50 results on '"Lisa A. Purcell"'
Search Results
2. A system-view of Bordetella pertussis booster vaccine responses in adults primed with whole-cell versus acellular vaccine in infancy
- Author
-
Ricardo da Silva Antunes, Ferran Soldevila, Mikhail Pomaznoy, Mariana Babor, Jason Bennett, Yuan Tian, Natalie Khalil, Yu Qian, Aishwarya Mandava, Richard H. Scheuermann, Mario Cortese, Bali Pulendran, Christopher D. Petro, Adrienne P. Gilkes, Lisa A. Purcell, Alessandro Sette, and Bjoern Peters
- Subjects
Immunology ,Vaccines ,Medicine - Abstract
The increased incidence of whooping cough worldwide suggests that current vaccination against Bordetella pertussis infection has limitations in quality and duration of protection. The resurgence of infection has been linked to the introduction of acellular vaccines (aP), which have an improved safety profile compared with the previously used whole-cell (wP) vaccines. To determine immunological differences between aP and wP priming in infancy, we performed a systems approach of the immune response to booster vaccination. Transcriptomic, proteomic, cytometric, and serologic profiling revealed multiple shared immune responses with different kinetics across cohorts, including an increase of blood monocyte frequencies and strong antigen-specific IgG responses. Additionally, we found a prominent subset of aP-primed individuals (30%) with a strong differential signature, including higher levels of expression for CCL3, NFKBIA, and ICAM1. Contrary to the wP individuals, this subset displayed increased PT-specific IgE responses after boost and higher antigen-specific IgG4 and IgG3 antibodies against FHA and FIM2/3 at baseline and after boost. Overall, the results show that, while broad immune response patterns to Tdap boost overlap between aP- and wP-primed individuals, a subset of aP-primed individuals present a divergent response. These findings provide candidate targets to study the causes and correlates of waning immunity after aP vaccination.
- Published
- 2021
- Full Text
- View/download PDF
3. The psychological processes of classic psychedelics in the treatment of depression: a systematic review protocol
- Author
-
Lauren Johansen, Paul Liknaitzky, Maja Nedeljkovic, Lisa Mastin-Purcell, and Greg Murray
- Subjects
Psychedelic-assisted psychotherapy ,Psilocybin ,LSD ,Depression ,Psychological processes ,Systematic review ,Medicine - Abstract
Abstract Background There is currently renewed interest in the use of psychedelic therapy in the treatment of psychiatric disorders, including depression. The proposed systematic review will aim to identify, evaluate and summarise the psychological processes of change underlying psychedelic therapy for depression in the current literature and consider the implications these processes may have on the psychotherapy component of treatment. Methods Scopus, PsycINFO, PubMed and Web of Science databases will be searched using relevant terms. Studies will be included if they discuss the use of a classic psychedelic to treat depression symptomology in an adult population and report or propose psychological processes responsible for depression symptom change. Two authors will independently screen articles, complete quality assessment tools and conduct data extraction. Empirical and non-empirical research will be extracted and synthesised separately. A narrative synthesis approach will be used to report psychological processes identified in the literature. Discussion This systematic review will be the first to collate available evidence on the psychological processes associated with psychedelic therapy for depression. The preliminary nature of this research field is expected to result in the review having several limitations, namely heterogeneity between studies and the inclusion of limited empirical research. We intend for this review to present the current state of the literature, identify gaps and generate candidate variables that warrant further investigation. Systematic review PROSPERO CRD42020197202
- Published
- 2022
- Full Text
- View/download PDF
4. A pan-influenza antibody inhibiting neuraminidase via receptor mimicry
- Author
-
Corey Momont, Ha V. Dang, Fabrizia Zatta, Kevin Hauser, Caihong Wang, Julia di Iulio, Andrea Minola, Nadine Czudnochowski, Anna De Marco, Kaitlin Branch, David Donermeyer, Siddhant Vyas, Alex Chen, Elena Ferri, Barbara Guarino, Abigail E. Powell, Roberto Spreafico, Samantha S. Yim, Dale R. Balce, Istvan Bartha, Marcel Meury, Tristan I. Croll, David M. Belnap, Michael A. Schmid, William Timothy Schaiff, Jessica L. Miller, Elisabetta Cameroni, Amalio Telenti, Herbert W. Virgin, Laura E. Rosen, Lisa A. Purcell, Antonio Lanzavecchia, Gyorgy Snell, Davide Corti, Matteo Samuele Pizzuto, di Iulio, Julia [0000-0001-9343-127X], Chen, Alex [0000-0003-2620-5066], Spreafico, Roberto [0000-0001-8282-7658], Yim, Samantha S [0009-0007-8567-3501], Belnap, David M [0000-0002-0619-7487], Schmid, Michael A [0000-0002-1137-9322], Telenti, Amalio [0000-0001-6290-7677], Rosen, Laura E [0000-0002-8030-0219], Purcell, Lisa A [0000-0002-9565-2030], Snell, Gyorgy [0000-0003-1475-659X], Corti, Davide [0000-0002-5797-1364], Pizzuto, Matteo Samuele [0000-0001-5776-654X], and Apollo - University of Cambridge Repository
- Subjects
Multidisciplinary ,Influenza A Virus, H3N2 Subtype ,Molecular Mimicry ,Antibodies, Monoclonal ,Hemagglutinins, Viral ,Neuraminidase ,Antibodies, Viral ,Arginine ,Mice ,Influenza B virus ,Orthomyxoviridae Infections ,Antibody Specificity ,Influenza A virus ,Influenza Vaccines ,Catalytic Domain ,Influenza, Human ,Sialic Acids ,Animals ,Humans ,Seasons - Abstract
Rapidly evolving influenza A viruses (IAVs) and influenza B viruses (IBVs) are major causes of recurrent lower respiratory tract infections. Current influenza vaccines elicit antibodies predominantly to the highly variable head region of haemagglutinin and their effectiveness is limited by viral drift1 and suboptimal immune responses2. Here we describe a neuraminidase-targeting monoclonal antibody, FNI9, that potently inhibits the enzymatic activity of all group 1 and group 2 IAVs, as well as Victoria/2/87-like, Yamagata/16/88-like and ancestral IBVs. FNI9 broadly neutralizes seasonal IAVs and IBVs, including the immune-evading H3N2 strains bearing an N-glycan at position 245, and shows synergistic activity when combined with anti-haemagglutinin stem-directed antibodies. Structural analysis reveals that D107 in the FNI9 heavy chain complementarity-determinant region 3 mimics the interaction of the sialic acid carboxyl group with the three highly conserved arginine residues (R118, R292 and R371) of the neuraminidase catalytic site. FNI9 demonstrates potent prophylactic activity against lethal IAV and IBV infections in mice. The unprecedented breadth and potency of the FNI9 monoclonal antibody supports its development for the prevention of influenza illness by seasonal and pandemic viruses.
- Published
- 2023
5. ACE2-binding exposes the SARS-CoV-2 fusion peptide to broadly neutralizing coronavirus antibodies
- Author
-
Jun Siong Low, Josipa Jerak, M. Alejandra Tortorici, Matthew McCallum, Dora Pinto, Antonino Cassotta, Mathilde Foglierini, Federico Mele, Rana Abdelnabi, Birgit Weynand, Julia Noack, Martin Montiel-Ruiz, Siro Bianchi, Fabio Benigni, Nicole Sprugasci, Anshu Joshi, John E. Bowen, Cameron Stewart, Megi Rexhepaj, Alexandra C. Walls, David Jarrossay, Diego Morone, Philipp Paparoditis, Christian Garzoni, Paolo Ferrari, Alessandro Ceschi, Johan Neyts, Lisa A. Purcell, Gyorgy Snell, Davide Corti, Antonio Lanzavecchia, David Veesler, Federica Sallusto, University of Zurich, Low, Jun Siong, Veesler, David, and Sallusto, Federica
- Subjects
1000 Multidisciplinary ,Multidisciplinary ,SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,610 Medicine & health ,Antibodies, Viral ,Antibodies, Neutralizing ,10199 Clinic for Clinical Pharmacology and Toxicology ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,Angiotensin-Converting Enzyme 2 ,Peptides ,Broadly Neutralizing Antibodies ,Protein Binding - Abstract
The coronavirus spike glycoprotein attaches to host receptors and mediates viral fusion. Using a broad screening approach, we isolated seven monoclonal antibodies (mAbs) that bind to all human-infecting coronavirus spike proteins from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune donors. These mAbs recognize the fusion peptide and acquire affinity and breadth through somatic mutations. Despite targeting a conserved motif, only some mAbs show broad neutralizing activity in vitro against alpha- and betacoronaviruses, including animal coronaviruses WIV-1 and PDF-2180. Two selected mAbs also neutralize Omicron BA.1 and BA.2 authentic viruses and reduce viral burden and pathology in vivo. Structural and functional analyses showed that the fusion peptide-specific mAbs bound with different modalities to a cryptic epitope hidden in prefusion stabilized spike, which became exposed upon binding of angiotensin-converting enzyme 2 (ACE2) or ACE2-mimicking mAbs. ispartof: SCIENCE vol:377 issue:6607 pages:735-741 ispartof: location:United States status: published
- Published
- 2022
6. Accelerating antiviral drug discovery: lessons from COVID-19
- Author
-
Annette von Delft, Matthew D. Hall, Ann D. Kwong, Lisa A. Purcell, Kumar Singh Saikatendu, Uli Schmitz, John A. Tallarico, and Alpha A. Lee
- Subjects
Pharmacology ,Drug Discovery ,General Medicine - Published
- 2023
7. Pandemic preparedness strategies must go beyond vaccines
- Author
-
Rajesh Gupta, Lisa A. Purcell, Davide Corti, and Herbert W. Virgin
- Subjects
General Medicine - Abstract
Monoclonal antibodies can fill a critical gap to help stop the next infectious disease outbreak from becoming the next pandemic.
- Published
- 2023
8. Preclinical and clinical experience with dupilumab on the correlates of live attenuated vaccines
- Author
-
Michael E. Wechsler, Adelmir Souza-Machado, Christine Xu, Xuezhou Mao, Upender Kapoor, Faisal A. Khokhar, John T. O’Malley, Christopher D. Petro, Veronica Mas Casullo, Leda P. Mannent, Paul J. Rowe, Juby A. Jacob-Nara, Marcella Ruddy, Elizabeth Laws, Lisa A. Purcell, and Megan Hardin
- Published
- 2022
9. Therapeutic and vaccine-induced cross-reactive antibodies with effector function against emerging Omicron variants
- Author
-
Amin Addetia, Luca Piccoli, James Brett Case, Young-Jun Park, Martina Beltramello, Barbara Guarino, Ha Dang, Dora Pinto, Suzanne M. Scheaffer, Kaitlin Sprouse, Jessica Bassi, Chiara Silacci-Fregni, Francesco Muoio, Marco Dini, Lucia Vincenzetti, Rima Acosta, Daisy Johnson, Sambhavi Subramanian, Christian Saliba, Martina Giurdanella, Gloria Lombardo, Giada Leoni, Katja Culap, Carley McAlister, Anushka Rajesh, Exequiel Dellota, Jiayi Zhou, Nisar Farhat, Dana Bohan, Julia Noack, Florian A. Lempp, Elisabetta Cameroni, Bradley Whitener, Olivier Giannini, Alessandro Ceschi, Paolo Ferrari, Alessandra Franzetti-Pellanda, Maira Biggiogero, Christian Garzoni, Stephanie Zappi, Luca Bernasconi, Min Jeong Kim, Gretja Schnell, Nadine Czudnochowski, Nicholas Franko, Jennifer K. Logue, Courtney Yoshiyama, Cameron Stewart, Helen Chu, Michael A. Schmid, Lisa A. Purcell, Gyorgy Snell, Antonio Lanzavecchia, Michael S. Diamond, Davide Corti, and David Veesler
- Subjects
Article - Abstract
Currently circulating SARS-CoV-2 variants acquired convergent mutations at receptor-binding domain (RBD) hot spots1. Their impact on viral infection, transmission, and efficacy of vaccines and therapeutics remains poorly understood. Here, we demonstrate that recently emerged BQ.1.1. and XBB.1 variants bind ACE2 with high affinity and promote membrane fusion more efficiently than earlier Omicron variants. Structures of the BQ.1.1 and XBB.1 RBDs bound to human ACE2 and S309 Fab (sotrovimab parent) explain the altered ACE2 recognition and preserved antibody binding through conformational selection. We show that sotrovimab binds avidly to all Omicron variants, promotes Fc-dependent effector functions and protects mice challenged with BQ.1.1, the variant displaying the greatest loss of neutralization. Moreover, in several donors vaccine-elicited plasma antibodies cross-react with and trigger effector functions against Omicron variants despite reduced neutralizing activity. Cross-reactive RBD-directed human memory B cells remained dominant even after two exposures to Omicron spikes, underscoring persistent immune imprinting. Our findings suggest that this previously overlooked class of cross-reactive antibodies, exemplified by S309, may contribute to protection against disease caused by emerging variants through elicitation of effector functions.
- Published
- 2023
10. Imprinted antibody responses against SARS-CoV-2 Omicron sublineages
- Author
-
Young-Jun Park, Dora Pinto, Alexandra C. Walls, Zhuoming Liu, Anna De Marco, Fabio Benigni, Fabrizia Zatta, Chiara Silacci-Fregni, Jessica Bassi, Kaitlin R. Sprouse, Amin Addetia, John E. Bowen, Cameron Stewart, Martina Giurdanella, Christian Saliba, Barbara Guarino, Michael A. Schmid, Nicholas M. Franko, Jennifer K. Logue, Ha V. Dang, Kevin Hauser, Julia di Iulio, William Rivera, Gretja Schnell, Anushka Rajesh, Jiayi Zhou, Nisar Farhat, Hannah Kaiser, Martin Montiel-Ruiz, Julia Noack, Florian A. Lempp, Javier Janer, Rana Abdelnabi, Piet Maes, Paolo Ferrari, Alessandro Ceschi, Olivier Giannini, Guilherme Dias de Melo, Lauriane Kergoat, Hervé Bourhy, Johan Neyts, Leah Soriaga, Lisa A. Purcell, Gyorgy Snell, Sean P. J. Whelan, Antonio Lanzavecchia, Herbert W. Virgin, Luca Piccoli, Helen Y. Chu, Matteo Samuele Pizzuto, Davide Corti, David Veesler, University of Washington [Seattle], Humabs BioMed SA, Washington University School of Medicine [Saint Louis, MO], Vir Biotechnology Inc [San Francisco], Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), Università della Svizzera italiana = University of Italian Switzerland (USI), Lugano Regional Hospital [Lugano], University of New South Wales [Sydney] (UNSW), University hospital of Zurich [Zurich], Bellinzona Regional Hospital [Bellinzona], Lyssavirus, épidémiologie et neuropathologie - Lyssavirus Epidemiology and Neuropathology, Institut Pasteur [Paris] (IP)-Université Paris Cité (UPCité), University of Texas Southwestern Medical Center [Dallas], and This study was supported by the National Institute of Allergy and Infectious Diseases (DP1AI158186 and HHSN272201700059C to D.V.), a Pew Biomedical Scholars Award (D.V.), an Investigators in the Pathogenesis of Infectious Disease Awards from the Burroughs Wellcome Fund (D.V.), Fast Grants (D.V.), the University of Washington Arnold and Mabel Beckman cryoEM center and the National Institute of Health grant S10OD032290 (to D.V.). S.P.J.W. supported be NIH grant AI163019. D.V. is an Investigator of the Howard Hughes Medical Institute. O.G. is funded by the Swiss Kidney Foundation.
- Subjects
Multidisciplinary ,SARS-CoV-2 ,COVID-19 ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Memory B Cells ,Neutralization Tests ,Antibody Formation ,Spike Glycoprotein, Coronavirus ,Humans ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Immunologic Memory ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Immune Evasion - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron sublineages carry distinct spike mutations resulting in escape from antibodies induced by previous infection or vaccination. We show that hybrid immunity or vaccine boosters elicit plasma-neutralizing antibodies against Omicron BA.1, BA.2, BA.2.12.1, and BA.4/5, and that breakthrough infections, but not vaccination alone, induce neutralizing antibodies in the nasal mucosa. Consistent with immunological imprinting, most antibodies derived from memory B cells or plasma cells of Omicron breakthrough cases cross-react with the Wuhan-Hu-1, BA.1, BA.2, and BA.4/5 receptor-binding domains, whereas Omicron primary infections elicit B cells of narrow specificity up to 6 months after infection. Although most clinical antibodies have reduced neutralization of Omicron, we identified an ultrapotent pan-variant-neutralizing antibody that is a strong candidate for clinical development. ispartof: SCIENCE vol:378 issue:6620 pages:619-+ ispartof: location:United States status: published
- Published
- 2022
11. The potent broadly neutralizing antibody VIR-3434 controls Hepatitis B and D Virus infection and reduces HBsAg in humanized mice
- Author
-
Florian A. Lempp, Tassilo Volz, Elisabetta Cameroni, Fabio Benigni, Jiayi Zhou, Laura E. Rosen, Julia Noack, Fabrizia Zatta, Hannah Kaiser, Siro Bianchi, Gloria Lombardo, Stefano Jaconi, Hasan Imam, Leah B. Soriaga, Nadia Passini, David M. Belnap, Andreas Schulze, Marc Lütgehetmann, Amalio Telenti, Andrea L. Cathcart, Gyorgy Snell, Lisa A. Purcell, Christy M. Hebner, Stephan Urban, Maura Dandri, Davide Corti, and Michael A. Schmid
- Abstract
Background & AimsChronic hepatitis B is a major global public health problem, and coinfection with hepatitis delta virus (HDV) worsens disease outcome. Here, we describe a hepatitis B virus (HBV) surface antigen (HBsAg)-targeting monoclonal antibody (mAb) with the potential to promote functional cure of chronic hepatitis B and D to address this unmet medical need.MethodsHBsAg-specific mAbs were isolated from memory B cells of HBV vaccinated individuals. In vitro neutralization was determined against HBV and HDV enveloped with HBsAg representing eight HBV genotypes. Human liver-chimeric mice were treated twice weekly with a candidate mAb starting three weeks post HBV inoculation (spreading phase) or during stable HBV or HBV/HDV coinfection (chronic phase).ResultsFrom a panel of human anti-HBs mAbs, VIR-3434 was selected and engineered for pre-clinical development. VIR-3434 targets a putative conserved, conformational epitope within the antigenic loop of HBsAg and neutralized HBV and HDV infection with >12,000-fold higher potency than Hepatitis B Immunoglobulins in vitro. Neutralization was pan-genotypic against strains representative of HBV genotypes A-H. In the spreading phase of HBV infection in human liver-chimeric mice, a parental mAb of VIR-3434 (HBC34) prevented HBV dissemination and intrahepatic HBV RNA and cccDNA increase. In the chronic phase of HBV infection or co-infection with HDV, HBC34 treatment decreased circulating HBsAg by >1 log and HDV RNA by >2 logs.ConclusionsThis in vitro and in vivo characterization identified the potent anti-HBs mAb VIR-3434, which reduces circulating HBsAg and HBV/HDV viremia in human liver-chimeric mice. VIR-3434 is currently in clinical development for treatment of patients with chronic hepatitis B or D.Lay summaryChronic infection with hepatitis B virus places approximately 290 million individuals worldwide at risk for severe liver disease and cancer. Currently available treatments result in low rates of functional cure or require lifelong therapy that does not eliminate the risk of liver disease. We isolated and characterized a potent, human antibody that neutralizes hepatitis B and D viruses and reduces infection in a mouse model. This antibody could provide a new treatment for patients with chronic hepatitis B and D.HighlightsIdentification of a human mAb VIR-3434 that potently neutralizes HBV and HDVVIR-3434 targets a conserved, conformational epitope of the HBsAg antigenic loopVIR-3434 treatment blocks intrahepatic HBV spread in human liver-chimeric miceVIR-3434 treatment reduces circulating HBsAg and HDV RNA in co-infected miceData have enabled clinical development of VIR-3434 against chronic hepatitis B/D
- Published
- 2022
12. Lectins enhance SARS-CoV-2 infection and influence neutralizing antibodies
- Author
-
Antonio Lanzavecchia, Lisa A. Purcell, Young-Jun Park, Nuria Izquierdo-Useros, Siro Bianchi, Stefano Jaconi, Marcel Meury, Maria De Agostini, Exequiel Dellota, Davide Corti, Fabio Benigni, David Veesler, Amalio Telenti, Elisabetta Cameroni, Florian A. Lempp, Herbert W. Virgin, Júlia Vergara-Alert, Martin Montiel-Ruiz, Hannah Kaiser, Jiayi Zhou, Javier Martinez-Picado, Anshu Joshi, Julia Noack, Alexandra C. Walls, Leah Soriaga, and John E. Bowen
- Subjects
chemistry.chemical_classification ,Multidisciplinary ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Cell ,Lectin ,Biology ,Neutralization ,Microbiology ,medicine.anatomical_structure ,Enzyme ,chemistry ,medicine ,biology.protein ,Antibody ,Receptor ,hormones, hormone substitutes, and hormone antagonists ,Function (biology) - Abstract
SARS-CoV-2 infection—which involves both cell attachment and membrane fusion—relies on the angiotensin-converting enzyme 2 (ACE2) receptor, which is paradoxically found at low levels in the respiratory tract1–3, suggesting that there may be additional mechanisms facilitating infection. Here we show that C-type lectin receptors, DC-SIGN, L-SIGN and the sialic acid–binding immunoglobulin-like lectin 1 (SIGLEC1) function as attachment receptors by enhancing ACE2-mediated infection and modulating the neutralizing activity of different classes of spike-specific antibodies. Antibodies to the amino-terminal domain or to the conserved site at the base of the receptor-binding domain, while poorly neutralizing infection of ACE2-overexpressing cells, effectively block lectin-facilitated infection. Conversely, antibodies to the receptor binding motif, while potently neutralizing infection of ACE2-overexpressing cells, poorly neutralize infection of cells expressing DC-SIGN or L-SIGN and trigger fusogenic rearrangement of the spike, promoting cell-to-cell fusion. Collectively, these findings identify a lectin-dependent pathway that enhances ACE2-dependent infection by SARS-CoV-2 and reveal distinct mechanisms of neutralization by different classes of spike-specific antibodies. C-type lectins and SIGLEC1 function as attachment receptors for SARS-CoV-2 and enhance ACE2-mediated infection.
- Published
- 2021
13. ACE2 engagement exposes the fusion peptide to pan-coronavirus neutralizing antibodies
- Author
-
Jun Siong Low, Josipa Jerak, M. Alejandra Tortorici, Matthew McCallum, Dora Pinto, Antonino Cassotta, Mathilde Foglierini, Federico Mele, Rana Abdelnabi, Birgit Weynand, Julia Noack, Martin Montiel-Ruiz, Siro Bianchi, Fabio Benigni, Nicole Sprugasci, Anshu Joshi, John E. Bowen, Alexandra C. Walls, David Jarrossay, Diego Morone, Philipp Paparoditis, Christian Garzoni, Paolo Ferrari, Alessandro Ceschi, Johan Neyts, Lisa A. Purcell, Gyorgy Snell, Davide Corti, Antonio Lanzavecchia, David Veesler, and Federica Sallusto
- Abstract
Coronaviruses use diverse Spike (S) glycoproteins to attach to host receptors and fuse with target cells. Using a broad screening approach, we isolated from SARS-CoV-2 immune donors seven monoclonal antibodies (mAbs) that bind to all human alpha and beta coronavirus S proteins. These mAbs recognize the fusion peptide and acquire high affinity and breadth through somatic mutations. Despite targeting a conserved motif, only some mAbs show broad neutralizing activity in vitro against alpha and beta coronaviruses, including Omicron BA.1 variant and bat WIV-1, and reduce viral titers and pathology in vivo. Structural and functional analyses show that the fusion peptide-specific mAbs bind with different modalities to a cryptic epitope which is concealed by prefusion-stabilizing ‘2P’ mutations and becomes exposed upon binding of ACE2 or ACE2-mimicking mAbs. This study identifies a new class of pan-coronavirus neutralizing mAbs and reveals a receptor-induced conformational change in the S protein that exposes the fusion peptide region.
- Published
- 2022
14. Effect of Subcutaneous Casirivimab and Imdevimab Antibody Combination vs Placebo on Development of Symptomatic COVID-19 in Early Asymptomatic SARS-CoV-2 Infection: A Randomized Clinical Trial
- Author
-
Meagan P, O'Brien, Eduardo, Forleo-Neto, Neena, Sarkar, Flonza, Isa, Peijie, Hou, Kuo-Chen, Chan, Bret J, Musser, Katharine J, Bar, Ruanne V, Barnabas, Dan H, Barouch, Myron S, Cohen, Christopher B, Hurt, Dale R, Burwen, Mary A, Marovich, Elizabeth R, Brown, Ingeborg, Heirman, John D, Davis, Kenneth C, Turner, Divya, Ramesh, Adnan, Mahmood, Andrea T, Hooper, Jennifer D, Hamilton, Yunji, Kim, Lisa A, Purcell, Alina, Baum, Christos A, Kyratsous, James, Krainson, Richard, Perez-Perez, Rizwana, Mohseni, Bari, Kowal, A Thomas, DiCioccio, Gregory P, Geba, Neil, Stahl, Leah, Lipsich, Ned, Braunstein, Gary, Herman, George D, Yancopoulos, David M, Weinreich, and Anya T., Weerasinghe
- Subjects
Adult ,Aged, 80 and over ,Male ,Adolescent ,SARS-CoV-2 ,Incidence ,Injections, Subcutaneous ,COVID-19 ,Correction ,General Medicine ,Middle Aged ,Viral Load ,Antibodies, Monoclonal, Humanized ,COVID-19 Drug Treatment ,Drug Combinations ,Double-Blind Method ,Risk Factors ,COVID-19 Nucleic Acid Testing ,Disease Progression ,Humans ,Female ,Child ,Asymptomatic Infections ,Aged ,Original Investigation - Abstract
IMPORTANCE: Easy-to-administer anti–SARS-CoV-2 treatments may be used to prevent progression from asymptomatic infection to symptomatic disease and to reduce viral carriage. OBJECTIVE: To evaluate the effect of combination subcutaneous casirivimab and imdevimab on progression from early asymptomatic SARS-CoV-2 infection to symptomatic COVID-19. DESIGN, SETTING, AND PARTICIPANTS: Randomized, double-blind, placebo-controlled, phase 3 trial of close household contacts of a SARS-CoV-2–infected index case at 112 sites in the US, Romania, and Moldova enrolled July 13, 2020–January 28, 2021; follow-up ended March 11, 2021. Asymptomatic individuals (aged ≥12 years) were eligible if identified within 96 hours of index case positive test collection. Results from 314 individuals positive on SARS-CoV-2 reverse transcriptase–quantitative polymerase chain reaction (RT-qPCR) testing are reported. INTERVENTIONS: Individuals were randomized 1:1 to receive 1 dose of subcutaneous casirivimab and imdevimab, 1200 mg (600 mg of each; n = 158), or placebo (n = 156). MAIN OUTCOMES AND MEASURES: The primary end point was the proportion of seronegative participants who developed symptomatic COVID-19 during the 28-day efficacy assessment period. The key secondary efficacy end points were the number of weeks of symptomatic SARS-CoV-2 infection and the number of weeks of high viral load (>4 log(10) copies/mL). RESULTS: Among 314 randomized participants (mean age, 41.0 years; 51.6% women), 310 (99.7%) completed the efficacy assessment period; 204 were asymptomatic and seronegative at baseline and included in the primary efficacy analysis. Subcutaneous casirivimab and imdevimab, 1200 mg, significantly prevented progression to symptomatic disease (29/100 [29.0%] vs 44/104 [42.3%] with placebo; odds ratio, 0.54 [95% CI, 0.30-0.97]; P = .04; absolute risk difference, −13.3% [95% CI, −26.3% to −0.3%]). Casirivimab and imdevimab reduced the number of symptomatic weeks per 1000 participants (895.7 weeks vs 1637.4 weeks with placebo; P = .03), an approximately 5.6-day reduction in symptom duration per symptomatic participant. Treatment with casirivimab and imdevimab also reduced the number of high viral load weeks per 1000 participants (489.8 weeks vs 811.9 weeks with placebo; P = .001). The proportion of participants receiving casirivimab and imdevimab who had 1 or more treatment-emergent adverse event was 33.5% vs 48.1% for placebo, including events related (25.8% vs 39.7%) or not related (11.0% vs 16.0%) to COVID-19. CONCLUSIONS AND RELEVANCE: Among asymptomatic SARS-CoV-2 RT-qPCR–positive individuals living with an infected household contact, treatment with subcutaneous casirivimab and imdevimab antibody combination vs placebo significantly reduced the incidence of symptomatic COVID-19 over 28 days. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04452318
- Published
- 2022
15. Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift
- Author
-
Elisabetta Cameroni, Christian Saliba, John E. Bowen, Laura E. Rosen, Katja Culap, Dora Pinto, Laura A. VanBlargan, Anna De Marco, Samantha K. Zepeda, Julia di Iulio, Fabrizia Zatta, Hannah Kaiser, Julia Noack, Nisar Farhat, Nadine Czudnochowski, Colin Havenar-Daughton, Kaitlin R. Sprouse, Josh R. Dillen, Abigail E. Powell, Alex Chen, Cyrus Maher, Li Yin, David Sun, Leah Soriaga, Jessica Bassi, Chiara Silacci-Fregni, Claes Gustafsson, Nicholas M. Franko, Jenni Logue, Najeeha Talat Iqbal, Ignacio Mazzitelli, Jorge Geffner, Renata Grifantini, Helen Chu, Andrea Gori, Agostino Riva, Olivier Giannini, Alessandro Ceschi, Paolo Ferrari, Pietro Cippà, Alessandra Franzetti-Pellanda, Christian Garzoni, Peter J. Halfmann, Yoshihiro Kawaoka, Christy Hebner, Lisa A. Purcell, Luca Piccoli, Matteo Samuele Pizzuto, Alexandra C. Walls, Michael S. Diamond, Amalio Telenti, Herbert W. Virgin, Antonio Lanzavecchia, David Veesler, Gyorgy Snell, and Davide Corti
- Subjects
COVID-19 Vaccines ,Multidisciplinary ,SARS-CoV-2 ,Antibodies, Monoclonal ,Convalescence ,Vesiculovirus ,Antibodies, Monoclonal, Humanized ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Cell Line ,Mice ,Neutralization Tests ,Spike Glycoprotein, Coronavirus ,Animals ,Epitopes, B-Lymphocyte ,Humans ,Angiotensin-Converting Enzyme 2 ,Antigenic Drift and Shift ,Broadly Neutralizing Antibodies ,Immune Evasion - Abstract
SUMMARYThe recently emerged SARS-CoV-2 Omicron variant harbors 37 amino acid substitutions in the spike (S) protein, 15 of which are in the receptor-binding domain (RBD), thereby raising concerns about the effectiveness of available vaccines and antibody therapeutics. Here, we show that the Omicron RBD binds to human ACE2 with enhanced affinity relative to the Wuhan-Hu-1 RBD and acquires binding to mouse ACE2. Severe reductions of plasma neutralizing activity were observed against Omicron compared to the ancestral pseudovirus for vaccinated and convalescent individuals. Most (26 out of 29) receptor-binding motif (RBM)-directed monoclonal antibodies (mAbs) lost in vitro neutralizing activity against Omicron, with only three mAbs, including the ACE2-mimicking S2K146 mAb1, retaining unaltered potency. Furthermore, a fraction of broadly neutralizing sarbecovirus mAbs recognizing antigenic sites outside the RBM, including sotrovimab2, S2X2593 and S2H974, neutralized Omicron. The magnitude of Omicron-mediated immune evasion and the acquisition of binding to mouse ACE2 mark a major SARS-CoV-2 mutational shift. Broadly neutralizing sarbecovirus mAbs recognizing epitopes conserved among SARS-CoV-2 variants and other sarbecoviruses may prove key to controlling the ongoing pandemic and future zoonotic spillovers.
- Published
- 2021
16. An infectious SARS-CoV-2 B.1.1.529 Omicron virus escapes neutralization by several therapeutic monoclonal antibodies
- Author
-
Laura A. VanBlargan, John M. Errico, Peter J. Halfmann, Seth J. Zost, James E. Crowe, Lisa A. Purcell, Yoshihiro Kawaoka, Davide Corti, Daved H. Fremont, and Michael S. Diamond
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global COVID-19 pandemic resulting in millions of deaths worldwide. Despite the development and deployment of highly effective antibody and vaccine countermeasures, rapidly-spreading SARS-CoV-2 variants with mutations at key antigenic sites in the spike protein jeopardize their efficacy. Indeed, the recent emergence of the highly-transmissible B.1.1.529 Omicron variant is especially concerning because of the number of mutations, deletions, and insertions in the spike protein. Here, using a panel of anti-receptor binding domain (RBD) monoclonal antibodies (mAbs) corresponding to those with emergency use authorization (EUA) or in advanced clinical development by Vir Biotechnology (S309, the parent mAbs of VIR-7381), AstraZeneca (COV2-2196 and COV2-2130, the parent mAbs of AZD8895 and AZD1061), Regeneron (REGN10933 and REGN10987), Lilly (LY-CoV555 and LY-CoV016), and Celltrion (CT-P59), we report the impact on neutralization of a prevailing, infectious B.1.1.529 Omicron isolate compared to a historical WA1/2020 D614G strain. Several highly neutralizing mAbs (LY-CoV555, LY-CoV016, REGN10933, REGN10987, and CT-P59) completely lost inhibitory activity against B.1.1.529 virus in both Vero-TMPRSS2 and Vero-hACE2-TMPRSS2 cells, whereas others were reduced (∼12-fold decrease, COV2-2196 and COV2-2130 combination) or minimally affected (S309). Our results suggest that several, but not all, of the antibody products in clinical use will lose efficacy against the B.1.1.529 Omicron variant and related strains.
- Published
- 2021
17. Subcutaneous REGEN-COV Antibody Combination for Covid-19 Prevention
- Author
-
Meagan P, O'Brien, Eduardo, Forleo-Neto, Bret J, Musser, Flonza, Isa, Kuo-Chen, Chan, Neena, Sarkar, Katharine J, Bar, Ruanne V, Barnabas, Dan H, Barouch, Myron S, Cohen, Christopher B, Hurt, Dale R, Burwen, Mary A, Marovich, Peijie, Hou, Ingeborg, Heirman, John D, Davis, Kenneth C, Turner, Divya, Ramesh, Adnan, Mahmood, Andrea T, Hooper, Jennifer D, Hamilton, Yunji, Kim, Lisa A, Purcell, Alina, Baum, Christos A, Kyratsous, James, Krainson, Richard, Perez-Perez, Rizwana, Mohseni, Bari, Kowal, A Thomas, DiCioccio, Neil, Stahl, Leah, Lipsich, Ned, Braunstein, Gary, Herman, George D, Yancopoulos, David M, Weinreich, and Sheryl, Zwerski
- Subjects
Male ,viruses ,Disease ,Subcutaneous injection ,Young adult ,Child ,Aged, 80 and over ,biology ,Incidence ,Incidence (epidemiology) ,virus diseases ,General Medicine ,Middle Aged ,Viral Load ,Drug Combinations ,Female ,Original Article ,medicine.symptom ,Antibody ,Viral load ,Adult ,medicine.medical_specialty ,Adolescent ,Coronavirus disease 2019 (COVID-19) ,medicine.drug_class ,Injections, Subcutaneous ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Placebo ,Asymptomatic ,Article ,Virus ,Young Adult ,Double-Blind Method ,Immunity ,Internal medicine ,medicine ,Humans ,Aged ,Asymptomatic Diseases ,SARS-CoV-2 ,business.industry ,Patient Acuity ,COVID-19 ,biochemical phenomena, metabolism, and nutrition ,Virology ,COVID-19 Drug Treatment ,respiratory tract diseases ,biology.protein ,business - Abstract
Background REGEN-COV (previously known as REGN-COV2), a combination of the monoclonal antibodies casirivimab and imdevimab, has been shown to markedly reduce the risk of hospitalization or death among high-risk persons with coronavirus disease 2019 (Covid-19). Whether subcutaneous REGEN-COV prevents severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and subsequent Covid-19 in persons at high risk for infection because of household exposure to a person with SARS-CoV-2 infection is unknown. Methods We randomly assigned, in a 1:1 ratio, participants (≥12 years of age) who were enrolled within 96 hours after a household contact received a diagnosis of SARS-CoV-2 infection to receive a total dose of 1200 mg of REGEN-COV or matching placebo administered by means of subcutaneous injection. At the time of randomization, participants were stratified according to the results of the local diagnostic assay for SARS-CoV-2 and according to age. The primary efficacy end point was the development of symptomatic SARS-CoV-2 infection through day 28 in participants who did not have SARS-COV-2 infection (as measured by reverse-transcriptase–quantitative polymerase-chain-reaction assay) or previous immunity (seronegativity). Results Symptomatic SARS-CoV-2 infection developed in 11 of 753 participants in the REGEN-COV group (1.5%) and in 59 of 752 participants in the placebo group (7.8%) (relative risk reduction [1 minus the relative risk], 81.4%; P104 copies per milliliter) was shorter (0.4 weeks and 1.3 weeks, respectively). No dose-limiting toxic effects of REGEN-COV were noted. Conclusions Subcutaneous REGEN-COV prevented symptomatic Covid-19 and asymptomatic SARS-CoV-2 infection in previously uninfected household contacts of infected persons. Among the participants who became infected, REGEN-COV reduced the duration of symptomatic disease and the duration of a high viral load. (Funded by Regeneron Pharmaceuticals and others; ClinicalTrials.gov number, NCT04452318.)
- Published
- 2021
18. Subcutaneous REGEN-COV Antibody Combination in Early Asymptomatic SARS-CoV-2 Infection: A Randomized Clinical Trial
- Author
-
Richard Perez-Perez, Gary Herman, Jennifer D. Hamilton, Leah Lipsich, Meagan P. O'Brien, Divya Ramesh, George D. Yancopoulos, John D. Davis, Bari Kowal, James Krainson, Ingeborg Heirman, Alina Baum, Neena Sarkar, Adnan Mahmood, Kenneth C. Turner, Christos A. Kyratsous, Rizwana Mohseni, Ruanne V. Barnabas, Dale R. Burwen, Eduardo Forleo-Neto, Ned Braunstein, Katharine J. Bar, Covid Phase Prevention Trial Team, A. Thomas DiCioccio, Lisa A. Purcell, Flonza Isa, Bret J Musser, David M. Weinreich, Dan H. Barouch, Peijie Hou, Myron S. Cohen, Mary A. Marovich, Kuo-Chen Chan, Neil Stahl, Christopher B. Hurt, Andrea T. Hooper, and Yunji Kim
- Subjects
Relative risk reduction ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Population ,Placebo ,Asymptomatic ,Internal medicine ,Multicenter trial ,medicine ,Clinical endpoint ,medicine.symptom ,Adverse effect ,business ,education ,Viral load - Abstract
ImportanceEasy-to-administer antiviral treatments may be used to prevent progression from asymptomatic infection to COVID-19 and to reduce viral carriage.ObjectiveEvaluate the efficacy and safety of subcutaneous casirivimab and imdevimab antibody combination (REGEN-COV) to prevent progression from early asymptomatic SARS-CoV-2 infection to COVID-19.DesignRandomized, double-blind, placebo-controlled, phase 3 study that enrolled asymptomatic close contacts living with a SARS-CoV-2–infected household member (index case). Participants who were SARS-CoV-2 RT-qPCR–positive at baseline were included in the analysis reported here.SettingMulticenter trial conducted at 112 sites in the United States, Romania, and Moldova.ParticipantsAsymptomatic individuals ≥12 years of age were eligible if identified within 96 hours of collection of the index case’s positive SARS-CoV-2 test sample.InterventionsA total of 314 asymptomatic, SARS-CoV-2 RT-qPCR–positive individuals living with an infected household contact were randomized 1:1 to receive a single dose of subcutaneous REGEN-COV 1200mg (n=158) or placebo (n=156).Main Outcome(s) and Measure(s)The primary endpoint was the proportion of participants who developed symptomatic COVID-19 during the 28-day efficacy assessment period. The key secondary efficacy endpoints were the number of weeks of symptomatic SARS-CoV-2 infection and the number of weeks of high viral load (>4 log10 copies/mL). Safety was assessed in all treated participants.ResultsSubcutaneous REGEN-COV 1200mg significantly prevented progression from asymptomatic to symptomatic disease compared with placebo (31.5% relative risk reduction; 29/100 [29.0%] vs 44/104 [42.3%], respectively; P=.0380). REGEN-COV reduced the overall population burden of high–viral load weeks (39.7% reduction vs placebo; 48 vs 82 total weeks; P=.0010) and of symptomatic weeks (45.3% reduction vs placebo; 89.6 vs 170.3 total weeks; P=.0273), the latter corresponding to an approximately 5.6-day reduction in symptom duration per symptomatic participant. Six placebo-treated participants had a COVID-19–related hospitalization or ER visit versus none for those receiving REGEN-COV. The proportion of participants receiving placebo who had ≥1 treatment-emergent adverse events was 48.1% compared with 33.5% for those receiving REGEN-COV, including events related (39.7% vs 25.8%, respectively) or not related (16.0% vs 11.0%, respectively) to COVID-19.Conclusions and RelevanceSubcutaneous REGEN-COV 1200mg prevented progression from asymptomatic SARS-CoV-2 infection to COVID-19, reduced the duration of high viral load and symptoms, and was well tolerated.Trial RegistrationClinicalTrials.gov Identifier, NCT04452318KEY POINTSQuestionCan treatment with the anti–SARS-CoV-2 antibody combination REGEN-COV prevent COVID-19 and reduce viral load when given to recently exposed and asymptomatic individuals?FindingsIn this randomized, double-blind, phase 3 trial, subcutaneously administered REGEN-COV 1200 mg significantly reduced progression of asymptomatic SARS-CoV-2 infection to symptomatic infection (ie, COVID-19) by 31.5% compared with placebo. REGEN-COV also reduced the overall population burden of high viral load weeks (39.7% reduction vs placebo; 48 vs 82 total weeks; P=.0010).MeaningIn the current pandemic, utilization of subcutaneous REGEN-COV prevents progression of early asymptomatic infection to COVID-19 and reduces viral carriage.
- Published
- 2021
19. VIR-2218 plus VIR-3434 combination therapy reduces hepatitis B virus surface antigen levels in vivo
- Author
-
Julia Noack, Jonathan Gall, Hasan Imam, Yesseinia Anglero-Rodriguez, Vasant Jadhav, Michael A. Schmid, Anna Bakardjiev, Lisa A. Purcell, Christy Hebner, and Florian Lempp
- Subjects
Hepatology - Published
- 2022
20. In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains
- Author
-
Baoling Ying, Pei Yong Shi, Spencer Stumpf, W. Blaine Stine, Charles Y. Chiu, James Brett Case, Davide Corti, Mehul S. Suthar, Sean P. J. Whelan, Laura A. VanBlargan, Lindsay Droit, Raul Andino, Adrianus C. M. Boon, Daved H. Fremont, Seth J. Zost, Miguel Garcia Knight, Zhuoming Liu, Rita E. Chen, Xuping Xie, Traci L. Bricker, Ali H. Ellebedy, John M. Errico, Michael S. Diamond, Lisa A. Purcell, Ishmael D. Aziati, Tamarand L. Darling, David Wang, Pavlo Gilchuk, Meredith E. Davis-Gardner, Scott A. Handley, Swathi Shrihari, James E. Crowe, Emma S. Winkler, and Astha Joshi
- Subjects
Male ,Antibodies, Viral ,Transgenic ,Mice ,Monoclonal ,Chlorocebus aethiops ,antibodies ,Viral ,Neutralizing ,Lung ,Multidisciplinary ,Serine Endopeptidases ,Antibodies, Monoclonal ,Spike Glycoprotein ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,Pneumonia & Influenza ,Female ,Angiotensin-Converting Enzyme 2 ,Antibody ,Post-Exposure Prophylaxis ,Infection ,Biotechnology ,Genetically modified mouse ,General Science & Technology ,medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Mice, Transgenic ,Biology ,Monoclonal antibody ,Antibodies ,Article ,Vaccine Related ,Neutralization Tests ,In vivo ,Biodefense ,medicine ,Animals ,Humans ,Potency ,variations ,Gene ,Vero Cells ,Mesocricetus ,SARS-CoV-2 ,Prevention ,COVID-19 ,Pneumonia ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,In vitro ,Coronavirus ,Emerging Infectious Diseases ,Cell culture ,Vero cell ,biology.protein ,SARS-CoV-2 variant strains ,Pre-Exposure Prophylaxis ,Immunization - Abstract
Rapidly-emerging variants jeopardize antibody-based countermeasures against SARS-CoV-2. While recent cell culture experiments have demonstrated loss of potency of several anti-spike neutralizing antibodies against SARS-CoV-2 variant strains1-3, the in vivo significance of these results remains uncertain. Here, using a panel of monoclonal antibodies (mAbs) corresponding to many in advanced clinical development by Vir Biotechnology, AbbVie, AstraZeneca, Regeneron, and Lilly we report the impact on protection in animals against authentic SARS-CoV-2 variants including WA1/2020 strains, a B.1.1.7 isolate, and chimeric strains with South African (B.1.351) or Brazilian (B.1.1.28) spike genes. Although some individual mAbs showed reduced or abrogated neutralizing activity against B.1.351 and B.1.1.28 viruses with E484K spike protein mutations in cell culture, low prophylactic doses of mAb combinations protected against infection in K18-hACE2 transgenic mice, 129S2 immunocompetent mice, and hamsters without emergence of resistance. Two exceptions were mAb LY-CoV555 monotherapy which lost all protective activity in vivo, and AbbVie 2B04/47D11, which showed partial loss of activity. When administered after infection as therapy, higher doses of mAb cocktails protected in vivo against viruses displaying a B.1.351 spike gene. Thus, many, but not all, of the antibody products with Emergency Use Authorization should retain substantial efficacy against the prevailing SARS-CoV-2 variant strains.
- Published
- 2021
21. A Randomized Placebo-Controlled Trial of Sarilumab in Hospitalized Patients with Covid-19
- Author
-
Meagan P. O'Brien, Michelle N. Gong, Suraj Saggar, Janie Parrino, Sumathi Sivapalasingam, A. Thomas DiCioccio, Vidya Menon, Romana Hosain, Judith A. Aberg, Samuel M. Brown, Ned Braunstein, Alpana Waldron, Sarilumab-COVID Study Team, Divya Ramesh, Gary Herman, Manuel A. R. Ferreira, Jack A. Kosmicki, Gerard J. Criner, D.J. Lederer, Magdalena E. Sobieszczyk, Michael C Nivens, William Park, Angeliki Giannelou, Adnan Mahmood, Lisa A. Purcell, Steven J. Sperber, Wendy Kampman, George D. Yancopoulos, Selin Somersan-Karakaya, Rafia Bhore, Julie E. Horowitz, Bret J Musser, Bari Kowal, David M. Weinreich, Aris Baras, Emily Labriola-Tompkins, Anita Boyapati, Negin Hajizadeh, Bolanle Akinlade, David Stein, and Daya Gulabani
- Subjects
Relative risk reduction ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Population ,Hazard ratio ,Placebo-controlled study ,Placebo ,Confidence interval ,Sarilumab ,Internal medicine ,Relative risk ,Medicine ,business ,education - Abstract
BACKGROUNDSarilumab (anti-interleukin-6 receptor-α monoclonal antibody) may attenuate the inflammatory response in Covid-19.METHODSWe performed an adaptive, phase 2/3, randomized, double-blind, placebo-controlled trial of intravenous sarilumab 200 mg or 400 mg in adults hospitalized with Covid-19. The phase 3 primary analysis population (cohort 1) was patients with critical Covid-19 receiving mechanical ventilation (MV) randomized to sarilumab 400 mg or placebo. The primary end point for phase 3 was the proportion of patients with ≥1-point improvement in clinical status from baseline to day 22.RESULTSFour-hundred fifty-seven (457) and 1365 patients were randomized and treated in phases 2 and 3, respectively. Among phase 3 critical patients receiving MV (n=289; 34.3% on corticosteroids), the proportion with ≥1-point improvement in clinical status (alive not receiving MV) at day 22 was 43.2% in sarilumab 400 mg and 35.5% in placebo (risk difference [RD] +7.5%; 95% confidence interval [CI], –7.4 to 21.3; P=0.3261), representing a relative risk improvement of 21.7%. Day 29 all-cause mortality was 36.4% in sarilumab 400 mg versus 41.9% in placebo (RD –5.5%; 95% CI, –20.2 to 8.7; relative risk reduction 13.3%). In post hoc analyses pooling phase 2 and 3 critical patients receiving MV, the hazard ratio (HR) for death in sarilumab 400 mg compared with placebo was 0.76 (95% CI, 0.51 to 1.13) overall, improving to 0.49 (95% CI, 0.25 to 0.94) in patients receiving corticosteroids at baseline.CONCLUSIONIn hospitalized patients with Covid-19 receiving MV, numerical benefits with sarilumab did not achieve statistical significance, but benefit may be greater in patients receiving corticosteroids. A larger study is required to confirm this observed numerical benefit.(ClinicalTrials.gov number, NCT04315298)
- Published
- 2021
22. Membrane lectins enhance SARS-CoV-2 infection and influence the neutralizing activity of different classes of antibodies
- Author
-
Elisabetta Cameroni, Maria De Agostini, Amalio Telenti, Herbert W. Virgin, Lisa A. Purcell, Alexandra C. Walls, Siro Bianchi, Florian A. Lempp, John E. Bowen, David Veesler, Young-Jun Park, Leah Soriaga, Fabio Benigni, Davide Corti, Julia Noack, Jiayi Zhou, Stefano Jaconi, Exequiel Dellota, Antonio Lanzavecchia, Martin Montiel-Ruiz, Hanna Kaiser, and Marcel Meury
- Subjects
Pathogenesis ,medicine.anatomical_structure ,biology ,Immunity ,Cell ,biology.protein ,medicine ,Lectin ,Lipid bilayer fusion ,Antibody ,Receptor ,Neutralization ,Cell biology - Abstract
Investigating the mechanisms of SARS-CoV-2 cellular infection is key to better understand COVID-19 immunity and pathogenesis. Infection, which involves both cell attachment and membrane fusion, relies on the ACE2 receptor that is paradoxically found at low levels in the respiratory tract, suggesting that additional mechanisms facilitating infection may exist. Here we show that C-type lectin receptors, DC-SIGN, L-SIGN and the sialic acid-binding Ig-like lectin 1 (SIGLEC1) function as auxiliary receptors by enhancing ACE2-mediated infection and modulating the neutralizing activity of different classes of spike-specific antibodies. Antibodies to the N-terminal domain (NTD) or to the conserved proteoglycan site at the base of the Receptor Binding Domain (RBD), while poorly neutralizing infection of ACE2 over-expressing cells, effectively block lectin-facilitated infection. Conversely, antibodies to the Receptor Binding Motif (RBM), while potently neutralizing infection of ACE2 over-expressing cells, poorly neutralize infection of cells expressing DC-SIGN or L-SIGN and trigger fusogenic rearrangement of the spike promoting cell-to-cell fusion. Collectively, these findings identify a lectin-dependent pathway that enhances ACE2-dependent infection by SARS-CoV-2 and reveal distinct mechanisms of neutralization by different classes of spike-specific antibodies.
- Published
- 2021
23. A system-view of Bordetella pertussis booster vaccine responses in adults primed with whole-cell versus acellular vaccine in infancy
- Author
-
Yuan Tian, Mikhail Pomaznoy, Jason Bennett, Adrienne P. Gilkes, Lisa A. Purcell, Yu Qian, Natalie Khalil, Mariana Babor, Alessandro Sette, Ricardo da Silva Antunes, Aishwarya Mandava, Mario Cortese, Richard H. Scheuermann, Bjoern Peters, Christopher D. Petro, Ferran Soldevila, and Bali Pulendran
- Subjects
0301 basic medicine ,Bordetella pertussis ,Secondary ,and promotion of well-being ,Neutrophils ,Gene Expression ,Booster dose ,Serology ,0302 clinical medicine ,NF-KappaB Inhibitor alpha ,Chemokine CCL3 ,Pertussis Vaccine ,Vaccines ,biology ,Bacterial ,Humoral ,Imprinting ,General Medicine ,Intercellular Adhesion Molecule-1 ,Antibodies, Bacterial ,Bacterial vaccine ,Vaccination ,Infectious Diseases ,3.4 Vaccines ,030220 oncology & carcinogenesis ,Cytokines ,Medicine ,Antibody ,Bacterial vaccines ,Infection ,Research Article ,Immunology ,Immunization, Secondary ,Antibodies ,Vaccine Related ,03 medical and health sciences ,Immune system ,Vaccines, Acellular ,Biodefense ,medicine ,Humans ,Whooping cough ,business.industry ,Prevention ,Inflammatory and immune system ,Immunity ,biology.organism_classification ,medicine.disease ,Prevention of disease and conditions ,Immunity, Humoral ,030104 developmental biology ,Emerging Infectious Diseases ,Good Health and Well Being ,Acellular ,biology.protein ,Immunization ,business - Abstract
The increased incidence of whooping cough worldwide suggests that current vaccination against Bordetella pertussis infection has limitations in quality and duration of protection. The resurgence of infection has been linked to the introduction of acellular vaccines (aP), which have an improved safety profile compared with the previously used whole-cell (wP) vaccines. To determine immunological differences between aP and wP priming in infancy, we performed a systems approach of the immune response to booster vaccination. Transcriptomic, proteomic, cytometric, and serologic profiling revealed multiple shared immune responses with different kinetics across cohorts, including an increase of blood monocyte frequencies and strong antigen-specific IgG responses. Additionally, we found a prominent subset of aP-primed individuals (30%) with a strong differential signature, including higher levels of expression for CCL3, NFKBIA, and ICAM1. Contrary to the wP individuals, this subset displayed increased PT-specific IgE responses after boost and higher antigen-specific IgG4 and IgG3 antibodies against FHA and FIM2/3 at baseline and after boost. Overall, the results show that, while broad immune response patterns to Tdap boost overlap between aP- and wP-primed individuals, a subset of aP-primed individuals present a divergent response. These findings provide candidate targets to study the causes and correlates of waning immunity after aP vaccination.
- Published
- 2021
24. The dual function monoclonal antibodies VIR-7831 and VIR-7832 demonstrate potent in vitro and in vivo activity against SARS-CoV-2
- Author
-
Andrea L. Cathcart, Colin Havenar-Daughton, Florian A. Lempp, Daphne Ma, Michael A. Schmid, Maria L. Agostini, Barbara Guarino, Julia Di iulio, Laura E. Rosen, Heather Tucker, Joshua Dillen, Sambhavi Subramanian, Barbara Sloan, Siro Bianchi, Dora Pinto, Christian Saliba, Katja Culap, Jason A Wojcechowskyj, Julia Noack, Jiayi Zhou, Hannah Kaiser, Sooyoung Lee, Nisar Farhat, Arthur Chase, Martin Montiel-Ruiz, Exequiel Dellota, Arnold Park, Roberto Spreafico, Anna Sahakyan, Elvin J. Lauron, Nadine Czudnochowski, Elisabetta Cameroni, Sarah Ledoux, Yoshihiro Kawaoka, Adam Werts, Christophe Colas, Leah Soriaga, Amalio Telenti, Lisa A. Purcell, Seungmin Hwang, Gyorgy Snell, Herbert W. Virgin, Davide Corti, and Christy M. Hebner
- Subjects
medicine.drug_class ,biochemical phenomena, metabolism, and nutrition ,Biology ,Monoclonal antibody ,Virology ,Fragment crystallizable region ,Virus ,Epitope ,In vitro ,In vivo ,medicine ,biology.protein ,bacteria ,Antibody ,Viral load - Abstract
VIR-7831 and VIR-7832 are dual action monoclonal antibodies (mAbs) targeting the spike glycoprotein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). VIR-7831 and VIR-7832 were derived from a parent antibody (S309) isolated from memory B cells of a 2003 severe acute respiratory syndrome coronavirus (SARS-CoV) survivor. Both mAbs contain an “LS” mutation in the Fc region to prolong serum half-life and potentially enhance distribution to the respiratory mucosa. In addition, VIR-7832 encodes an Fc GAALIE mutation that has been shown previously to evoke CD8+ T-cells in the context of an in vivo viral respiratory infection. VIR-7831 and VIR-7832 potently neutralize wild-type and variant authentic virus in vitro as well as variant pseudotyped viruses. In addition, they retain activity against monoclonal antibody resistance mutations conferring reduced susceptibility to currently authorized mAbs. The VIR-7831/VIR-7832 epitope does not overlap with mutational sites in current variants of concern and continues to be highly conserved among circulating sequences consistent with the high barrier to resistance observed in vitro. Furthermore, both mAbs can recruit effector mechanisms in vitro that may contribute to clinical efficacy via elimination of infected host cells. In vitro studies with these mAbs demonstrated no enhancement of infection. In a Syrian Golden hamster proof-of concept wildtype SARS-CoV-2 infection model, animals treated with VIR-7831 had less weight loss, and significantly decreased total viral load and infectious virus levels in the lung compared to a control mAb. Taken together, these data indicate that VIR-7831 and VIR-7832 are promising new agents in the fight against COVID-19.
- Published
- 2021
25. Lectins enhance SARS-CoV-2 infection and influence neutralizing antibodies
- Author
-
Florian A, Lempp, Leah B, Soriaga, Martin, Montiel-Ruiz, Fabio, Benigni, Julia, Noack, Young-Jun, Park, Siro, Bianchi, Alexandra C, Walls, John E, Bowen, Jiayi, Zhou, Hannah, Kaiser, Anshu, Joshi, Maria, Agostini, Marcel, Meury, Exequiel, Dellota, Stefano, Jaconi, Elisabetta, Cameroni, Javier, Martinez-Picado, Júlia, Vergara-Alert, Nuria, Izquierdo-Useros, Herbert W, Virgin, Antonio, Lanzavecchia, David, Veesler, Lisa A, Purcell, Amalio, Telenti, and Davide, Corti
- Subjects
SARS-CoV-2 ,Sialic Acid Binding Ig-like Lectin 1 ,Receptors, Cell Surface ,Antibodies, Neutralizing ,Membrane Fusion ,Cell Line ,Cell Fusion ,Cricetinae ,Lectins ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,Female ,Lectins, C-Type ,Angiotensin-Converting Enzyme 2 ,Cell Adhesion Molecules - Abstract
SARS-CoV-2 infection-which involves both cell attachment and membrane fusion-relies on the angiotensin-converting enzyme 2 (ACE2) receptor, which is paradoxically found at low levels in the respiratory tract(1-3), suggesting that there may be additional mechanisms facilitating infection. Here we show that C-type lectin receptors, DC-SIGN, L-SIGN and the sialic acid-binding immunoglobulin-like lectin 1 (SIGLEC1) function as attachment receptors by enhancing ACE2-mediated infection and modulating the neutralizing activity of different classes of spike-specific antibodies. Antibodies to the amino-terminal domain or to the conserved site at the base of the receptor-binding domain, while poorly neutralizing infection of ACE2-overexpressing cells, effectively block lectin-facilitated infection. Conversely, antibodies to the receptor binding motif, while potently neutralizing infection of ACE2-overexpressing cells, poorly neutralize infection of cells expressing DC-SIGN or L-SIGN and trigger fusogenic rearrangement of the spike, promoting cell-to-cell fusion. Collectively, these findings identify a lectin-dependent pathway that enhances ACE2-dependent infection by SARS-CoV-2 and reveal distinct mechanisms of neutralization by different classes of spike-specific antibodies.
- Published
- 2021
26. The Psychological Processes of Classic Psychedelics in the Treatment of Depression: A Systematic Review Protocol
- Author
-
Lisa Mastin-Purcell, Paul Liknaitzky, Maja Nedeljkovic, Greg Murray, and Lauren Johansen
- Subjects
Adult ,Protocol (science) ,Psychotherapist ,Depression ,business.industry ,Mental Disorders ,Medicine (miscellaneous) ,Psychotherapy ,Hallucinogens ,Humans ,Medicine ,business ,Depression (differential diagnoses) ,Systematic Reviews as Topic - Abstract
Background There is currently renewed interest in the use of psychedelic therapy in the treatment of psychiatric disorders, including depression. The proposed systematic review will aim to identify, evaluate and summarise the psychological processes of change underlying psychedelic therapy for depression in the current literature and consider the implications these processes may have on the psychotherapy component of treatment. Methods Scopus, PsycINFO, PubMed and Web of Science databases will be searched using relevant terms. Studies will be included if they discuss the use of a classic psychedelic to treat depression symptomology in an adult population and report or propose psychological processes responsible for depression symptom change. Two authors will independently screen articles, complete quality assessment tools and conduct data extraction. Empirical and non-empirical research will be extracted and synthesised separately. A narrative synthesis approach will be used to report psychological processes identified in the literature. Discussion This systematic review will be the first to collate available evidence on the psychological processes associated with psychedelic therapy for depression. The preliminary nature of this research field is expected to result in the review having several limitations, namely heterogeneity between studies and the inclusion of limited empirical research. We intend for this review to present the current state of the literature, identify gaps and generate candidate variables that warrant further investigation. Systematic review PROSPERO CRD42020197202
- Published
- 2020
27. A system-view of B. pertussis booster vaccine responses in adults primed with whole-cell vs. acellular vaccine in infancy
- Author
-
Natalie Khalil, Ricardo da Silva Antunes, Mariana Babor, Adrienne P. Gilkes, Yuan Tian, Mario Cortese, Aishwaraya Mandava, Alessandro Sette, Jason Bennett, Christopher D. Petro, Mikhail Pomaznoy, Lisa A. Purcell, Yu Qian, Bali Pulendran, Ferran Soldevila, Bjoern Peters, and Richard H. Scheuermann
- Subjects
Bordetella pertussis ,biology ,business.industry ,Booster dose ,biology.organism_classification ,Vaccination ,Immune system ,Antigen ,Immunity ,Inactivated vaccine ,Immunology ,biology.protein ,Medicine ,Antibody ,business - Abstract
Whole-cell inactivated vaccine against Bordetella pertussis (wP) was substituted in many countries by an acellular subunit vaccine (aP) to reduce side effects. Recent epidemiological studies have shown that aP vaccination in infancy induces less durable immunity than wP vaccination. To determine immunological differences associated with aP vs. wP priming, we performed system-level profiling of the immune response in adults primed with aP vs. wP vaccine in infancy following the Tdap booster vaccination as a surrogate to antigen encounter in vivo. Shared immune responses across cohorts were identified, including an increase of the blood monocyte frequency on day 1, and strong antigen-specific IgG response seven days after boost. Comparing aP and wP primed individuals, we found a subset of aP-primed individuals with higher levels of expression for several genes including CCL3 on day 3 and NFKBIA and ICAM1 on day 7 post immunization. These observations were supported by increased CCL3 concentrations in plasma of aP primed individuals. Contrary to the wP individuals, the CCL3-high aP subset presented boosted PT-specific IgE responses. Furthermore, higher antigen specific IgG4 and IgG3 antibodies against specific vaccine antigens at baseline and post boost of aP individuals was observed, suggesting a long term maintained difference in the IgG subtype response. Overall our findings demonstrate that, while broad immune response patterns to Tdap boost overlap between aP and wP primed individuals, a subset of aP primed individuals present a divergent response. These findings provide candidate targets to study the causes and correlates of waning immunity after aP vaccination.
- Published
- 2020
28. Identification of a mutant PfCRT-mediated chloroquine tolerance phenotype in Plasmodium falciparum.
- Author
-
Stephanie G Valderramos, Juan-Carlos Valderramos, Lise Musset, Lisa A Purcell, Odile Mercereau-Puijalon, Eric Legrand, and David A Fidock
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Mutant forms of the Plasmodium falciparum transporter PfCRT constitute the key determinant of parasite resistance to chloroquine (CQ), the former first-line antimalarial, and are ubiquitous to infections that fail CQ treatment. However, treatment can often be successful in individuals harboring mutant pfcrt alleles, raising questions about the role of host immunity or pharmacokinetics vs. the parasite genetic background in contributing to treatment outcomes. To examine whether the parasite genetic background dictates the degree of mutant pfcrt-mediated CQ resistance, we replaced the wild type pfcrt allele in three CQ-sensitive strains with mutant pfcrt of the 7G8 allelic type prevalent in South America, the Oceanic region and India. Recombinant clones exhibited strain-dependent CQ responses that ranged from high-level resistance to an incremental shift that did not meet CQ resistance criteria. Nonetheless, even in the most susceptible clones, 7G8 mutant pfcrt enabled parasites to tolerate CQ pressure and recrudesce in vitro after treatment with high concentrations of CQ. 7G8 mutant pfcrt was found to significantly impact parasite responses to other antimalarials used in artemisinin-based combination therapies, in a strain-dependent manner. We also report clinical isolates from French Guiana that harbor mutant pfcrt, identical or related to the 7G8 haplotype, and manifest a CQ tolerance phenotype. One isolate, H209, harbored a novel PfCRT C350R mutation and demonstrated reduced quinine and artemisinin susceptibility. Our data: 1) suggest that high-level CQR is a complex biological process dependent on the presence of mutant pfcrt; 2) implicate a role for variant pfcrt alleles in modulating parasite susceptibility to other clinically important antimalarials; and 3) uncover the existence of a phenotype of CQ tolerance in some strains harboring mutant pfcrt.
- Published
- 2010
- Full Text
- View/download PDF
29. Adenylyl cyclase alpha and cAMP signaling mediate Plasmodium sporozoite apical regulated exocytosis and hepatocyte infection.
- Author
-
Takeshi Ono, Laura Cabrita-Santos, Ricardo Leitao, Esther Bettiol, Lisa A Purcell, Olga Diaz-Pulido, Lucy B Andrews, Takushi Tadakuma, Purnima Bhanot, Maria M Mota, and Ana Rodriguez
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Malaria starts with the infection of the liver of the host by Plasmodium sporozoites, the parasite form transmitted by infected mosquitoes. Sporozoites migrate through several hepatocytes by breaching their plasma membranes before finally infecting one with the formation of an internalization vacuole. Migration through host cells induces apical regulated exocytosis in sporozoites. Here we show that apical regulated exocytosis is induced by increases in cAMP in sporozoites of rodent (P. yoelii and P. berghei) and human (P. falciparum) Plasmodium species. We have generated P. berghei parasites deficient in adenylyl cyclase alpha (ACalpha), a gene containing regions with high homology to adenylyl cyclases. PbACalpha-deficient sporozoites do not exocytose in response to migration through host cells and present more than 50% impaired hepatocyte infectivity in vivo. These effects are specific to ACalpha, as re-introduction of ACalpha in deficient parasites resulted in complete recovery of exocytosis and infection. Our findings indicate that ACalpha and increases in cAMP levels are required for sporozoite apical regulated exocytosis, which is involved in sporozoite infection of hepatocytes.
- Published
- 2008
- Full Text
- View/download PDF
30. Th1/Th17 polarization persists following whole-cell pertussis vaccination despite repeated acellular boosters
- Author
-
Pandurangan Vijayanand, Lisa A. Purcell, Bjoern Peters, Alexander J. Mentzer, Mariana Babor, Christopher D. Petro, Bali Pulendran, Mario Cortese, Chelsea Carpenter, Ricardo da Silva Antunes, Grégory Seumois, Alessandro Sette, Shane Crotty, and Natalie Khalil
- Subjects
0301 basic medicine ,business.industry ,T cell ,Antibody titer ,General Medicine ,Acquired immune system ,In vitro ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Immunology ,medicine ,030212 general & internal medicine ,Th1 th17 ,business ,Adverse effect ,Ex vivo - Abstract
In the mid-1990s, whole-cell pertussis (wP) vaccines were associated with local and systemic adverse events that prompted their replacement with acellular pertussis (aP) vaccines in many high-income countries. In the past decade, rates of pertussis disease have increased in children receiving only aP vaccines. We compared the immune responses to aP boosters in individuals who received their initial doses with either wP or aP vaccines using activation-induced marker (AIM) assays. Specifically, we examined pertussis-specific memory CD4+ T cell responses ex vivo, highlighting a type 2/Th2 versus type 1/Th1 and Th17 differential polarization as a function of childhood vaccination. Remarkably, after a contemporary aP booster, cells from donors originally primed with aP were (a) associated with increased IL-4, IL-5, IL-13, IL-9, and TGF-β and decreased IFN-γ and IL-17 production, (b) defective in their ex vivo capacity to expand memory cells, and (c) less capable of proliferating in vitro. These differences appeared to be T cell specific, since equivalent increases of antibody titers and plasmablasts after aP boost were seen in both groups. In conclusion, our data suggest that there are long-lasting effects and differences in polarization and proliferation of T cell responses in adults originally vaccinated with aP compared with those that initially received wP, despite repeated acellular boosters.
- Published
- 2018
31. Differences in T cell responses to Bordetella Pertussis in adults as a function of whole cell versus acellular childhood vaccination
- Author
-
Ricardo Filipe da Silva Antunes, Mariana Babor, Mikhail Pomaznoy, Natalie Khalil, Mario Cortese, Alexander J Mentzer, Gregory Seumois, Christopher D Petro, Lisa A Purcell, Pandurangan Vijayanand, Shane Crotty, Bali Pulendran, Bjoern Peters, and Alessandro Sette
- Subjects
Immunology ,Immunology and Allergy - Abstract
In the mid-1990s vaccine-related side effects prompted the replacement of the whole Pertussis (wP) by a new acellular Pertussis vaccine (aP). Unexpectedly, whooping cough cases have recently increased, and the switch to the aP vaccine suspected to underlie this rise in morbidity, despite the use of routine boosters. Here we compared the immune responses to aP boosters in children who received their initial doses with either wP or aP vaccines. The use of ex vivo AIM assays highlighted a Th2 vs Th1/Th17 differential polarization of PT-specific memory CD4+ T cells as a function of childhood vaccination. The nature of the differences was further investigated for memory subset composition, activation, exhaustion marker expression, and transcriptomic profiles. Remarkably, donors originally primed with aP were defective in their ex vivo capacity to expand memory cells following a booster aP immunization and less capable to proliferate in vitro in response to PT epitopes. By contrast antibody responses are boosted in both aP and wP cohorts following IgG subclass distribution. Most recently, we have assessed early immune responses followed aP boosting for the aP and wP cohorts using longitudinal transcriptomics data from PBMC. We have identified several modules of genes that are co-expressed; using cell frequency data from CyTOF we infer modules correlated with specific cell types. Furthermore, we identified certain modules that show differential expression profiles between the two cohorts and are mostly enriched in immune related functions. In conclusion, our data suggest that the original priming after birth with aP and wP vaccines induce different T cell phenotypes associated with long-term T cell polarization.
- Published
- 2019
32. Dupilumab does not affect correlates of vaccine-induced immunity: A randomized, placebo-controlled trial in adults with moderate-to-severe atopic dermatitis
- Author
-
Andrew Blauvelt, Neil M.H. Graham, Eric L. Simpson, Bolanle Akinlade, Christopher D. Petro, Robert Evans, Brad Shumel, Lisa A. Purcell, Abhijit Gadkari, Laurent Eckert, Stephen K. Tyring, and Gianluca Pirozzi
- Subjects
Adult ,Male ,Dermatology ,Meningococcal vaccine ,Antibodies, Monoclonal, Humanized ,Severity of Illness Index ,Eczema Area and Severity Index ,Dermatitis, Atopic ,030207 dermatology & venereal diseases ,03 medical and health sciences ,Immunogenicity, Vaccine ,0302 clinical medicine ,Double-Blind Method ,medicine ,Humans ,Seroconversion ,Tetanus ,business.industry ,Toxoid ,Antibody titer ,Antibodies, Monoclonal ,medicine.disease ,Dupilumab ,Immunity, Humoral ,Vaccination ,030220 oncology & carcinogenesis ,Immunology ,Female ,business - Abstract
The impact of dupilumab, an anti-interleukin (IL) 4 receptor α antibody that inhibits IL-4 and IL-13 signaling, on vaccine responses of patients with atopic dermatitis (AD) is unknown.To assess T-cell-dependent and T-cell-independent humoral immune responses to tetanus and meningococcal vaccines, IgE seroconversion to tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis (Tdap) vaccination, and dupilumab efficacy and safety.In a randomized, double-blinded, placebo-controlled study (NCT02210780), adults with moderate-to-severe AD received dupilumab (300 mg) or placebo weekly for 16 weeks, and single doses of Tdap and quadrivalent meningococcal polysaccharide vaccines at week 12. Primary endpoint was proportion of patients achieving satisfactory IgG response to tetanus toxoid at week 16.In total, 178 patients completed the study. Similar positive immune responses (≥4-fold increase in antibody titer, or an antibody titer of ≥8) were achieved in the dupilumab and placebo groups to tetanus (83.3% and 83.7%, respectively) and meningococcal polysaccharide (86.7% and 87.0%, respectively). Dupilumab significantly decreased total serum IgE; most dupilumab-treated patients were Tdap-IgE seronegative at week 32 (62.2% dupilumab and 34.8% placebo). Dupilumab improved key AD efficacy endpoints (P .001). Injection-site reactions and conjunctivitis were more common with dupilumab; AD exacerbations more frequent with placebo.Patients' prior vaccination status was not available before enrollment.Dupilumab did not affect responses to the vaccines studied, significantly decreased IgE, and improved measures of AD severity versus placebo, with an acceptable safety profile.
- Published
- 2019
33. Dysregulation of angiopoietin-1 plays a mechanistic role in the pathogenesis of cerebral malaria
- Author
-
W. Conrad Liles, Andrea L. Conroy, Lisa A. Purcell, Valerie M. Crowley, Kevin C. Kain, Michael Hawkes, Gavin Thurston, John G. Hay, Robert O. Opoka, Susan E. Quaggin, Vanessa Tran, Karlee L. Silver, and Sarah J. Higgins
- Subjects
Male ,0301 basic medicine ,Plasmodium falciparum ,Malaria, Cerebral ,Artesunate ,Kaplan-Meier Estimate ,Biology ,Article ,Adenoviridae ,Angiopoietin ,Pathogenesis ,Antimalarials ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Disease severity ,parasitic diseases ,Angiopoietin-1 ,Animals ,Humans ,Uganda ,Malaria, Falciparum ,Prospective cohort study ,Endothelial Cells ,Infant ,General Medicine ,Receptor, TIE-2 ,Survival Analysis ,Artemisinins ,Recombinant Proteins ,Pathophysiology ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Kinetics ,Phenotype ,Treatment Outcome ,030104 developmental biology ,chemistry ,Blood-Brain Barrier ,Cerebral Malaria ,Child, Preschool ,030220 oncology & carcinogenesis ,Immunology ,Female ,Disease Susceptibility ,Gene Deletion - Abstract
Cerebral malaria is a leading cause of global morbidity and mortality. Interventions targeting the underlying pathophysiology of cerebral malaria may improve outcomes compared to treatment with antimalarials alone. Microvascular leak plays an important role in the pathogenesis of cerebral malaria. The angiopoietin (Ang)–Tie-2 system is a critical regulator of vascular function. We show that Ang-1 expression and soluble Tie-2 expression were associated with disease severity and outcome in a prospective study of Ugandan children with severe malaria and in a preclinical murine model of experimental cerebral malaria. Ang-1 was necessary for maintenance of vascular integrity and survival in a mouse model of cerebral malaria. Therapeutic administration of Ang-1 preserved blood-brain barrier integrity and, in combination with artesunate treatment, improved survival beyond that with artesunate alone. These data define a role for dysregulation of the Ang–Tie-2 axis in the pathogenesis of cerebral malaria and support the evaluation of Ang–Tie-2–based interventions as potential adjunctive therapies for treating severe malaria.
- Published
- 2016
34. A putative kinase-related protein (PKRP) from Plasmodium berghei mediates infection in the midgut and salivary glands of the mosquito
- Author
-
Ricardo Leitao, Gabriele Pradel, Terry W. Spithill, Lisa A. Purcell, Takeshi Ono, Ana Rodriguez, and Stephanie K. Yanow
- Subjects
Male ,Plasmodium berghei ,Virulence Factors ,Molecular Sequence Data ,Protozoan Proteins ,Plasmodium ,Article ,Salivary Glands ,Microbiology ,Mice ,03 medical and health sciences ,parasitic diseases ,medicine ,Animals ,Parasite hosting ,Amino Acid Sequence ,Protein kinase A ,Gene ,030304 developmental biology ,0303 health sciences ,Sequence Homology, Amino Acid ,biology ,Salivary gland ,030306 microbiology ,Gene Expression Profiling ,fungi ,Midgut ,biology.organism_classification ,Molecular biology ,Epithelium ,3. Good health ,Gastrointestinal Tract ,Molecular Weight ,Mutagenesis, Insertional ,Culicidae ,Infectious Diseases ,medicine.anatomical_structure ,Female ,Parasitology ,Protein Kinases ,Gene Deletion - Abstract
The completion of the Plasmodium (malaria) life cycle in the mosquito requires the parasite to traverse first the midgut and later the salivary gland epithelium. We have identified a putative kinase-related protein (PKRP) that is predicted to be an atypical protein kinase, which is conserved across many species of Plasmodium . The pkrp gene encodes a RNA of about 5300 nucleotides that is expressed as a 90 kDa protein in sporozoites. Targeted disruption of the pkrp gene in Plasmodium berghei , a rodent model of malaria, compromises the ability of parasites to infect different tissues within the mosquito host. Early infection of mosquito midgut is reduced by 58–71%, midgut oocyst production is reduced by 50–90% and those sporozoites that are produced are defective in their ability to invade mosquito salivary glands. Midgut sporozoites are not morphologically different from wild-type parasites by electron microscopy. Some sporozoites that emerged from oocysts were attached to the salivary glands but most were found circulating in the mosquito hemocoel. Our findings indicate that a signalling pathway involving PbPKRP regulates the level of Plasmodium infection in the mosquito midgut and salivary glands.
- Published
- 2010
35. The Fatty Acid Biosynthesis Enzyme FabI Plays a Key Role in the Development of Liver-Stage Malarial Parasites
- Author
-
Juan Carlos Valderramos, Brie Falkard, Celeste D. Li, Mark J. Siedner, Guy A. Schiehser, David A. Fidock, Photini Sinnis, Brendan J Kelly, Pedro A. Moura, Viswanathan Lakshmanan, Arba L. Ager, James C. Sacchettini, Catherine Vilchèze, Min Yu, William R. Jacobs, Chris J. Janse, Joel S. Freundlich, David P. Jacobus, Andrew P. Waters, Lisa A. Purcell, Alida Coppi, Jennifer H.C. Tsai, Volker Heussler, Laurent Kremer, Louis J. Nkrumah, T. R. Santha Kumar, Paul Gratraud, Amar Bir Singh Sidhu, Silke Retzlaff, University College of the Fraser Valley (UCFV), University College of the Fraser Valley (UFV), Courant Institute of Mathematical Sciences [New York] (CIMS), New York University [New York] (NYU), NYU System (NYU)-NYU System (NYU), Institute of Vertebrate Paleontology and Paleoanthropology, Institut Européen des membranes (IEM), Centre National de la Recherche Scientifique (CNRS)-Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Institut de Chimie du CNRS (INC)-Université de Montpellier (UM), Department of Biochemistry and Biophysics, Texas A&M University [College Station], Laboratory of Molecular Neurochemistry, The Research Building, Rom W222, Washington, Laboratory of Molecular Neurochemistry, Department of Pediatrics, Georgetown University, Institut de Neurobiologie Alfred Fessard (INAF), Centre National de la Recherche Scientifique (CNRS), Dynamique des interactions membranaires normales et pathologiques (DIMNP), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université Montpellier 1 (UM1), Dpt of Parasitology [Leiden], Leiden University Medical Center (LUMC), and Howard Hughes Medical Institute (HHMI)
- Subjects
Cancer Research ,MESH: Parasitemia ,MICROBIO ,Plasmodium berghei ,Protozoan Proteins ,Parasitemia ,MESH: Triclosan ,Plasmodium ,Mice ,chemistry.chemical_compound ,MESH: Animals ,MESH: Protozoan Proteins ,MESH: Plasmodium falciparum ,chemistry.chemical_classification ,0303 health sciences ,biology ,Transfection ,3. Good health ,Liver ,Biochemistry ,Plasmodium falciparum ,MESH: Malaria ,Microbiology ,Antimalarials ,03 medical and health sciences ,MESH: Mice, Inbred C57BL ,Immunology and Microbiology(all) ,Virology ,parasitic diseases ,Animals ,MESH: Plasmodium berghei ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Molecular Biology ,MESH: Mice ,Fatty acid synthesis ,030304 developmental biology ,Apicoplast ,030306 microbiology ,biology.organism_classification ,MESH: Antimalarials ,Triclosan ,In vitro ,Malaria ,Mice, Inbred C57BL ,Mutagenesis, Insertional ,CHEMBIO ,Enzyme ,MESH: Mutagenesis, Insertional ,chemistry ,MESH: Gene Deletion ,Parasitology ,Gene Deletion ,MESH: Liver - Abstract
International audience; The fatty acid synthesis type II pathway has received considerable interest as a candidate therapeutic target in Plasmodium falciparum asexual blood-stage infections. This apicoplast-resident pathway, distinct from the mammalian type I process, includes FabI. Here, we report synthetic chemistry and transfection studies concluding that Plasmodium FabI is not the target of the antimalarial activity of triclosan, an inhibitor of bacterial FabI. Disruption of fabI in P. falciparum or the rodent parasite P. berghei does not impede blood-stage growth. In contrast, mosquito-derived, FabI-deficient P. berghei sporozoites are markedly less infective for mice and typically fail to complete liver-stage development in vitro. This defect is characterized by an inability to form intrahepatic merosomes that normally initiate blood-stage infections. These data illuminate key differences between liver- and blood-stage parasites in their requirements for host versus de novo synthesized fatty acids, and create new prospects for stage-specific antimalarial interventions.
- Published
- 2008
36. Chemically attenuated Plasmodium sporozoites induce specific immune responses, sterile immunity and cross-protection against heterologous challenge
- Author
-
Stephanie K. Yanow, Kurt A Wong, Moses Lee, Lisa A. Purcell, Ana Rodriguez, and Terry W. Spithill
- Subjects
Protozoan Vaccines ,Indoles ,Plasmodium berghei ,Antibodies, Protozoan ,Heterologous ,Antigens, Protozoan ,Cross immunity ,CD8-Positive T-Lymphocytes ,Cross Reactions ,Vaccines, Attenuated ,Plasmodium ,Article ,Microbiology ,Interferon-gamma ,Mice ,Immune system ,Immunity ,parasitic diseases ,Animals ,Mice, Inbred BALB C ,General Veterinary ,General Immunology and Microbiology ,biology ,Vaccination ,fungi ,Public Health, Environmental and Occupational Health ,Plasmodium yoelii ,biology.organism_classification ,Virology ,Malaria ,Infectious Diseases ,Gene Expression Regulation ,Sporozoites ,Molecular Medicine ,Female - Abstract
Vaccination with Plasmodium sporozoites attenuated by irradiation or genetic manipulation induces a protective immune response in rodent malaria models. Recently, vaccination with chemically attenuated P. berghei sporozoites (CAS) has also been shown to elicit sterile immunity in mice. Here we show that vaccination with CAS of P. yoelii also protects against homologous infection and that a P. berghei CAS vaccine cross protects against heterologous challenge with P. yoelii sporozoites. Vaccination with P. yoelii or P. berghei CAS induced parasite-specific antibodies and IFN-gamma-producing CD8(+) T cells at levels not significantly different from radiation-attenuated sporozoites. Our findings provide an initial characterization of the immune response generated by CAS vaccination and suggest that this attenuation process could be used in the production of an effective cross-protective liver stage vaccine for malaria.
- Published
- 2008
37. Genomics-based drug design targets the AT-rich malaria parasite: implications for antiparasite chemotherapy
- Author
-
Moses Lee, Stephanie K. Yanow, Lisa A. Purcell, and Terry W. Spithill
- Subjects
Pharmacology ,Genetics ,Plasmodium falciparum ,Context (language use) ,Genomics ,Biology ,medicine.disease ,biology.organism_classification ,Genome ,genomic DNA ,Adozelesin ,Pharmacogenomics ,medicine ,Molecular Medicine ,Malaria - Abstract
Evaluation of: Woynarowski JM, Krugliak M, Ginsburg H: Pharmacogenomic analyses of targeting the AT-rich malaria parasite genome with AT-specific alkylating drugs. Mol. Biochem. Parasitol. 154(1), 70–81 (2007) [1] . The sequencing of the malaria genome sought to expose the parasite’s ability to cause disease and identify new targets for antimalarial drugs and vaccines. In this study, the authors discovered how malaria genomic DNA, which is unusually rich in adenine and thymine nucleotides, is intrinsically a target for a selective class of compounds. AT-specific DNA-binding agents have previously been shown to have potent antimalarial activity in vitro. The authors used high-resolution bioinformatic tools to explore the genomic basis for this drug susceptibility, first at the level of individual DNA-binding sites, then expanding to the entire genomic context of each malaria chromosome. Their findings revealed a nonrandom distribution and organization of drug-binding sites that can be further exploited to target these AT sequences. Based on these findings, comparative bioinformatics analyses with other parasite genomes may lead to the identification of new target organisms for these AT-specific drugs and have wide implications for the treatment of human and animal parasitic diseases.
- Published
- 2007
38. Effects of an azinphos-methyl runoff event on macroinvertebrates in the Wilmot River, Prince Edward Island, Canada
- Author
-
Donna J. Giberson and Lisa A. Purcell
- Subjects
Physiology ,Ecology ,Community structure ,STREAMS ,Population decline ,Geography ,Structural Biology ,Benthic zone ,Abundance (ecology) ,Insect Science ,Species richness ,Surface runoff ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Invertebrate - Abstract
High levels of azinphos-methyl (0.4–0.8 µg/L) were detected in the Wilmot River, Prince Edward Island, Canada, following runoff from an agricultural field after a heavy rainfall on 19 July 2002. Benthic macroinvertebrate abundance and diversity were sharply lower in samples collected 1 d after the event compared with samples collected in the same manner in July or October 2001. The greatest effects were noticed on the aquatic insects, whose abundance declined from >10 000 individuals per 3-min kick sample in July 2001 to etc.) confirmed that aquatic insects were more heavily targeted by the insecticide than non-insect invertebrates. This resulted in a shift in the community towards non-insect taxa that were better able to avoid or tolerate this type of pollution.
- Published
- 2007
39. The A/T-specific DNA alkylating agent adozelesin inhibits Plasmodium falciparum growth in vitro and protects mice against Plasmodium chabaudi adami infection
- Author
-
Terry W. Spithill, Stephanie K. Yanow, and Lisa A. Purcell
- Subjects
Indoles ,Cyclohexanecarboxylic Acids ,DNA damage ,Plasmodium falciparum ,Drug Evaluation, Preclinical ,Parasitemia ,Biology ,Plasmodium chabaudi ,Antimalarials ,Duocarmycins ,Mice ,In vivo ,Cyclohexenes ,parasitic diseases ,medicine ,Animals ,Malaria, Falciparum ,Base Pairing ,Molecular Biology ,Benzofurans ,Mice, Inbred BALB C ,Dose-Response Relationship, Drug ,DNA synthesis ,Adenine ,DNA replication ,DNA, Protozoan ,biology.organism_classification ,medicine.disease ,Virology ,Adozelesin ,Female ,Parasitology ,Injections, Intraperitoneal ,Thymine - Abstract
There is an urgent need for new anti-malarial drugs to combat the resurgence of resistance to current therapies. To exploit the A/T richness of malaria DNA as a potential target for anti-malarial drugs we tested an A/T-specific DNA synthesis inhibitor, adozelesin, for activity against Plasmodium falciparum in vitro and Plasmodium chabaudi adami in mice. Adozelesin is a DNA alkylating agent that exhibits specificity for the motif A/T, A/T and A. In P. falciparum 3D7 cultures, adozelesin acts as a powerful inhibitor of parasite growth (IC 50 of 70 pM) and is equally potent at killing the drug-resistant strains FCR3 and 7G8. Using a real-time PCR assay, we show that treatment with adozelesin in vitro results in damage of P. falciparum genomic DNA. In synchronized cultures, adozelesin exhibits a concentration-dependent effect on parasitemia and on the development of parasites through the asexual cycle. In asynchronous cultures, parasites arrest at all stages of the asexual cycle suggesting that adozelesin exerts other anti-parasitic effects in addition to inhibiting DNA replication. These anti-parasite effects are irreversible since cultures exposed to adozelesin for more than 6 h fail to recover upon removal of the drug. Furthermore, adozelesin is very effective at suppressing malaria infection in vivo; growth of P. c. adami DK in mice was highly impaired by a single injection of adozelesin (25 μg/kg) at 4 days post-infection. These results demonstrate that adozelesin irreversibly blocks parasite growth in vitro and suppresses parasite infection in vivo, suggesting that A/T-specific DNA damaging agents represent a new class of compounds with potential as anti-malarials.
- Published
- 2006
40. Insular black files (Diptera: Simuliidae) of North America: tests of colonization hypotheses
- Author
-
Lisa A. Purcell, Donna J. Giberson, and Peter H. Adler
- Subjects
Colonisation ,Ecology ,biology ,Habitat ,Insular biogeography ,Biogeography ,Biological dispersal ,Queen (butterfly) ,Mainland ,Simulium ,biology.organism_classification ,Ecology, Evolution, Behavior and Systematics - Abstract
Aim To understand factors that facilitate insular colonization by black flies, we tested six hypotheses related to life-history traits, phylogeny, symbiotes, island area, and distance from source areas. Location Four northern islands, all within 150 km of the North American mainland, were included in the study: Isle Royale, Magdalen Islands, Prince Edward Island, and Queen Charlotte Islands. Methods Immature black flies and their symbiotes were surveyed in streams on the Magdalen Islands, and the results combined with data from similar surveys on Isle Royale, Prince Edward Island, and the Queen Charlotte Islands. Black flies were analysed chromosomally to ensure that all sibling species were revealed. Tests of independence were used to examine the frequency of life-history traits and generic representation of black flies on islands vs. source areas. Results A total of 13‐20 species was found on each of the islands, but no species was unique to any of the islands. The simuliid faunas of the islands reflected the composition of their source areas in aspects of voltinism (univoltine vs. multivoltine), blood feeding (ornithophily vs. mammalophily), and phylogeny (genus Simulium vs. other genera). Five symbiotic species were found on the most distant island group, the Magdalen Islands, supporting the hypothesis that obligate symbiotes are effectively transported to near-mainland islands. An inverse relationship existed between the number of species per island and distance from the source. The Queen Charlotte Islands did not conform to the species‐ area relationship. Main conclusions The lack of precinctive insular species and an absence of lifehistory and phylogenetic characteristics related to the presence of black flies on these islands argue for gene flow and dispersal capabilities of black flies over open waters, possibly aided by winds. However, the high frequency of precinctive species on islands 500 km or more from the nearest mainland indicates that at some distance beyond 100 km, open water provides a significant barrier to colonization and gene exchange. An inverse relationship between number of species and distance from the source suggests that as long as suitable habitat is present, distance plays an important role in colonization. Failure of the Queen Charlotte Islands to conform to an area‐richness trend suggests that not all resident species have been found.
- Published
- 2005
41. INCREASED EXPRESSION OF IL-4 AND IL-10 AND DECREASED EXPRESSION OF IL-2 AND INTERFERON-γ IN LONG-SURVIVING MOUSE HEART ALLOGRAFTS AFTER BRIEF CD4-MONOCLONAL ANTIBODY THERAPY1,2
- Author
-
Patricia L. Mottram, Wen-Ruo Han, Lisa J. Purcell, Ian F.C. McKenzie, and Wayne W. Hancock
- Subjects
Transplantation - Published
- 1995
42. Changes in smooth muscle contractility of rainbow trout (Oncorhynchus mykiss Walbaum) intestine during acclimation to altered temperature
- Author
-
Heather A. Briand, Jeffrey G. Hogan, Gregory A. Mitton, Lisa M. Purcell, William P. Ireland, and John F. Burka
- Subjects
medicine.medical_specialty ,Carbachol ,Physiology ,Stimulation ,General Medicine ,Smooth muscle contraction ,Aquatic Science ,Biology ,biology.organism_classification ,Biochemistry ,Acclimatization ,Contractility ,Trout ,Endocrinology ,Internal medicine ,medicine ,Rainbow trout ,sense organs ,Acetylcholine ,medicine.drug - Abstract
The effects of altered water temperature in vivo on in vitro smooth muscle contractility of rainbow trout intestine were investigated. Temperature has a significant effect on receptor-mediated intestinal smooth muscle contractility in the rainbow trout. The efficacy of 5-HT, carbachol, and transmural stimulation increased with temperatures above 10°C, with an optimal increase at 15°C. There was also a modest increase in the potency of 5-HT and carbachol within 2 days of establishing trout at 20°C. By day 8, most of these changes had either stabilized or were returning to control values, suggesting that acclimation changes in membranes and enzyme activities were taking effect. However, the contractile responses to carbachol and transmural stimulation were still increasing at this time. This may imply that the muscarinic receptors are more resistant to membrane acclimation changes and may take longer to adapt. Because these experiments were controlled for handling stress and seasonal changes that affect contractility, we have been able to demonstrate some early changes in smooth muscle contractility that occur during acclimation to altered temperature.
- Published
- 1993
43. The effects of acute temperature change on smooth muscle contractility of rainbow trout (Oncorhynchus mykiss Walbaum) intestine
- Author
-
Lisa M. Purcell, John F. Burka, Heather A. Briand, and William P. Ireland
- Subjects
medicine.medical_specialty ,Carbachol ,Physiology ,Stimulation ,General Medicine ,Smooth muscle contraction ,Aquatic Science ,Biology ,biology.organism_classification ,Biochemistry ,Trout ,Endocrinology ,In vivo ,Internal medicine ,medicine ,Potency ,Rainbow trout ,Acetylcholine ,medicine.drug - Abstract
The effects of altered temperature in vivo on in vitro smooth muscle contractility of rainbow trout intestine were investigated. Initial analysis of the data revealed a seasonal variation in the maximal tension of intestinal smooth muscle attainable with 5-hydroxytryptamine (serotonin), carbachol, KCl, and transmural stimulation in vitro. Peaks occurred in spring and troughs in autumn. There was no seasonal cycling of the potency of the stimulants. All data regarding the efficacy of the stimulants were subsequently corrected for seasonal variation. The response of smooth muscle depends on the temperature of the water in which the fish are placed (2°C-20°C). There was a marked linear increase in efficacy and a slight increase in potency of the stimulants with increasing temperature. Changes in responsiveness of the intestinal smooth muscle occurred within 30 min of moving the fish between tanks. Smooth muscle reactivity returned to pretreatment values by 48h. Any changes in responsiveness with regards to time were unlikely to be as a consequence of water temperature, but may have been a result of handling stress.
- Published
- 1993
44. Potent antimalarial and transmission-blocking activities of centanamycin, a novel DNA-binding agent
- Author
-
Terry W. Spithill, Atsushi Sato, Ana Rodriguez, Stephanie K. Yanow, Moses Lee, Gabriele Pradel, and Lisa A. Purcell
- Subjects
Alkylating Agents ,Plasmodium ,Indoles ,DNA damage ,chemistry.chemical_compound ,Antimalarials ,Duocarmycins ,Mice ,parasitic diseases ,Anopheles ,medicine ,Immunology and Allergy ,Parasite hosting ,Animals ,Pyrroles ,biology ,fungi ,Oocysts ,medicine.disease ,biology.organism_classification ,Virology ,genomic DNA ,Disease Models, Animal ,Infectious Diseases ,chemistry ,Sporozoites ,Female ,DNA Binding Agent ,Malaria ,DNA - Abstract
Most treatments for malaria target the blood stage of infection in the human host, although few can also block transmission of the parasite to the mosquito. We show here that the compound centanamycin is very effective against blood-stage malarial infections in vitro and in vivo and has profound effects on sexual differentiation of the parasites in mosquitoes. After drug treatment, parasite development is arrested within the midguts of mosquitoes, failing to produce the infective forms that migrate to the salivary glands. The mechanism of parasite death is associated with modification of Plasmodium genomic DNA. We detected DNA damage in parasites isolated from mice 24 h after treatment with centanamycin, and, importantly, we also detected this DNA damage in parasites within mosquitoes that had fed on these mice 10 days earlier. This demonstrates that damage to parasite DNA during blood-stage infection persists from the vertebrate to the mosquito host and provides a novel biochemical strategy to block malaria transmission.
- Published
- 2008
45. Adenylyl cyclase alpha and cAMP signaling mediate Plasmodium sporozoite apical regulated exocytosis and hepatocyte infection
- Author
-
Lisa A. Purcell, Purnima Bhanot, Laura Cabrita-Santos, Takeshi Ono, Lucy B. Andrews, Ana Rodriguez, Olga Diaz-Pulido, Ricardo Leitao, Takushi Tadakuma, Esther Bettiol, and Maria M. Mota
- Subjects
Plasmodium ,Vacuole ,Adenylyl cyclase ,Animals, Genetically Modified ,chemistry.chemical_compound ,Mice ,Cyclic AMP ,Internalization ,lcsh:QH301-705.5 ,media_common ,0303 health sciences ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Sporozoites ,Hepatocyte ,Signal transduction ,Plasmodium yoelii ,Research Article ,Adenylyl Cyclases ,Signal Transduction ,lcsh:Immunologic diseases. Allergy ,media_common.quotation_subject ,Movement ,Immunology ,Longevity ,Antigens, Protozoan ,Biology ,Microbiology ,Exocytosis ,03 medical and health sciences ,Virology ,parasitic diseases ,Genetics ,medicine ,Animals ,Humans ,Gene Silencing ,RNA, Messenger ,Uracil ,Molecular Biology ,030304 developmental biology ,030306 microbiology ,Infectious Diseases/Protozoal Infections ,biology.organism_classification ,Rats ,Mice, Inbred C57BL ,Disease Models, Animal ,chemistry ,lcsh:Biology (General) ,Hepatocytes ,Parasitology ,lcsh:RC581-607 - Abstract
Malaria starts with the infection of the liver of the host by Plasmodium sporozoites, the parasite form transmitted by infected mosquitoes. Sporozoites migrate through several hepatocytes by breaching their plasma membranes before finally infecting one with the formation of an internalization vacuole. Migration through host cells induces apical regulated exocytosis in sporozoites. Here we show that apical regulated exocytosis is induced by increases in cAMP in sporozoites of rodent (P. yoelii and P. berghei) and human (P. falciparum) Plasmodium species. We have generated P. berghei parasites deficient in adenylyl cyclase α (ACα), a gene containing regions with high homology to adenylyl cyclases. PbACα-deficient sporozoites do not exocytose in response to migration through host cells and present more than 50% impaired hepatocyte infectivity in vivo. These effects are specific to ACα, as re-introduction of ACα in deficient parasites resulted in complete recovery of exocytosis and infection. Our findings indicate that ACα and increases in cAMP levels are required for sporozoite apical regulated exocytosis, which is involved in sporozoite infection of hepatocytes., Author Summary Malaria is transmitted through the bite of an infected mosquito that deposits Plasmodium sporozoites under the skin. These sporozoites migrate from the skin into the circulation and then enter the liver to start a new infection inside hepatocytes. Sporozoites have the capacity to traverse mammalian cells. They breach their membranes and migrate through their cytosol. This process is required for infection of the liver and triggers the exposure of adhesive proteins in the apical end of sporozoites, a process that facilitates invasion of hepatocytes. We found that elevations of cAMP inside sporozoites mediate the exposure of adhesive proteins and therefore the infection process. Mutant sporozoites that do not express adenylyl cyclase, the enzyme that synthesizes cAMP, are not able to expose the adhesive proteins and their infectivity is reduced by half. Reinsertion of adenylyl cyclase gene in the mutant sporozoites recovers their capacity to expose adhesive proteins and to infect hepatocytes, confirming the specific role of this protein in infection. These results demonstrate the importance of cAMP and the exposure of adhesive proteins in sporozoites, but also show that Plasmodium sporozoites have other mechanisms to invade host hepatocytes that are not inhibited in the mutant parasites.
- Published
- 2008
46. Chemical Attenuation of Plasmodium berghei Sporozoites Induces Sterile Immunity in Mice▿
- Author
-
Stephanie K. Yanow, Lisa A. Purcell, Moses Lee, Terry W. Spithill, and Ana Rodriguez
- Subjects
Protozoan Vaccines ,Plasmodium berghei ,Immunology ,Antiprotozoal Agents ,Parasitemia ,Biology ,Vaccines, Attenuated ,Microbiology ,Plasmodium ,Cell Line ,Apicomplexa ,Mice ,In vivo ,Immunity ,parasitic diseases ,medicine ,Animals ,Mice, Inbred BALB C ,Cell Membrane ,Plasmodium falciparum ,medicine.disease ,biology.organism_classification ,Virology ,Mice, Inbred C57BL ,Infectious Diseases ,Liver ,Sporozoites ,Hepatocytes ,Parasitology ,Female ,Fungal and Parasitic Infections ,Malaria ,Locomotion - Abstract
Radiation and genetic attenuation of Plasmodium sporozoites are two approaches for whole-organism vaccines that protect against malaria. We evaluated chemical attenuation of sporozoites as an alternative vaccine strategy. Sporozoites were treated with the DNA sequence-specific alkylating agent centanamycin, a compound that significantly affects blood stage parasitemia and transmission of murine malaria and also inhibits Plasmodium falciparum growth in vitro. Here we show that treatment of Plasmodium berghei sporozoites with centanamycin impaired parasite function both in vitro and in vivo. The infection of hepatocytes by sporozoites in vitro was significantly reduced, and treated parasites showed arrested liver stage development. Inoculation of mice with sporozoites that were treated in vitro with centanamycin failed to produce blood stage infections. Furthermore, BALB/c and C57BL/6 mice vaccinated with treated sporozoites were protected against subsequent challenge with wild-type sporozoites. Our findings demonstrate that chemically attenuated sporozoites could be a viable alternative for the production of an effective liver stage vaccine for malaria.
- Published
- 2008
47. The effects of temperature on contractile mechanisms of rainbow trout (Salmo gairdneri) intestine
- Author
-
John F. Burka, Heather A. Briand, Gillian F. Calder, Rosalind M. J. Blair, and Lisa M. Purcell
- Subjects
Serotonin ,medicine.medical_specialty ,Trout ,Physiology ,chemistry.chemical_element ,Calcium ,Calcium in biology ,Potassium Chloride ,Contractility ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Pharmacology ,Calcium metabolism ,biology ,Temperature ,Muscle, Smooth ,General Medicine ,Smooth muscle contraction ,biology.organism_classification ,Intestines ,Endocrinology ,chemistry ,Carbachol ,Rainbow trout ,Extracellular Space ,Gastrointestinal Motility ,Intracellular ,Muscle Contraction - Abstract
Experiments were designed to determine whether contractility of trout smooth muscle in vitro varied with temperature and if changes occurred at the receptor or intracellular levels. The role of calcium in contractility at various temperatures was also investigated. Isolated trout intestinal segments, approximately 2 cm in length, were suspended isometrically under 2 g tension in 10-mL organ baths containing trout Ringer's solution aerated with O2 and CO2 (95:5). Contractions of trout intestine were not statistically different at 10 and 20 °C for carbachol, 5-hydroxytryptamine, and KCl. However, the efficacy, but not the potency, of each agonist was decreased at 2 °C. Receptor-induced contractions were reduced to a greater extent at 2 °C and did not recover to the same extent when returned to 10 °C in comparison with those induced by depolarization. The calcium source for contractility was also dependent on temperature. As temperatures increased, utilization of intracellular calcium increased, as indicated by increased contractility in the absence of extracellular calcium. Thus, low temperatures decrease smooth muscle contractility by affecting receptor-mediated events rather than the intracellular contractile mechanisms. Receptor-operated agonists appear to have a higher capability of using intracellular calcium than depolarizing agents.Key words: trout, intestine, smooth muscle, calcium, temperature, 5-hydroxytryptamine, muscarinic receptors, potassium.
- Published
- 1990
48. Contractile mechanisms of rainbow trout intestines: transmurally stimulated contractions
- Author
-
Lisa M. Purcell, John F. Burka, and Gregory A. Mitton
- Subjects
medicine.medical_specialty ,Endocrinology ,Internal medicine ,medicine ,Rainbow trout ,Aquatic Science ,Biology - Published
- 1992
49. Trout intestine as a model for studying gastrointestinal motility in teleosts
- Author
-
William P. Ireland, Heather A. Briand, Lisa M. Purcell, C. Chong, and John F. Burka
- Subjects
Pharmacology ,Trout ,biology ,Motility ,Zoology ,biology.organism_classification - Published
- 1990
50. Pandemic preparedness strategies must go beyond vaccines.
- Author
-
Gupta R, Purcell LA, Corti D, and Virgin HW
- Subjects
- Humans, Pandemics prevention & control, Disease Outbreaks prevention & control, Influenza, Human epidemiology, Vaccines
- Abstract
Monoclonal antibodies can fill a critical gap to help stop the next infectious disease outbreak from becoming the next pandemic.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.