Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection

K El Bakkouri, F Descamps, M De Filette… - The Journal of …, 2011 - journals.aai.org
K El Bakkouri, F Descamps, M De Filette, A Smet, E Festjens, A Birkett, N Van Rooijen
The Journal of Immunology, 2011journals.aai.org
The ectodomain of matrix protein 2 (M2e) of influenza A virus is an attractive target for a
universal influenza A vaccine: the M2e sequence is highly conserved across influenza virus
subtypes, and induced humoral anti-M2e immunity protects against a lethal influenza virus
challenge in animal models. Clinical phase I studies with M2e vaccine candidates have
been completed. However, the in vivo mechanism of immune protection induced by M2e-
carrier vaccination is unclear. Using passive immunization experiments in wild-type …
Abstract
The ectodomain of matrix protein 2 (M2e) of influenza A virus is an attractive target for a universal influenza A vaccine: the M2e sequence is highly conserved across influenza virus subtypes, and induced humoral anti-M2e immunity protects against a lethal influenza virus challenge in animal models. Clinical phase I studies with M2e vaccine candidates have been completed. However, the in vivo mechanism of immune protection induced by M2e-carrier vaccination is unclear. Using passive immunization experiments in wild-type, FcRγ−/−, FcγRI−/−, FcγRIII−/−, and (FcγRI, FcγRIII)−/− mice, we report in this study that Fc receptors are essential for anti-M2e IgG-mediated immune protection. M2e-specific IgG1 isotype Abs are shown to require functional FcγRIII for in vivo immune protection but other anti-M2e IgG isotypes can rescue FcγRIII−/− mice from a lethal challenge. Using a conditional cell depletion protocol, we also demonstrate that alveolar macrophages (AM) play a crucial role in humoral M2e-specific immune protection. Additionally, we show that adoptive transfer of wild-type AM into (FcγRI, FcγRIII)−/− mice restores protection by passively transferred anti-M2e IgG. We conclude that AM and Fc receptor-dependent elimination of influenza A virus-infected cells are essential for protection by anti-M2e IgG.
journals.aai.org