Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Infect Immun ; 92(4): e0001824, 2024 Apr 09.
Article En | MEDLINE | ID: mdl-38514468

Borrelia burgdorferi, the spirochetal agent of Lyme disease, utilizes a variety of strategies to evade and suppress the host immune response, which enables it to chronically persist in the host. The resulting immune response is characterized by unusually strong IgM production and a lack of long-term protective immunity. Previous studies in mice have shown that infection with B. burgdorferi also broadly suppresses host antibody responses against unrelated antigens. Here, we show that mice infected with B. burgdorferi and concomitantly immunized with recombinant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein had an abrogated antibody response to the immunization. To further define how long this humoral immune suppression lasts, mice were immunized at 2, 4, and 6 weeks post-infection. Suppression of host antibody production against the SARS-CoV-2 spike protein peaked at 2 weeks post-infection but continued for all timepoints measured. Antibody responses against the SARS-CoV-2 spike protein were also assessed following antibiotic treatment to determine whether this immune suppression persists or resolves following clearance of B. burgdorferi. Host antibody production against the SARS-CoV-2 spike protein returned to baseline following antibiotic treatment; however, anti-SARS-CoV-2 IgM remained high, comparable to levels found in B. burgdorferi-infected but untreated mice. Thus, our data demonstrate restored IgG responses following antibiotic treatment but persistently elevated IgM levels, indicating lingering effects of B. burgdorferi infection on the immune system following treatment.


Borrelia burgdorferi , Lyme Disease , Spike Glycoprotein, Coronavirus , Mice , Humans , Animals , Immunity, Humoral , Immunoglobulin M , Anti-Bacterial Agents , Antibodies, Bacterial
2.
Cell Rep Med ; 4(11): 101253, 2023 11 21.
Article En | MEDLINE | ID: mdl-37918405

Colonization of the gut and airways by pathogenic bacteria can lead to local tissue destruction and life-threatening systemic infections, especially in immunologically compromised individuals. Here, we describe an mRNA-based platform enabling delivery of pathogen-specific immunoglobulin A (IgA) monoclonal antibodies into mucosal secretions. The platform consists of synthetic mRNA encoding IgA heavy, light, and joining (J) chains, packaged in lipid nanoparticles (LNPs) that express glycosylated, dimeric IgA with functional activity in vitro and in vivo. Importantly, mRNA-derived IgA had a significantly greater serum half-life and a more native glycosylation profile in mice than did a recombinantly produced IgA. Expression of an mRNA encoded Salmonella-specific IgA in mice resulted in intestinal localization and limited Peyer's patch invasion. The same mRNA-LNP technology was used to express a Pseudomonas-specific IgA that protected from a lung challenge. Leveraging the mRNA antibody technology as a means to intercept bacterial pathogens at mucosal surfaces opens up avenues for prophylactic and therapeutic interventions.


Mucous Membrane , Peyer's Patches , Mice , Animals , Immunoglobulin A , Antibodies, Monoclonal
3.
J Med Chem ; 64(14): 10155-10166, 2021 07 22.
Article En | MEDLINE | ID: mdl-34191513

A major antimicrobial resistance mechanism in Gram-negative bacteria is the production of ß-lactamase enzymes. The increasing emergence of ß-lactamase-producing multi-drug-resistant "superbugs" has resulted in increases in costly hospital Emergency Department (ED) visits and hospitalizations due to the requirement for parenteral antibiotic therapy for infections caused by these difficult-to-treat bacteria. To address the lack of outpatient treatment, we initiated an iterative program combining medicinal chemistry, biochemical testing, microbiological profiling, and evaluation of oral pharmacokinetics. Lead optimization focusing on multiple smaller, more lipophilic active compounds, followed by an exploration of oral bioavailability of a variety of their respective prodrugs, provided 36 (VNRX-7145/VNRX-5236 etzadroxil), the prodrug of the boronic acid-containing ß-lactamase inhibitor 5 (VNRX-5236). In vitro and in vivo studies demonstrated that 5 restored the activity of the oral cephalosporin antibiotic ceftibuten against Enterobacterales expressing Ambler class A extended-spectrum ß-lactamases, class A carbapenemases, class C cephalosporinases, and class D oxacillinases.


Anti-Bacterial Agents/pharmacology , Drug Discovery , Enterobacteriaceae/drug effects , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enterobacteriaceae/enzymology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamase Inhibitors/chemistry
4.
Antimicrob Agents Chemother ; 65(8): e0055221, 2021 07 16.
Article En | MEDLINE | ID: mdl-34001510

There is an urgent need for oral agents to combat resistant Gram-negative pathogens. Here, we describe the characterization of VNRX-5236, a broad-spectrum boronic acid ß-lactamase inhibitor (BLI), and its orally bioavailable etzadroxil prodrug, VNRX-7145. VNRX-7145 is being developed in combination with ceftibuten, an oral cephalosporin, to combat strains of Enterobacterales expressing extended-spectrum ß-lactamases (ESBLs) and serine carbapenemases. VNRX-5236 is a reversible covalent inhibitor of serine ß-lactamases, with inactivation efficiencies on the order of 104 M-1 · sec-1, and prolonged active site residence times (t1/2, 5 to 46 min). The spectrum of inhibition includes Ambler class A ESBLs, class C cephalosporinases, and class A and D carbapenemases (KPC and OXA-48, respectively). Rescue of ceftibuten by VNRX-5236 (fixed at 4 µg/ml) in isogenic strains of Escherichia coli expressing class A, C, or D ß-lactamases demonstrated an expanded spectrum of activity relative to oral comparators, including investigational penems, sulopenem, and tebipenem. VNRX-5236 rescued ceftibuten activity in clinical isolates of Enterobacterales expressing ESBLs (MIC90, 0.25 µg/ml), KPCs (MIC90, 1 µg/ml), class C cephalosporinases (MIC90, 1 µg/ml), and OXA-48-type carbapenemases (MIC90, 1 µg/ml). Frequency of resistance studies demonstrated a low propensity for recovery of resistant variants at 4× the MIC of the ceftibuten/VNRX-5236 combination. In vivo, whereas ceftibuten alone was ineffective (50% effective dose [ED50], >128 mg/kg), ceftibuten/VNRX-7145 administered orally protected mice from lethal septicemia caused by Klebsiella pneumoniae producing KPC carbapenemase (ED50, 12.9 mg/kg). The data demonstrate potent, broad-spectrum rescue of ceftibuten activity by VNRX-5236 in clinical isolates of cephalosporin-resistant and carbapenem-resistant Enterobacterales.


Cephalosporins , beta-Lactamase Inhibitors , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins , Carbapenems/pharmacology , Ceftibuten , Cephalosporins/pharmacology , Mice , Microbial Sensitivity Tests , Serine , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics
5.
Sci Rep ; 9(1): 2904, 2019 02 27.
Article En | MEDLINE | ID: mdl-30814582

In Gram-negative bacterial sepsis, production of excess pro-inflammatory cytokines results in hyperinflammation and tissue injury. Anti-inflammatory cytokines such as IL-10 inhibit inflammation and enhance tissue healing. Here, we report a novel approach to treat septicemia associated with intra-abdominal infection in a murine model by delicately balancing pro- and anti-inflammatory cytokines. A novel oligosaccharide compound AVR-25 selectively binds to the TLR4 protein (IC50 = 0.15 µM) in human peripheral blood monocytes and stimulates IL-10 production. Following the cecal ligation and puncture (CLP) procedure, intravenous dosing of AVR-25 (10 mg/kg, 6-12 h post-CLP) alone and in combination with antibiotic imipenem protected both young adult (10-12 week old) and aged (16-18 month old) mice against polymicrobial infection, organ dysfunction, and death. Proinflammatory cytokines (TNF-α, MIP-1, i-NOS) were decreased significantly and restoration of tissue damage was observed in all organs. A decrease in serum C-reactive protein (CRP) and bacterial colony forming unit (CFU) confirmed improved bacterial clearance. Together, these findings demonstrate the therapeutic ability of AVR-25 to mitigate the storm of inflammation and minimize tissue injury with high potential for adjunctive therapy in intra-abdominal sepsis.


Aging/physiology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chitin/therapeutic use , Gram-Negative Bacterial Infections/drug therapy , Intraabdominal Infections/drug therapy , Leukocytes, Mononuclear/drug effects , Oligosaccharides/therapeutic use , Sepsis/prevention & control , Animals , Cecum/surgery , Cells, Cultured , Chitin/chemistry , Cytokines/metabolism , Disease Models, Animal , Gram-Negative Bacterial Infections/complications , Humans , Inflammation Mediators/metabolism , Intraabdominal Infections/complications , Leukocytes, Mononuclear/physiology , Mice , Mice, Inbred C57BL , Oligosaccharides/chemistry , Sepsis/etiology , Toll-Like Receptor 4/metabolism
6.
Article En | MEDLINE | ID: mdl-30670434

Lantibiotics present an attractive scaffold for the development of novel antibiotics. We report here a novel lantibiotic for the treatment of Clostridium difficile infection. The lead compounds were selected from a library of over 700 single- and multiple-substitution variants of the lantibiotic mutacin 1140 (MU1140). The best performers in vitro and in vivo were further used to challenge Golden Syrian hamsters orally in a Golden Syrian hamster model of Clostridium difficile-associated disease (CDAD) in a dose-response format, resulting in the selection of OG716 as the lead compound. This lantibiotic was characterized by a 50% effective dose of 23.85 mg/kg of body weight/day (10.97 µmol/kg/day) in this model. Upon oral administration of the maximum feasible dose (≥1,918 mg/kg/day), no observable toxicities or side effects were noted, and no effect on intestinal motility was observed. Compartmentalization to the gastrointestinal tract was confirmed. MU1140-derived variants offer a large pipeline for the development of novel antibiotics for the treatment of several indications and are particularly attractive considering their novel mechanism of action. Based on the currently available data, OG716 has an acceptable profile for further development for the treatment of CDAD.


Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Clostridium Infections/drug therapy , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemistry , Bacteriocins/administration & dosage , Bacteriocins/adverse effects , Bacteriocins/chemistry , Biological Availability , Cecum/microbiology , Clostridium Infections/mortality , Colony Count, Microbial , Dose-Response Relationship, Drug , Female , Gastric Emptying/drug effects , Male , Maximum Tolerated Dose , Mesocricetus , Rats, Wistar
7.
PLoS One ; 13(6): e0197467, 2018.
Article En | MEDLINE | ID: mdl-29894469

Lantibiotics continue to offer an untapped pipeline for the development of novel antibiotics. We report here the discovery of a novel lantibiotic for the treatment of C. difficile infection (CDI). The leads were selected from a library of over 300 multiple substitution variants of the lantibiotic Mutacin 1140 (MU1140). Top performers were selected based on testing for superior potency, solubility, manufacturability, and physicochemical and/or metabolic stability in biologically-relevant systems. The best performers in vitro were further evaluated orally in the Golden Syrian hamster model of CDAD. In vivo testing ultimately identified OG716 as the lead compound, which conferred 100% survival and no relapse at 3 weeks post infection. MU1140-derived variants are particularly attractive for further clinical development considering their novel mechanism of action.


Bacteriocins/administration & dosage , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Animals , Clostridioides difficile/pathogenicity , Clostridium Infections/microbiology , Cricetinae , Disease Models, Animal , Humans , Mesocricetus
8.
Front Microbiol ; 9: 415, 2018.
Article En | MEDLINE | ID: mdl-29615987

Lantibiotics offer an untapped pipeline for the development of novel antibiotics to treat serious Gram-positive (+) infections including Clostridium difficile. Mutacin 1140 (MU1140) is a lantibiotic produced by Streptococcus mutans and acts via a novel mechanism of action, which may limit the development of resistance. This study sought to identify a lead compound for the treatment of C. difficile associated diarrhea (CDAD). Compounds were selected from a saturation mutagenesis library of 418 single amino acid variants of MU1140. Compounds were produced by small scale fermentation, purified, characterized and then subjected to a panel of assays aimed at identifying the best performers. The screening assays included: in vitro susceptibility testing [MIC against Micrococcus luteus, Clostridium difficile, vancomycin-resistant enterococci (VRE), Staphylococcus aureus, Streptococcus pneumonia, Mycobacterium phlei, and Pseudomonas aeruginosa; cytotoxicity screening on HepG2 hepatocytes; in vitro pharmacological profiling with the Safety Screen 44TM, metabolic and chemical stability in biologically relevant fluids (FaSSGF, FaSSIF and serum); and efficacy in vivo]. Several lantibiotic compounds had better MIC against C. difficile, compared to vancomycin, but not against other bacterial species tested. The Safety Screen 44TMin vitro pharmacological profiling assay suggested that this class of compounds has relatively low overall toxicity and that compound OG253 (MU1140, Phe1Ile) is not likely to present inadvertent off-target effects, as evidenced by a low promiscuity score. The in vitro cytotoxicity assay also indicated that this class of compounds was characterized by low toxicity; the EC50 of OG253 was 636 mg/mL on HepG2 cells. The half-life in simulated gastric fluid was >240 min. for all compound tested. The stability in simulated intestinal fluid ranged between a half-life of 5 min to >240 min, and paralleled the half-life in serum. OG253 ultimately emerged as the lead compound based on superior in vivo efficacy along with an apparent lack of relapse in a hamster model of infection. The lessons learned from this report are applicable to therapeutic lanthipeptides in general and may assist in the design of novel molecules with improved pharmacological, therapeutic and physicochemical profiles. The data presented also support the continued clinical development of OG253 as a novel antibiotic against CDAD that could prevent recurrence of the infection.

9.
Article En | MEDLINE | ID: mdl-29038270

The recently approved combination of meropenem and vaborbactam (Vabomere) is highly active against Gram-negative pathogens, especially Klebsiella pneumoniae carbapenemase (KPC)-producing, carbapenem-resistant Enterobacteriaceae We evaluated the efficacy of meropenem-vaborbactam against three clinically relevant isolates in a murine pyelonephritis model. The data indicate that the combination of meropenem and vaborbactam significantly increased bacterial killing compared to that with the untreated controls. These data suggest that this combination may have utility in the treatment of complicated urinary tract infections due to KPC-producing, carbapenem-resistant Enterobacteriaceae.


Anti-Bacterial Agents/therapeutic use , Boronic Acids/therapeutic use , Carbapenem-Resistant Enterobacteriaceae/drug effects , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Meropenem/therapeutic use , Pyelonephritis/drug therapy , Urinary Tract Infections/drug therapy , beta-Lactamase Inhibitors/therapeutic use , Animals , Bacterial Proteins/metabolism , Carbapenem-Resistant Enterobacteriaceae/isolation & purification , Disease Models, Animal , Drug Combinations , Humans , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/metabolism , Mice , Microbial Sensitivity Tests , Pyelonephritis/microbiology , Urinary Tract Infections/microbiology , beta-Lactamases/metabolism
10.
Article En | MEDLINE | ID: mdl-28167541

Based upon knowledge of the hydrolytic profile of major ß-lactamases found in Gram-negative bacteria, we tested the efficacy of the combination of ceftazidime-avibactam (CAZ-AVI) with aztreonam (ATM) against carbapenem-resistant enteric bacteria possessing metallo-ß-lactamases (MBLs). Disk diffusion and agar-based antimicrobial susceptibility testing were initially performed to determine the in vitro efficacy of a unique combination of CAZ-AVI and ATM against 21 representative Enterobacteriaceae isolates with a complex molecular background that included blaIMP, blaNDM, blaOXA-48, blaCTX-M, blaAmpC, and combinations thereof. Time-kill assays were conducted, and the in vivo efficacy of this combination was assessed in a murine neutropenic thigh infection model. By disk diffusion assay, all 21 isolates were resistant to CAZ-AVI alone, and 19/21 were resistant to ATM. The in vitro activity of CAZ-AVI in combination with ATM against diverse Enterobacteriaceae possessing MBLs was demonstrated in 17/21 isolates, where the zone of inhibition was ≥21 mm. All isolates demonstrated a reduction in CAZ-AVI agar dilution MICs with the addition of ATM. At 2 h, time-kill assays demonstrated a ≥4-log10-CFU decrease for all groups that had CAZ-AVI with ATM (8 µg/ml) added, compared to the group treated with CAZ-AVI alone. In the murine neutropenic thigh infection model, an almost 4-log10-CFU reduction was noted at 24 h for CAZ-AVI (32 mg/kg every 8 h [q8h]) plus ATM (32 mg/kg q8h) versus CAZ-AVI (32 mg/kg q8h) alone. The data presented herein require us to carefully consider this new therapeutic combination to treat infections caused by MBL-producing Enterobacteriaceae.


Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/pharmacology , Aztreonam/pharmacology , Ceftazidime/pharmacology , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae/drug effects , Animals , Colony Count, Microbial , Cyclophosphamide , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Enterobacteriaceae/enzymology , Enterobacteriaceae/genetics , Enterobacteriaceae/growth & development , Enterobacteriaceae Infections/microbiology , Female , Gene Expression , Humans , Klebsiella Infections/complications , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/growth & development , Mice , Microbial Sensitivity Tests , Neutropenia/chemically induced , Neutropenia/complications , Neutropenia/drug therapy , Neutropenia/microbiology , Plasmids/chemistry , Plasmids/metabolism , Soft Tissue Infections/complications , Soft Tissue Infections/drug therapy , Soft Tissue Infections/microbiology , Thigh , beta-Lactam Resistance/drug effects , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism
11.
Antimicrob Agents Chemother ; 55(1): 82-5, 2011 Jan.
Article En | MEDLINE | ID: mdl-21041503

We evaluated the efficacy of NXL104, a novel ß-lactamase inhibitor, in combination with ceftazidime (CAZ) in two murine infection models (septicemia and thigh infection). We chose two KPC-producing Klebsiella pneumoniae strains (VA-361 and VA-406) showing MICs of CAZ of ≥256 µg/ml. Septicemia was induced by the intraperitoneal injection of KPC-producing K. pneumoniae followed 30 min later by a single subcutaneous treatment with CAZ alone or CAZ-NXL104 in ratios of 2:1, 4:1, 8:1, and 16:1. In this model, the median effective doses for 50% (ED(50)) of the animals for CAZ alone versus VA-361 and VA-406 were 1,578 and 709 mg/kg of body weight, respectively. When combined with NXL104 at 2:1, 4:1, 8:1, and 16:1 ratios, the CAZ ED(50)s for VA-361 and VA-406 were reduced to 8.1 and 3.5 mg/kg, 15.1 and 3.8 mg/kg, 16.9 and 7.2 mg/kg, and 29.5 and 12.1 mg/kg, respectively. For thigh infection, neutropenia was induced by the intraperitoneal injection of cyclophosphamide at days -4 and -1 preinfection. Infection was established by the intramuscular injection of KPC-producing K. pneumoniae into the right thigh. Mice were treated 1.5 h postinfection with either CAZ alone or CAZ-NXL104 at constant ratios of 4:1. When thighs were removed at 24 h postinfection, a >2-log CFU reduction was observed for mice treated with CAZ-NXL104 at doses of ≥128:32 mg/kg. In contrast, CAZ doses of ≥1,024 mg/kg were unable to reduce the numbers of CFU. Despite resistance to CAZ and possessing a complex ß-lactamase background, NXL104 combined with CAZ proved to be very effective in murine models of infection due to contemporary highly resistant KPC-producing K. pneumoniae isolates.


Anti-Bacterial Agents/therapeutic use , Azabicyclo Compounds/therapeutic use , Ceftazidime/therapeutic use , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/physiology , Animals , Disease Models, Animal , Female , Mice , Microbial Sensitivity Tests , Sepsis/drug therapy
12.
J Pharm Biomed Anal ; 46(4): 723-7, 2008 Mar 13.
Article En | MEDLINE | ID: mdl-18178051

The aim of this study was to develop a specific and sensitive liquid chromatography mass spectrometry (LC/MS) method for the determination of rifampicin and levofloxacin concentrations from infected tissues within teflon catheter segments which were subcutaneously implanted in mice. A solid-phase extraction procedure was used to extract analytes from tissue homogenates of the catheter segments and reverse-phase HPLC combined with positive electrospray ionization mass spectrometry was used for analyte separation and quantification. The assay was found to be linear over the concentration range of 0.02-2 microg/g for rifampicin and levofloxacin in tissues and provided good validation data for accuracy and precision. The intra-day accuracy as determined by the relative error was -1.3% for levofloxacin and 6.1% for rifampicin, and precision was evaluated by R.S.D.s with a maximum of 5.1% for levofloxacin and 8.1% for rifampicin. The inter-day accuracy was -3.3% for levofloxacin and -4.6% for rifampicin, and precision was 8.6% for levofloxacin and 7.1% for rifampicin. The assay uses less tissue than previously described methods and can be applied to determine the penetration of rifampicin and the fluoroquinolone in catheter segments from a mouse model of a device-related infection. Finally, the HPLC-MS assay should be applicable to studies of rifamycin+quinolone combination therapies in other animal models of bacterial infection.


Anti-Bacterial Agents/analysis , Catheters, Indwelling/microbiology , Chromatography, High Pressure Liquid/methods , Levofloxacin , Ofloxacin/analysis , Rifampin/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C , Ofloxacin/pharmacology , Rifampin/pharmacology , Sensitivity and Specificity , Staphylococcus aureus/drug effects
...