C. Kendall Stover
MedImmune
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by C. Kendall Stover.
Proceedings of the National Academy of Sciences of the United States of America | 2009
J. Richard Miller; Steve Dunham; Igor Mochalkin; Craig Banotai; Matthew Bowman; Susan Buist; Bill Dunkle; Debra Hanna; H. James Harwood; Michael D. Huband; Alla Karnovsky; Michael Kuhn; Chris Limberakis; Jia Y. Liu; Shawn Mehrens; W. Thomas Mueller; Lakshmi Narasimhan; Adam Ogden; Jeff Ohren; J. V. N. Vara Prasad; John A. Shelly; Laura Skerlos; Mark C. Sulavik; V. Hayden Thomas; Steve VanderRoest; Li Ann Wang; Zhigang Wang; Amy Whitton; Tong Zhu; C. Kendall Stover
As the need for novel antibiotic classes to combat bacterial drug resistance increases, the paucity of leads resulting from target-based antibacterial screening of pharmaceutical compound libraries is of major concern. One explanation for this lack of success is that antibacterial screening efforts have not leveraged the eukaryotic bias resulting from more extensive chemistry efforts targeting eukaryotic gene families such as G protein-coupled receptors and protein kinases. Consistent with a focus on antibacterial target space resembling these eukaryotic targets, we used whole-cell screening to identify a series of antibacterial pyridopyrimidines derived from a protein kinase inhibitor pharmacophore. In bacteria, the pyridopyrimidines target the ATP-binding site of biotin carboxylase (BC), which catalyzes the first enzymatic step of fatty acid biosynthesis. These inhibitors are effective in vitro and in vivo against fastidious Gram-negative pathogens including Haemophilus influenzae. Although the BC active site has architectural similarity to those of eukaryotic protein kinases, inhibitor binding to the BC ATP-binding site is distinct from the protein kinase-binding mode, such that the inhibitors are selective for bacterial BC. In summary, we have discovered a promising class of potent antibacterials with a previously undescribed mechanism of action. In consideration of the eukaryotic bias of pharmaceutical libraries, our findings also suggest that pursuit of a novel inhibitor leads for antibacterial targets with active-site structural similarity to known human targets will likely be more fruitful than the traditional focus on unique bacterial target space, particularly when structure-based and computational methodologies are applied to ensure bacterial selectivity.
Science Translational Medicine | 2014
Antonio DiGiandomenico; Ashley E. Keller; Cuihua Gao; Godfrey Rainey; Paul Warrener; Mareia M. Camara; Ryan Fleming; Binyam Bezabeh; Nazzareno Dimasi; Bret R. Sellman; Jamese J. Hilliard; Caitlin M. Guenther; Vivekananda Datta; Wei Zhao; Changshou Gao; Xiang-Qing Yu; JoAnn Suzich; C. Kendall Stover
A new antibody platform combining anti-Psl and anti-PcrV activities provides enhanced protection and acts synergistically with antibiotics against Pseudomonas aeruginosa. Bispecific Antibodies Protect Against Pseudomonas Multifunctional bispecific antibodies were constructed conferring three mechanisms of action against the bacterial pathogen Pseudomonas aeruginosa by targeting both the type III secretion injectisome virulence factor PcrV and the persistence factor Psl exopolysaccharide (DiGiandomenico et al.). A new multimechanistic bispecific antibody platform called BiS4αPa exhibited targeted synergistic protection against P. aeruginosa in a mouse model of lung infection compared to the parent monoclonal antibody combination. This BiS4αPa construct, now designated clinical candidate MEDI3902, was also protective in mouse models of thermal injury, bacteremia, and immunosuppression and synergistically enhanced treatment with multiple antibiotic classes. This study suggests that multifunctional bispecific antibodies may be a promising platform for targeting other antibiotic-resistant bacterial pathogens. Widespread drug resistance due to empiric use of broad-spectrum antibiotics has stimulated development of bacteria-specific strategies for prophylaxis and therapy based on modern monoclonal antibody (mAb) technologies. However, single-mechanism mAb approaches have not provided adequate protective activity in the clinic. We constructed multifunctional bispecific antibodies, each conferring three mechanisms of action against the bacterial pathogen Pseudomonas aeruginosa by targeting the serotype-independent type III secretion system (injectisome) virulence factor PcrV and persistence factor Psl exopolysaccharide. A new bispecific antibody platform, BiS4, exhibited superior synergistic protection against P. aeruginosa–induced murine pneumonia compared to parent mAb combinations or other available bispecific antibody structures. BiS4αPa was protective in several mouse infection models against disparate P. aeruginosa strains and unexpectedly further synergized with multiple antibiotic classes even against drug-resistant clinical isolates. In addition to resulting in a multimechanistic clinical candidate (MEDI3902) for the prevention or treatment of P. aeruginosa infections, these antibody studies suggest that multifunctional antibody approaches may be a promising platform for targeting other antibiotic-resistant bacterial pathogens.
ACS Chemical Biology | 2009
Igor Mochalkin; J. Richard Miller; Lakshmi Narasimhan; Venkataraman Thanabal; Paul Erdman; Philip B. Cox; J. V. N. Vara Prasad; Sandra Lightle; Michael D. Huband; C. Kendall Stover
As part of our effort to inhibit bacterial fatty acid biosynthesis through the recently validated target biotin carboxylase, we employed a unique combination of two emergent lead discovery strategies. We used both de novo fragment-based drug discovery and virtual screening, which employs 3D shape and electrostatic property similarity searching. We screened a collection of unbiased low-molecular-weight molecules and identified a structurally diverse collection of weak-binding but ligand-efficient fragments as potential building blocks for biotin carboxylase ATP-competitive inhibitors. Through iterative cycles of structure-based drug design relying on successive fragment costructures, we improved the potency of the initial hits by up to 3000-fold while maintaining their ligand-efficiency and desirable physicochemical properties. In one example, hit-expansion efforts resulted in a series of amino-oxazoles with antibacterial activity. These results successfully demonstrate that virtual screening approaches can substantially augment fragment-based screening approaches to identify novel antibacterial agents.
Vaccine | 1999
Robert Edelman; Kathleen Palmer; Karin Russ; Helen P Secrest; Jo Anna L. Becker; Sacared A Bodison; Judith Perry; Alvin R Sills; Alan G. Barbour; Catherine J. Luke; Mark S. Hanson; C. Kendall Stover; Jeanne Burlein; Geetha P. Bansal; Edward M. Connor; Scott Koenig
This phase I clinical trial was designed to determine the feasibility of using rBCG as a live bacterial vaccine vector for the outer surface protein A (OspA) of Borrelia burgdorferi and as model for other vaccines based on a rBCG vector. To construct the vaccine, a signal peptide derived from a mycobacterial lipoprotein was used to direct the export, and membrane-associated surface expression, of OspA in a standard strain of BCG (Connaught). The rBCG OspA vaccine was safe and immunogenic in several animal species, and protective in a mouse model of Lyme borreliosis. An intradermal injection (0.1 ml) of rBCG OspA was administered to 24 healthy adult volunteers sequentially at one of four dose levels, ranging from 2.0 x 10(4) CFU to 2 x 10(7) CFU, using a dose-escalation design. All volunteers were initially PPD-skin test and OspA antibody negative, and they were monitored for 2 years after immunization. Three volunteers had mild flu-like reactions 1-2 days after vaccination. Local ulceration and drainage at the site of injection, which occurred in 50% and 83% of volunteers in the two highest dose groups, persisted for 1-70 days before the ulcers healed. Most of the drainage samples yielded rBCG colonies that contained the OspA plasmid. Thirteen of 24 vaccinees, principally in the two highest dose groups, converted their PPD skin tests from negative to positive. None of the 24 volunteers developed OspA antibody. In conclusion, the current rBCG vaccine construct, the first such construct tested in humans, had a safety profile comparable to that of licensed BCG, but it did not elicit primary humoral responses to the vectored antigen.
Antimicrobial Agents and Chemotherapy | 2014
Paul Warrener; Reena Varkey; Antonio DiGiandomenico; Maria Margarita Camara; Kimberly E. Cook; Li Peng; Jingying Zha; Partha Chowdury; Bret R. Sellman; C. Kendall Stover
ABSTRACT Pseudomonas aeruginosa is a major cause of hospital-acquired infections, particularly in mechanically ventilated patients, and it is the leading cause of death in cystic fibrosis patients. A key virulence factor associated with disease severity is the P. aeruginosa type III secretion system (T3SS), which injects bacterial toxins directly into the cytoplasm of host cells. The PcrV protein, located at the tip of the T3SS injectisome complex, is required for T3SS function and is a well-validated target in animal models of immunoprophylactic strategies targeting P. aeruginosa. In an effort to identify a highly potent and protective monoclonal antibody (MAb) that inhibits the T3SS, we generated and characterized a panel of novel anti-PcrV MAbs. Interestingly, some MAbs exhibiting potent inhibition of T3SS in vitro failed to provide protection in a mouse model of P. aeruginosa infection, suggesting that effective in vivo inhibition of T3SS with anti-PcrV MAbs is epitope dependent. V2L2MD, while not the most potent MAb as assessed by in vitro cytotoxicity inhibition assays, provided strong prophylactic protection in several murine infection models and a postinfection therapeutic model. V2L2MD mediated significantly (P < 0.0001) better in vivo protection than that provided by a comparator antibody, MAb166, a well-characterized anti-PcrV MAb and the progenitor of a clinical candidate, KB001-A. The results described here support further development of a V2L2MD-containing immunotherapeutic and may suggest even greater potential than was previously recognized for the prevention and treatment of P. aeruginosa infections in high-risk populations.
Mbio | 2015
Amy S. Jenkins; Binh An Diep; Thuy T. Mai; Nhung H. Vo; Paul Warrener; JoAnn Suzich; C. Kendall Stover; Bret R. Sellman
ABSTRACT Staphylococcus aureus is a Gram-positive, commensal bacterium known to asymptomatically colonize the human skin, nares, and gastrointestinal tract. Colonized individuals are at increased risk for developing S. aureus infections, which range from mild skin and soft tissue infections to more severe diseases, such as endocarditis, bacteremia, sepsis, and osteomyelitis. Different virulence factors are required for S. aureus to infect different body sites. In this study, virulence gene expression was analyzed in two S. aureus isolates during nasal colonization, bacteremia and in the heart during sepsis. These models were chosen to represent the stepwise progression of S. aureus from an asymptomatic colonizer to an invasive pathogen. Expression of 23 putative S. aureus virulence determinants, representing protein and carbohydrate adhesins, secreted toxins, and proteins involved in metal cation acquisition and immune evasion were analyzed. Consistent upregulation of sdrC, fnbA, fhuD, sstD, and hla was observed in the shift between colonization and invasive pathogen, suggesting a prominent role for these genes in staphylococcal pathogenesis. Finally, gene expression data were correlated to the roles of the genes in pathogenesis by using knockout mutants in the animal models. These results provide insights into how S. aureus modifies virulence gene expression between commensal and invasive pathogens. IMPORTANCE Many bacteria, such as Staphylococcus aureus, asymptomatically colonize human skin and nasal passages but can also cause invasive diseases, such as bacteremia, pneumonia, sepsis, and osteomyelitis. The goal of this study was to analyze differences in the expression of selected S. aureus genes during a commensal lifestyle and as an invasive pathogen to gain insight into the commensal-to-pathogen transition and how a bacterial pathogen adapts to different environments within a host (e.g., from nasal colonization to invasive pathogen). The gene expression data were also used to select genes for which to construct knockout mutants to assess the role of several proteins in nasal colonization and lethal bacteremia. These results not only provide insight into the factors involved in S. aureus disease pathogenesis but also provide potential therapeutic targets. Many bacteria, such as Staphylococcus aureus, asymptomatically colonize human skin and nasal passages but can also cause invasive diseases, such as bacteremia, pneumonia, sepsis, and osteomyelitis. The goal of this study was to analyze differences in the expression of selected S. aureus genes during a commensal lifestyle and as an invasive pathogen to gain insight into the commensal-to-pathogen transition and how a bacterial pathogen adapts to different environments within a host (e.g., from nasal colonization to invasive pathogen). The gene expression data were also used to select genes for which to construct knockout mutants to assess the role of several proteins in nasal colonization and lethal bacteremia. These results not only provide insight into the factors involved in S. aureus disease pathogenesis but also provide potential therapeutic targets.
Antimicrobial Agents and Chemotherapy | 2015
Jamese J. Hilliard; Vivekananda Datta; Christine Tkaczyk; Melissa Hamilton; Agnieszka Sadowska; Omari Jones-Nelson; Terrence O'Day; William J. Weiss; Szabolcs Szarka; Vien Nguyen; Laszlo Prokai; JoAnn Suzich; C. Kendall Stover; Bret R. Sellman
ABSTRACT Alpha-toxin (AT) is a major virulence determinant in Staphylococcus aureus skin and soft tissue infection models. We previously demonstrated that prophylactic administration of 2A3, an AT-neutralizing monoclonal antibody (MAb), prevents S. aureus disease in a mouse dermonecrosis model by neutralizing AT-mediated tissue necrosis and immune evasion. In the present study, MEDI4893*, an affinity-optimized version of 2A3, was characterized for therapeutic activity in the dermonecrosis model as a single agent and in combination with two frontline antibiotics, vancomycin and linezolid. MEDI4893* postinfection therapy was found to exhibit a therapeutic treatment window similar to that for linezolid but longer than that for vancomycin. Additionally, when combined with either vancomycin or linezolid, MEDI4893* resulted in reduced tissue damage, increased neutrophil and macrophage infiltration and abscess formation, and accelerated healing relative to those with the antibiotic monotherapies. These data suggest that AT neutralization with a potent MAb holds promise for both prophylaxis and adjunctive therapy with antibiotics and may be a valuable addition to currently available options for the treatment of S. aureus skin and soft tissue infections.
Science Translational Medicine | 2016
Taylor S. Cohen; Jamese J. Hilliard; Omari Jones-Nelson; Ashley E. Keller; Terrence O’Day; Christine Tkaczyk; Antonio DiGiandomenico; Melissa Hamilton; Mark Pelletier; Qun Wang; Binh An Diep; Vien T. M. Le; Lily Cheng; JoAnn Suzich; C. Kendall Stover; Bret R. Sellman
S. aureus α toxin promotes opportunistic co-infection, which can be neutralized with a pathogen-specific antibody. Toxic eviction There are many benefits to a good roommate, but the wrong choice can be toxic. Now, Cohen et al. examine the effects of co-habitation on lung infection. They found that α toxin produced by Staphylococcus aureus can worsen lung co-infection by Gram-negative bacteria by preventing acidification of bacteria-containing phagosomes, increasing proliferation, spread, and lethality. However, early treatment or prophylaxis with a neutralizing antibody to α toxin prevented this effect and promoted S. aureus clearance in a humanized mouse model. If this eviction occurs in humans, this approach may reduce co-infection risk in patients colonized with S. aureus. Broad-spectrum antibiotic use may adversely affect a patient’s beneficial microbiome and fuel cross-species spread of drug resistance. Although alternative pathogen-specific approaches are rationally justified, a major concern for this precision medicine strategy is that co-colonizing or co-infecting opportunistic bacteria may still cause serious disease. In a mixed-pathogen lung infection model, we find that the Staphylococcus aureus virulence factor α toxin potentiates Gram-negative bacterial proliferation, systemic spread, and lethality by preventing acidification of bacteria-containing macrophage phagosomes, thereby reducing effective killing of both S. aureus and Gram-negative bacteria. Prophylaxis or early treatment with a single α toxin neutralizing monoclonal antibody prevented proliferation of co-infecting Gram-negative pathogens and lethality while also promoting S. aureus clearance. These studies suggest that some pathogen-specific, antibody-based approaches may also work to reduce infection risk in patients colonized or co-infected with S. aureus and disparate drug-resistant Gram-negative bacterial opportunists.
The Journal of Infectious Diseases | 2016
Qun Wang; Chew-Shun Chang; Meghan E. Pennini; Mark Pelletier; Saravanan Rajan; Jingying Zha; Yan Chen; Romana Cvitkovic; Agnieszka Sadowska; Jenny Heidbrink Thompson; Hung Yu Lin; Arnita Barnes; Keith W. Rickert; Susan Wilson; C. Kendall Stover; William F. Dall'Acqua; Partha S. Chowdhury; Xiaodong Xiao
The increasing incidence of Klebsiella pneumoniae infections refractory to treatment with current broad-spectrum antibiotic classes warrants the exploration of alternative approaches, such as antibody therapy and/or vaccines, for prevention and treatment. However, the lack of validated targets shared by spectrums of clinical strains poses a significant challenge. We adopted a target-agnostic approach to identify protective antibodies against K. pneumoniae Several monoclonal antibodies were isolated from phage display and hybridoma platforms by functional screening for opsonophagocytic killing activity. We further identified their common target antigen to be MrkA, a major protein in the type III fimbriae complex, and showed that these serotype-independent anti-MrkA antibodies reduced biofilm formation in vitro and conferred protection in multiple murine pneumonia models. Importantly, mice immunized with purified MrkA proteins also showed reduced bacterial burden following K. pneumoniae challenge. Taken together, these results support MrkA as a promising target for K. pneumoniae antibody therapeutics and vaccines.
The Journal of Infectious Diseases | 2016
Joshua T. Thaden; Ashley E. Keller; Norah J. Shire; M. Margarita Camara; Linda G. Otterson; Mike Huband; Caitlin M. Guenther; Wei Zhao; Paul Warrener; C. Kendall Stover; Vance G. Fowler; Antonio DiGiandomenico
BACKGROUND The type 3 secretion protein PcrV and Psl exopolysaccharide are promising therapeutic antibody targets against Pseudomonas aeruginosa. We examined P. aeruginosa bloodstream infection (BSI) isolates for the ability to express PcrV and Psl and evaluated corresponding patient serum for active titers to these targets. METHODS We identified 114 patients with acute P. aeruginosa BSI; 56 cases were accompanied by acute sera. Serum was evaluated for PcrV- and Psl-specific immunoglobulin G (IgG) and for cytotoxicity and opsonophagocytosis. Isolates were evaluated for susceptibility to antibiotics, expression of PcrV and Psl, and susceptibility to the anti-PcrV/Psl bispecific antibody and clinical candidate MEDI3902. RESULTS In-hospital mortality for patients with P. aeruginosa BSI was 39%. A total of 26% of isolates were resistant to ≥3 antibiotic classes. Although PcrV and/or Psl were detected in 99% of isolates, a majority of patients lacked active titers to PcrV (100%) and Psl (98%). In addition, MEDI3902 was active against all tested isolates. CONCLUSIONS A vast majority of P. aeruginosa BSI isolates express PcrV and Psl; however, patient sera most often lacked IgG and functionally active responses to these targets. These results suggest that therapies directed at PcrV and Psl could be a promising approach for combating P. aeruginosa bloodstream infections.