aureus/growth & development , Vancomycin/pharmacology , Vancomycin Resistance
SELECTION OF CITATIONS
SEARCH DETAIL
...