Randomized Phase I: Safety, Immunogenicity and Mucosal Antiviral Activity in Young Healthy Women Vaccinated with HIV-1 Gp41 P1 Peptide on Virosomes

Geert Leroux-Roels, Cathy Maes, Frédéric Clement, Frank van Engelenburg, Marieke van den Dobbelsteen, Michael Adler, Mario Amacker, Lucia Lopalco, Morgane Bomsel, Anick Chalifour, Sylvain Fleury, Geert Leroux-Roels, Cathy Maes, Frédéric Clement, Frank van Engelenburg, Marieke van den Dobbelsteen, Michael Adler, Mario Amacker, Lucia Lopalco, Morgane Bomsel, Anick Chalifour, Sylvain Fleury

Abstract

Mucosal antibodies harboring various antiviral activities may best protect mucosal surfaces against early HIV-1 entry at mucosal sites and they should be ideally induced by prophylactic HIV-1 vaccines for optimal prevention of sexually transmitted HIV-1. A phase I, double-blind, randomized, placebo-controlled trial was conducted in twenty-four healthy HIV-uninfected young women. The study objectives were to assess the safety, tolerability and immunogenicity of virosomes harboring surface HIV-1 gp41-derived P1 lipidated peptides (MYM-V101). Participants received placebo or MYM-V101 vaccine at 10 μg/dose or 50 μg/dose intramuscularly at week 0 and 8, and intranasally at week 16 and 24. MYM-V101 was safe and well-tolerated at both doses administered by the intramuscular and intranasal routes, with the majority of subjects remaining free of local and general symptoms. P1-specific serum IgGs and IgAs were induced in all high dose recipients after the first injection. After the last vaccination, vaginal and rectal P1-specific IgGs could be detected in all high dose recipients. Approximately 63% and 43% of the low and high dose recipients were respectively tested positive for vaginal P1-IgAs, while 29% of the subjects from the high dose group tested positive for rectal IgAs. Serum samples had total specific IgG and IgA antibody concentrations ≥ 0.4 μg/mL, while mucosal samples were usually below 0.01 μg/mL. Vaginal secretions from MYM-V101 vaccinated subjects were inhibiting HIV-1 transcytosis but had no detectable neutralizing activity. P1-specific Th1 responses could not be detected on PBMC. This study demonstrates the excellent safety and tolerability of MYM-V101, eliciting systemic and mucosal antibodies in the majority of subjects. Vaccine-induced mucosal anti-gp41 antibodies toward conserved gp41 motifs were harboring HIV-1 transcytosis inhibition activity and may contribute to reduce sexually-transmitted HIV-1.

Trial registration: ClinicalTrials.gov NCT01084343.

Conflict of interest statement

Competing Interests: GL-R, CM, FC, FE, MD, M. Adler, M. Amacker, LL and MB received hononaria as consultants for Mymetics as sponsor. AC and SF are Mymetics employees. MB and SF own equity in the company. The commercial company's affiliations of FE, MD, M. Adler, M. Amacker, AC and SF do not alter the authors' ability to adhere to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Study flow chart.
Figure 1. Study flow chart.
All subjects screened, enrolled and randomized are indicated in the chart. A total of 24 subjects divided over 2 panels of 12 subjects, which was deemed sufficient to meet the objectives of this trial. The study encompassed 12 medical visits, supplemented by physical examination, recording of vital signs and body temperature, as well as collection of blood samples for safety evaluation. Interim safety reviews have been done prior to the second vaccine administration for each route and prior to start of the HD regimen. Serum (week −3, 0, 1, 4, 8, 9, 16, 17, 24, 25 and 29), vaginal (week −3, 17, 25 and 29) and rectal samples (week −3 and 25) were taken for secondary endpoint and ancillary studies. To evaluate the cellular immune response, PBMC were isolated on weeks −3, 1, 9, 17 and 25. One subject from HD withdrew consent after Visit 5/week 9, which includes the first two intramuscular injections and performed the early withdrawal visit at the time point of week 16. Consequently, safety and immunogenicity analyses were done only from visit 1 to 5. Rectal samples of 2 LD subjects at Baseline were discarded by mistake during sample work-up. Without baseline data, induction of rectal specific antibodies above baseline could not be estimated. LD: Low dose; HD: High dose. Allocation to a panel was done in consecutive order; Panel 1 (LD: number 101 to 112) was filled first followed by Panel 2 (HD: number 201 to 212). In each panel, 8 subjects received active treatment with MYM-V101 and 4 subjects received placebo MYM-IRIVm or MYM-IRIVn in a double-blind way.
Figure 2. Serum and mucosal anti-P1 specific…
Figure 2. Serum and mucosal anti-P1 specific antibody responses.
The presence of P1-specific IgGs and IgAs was measured by Imperacer® and data are presented as the mean ΔCT value from pre-immune (Baseline/week −3) and immune samples (week 0 to week 29). Panels A, D, G and J are for the placebo, Panels B, E, H and K are for the LD, Panels C, F, I and L are for the HD. To determine if ΔCT values of immune samples were significantly increased, respective to pre-immune samples (week −3), p values for serum samples were calculated for weeks 8, 9, 16, 17, 24, 25, and for vaginal samples weeks 17, 25 and 29. For determining if the injected vaccine had a boost effect, respective to the previous vaccination, p values were also estimated between weeks 9–17 (2nd versus 3rd injection) or 17–25 (3rd versus 4th injection): *0.01<p<0.05, **0.001<p<0.01, ***p<0.001. NS (not significant), t (trend), LD (low dose), HD (high dose). Whisker 10–90% percentile with minimum, maximum and median. Normally distributed immunological data were tested by paired t-test whereas for non-normal distributed data, the Wilcoxon signed rank-sum test was used to compare pre- and post-vaccination results.
Figure 3. In vitro HIV-1 transcytosis inhibition…
Figure 3. In vitro HIV-1 transcytosis inhibition by vaginal antibodies.
Vaginal samples were collected from placebo and MYM-V101 vaccinated groups for exploratory work on transcytosis. Panel A (Visit 9/week 25) and B (Visit 10/week 29) are showing the measured mean values of transcytosis inhibition with standard deviations (histogram bars), with the indicated median values for LD and HD subjects within the bars. Percentages of transcytosis inhibitions were determined in three independent experiments. Samples were defined as positive when the transcytosis inhibitions were above 50% and reproduced in at least 2 experiments. Samples indicated by the (x) symbol means that they are negative for transcytosis, as they had only 1 measurement above 50% but the mean of the different experiments has generated a mean value above 50% (see method section). For each subject, the corresponding ΔΔCT value for P1-specific IgGs and IgAs is indicated: Samples with values above 1.996 are positive for the presence of specific antibodies. Low Dose (LD) dark grey bars, High Dose (HD) light grey bars and placebo with white bars. Panel C is showing all individual transcytosis data of each placebo and MYM-V101 subject from each representative experiment at week 25 and 29, allowing an overall qualitative comparison of the presence of transcytosis inhibition activity in vaginal samples. Placebo week 25 (opened square) compared with MYM-V101 week 25 (black circle), and placebo week 29 (opened triangle), as compared to MYM-V101 week 29 (black triangle).

References

    1. Shattock RJ, Moore JP (2003) Inhibiting sexual transmission of HIV-1 infection. Nat Rev Microbiol 1: 25–34.
    1. Hartley O, Klasse PJ, Sattentau QJ, Moore JP (2005) V3: HIV's switch-hitter. AIDS Res Hum Retroviruses 21: 171–189.
    1. Montero M, van Houten NE, Wang X, Scott1 JK (2008) The membrane-proximal external region of the human immunodeficiency virus type 1 envelope: Dominant site of antibody neutralization and target for vaccine design. Microbiology and molecular biology reviews 72: 54–84.
    1. Plotkin SA, Orenstein WA, Offit PA (2008) Vaccines 5th Edition.
    1. Mascola JR, Montefiori DC (2010) The role of antibodies in HIV vaccines. Annu Rev Immunol 28: 413–444.
    1. Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, et al. (1983) Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 220: 868–871.
    1. Fuchs JD, Sobieszczyk ME, Hammer SM, Buchbinder SP (2010) Lessons drawn from recent HIV vaccine efficacy trials. J Acquir Immune Defic Syndr 55 Suppl 2S128–131.
    1. IAVI (2010) Database of AIDS vaccine candidate in clinical trials (database online). IAVI Report July 7.
    1. Billich A (2004) AIDSVAX VaxGen. Curr Opin Investig Drugs 5: 214–221.
    1. Burton DR, Desrosiers RC, Doms RW, Koff WC, Kwong PD, et al. (2004) HIV vaccine design and the neutralizing antibody problem. Nat Immunol 5: 233–236.
    1. Hessell AJ, Rakasz EG, Poignard P, Hangartner L, Landucci G, et al. (2009) Broadly neutralizing human anti-HIV antibody 2G12 is effective in protection against mucosal SHIV challenge even at low serum neutralizing titers. PLoS Pathog 5: e1000433.
    1. Mascola JR (2002) Passive transfer studies to elucidate the role of antibody-mediated protection against HIV-1. Vaccine 20: 1922–1925.
    1. Zolla-Pazner S (2004) Identifying epitopes of HIV-1 that induce protective antibodies. Nat Rev Immunol 4: 199–210.
    1. Bernard NF, Pederson K, Chung F, Ouellet L, Wainberg MA, et al. (1998) HIV-specific cytotoxic T-lymphocyte activity in immunologically normal HIV-infected persons. AIDS 12: 2125–2139.
    1. Casimiro DR, Wang F, Schleif WA, Liang X, Zhang ZQ, et al. (2005) Attenuation of simian immunodeficiency virus SIVmac239 infection by prophylactic immunization with dna and recombinant adenoviral vaccine vectors expressing Gag. J Virol 79: 15547–15555.
    1. Pantaleo G, Koup RA (2004) Correlates of immune protection in HIV-1 infection: what we know, what we don't know, what we should know. Nat Med 10: 806–810.
    1. Pontesilli O, Klein MR, Kerkhof-Garde SR, Pakker NG, de Wolf F, et al. (1998) Longitudinal analysis of human immunodeficiency virus type 1-specific cytotoxic T lymphocyte responses: a predominant gag-specific response is associated with nonprogressive infection. J Infect Dis 178: 1008–1018.
    1. Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon MA, et al. (1999) Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 283: 857–860.
    1. Sekaly RP (2008) The failed HIV Merck vaccine study: a step back or a launching point for future vaccine development? J Exp Med 205: 7–12.
    1. Shiver JW, Fu TM, Chen L, Casimiro DR, Davies ME, et al. (2002) Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 415: 331–335.
    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, et al. (2009) Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 361: 2209–2220.
    1. Bonsignori M, Pollara J, Moody MA, Alpert MD, Chen X, et al... (2012) ADCC-Mediating Antibodies from an HIV-1 Vaccine Efficacy Trial Target Multiple Epitopes and Preferentially Use the VH1 Gene Family. J Virol.
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, et al. (2012) Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366: 1275–1286.
    1. Anton PA, Ibarrondo FJ, Boscardin WJ, Zhou Y, Schwartz EJ, et al. (2008) Differential immunogenicity of vaccinia and HIV-1 components of a human recombinant vaccine in mucosal and blood compartments. Vaccine 26: 4617–4623.
    1. Pialoux G, Hocini H, Perusat S, Silberman B, Salmon-Ceron D, et al. (2008) Phase I study of a candidate vaccine based on recombinant HIV-1 gp160 (MN/LAI) administered by the mucosal route to HIV-seronegative volunteers: the ANRS VAC14 study. Vaccine 26: 2657–2666.
    1. Berneman A, Belec L, Fischetti VA, Bouvet JP (1998) The specificity patterns of human immunoglobulin G antibodies in serum differ from those in autologous secretions. Infect Immun 66: 4163–4168.
    1. Bomsel M, Tudor D, Drillet AS, Alfsen A, Ganor Y, et al. (2011) Immunization with HIV-1 gp41 subunit virosomes induces mucosal antibodies protecting nonhuman primates against vaginal SHIV challenges. Immunity 34: 269–280.
    1. Plotkin SA (2010) Correlates of protection induced by vaccination. Clin Vaccine Immunol 17: 1055–1065.
    1. Haase AT (2010) Targeting early infection to prevent HIV-1 mucosal transmission. Nature 464: 217–223.
    1. Cerutti A, Chen K, Chorny A (2011) Immunoglobulin responses at the mucosal interface. Annu Rev Immunol 29: 273–293.
    1. Mestecky J, Raska M, Novak J, Alexander RC, Moldoveanu Z (2010) Antibody-mediated protection and the mucosal immune system of the genital tract: relevance to vaccine design. J Reprod Immunol 85: 81–85.
    1. Pavot V, Rochereau N, Genin C, Verrier B, Paul S (2012) New insights in mucosal vaccine development. Vaccine 30: 142–154.
    1. Mascola JR, Stiegler G, VanCott TC, Katinger H, Carpenter CB, et al. (2000) Protection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodies. Nat Med 6: 207–210.
    1. Parren PW, Marx PA, Hessell AJ, Luckay A, Harouse J, et al. (2001) Antibody protects macaques against vaginal challenge with a pathogenic R5 simian/human immunodeficiency virus at serum levels giving complete neutralization in vitro. J Virol 75: 8340–8347.
    1. Poignard P, Moldt B, Maloveste K, Campos N, Olson WC, et al. (2012) Protection against High-Dose Highly Pathogenic Mucosal SIV Challenge at Very Low Serum Neutralizing Titers of the Antibody-Like Molecule CD4-IgG2. PLoS One 7: e42209.
    1. Broliden K, Hinkula J, Devito C, Kiama P, Kimani J, et al. (2001) Functional HIV-1 specific IgA antibodies in HIV-1 exposed, persistently IgG seronegative female sex workers. Immunol Lett 79: 29–36.
    1. Devito C, Broliden K, Kaul R, Svensson L, Johansen K, et al. (2000) Mucosal and plasma IgA from HIV-1-exposed uninfected individuals inhibit HIV-1 transcytosis across human epithelial cells. J Immunol 165: 5170–5176.
    1. Devito C, Hinkula J, Kaul R, Lopalco L, Bwayo JJ, et al. (2000) Mucosal and plasma IgA from HIV-exposed seronegative individuals neutralize a primary HIV-1 isolate. AIDS 14: 1917–1920.
    1. Horton RE, Ball TB, Wachichi C, Jaoko W, Rutherford WJ, et al. (2009) Cervical HIV-specific IgA in a population of commercial sex workers correlates with repeated exposure but not resistance to HIV. AIDS Res Hum Retroviruses 25: 83–92.
    1. Kaul R, Trabattoni D, Bwayo JJ, Arienti D, Zagliani A, et al. (1999) HIV-1-specific mucosal IgA in a cohort of HIV-1-resistant Kenyan sex workers. AIDS 13: 23–29.
    1. Miyazawa M, Lopalco L, Mazzotta F, Lo Caputo S, Veas F, et al. (2009) The 'immunologic advantage' of HIV-exposed seronegative individuals. AIDS 23: 161–175.
    1. Clerici M, Barassi C, Devito C, Pastori C, Piconi S, et al. (2002) Serum IgA of HIV-exposed uninfected individuals inhibit HIV through recognition of a region within the alpha-helix of gp41. AIDS 16: 1731–1741.
    1. Devito C, Hinkula J, Kaul R, Kimani J, Kiama P, et al. (2002) Cross-clade HIV-1-specific neutralizing IgA in mucosal and systemic compartments of HIV-1-exposed, persistently seronegative subjects. J Acquir Immune Defic Syndr 30: 413–420.
    1. Pastori C, Barassi C, Piconi S, Longhi R, Villa ML, et al. (2000) HIV neutralizing IgA in exposed seronegative subjects recognise an epitope within the gp41 coiled-coil pocket. J Biol Regul Homeost Agents 14: 15–21.
    1. Choi RY, Levinson P, Guthrie BL, Lohman-Payne B, Bosire R, et al. (2012) Cervicovaginal HIV-1-neutralizing immunoglobulin A detected among HIV-1-exposed seronegative female partners in HIV-1-discordant couples. AIDS 26: 2155–2163.
    1. Huang J, Ofek G, Laub L, Louder MK, Doria-Rose NA, et al. (2012) Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491: 406–412.
    1. Hessell AJ, Rakasz EG, Tehrani DM, Huber M, Weisgrau KL, et al. (2010) Broadly neutralizing monoclonal antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian-human immunodeficiency virus SHIVBa-L. J Virol 84: 1302–1313.
    1. Guenaga J, Wyatt RT (2012) Structure-guided Alterations of the gp41-directed HIV-1 Broadly Neutralizing Antibody 2F5 Reveal New Properties Regarding its Neutralizing Function. PLoS Pathog 8: e1002806.
    1. Muster T, Steindl F, Purtscher M, Trkola A, Klima A, et al. (1993) A conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1. J Virol 67: 6642–6647.
    1. Stiegler G, Kunert R, Purtscher M, Wolbank S, Voglauer R, et al. (2001) A potent cross-clade neutralizing human monoclonal antibody against a novel epitope on gp41 of human immunodeficiency virus type 1. AIDS Res Hum Retroviruses 17: 1757–1765.
    1. Zwick MB, Jensen R, Church S, Wang M, Stiegler G, et al. (2005) Anti-human immunodeficiency virus type 1 (HIV-1) antibodies 2F5 and 4E10 require surprisingly few crucial residues in the membrane-proximal external region of glycoprotein gp41 to neutralize HIV-1. J Virol 79: 1252–1261.
    1. Zwick MB, Labrijn AF, Wang M, Spenlehauer C, Saphire EO, et al. (2001) Broadly neutralizing antibodies targeted to the membrane-proximal external region of human immunodeficiency virus type 1 glycoprotein gp41. J Virol 75: 10892–10905.
    1. Tudor D, Derrien M, Diomede L, Drillet AS, Houimel M, et al. (2009) HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis. Mucosal Immunol 2: 412–426.
    1. Gallo SA, Finnegan CM, Viard M, Raviv Y, Dimitrov A, et al. (2003) The HIV Env-mediated fusion reaction. Biochim Biophys Acta 1614: 36–50.
    1. Alfsen A, Bomsel M (2002) HIV-1 gp41 envelope residues 650–685 exposed on native virus act as a lectin to bind epithelial cell galactosyl ceramide. J Biol Chem 277: 25649–25659.
    1. Moser C, Amacker M, Kammer AR, Rasi S, Westerfeld N, et al. (2007) Influenza virosomes as a combined vaccine carrier and adjuvant system for prophylactic and therapeutic immunizations. Expert Rev Vaccines 6: 711–721.
    1. Moser C, Amacker M, Zurbriggen R (2011) Influenza virosomes as a vaccine adjuvant and carrier system. Expert Rev Vaccines 10: 437–446.
    1. Wilschut J (2009) Influenza vaccines: the virosome concept. Immunol Lett 122: 118–121.
    1. Coutant J, Yu H, Clement MJ, Alfsen A, Toma F, et al. (2008) Both lipid environment and pH are critical for determining physiological solution structure of 3-D-conserved epitopes of the HIV-1 gp41-MPER peptide P1. FASEB J 22: 4338–4351.
    1. Tamura M, Webster RG, Ennis FA (1991) Antibodies to HA and NA augment uptake of influenza A viruses into cells via Fc receptor entry. Virology 182: 211–219.
    1. Tamura M, Webster RG, Ennis FA (1994) Subtype cross-reactive, infection-enhancing antibody responses to influenza A viruses. J Virol 68: 3499–3504.
    1. Herzog C, Hartmann K, Kunzi V, Kursteiner O, Mischler R, et al. (2009) Eleven years of Inflexal V-a virosomal adjuvanted influenza vaccine. Vaccine 27: 4381–4387.
    1. Zuccotti G, Amendola A, Vigano A, Pariani E, Zappa A, et al. (2007) Long-term immunogenicity of a virosomal subunit inactivated influenza vaccine in children with asthma. Vaccine 25: 6692–6698.
    1. Anderson DJ, Politch JA, Nadolski AM, Blaskewicz CD, Pudney J, et al. (2010) Targeting Trojan Horse leukocytes for HIV prevention. AIDS 24: 163–187.
    1. UNAIDS (2012) Global report: UNAIDS report on the global AIDS epidemic. 1–110.
    1. Mischler R, Metcalfe IC (2002) Inflexal V a trivalent virosome subunit influenza vaccine: production. Vaccine 20 Suppl 5B17–23.
    1. Hildesheim A, McShane LM, Schiffman M, Bratti MC, Rodriguez AC, et al. (1999) Cytokine and immunoglobulin concentrations in cervical secretions: reproducibility of the Weck-cel collection instrument and correlates of immune measures. J Immunol Methods 225: 131–143.
    1. Kozlowski PA, Lynch RM, Patterson RR, Cu-Uvin S, Flanigan TP, et al. (2000) Modified wick method using Weck-Cel sponges for collection of human rectal secretions and analysis of mucosal HIV antibody. J Acquir Immune Defic Syndr 24: 297–309.
    1. Adler M, Wacker R, Niemeyer CM (2003) A real-time immuno-PCR assay for routine ultrasensitive quantification of proteins. Biochem Biophys Res Commun 308: 240–250.
    1. Adler M, Wacker R, Niemeyer CM (2008) Sensitivity by combination: immuno-PCR and related technologies. Analyst 133: 702–718.
    1. Niemeyer CM, Adler M, Wacker R (2007) Detecting antigens by quantitative immuno-PCR. Nat Protoc 2: 1918–1930.
    1. Sano T, Smith CL, Cantor CR (1992) Immuno-PCR: very sensitive antigen detection by means of specific antibody-DNA conjugates. Science 258: 120–122.
    1. Frey A, Di Canzio J, Zurakowski D (1998) A statistically defined endpoint titer determination method for immunoassays. J Immunol Methods 221: 35–41.
    1. Maecker HT (2009) Multiparameter flow cytometry monitoring of T cell responses. Methods Mol Biol 485: 375–391.
    1. Donadoni C, Bisighini C, Scotti L, Diomede L, Ngyen M, et al. (2010) Setting of methods for analysis of mucosal antibodies in seminal and vaginal fluids of HIV seropositive subjects from Cambodian and Italian cohorts. PLoS One 5: e9920.
    1. Mestecky J, Wright PF, Lopalco L, Staats HF, Kozlowski PA, et al. (2011) Scarcity or absence of humoral immune responses in the plasma and cervicovaginal lavage fluids of heavily HIV-1-exposed but persistently seronegative women. AIDS Res Hum Retroviruses 27: 469–486.
    1. Diomede L, Nyoka S, Pastori C, Scotti L, Zambon A, et al... (2012) Passively transmitted gp41 antibodies in babies born from subtype C HIV-1 seropositive women: correlation between fine specificity and protection. J Virol.
    1. Li M, Gao F, Mascola JR, Stamatatos L, Polonis VR, et al. (2005) Human immunodeficiency virus type 1 env clones from acute and early subtype B infections for standardized assessments of vaccine-elicited neutralizing antibodies. J Virol 79: 10108–10125.
    1. Alfsen A, Iniguez P, Bouguyon E, Bomsel M (2001) Secretory IgA specific for a conserved epitope on gp41 envelope glycoprotein inhibits epithelial transcytosis of HIV-1. J Immunol 166: 6257–6265.
    1. Hocini H, Belec L, Iscaki S, Garin B, Pillot J, et al. (1997) High-level ability of secretory IgA to block HIV type 1 transcytosis: contrasting secretory IgA and IgG responses to glycoprotein 160. AIDS Res Hum Retroviruses 13: 1179–1185.
    1. Hocini H, Bomsel M (1999) Infectious human immunodeficiency virus can rapidly penetrate a tight human epithelial barrier by transcytosis in a process impaired by mucosal immunoglobulins. J Infect Dis 179 Suppl 3S448–453.
    1. Johansson EL, Wassen L, Holmgren J, Jertborn M, Rudin A (2001) Nasal and vaginal vaccinations have differential effects on antibody responses in vaginal and cervical secretions in humans. Infect Immun 69: 7481–7486.
    1. Kozlowski PA, Cu-Uvin S, Neutra MR, Flanigan TP (1997) Comparison of the oral, rectal, and vaginal immunization routes for induction of antibodies in rectal and genital tract secretions of women. Infect Immun 65: 1387–1394.
    1. Kozlowski PA, Williams SB, Lynch RM, Flanigan TP, Patterson RR, et al. (2002) Differential induction of mucosal and systemic antibody responses in women after nasal, rectal, or vaginal immunization: influence of the menstrual cycle. J Immunol 169: 566–574.
    1. Lewis DJ, Fraser CA, Mahmoud AN, Wiggins RC, Woodrow M, et al. (2011) Phase I randomised clinical trial of an HIV-1(CN54), clade C, trimeric envelope vaccine candidate delivered vaginally. PLoS One 6: e25165.
    1. Ansari AA (2004) Autoimmunity, anergy, lentiviral immunity and disease. Autoimmun Rev 3: 530–540.
    1. Haynes BF, Fleming J, St Clair EW, Katinger H, Stiegler G, et al. (2005) Cardiolipin polyspecific autoreactivity in two broadly neutralizing HIV-1 antibodies. Science 308: 1906–1908.
    1. Silvestris F, Williams RC Jr, Dammacco F (1995) Autoreactivity in HIV-1 infection: the role of molecular mimicry. Clin Immunol Immunopathol 75: 197–205.
    1. Chen K, Cerutti A (2010) Vaccination strategies to promote mucosal antibody responses. Immunity 33: 479–491.
    1. Kunkel EJ, Butcher EC (2003) Plasma-cell homing. Nat Rev Immunol 3: 822–829.
    1. Kunkel EJ, Kim CH, Lazarus NH, Vierra MA, Soler D, et al. (2003) CCR10 expression is a common feature of circulating and mucosal epithelial tissue IgA Ab-secreting cells. J Clin Invest 111: 1001–1010.
    1. Tudor D, Yu H, Maupetit J, Drillet AS, Bouceba T, et al... (2012) Isotype modulates epitope specificity, affinity, and antiviral activities of anti-HIV-1 human broadly neutralizing 2F5 antibody. Proc Natl Acad Sci U S A.
    1. Anderson DJ, Politch JA, Tucker LD, Fichorova R, Haimovici F, et al. (1998) Quantitation of mediators of inflammation and immunity in genital tract secretions and their relevance to HIV type 1 transmission. AIDS Res Hum Retroviruses 14 Suppl 1S43–49.
    1. Pilcher CD, Joaki G, Hoffman IF, Martinson FE, Mapanje C, et al. (2007) Amplified transmission of HIV-1: comparison of HIV-1 concentrations in semen and blood during acute and chronic infection. AIDS 21: 1723–1730.
    1. Anderson D, Politch JA, Pudney J (2011) HIV infection and immune defense of the penis. Am J Reprod Immunol 65: 220–229.
    1. Pudney J, Anderson D (2011) Innate and acquired immunity in the human penile urethra. J Reprod Immunol 88: 219–227.
    1. Rothaeusler K, Ma ZM, Qureshi H, Carroll TD, Rourke T, et al. (2012) Antiviral antibodies and T cells are present in the foreskin of simian immunodeficiency virus-infected rhesus macaques. J Virol 86: 7098–7106.
    1. Peduzzi E, Westerfeld N, Zurbriggen R, Pluschke G, Daubenberger CA (2008) Contribution of influenza immunity and virosomal-formulated synthetic peptide to cellular immune responses in a phase I subunit malaria vaccine trial. Clin Immunol 127: 188–197.
    1. Schroeder HW Jr, Cavacini L (2010) Structure and function of immunoglobulins. J Allergy Clin Immunol 125: S41–52.
    1. Yu Q, Yu R, Qin X (2010) The good and evil of complement activation in HIV-1 infection. Cell Mol Immunol 7: 334–340.
    1. Ackerman ME, Moldt B, Wyatt RT, Dugast AS, McAndrew E, et al. (2011) A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples. J Immunol Methods 366: 8–19.
    1. Dugast AS, Tonelli A, Berger CT, Ackerman ME, Sciaranghella G, et al. (2011) Decreased Fc receptor expression on innate immune cells is associated with impaired antibody-mediated cellular phagocytic activity in chronically HIV-1 infected individuals. Virology 415: 160–167.
    1. Bakema JE, van Egmond M (2011) The human immunoglobulin A Fc receptor FcalphaRI: a multifaceted regulator of mucosal immunity. Mucosal Immunol 4: 612–624.
    1. Duval M, Posner MR, Cavacini LA (2008) A bispecific antibody composed of a nonneutralizing antibody to the gp41 immunodominant region and an anti-CD89 antibody directs broad human immunodeficiency virus destruction by neutrophils. J Virol 82: 4671–4674.
    1. Holl V, Peressin M, Decoville T, Schmidt S, Zolla-Pazner S, et al. (2006) Nonneutralizing antibodies are able to inhibit human immunodeficiency virus type 1 replication in macrophages and immature dendritic cells. J Virol 80: 6177–6181.
    1. Otten MA, Rudolph E, Dechant M, Tuk CW, Reijmers RM, et al. (2005) Immature neutrophils mediate tumor cell killing via IgA but not IgG Fc receptors. J Immunol 174: 5472–5480.
    1. van Spriel AB, van den Herik-Oudijk IE, van Sorge NM, Vile HA, van Strijp JA, et al. (1999) Effective phagocytosis and killing of Candida albicans via targeting FcgammaRI (CD64) or FcalphaRI (CD89) on neutrophils. J Infect Dis 179: 661–669.
    1. Mabuka J, Nduati R, Odem-Davis K, Peterson D, Overbaugh J (2012) HIV-Specific Antibodies Capable of ADCC Are Common in Breastmilk and Are Associated with Reduced Risk of Transmission in Women with High Viral Loads. PLoS Pathog 8: e1002739.

Source: PubMed

3
購読する