72 results on '"Lisa A. Cavacini"'
Search Results
2. Formulation Studies to Develop Low-Cost, Orally-Delivered Secretory IgA Monoclonal Antibodies for Passive Immunization Against Enterotoxigenic Escherichia coli
- Author
-
Sakshi Bajoria, Lorena R. Antunez, Ozan S. Kumru, Mark Klempner, Yang Wang, Lisa A. Cavacini, Sangeeta B. Joshi, and David B. Volkin
- Subjects
Pharmaceutical Science - Published
- 2023
- Full Text
- View/download PDF
3. Mucosal nanobody IgA as inhalable and affordable prophylactic and therapeutic treatment against SARS-CoV-2 and emerging variants
- Author
-
Qi Li, Fiachra Humphries, Roxie C. Girardin, Aaron Wallace, Monir Ejemel, Alla Amcheslavsky, Conor T. McMahon, Zachary A. Schiller, Zepei Ma, John Cruz, Alan P. Dupuis, Anne F. Payne, Arooma Maryam, Nese Kurt Yilmaz, Kathleen A. McDonough, Brian G. Pierce, Celia A. Schiffer, Andrew C. Kruse, Mark S. Klempner, Lisa A. Cavacini, Katherine A. Fitzgerald, and Yang Wang
- Subjects
SARS-CoV-2 ,Immunology ,COVID-19 ,Single-Domain Antibodies ,Antibodies, Viral ,Immunoglobulin A ,Epitopes ,Mice ,Immunoglobulin G ,Spike Glycoprotein, Coronavirus ,Immunology and Allergy ,Animals ,Humans ,Angiotensin-Converting Enzyme 2 - Abstract
Anti-COVID antibody therapeutics have been developed but not widely used due to their high cost and escape of neutralization from the emerging variants. Here, we describe the development of VHH-IgA1.1, a nanobody IgA fusion molecule as an inhalable, affordable and less invasive prophylactic and therapeutic treatment against SARS-CoV-2 Omicron variants. VHH-IgA1.1 recognizes a conserved epitope of SARS-CoV-2 spike protein Receptor Binding Domain (RBD) and potently neutralizes major global SARS-CoV-2 variants of concern (VOC) including the Omicron variant and its sub lineages BA.1.1, BA.2 and BA.2.12.1. VHH-IgA1.1 is also much more potent against Omicron variants as compared to an IgG Fc fusion construct, demonstrating the importance of IgA mediated mucosal protection for Omicron infection. Intranasal administration of VHH-IgA1.1 prior to or after challenge conferred significant protection from severe respiratory disease in K18-ACE2 transgenic mice infected with SARS-CoV-2 VOC. More importantly, for cost-effective production, VHH-IgA1.1 produced in Pichia pastoris had comparable potency to mammalian produced antibodies. Our study demonstrates that intranasal administration of affordably produced VHH-IgA fusion protein provides effective mucosal immunity against infection of SARS-CoV-2 including emerging variants.
- Published
- 2022
4. Oral administration of an anti-CfaE secretory IgA antibody protects against Enterotoxigenic Escherichia coli diarrheal disease in a nonhuman primate model
- Author
-
Mark S. Klempner, Gladys Nunez, James Regeimbal, Yang Wang, Carlos Gaspar, Zachary A. Schiller, Lisa A. Cavacini, Melissa A. Gawron, Matteo Stoppato, Matthew I. Schneider, Serena Giuntini, Joseph C. Martin, and Jessica R. Pondish
- Subjects
Diarrhea ,030231 tropical medicine ,Attack rate ,Administration, Oral ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Oral administration ,Immunity ,Enterotoxigenic Escherichia coli ,Animals ,Medicine ,030212 general & internal medicine ,Aotus nancymaae ,Escherichia coli Infections ,Colony-forming unit ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,Antibodies, Bacterial ,Disease Models, Animal ,Infectious Diseases ,Immunoglobulin A, Secretory ,Immunology ,biology.protein ,Aotidae ,Molecular Medicine ,Antibody ,medicine.symptom ,business - Abstract
ABSTACTEnterotoxigenicEscherichia coli(ETEC) is a leading cause of diarrhea-associated illness in developing countries. There is currently no vaccine licensed to prevent ETEC and the development of an efficacious prophylaxis would provide an intervention with significant impact. Recent studies suggested that effective protection could be achieved by inducing immunity to block colonization of ETEC. Here, we evaluated the efficacy of secretory (s) IgA2 and dimeric (d) IgA2 of an anti-colonization factor antigen antibody, 68-61, in theAotus nancymaaenon-human primate (NHP) ETEC challenge model via oral and parental delivery. Thirty-nine animals were distributed across 3 groups of 13, and challenged with 5.0×1011cfu of H10407 on Day 0. Group 1 received a dIgA2 68-61 subcutaneously on day 0. Group 2 received a SIgA2 68-61 orally on days −1, 0, and +1, and Group 3 received an irrelevant SIgA2 antibody orally on days −1, 0, and +1. All animals were observed for symptoms of diarrhea, and stools were collected for ETEC colony counts. SIgA2 treatment significantly lowered the attack rate, resulting in a protective efficacy of 71.4% (p=0.025) in Group 2 as compared to Group 3. Anti-CfaE dIgA2 treatment group reduced the diarrheal attack rate, although the reduction did not reach significance (57.1%; P=0.072) as compared to the irrelevant SIgA2 Group 3. Our results demonstrated the feasibility of oral administration of SIgA as a potential immunoprophylaxis against enteric infections. To our knowledge, this is the first study to demonstrate the efficacy of administrated SIgA in a non-human primate model.
- Published
- 2020
- Full Text
- View/download PDF
5. Preformulation Characterization and Stability Assessments of Secretory IgA Monoclonal Antibodies as Potential Candidates for Passive Immunization by Oral Administration
- Author
-
Jian Xiong, Lorena R. Antunez, Mark S. Klempner, Ozan S. Kumru, Sangeeta B. Joshi, Yue Hu, John M. Hickey, Yang Wang, David B. Volkin, and Lisa A. Cavacini
- Subjects
enterotoxigenic Escherichia coli ,Administration, Oral ,Pharmaceutical Science ,02 engineering and technology ,Enterotoxin ,ETEC, enterotoxigenic Escherichia coli ,medicine.disease_cause ,030226 pharmacology & pharmacy ,MW, molecular weight ,PTM, post translational modifications ,Immunoglobulin G ,immunoglobulin G ,0302 clinical medicine ,Drug Stability ,Pepsin ,physicochemical characterization ,Oral administration ,Enterotoxigenic Escherichia coli ,Escherichia coli Infections ,SDS-PAGE, sodium dodecyl sulfate–polyacrylamide gel electrophoresis ,biology ,Escherichia coli Vaccines ,Chemistry ,Escherichia coli Proteins ,Antibodies, Monoclonal ,ELISA, enzyme-linked immunosorbent assay ,021001 nanoscience & nanotechnology ,oral delivery ,GdnHCl, guanidine hydrochloride ,LT, heat labile enterotoxin ,SV-AUC, sedimentation velocity analytical ultracentrifugation ,secretory immunoglobulin A ,0210 nano-technology ,Diarrhea ,Glycan ,medicine.drug_class ,Drug Compounding ,IgG, immunoglobulin G ,formulation ,CHO Cells ,LC, light chain ,immunization ,Monoclonal antibody ,Article ,Fc, crystallizable fragment ,Microbiology ,03 medical and health sciences ,Cricetulus ,medicine ,Animals ,mAb, monoclonal antibody ,SE-HPLC, size exclusion high performance liquid chromatography ,SGF, simulated gastric fluid ,PEG, polyethylene glycol ,Fab, antigen binding fragment ,Immunization, Passive ,HC, heavy chain ,stability ,Immunization ,Immunoglobulin A, Secretory ,biology.protein ,SC, secretory component ,sIgA, secretory immunoglobulin A - Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrheal disease among children in developing countries, and there are no licensed vaccines to protect against ETEC. Passive immunization by oral delivery of ETEC-specific secretory IgAs (sIgAs) could potentially provide an alternative approach for protection in targeted populations. In this study, a series of physiochemical techniques and an in vitro gastric digestion model were used to characterize and compare key structural attributes and stability profiles of 3 anti–heat-labile enterotoxin mAbs (sIgA1, sIgA2, and IgG1 produced in CHO cells). The mAbs were evaluated in terms of primary structure, N-linked glycan profiles, size and aggregate content, relative apparent solubility, conformational stability, and in vitro antigen binding. Compared to IgG1 mAb, sIgA1 and sIgA2 mAbs showed increased sample heterogeneity, especially in terms of N-glycan composition and the presence of higher molecular weight species. The sIgA mAbs showed overall better physical stability and were more resistant to loss of antigen binding activity during incubation at low pH, 37°C with pepsin. These results are discussed in terms of future challenges to design stable, low-cost formulations of sIgA mAbs as an oral supplement for passive immunization to protect against enteric diseases in the developing world.
- Published
- 2020
- Full Text
- View/download PDF
6. Investigation of a monoclonal antibody against enterotoxigenic Escherichia coli, expressed as secretory IgA1 and IgA2 in plants
- Author
-
Ozan S. Kumru, Jian Xiong, Richard L. Guerrant, Mark S. Klempner, Clemens Grünwald-Gruber, Yang Wang, Yue Hu, Lisa A. Cavacini, David B. Volkin, David T. Bolick, Sangeeta B. Joshi, Julian K.-C. Ma, Friedrich Altmann, and Audrey Y.-H. Teh
- Subjects
0301 basic medicine ,Microbiology (medical) ,Glycosylation ,Secretory component ,medicine.drug_class ,RC799-869 ,medicine.disease_cause ,Monoclonal antibody ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,N-linked glycosylation ,nicotiana benthamiana ,Enterotoxigenic Escherichia coli ,medicine ,Hemagglutination assay ,biology ,enterotoxigenic escherichia coli ,Gastroenterology ,food and beverages ,Diseases of the digestive system. Gastroenterology ,J chain ,030104 developmental biology ,Infectious Diseases ,secretory iga ,chemistry ,monoclonal antibody ,biology.protein ,passive immunization ,030211 gastroenterology & hepatology ,immunotherapy ,Antibody - Abstract
Passive immunization with antibodies is a promising approach against enterotoxigenic Escherichia coli diarrhea, a prevalent disease in LMICs. The objective of this study was to investigate expression of a monoclonal anti-ETEC CfaE secretory IgA antibody in N. benthamiana plants, with a view to facilitating access to ETEC passive immunotherapy. SIgA1 and SIgA2 forms of mAb 68-81 were produced by co-expressing the light and engineered heavy chains with J chain and secretory component in N. benthamiana. Antibody expression and assembly were compared with CHO-derived antibodies by SDS-PAGE, western blotting, size-exclusion chromatography and LC-MS peptide mapping. N-linked glycosylation was assessed by rapid fluorescence/mass spectrometry and LC-ESI-MS. Susceptibility to gastric digestion was assessed in an in vitro model. Antibody function was compared for antigen binding, a Caco-2 cell-based ETEC adhesion assay, an ETEC hemagglutination inhibition assay and a murine in vivo challenge study. SIgA1 assembly appeared superior to SIgA2 in plants. Both sub-classes exhibited resistance to degradation by simulated gastric fluid, comparable to CHO-produced 68-61 SIgA1. The plant expressed SIgAs had more homogeneous N-glycosylation than CHO-derived SIgAs, but no alteration of in vitro functional activity was observed, including antibodies expressed in a plant line engineered for mammalian-like N glycosylation. The plant-derived SIgA2 mAb demonstrated protection against diarrhea in a murine infection model. Although antibody yield and purification need to be optimized, anti-ETEC SIgA antibodies produced in a low-cost plant platform are functionally equivalent to CHO antibodies, and provide promise for passive immunotherapy in LMICs.
- Published
- 2021
- Full Text
- View/download PDF
7. Oral administration of a single anti-CfaE nanobody provides broadly cross-protective immunity against major pathogenic Enterotoxigenic Escherichia coli strains
- Author
-
Jessica R. Pondish, Ryan Schneider, Qi Li, Mark S. Klempner, Monir Ejemel, Jordan Meisinger, Eileen M. Barry, Yang Wang, Andrew C. Kruse, Raimond Heukers, Zachary A. Schiller, Brian G. Pierce, Conor McMahon, Matteo Stoppato, Lisa A. Cavacini, Jacqueline R. Toomey, Aaron Wallace, Serena Giuntini, and Alla Amcheslavsky
- Subjects
Bacterial adhesin ,Hemagglutination ,Infectious disease (medical specialty) ,medicine.medical_treatment ,Enterotoxigenic Escherichia coli ,medicine ,Outbreak ,Immunotherapy ,Yeast display ,Biology ,medicine.disease_cause ,Epitope ,Microbiology - Abstract
Enterotoxigenic Escherichia coli (ETEC) is estimated to cause approximately 380,000 deaths annually during sporadic or epidemic outbreaks worldwide. There is currently no vaccine licensed to prevent ETEC. Development of prophylaxis against ETEC is challenging due to the vast heterogeneity of the ETEC strains. The discovery of nanobodies has emerged as a successful new biologics in treating mucosal infectious disease as nanobodies can recognize conserved epitopes on hypervariable pathogens. In this study, we performed large screens using immunized llamas and a naïve nanobody yeast display library against adhesins of colonization factors. Cross-protective nanobodies were selected with in vitro activities inhibiting mannose-resistant hemagglutination (MRHA) against all eleven major pathogenic ETEC strains. Oral administration of nanobodies led to significant reduction of bacterial colonization in animals challenged with multiple ETEC strains. Structural analysis revealed novel conserved epitopes as critical structural features for pan-ETEC vaccine design.Two of the lead nanobodies, 2R215 and 1D7, were further engineered as trimer or fused with human IgA Fc-fragments as fusionbodies. Oral administration of the trimers or fusionbodies protected mice from infection at a much lower dose compared to the monomeric format. Importantly, fusionbodies prevented infection as a pre-treatment when administrated 2 hours before ETEC challenge to the animals. Together, our study provides the first proof of concept that oral administration of a single nanobody could confer broad protection against major pathogenic ETEC strains. Technological advances in large-scale manufacturing of biological proteins in plants and microorganisms will make nanobody-based immunotherapy a potent and cost-effective prophylaxis or treatment for ETEC.
- Published
- 2020
- Full Text
- View/download PDF
8. IgA MAb blocks SARS-CoV-2 Spike-ACE2 interaction providing mucosal immunity
- Author
-
Monir Ejemel, Qi Li, Shurong Hou, Zachary A. Schiller, Aaron L. Wallace, Alla Amcheslavsky, Nese Kurt Yilmaz, Jacqueline R. Toomey, Ryan Schneider, Brianna J. Close, Da-Yuan Chen, Hasahn L. Conway, Saeed Mohsan, Lisa A. Cavacini, Mark S. Klempner, Celia A. Schiffer, and Yang Wang
- Subjects
Coronavirus disease 2019 (COVID-19) ,medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,fungi ,virus diseases ,Biology ,biochemical phenomena, metabolism, and nutrition ,Monoclonal antibody ,Virology ,Article ,body regions ,Viral infection ,medicine ,biology.protein ,Mucosal immunology ,Spike (database) ,Antibody therapy ,Molecular modelling ,Antibody ,skin and connective tissue diseases ,Mucosal immunity - Abstract
COVID-19 caused by SARS-CoV-2 has become a global pandemic requiring the development of interventions for the prevention or treatment to curtail mortality and morbidity. No vaccine to boost mucosal immunity, or as a therapeutic, has yet been developed to SARS-CoV-2. In this study, we discover and characterize a cross-reactive human IgA monoclonal antibody, MAb362. MAb362 binds to both SARS-CoV and SARS-CoV-2 spike proteins and competitively blocks ACE2 receptor binding, by overlapping the ACE2 structural binding epitope. Furthermore, MAb362 IgA neutralizes both pseudotyped SARS-CoV and SARS-CoV-2 in 293 cells expressing ACE2. When converted to secretory IgA, MAb326 also neutralizes authentic SARS-CoV-2 virus while the IgG isotype shows no neutralization. Our results suggest that SARS-CoV-2 specific IgA antibodies, such as MAb362, may provide effective immunity against SARS-CoV-2 by inducing mucosal immunity within the respiratory system, a potentially critical feature of an effective vaccine., Here, Ejemel et al. report the identification and characterization of a cross-neutralizing human IgA monoclonal antibody, named MAb362, that binds the receptor-binding domain of SARS-CoV-2 Spike, blocking its interaction with the ACE2 host receptor.
- Published
- 2020
9. Transgenic goats producing an improved version of cetuximab in milk
- Author
-
Lisa A. Cavacini, Nathalie Fournier, Götz Laible, Paul S. MacLean, Harry M. Meade, Li How Chen, Dan Pollock, Brigid Brophy, Nicholas C. Masiello, Sally Cole, David N. Wells, Christophe De Romeuf, and William G. Gavin
- Subjects
Cancer Research ,Physiology ,medicine.drug_class ,EGFR ,Transgene ,Pharmacology ,CD16 ,Biology ,Monoclonal antibody ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Epitope ,cetuximab ,medicine ,biobetter ,Epidermal growth factor receptor ,Cytotoxicity ,lcsh:QH301-705.5 ,Research Articles ,Brand names ,Cetuximab ,Virology ,lcsh:Biology (General) ,monoclonal antibody ,biology.protein ,Molecular Medicine ,biosimilar ,Research Article ,medicine.drug - Abstract
Therapeutic monoclonal antibodies (mAbs) represent one of the most important classes of pharmaceutical proteins to treat human diseases. Most are produced in cultured mammalian cells which is expensive, limiting their availability. Goats, striking a good balance between a relatively short generation time and copious milk yield, present an alternative platform for the cost-effective, flexible, large-scale production of therapeutic mAbs. Here, we focused on cetuximab, a mAb against epidermal growth factor receptor, that is commercially produced under the brand name Erbitux and approved for anti-cancer treatments. We generated several transgenic goat lines that produce cetuximab in their milk. Two lines were selected for detailed characterization. Both showed stable genotypes and cetuximab production levels of up to 10g/L. The mAb could be readily purified and showed improved characteristics compared to Erbitux. The goat-produced cetuximab (gCetuximab) lacked a highly immunogenic epitope that is part of Erbitux. Moreover, it showed enhanced binding to CD16 and increased antibody-dependent cell-dependent cytotoxicity compared to Erbitux. This indicates that these goats produce an improved cetuximab version with the potential for enhanced effectiveness and better safety profile compared to treatments with Erbitux. In addition, our study validates transgenic goats as an excellent platform for large-scale production of therapeutic mAbs.
- Published
- 2020
- Full Text
- View/download PDF
10. Human Anti–HIV-1 gp120 Monoclonal Antibodies with Neutralizing Activity Cloned from Humanized Mice Infected with HIV-1
- Author
-
Joseph C. Martin, Ann Dauphin, Dale L. Greiner, Michael A. Brehm, Leonard D. Shultz, Lisa A. Cavacini, Aaron Wallace, Claudia Carbone, Mark Duval, Melissa A. Gawron, Jeremy Luban, and Smita Jaiswal
- Subjects
Anti hiv 1 ,medicine.drug_class ,Immunology ,HIV Infections ,Mice, SCID ,HIV Antibodies ,HIV Envelope Protein gp120 ,Biology ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Article ,Animals, Genetically Modified ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Germline mutation ,Mice, Inbred NOD ,Neutralization Tests ,medicine ,Animals ,Humans ,Immunology and Allergy ,Gene family ,virus diseases ,Antibodies, Neutralizing ,Virology ,Genetically modified organism ,Monoclonal ,HIV-1 ,biology.protein ,Antibody ,030215 immunology - Abstract
Broadly neutralizing, anti-HIV-1 gp120 monoclonal antibodies have been isolated from infected individuals, and there is considerable interest in developing these reagents for antibody-based immunoprophylaxis and treatment. As a means to identify potentially new anti-HIV antibodies, we exploited humanized NOD-scid IL2rg(null) (NSG) mice systemically infected with HIV-1, to generate a wide variety of antigen-specific human monoclonal antibodies. The antibodies were encoded by a diverse range of variable gene families and immunoglobulin classes, including IgA, and several showed significant levels of somatic mutation. Moreover, the isolated antibodies not only bound target antigens with similar affinity as broadly neutralizing antibodies, they also demonstrated neutralizing ability against multiple HIV-1 clades. The use of humanized mice will allow us to utilize our knowledge of HIV-1 gp120 structure and function, and the immune response targeting this protein, to generate native human prophylactic antibodies to reduce the infection and spread of HIV-1.
- Published
- 2019
- Full Text
- View/download PDF
11. Human genital antibody-mediated inhibition of Chlamydia trachomatis infection and evidence for ompA genotype-specific neutralization
- Author
-
Lisa A. Cavacini, Caitlyn E. L. Bagnetto, Pamela A. Kozlowski, Hannah L. Albritton, Rebecca A. Lillis, Alison J. Quayle, Li Shen, and Caleb M. Ardizzone
- Subjects
Physiology ,Cell Membranes ,Chlamydia trachomatis ,Cervix Uteri ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Biochemistry ,Neutralization ,Chlamydia Infection ,Medical Conditions ,Immune Physiology ,Genotype ,Medicine and Health Sciences ,Chlamydia ,Enzyme-Linked Immunoassays ,Phylogeny ,Immune System Proteins ,Multidisciplinary ,Antibodies, Bacterial ,Bacterial Pathogens ,Infectious Diseases ,Medical Microbiology ,Medicine ,Female ,Pathogens ,Cellular Structures and Organelles ,Antibody ,Bacterial outer membrane ,Bacterial Outer Membrane Proteins ,Research Article ,Adult ,Science ,Immunology ,Sexually Transmitted Diseases ,Biology ,Research and Analysis Methods ,Microbiology ,Antibodies ,Cell Line ,Young Adult ,Antibody Repertoire ,Antibody Isotypes ,medicine ,Humans ,Animal Models of Disease ,Immunoassays ,Microbial Pathogens ,Genotyping ,Secretion ,Bacteria ,Organisms ,Biology and Life Sciences ,Proteins ,Membrane Proteins ,Epithelial Cells ,Sequence Analysis, DNA ,Cell Biology ,Chlamydia Infections ,Outer Membrane Proteins ,Antibodies, Neutralizing ,In vitro ,Immunoglobulin A ,Animal Models of Infection ,Immunoglobulin G ,Immunologic Techniques ,Animal Studies ,biology.protein ,Physiological Processes - Abstract
The endocervix, the primary site ofChlamydia trachomatis(Ct) infection in women, has a unique repertoire of locally synthesized IgG and secretory IgA (SIgA) with contributions from serum IgG. Here, we assessed the ability of genital and serum-derived IgG and IgA from women with a recent positive Ct test to neutralize Ct elementary bodies (EBs) and inhibit inclusion formationin vitroin human endocervical epithelial cells. We also determined if neutralization was influenced by the major outer membrane protein (MOMP) of the infecting strain, as indicated byompAgene sequencing and genotyping. At equivalent low concentrations of Ct EB (D/UW-3/Cx + E/UW-5/Cx)-specific antibody, genital-derived IgG and IgA and serum IgA, but not serum IgG, significantly inhibited inclusion formation, with genital IgA being most effective, followed by genital IgG, then serum IgA. The well-characterized Ct genotype D strain, D/UW-3/Cx, was neutralized by serum-derived IgG from patients infected with genotype D strains, genital IgG from patients infected with genotype D or E strains, and by genital IgA from patients infected with genotype D, E, or F strains. Additionally, inhibition of D/UW-3/Cx infection by whole serum, rather than purified immunoglobulin, was associated with levels of serum EB-specific IgG rather than the genotype of infecting strain. In contrast, a Ct genotype Ia clinical isolate, Ia/LSU-56/Cx, was neutralized by whole serum in a genotype and genogroup-specific manner, and inhibition also correlated with EB-specific IgG concentrations in serum. Taken together, these data suggest that (i) genital IgA most effectively inhibits Ct infectionin vitro, (ii) human antibody-mediated inhibition of Ct infection is significantly influenced by theompAgenotype of the infecting strain, (iii) the genital antibody repertoire develops or matures differently compared to systemic antibody, and (iv)ompAgenotype-specificity of inhibition of infection by whole serum can be overcome by high concentrations of Ct-specific IgG.
- Published
- 2021
- Full Text
- View/download PDF
12. Isolation of potent neutralizing antibodies from a survivor of the 2014 Ebola virus outbreak
- Author
-
Joshua D. Shamblin, Wen Li, Kathleen B. J. Pommert, Mark S. Klempner, Ashley E. Piper, Thomas R. Sprague, Pamela J. Glass, Arthur J. Goff, Colby A. Souders, Keith A. Reimann, Andrew B. Ward, Laura M. Walker, Karl Dane Wittrup, Erica Ollmann Saphire, Dennis R. Burton, Heidi L. Smith, Charles D. Murin, Eric Krauland, Tillman U. Gerngross, Suzanne E. Wollen, Lisa A. Cavacini, Devin Sok, Hannah L. Turner, Marnie L. Fusco, and Zachary A. Bornholdt
- Subjects
0301 basic medicine ,medicine.drug_class ,Antigen-Antibody Complex ,Antibodies, Viral ,medicine.disease_cause ,Monoclonal antibody ,Article ,Disease Outbreaks ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Viral Envelope Proteins ,medicine ,Animals ,Humans ,Survivors ,Ebola Vaccines ,Ebolavirus ,Multidisciplinary ,Ebola virus ,Ebola vaccine ,biology ,Immunization, Passive ,Virion ,Antibodies, Monoclonal ,Hemorrhagic Fever, Ebola ,Viral membrane ,Antibodies, Neutralizing ,Virology ,Tissue Donors ,Bundibugyo virus ,030104 developmental biology ,Immunization ,Antibody Formation ,Democratic Republic of the Congo ,biology.protein ,Antibody ,030217 neurology & neurosurgery - Abstract
Profiling the antibody response to Ebola The recent Ebola virus outbreak in West Africa illustrates the need not only for a vaccine but for potential therapies, too. One promising therapy is monoclonal antibodies that target Ebola's membrane-anchored glycoprotein (GP). Bornholdt et al. isolated and characterized 349 antibodies from a survivor of the 2014 outbreak. A large fraction showed some neutralizing activity and several were quite potent. Structural analysis revealed an important site of vulnerability on the membrane stalk region of GP. Antibodies targeting this area were therapeutically effective in Ebola virus–infected mice. Science , this issue p. 1078
- Published
- 2016
- Full Text
- View/download PDF
13. List of Contributors
- Author
-
Roshini Sarah Abraham, Cristina Albanesi, Ilias Alevizos, Juan Anguita, Brendan Antiochos, Cynthia Aranow, John P. Atkinson, Howard A. Austin, Subash Babu, Mark C. Ballow, James E. Balow, John W. Belmont, Claudia Berek, Timothy Beukelman, Tapan Bhavsar, J. Andrew Bird, Sarah E. Blutt, Mark Boguniewicz, Rafael Bonamichi-Santos, Bertrand Boisson, Elena Borzova, Prosper N. Boyaka, Joshua Boyce, Sarah K. Browne, Wesley Burks, Jacinta Bustamante, Virginia L. Calder, Matthew Campbell, Adela Rambi G. Cardones, Jean-Laurent Casanova, Mariana Castells, Lisa A. Cavacini, Edwin S.L. Chan, David D. Chaplin, W. Winn Chatham, Edward S. Chen, Javier Chinen, Lisa Christopher-Stine, Michael Ciancanelli, Andrew P. Cope, David B. Corry, Filippo Crea, Randy Q. Cron, Jennifer M. Cuellar-Rodriguez, Marinos C. Dalakas, Sara M. Dann, Betty Diamond, Terry W. Du, Stéphanie Dupuis-Boisson, Todd N. Eagar, Craig A. Elmets, Doruk Erkan, Laura Fanning, Erol Fikrig, Davide Flego, Thomas A. Fleisher, Luz Fonacier, Andrew P. Fontenot, Alexandra F. Freeman, Anthony J. Frew, Kohtaro Fujihashi, Massimo Gadina, Moshe E. Gatt, M. Eric Gershwin, Susan L. Gillespie, Jörg J. Goronzy, Sangeeta Goswami, Clive E.H. Grattan, Neil S. Greenspan, Sarthak Gupta, Claire E. Gustafson, Russell P. Hall, Robert G. Hamilton, Laurie E. Harrington, Leonard C. Harrison, Sarfaraz A. Hasni, Arthur Helbling, Joanna Hester, Steven M. Holland, Dennis Hourcade, Nicholas D. Huntington, Tracy Hwangpo, John B. Imboden, Fadi Issa, Shai Izraeli, Elaine S. Jaffe, Sirpa Jalkanen, Stacie Jones, Emmanuelle Jouanguy, Sarah Kabbani, Stefan H.E. Kaufmann, Farrah Kheradmand, Donald B. Kohn, Robert Korngold, Anna Kovalszki, Douglas B. Kuhns, Hrishikesh Kulkarni, Caroline Y. Kuo, Arash Lahouti, C. Ola Landgren, Arian Laurence, Joyce S. Lee, Catherine Lemière, Donald Y.M. Leung, Arnold I. Levinson, Ofer Levy, Dorothy E. Lewis, Phoebe Lin, Andreas Linkermann, Giovanna Liuzzo, Michael D. Lockshin, Allison K. Lord, Jay N. Lozier, Amber Luong, Raashid Luqmani, Meggan Mackay, Jonathan S. Maltzman, Peter J. Mannon, Michael P. Manns, James G. Martin, Craig L. Maynard, Samual McCash, Douglas R. McDonald, Peter C. Melby, Stephen D. Miller, Anna L. Mitchell, Amirah Mohd-Zaki, Carolyn Mold, David R. Moller, Dimitrios S. Monos, Scott N. Mueller, Catharina M. Mulders-Manders, Mark J. Mulligan, Ulrich R. Müller, Pashna N. Munshi, Kazunori Murata, Philip M. Murphy, Nicolás Navasa, Pierre Noel, Luigi D. Notarangelo, Robert L. Nussbaum, Thomas B. Nutman, Stephen L. Nutt, João B. Oliveira, Thomas L. Ortel, John J. O'Shea, Sung-Yun Pai, Lavannya Pandit, Mary E. Paul, Simon H.S. Pearce, Daniela Pedicino, Erik J. Peterson, Capucine Picard, Stefania Pittaluga, Debra Long Priel, Jennifer Puck, Anne Puel, Andreas Radbruch, Stephen T. Reece, John D. Reveille, Robert R. Rich, Chaim M. Roifman, Antony Rosen, James T. Rosenbaum, Sergio D. Rosenzweig, Barry T. Rouse, Scott D. Rowley, Shimon Sakaguchi, Marko Salmi, Andrea J. Sant, Sarah W. Satola, Valerie Saw, Marcos C. Schechter, Harry W. Schroeder, Benjamin M. Segal, Carlo Selmi, Sushma Shankar, Anu Sharma, Padmanee Sharma, William T. Shearer, Richard M. Siegel, Anna Simon, Gideon P. Smith, David S. Stephens, Robin Stephens, Alex Straumann, Leyla Y. Teos, Laura Timares, Wulf Tonnus, Raul M. Torres, Gülbü Uzel, Jeroen C.H. van der Hilst, Jos W.M. van der Meer, John Varga, Jatin M. Vyas, Meryl Waldman, Peter Weiser, Peter F. Weller, Cornelia M. Weyand, Fredrick M. Wigley, Robert J. Winchester, James B. Wing, Kathryn J. Wood, Xiaobo Wu, Hui Xu, Cassian Yee, and Shen-Ying Zhang
- Published
- 2019
- Full Text
- View/download PDF
14. Correction for Giuntini et al., 'Identification and Characterization of Human Monoclonal Antibodies for Immunoprophylaxis against Enterotoxigenic Escherichia coli Infection'
- Author
-
William D. Thomas, Zachary A. Schiller, Mark S. Klempner, Eileen M. Barry, Maja Sedic, Monir Ejemel, Yang Wang, Danielle Wisheart, Matteo Stoppato, Serena Giuntini, Lisa A. Cavacini, and Jessica R. Pondish
- Subjects
0301 basic medicine ,Escherichia coli Vaccines ,medicine.drug_class ,Immunology ,Antibodies, Monoclonal ,Biology ,Monoclonal antibody ,Microbiology ,Virology ,Enterotoxigenic Escherichia coli infection ,Mice ,03 medical and health sciences ,030104 developmental biology ,Infectious Diseases ,medicine ,Animals ,Enterotoxigenic Escherichia coli ,Humans ,Parasitology ,Author Correction ,Escherichia coli Infections - Abstract
Volume 86, no. 8, e00355-18, 2018, [https://doi.org/10.1128/IAI.00355-18][1]. Page 4: Figure 2B should appear as shown below. ![Figure][2] [1]: /lookup/doi/10.1128/IAI.00355-18 [2]: pending:yes
- Published
- 2018
- Full Text
- View/download PDF
15. Neutralization of HIV by Milk Expressed Antibody
- Author
-
Harry M. Meade, Kristin Joseph, Daniel Pollock, Mark Duval, Xiaocong Yu, Lisa A. Cavacini, and Christopher J. Lewis
- Subjects
Immunoglobulin A ,medicine.drug_class ,Mammary gland ,Gene Expression ,Mice, Transgenic ,CHO Cells ,HIV Antibodies ,Breast milk ,Immunoglobulin light chain ,Monoclonal antibody ,Article ,Neutralization ,Mice ,Cricetulus ,fluids and secretions ,Neutralization Tests ,Cricetinae ,medicine ,Animals ,Humans ,Pharmacology (medical) ,biology ,Antibodies, Monoclonal ,HIV ,Antibodies, Neutralizing ,Virology ,Milk ,Infectious Diseases ,Cell killing ,medicine.anatomical_structure ,Immunology ,biology.protein ,Female ,Antibody ,Plasmids - Abstract
Background In some areas of the world, mother-to-child transmission of HIV remains a significant problem in part due to widespread breastfeeding, which is essential because of scarce supply of a safe replacement, protection conferred by breast milk against many enteric illnesses, and cultural norms. We propose that sustained adequate levels of protective antibodies in breast milk will prevent transmission of HIV. Methods The HIV-neutralizing human monoclonal antibody b12 (IgG1) has been expressed as an IgA2 in CHO cells and shown to retain full immunoreactivity and neutralizing activity as the parental IgG1. The expression plasmids containing the b12 heavy and light chains were also used to construct milk-specific expression vectors using the GTC goat β-casein expression vector to direct expression of linked genes to the mammary gland with subsequent secretion into the milk. Female transgenic mice were generated and following parturition, their milk was tested for antibody immunoreactivity with gp120 and neutralization of HIV. Results When milk-derived b12 IgA2 was compared with CHO-derived b12 IgA2 (or IgG1), immunoreactivity was retained. When tested for neutralization, milk-derived b12 IgA2 was at least comparable to CHO-derived antibody and in some cases, superior to CHO-derived antibody. Furthermore, milk that expressed b12 IgA2 was significantly more effective at mediating antibody-dependent cell killing. Conclusions These results suggest that it is possible to achieve functional HIV-specific mAb in the milk of transgenic mice, and further investigations are warranted to explore ways for inducing this type of antibody response in the breast milk of HIV-infected women.
- Published
- 2013
- Full Text
- View/download PDF
16. Enhanced Neutralization of HIV by Antibodies Displayed on the S-Layer of Caulobacter crescentus
- Author
-
John Smit, Marc S. Horwitz, John F. Nomellini, Christopher J. Lewis, Mark Duval, and Lisa A. Cavacini
- Subjects
Caulobacter ,Anti-HIV Agents ,HIV Infections ,HIV Antibodies ,Antiviral Agents ,Epitope ,Neutralization ,Microbiology ,law.invention ,Drug Delivery Systems ,Neutralization Tests ,law ,Caulobacter crescentus ,Humans ,Pharmacology (medical) ,Pharmacology ,Membrane Glycoproteins ,biology ,biology.organism_classification ,Virology ,Recombinant Proteins ,Infectious Diseases ,CD4 Antigens ,HIV-1 ,biology.protein ,Recombinant DNA ,Female ,Protein G ,Antibody ,Genetic Engineering ,S-layer - Abstract
Innovative methods of prevention are needed to stop the more than two million new HIV-1 infections annually, particularly in women. Local application of anti-HIV antibodies has been shown to be effective at preventing infection in nonhuman primates; however, the concentrations needed are cost prohibitive. Display of antibodies on a particulate platform will likely prolong effectiveness of these anti-HIV agents and lower the cost of goods. Here, we demonstrate that the bacterium Caulobacter crescentus and its highly expressed surface-layer (S-layer) protein can provide this antibody display platform. Caulobacters displaying protein G, alone or with CD4 codisplay, successfully captured HIV-1-specific antibodies and demonstrated functional neutralization. Compared to soluble antibodies, a neutralizing anti-HIV antibody displayed on Caulobacter was as effective or more effective at neutralizing diverse HIV-1 isolates. Moreover, when an antibody reactive with an epitope induced by CD4 binding (CD4i) was codisplayed with CD4, there was significant enhancement in HIV-1 neutralization. These results suggest that caulobacters displaying anti-HIV antibodies offer a distinct improvement in the use of antibodies as microbicides. Furthermore, these reagents can specifically evaluate anti-HIV antibodies in concert with other HIV-1 blocking agents to assess the most suitable tools for conversion to scFvs, allowing for direct display within the S-layer protein and further reducing cost of goods. In summary, C. crescentus , which can be easily produced and chemically stabilized at low cost, is well suited for engineering as an effective platform, offering an inexpensive way to produce and deliver HIV-1-specific microbicides.
- Published
- 2011
- Full Text
- View/download PDF
17. Neutralizing activity of antibodies to the V3 loop region of HIV-1 gp120 relative to their epitope fine specificity
- Author
-
James E. Robinson, Terri Wrin, Lisa A. Cavacini, Ralph Pantophlet, and Dennis R. Burton
- Subjects
Models, Molecular ,medicine.drug_class ,V3 accessibility ,Epitope mapping ,Amino Acid Motifs ,Human immunodeficiency virus (HIV) ,Cross-neutralization ,Cross Reactions ,HIV Antibodies ,HIV Envelope Protein gp120 ,V3 loop ,medicine.disease_cause ,Monoclonal antibody ,Article ,Neutralization ,Virus ,Epitope ,Cell Line ,Epitopes ,03 medical and health sciences ,Antibody Specificity ,Neutralization Tests ,Virology ,medicine ,Amino Acid Sequence ,030304 developmental biology ,0303 health sciences ,Alanine ,biology ,030306 microbiology ,Antibodies, Monoclonal ,V3 antibodies ,HIV vaccine design ,Peptide Fragments ,Protein Structure, Tertiary ,3. Good health ,HIV-1 ,biology.protein ,Antibody ,Sequence Alignment - Abstract
The V3 loop of HIV-1 gp120 is considered occluded on many primary viruses. However, virus sensitivity to neutralization by different V3 mAbs often varies, indicating that access to V3 is not restricted equally for all antibodies. Here, we have sought to gain a better understanding of these restrictions by determining the neutralizing activities of 7 V3 mAbs (19b, 39F, CO11, F2A3, F530, LA21, and LE311) against 15 subtype B primary isolates and relating these activities to the fine specificity of the mAbs. Not surprisingly, we found that most mAbs neutralized the same 2–3 viruses, with only mAb F530 able to neutralize 2 additional viruses not neutralized by the other mAbs. Epitope mapping revealed that positively-charged residues in or near the V3 stem are important for the binding of all the mAbs and that most mAbs seem to require the Pro residue that forms the GPGR β hairpin turn in the V3 tip for binding. Based on the mapping, we determined that V3 sequence variation accounted for neutralization resistance of approximately half the viruses tested. Comparison of these results to those of select V3 mAbs with overall better neutralizing activities in the light of structural information illustrates how an antibody's mode of interaction with V3, driven by contact residue requirements, may restrict the antibody from accessing its epitope on different viruses. Based on the data we propose an angle of interaction with V3 that is less stringent on access for antibodies with cross-neutralizing activity compared to antibodies that neutralize relatively fewer viruses.
- Published
- 2008
- Full Text
- View/download PDF
18. Pathogenic human monoclonal antibody against desmoglein 3
- Author
-
Mukesh Kumar, Kailash C. Bhol, Mong-Shang Lin, Lisa A. Cavacini, Shih-Wei Yeh, Mark Duval, Marshall R. Posner, and A. Razzaque Ahmed
- Subjects
Keratinocytes ,medicine.drug_class ,Blotting, Western ,Immunology ,Biology ,Monoclonal antibody ,Mice ,medicine ,Animals ,Humans ,Immunology and Allergy ,Fluorescent Antibody Technique, Indirect ,education ,Mice, Inbred BALB C ,education.field_of_study ,Desmoglein 3 ,integumentary system ,Acantholysis ,Pemphigus vulgaris ,Autoantibody ,Antibodies, Monoclonal ,Intradermal Tests ,medicine.disease ,Immunohistochemistry ,medicine.anatomical_structure ,Animals, Newborn ,Desmoglein 1 ,Immunoglobulin G ,biology.protein ,Immunotherapy ,Antibody ,Keratinocyte ,Epitope Mapping ,Pemphigus - Abstract
Pemphigus vulgaris (PV) is a potentially fatal autoimmune mucocutaneous disease associated with production of IgG autoantibodies to desmoglein 3 (Dsg3), a 130-kDa epidermal cadherin protein. The binding of pathogenic antibody to Dsg3 on epidermal keratinocytes leads to loss of intercellular adhesion and results in intraepithelial blister formation. Here, we describe a human monoclonal antibody, PVMAB786, a Dsg3-specific IgG4 antibody, from an untreated patient with active PV. The antibody reacts with a 130-kDa protein on keratinocyte cell surfaces and recombinant Dsg3 protein, but not desmoglein 1 protein. PVMAB786 induces acantholysis in normal human skin and mucous membranes and induces a clinical and histological profile similar to human PV when injected into neonatal mice. PVMAB786 will be a valuable tool in identifying the role of Dsg3 in epithelial cell adherence and acantholysis, mechanisms of Dsg3 processing/presentation and V gene and isotype usage in PV pathogenesis.
- Published
- 2006
- Full Text
- View/download PDF
19. Structure of the Fab Fragment of F105, a Broadly Reactive Anti-Human Immunodeficiency Virus (HIV) Antibody That Recognizes the CD4 Binding Site of HIV Type 1 gp120
- Author
-
Chayne L. Piscitelli, C. Martin Lawrence, Martin Teintze, Marshall R. Posner, Lisa A. Cavacini, and Royce A. Wilkinson
- Subjects
Models, Molecular ,Conformational change ,medicine.drug_class ,Molecular Sequence Data ,Immunology ,HIV Antibodies ,HIV Envelope Protein gp120 ,medicine.disease_cause ,Monoclonal antibody ,Microbiology ,Virus ,Epitope ,Immunoglobulin Fab Fragments ,Antibody Specificity ,Virology ,medicine ,Amino Acid Sequence ,Binding site ,Crystallography ,biology ,Lipid bilayer fusion ,Simian immunodeficiency virus ,Complementarity Determining Regions ,Insect Science ,CD4 Antigens ,HIV-1 ,biology.protein ,Pathogenesis and Immunity ,Antibody - Abstract
A key step in the development of a successful vaccine against human immunodeficiency virus (HIV) will be the design of immunogens capable of generating an effective humoral immune response (3). Antibodies with two features characterize such a response. They must show, at the same time, both potency and broad specificity. A number of human antibodies that recognize elements of the conserved CD4 binding site of HIV type 1 (HIV-1) gp120 have now been isolated from HIV-infected patients (2, 11, 16, 24, 38, 40, 47, 49, 59, 65). This includes immunoglobulin G1 (IgG1) b12 (38, 47, 53), one of the most potent and broadly reactive anti-HIV antibodies known. In contrast, other CD4 binding-site antibodies are less potent towards many clinical isolates, even though they may be broadly cross-reactive. F105, the subject of this paper, is representative of the latter group. F105 is an IgG1 κ human monoclonal antibody isolated from an HIV-infected individual (49). It binds to the CD4 binding sites of both trimeric and monomeric gp120 and is capable of neutralizing various strains of HIV (e.g., IIIB [HXBc2], MN, RF, and SF2) (7, 49, 58) but is less successful against many primary clinical isolates (12, 32). F105 did not show evidence of anti-HIV-1 activity or a viral load decrease in a phase I dose-escalation study (5, 70). However, in triple and quadruple combination therapies with anti-HIV monoclonal antibodies (2F5, 2G12, and 694/98D) with other specificities, a complete and synergistic neutralization of the SHIV-Vpu+ chimeric simian-human immunodeficiency virus was seen in macaque peripheral blood mononuclear cells in vitro and in an in vivo macaque model that mimics mucosal exposure during intrapartum virus transmission (1, 31). Crystallographic studies of the ternary complex of the HIV gp120 core, CD4, and antibody 17b provided the first look at the structure of gp120 and its interactions with CD4 (26-28, 65). CD4 was found to bind at the nexus of the inner domain, the outer domain, and the bridging sheet of gp120 (26, 27). A large body of biochemical and biophysical data indicates a considerable conformational change in gp120 upon binding to CD4 (4, 6, 15, 24, 26, 27, 39, 41, 50, 55, 60, 62-69, 72, 73). The conformational change results in the formation and/or exposure of the chemokine receptor sites (62, 64), thus promoting further viral attachment and membrane fusion. The molecular reorganization that results upon binding of CD4 is revealed by the structure of an unliganded simian immunodeficiency virus (SIV) gp120 core (6). With a few important exceptions, the structure of the outer domain is quite similar to that seen in the CD4-bound state. In contrast, the structure of the inner domain is markedly different. A comparison of CD4-bound and unliganded gp120 shows that the conformational change is not a simple movement of the inner domain as a rigid body. Rather, the inner domain is comprised of a set of distinct substructures that move relatively independently of one another (6). The binding of CD4 results in a rearrangement of these secondary structural elements within the inner domain (6). As predicted (24), the bridging sheet is not present in the structure of unliganded gp120 (6). The structure of antibody b12 was determined by Saphire et al. (52-54). The structure revealed an extended CDR H3 loop with an apical tryptophan residue that is thought to recognize the Phe43 binding pocket of gp120 (53, 75). This putative interaction places significant constraints on possible gp120/b12 interactions, allowing an interaction between b12 and the CD4-bound conformation of gp120 to be modeled (53, 75). This model suggests that the broad neutralizing activity of b12 lies in its ability to interact with conserved features of the CD4 binding site through recognition of the Phe43 pocket and interactions with main chain atoms of gp120 and in its ability to recognize trimeric gp120 on the native viral surface. A further understanding of the molecular properties that confer broad reactivity and potency upon a CD4 binding-site antibody are of substantial interest. The underlying principles are likely to impact the design of new immunogens with the potential to elicit a successful immune response. In this regard, the structure of F105 provides an opportunity to compare and contrast the structure of a broadly reactive but nonpotent CD4 binding-site antibody (F105) with that of a broadly neutralizing antibody with an overlapping epitope (b12). The comparison provides significant insight into the molecular properties that impart broad reactivity and potency upon a CD4 binding-site antibody.
- Published
- 2005
- Full Text
- View/download PDF
20. CD40 function in squamous cell cancer of the head and neck
- Author
-
Wenhui Cao, Karl C. Tillman, Marshall R. Posner, and Lisa A. Cavacini
- Subjects
Cancer Research ,Cell signaling ,Fas Ligand Protein ,CD40 Ligand ,Apoptosis ,Cell Communication ,Biology ,Fas ligand ,Proinflammatory cytokine ,chemistry.chemical_compound ,Epidermal growth factor ,Cell Line, Tumor ,Humans ,Secretion ,Neoplasms, Squamous Cell ,CD40 Antigens ,Phosphorylation ,Membrane Glycoproteins ,Epidermal Growth Factor ,hemic and immune systems ,Tyrosine phosphorylation ,ErbB Receptors ,stomatognathic diseases ,Oncology ,chemistry ,Head and Neck Neoplasms ,Prostaglandins ,Cancer research ,Cytokines ,Oral Surgery - Abstract
CD40 is expressed on basal keratinocytes and Squamous Cell Cancer of the Head and Neck (SCCHN) tumor cells in vivo and in vitro. CD40 ligation reduces proliferation of SCCHN cell lines and enhances EGFr mediated inhibition of proliferation. We investigated the mechanisms of CD40 function and EGFr cross-communication in SCCHN cell lines. CD40 ligation inhibited spontaneous and Fas-induced apoptosis. CD40 ligation specifically increased the secretion of IL-8, VEGF and PGE(2) but not IL-6, IL-10, FasL, GM-CSF, or TGFalpha. Co-ligation with EGFr further increased IL-8, VEGF and PGE(2) secretion. CD40 ligation also induced delayed activation and tyrosine phosphorylation of EGFr. CD40 induces secretion of specific proinflammatory and proangiogenic cytokines, inhibits spontaneous and Fas-induced apoptosis and increases EGFr phosphorylation. CD40 signaling may enhance the survival of SCCHN and tumor stroma.
- Published
- 2005
- Full Text
- View/download PDF
21. Dichotomy in cross-clade reactivity and neutralization by HIV-1 sera: Implications for active and passive immunotherapy
- Author
-
Lisa A. Cavacini, Kenneth H. Mayer, Mark Duval, Charles E. Wood, Ajay Patil, Marshall R. Posner, and Ruth M. Ruprecht
- Subjects
AIDS Vaccines ,biology ,viruses ,Immunization, Passive ,HIV Infections ,Cross Reactions ,HIV Antibodies ,biology.organism_classification ,Virology ,Peripheral blood mononuclear cell ,Neutralization ,Virus ,Pathogenesis ,Infectious Diseases ,Immunization ,Neutralization Tests ,Lentivirus ,HIV-1 ,biology.protein ,Humans ,Antibody ,Clade - Abstract
The identification of broadly reactive and cross-clade neutralizing antibodies will facilitate the development of a more universally effective vaccine for human immunodeficiency virus (HIV). Antibodies in sera from individuals infected with Clade B HIV bind native primary viral isolates, and virus binding correlates with neutralization and stable clinical disease. In this study, we quantified cross-clade antibody reactivity and neutralization by Clades B and C sera. Primary viral isolates were captured by serum IgG bound to anti-human IgG and quantitated as p24 released by lysis of captured virus. Neutralization was determined using PHA-stimulated PBMC. Clade B antibodies reacted more frequently with Clade B R5 virus, but positive sera captured quantitatively more X4 virus than R5 and R5X4 virus. Clade B sera reacted less frequently and captured less Clade C virus than Clade B virus. Antibodies in Clade C sera captured Clades B and C isolates with equal frequency and quantity. There was no difference in neutralization of Clade B virus by either group of sera; however, Clade C sera neutralized Clade C virus, whereas Clade B sera were ineffective against Clade C virus. Thus, there are distinct differences in cross-clade reactivity of and neutralization by antibodies induced in response to Clade C infection compared to Clade B infection. Understanding antibody responses to native virions after Clade C infection and cross clade antibody behavior has implications for understanding pathogenesis and vaccine development.
- Published
- 2005
- Full Text
- View/download PDF
22. Human Monoclonal Antibodies to Pseudomonas aeruginosa Alginate That Protect against Infection by Both Mucoid and Nonmucoid Strains
- Author
-
Nicolas Llosa, Marshall R. Posner, Michael J. Preston, Fadie T. Coleman, Christian Theilacker, Debra Boyer, Gregory P. Priebe, Hannah Goldenberg, Jeffrey Uchin, Simone Mueschenborn-Koglin, Gerald B. Pier, Lisa A. Cavacini, and Martha Grout
- Subjects
Alginates ,medicine.drug_class ,Immunology ,Immunoglobulin Variable Region ,Biology ,medicine.disease_cause ,Monoclonal antibody ,Epitope ,Microbiology ,Mice ,Glucuronic Acid ,Phagocytosis ,Species Specificity ,In vivo ,Pneumonia, Bacterial ,medicine ,Mannuronic acid ,Animals ,Humans ,Immunology and Allergy ,Pseudomonas Infections ,Lung ,Mice, Inbred C3H ,Hybridomas ,Pseudomonas aeruginosa ,Hexuronic Acids ,Antibodies, Monoclonal ,medicine.disease ,Antibodies, Bacterial ,Recombinant Proteins ,In vitro ,respiratory tract diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,Murine lung ,Acute Disease ,Female ,Binding Sites, Antibody ,Pneumonia (non-human) - Abstract
Two fully human mAbs specific for epitopes dependent on intact carboxylate groups on the C6 carbon of the mannuronic acid components of Pseudomonas aeruginosa alginate were found to promote phagocytic killing of both mucoid and nonmucoid strains as well as protection against both types of strains in a mouse model of acute pneumonia. The specificity of the mAbs for alginate was determined by ELISA and killing assays. Some strains of P. aeruginosa did not make detectable alginate in vitro, but in vivo protection against lethal pneumonia was obtained and shown to be due to rapid induction of expression of alginate in the murine lung. No protection against strains genetically unable to make alginate was achieved. These mAbs have potential to be passive therapeutic reagents for all strains of P. aeruginosa and the results document that alginate is a target for the proper type of protective Ab even when expressed at low levels on phenotypically nonmucoid strains.
- Published
- 2004
- Full Text
- View/download PDF
23. Binding and Neutralization Activity of Human IgG1 and IgG3 from Serum of HIV-Infected Individuals
- Author
-
Kenneth H. Mayer, Lisa A. Cavacini, Marshall R. Posner, Mark Duval, and David Kuhrt
- Subjects
medicine.drug_class ,Immunology ,HIV Infections ,HIV Antibodies ,HIV Envelope Protein gp120 ,Monoclonal antibody ,complex mixtures ,Neutralization ,Cell Line ,Antigen ,Neutralization Tests ,Virology ,parasitic diseases ,medicine ,Humans ,Neutralizing antibody ,biology ,Antibodies, Monoclonal ,biology.organism_classification ,Isotype ,HIV Envelope Protein gp41 ,Immunoglobulin Isotypes ,Infectious Diseases ,Immunoglobulin G ,Humoral immunity ,Lentivirus ,biology.protein ,Antibody - Abstract
The IgG1 and IgG3 subclasses represent the predominant antibody response to viral infections, including HIV. IgG subclasses differ in their interaction with antigen and functional effects due to specific physiochemical features. With an elongated hinge, IgG3 antibodies tend to have more segmental flexibility, which can render the antibody more effective at interacting with antigen. We have previously shown that the change of the human anti-CD4-binding site monoclonal antibody F105 from IgG1 to IgG3 results in neutralization of a T cell line-adapted isolate (TCLA) resistant to neutralization by the parental IgG1. In the studies presented here, we have purified IgG1 and IgG3 subclasses from the sera of HIV-infected individuals and tested for immunoreactivity with and neutralization of HIV. Purified total IgG3 tended to have less relative reactivity and mediated relatively poorer neutralization of either laboratory or primary isolates. IgG3 also tended to react relatively less well with gp160 and gp120 and more robustly with gp41 and p24. The contrasting results with serum, as opposed to F105, may result from the polyclonal nature of serum antibodies. There is also a failure to make a robust IgG3 response to neutralizing epitopes on envelope glycoproteins during natural infection. These studies suggest that the investigation of isotype effects on neutralization will require isotype-switched human monoclonal antibodies. Understanding isotype and neutralization will provide important data necessary for designing the most effective possible vaccines.
- Published
- 2003
- Full Text
- View/download PDF
24. Expression and Functional Activity of Isotype and Subclass Switched Human Monoclonal Antibody Reactive with the Base of the V3 Loop of HIV-1 gp120
- Author
-
Pablo Lopez Bergami, Mark Duval, Lisa A. Cavacini, David Kuhrt, Fangbing Liu, and Marshall R. Posner
- Subjects
HIV Antigens ,medicine.drug_class ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Immunology ,Immunoglobulin Variable Region ,HIV Antibodies ,HIV Envelope Protein gp120 ,Biology ,V3 loop ,Transfection ,Monoclonal antibody ,Epitope ,Antigen-Antibody Reactions ,Epitopes ,Mice ,Structure-Activity Relationship ,Antigen ,Antibody Specificity ,Neutralization Tests ,Virology ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Primary isolate ,Hybridomas ,Base Sequence ,Genes, Immunoglobulin ,Virion ,Antibodies, Monoclonal ,Immunoglobulin Class Switching ,Isotype ,Peptide Fragments ,Immunoglobulin A ,Immunoglobulin Isotypes ,Infectious Diseases ,Immunoglobulin class switching ,Immunoglobulin G ,HIV-1 ,Mutagenesis, Site-Directed ,biology.protein ,Antibody ,Multiple Myeloma ,Sequence Alignment - Abstract
Immunoglobulins undergo isotype switching in response to antigenic stimulation. The C(H) domains, in particular the hinge region, impose structural constraints on the interaction of antibody with antigen, especially multivalent antigens such as HIV. We previously showed that switching the IgG1 anti-HIV human monoclonal antibody (HMAb) F105 to an IgG3 resulted in significantly enhanced neutralization of HIV. To further investigate the influence of isotype, including the functional activity of HMAbs switched to IgA, which may be important in mucosal defenses, isotype switched antibodies have been generated for the anti-V3 loop base IgG2 HMAb F425B4e8. Reactivity of the IgG1 antibody was greater than the parental IgG2 antibody for SF2 infected cells but less for primary isolate virions. In contrast, there was less reactivity of the IgG3 with either infected cells or virions. IgA reacted significantly more with infected cells and virions as compared to the IgG subclasses. In contrast to previous studies whereby IgG3 enhanced neutralization, comparable neutralization of primary isolate virus was observed for IgG subclasses (IgG1, IgG2, IgG3) and IgA. This may reflect differences in the exposure of epitopes recognized by the HMAb with antibody flexibility being important to neutralization by antibodies reactive with obscured epitopes (e.g., CD4 binding site). Further analysis of the in vitro activity of isotype or subclass switched antibodies, IgA in particular, alone and in combination with other HMAbs, will provide important information on the role of IgG subclass and IgA antibodies on protective immunity to HIV.
- Published
- 2003
- Full Text
- View/download PDF
25. Conformational changes in env oligomer induced by an antibody dependent on the V3 loop base
- Author
-
Joseph Sodroski, Mark Duval, Shi Hua Xiang, Leslie Song, Rebecca Sangster, Marshall R. Posner, and Lisa A. Cavacini
- Subjects
Protein Conformation ,medicine.drug_class ,viruses ,Immunology ,HIV Infections ,HIV Envelope Protein gp120 ,V3 loop ,Monoclonal antibody ,Epitope ,Neutralization ,Virus ,Epitopes ,Neutralization Tests ,medicine ,Humans ,Immunology and Allergy ,Binding site ,Primary isolate ,Cells, Cultured ,biology ,Virion ,Antibodies, Monoclonal ,Gene Products, env ,Virology ,Molecular biology ,Peptide Fragments ,Infectious Diseases ,Immunoglobulin G ,HIV-1 ,biology.protein ,Antibody - Abstract
Objective: The HIV-1 env oligomer is structured such that conserved, neutralizing epitopes are obscured by gp120 variable loops. We have studied the ability of an IgG2 human monoclonal antibody (hmAb), F425 B4e8 (B4e8), dependent upon the base of the V3 loop, to induce conformational changes in the env oligomer. Design: The effect of B4e8 antibody on the exposure of neutralizing epitopes and viral neutralization was studied in combination with other hmAb. Methods: Epitope exposure and viral neutralization was determined using native, intact primary isolate virions. Results: B4e8 antibody neutralizes infection and binds to HIV-infected cells and primary isolate virions. B4e8 and 2G12 enhanced the binding of each other to infected cells or virus and the combination resulted in synergistic neutralization. B4e8 also enhanced the binding of CD4i and CD4 binding site antibodies. Conclusions: The conserved epitopes exposed by B4e8 are similar to those exposed by the movement of the variable loops following CD4 engagement. Further studies with select antibody combinations should provide important information for the design of effective immunotherapeutic agents.
- Published
- 2003
- Full Text
- View/download PDF
26. Post-exposure prophylaxis with human monoclonal antibodies prevented SHIV89.6P infection or disease in neonatal macaques
- Author
-
Lisa A. Cavacini, Robert A. Rasmussen, P.-L. Li, Daniel C. Anderson, Gabriela Stiegler, Weidong Xu, Harold M. McClure, David C. Montefiori, Hermann Katinger, Ruth M. Ruprecht, Tao Wang, Flavia Ferrantelli, and Regina Hofmann-Lehmann
- Subjects
Immunology ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Viremia ,Biology ,medicine.disease_cause ,Macaque ,Virus ,biology.animal ,medicine ,Animals ,Immunology and Allergy ,Prospective Studies ,Immunodeficiency ,Immunity, Cellular ,Chimera ,Immunization, Passive ,Antibodies, Monoclonal ,Simian immunodeficiency virus ,biology.organism_classification ,medicine.disease ,Macaca mulatta ,Virology ,Infectious Disease Transmission, Vertical ,CD4 Lymphocyte Count ,Infectious Diseases ,Animals, Newborn ,Immunization ,Immunoglobulin G ,Lentivirus ,Viral disease - Abstract
BACKGROUND: The majority of infants infected through maternal transmission acquire the virus during birth or postpartum through breastfeeding: mucosal exposure is considered to be a major route of infection. OBJECTIVES: To develop passive immunization with human neutralizing monoclonal antibodies (mAbs) against mother-to-child transmission of HIV during delivery and through breastfeeding. DESIGN: An oral challenge model in newborn rhesus macaques mimicked peri- and postpartum virus transmission. METHODS: Neonatal rhesus macaques were challenged orally with the highly pathogenic, chimeric simian-human immunodeficiency virus SHIV89.6P and given post-exposure prophylaxis with a quadruple combination of neutralizing human mAbs, IgG1b12, 2G12, 2F5, and 4E10, directed against conserved epitopes of HIV envelope glycoproteins. Control animals were virus challenged but left untreated. All infants were followed prospectively for signs of viremia and immunodeficiency. RESULTS: Two out of four macaque infants treated with neutralizing mAbs showed no evidence of infection; the other two maintained normal CD4 T cell counts. In contrast, all control animals became highly viremic and had profound CD4 T cell losses; three out of four died from AIDS within 1.5-6 weeks of the challenge. CONCLUSIONS: Passive immunization with this quadruple neutralizing mAbs combination may represent a promising approach to prevent peri- and postnatal HIV transmission. Furthermore, the epitopes recognized by the four neutralizing mAbs are key determinants to achieve complete protection and represent important targets against which to develop active, antibody-response-based AIDS vaccines.
- Published
- 2003
- Full Text
- View/download PDF
27. Interactions of human antibodies, epitope exposure, antibody binding and neutralization of primary isolate HIV-1 virions
- Author
-
Mark Duval, Marshall R. Posner, James A. Robinson, and Lisa A. Cavacini
- Subjects
medicine.drug_class ,viruses ,Immunology ,Virion ,Antibodies, Monoclonal ,Enzyme-Linked Immunosorbent Assay ,HIV Antibodies ,V3 loop ,Biology ,Monoclonal antibody ,Virology ,Neutralization ,Epitope ,Epitopes ,Infectious Diseases ,Virion binding ,CD4 Antigens ,HIV-1 ,medicine ,biology.protein ,Humans ,Immunology and Allergy ,Primary isolate ,Antibody ,HIV vaccine - Abstract
Objective Development of an effective HIV vaccine has been limited because of the inherent structural properties of the HIV envelope on native virions and the failure of the immune system to respond in an effective manner. Identification of the interactions of human antibodies with virions resulting in neutralization will facilitate vaccine design. Design Combinations of human monoclonal antibodies (hMAb) were studied for binding to and neutralization of primary isolate virions. Methods Virion binding and neutralization were measured using primary isolate virions. Results Antibodies and combinations of antibodies to epitopes exposed upon CD4 binding (CD4i) and V3 loop antibodies resulted in additive binding and neutralization of R5X4 virus. Antibodies did not bind to or neutralize R5 virus as well. The combination of V3 loop antibody with 2G12 resulted in enhanced neutralization and binding to the R5X4 isolate but not the R5 isolate. Preincubation of the R5X4 isolate with F240, a non-neutralizing anti-gp41 antibody, significantly enhanced binding and neutralization by CD4i hMAb and 2F5. F240 also enhanced the binding of 2F5 to the R5 isolate and the neutralization of the R5 isolate mediated by 2G12. Conclusions Neutralizing epitopes are obscured on intact primary isolate virions and are dynamically exposed upon ligand (CD4) interactions. Interestingly, a non-neutralizing antibody to gp41 also increased binding and neutralizing activity of some hMAb that poorly neutralized R5 virus. These data suggest that non-neutralizing epitopes may be appropriate targets for vaccine design and epitope exposure should be considered in the development of immunotherapeutic strategies for HIV.
- Published
- 2002
- Full Text
- View/download PDF
28. Postnatal pre- and postexposure passive immunization strategies: protection of neonatal macaques against oral simian-human immunodeficiency virus challenge
- Author
-
Bruce J. Bernacky, Lorraine R Hill, P.-L. Li, David C. Montefiori, Hermann Katinger, Robert A. Rasmussen, Shisong Jiang, Regina Hofmann-Lehmann, Ruth M. Ruprecht, Timothy W. Baba, Josef Vlasak, Tahir A. Rizvi, Michale E. Keeling, Gabriela Stiegler, Marshall R. Posner, Russell D. Schmidt, and Lisa A. Cavacini
- Subjects
chemistry.chemical_classification ,General Veterinary ,Low toxicity ,medicine.drug_class ,Virus transmission ,business.industry ,Simian human immunodeficiency virus ,Human immunodeficiency virus (HIV) ,medicine.disease_cause ,Monoclonal antibody ,medicine.disease ,Virology ,Immunization ,chemistry ,Immunology ,medicine ,Animal Science and Zoology ,Glycoprotein ,business ,Immunodeficiency - Abstract
Simian-human immunodeficiency viruses (SHIV) allow the evaluation of antiviral strategies that target the envelope glycoproteins of the human immunodeficiency virus 1 (HIV-1) in macaques. We previously protected neonates from oral challenge with cell-free SHIV-vpu+ by passive immunization with synergistic human neutralizing monoclonal antibodies (mAbs) (Baba et al., Nat Med 6:200-206, 2000). mAbs were administered prenatally to pregnant dams and postnatally to the neonates. Here, we used solely postnatal or postexposure mAb treatment, thus significantly reducing the amount of mAbs necessary. All neonatal monkeys were also protected with these abbreviated mAb regimens. Our results are directly relevant for humans because we used mAbs that target HIV-1 envelope glycoproteins. Thus, the large-scale use of passive immunization with neutralizing mAbs may be feasible in human neonates. The mAbs, being natural human proteins, can be expected to have low toxicity. Passive immunization has promise to prevent intrapartum as well as milk-borne virus transmission from HIV-1-infected women to their infants.
- Published
- 2002
- Full Text
- View/download PDF
29. Shared VH1-46 gene usage by pemphigus vulgaris autoantibodies indicates common humoral immune responses among patients
- Author
-
Christoph M. Hammers, Eun Jung Choi, Sara A. Farber, Courtney B. Rubin, Marshall R. Posner, Agnes Lo, Lisa A. Cavacini, Arielle R. Nagler, Bruce S. Sachais, Steven H. Kleinstein, Hong Li, Mohamed Uduman, Michael Jeffrey Cho, Eric M. Mukherjee, Preety M. Sharma, Christoph T. Ellebrecht, Ann H. Rux, Xuming Mao, and Aimee S. Payne
- Subjects
Clone (cell biology) ,General Physics and Astronomy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Germline ,Immune system ,Germline mutation ,medicine ,Humans ,education ,Autoantibodies ,Genetics ,education.field_of_study ,Multidisciplinary ,Desmoglein 3 ,Pemphigus vulgaris ,Autoantibody ,General Chemistry ,medicine.disease ,Complementarity Determining Regions ,Immunity, Humoral ,3. Good health ,Immunology ,biology.protein ,Antibody ,Pemphigus - Abstract
Pemphigus vulgaris (PV) is a potentially fatal blistering disease caused by autoantibodies against desmoglein 3 (Dsg3). Here, we clone anti-Dsg3 antibodies from four PV patients and identify pathogenic VH1-46 autoantibodies from all four patients. Unexpectedly, VH1-46 autoantibodies had relatively few replacement mutations. We reverted antibody somatic mutations to their germline sequences to determine the requirement of mutations for autoreactivity. Three of five VH1-46 germline-reverted antibodies maintain Dsg3 binding, compared to zero of five non-VH1-46 germline-reverted antibodies. Site-directed mutagenesis of VH1-46 antibodies demonstrate that acidic amino acid residues introduced by somatic mutation or heavy chain VDJ recombination are necessary and sufficient for Dsg3 binding. Our data suggest that VH1-46 autoantibody gene usage is commonly found in PV because VH1-46 antibodies require few to no mutations to acquire Dsg3 autoreactivity, which may favor their early selection. Common VH gene usage indicates common humoral immune responses, even among unrelated patients.
- Published
- 2014
- Full Text
- View/download PDF
30. Postnatal Passive Immunization of Neonatal Macaques with a Triple Combination of Human Monoclonal Antibodies against Oral Simian-Human Immunodeficiency Virus Challenge
- Author
-
Vladimir Liska, Josef Vlasak, Timothy W. Baba, Gabriela Stiegler, Regina Hofmann-Lehmann, Harold M. McClure, Russell D. Schmidt, Daniel C. Anderson, Beverly A. Smith, Tahir A. Rizvi, Ruth M. Ruprecht, Lori R. Hill, Hermann Katinger, Ting-Chao Chou, Marshall R. Posner, Flavia Ferrantelli, Lisa A. Cavacini, Janet Andersen, Robert A. Rasmussen, David C. Montefiori, Bruce J. Bernacky, Michale E. Keeling, University of Zurich, and Ruprecht, R M
- Subjects
1109 Insect Science ,medicine.drug_class ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Administration, Oral ,Biology ,Monoclonal antibody ,medicine.disease_cause ,Microbiology ,Virus ,Acquired immunodeficiency syndrome (AIDS) ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,Immunity, Mucosal ,2403 Immunology ,630 Agriculture ,Transmission (medicine) ,2404 Microbiology ,Immunization, Passive ,Antibodies, Monoclonal ,Drug Synergism ,Simian immunodeficiency virus ,medicine.disease ,In vitro ,10187 Department of Farm Animals ,Animals, Newborn ,Immunization ,Insect Science ,2406 Virology ,biology.protein ,570 Life sciences ,biology ,Macaca ,Simian Immunodeficiency Virus ,Antibody - Abstract
To develop prophylaxis against mother-to-child human immunodeficiency virus (HIV) transmission, we established a simian-human immunodeficiency virus (SHIV) infection model in neonatal macaques that mimics intrapartum mucosal virus exposure (T. W. Baba et al., AIDS Res. Hum. Retroviruses 10:351–357, 1994). Using this model, neonates were protected from mucosal SHIV-vpu+challenge by pre- and postnatal treatment with a combination of three human neutralizing monoclonal antibodies (MAbs), F105, 2G12, and 2F5 (Baba et al., Nat. Med. 6:200–206, 2000). In the present study, we used this MAb combination only postnatally, thereby significantly reducing the quantity of antibodies necessary and rendering their potential use in humans more practical. We protected two neonates with this regimen against oral SHIV-vpu+challenge, while four untreated control animals became persistently infected. Thus, synergistic MAbs protect when used as immunoprophylaxis without the prenatal dose. We then determined in vitro the optimal MAb combination against the more pathogenic SHIV89.6P, a chimeric virus encodingenvof the primary HIV89.6. Remarkably, the most potent combination included IgG1b12, which alone does not neutralize SHIV89.6P. We administered the combination of MAbs IgG1b12, 2F5, and 2G12 postnatally to four neonates. One of the four infants remained uninfected after oral challenge with SHIV89.6P, and two infants had no or a delayed CD4+T-cell decline. In contrast, all control animals had dramatic drops in their CD4+T cells by 2 weeks postexposure. We conclude that our triple MAb combination partially protected against mucosal challenge with the highly pathogenic SHIV89.6P. Thus, combination immunoprophylaxis with passively administered synergistic human MAbs may play a role in the clinical prevention of mother-to-infant transmission of HIV type 1.
- Published
- 2001
- Full Text
- View/download PDF
31. The N-Terminal V3 Loop Glycan Modulates the Interaction of Clade A and B Human Immunodeficiency Virus Type 1 Envelopes with CD4 and Chemokine Receptors
- Author
-
Lisa A. Cavacini, Marshall R. Posner, David Yang, James E. Robinson, Cecilia Cheng-Mayer, and Susan E. Malenbaum
- Subjects
Receptors, CXCR4 ,Glycan ,Glycosylation ,Receptors, CCR5 ,Immunology ,HIV Envelope Protein gp120 ,V3 loop ,Biology ,Microbiology ,Epitope ,Epitopes ,Structure-Activity Relationship ,Chemokine receptor ,Polysaccharides ,Virology ,Tumor Cells, Cultured ,Humans ,Binding site ,Clade ,chemistry.chemical_classification ,Binding Sites ,Chemokine receptor binding ,Virus-Cell Interactions ,chemistry ,Insect Science ,CD4 Antigens ,HIV-1 ,biology.protein ,Receptors, Chemokine ,Glycoprotein - Abstract
We investigated the underlying mechanism by which the highly conserved N-terminal V3 loop glycan of gp120 conferred resistance to neutralization of human immunodeficiency virus type 1 (HIV-1). We find that the presence or absence of this V3 glycan on clade A and B viruses accorded various degrees of susceptibility to neutralization by antibodies to the CD4 binding site, CD4-induced epitopes, and chemokine receptors. Our data suggest that this carbohydrate moiety on gp120 blocks access to the binding site for CD4 and modulates the chemokine receptor binding site of phenotypically diverse clade A and clade B isolates. Its presence also contributes to the masking of CD4-induced epitopes on clade B envelopes. These findings reveal a common mechanism by which diverse HIV-1 isolates escape immune recognition. Furthermore, the observation that conserved functional epitopes of HIV-1 are more exposed on V3 glycan-deficient envelope glycoproteins provides a basis for exploring the use of these envelopes as vaccine components.
- Published
- 2000
- Full Text
- View/download PDF
32. Review: Human Antibody Variable Region Gene Usage in HIV-1 Infection
- Author
-
Lisa A. Cavacini, Marshall R. Posner, and Adam V. Wisnewski
- Subjects
Phage display ,medicine.drug_class ,Immunology ,Biology ,Monoclonal antibody ,Virology ,Virus ,Serology ,Gene expression ,biology.protein ,medicine ,Immunology and Allergy ,Gene family ,Antibody ,Gene - Abstract
Human antibody variable region gene usage during human immunodeficiency virus type 1 (HIV-1) infection is examined in the following review, and several hypotheses are presented to account for the distinct patterns of antibody gene expression associated with infection. Evidence supporting qualitatively biased antibody gene expression has been derived from analysis of the human humoral immune response by isoelectric focusing (IEF) and serological and molecular studies of immunoglobulin (Ig) from different lymphoid compartments of HIV-1-infected patients. Preferential usage of heavy-chain variable region (VH) gene families 1 and 4 is supported by serological studies of serum Ig and molecular characterization of anti-HIV-1 human monoclonal antibodies derived from infected patients. Negative biases against VH3 family gene usage are detected by polymerase chain reaction (PCR) studies of peripheral blood lymphocytes from AIDS patients but not by combinatorial phage display library techniques. Biased antibody gene usage and expression during HIV-1 infection may be related to HIV-1 pathogenesis by limiting the available HIV-1 neutralizing repertoire. Further molecular characterization of anti-HIV-1 antibodies and in vivo expression of V-region genes during HIV-1 infection should provide important information regarding antibody gene expression and its relationship to HIV-1 pathogenesis.
- Published
- 1996
- Full Text
- View/download PDF
33. Immunoglobulin function
- Author
-
Lisa A. Cavacini and Neil S. Greenspan
- Subjects
biology ,Chemistry ,Immunology ,biology.protein ,Antibody ,Function (biology) - Published
- 2013
- Full Text
- View/download PDF
34. List of contributors
- Author
-
Roshini Sarah Abraham, Cristina Albanesi, Ilias Alevizos, Juan Anguita, Gregory M. Anstead, Cynthia Aranow, Howard A. Austin, Subash Babu, Mark C. Ballow, James E. Balow, David R. Barnidge, John W. Belmont, Gabrielle T. Belz, Dina Ben-Yehuda, Claudia Berek, Timothy Beukelman, Thomas Bieber, Johannes W.J. Bijlsma, Jack J.H. Bleesing, Sarah E. Blutt, Barbara Bohle, Elena Borzova, Prosper N. Boyaka, Brockow Knut, Jacinta Bustamante, Frank Buttgereit, Mary Byrne, Virginia L. Calder, Magda Carneiro-Sampaio, Sebastian Carotta, Jean-Laurent Casanova, Lisa A. Cavacini, Edwin S.L. Chan, Javier Chinen, Tanuja Chitnis, Monique Cho, Lisa Christopher-Stine, Andrew P. Cope, David B. Corry, Tricia Cottrell, Antonio Coutinho, Marco Craveiro, Randy Q. Cron, Jennifer Cuellar-Rodriguez, Marinos C. Dalakas, Stephanie C. de Barros, Blythe H. Devlin, Betty Diamond, Angela Dispenzieri, Terry W. Du Clos, Stéphanie Dupuis-Boisson, Todd N. Eagar, Kim D. Edhegard, George S. Eisenbarth, Craig A. Elmets, Doruk Erkan, Mark B. Feinberg, Erol Fikrig, Thomas A. Fleisher, Andrew P. Fontenot, Luis M. Franco, Alexandra F. Freeman, Anthony J. Frew, Thea Friedman, Kohtaro Fujihashi, Massimo Gadina, Stephen J. Galli, H. Bobby Gaspar, Moshe E. Gatt, M. Eric Gershwin, Kamran Ghoreschi, Susan L. Gillespie, Jörg J. Goronzy, Clive E.H. Grattan, Neil S. Greenspan, Eyal Grunebaum, Gabrielle Haeberli, Russell P. Hall, Robert G. Hamilton, Gregory R. Harriman, Sarfaraz A. Hasni, Arthur Helbling, Melanie Hingorani, Steven M. Holland, Petr L. Hruz, Gabor Illei, John B. Imboden, Shai Izraeli, Elaine S. Jaffe, Caroline Jagobi, Sirpa Jalkanen, Pim Jetanalin, Emmanuelle Jouanguy, Carl H. June, Axel Kallies, Stefan H.E. Kaufmann, Arthur Kavanaugh, Sabiha Khan, Farrah Kheradmand, Samia J. Khoury, Gary A. Koretzky, Robert Korngold, Anna Kovalszki, Douglas B. Kuhns, Robert A. Kyle, Ian R. Lanza, Arian Laurence, Susan J. Lee, Michael J. Lenardo, Arnold I. Levinson, Ofer Levy, David B. Lewis, Dorothy E. Lewis, Sue L. Lightman, Michael D. Lockshin, Michael T. Lotze, Amber Luong, Meggan Mackay, Jean-Luc Malo, Jonathan S. Maltzman, Peter J. Mannon, Michael P. Manns, Mary Louise Markert, Elizabeth A. McCarthy, Douglas R. McDonald, Jerry R. McGhee, Peter C. Melby, Dean D. Metcalfe, Martin Metz, Stephen D. Miller, Anna L. Mitchell, Shruti Mittal, Makoto Miyara, Carolyn Mold, David R. Moller, Scott N. Mueller, Ulrich R. Müller, Philip M. Murphy, Pierre Noel, Luigi Notarangelo, Thomas B. Nutman, Stephen L. Nutt, João B. Oliveira, Chris M. Olson, John J. O'Shea, Sung-Yun Pai, Lavannya Pandit, Mary E. Paul, Simon H.S. Pearce, Erik J. Peterson, Capucine Picard, Werner J. Pichler, Stefania Pittaluga, Anne Puel, Andreas Radbruch, Stephen T. Reece, John D. Reveille, Robert R. Rich, Christine Rivat, Bruce W.S. Robinson, John R. Rodgers, Chaim M. Roifman, Antony Rosen, James T. Rosenbaum, Barry T. Rouse, Scott D. Rowley, Shimon Sakaguchi, Marko Salmi, Harry W. Schroeder, Markus J.H. Seibel, Carlo Selmi, William M. Shafer, Prediman K. Shah, Sushma Shankar, Alan R. Shaw, William T. Shearer, Javed Sheikh, Richard Siegel, Anna Simon, Philip L. Simonian, Gideon P. Smith, Justine R. Smith, Andrew L. Snow, David S. Stephens, John H. Stone, Alex Straumann, Helen C. Su, Louise Swainson, Ewa Szymanska-Mroczek, Naomi Taylor, Adrian J. Thrasher, Laura Timares, Raul M. Torres, Gülbŭ Uzel, Jos W.M. van der Meer, Jeroen C.H. van der Hilst, John Varga, Meryl Waldman, Peter Weiser, Peter F. Weller, Cornelia M. Weyand, Theresa L. Whiteside, Fredrick M. Wigley, Robert J. Winchester, Kajsa Wing, Kathryn Wood, Hui Xu, Shen-Ying Zhang, and Valérie S. Zimmermann
- Published
- 2013
- Full Text
- View/download PDF
35. Vertical Transmission of HIV-1. Correlation with maternal viral load and plasma levels of CD4 binding site anti-gp120 antibodies
- Author
-
Ruth Tuomala, Y F Khouri, M Pagano, Mitchell C. Posner, Lisa A. Cavacini, Kenneth McIntosh, and Wayne A. Marasco
- Subjects
Adult ,medicine.drug_class ,HIV Antibodies ,HIV Envelope Protein gp120 ,V3 loop ,Biology ,Monoclonal antibody ,law.invention ,Pregnancy ,law ,HIV Seropositivity ,medicine ,Humans ,Pregnancy Complications, Infectious ,Binding site ,chemistry.chemical_classification ,Acquired Immunodeficiency Syndrome ,Analysis of Variance ,virus diseases ,General Medicine ,medicine.disease ,Virology ,Infectious Disease Transmission, Vertical ,CD4 Lymphocyte Count ,Transmission (mechanics) ,chemistry ,Immunology ,HIV-1 ,biology.protein ,Female ,Antibody ,Glycoprotein ,Viral load ,Biomarkers ,Research Article - Abstract
Almost all childhood HIV-1 is now acquired through vertical transmission. Identifying factors that affect the rate of transmission may lead to the initiation of specific preventive strategies. In this study, antibody levels against different neutralizing epitopes on the envelope glycoprotein of HIV-1 (gp120) were measured in HIV-1-infected pregnant women that either transmitted HIV-1 to their infants (18 women) or did not (29 women). Differences in levels of antibodies directed against the monomeric gp120 molecule and against the V3 loop region of gp120 were not significantly different between the two groups studied. However, significant differences were observed in the levels of CD4 binding site antibodies, as determined by the ability of diluted maternal plasma to inhibit binding of the CD4 binding site monoclonal antibody F105 (mAb F105) to monomeric gp120. In addition, more nontransmitting mothers had low viral load as defined by having two or more negative HIV-1 viral cultures during pregnancy compared with transmitters. This pilot study suggests that in addition to higher viral load, low levels of CD4 binding site antibodies correlate with increased risk of HIV-1 vertical transmission. Passive immunotherapy with broadly neutralizing CD4 binding site antibodies should be considered as a strategy to reduce this risk.
- Published
- 1995
- Full Text
- View/download PDF
36. Impact of IgA constant domain on HIV-1 neutralizing function of monoclonal antibody F425-A1g8
- Author
-
Lisa A. Cavacini, Mark Duval, Christopher J. Lewis, and Xiaocong Yu
- Subjects
lcsh:Immunologic diseases. Allergy ,biology ,medicine.drug_class ,business.industry ,Constant domain ,Human immunodeficiency virus (HIV) ,Monoclonal antibody ,medicine.disease_cause ,Virology ,Infectious Diseases ,Protein structure ,Poster Presentation ,medicine ,biology.protein ,Antibody ,lcsh:RC581-607 ,business ,Function (biology) - Published
- 2012
- Full Text
- View/download PDF
37. Plasma Pharmacokinetics and Biological Activity of a Human Immunodeficiency Virus Type 1 Neutralizing Human Monoclonal Antibody, F105, in Cynomolgus Monkeys
- Author
-
Lisa A. Cavacini, Marshall R. Posner, Kenneth Mace, George Treacy, Jennifer Power, and Emes Cl
- Subjects
Male ,Cancer Research ,medicine.drug_class ,Immunology ,Enzyme-Linked Immunosorbent Assay ,HIV Infections ,HIV Antibodies ,Monoclonal antibody ,Bolus (medicine) ,Pharmacokinetics ,Neutralization Tests ,In vivo ,medicine ,Animals ,Humans ,Immunology and Allergy ,Pharmacology ,Volume of distribution ,biology ,Antibodies, Monoclonal ,Biological activity ,Molecular biology ,Macaca fascicularis ,Titer ,HIV-1 ,biology.protein ,Female ,Antibody - Abstract
The IgG1 kappa human monoclonal antibody (HMab), F105 reacts with a discontinuous epitope on the CD4 binding site (CD4BS) of human immunodeficiency virus type 1 (HIV-1)/gp120 and has broad neutralizing activity. F105 HMab (60 mg/kg bolus) was administered intravenously to four monkeys and serum was collected at intervals to determine pharmacokinetics in a primate model. Average serum F105 concentrations, as determined by enzyme-linked immunosorbent assay, were analyzed with MINSQ software using a two-compartment, first-order model. The half-life for the alpha phase of the distribution curve is 6.7 h and for the beta elimination phase, 9.6 days. The volume of distribution is 0.65 L/kg and the rate of clearance 2 ml/kg/h. Serum levels of 1.3-1.6 mg/ml of F105 were maintained for 24 h. When monkey serum from day 15 postdose was tested, total serum F105 was 230 +/- 79 micrograms/ml and was immunoreactive with cells infected with the MN and IIIB strains of HIV-1 as determined by flow cytometry. Binding activity was identical to that obtained with stock F105 HMab. Identical neutralizing activity between the injected and uninjected antibody was also observed. Thus, serum neutralizing titers (90%) of 1:2000 at peak and 1:30 at day 15 postdose for MN virus were observed. These data indicate that high in vivo levels of HMab F105 can be attained by single bolus administration with full retention of biological activity. Of importance, levels of antibody necessary for effective neutralization can be achieved and maintained.
- Published
- 1994
- Full Text
- View/download PDF
38. Synergistic Inhibition of HIV-1 by CD4 Binding Domain Reagents and V3-Directed Monoclonal Antibodies
- Author
-
Yaming Wu, Lisa A. Cavacini, Keith G. Field, Barbara J. Potts, Mary E. White-Scharf, and Marshall R. Posner
- Subjects
medicine.drug_class ,T-Lymphocytes ,HIV Envelope Protein gp120 ,Monoclonal antibody ,Neutralization ,Virus ,Cell Line ,Mice ,Neutralization Tests ,Viral entry ,Virology ,medicine ,Animals ,Humans ,Lymphocytes ,Binding site ,Binding Sites ,biology ,Antibodies, Monoclonal ,Flow Cytometry ,Molecular biology ,Recombinant Proteins ,Kinetics ,CD4 Antigens ,HIV-1 ,biology.protein ,Antibody ,Viral load ,Binding domain - Abstract
Human immunodeficiency virus (HIV) binds to the surface of CD4 positive lymphocytes and monocyte/macrophages via a high affinity interaction between CD4 and the HIV envelope glycoprotein gp120 and is internalized by fusion of the virus and the cell membrane. The third variable (V3) domain of gp120 also plays a central role in the fusion and viral entry process. Reagents that neutralize HIV by binding to the CD4 binding domain or V3 domain of gp120 have been proposed as therapeutics in the post-HIV exposure and perinatal setting. However, the neutralization potency of these proposed reagents, sCD4, V3 directed and CD4 binding domain directed monoclonal antibodies (MAbs), is intermediate at best and may be of limited use in a clinical setting. We have demonstrated that the combination of reagents to these two primary targets for neutralization resulted in synergy with 10- to 1000-fold increase in virus neutralization. The addition of these combined reagents a second time 3 and 5 days postinfection resulted in an additional 10-fold increase in neutralization suggesting a block in HIV spread from cell to cell. These data suggest that a combination of CD4 binding domain reagents and V3 antibody infused at intervals may significantly reduce the viral load in AIDS patients.
- Published
- 1993
- Full Text
- View/download PDF
39. Characterization of the cDNA of a broadly reactive neutralizing human anti-gp120 monoclonal antibody
- Author
-
Wayne A. Marasco, C Zani, William A. Haseltine, Mitchell C. Posner, Lisa A. Cavacini, Jessamyn Bagley, and Joseph Sodroski
- Subjects
Molecular Sequence Data ,Immunoglobulin Variable Region ,HIV Envelope Protein gp120 ,Immunoglobulin light chain ,Germline ,Immunoglobulin kappa-Chains ,Immunoglobulin Idiotypes ,Complementary DNA ,Humans ,Gene family ,Amino Acid Sequence ,Gene ,Gene Rearrangement ,Base Sequence ,Genes, Immunoglobulin ,biology ,Point mutation ,Antibodies, Monoclonal ,Idiotopes ,DNA ,General Medicine ,Molecular biology ,biology.protein ,Antibody ,Immunoglobulin Heavy Chains ,Research Article - Abstract
The F105 mAb, identified in an HIV-1-infected individual, binds to a discontinuous epitope on the HIV-1 gp120 envelope glycoprotein, blocks the binding of gp120 to the CD4 viral receptor, and neutralizes a broad range of HIV-1 isolates. This study reports the primary nucleotide and deduced amino acid sequences of the rearranged heavy and light chains of the mAb F105. This IgG1k mAb uses a VH gene member of the VH4 gene family (V71-4) and is productively rearranged with a D-D fusion product of the dlr4 and da4 germline DH genes and the JH5 gene. This rearranged heavy chain gene expresses the VH4-HV2a idiotope, which is seen in human monoclonal IgM cold agglutinins. The F105 Vk appears to be derived from the Humvk325 germline gene and is rearranged with a Jk2 gene. For both chains, the mutational pattern in the rearranged VH and VL genes is indicative of an antigen-driven process. These studies show that production of a broadly neutralizing anti-HIV-1 antibody that recognizes determinants within the CD4 recognition site of the envelope glycoprotein is achieved by rearrangement of the V71-4 and Humvk325 germline variable region genes along with selected individual point mutations in the rearranged genes.
- Published
- 1992
- Full Text
- View/download PDF
40. Structural basis of immune evasion at the site of CD4 attachment on HIV-1 gp120
- Author
-
Tongqing Zhou, John R. Mascola, Ling Xu, Lisa A. Cavacini, James Arthos, Zhi Yong Yang, Ann J. Hessell, Marie Pancera, Mei-Yun Zhang, Dennis R. Burton, Dimiter S. Dimitrov, Richard T. Wyatt, Joseph Sodroski, Lei Chen, Sijy O'Dell, Min Tang, Gary J. Nabel, Xueling Wu, Young Do Kwon, Peter D. Kwong, and Marshall R. Posner
- Subjects
Models, Molecular ,Protein Conformation ,Molecular Sequence Data ,HIV Antibodies ,HIV Envelope Protein gp120 ,Crystallography, X-Ray ,Epitope ,Virus ,Article ,Epitopes ,Protein structure ,Immune system ,Antigens, CD4 - chemistry - metabolism ,Humans ,Amino Acid Sequence ,Binding site ,Receptor ,Immune Evasion ,Multidisciplinary ,Binding Sites ,biology ,virus diseases ,Antibodies, Neutralizing - chemistry - immunology - metabolism ,HIV Antibodies - chemistry - immunology - metabolism ,biology.organism_classification ,HIV Envelope Protein gp120 - chemistry - immunology - metabolism ,Virology ,Antibodies, Neutralizing ,Peptide Fragments ,Lentivirus ,CD4 Antigens ,biology.protein ,HIV-1 ,Binding Sites, Antibody ,Antibody ,Hydrophobic and Hydrophilic Interactions - Abstract
The site on HIV-1 gp120 that binds to the CD4 receptor is vulnerable to antibodies. However, most antibodies that interact with this site cannot neutralize HIV-1. To understand the basis of this resistance, we determined co-crystal structures for two poorly neutralizing, CD4-binding site (CD4BS) antibodies, F105 and b13, in complexes with gp120. Both antibodies exhibited approach angles to gp120 similar to those of CD4 and a rare, broadly neutralizing CD4BS antibody, b12. Slight differences in recognition, however, resulted in substantial differences in F105- and b13-bound conformations relative to b12-bound gp120. Modeling and binding experiments revealed these conformations to be poorly compatible with the viral spike. This incompatibility, the consequence of slight differences in CD4BS recognition, renders HIV-1 resistant to all but the most accurately targeted antibodies., link_to_OA_fulltext
- Published
- 2009
41. Cell-mediated immunity to the asexual blood stages of malarial parasites: Animal models
- Author
-
Johanne Melancon-Kaplan, William P. Weidanz, and Lisa A. Cavacini
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Erythrocytes ,CD8 Antigens ,T-Lymphocytes ,T cell ,Immunology ,Parasitemia ,Biology ,Immunotherapy, Adoptive ,Cell Line ,Interleukin 21 ,Immune system ,Antigen ,Antigens, CD ,Recurrence ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,B-Lymphocytes ,Immunity, Cellular ,T lymphocyte ,medicine.disease ,Natural killer T cell ,Clone Cells ,Malaria ,Disease Models, Animal ,medicine.anatomical_structure ,Spleen - Abstract
Studies using experimental models of malaria in immunodeficient mice and chickens have shown that resistance to blood-stage infection is mediated by protective antibodies and T cell-dependent cell-mediated mechanisms of immunity. Depending on the infecting species of Plasmodium and prior experience of the host, either humoral or cell-mediated immune mechanisms predominate. Cell-mediated immunity has been adoptively transferred with CD4 + splenic T cells, and with antigen-specific T cell lines and clones. Since ascending parasitemia occurs in all instances, the transferred cells do not kill plasmodia directly but appear to activate effector mechanisms capable of destroying the invading parasites. Both CD4 + and CD8 + T cells were found to increase in the spleens of malarious mice. Depletion of CD4 + T cells prevented nude mice adoptively transferred with immune splenic T cells from clearing parasitemia. In contrast, late treatment with anti-CD4 antibody had little if any effect. The converse was true when anti-CD8 antibody was utilized, i.e., a significant number of mice treated with anti-CD8 antibody after parasitemia became patent and had difficulty clearing blood parasites. These data suggest that during infection CD8 + T cells may become activated by CD4 + T cells responding to malarial antigens. These CD8 + T cells may be directly cytotoxic or secrete additional cytokines thereby amplifying the role of CD4 + T cells in the activation of anti-parasite effector mechanisms. Finally, a hypothesis is presented to explain how the parasite in natural infections may activate T cell-dependent effector mechanisms in order to control its numbers in host tissues thereby ensuring the survival of both parasite and host.
- Published
- 1990
- Full Text
- View/download PDF
42. The neutralization properties of a HIV-specific antibody are markedly altered by glycosylation events outside the antigen-binding domain
- Author
-
Heather Doherty, Luis R. Miranda, Mark Duval, Marshall R. Posner, Michael S. Seaman, and Lisa A. Cavacini
- Subjects
Glycosylation ,Anti-HIV Agents ,Immunology ,Immunoglobulin Variable Region ,CHO Cells ,HIV Antibodies ,Gp41 ,Neutralization ,Epitope ,Cell Line ,chemistry.chemical_compound ,Cricetulus ,Antibody Specificity ,Neutralization Tests ,Cricetinae ,Sequence Homology, Nucleic Acid ,Immunology and Allergy ,Animals ,Humans ,biology ,Chemistry ,Chinese hamster ovary cell ,Antibodies, Monoclonal ,Acquired immune system ,Virology ,Molecular biology ,Immunoglobulin Class Switching ,HIV Envelope Protein gp41 ,Recombinant Proteins ,Protein Structure, Tertiary ,Cell culture ,biology.protein ,Binding Sites, Antibody ,Antibody ,HeLa Cells - Abstract
Neutralizing Abs constitute a pivotal mechanism of the adaptive immune response against HIV-1 infection. Yet, most of the Abs that appear in the circulation during HIV infection are nonneutralizing. In this study, we report a dramatic change of the neutralizing properties of a human Ab reactive with the nonneutralizing epitope termed cluster I on the HIV-1 transmembrane protein gp41 when the Ab was produced in Chinese hamster ovary (CHO)-K1 cells. Our laboratory has previously reported that the Ab F240, when produced in a hybridoma, is nonneutralizing as assessed by standard neutralization assays. The F240 IgG1 Ab expressed in CHO cells acquired a strong neutralization activity against a broad range of HIV isolates without a change in immunoreactivity. Sequencing of the F240 mRNAs produced in the parental hybridoma and CHO cells revealed identical sequences, suggesting that acquired neutralization resulted from cell-specific posttranslational modifications. We found that the Ab produced by CHO cells is glycosylated to a greater extent than the parental Ab produced by the hybridoma. Moreover, treatment with peptide N-glycosidase F abrogated F240 neutralization, in an isolate-specific manner, but not Ab b12 neutralization. Interestingly, the F240 isotype-switched variants IgG3 and IgG4, also expressed in CHO cells, exhibited identical immunoreactivity to IgG1 isotypes but had clear differences in viral neutralization. These results suggest that structural features of the Ig molecule other than the primary sequence of the variable regions play a more prominent role in HIV neutralization than anticipated.
- Published
- 2007
43. Analysis of the neutralization breadth of the anti-V3 antibody F425-B4e8 and re-assessment of its epitope fine specificity by scanning mutagenesis
- Author
-
Rowena O. Aguilar-Sino, Dennis R. Burton, Terri Wrin, Lisa A. Cavacini, and Ralph Pantophlet
- Subjects
Models, Molecular ,medicine.drug_class ,Cross-clade neutralization ,Molecular Sequence Data ,Mutagenesis (molecular biology technique) ,Cross Reactions ,HIV Antibodies ,HIV Envelope Protein gp120 ,In Vitro Techniques ,Monoclonal antibody ,Epitope ,Neutralization ,Virus ,Article ,Cell Line ,Epitopes ,Antigen ,Neutralization Tests ,Virology ,medicine ,Humans ,Amino Acid Sequence ,V3 antibody ,biology ,Virulence ,Antibodies, Monoclonal ,Peptide Fragments ,F425-B4e8 ,Epitope mapping ,Scanning mutagenesis ,Amino Acid Substitution ,biology.protein ,HIV-1 ,Mutagenesis, Site-Directed ,Antibody ,Epitope Mapping - Abstract
The identification of cross-neutralizing antibodies to HIV-1 is important for designing antigens aimed at eliciting similar antibodies upon immunization. The monoclonal antibody (mAb) F425-B4e8 had been suggested previously to bind an epitope at the base of V3 and shown to neutralize two primary HIV isolates. Here, we have assessed the neutralization breadth of mAb F425-B4e8 using a 40-member panel of primary HIV-1 and determined the epitope specificity of the mAb. The antibody was able to neutralize 8 clade B viruses (n = 16), 1 clade C virus (n = 11), and 2 clade D viruses (n = 6), thus placing it among the more broadly neutralizing anti-V3 antibodies described so far. Contrary to an initial report, results from our scanning mutagenesis of the V3 region suggest that mAb F425-B4e8 interacts primarily with the crown/tip of V3, notably Ile309, Arg315, and Phe317. Despite the somewhat limited neutralization breadth of mAb F425-B4e8, the results presented here, along with analyses from other cross-neutralizing anti-V3 mAbs, may facilitate the template-based design of antigens that target V3 and permit neutralization of HIV-1 strains in which the V3 region is accessible to antibodies.
- Published
- 2007
44. Treatment of pemphigus vulgaris with rituximab and intravenous immune globulin
- Author
-
A. Razzaque Ahmed, Zachary Spigelman, Lisa A. Cavacini, and Marshall R. Posner
- Subjects
Adult ,Keratinocytes ,Male ,Adolescent ,medicine.medical_treatment ,Mucocutaneous zone ,Antibodies, Monoclonal, Murine-Derived ,Prednisone ,Recurrence ,Immunopathology ,medicine ,Humans ,Immunologic Factors ,Lymphocyte Count ,Aged ,Autoantibodies ,Autoimmune disease ,B-Lymphocytes ,biology ,business.industry ,Pemphigus vulgaris ,Remission Induction ,Antibodies, Monoclonal ,Immunoglobulins, Intravenous ,General Medicine ,Immunotherapy ,Middle Aged ,medicine.disease ,Immunology ,biology.protein ,Rituximab ,Drug Therapy, Combination ,Female ,Antibody ,business ,Desmogleins ,Pemphigus ,medicine.drug ,Follow-Up Studies - Abstract
BACKGROUND Pemphigus vulgaris is a potentially fatal autoimmune mucocutaneous blistering disease. Conventional therapy consists of high-dose corticosteroids, immunosuppressive agents, and intravenous immune globulin. METHODS We studied patients with refractory pemphigus vulgaris involving 30% or more of their body-surface area, three or more mucosal sites, or both who had inadequate responses to conventional therapy and intravenous immune globulin. We treated the patients with two cycles of rituximab (375 mg per square meter of body-surface area) once weekly for 3 weeks and intravenous immune globulin (2 g per kilogram of body weight) in the fourth week. This induction therapy was followed by a monthly infusion of rituximab and intravenous immune globulin for 4 consecutive months. Titers of serum antibodies against keratinocytes and numbers of peripheral-blood B cells were monitored. RESULTS Of 11 patients, 9 had rapid resolution of lesions and a clinical remission lasting 22 to 37 months (mean, 31.1). All immunosuppressive therapy, including prednisone, could be discontinued before ending rituximab treatment in all patients. Two patients were treated with rituximab only during recurrences and had sustained remissions. Titers of IgG4 antikeratinocyte antibodies correlated with disease activity. Peripheral-blood B cells became undetectable shortly after initiating rituximab therapy but subsequently returned to normal values. Side effects that have been associated with rituximab were not observed, nor were infections. CONCLUSIONS The combination of rituximab and intravenous immune globulin is effective in patients with refractory pemphigus vulgaris.
- Published
- 2006
45. Characterization of the opsonic and protective activity against Staphylococcus aureus of fully human monoclonal antibodies specific for the bacterial surface polysaccharide poly-N-acetylglucosamine
- Author
-
Marshall R. Posner, Lisa A. Cavacini, Gerald B. Pier, Casie Anne Kelly-Quintos, and Donald A. Goldmann
- Subjects
Staphylococcus aureus ,medicine.drug_class ,Immunology ,Molecular Sequence Data ,medicine.disease_cause ,Monoclonal antibody ,Microbiology ,Epitope ,Acetylglucosamine ,Epitopes ,Mice ,Immune system ,Antigen ,Phagocytosis ,medicine ,Animals ,Humans ,Opsonin ,biology ,Base Sequence ,Antibodies, Monoclonal ,Complement C3 ,Staphylococcal Infections ,Antibodies, Bacterial ,Infectious Diseases ,Microbial Immunity and Vaccines ,biology.protein ,Hybridoma technology ,Parasitology ,Female ,Antibody - Abstract
Carbohydrate antigens are important targets of the immune system in clearing bacterial pathogens. Although the immune system almost exclusively uses antibodies in response to foreign carbohydrates, there is still much to learn about the role of different epitopes on the carbohydrate as targets of protective immunity. We examined the role of acetyl group-dependent and -independent epitopes on the staphylococcal surface of polysaccharide poly- N -acetylated glucosamine (PNAG) by use of human monoclonal antibodies (MAbs) specific for such epitopes. We utilized hybridoma technology to produce fully human immunoglobulin G2 (IgG2) MAbs from B cells of an individual post- Staphylococcus aureus infection and cloned the antibody variable regions to produce an IgG1 form of each original MAb. Specificity and functionality of the purified MAbs were tested in vitro using enzyme-linked immunosorbent assays, complement deposition, and opsonophagocytic assays. We found that a MAb (MAb F598) that bound the best to nonacetylated or backbone epitopes on PNAG had superior complement deposition and opsonophagocytic activity compared to two MAbs that bound optimally to PNAG that was expressed with a native level (>90%) of N -acetyl groups (MAbs F628 and F630). Protection of mice against lethality due to S. aureus strains Mn8 and Reynolds further showed that the backbone-specific MAb had optimal protective efficacy compared with the acetate-specific MAbs. These results provide evidence for the importance of epitope specificity in inducing the optimal protective antibody response to PNAG and indicate that MAbs to the deacetylated form of PNAG could be immunotherapeutic agents for preventing or treating staphylococcal infections.
- Published
- 2006
46. Human single-chain antibodies inhibit replication of human immunodeficiency virus type 1 (HIV-1)
- Author
-
Mark Duval, Lisa A. Cavacini, Richard P. Junghans, Fangbing Liu, David Kuhrt, Mukesh Kumar, Qiangzhong Ma, and Marshall R. Posner
- Subjects
Signal peptide ,medicine.drug_class ,Immunology ,Immunoglobulin Variable Region ,chemical and pharmacologic phenomena ,Immunoglobulin light chain ,Monoclonal antibody ,Gp41 ,Virus Replication ,Epitope ,Mice ,Neutralization Tests ,Virology ,medicine ,Animals ,Humans ,DNA Primers ,Expression vector ,biology ,Base Sequence ,Antibodies, Monoclonal ,respiratory system ,Molecular biology ,HIV Envelope Protein gp41 ,Infectious Diseases ,biology.protein ,HIV-1 ,NIH 3T3 Cells ,Antibody ,Single-Chain Antibodies - Abstract
The F240 human monoclonal antibody specifically recognizes the disulfide loop-bonded immunodominant epitope of gp41 spanning residues 592-606 and expressed broadly on HIV-1 primary isolates. Despite broad reactivity with native virions and HIV-infected cells, the antibody fails to neutralize infection. However, cytoplasmic expression of single-chain antibody (scFv) directed against gp41 of HIV-1 provides a rationale means to inhibit the maturation of envelope protein. The variable regions of the heavy chain and light chain of human monoclonal antibody were amplified by PCR and linked by a 15 amino acid (GGSGS)3 linker in an orientation of VL-linker-VH and retroviral expression vectors were constructed to simultaneously express F240 scFv and eGFP to facilitate selection of scFv-producing cells. Incorporation of a human immunoglobulin signal sequence directed secretion of the F240 scFv (s-scFv) while an otherwise identical vector lacked this sequence (scFv) resulting in intracellular expression of scFv. Transduced human CD4+ H9 T cells were challenged with HIV. While both secreted and nonsecreted F240 scFv inhibited viral production, secretory F240 scFv was more potent. Thus, this novel approach to direct expression of a nonneutralizing scFv using the Ig signal sequence suggests that targeted therapy using antibodies to conserved, highly expressed epitopes may result in a decrease in viral production due to a reduction of viral assembly and/or transport and expression.
- Published
- 2005
47. Native HIV type 1 virion surface structures: relationships between antibody binding and neutralization or lessons from the viral capture assay
- Author
-
Marshall R. Posner and Lisa A. Cavacini
- Subjects
AIDS Vaccines ,biology ,Immunology ,Antibody Affinity ,virus diseases ,HIV Infections ,HIV Antibodies ,biology.organism_classification ,Virology ,Epitope ,Neutralization ,Virus ,Epitopes ,Infectious Diseases ,Neutralization Tests ,Lentivirus ,biology.protein ,HIV-1 ,Humans ,Viral disease ,Primary isolate ,Antibody - Abstract
Despite a vigorous antibody response following HIV-1 infection, antibodies which neutralize primary isolates tend to be of low titer or sporadic. Similarly, antibodies produced in response to HIV-1 vaccines in human and animals react with HIV but, only on occasion, do these antibodies neutralize primary isolates. The failure of the immune system to respond in an effective manner is related to the inherent structural properties of the HIV-1 envelope expressed on the native virion and the pathogenesis of HIV infection. Identification of effective antibody interactions with HIV, as judged by inhibition of virus, is crucial for the development of broadly effective HIV vaccines and immune therapeutics. It has been proposed that antibodies must bind and neutralize virus to be effective at controlling HIV infection. We propose that this hypothesis may limit the identification of effective antibodies that are desperately needed given the difficulty in preventing and treating HIV. We provide evidence that the viral capture assay (VCA) is an important adjunct to the study of antibody interactions with primary isolate virus. Further, we propose that antibodies that are ineffective in traditional neutralization assays may also be effective at limiting viral spread and preventing viral infection.
- Published
- 2004
48. Serum concentrations of interleukin-8, vascular endothelial growth factor, and epidermal growth factor receptor in patients with squamous cell cancer of the head and neck
- Author
-
Abhay S. Gokhale, Jarrod Faucher, Lori J. Wirth, Melissa Hallar, Marshall R. Posner, Robert I. Haddad, Lisa A. Cavacini, and Linda Weeks
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Pilot Projects ,chemistry.chemical_compound ,Growth factor receptor ,Epidermal growth factor ,Internal medicine ,Biomarkers, Tumor ,Medicine ,Humans ,Interleukin 8 ,Epidermal growth factor receptor ,Aged ,Neoplasm Staging ,biology ,business.industry ,Head and neck cancer ,Interleukin-8 ,Middle Aged ,medicine.disease ,Neoplasm Proteins ,Vascular endothelial growth factor ,ErbB Receptors ,stomatognathic diseases ,Endocrinology ,Cytokine ,Oncology ,chemistry ,Head and Neck Neoplasms ,Cancer research ,biology.protein ,Carcinoma, Squamous Cell ,Biomarker (medicine) ,Female ,Oral Surgery ,Neoplasm Recurrence, Local ,business - Abstract
Squamous cell cancer of the head and neck (SCCHN) is associated with production of pro-inflammatory and pro-angiogenic cytokines. We hypothesized that cytokine serum levels will correlate with tumor volume and aggressiveness. We investigated interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), and epidermal growth factor receptor (EGFR) in SCCHN. The patient population consisted of normal and irradiated controls: patients with newly diagnosed SCCHN, and patients with recurrent or metastatic disease. Pretreatment sera were studied by ELISA. Serum IL-8 levels, as opposed to VEGF or EGFR, were consistently elevated in patients with recurrent or metastatic disease. The differences in mean serum IL-8, compared to controls, were significant (p=0.02). Serum levels of IL-8 are consistently elevated in patients with recurrent or metastatic SCCHN and elevated levels may correlate with advanced or aggressive disease. Further, more intensive, study of IL-8 as a biomarker in SCCHN is warranted.
- Published
- 2004
49. Primary African HIV clade A and D isolates: effective cross-clade neutralization with a quadruple combination of human monoclonal antibodies raised against clade B
- Author
-
Hermann Katinger, Gabriela Stiegler, Alistair Chalmers, Tao Wang, Ting-Chao Chou, Ruth M. Ruprecht, Flavia Ferrantelli, Lisa A. Cavacini, and Moiz Kitabwalla
- Subjects
medicine.drug_class ,Immunology ,Human immunodeficiency virus (HIV) ,HIV Infections ,Cross Reactions ,HIV Antibodies ,medicine.disease_cause ,Monoclonal antibody ,Virus ,Epitope ,Neutralization ,Neutralization Tests ,Virology ,Genotype ,medicine ,Humans ,Clade ,biology ,virus diseases ,Antibodies, Monoclonal ,biology.organism_classification ,Infectious Diseases ,Lentivirus ,Africa ,HIV-1 - Abstract
We investigated the ability of several human neutralizing monoclonal antibodies (nmAbs), originally raised against human immunodeficiency virus (HIV) clade B isolates, to neutralize primary clade A and D isolates as single agents and in combinations. All four primary HIV clade A isolates and five primary HIV clade D isolates tested were neutralized >99% by the quadruple combination of nmAbs IgG1b12, 2G12, 2F5, and 4E10. These mAbs recognize conserved epitopes on HIV-1 envelope (Env), resulting in strong cross-clade neutralization. Previously, we showed synergistic neutralization of primary HIV-1 clade C isolates in vitro by the same nMAb combination. We and others also showed neutralization of primary HIV clade B strains. Together, our data show that the quadruple combination of mAbs effectively neutralized primary HIV clade A, B, C, and D isolates.
- Published
- 2003
50. Evidence of determinant spreading in the antibody responses to prostate cell surface antigens in patients immunized with prostate-specific antigen
- Author
-
Lisa A, Cavacini, Mark, Duval, J Paul, Eder, and Marshall R, Posner
- Subjects
Male ,Epitopes ,Time Factors ,Immunoglobulin G ,Cell Membrane ,Tumor Cells, Cultured ,Humans ,Prostatic Neoplasms ,Prostate-Specific Antigen ,Flow Cytometry ,Cancer Vaccines - Abstract
Prostate cancer consistently remains a difficult clinical problem. The development of novel therapy strategies for effective control and treatment of prostate cancer is essential. The prostate represents a unique site for immunotherapy, in part because prostate-specific immunity would most probably be without significant long-term sequellae. Antibodies and cell-mediated immunity, induced by either active or passive immunization, represent potential means to specifically target prostate tumor cells.The serum IgG response to cell surface antigens expressed on LNCAP [prostate-specific antigen (PSA)-positive] and PC-3 (PSA-negative) were analyzed in individuals with advanced disease receiving vaccinia- or fowlpox-expressed PSA (v-PSA or f-PSA, respectively) by flow cytometry.Sera from all seven patients in a Phase I study of v-PSA, collected prior to the third immunization, reacted with both prostate tumor cell lines. The majority of individuals (n = 12) in a Phase II trial of v-PSA and f-PSA developed sustainable antibody responses to cell surface antigens on the prostate tumor cell lines. The magnitude and kinetics of these responses were dependent on the immunization schedule. Of importance, the baseline serum of only one of nine patients tested had reactivity with nonprostate tumor cell lines. Sera from three normal males also lacked reactivity with prostate tumor cells.PSA vaccine constructs are immunogenic and induce antibody responses to a multitude of surface antigens on prostate tumor cell lines by epitope or determinant spreading after stimulation of the immune system by PSA immunization.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.