51
|
Hamelin ME, Gagnon C, Prince GA, Kiener P, Suzich J, Ulbrandt N, Boivin G. Prophylactic and therapeutic benefits of a monoclonal antibody against the fusion protein of human metapneumovirus in a mouse model. Antiviral Res 2010; 88:31-7. [PMID: 20619294 DOI: 10.1016/j.antiviral.2010.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 05/31/2010] [Accepted: 07/01/2010] [Indexed: 11/28/2022]
Abstract
Human metapneumovirus (HMPV) is a paramyxovirus causing acute respiratory tract infections in humans. The effects of a monoclonal antibody (MAb 338, MedImmune, Inc.) directed against the HMPV fusion protein were assessed in vivo. Different groups of BALB/c mice received an intraperitoneal injection of 25 or 50mg/kg of MAb 338 either 24h before or 48h after viral infection. Lung samples were collected on days 5 and 42 after infection for determination of viral titers and histopathological changes. Pulmonary functions were also evaluated by plethysmography. On day 5 post-infection, lung viral titers were significantly decreased in mice treated with 25 or 50mg/kg before or after viral infection compared to HMPV-infected control mice. Similarly, HMPV copy numbers on day 42 were decreased for all prophylactic and therapeutic interventions. Histopathological changes were also less severe in all treated groups of mice on days 5 and 42 post-infection, correlating with decreased airways obstruction. Finally, on day 42, all treated groups had a significant decrease in airways hyperresponsiveness following treatment with MAb 338. Both prophylactic and, to a lesser extent, therapeutic administration of MAb 338 improved acute and late consequences of HMPV infection in a relevant mouse model.
Collapse
Affiliation(s)
- Marie-Eve Hamelin
- Research Center in Infectious Diseases of the Centre Hospitalier Universitaire de Québec and Laval University, 2705 Laurier blvd, Québec City, QC, Canada G1V 4G2
| | | | | | | | | | | | | |
Collapse
|
52
|
Fernández P, Trenholme A, Abarca K, Griffin MP, Hultquist M, Harris B, Losonsky GA. A phase 2, randomized, double-blind safety and pharmacokinetic assessment of respiratory syncytial virus (RSV) prophylaxis with motavizumab and palivizumab administered in the same season. BMC Pediatr 2010; 10:38. [PMID: 20525274 PMCID: PMC2898783 DOI: 10.1186/1471-2431-10-38] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 06/03/2010] [Indexed: 11/18/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) is an important pathogen causing annual epidemics of bronchiolitis and pneumonia among infants worldwide. High-risk infants currently receive RSV prophylaxis with palivizumab, a humanized RSV monoclonal antibody (MAb). In preclinical in vitro and in vivo (cotton-rat model) studies, motavizumab, a new RSV MAb, was shown to have greater anti-RSV activity than palivizumab. Motavizumab is currently under review for licensing approval. Since both MAbs may be available concurrently, this study evaluated their safety and tolerability when administered sequentially during the same RSV season. Methods Between April 2006 and May 2006, 260 high-risk infants were randomly assigned 1:1:1 to receive monthly intramuscular injections: 2 doses of motavizumab followed by 3 doses of palivizumab (M/P); 2 doses of palivizumab followed by 3 doses of motavizumab (P/M); or 5 doses of motavizumab (control). Adverse events (AEs, serious AEs [SAEs]), development of antidrug antibody (ADA), and serum drug trough concentrations were assessed. Results Most children received all 5 doses (246/260 [94.6%]) and completed the study (241/260 [92.7%]). While overall AE rates were similar (mostly level 1 or 2 in severity), SAEs and level 3 AEs occurred more frequently in the M/P group (SAEs: 22.9% M/P, 8.4% P/M, 11.8% motavizumab only; level 3 AEs: 15.7% M/P, 6.0% P/M, 6.5% motavizumab only). This trend in AE rates occurred before and after switching from motavizumab to palivizumab, suggesting a cause other than the combined regimen. Frequencies of AEs judged by the investigator to be related to study drug were similar among groups. Two deaths occurred on study (both in the M/P group, before palivizumab administration); neither was considered by the site investigator to be related to study drug. Mean serum drug trough concentrations were comparable among groups; ADA detection was infrequent (5.1% or less of any group). Conclusions The conclusions drawn from this study are limited by the small sample size per group. However, within this small study, overall AE rates, serum drug trough concentrations, and development of ADA associated with administering motavizumab and palivizumab sequentially to high-risk children appear comparable to administering motavizumab alone during the same RSV season. Trial Registration clinicaltrials.gov NCT00316264
Collapse
|
53
|
Carbonell-Estrany X, Simões EAF, Dagan R, Hall CB, Harris B, Hultquist M, Connor EM, Losonsky GA. Motavizumab for prophylaxis of respiratory syncytial virus in high-risk children: a noninferiority trial. Pediatrics 2010; 125:e35-51. [PMID: 20008423 DOI: 10.1542/peds.2008-1036] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Palivizumab reduces respiratory syncytial virus (RSV) hospitalization in children at high risk by approximately 50% compared with placebo. We compared the efficacy and safety of motavizumab, an investigational monoclonal antibody with enhanced anti-RSV activity in preclinical studies, with palivizumab. METHODS This randomized, double-blind, multinational, phase 3, noninferiority trial assessed safety and RSV hospitalization in 6635 preterm infants aged <or=6 months at enrollment or children aged <or=24 months with chronic lung disease of prematurity who received 15 mg/kg palivizumab or motavizumab monthly. Secondary end points included outpatient medically attended lower respiratory tract infections (MALRIs), RSV-specific LRIs, otitis media, antibiotic use, development of antimotavizumab antibodies, and motavizumab serum concentrations. RESULTS Motavizumab recipients had a 26% relative reduction in RSV hospitalization compared with palivizumab recipients, achieving noninferiority. Motavizumab was superior to palivizumab for reduction of RSV-specific outpatient MALRIs (50% relative reduction). Overall, adverse events (AEs) were not significantly different between groups. Cutaneous events were reported in 2 percentage points more motavizumab recipients (7.2% vs 5.1%); most were mild, but 0.3% resulted in dosing discontinuation. Antidrug antibodies (ADA) were detected in 1.8% of motavizumab recipients. Patients with anti-drug antibody reported 6 RSV events and 17 cutaneous events. CONCLUSIONS Children receiving prophylaxis with motavizumab or palivizumab had low rates of RSV hospitalization; motavizumab recipients experienced 50% fewer RSV MALRIs than palivizumab recipients. AEs were similar in both groups, although cutaneous AEs were higher for motavizumab recipients. Motavizumab may offer an improved alternative in prophylaxis for serious RSV disease in infants and children at high risk.
Collapse
Affiliation(s)
- Xavier Carbonell-Estrany
- Neonatology Service, Hospital Clínic, Agrupació Sanitèria Clínic, Hospital de Sant Joan de Déu, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Abstract
Motavizumab (MEDI-524, Numax) is a second generation monoclonal antibody (mAb) derived from palivizumab (Synagis) using affinity maturation techniques. Motavizumab is currently undergoing US Food and Drug Administration review as a treatment for respiratory syncytial virus (RSV) prophylaxis. It has been evaluated in large-scale clinical studies, and has demonstrated efficacy in reducing the disease burden of RSV in high-risk infant populations.
Collapse
Affiliation(s)
- Oya Cingoz
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
55
|
Abstract
Despite the considerable impact that respiratory syncytial virus (RSV) infections have in the health of children and adults, the development of therapeutic interventions has been remarkably slow. In the 1980s, advances in the characterisation of neutralising antibodies against RSV led to the development and clinical application of a polyclonal immunoglobulin preparation with high titres of neutralising antibodies against RSV (RSV IVIG). The next step was the development of neutralising monoclonal antibodies. Only palivizumab demonstrated clinical efficacy and was licensed in 1998 by the US FDA, and subsequently by the different world drug regulatory agencies. Motavizumab (MEDI-524) is a novel recombinant humanised IgG1 mAb derived from palivizumab with enhanced anti-RSV neutralising activity. In recent years, in addition to the anti-RSV antibodies, a number of alternative strategies against RSV are being developed. The most innovative approach is the use of siRNAs (small inhibitory RNAs) with specific anti-RSV activity.
Collapse
Affiliation(s)
- Octavio Ramilo
- University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
56
|
Sorce LR. Respiratory syncytial virus: from primary care to critical care. J Pediatr Health Care 2009; 23:101-8. [PMID: 19232926 DOI: 10.1016/j.pedhc.2007.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 11/28/2007] [Accepted: 11/29/2007] [Indexed: 10/22/2022]
Abstract
Respiratory syncytial virus (RSV) is a common disease in pediatrics. Certain subpopulations of children are at greatest risk for serious disease. However, previously healthy children also may become critically ill. In the clinic or the intensive care unit, children with RSV pose the challenge of how to treat a disease when evidence to support therapeutic options is severely limited. Prophylaxis is an option for certain children, although many do not qualify. RSV has been implicated in continued wheezing and the subsequent development of asthma. While evidence for this implication is still being sought, researchers are working on finding new ways to treat and prevent RSV.
Collapse
Affiliation(s)
- Lauren R Sorce
- Pediatric Critical Care, Children's Memorial Hospital, Chicago, IL, USA.
| |
Collapse
|
57
|
Abstract
Respiratory syncytial virus (RSV) is a common cause of respiratory tract infections. In severely immunosuppressed patients RSV can cause significant morbidity and mortality. The only FDA-approved drug for RSV is aerosolized ribavirin. Given the high morbidity and mortality in high-risk populations and inconsistent results with aerosolized ribavirin, new strategies for prevention and treatment of RSV are being sought. Palivizumab is an RSV-specific monoclonal antibody. A randomized, double-blind, placebo-controlled multicenter study showed significant reduction in hospitalization rates among children at high risk of RSV infection who had been given prophylactic palivizumab; these findings led to palivizumab's approval by the FDA in June 1998. Palivizumab also has a role in prevention of severe respiratory tract infections in high-risk infants. In immunocompromised patients, palivizumab has an excellent safety profile and may be beneficial in the prevention and treatment of RSV infections; however, clinical trials are needed to determine its effectiveness. In this article, we review the role of palivizumab in prevention and treatment of RSV infections in immunocompetent and immunocompromised patients.
Collapse
Affiliation(s)
- Georgiana Georgescu
- University of Texas MD Anderson Cancer Center, Department of Infectious Diseases, Infection Control and Employee Health, Houston, TX 77030, USA
| | | |
Collapse
|
58
|
Estripeaut D, Torres JP, Somers CS, Tagliabue C, Khokhar S, Bhoj VG, Grube SM, Wozniakowski A, Gomez AM, Ramilo O, Jafri HS, Mejias A. Respiratory syncytial virus persistence in the lungs correlates with airway hyperreactivity in the mouse model. J Infect Dis 2009; 198:1435-43. [PMID: 18828742 DOI: 10.1086/592714] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Previous studies in mice showed that respiratory syncytial virus (RSV) infection was associated with RSV RNA persistence. This study was designed to characterize the significance of RSV RNA persistence and its relation to RSV-induced chronic airway disease. METHODS Mice were inoculated with live RSV, UV light-treated RSV, heat-inactivated RSV, or medium. Bronchoalveolar lavage fluid samples were obtained and lung specimens were harvested on days 1, 5, and 42 after inoculation to assess lung inflammation, lung mRNA expression of interleukin (IL)-4, IL-5, IL-15, and interferon (IFN)-gamma; RSV loads were assessed by culture and real-time polymerase chain reaction (PCR) and correlated with pulmonary function. RESULTS During the acute phase of infection, RSV loads as indicated by culture and PCR were significantly higher in mice inoculated with live RSV. On day 42, RSV RNA remained detectable only in mice inoculated with live or UV light-treated RSV. Lung inflammation, IFN-gamma:IL-4 mRNA expression ratios, airway obstruction (AO), and airway hyperreactivity (AHR) were significantly increased in mice inoculated with live RSV. AO on day 5 and AHR on day 42 were significantly correlated with RSV RNA copy number in lung samples. CONCLUSIONS Infection with live RSV induced acute and chronic airway disease that was associated with a predominantly Th-1 immune response and RSV RNA persistence that significantly correlated with pulmonary function abnormalities.
Collapse
Affiliation(s)
- Dora Estripeaut
- Department of Pediatrics, University of Texas Southwestern Medical Center and Children's Medical Center Dallas, Texas 75390-9063, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Abstract
Several studies have described a clear association between respiratory syncytial virus (RSV) lower respiratory tract infection in infancy and the subsequent development of persistent wheezing in children. Using the mouse model we demonstrated that RSV induces long-term airway disease characterized by chronic airway inflammation and airway hyperreactivity (AHR). The RSV murine model offers great advantages to study the immunopathogenesis of RSV-induced long-term airway disease. Mice can be challenged with aerosolized methylcholine to determine the presence of AHR. We can apply the reverse transcription-polymerase chain reaction assay (RT-PCR) to detect RSV RNA in the respiratory tract and we can perform lung gene expression analysis to further characterize the chronic changes induced by RSV infection. Compared with sham-inoculated controls, RSV-infected mice developed chronic airway disease characterized by AHR and persistent airway inflammation. Forty-two days after RSV infection, a time point when RSV could no longer be isolated, RT-PCR demonstrated, quite unexpectedly, the presence of RSV RNA in the lower respiratory tract of mice. The presence of genomic RNA persisted for months after inoculation. Furthermore, preliminary studies also demonstrated that on day 42 there were a number of genes differentially expressed in RSV-infected mice compared with controls. RSV-infected mice with persistent AHR exhibited presence of abnormal chronic inflammatory changes, altered gene expression profiles, and persistence of RSV RNA, which may contribute to long-term airway disease induced by RSV. Future studies are needed to define the significance of persistent RSV RNA in the mouse model, and its potential role in the pathogenesis of RSV-induced persistent wheezing in children.
Collapse
|
60
|
Mejías A, Ramilo O. Review of palivizumab in the prophylaxis of respiratory syncytial virus (RSV) in high-risk infants. Biologics 2008; 2:433-9. [PMID: 19707374 PMCID: PMC2721379 DOI: 10.2147/btt.s3104] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In respiratory syncytial virus (RSV) disease the balance between the innate and adaptive immune responses determines the expression of the pathological phenotype favoring the development of acute bronchiolitis, and in certain children the development of recurrent wheezing. While humoral antibody plays a major role in protection against disease, T-cell immunity targeted to viral proteins appears to terminate viral infection. At the moment, treatment modalities for acute RSV infection do not effectively modify the course of the disease, and RSV vaccine development has shown conflicting results. To date, however, passive immunoprophylaxis with monoclonal antibodies is the only strategy that has demonstrated consistent efficacy in reducing RSV hospitalizations in high-risk children. The potential benefit of new strategies for prevention and treatment of RSV infections should be evaluated with respect to both the acute infection as well as the chronic respiratory manifestations induced by RSV.
Collapse
Affiliation(s)
- Asunción Mejías
- Division of Pediatric Infectious Diseases, The University of Texas, Southwestern Medical Center and Children’s Medical Center, Dallas, Texas, USA
| | - Octavio Ramilo
- Division of Pediatric Infectious Diseases, The University of Texas, Southwestern Medical Center and Children’s Medical Center, Dallas, Texas, USA
| |
Collapse
|
61
|
Kota S, Sabbah A, Chang TH, Harnack R, Xiang Y, Meng X, Bose S. Role of human beta-defensin-2 during tumor necrosis factor-alpha/NF-kappaB-mediated innate antiviral response against human respiratory syncytial virus. J Biol Chem 2008; 283:22417-29. [PMID: 18567888 PMCID: PMC2504899 DOI: 10.1074/jbc.m710415200] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 06/10/2008] [Indexed: 01/01/2023] Open
Abstract
Human respiratory syncytial virus (RSV) constitutes a highly pathogenic virus that infects lung epithelial cells to cause a wide spectrum of respiratory diseases. Our recent studies have revealed the existence of an interferon-alpha/beta-independent, innate antiviral response against RSV that was dependent on activation of NF-kappaB. We demonstrated that NF-kappaB inducing pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF) confers potent antiviral function against RSV in an NF-kappaB-dependent fashion, independent of interferon-alpha/beta. During our efforts to study this pathway, we identified HBD2 (human beta-defensin-2), a soluble secreted cationic protein as an antiviral factor induced during NF-kappaB-dependent innate antiviral activity in human lung epithelial cells. Our results demonstrated that HBD2 is induced by TNF and RSV in an NF-kappaB-dependent manner. Induction of HBD2 in infected cells was mediated by the paracrine/autocrine action of TNF produced upon RSV infection. HBD2 plays a critical role during host defense, because purified HBD2 drastically inhibited RSV infection. We also show that the antiviral mechanism of HBD2 involves blocking of viral cellular entry possibly because of destabilization/disintegration of the viral envelope. The important role of HBD2 in the innate response was also evident from loss of antiviral activity of TNF upon HBD2 silencing by short interfering RNA. The in vivo physiological relevance of HBD2 in host defense was apparent from induction of murine beta-defensin-4 (murine counterpart of HBD2) in lung tissues of RSV-infected mice. Thus, HBD2 functions as an antiviral molecule during NF-kappaB-dependent innate antiviral immunity mediated by the autocrine/paracrine action of TNF.
Collapse
Affiliation(s)
- Srikanth Kota
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | | | | | | | | | | | | |
Collapse
|
62
|
Ramilo O, Mejías A. Infecciones por virus respiratorio sincitial. Antiguos retos y nuevas estrategias. An Pediatr (Barc) 2008; 69:1-4. [DOI: 10.1157/13124210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
63
|
Skaricic D, Traube C, De B, Joh J, Boyer J, Crystal RG, Worgall S. Genetic delivery of an anti-RSV antibody to protect against pulmonary infection with RSV. Virology 2008; 378:79-85. [PMID: 18556039 DOI: 10.1016/j.virol.2008.04.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 03/28/2008] [Accepted: 04/08/2008] [Indexed: 11/16/2022]
Abstract
Respiratory syncytial virus (RSV) is a common cause of severe lower respiratory tract infections. Protection against infection with RSV can be achieved by monthly administration of the humanized monoclonal antibody palivizumab. The present study analyzes if genetic delivery of a murine version of palivizumab by single administration would achieve high-level and sustained antibody expression to protect mice against pulmonary infection with RSV. A murine version of the palivizumab antibody was constructed by replacing the human sequences with sequences from the constant region of a murine IgG1 antibody, while preserving the complementarity-determining region. As a proof-of-principle to test the validity of the strategy, the coding sequence for the heavy and light chains were cloned into a replication-defective serotype 5 human adenovirus vector (AdalphaRSV). Antibody expression and specificity for RSV was confirmed by Western analysis. To determine if AdalphaRSV would mediate production of anti-RSV antibodies in vivo, 5x10(10) particle units of AdalphaRSV or a control vector without transgene (AdNull), were administered intravenously to BALB/c mice. RSV neutralizing antibodies were detected in the serum after 4 days in mice receiving AdalphaRSV but not in AdNull-infected or naive mice (p<0.05). The mice that had received AdalphaRSV had at least 5.4-fold lower RSV titers in the lung 4 days following intranasal challenge with RSV compared to the AdNull or naive group (p<0.01). To evaluate long-term protection, the antibody construct was expressed in a non-human primate serotype rh.10 adeno-associated virus vector (AAVrh.10alphaRSV). RSV neutralizing antibodies were detected in serum and bronchoalveolar lavage fluid for up to 21 wk following intrapleural administration of AAVrh.10alphaRSV, but not with a control AAV vector expressing an unrelated transgene (AAVrh.10alpha1AT). Following challenge with RSV at 7 or 21 wk, 14.3-fold and 10.6-fold lower RSV titers were observed after 4 days in the lungs of mice that had received AAVrh.10alphaRSV compared to AAVrh.10alpha1AT (p<0.05). Together these data demonstrate that a gene transfer strategy for delivery of an anti-RSV antibody can generate protective immunity in mice against RSV infection in the respiratory tract and may provide an alternative to the administration of the antibody itself.
Collapse
Affiliation(s)
- Davor Skaricic
- Department of Pediatrics, Weill Medical College of Cornell University, New York, New York 10065, USA
| | | | | | | | | | | | | |
Collapse
|
64
|
Abstract
Palivizumab (Synagis®, MedImmune, Inc., USA) is a humanized monoclonal antibody that provides immunoprophylaxis against respiratory syncytial virus (RSV). RSV causes seasonal epidemics (winter or wet-season) of serious lower respiratory tract infections in young infants with subsequent increased frequency of recurrent wheezing during early childhood. Two large, double-blind, placebo-controlled trials including infants at high risk for severe RSV infection showed significant reduction of RSV-related hospitalizations: a 55% reduction in 1502 infants with prematurity and/or bronchopulmonary dysplasia and a 45% reduction in 1287 infants with hemodynamically significant congenital heart disease. Palivizumab proved to be as safe as an intramuscular injection of 15 mg/kg every 30 days for 5 months according to local epidemiology. Recent data suggest further benefits for palivizumab prophylaxis by reduction of recurrent post-RSV wheezing.
Collapse
Affiliation(s)
- Bernhard Resch
- Medical University Graz, Division of Neonatology, Pediatric Department, Auenbruggerplatz 30, 8036 Graz, Austria
| |
Collapse
|
65
|
Boivin G, Caouette G, Frenette L, Carbonneau J, Ouakki M, De Serres G. Human respiratory syncytial virus and other viral infections in infants receiving palivizumab. J Clin Virol 2008; 42:52-7. [PMID: 18164233 PMCID: PMC7172843 DOI: 10.1016/j.jcv.2007.11.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 11/12/2007] [Accepted: 11/14/2007] [Indexed: 01/14/2023]
Abstract
BACKGROUND Palivizumab is a humanized monoclonal antibody that prevents severe human respiratory syncytial virus (HRSV) infections. OBJECTIVES We determined the etiology of respiratory viral infections in palivizumab recipients, and monitored the clinical outcome and HRSV genotype in HRSV-infected infants. STUDY DESIGN Nasopharyngeal aspirates (NPAs) were collected from children receiving palivizumab who consulted or were hospitalized for acute respiratory tract infection (ARTI) during the 2004-2005 season. Viral cultures and multiplex RT-PCR for influenza A/B, HRSV and human metapneumovirus were performed. The fusion (F) gene of HRSV amplicons was also sequenced. RESULTS Among 116 enrolled patients, 51 (44%) had > or = 1 episode of ARTI for a total of 93 visits. At least one virus was identified in 33 (36%) of the 93 NPA samples; HRSV accounted for 11 (33%) of confirmed viral etiologies. Compared to subjects who had other viral ARTI, HRSV-positive subjects had less fever (p=0.01) and tended to have more bronchiolitis (p=0.07). Ten subjects (11 visits) developed HRSV infection, although only one was hospitalized. HRSV was detected after a median of 5.5 palivizumab doses and a median of 14 days after the last dose. One of the 11 HRSV strains tested had a F mutation located in the palivizumab-binding site. CONCLUSION HRSV is still a major cause of ARTI in children receiving palivizumab, although the outcome of infected children appears mild.
Collapse
Affiliation(s)
- Guy Boivin
- Research Center in Infectious Diseases, CHUQ-CHUL, Québec City, QC, Canada.
| | | | | | | | | | | |
Collapse
|
66
|
Prevention of allergic respiratory disease in infants: current aspects and future perspectives. Curr Opin Allergy Clin Immunol 2008; 7:547-55. [PMID: 17989533 DOI: 10.1097/aci.0b013e3282f14a17] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Primary and secondary prevention of severe atopic disease exemplified by atopic asthma represents an increasingly prominent focus of research in paediatric allergy. We review below the rationale for this approach and recent progress in development and testing of a range of potential preventive strategies. RECENT FINDINGS The principal areas reviewed relate to potential and currently available treatments targeting enhancement of overall immune functions, allergen-specific immunomodulation and respiratory viral infections, particularly during early childhood. SUMMARY The scientific rationale for prophylaxis of atopic diseases via early intervention strategies targeting young children is increasingly supported by findings from the experimental and clinical literature and significant progress may be expected in the mid-term future.
Collapse
|
67
|
Marasco WA, Sui J. The growth and potential of human antiviral monoclonal antibody therapeutics. Nat Biotechnol 2008; 25:1421-34. [PMID: 18066039 PMCID: PMC7097443 DOI: 10.1038/nbt1363] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Monoclonal antibodies (mAbs) have long provided powerful research tools for virologists to understand the mechanisms of virus entry into host cells and of antiviral immunity. Even so, commercial development of human (or humanized) mAbs for the prophylaxis, preemptive and acute treatment of viral infections has been slow. This is surprising, as new antibody discovery tools have increased the speed and precision with which potent neutralizing human antiviral mAbs can be identified. As longstanding barriers to antiviral mAb development, such as antigenic variability of circulating viral strains and the ability of viruses to undergo neutralization escape, are being overcome, deeper insight into the mechanisms of mAb action and engineering of effector functions are also improving the efficacy of antiviral mAbs. These successes, in both industrial and academic laboratories, coupled with ongoing changes in the biomedical and regulatory environments, herald an era when the commercial development of human antiviral mAb therapies will likely surge.
Collapse
Affiliation(s)
- Wayne A Marasco
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School 44, Binney Street, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
68
|
The fusion protein of respiratory syncytial virus triggers p53-dependent apoptosis. J Virol 2008; 82:3236-49. [PMID: 18216092 DOI: 10.1128/jvi.01887-07] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infection with respiratory syncytial virus (RSV) frequently causes inflammation and obstruction of the small airways, leading to severe pulmonary disease in infants. We show here that the RSV fusion (F) protein, an integral membrane protein of the viral envelope, is a strong elicitor of apoptosis. Inducible expression of F protein in polarized epithelial cells triggered caspase-dependent cell death, resulting in rigorous extrusion of apoptotic cells from the cell monolayer and transient loss of epithelial integrity. A monoclonal antibody directed against F protein inhibited apoptosis and was also effective if administered to A549 lung epithelial cells postinfection. F protein expression in epithelial cells caused phosphorylation of tumor suppressor p53 at serine 15, activation of p53 transcriptional activity, and conformational activation of proapoptotic Bax. Stable expression of dominant-negative p53 or p53 knockdown by RNA interference inhibited the apoptosis of RSV-infected A549 cells. HEp-2 tumor cells with low levels of p53 were not sensitive to RSV-triggered apoptosis. We propose a new model of RSV disease with the F protein as an initiator of epithelial cell shedding, airway obstruction, secondary necrosis, and consequent inflammation. This makes the RSV F protein a key target for the development of effective postinfection therapies.
Collapse
|
69
|
Hamelin ME, Couture C, Sackett M, Kiener P, Suzich J, Ulbrandt N, Boivin G. The Prophylactic Administration of a Monoclonal Antibody against Human Metapneumovirus Attenuates viral Disease and Airways Hyperresponsiveness in Mice. Antivir Ther 2008. [DOI: 10.1177/135965350801300104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Human metapneumovirus (hMPV) is one of the most frequent causes of respiratory tract infections in children. Our objective was to assess the prophylactic benefit of a monoclonal antibody (mAb) against the hMPV fusion protein in a murine model. Methods BALB/c mice received one intramuscular injection of either 5 or 10 mg/kg of mAb 338 (MedImmune, Inc.) and were infected intranasally 24 h later with 1x108 TCID50 (50% tissue culture infectious dose) of hMPV. On days 5 and 42 post-infection, lung samples were collected for determination of viral titres and for histopathological studies. Pulmonary function was characterized by plethysmography. Results Mean lung viral titres were significantly lower in mice treated with 5 or 10 mg/kg of mAb 338 compared with infected controls on day 5 (283, 45.6 and 1.49x105 TCID50/g, respectively; P<0.05). Similarly, lung viral RNA copies were significantly reduced in treated mice on day 42 (292, 101 and 607 copies per 0.01 g of lungs for mice that received 5 mg/kg, 10 mg/kg or no mAb, respectively; P<0.05). Histopathological changes characterized by important alveolar and interstitial inflammation were less severe in treated mice on days 5 and 42 compared with control. Airways obstruction was also significantly reduced in both treated groups on days 5 and 42, but development of hyperresponsiveness following the acute phase of infection was only significantly reduced in 10 mg/kg treated mice. Conclusions Prophylactic administration of mAb 338 attenuates acute and late consequences of hMPV disease in this mouse model.
Collapse
Affiliation(s)
- Marie-Eve Hamelin
- Research Center in Infectious Diseases of the ‘Centre Hospitalier Universitaire de Québec’ and Laval University, Québec City, Canada
| | - Christian Couture
- Anatomic Pathology Service, Laval Hospital and Cardiology-Pulmonary Institute of Laval University, Québec City, Canada
| | - Melanie Sackett
- Anatomic Pathology Service, Laval Hospital and Cardiology-Pulmonary Institute of Laval University, Québec City, Canada
| | | | | | | | - Guy Boivin
- Research Center in Infectious Diseases of the ‘Centre Hospitalier Universitaire de Québec’ and Laval University, Québec City, Canada
| |
Collapse
|
70
|
|
71
|
Beck A, Klinguer-Hamour C, Bussat MC, Champion T, Haeuw JF, Goetsch L, Wurch T, Sugawara M, Milon A, Van Dorsselaer A, Nguyen T, Corvaïa N. Peptides as tools and drugs for immunotherapies. J Pept Sci 2007; 13:588-602. [PMID: 17602441 DOI: 10.1002/psc.852] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Peptides are essential tools for discovery and pre-clinical and pharmaceutical development of viral and cancer vaccines ('active immunotherapies') as well as for therapeutic antibodies ('passive immunotherapies'). They help to trigger and analyze immune responses at a molecular level (B-cell, T-helper and CTL epitopes). They contribute largely to the design of new vaccine candidates and to the generation of monoclonal antibodies. They are also valuable analytical reference compounds for the structural characterisation by liquid chromatography and mass spectrometry of recombinant proteins used as biopharmaceuticals. As for other therapeutic applications, formulation, solubilisation, batch consistency and stability, issues have to be addressed to allow the pre-clinical and clinical development of this class of compounds as immunotherapeutic drugs. In the present review, three case studies dealing with (i) the design and the characterisation of Respiratory Syntycial Virus subunit vaccines, (ii) peptide-based melanoma vaccines, and (iii) therapeutic monoclonal antibodies, all investigated in clinical trials, are reported and discussed.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Epitopes, B-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/genetics
- Humans
- Immunotherapy/methods
- Mice
- Models, Immunological
- Models, Molecular
- Molecular Sequence Data
- Peptide Mapping
- Peptides/immunology
- Peptides/therapeutic use
- Respiratory Syncytial Virus Vaccines/genetics
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus, Human/genetics
- Respiratory Syncytial Virus, Human/immunology
Collapse
Affiliation(s)
- Alain Beck
- Centre d'Immunologie Pierre Fabre, 5 Avenue Napoléon III, F74164 Saint-Julien-en-Genevois, Cedex, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Motavizumab, a neutralizing anti-Respiratory Syncytial Virus (Rsv) monoclonal antibody significantly modifies the local and systemic cytokine responses induced by Rsv in the mouse model. Virol J 2007; 4:109. [PMID: 17961258 PMCID: PMC2222633 DOI: 10.1186/1743-422x-4-109] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Accepted: 10/25/2007] [Indexed: 11/10/2022] Open
Abstract
Motavizumab (MEDI-524) is a monoclonal antibody with enhanced neutralizing activity against RSV. In mice, motavizumab suppressed RSV replication which resulted in significant reduction of clinical parameters of disease severity. We evaluated the effect of motavizumab on the local and systemic immune response induced by RSV in the mouse model. Balb/c mice were intranasally inoculated with 106.5 PFU RSV A2 or medium. Motavizumab was given once intraperitoneally (1.25 mg/mouse) as prophylaxis, 24 h before virus inoculation. Bronchoalveolar lavage (BAL) and serum samples were obtained at days 1, 5 (acute) and 28 (long-term) post inoculation and analyzed with a multiplex assay (Beadlyte Upstate, NY) for simultaneous quantitation of 18 cytokines: IL-1alpha, IL-1beta, IL-2, IL-3, IL-4, IL-5, IL-6, KC (similar to human IL-8), IL-10, IL-12p40, IL-12p70, IL-13, IL-17, TNF-alpha, MCP-1, RANTES, IFN-gamma and GM-CSF. Overall, cytokine concentrations were lower in serum than in BAL samples. By day 28, only KC was detected in BAL specimens at low concentrations in all groups. Administration of motavizumab significantly reduced (p < 0.05) BAL concentrations of IL-1alpha, IL-12p70 and TNF-alpha on day 1, and concentrations of IFN-gamma on days 1 and 5 compared with RSV-infected untreated controls. In the systemic compartment, the concentrations of IL-10, IFN-gamma and KC were significantly reduced in the motavizumab-treated mice compared with the untreated controls. In summary, prophylactic administration of motavizumab was associated with significant reductions on RSV replication and concentrations of cytokine and chemokines, which are likely related to the improvement observed in clinical markers of disease severity.
Collapse
|
73
|
Feltes TF, Sondheimer HM. Palivizumab and the prevention of respiratory syncytial virus illness in pediatric patients with congenital heart disease. Expert Opin Biol Ther 2007; 7:1471-80. [PMID: 17727335 DOI: 10.1517/14712598.7.9.1471] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Respiratory syncytial virus (RSV) is a significant pathogen for infants and children with congenital heart disease. Non-sustained immunity and failure to develop an effective vaccine has steered RSV management toward a passive immunotherapy strategy in at-risk children. Palivizumab is a humanized murine monoclonal antibody targeting the RSV envelope F glycoprotein. In a Phase III clinical trial palivizumab significantly reduced RSV hospitalization in children with significant congenital heart disease and was proven to be safe. Palivizumab is one of the first monoclonal antibodies to significantly impact a pediatric disease.
Collapse
Affiliation(s)
- Timothy F Feltes
- The Ohio State University, Columbus Children's Hospital, 700 Children's Drive, ED 617, Columbus, Ohio 43205, USA.
| | | |
Collapse
|
74
|
Panitch HB. The relationship between early respiratory viral infections and subsequent wheezing and asthma. Clin Pediatr (Phila) 2007; 46:392-400. [PMID: 17556735 DOI: 10.1177/0009922806298641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Howard B Panitch
- University of Pennsylvania School of Medicine, Division of Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
75
|
Geskey JM, Thomas NJ, Brummel GL. Palivizumab: a review of its use in the protection of high risk infants against respiratory syncytial virus (RSV). Biologics 2007; 1:33-43. [PMID: 19707346 PMCID: PMC2721348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of hospitalization in children less than 1 year of age and causes substantial morbidity. Although there is not currently a vaccine available to prevent RSV infection, prophylaxis with the humanized monoclonal antibody palivizumab has been shown to reduce the rate of RSV hospitalization in premature infants and those infants with chronic lung disease or congenital heart disease. Because palivizumab has not been shown to have a beneficial clinical effect on established RSV disease such as reducing the rate of mechanical ventilation and mortality in children afflicted with RSV, there has been considerable debate as to the cost-benefit ratio of administering palivizumab according to international guidelines. Palivizumab has demonstrated a favorable side-effect profile in clinical trials without the development of anti-palivizumab antibodies. Future studies are needed to determine whether palivizumab, or other more potent monoclonal antibodies which are currently undergoing clinical trials, will reduce the long-term sequelae of RSV infection such as the development of wheezing and asthma.
Collapse
Affiliation(s)
| | - Neal J Thomas
- Critical Care Medicine, Department of Pediatrics, Department of Pediatrics and Pharmacy
| | - Gretchen L Brummel
- Pennsylvania State Children’s Hospital, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
76
|
Abstract
Originally considered as only a paediatric pathogen, respiratory syncytial virus (RSV) has recently been shown to be a significant cause of respiratory illness among elderly and high-risk adults. Approximately 170,000 hospitalizations and 10,000 deaths associated with RSV occur annually in people over the age of 65 years in the United States. Although rhinorrhoea and wheezing are common symptoms among adults, the clinical syndrome associated with RSV is not distinctive and thus laboratory methods are required for specific diagnosis. Presently, the combination of reverse transcription PCR and enzyme immunoassay serology offers the best sensitivity and specificity for diagnosis of RSV. Treatment options are limited at present, with inhaled ribavirin being the only licensed drug for use in hospitalized children. Vaccines against RSV remain an unachieved goal. Promising new agents that inhibit the virus-cell fusion, cell-cell fusion, or viral gene expression are currently in development.
Collapse
Affiliation(s)
- Yoshihiko Murata
- Infectious Diseases Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester Genera Hospital, New York, USA
| | - Ann R Falsey
- Infectious Diseases Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester Genera Hospital, New York, USA
| |
Collapse
|
77
|
Effect of respiratory syncytial virus on the activity of matrix metalloproteinase in mice. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200701010-00002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
78
|
Abstract
Monoclonal antibodies (mAbs) developed for either the prevention or treatment of viral diseases represent a small, but valuable, class of products. Since 1985, commercial firms have initiated clinical studies involving a total of 28 mAbs. To date, one product (palivizumab) has been approved and eight candidates are currently in clinical study. Most commercial mAbs studied as antiviral agents in the clinic have either directly or indirectly targeted human immunodeficiency virus, respiratory syncytial virus, or hepatitis C virus infections. However, the ability of mAbs to bind to specific targets and utilize various anti-infective modes of action would seem to make them well suited for the prevention and/or treatment of a wider variety of viral diseases. A number of factors, including the continuing need for innovative medicines for viral infections, the global spread of viral infections, and increased government funding for the study of pathogen countermeasures, have prompted companies to reconsider mAbs as antiviral agents. Public sector research into the use of mAbs against emerging pathogens, such as severe acute respiratory syndrome coronavirus, may have already provided candidates for further development.
Collapse
Affiliation(s)
- Janice M Reichert
- Tufts University, Tufts Center for the Study of Drug Development, Boston, Massachusetts 02111, USA.
| |
Collapse
|
79
|
Moore ML, Peebles RS. Respiratory syncytial virus disease mechanisms implicated by human, animal model, and in vitro data facilitate vaccine strategies and new therapeutics. Pharmacol Ther 2006; 112:405-24. [PMID: 16820210 DOI: 10.1016/j.pharmthera.2006.04.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 04/26/2006] [Indexed: 02/07/2023]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis, pneumonia, mechanical ventilation, and respiratory failure in infants in the US. No effective post-infection treatments are widely available, and currently there is no vaccine. RSV disease is the result of virus-induced airway damage and complex inflammatory processes. The outcome of infection depends on host and viral genetics. Here, we review disease mechanisms in primary RSV infection that are implicated by clinical studies, in vitro systems, and animal models. Defining RSV disease mechanisms is difficult because there is a wide range of RSV disease phenotypes in humans, and there are disparities in RSV disease phenotypes among the animal models of RSV infection. However, host factors identified by multiple lines of investigation as playing important roles in RSV pathogenesis are providing key insights. A better understanding of RSV molecular biology and RSV pathogenesis is facilitating rational vaccine design strategies and molecular targets for new therapeutics.
Collapse
Affiliation(s)
- Martin L Moore
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, T-1218 MCN, Vanderbilt University School of Medicine, Nashville, TN 37232-2650, USA
| | | |
Collapse
|
80
|
Sidwell RW, Barnard DL. Respiratory syncytial virus infections: Recent prospects for control. Antiviral Res 2006; 71:379-90. [PMID: 16806515 DOI: 10.1016/j.antiviral.2006.05.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 05/18/2006] [Accepted: 05/22/2006] [Indexed: 10/24/2022]
Abstract
Respiratory syncytial virus (RSV) infections remain a significant public health problem throughout the world, although recently developed and clinically approved anti-RSV antibodies administered prophylactically to at-risk populations appear to have significantly affected the disease development. Much effort has been expended to develop effective anti-RSV therapies, using both in vitro assay systems and mouse, cotton rat, and primate models, with several products now in various stages of clinical study. Several products are also being considered for the treatment of clinical symptoms of RSV. In this review, updates on the status of the approved anti-RSV antibodies, ribavirin, and recent results of studies with potential new anti-RSV compounds are summarized and discussed.
Collapse
Affiliation(s)
- Robert W Sidwell
- Institute for Antiviral Research, Utah State University, 5600 Old Main Hill, Logan, UT 84322-5600, United States.
| | | |
Collapse
|
81
|
Dall'Acqua WF, Kiener PA, Wu H. Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn). J Biol Chem 2006; 281:23514-24. [PMID: 16793771 DOI: 10.1074/jbc.m604292200] [Citation(s) in RCA: 446] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe here the functional implications of an increase in IgG binding to the neonatal Fc receptor. We have defined in a systematic fashion the relationship between enhanced FcRn binding of a humanized anti-respiratory syncytial virus (RSV) monoclonal antibody (MEDI-524) and the corresponding biological consequences in cynomolgus monkeys. The triple mutation M252Y/S254T/T256E (YTE) was introduced into the Fc portion of MEDI-524. Whereas these substitutions did not affect the ability of MEDI-524 to bind to its cognate antigen and inhibit RSV replication, they resulted in a 10-fold increase in its binding to both cynomolgus monkey and human FcRn at pH 6.0. MEDI-524-YTE was efficiently released from FcRn at pH 7.4 in both cases. We show that MEDI-524-YTE consistently exhibited a nearly 4-fold increase in serum half-life in cynomolgus monkeys when compared with MEDI-524. This constituted the largest half-life improvement described to date for an IgG in a primate. For the first time, we demonstrate that these sustained serum levels resulted in an up to 4-fold increase in lung bioavailability. Importantly, we also establish that our non-human primate model is relevant to human. Finally, we report that the YTE triple substitution provided a means to modulate the antibody-dependent cell-mediated cytotoxicity (ADCC) activity of a humanized IgG1 directed against the human integrin alpha(v)beta3. Therefore, the YTE substitutions allow the simultaneous modulation of serum half-life, tissue distribution and activity of a given human IgG1.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized
- Antibody Diversity
- Antibody-Dependent Cell Cytotoxicity
- Binding Sites, Antibody
- Half-Life
- Histocompatibility Antigens Class I/blood
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/chemistry
- Immunoglobulin G/metabolism
- Macaca fascicularis
- Organ Specificity
- Protein Binding
- Protein Engineering/methods
- Receptors, Fc/blood
- Receptors, Fc/metabolism
- Receptors, IgG/metabolism
Collapse
Affiliation(s)
- William F Dall'Acqua
- Department of Antibody Discovery and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland 20878, USA. dall'
| | | | | |
Collapse
|
82
|
Abstract
Antibodies constitute the most rapidly growing class of human therapeutics and the second largest class of drugs after vaccines. The generation of potent antibody therapeutics, which I review here, is an iterative design process that involves the generation and optimization of antibodies to improve their clinical potential.
Collapse
Affiliation(s)
- Paul J Carter
- Department of Antibody Technologies, Seattle Genetics Incorporated, 21823 30th Drive South East, Bothell, Washington 98021, USA.
| |
Collapse
|