Your browser doesn't support javascript.
loading
Potent Killing of Pseudomonas aeruginosa by an Antibody-Antibiotic Conjugate.
Kajihara, Kimberly K; Pantua, Homer; Hernandez-Barry, Hilda; Hazen, Meredith; Deshmukh, Kiran; Chiang, Nancy; Ohri, Rachana; Castellanos, Erick R; Martin, Lynn; Matsumoto, Marissa L; Payandeh, Jian; Storek, Kelly M; Schneider, Kellen; Smith, Peter A; Koehler, Michael F T; Tsai, Siao Ping; Vandlen, Richard; Loyet, Kelly M; Nakamura, Gerald; Pillow, Thomas; Seshasayee, Dhaya; Kapadia, Sharookh B; Hazenbos, Wouter L W.
Afiliación
  • Kajihara KK; Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA.
  • Pantua H; Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA.
  • Hernandez-Barry H; Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, USA.
  • Hazen M; Department of Antibody Engineering, Genentech, Inc., South San Francisco, California, USA.
  • Deshmukh K; Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, USA.
  • Chiang N; Department of Antibody Engineering, Genentech, Inc., South San Francisco, California, USA.
  • Ohri R; Department of Protein Chemistry, Genentech, Inc., South San Francisco, California, USA.
  • Castellanos ER; Department of Structural Biology, Genentech, Inc., South San Francisco, California, USA.
  • Martin L; Department of BioMolecular Resources, Genentech, Inc., South San Francisco, California, USA.
  • Matsumoto ML; Department of Structural Biology, Genentech, Inc., South San Francisco, California, USA.
  • Payandeh J; Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA.
  • Storek KM; Department of Antibody Engineering, Genentech, Inc., South San Francisco, California, USA.
  • Schneider K; Department of Structural Biology, Genentech, Inc., South San Francisco, California, USA.
  • Smith PA; Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA.
  • Koehler MFT; Department of Antibody Engineering, Genentech, Inc., South San Francisco, California, USA.
  • Tsai SP; Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA.
  • Vandlen R; Department of Medicinal Chemistry; Genentech, Inc., South San Francisco, California, USA.
  • Loyet KM; Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, USA.
  • Nakamura G; Department of Protein Chemistry, Genentech, Inc., South San Francisco, California, USA.
  • Pillow T; Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, USA.
  • Seshasayee D; Department of Antibody Engineering, Genentech, Inc., South San Francisco, California, USA.
  • Kapadia SB; Department of Medicinal Chemistry; Genentech, Inc., South San Francisco, California, USA.
  • Hazenbos WLW; Department of Antibody Engineering, Genentech, Inc., South San Francisco, California, USA.
mBio ; 12(3): e0020221, 2021 06 29.
Article en En | MEDLINE | ID: mdl-34061593
ABSTRACT
Pseudomonas aeruginosa causes life-threatening infections that are associated with antibiotic failure. Previously, we identified the antibiotic G2637, an analog of arylomycin, targeting bacterial type I signal peptidase, which has moderate potency against P. aeruginosa. We hypothesized that an antibody-antibiotic conjugate (AAC) could increase its activity by colocalizing P. aeruginosa bacteria with high local concentrations of G2637 antibiotic in the intracellular environment of phagocytes. Using a novel technology of screening for hybridomas recognizing intact bacteria, we identified monoclonal antibody 26F8, which binds to lipopolysaccharide O antigen on the surface of P. aeruginosa bacteria. This antibody was engineered to contain 6 cysteines and was conjugated to the G2637 antibiotic via a lysosomal cathepsin-cleavable linker, yielding a drug-to-antibody ratio of approximately 6. The resulting AAC delivered a high intracellular concentration of free G2637 upon phagocytosis of AAC-bound P. aeruginosa by macrophages, and potently cleared viable P. aeruginosa bacteria intracellularly. The molar concentration of AAC-associated G2637 antibiotic that resulted in elimination of bacteria inside macrophages was approximately 2 orders of magnitude lower than the concentration of free G2637 required to eliminate extracellular bacteria. This study demonstrates that an anti-P. aeruginosa AAC can locally concentrate antibiotic and kill P. aeruginosa inside phagocytes, providing additional therapeutic options for antibiotics that are moderately active or have an unfavorable pharmacokinetics or toxicity profile. IMPORTANCE Antibiotic treatment of life-threatening P. aeruginosa infections is associated with low clinical success, despite the availability of antibiotics that are active in standard microbiological in vitro assays, affirming the need for new therapeutic approaches. Antibiotics often fail in the preclinical stage due to insufficient efficacy against P. aeruginosa. One potential strategy is to enhance the local concentration of antibiotics with limited inherent anti-P. aeruginosa activity. This study presents proof of concept for an antibody-antibiotic conjugate, which releases a high local antibiotic concentration inside macrophages upon phagocytosis, resulting in potent intracellular killing of phagocytosed P. aeruginosa bacteria. This approach may provide new therapeutic options for antibiotics that are dose limited.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Pseudomonas aeruginosa / Macrófagos / Antibacterianos / Anticuerpos Monoclonales Límite: Animals / Humans Idioma: En Revista: MBio Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Pseudomonas aeruginosa / Macrófagos / Antibacterianos / Anticuerpos Monoclonales Límite: Animals / Humans Idioma: En Revista: MBio Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos