Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Viruses ; 14(10)2022 10 12.
Article in English | MEDLINE | ID: mdl-36298792

ABSTRACT

Human rhinoviruses (HRVs) are small non-enveloped RNA viruses that belong to the Enterovirus genus within the Picornaviridae family and are known for causing the common cold. Though symptoms are generally mild in healthy individuals, the economic burden associated with HRV infection is significant. A vaccine could prevent disease. The Vero-cell-based viral vaccine platform technology was considered for such vaccine development. Unfortunately, most HRV strains are unable to propagate on Vero cells due to a lack of the major receptor of HRV group A and B, intercellular adhesion molecule (ICAM1, also known as CD54). Therefore, stable human ICAM1 expressing Vero cell clones were generated by transfecting the ICAM1 gene in Vero cells and selecting clones that overexpressed ICAM1 on the cell surface. Cell banks were made and expression of ICAM1 was stable for at least 30 passages. The Vero_ICAM1 cells and parental Vero cells were infected with four HRV prototypes, B14, A16, B37 and A57. Replication of all four viruses was detected in Vero_ICAM1, but not in the parental Vero cells. Altogether, Vero cells expressing ICAM1 could efficiently propagate the tested HRV strains. Therefore, ICAM1-expressing cells could be a useful tool for the development and future production of polyvalent HRV vaccines or other viruses that use ICAM1 as a receptor.


Subject(s)
Intercellular Adhesion Molecule-1 , Picornaviridae Infections , Rhinovirus , Vero Cells , Viral Vaccines , Animals , Humans , Chlorocebus aethiops , Enterovirus/genetics , Enterovirus/immunology , Enterovirus Infections/genetics , Enterovirus Infections/immunology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Picornaviridae Infections/genetics , Picornaviridae Infections/immunology , Rhinovirus/genetics , Rhinovirus/immunology , Vero Cells/immunology , Viral Vaccines/immunology
3.
Indian J Med Res ; 152(1 & 2): 82-87, 2020.
Article in English | MEDLINE | ID: mdl-32859866

ABSTRACT

BACKGROUND & OBJECTIVES: The global pandemic caused by SARS-CoV-2 virus has challenged public health system worldwide due to the unavailability of approved preventive and therapeutic options. Identification of neutralizing antibodies (NAb) and understanding their role is important. However, the data on kinetics of NAb response among COVID-19 patients are unclear. To understand the NAb response in COVID-19 patients, we compared the findings of microneutralization test (MNT) and plaque reduction neutralization test (PRNT) for the SARS-CoV-2. Further, the kinetics of NAb response among COVID-19 patients was assessed. METHODS: A total of 343 blood samples (89 positive, 58 negative for SARS-CoV-2 and 17 cross-reactive and 179 serum from healthy individuals) were collected and tested by MNT and PRNT. SARS-CoV-2 virus was prepared by propagating the virus in Vero CCL-81 cells. The intra-class correlation was calculated to assess the correlation between MNT and PRNT. The neutralizing endpoint as the reduction in the number of plaque count by 90 per cent (PRNT90) was also calculated. RESULTS: The analysis of MNT and PRNT quantitative results indicated that the intra-class correlation was 0.520. Of the 89 confirmed COVID-19 patients, 64 (71.9%) showed NAb response. INTERPRETATION & CONCLUSIONS: The results of MNT and PRNT were specific with no cross-reactivity. In the early stages of infection, the NAb response was observed with variable antibody kinetics. The neutralization assays can be used for titration of NAb in recovered/vaccinated or infected COVID-19 patients.


Subject(s)
Antibodies, Neutralizing/isolation & purification , Coronavirus Infections/blood , Neutralization Tests , Pandemics , Pneumonia, Viral/blood , Adolescent , Adult , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Child , Chlorocebus aethiops/immunology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Female , Humans , Immunoglobulin G/blood , Male , Middle Aged , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , Vero Cells/immunology , Young Adult
4.
Nat Med ; 26(9): 1428-1434, 2020 09.
Article in English | MEDLINE | ID: mdl-32661393

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has dramatically expedited global vaccine development efforts1-3, most targeting the viral 'spike' glycoprotein (S). S localizes on the virion surface and mediates recognition of cellular receptor angiotensin-converting enzyme 2 (ACE2)4-6. Eliciting neutralizing antibodies that block S-ACE2 interaction7-9, or indirectly prevent membrane fusion10, constitute an attractive modality for vaccine-elicited protection11. However, although prototypic S-based vaccines show promise in animal models12-14, the immunogenic properties of S in humans are poorly resolved. In this study, we characterized humoral and circulating follicular helper T cell (cTFH) immunity against spike in recovered patients with coronavirus disease 2019 (COVID-19). We found that S-specific antibodies, memory B cells and cTFH are consistently elicited after SARS-CoV-2 infection, demarking robust humoral immunity and positively associated with plasma neutralizing activity. Comparatively low frequencies of B cells or cTFH specific for the receptor binding domain of S were elicited. Notably, the phenotype of S-specific cTFH differentiated subjects with potent neutralizing responses, providing a potential biomarker of potency for S-based vaccines entering the clinic. Overall, although patients who recovered from COVID-19 displayed multiple hallmarks of effective immune recognition of S, the wide spectrum of neutralizing activity observed suggests that vaccines might require strategies to selectively target the most potent neutralizing epitopes.


Subject(s)
Antibodies, Neutralizing/pharmacology , Coronavirus Infections/immunology , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Antigens, Viral/immunology , COVID-19 , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/virology , Epitopes/immunology , Humans , Immunity, Cellular/immunology , Pandemics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/immunology , Vero Cells/immunology
5.
Vaccine ; 37(40): 5962-5971, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31471148

ABSTRACT

Type B Coxsackieviruses (CVBs) belong to the enterovirus genus, and they cause both acute and chronic diseases in humans. CVB infections usually lead to flu-like symptoms but can also result in more serious diseases such as myocarditis, aseptic meningitis and life-threatening multi-organ infections in young infants. Thus, CVBs have long been considered as important targets of future vaccines. We have previously observed CVB1 capsid disintegration and virus concentration decrease with 12-day long formalin inactivation protocol. Here a scalable ion exchange chromatography purification method was developed, and purified CVB1 was inactivated with UV-C or formalin. Virus morphology and concentration remained unchanged, when the UV (2 min) or formalin (5 days) inactivation were performed in the presence of tween80 detergent. The concentration of the native and UV inactivated CVB1 remained constant at 4 °C during a six months stability study, whereas the concentration of the formalin inactivated vaccine decreased 29% during this time. UV treatment decreased, whereas formalin treatment increased the thermal stability of the capsid. The formalin inactivated CVB1 vaccine was more immunogenic than the UV inactivated vaccine; the protective neutralizing antibody levels were higher in mice immunized with formalin inactivated vaccine. High levels of CVB1 neutralizing antibodies as well as IgG1 antibodies were detected in mice that were protected against viremia induced by experimental CVB1 infection. In conclusion, this study describes a scalable ion exchange chromatography purification method and optimized 5-day long formalin inactivation method that preserves CVB1 capsid structure and immunogenicity. Formalin treatment stabilizes the virus particle at elevated temperatures, and the formalin inactivated vaccine induces high levels of serum IgG1 antibodies (Th2 type response) and protective levels of neutralizing antibodies. Formalin inactivated CVB vaccines are promising candidates for human clinical trials.


Subject(s)
Coxsackievirus Infections/immunology , Coxsackievirus Infections/prevention & control , Enterovirus B, Human/immunology , Immunogenicity, Vaccine/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid/immunology , Capsid Proteins/immunology , Chlorocebus aethiops , Formaldehyde , Mice , Mice, Inbred C57BL , Ultraviolet Rays , Vaccination/methods , Vaccines, Inactivated/immunology , Vero Cells/immunology
6.
Hum Vaccin Immunother ; 14(4): 900-905, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29227177

ABSTRACT

Inactivated mouse-brain-derived Japanese encephalitis vaccine has a worrisome safety profile and the live attenuated vaccine is unsuitable in immunodeficiency. This study aimed to evaluate the immunogenicity and safety of an inactivated chromatographically purified Vero-cell-derived JE vaccine (CVI-JE, Beijing P-3 strain) in children. 152 healthy Thai children, with an average (SD) age of 14.4 (3.8) months, received 3 doses of CVI-JE on days 0, 7-28, and one year. Homologous JE neutralizing antibody titers (NT) were measured. All subjects had seroprotection [geometric mean titer (GMT) 150] 28 days' post 2nd vaccination. The seroprotection rates at 1 year after primary series and and 1 month after the booster were 89.3% (GMT 49) and 100% (GMT 621), respectively. Local and systemic reactions-fever (17.6%), vomiting (8%), and poor appetite (5.3%)-were noted within 28 days' post-vaccination. All these symptoms were self-limited. CONCLUSIONS: CVI-JE is safe, immunogenic, and provided high NT.


Subject(s)
Encephalitis, Japanese/prevention & control , Immunogenicity, Vaccine/immunology , Japanese Encephalitis Vaccines/adverse effects , Japanese Encephalitis Vaccines/immunology , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology , Vero Cells/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Child, Preschool , Chlorocebus aethiops , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/immunology , Female , Humans , Immunization, Secondary/methods , Infant , Male , Thailand , Vaccination/methods , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology
7.
Hum Vaccin Immunother ; 14(3): 615-622, 2018 03 04.
Article in English | MEDLINE | ID: mdl-29064728

ABSTRACT

Live attenuated influenza vaccine (LAIV)-based Vero cells could provide a better choice to control and prevent influenza virus infections. This study used the human influenza virus A/Yunnan/1/2005Vca(H3N2) (YN/05Vca) as a donor strain. YN/05Vca has a double phenotype of cold adaption (ca) and Vero cell adaption (va). The parental virus strain used was the wild-type A/Solomon Islands/3/2006 (H1N1) (SI/06wt). The study employed the modified classical reassortment method to generate a new virus strain. After co-infection of Vero cells, some different sub-types of the reassorted viruses were generated randomly. Then, the specific anti-serum (anti-YN/05Vca) could combine with and neutralize the donor virus, and the original parental virus could not grow in Vero cells at a low temperature until it was re-structured with the meaningful gene fragment from the donor virus in Vero cells. According to the plaques and RT-PCR results, a new monoclonal strain of Vero cell cold adaption virus was screened: SI/06Vca. After immunological and biological identification, this new strain virus could be used as a seed bank for LAIV, which has maintained surface antigenicity with SI/06wt. Consequently, this new Vero cell cold adaption virus SI/06Vca could be used for large-scale vaccine production with sufficient safety and efficacy, as confirmed by animal experiments with mice and ferrets.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Reassortant Viruses/immunology , Vaccines, Attenuated/immunology , Animals , Chlorocebus aethiops , Ferrets/immunology , Ferrets/virology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Mice , Vero Cells/immunology , Vero Cells/virology , Virus Replication/immunology
8.
Microbes Infect ; 17(3): 228-36, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25479555

ABSTRACT

Immunization programs have implemented live attenuated mumps vaccines which reduced mumps incidence ≥97%. Some of the vaccine strains were abandoned due to unwanted side effects and the genetic marker of attenuation has not been identified so far. Our hypothesis was that non-infectious viral particles, in particular defective interfering particles (DIPs), contribute to neuroattenuation. We showed that non-infectious particles of the mumps vaccine L-Zagreb attenuated neurovirulence of wild type mumps virus 9218/Zg98. Then, we attenuated recent wild type mumps virus MuVi/Zagreb.HRV/28.12 in Vero cells through 16 passages but already the fifth passage (p5) showed accumulation of DIPs and attenuated neurovirulence in a newborn rat model when compared to the second passage (p2). Sequence analysis of the p2 and p5 revealed a single mutation in the 5' untranslated region of the HN gene. Analysis of the expression level of the HN protein showed that this mutation does not affect the expression of the protein. We conclude that the passages of MuVi/Zagreb.HRV/28.12 in Vero cells for only three passages accumulated DIPs which attenuate neurovirulence. These findings reveal DIPs as a very promising and general neuroattenuating factor which should be considered in the rational design of the new mumps vaccine.


Subject(s)
Defective Viruses/immunology , Mumps virus/immunology , Virion , Animals , Base Sequence , Cell Line, Tumor , Chlorocebus aethiops , Humans , Molecular Sequence Data , Mumps virus/genetics , Rats , Vaccines, Attenuated/genetics , Vero Cells/immunology , Vero Cells/virology , Virulence/genetics
9.
J Virol ; 88(16): 9350-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920801

ABSTRACT

UNLABELLED: Respiratory syncytial virus (RSV) infection is the number one cause of bronchiolitis in infants, yet no vaccines are available because of a lack of knowledge of the infant immune system. Using a neonatal mouse model, we previously revealed that mice initially infected with RSV as neonates develop Th2-biased immunopathophysiologies during reinfection, and we demonstrated a role for enhanced interleukin-4 receptor α (IL-4Rα) expression on T helper cells in these responses. Here we show that RSV infection in neonates induced limited type I interferon (IFN) and plasmacytoid dendritic cell (pDC) responses. IFN alpha (IFN-α) treatment or adoptive transfer of adult pDCs capable of inducing IFN-α prior to neonatal RSV infection decreased Th2-biased immunopathogenesis during reinfection. A reduced viral load and downregulation of IL-4Rα on Th2 cells were observed in IFN-α-treated neonatal mice, suggesting dual mechanisms of action. IMPORTANCE: Respiratory syncytial virus (RSV) is the most significant cause of lower respiratory tract infection in infancy worldwide. Despite the dire need, we have failed to produce efficacious RSV vaccines or therapeutics. Part of the reason for this failure is our lack of understanding of how RSV interacts with the infant immune system to suppress the development of protective immunity. In the study described in the present paper, we used a neonatal mouse model, which more closely mimics human infants, to study the role of the innate immune system, particularly type I interferons (IFNs) and plasmacytoid dendritic cells (pDCs), in the pathogenesis of RSV infection. RSV infection in neonates induced limited type I IFN and pDC responses. IFN-α treatment or adoptive transfer of adult pDCs capable of producing IFN-α prior to neonatal RSV infection decreased Th2-biased immunopathogenesis during reinfection. These data suggest that IFN-α is a promising target for future RSV vaccine design.


Subject(s)
Dendritic Cells/immunology , Interferon-alpha/immunology , Interferon-alpha/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Dendritic Cells/metabolism , Dendritic Cells/virology , Humans , Lung/immunology , Lung/metabolism , Lung/virology , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/virology , Vero Cells/immunology , Vero Cells/metabolism , Vero Cells/virology , Viral Load/immunology
10.
J Virol ; 88(16): 9182-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899195

ABSTRACT

UNLABELLED: Neonatal immune responses to infection and vaccination are biased toward TH2 at the cost of proinflammatory TH1 responses needed to combat intracellular pathogens. However, upon appropriate stimulation, the neonatal immune system can induce adult-like TH1 responses. Here we report that a new class of vaccine adjuvant is especially well suited to enhance early life immunity. The GVI3000 adjuvant is a safe, nonpropagating, truncated derivative of Venezuelan equine encephalitis virus that targets dendritic cells (DCs) in the draining lymph node (DLN) and produces intracellular viral RNA without propagating to other cells. RNA synthesis strongly activates the innate immune response so that in adult animals, codelivery of soluble protein antigens induces robust humoral, cellular, and mucosal responses. The adjuvant properties of GVI3000 were tested in a neonatal BALB/c mouse model using inactivated influenza virus (iFlu). After a single immunization, mice immunized with iFlu with the GVI3000 adjuvant (GVI3000-adjuvanted iFlu) had significantly higher and sustained influenza virus-specific IgG antibodies, mainly IgG2a (TH1), compared to the mice immunized with antigen only. GVI3000 significantly increased antigen-specific CD4(+) and CD8(+) T cells, primed mucosal immune responses, and enhanced protection from lethal challenge. As seen in adult mice, the GVI3000 adjuvant increased the DC population in the DLNs, caused activation and maturation of DCs, and induced proinflammatory cytokines and chemokines in the DLNs soon after immunization, including gamma interferon (IFN-γ), tumor necrosis factor alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), and interleukin 6 (IL-6). In summary, the GVI3000 adjuvant induced an adult-like adjuvant effect with an influenza vaccine and has the potential to improve the immunogenicity and protective efficacy of new and existing neonatal vaccines. IMPORTANCE: The suboptimal immune responses in early life constitute a significant challenge for vaccine design. Here we report that a new class of adjuvant is safe and effective for early life immunization and demonstrate its ability to significantly improve the protective efficacy of an inactivated influenza virus vaccine in a neonatal mouse model. The GVI3000 adjuvant delivers a truncated, self-replicating viral RNA into dendritic cells in the draining lymph node. Intracellular RNA replication activates a strong innate immune response that significantly enhances adaptive antibody and cellular immune responses to codelivered antigens. A significant increase in protection results from a single immunization. Importantly, this adjuvant also primed a mucosal IgA response, which is likely to be critical for protection during many early life infections.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alphavirus/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , Immunity, Mucosal/immunology , Influenza A virus/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn/immunology , Animals, Newborn/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line , Chlorocebus aethiops/immunology , Chlorocebus aethiops/virology , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , T-Lymphocytes/virology , Vaccination/methods , Vaccines, Inactivated/immunology , Vero Cells/immunology , Vero Cells/virology
11.
Drugs ; 72(11): 1543-57, 2012 Jul 30.
Article in English | MEDLINE | ID: mdl-22788239

ABSTRACT

The influenza A subtype H5N1 virus is a likely causative agent for the next human influenza pandemic. Pandemic influenza vaccine production can begin only after a novel pandemic virus emerges. Cell-based vaccine production has advantages over conventional egg-based methods, allowing more rapid large-scale vaccine production. A reliable Vero cell culture system is available for pandemic and prepandemic influenza vaccine production. Prepandemic influenza vaccines are an important component of influenza pandemic preparedness plans, as their targeted use in the pandemic alert period or early in a pandemic is likely to mitigate the consequences of an influenza outbreak. Vepacel® is a prepandemic influenza vaccine (whole virion, Vero cell-derived, inactivated) containing antigen of H5N1 strain A/Vietnam/1203/2004 and is approved for use in the EU. Clinical immunogenicity studies with the vaccine have demonstrated good rates of functional neutralizing antibody responses against the vaccine strain (A/Vietnam/1203/2004), meeting established immunogenicity criteria for seasonal influenza vaccines, and cross-reactivity against H5N1 strains from other clades. In phase I/II and III studies, a heterologous (A/Indonesia/05/2005) booster vaccine administered to healthy adult and elderly volunteers 6-24 months after the two-dose priming vaccine (A/Vietnam/1203/2004) regimen induced good immunogenic responses against both H5N1 strains, demonstrating strong immunological memory. Broadly similar, albeit less robust, responses were observed in two special risk cohorts of immunocompromised and chronically ill patients. In general, adverse events observed in clinical immunogenicity studies with H5N1 vaccine (A/Vietnam/1203/2004) were similar to those reported with non-adjuvanted, inactivated, seasonal influenza vaccines.


Subject(s)
Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Vero Cells/immunology , Virion/immunology , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/immunology , Vaccines, Inactivated/therapeutic use
12.
J Virol Methods ; 163(1): 153-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19761798

ABSTRACT

Although the plaque reduction neutralization test (PRNT) is considered the "gold-standard" assay for measuring neutralizing antibodies for mumps, it is technically demanding, slow and requires large serum volumes, which limits its use for investigating mumps vaccine efficacy and population susceptibility. Therefore, an immunocolourimetric-based focus reduction neutralization test (FRNT) was developed and validated against PRNT using 30 blood donor plasma samples (16 positive, 5 equivocal, and 9 negative for mumps IgG by EIA). The samples were tested in triplicate by FRNT and PRNT in 10 and 4 separate assay runs, respectively, and 50% neutralizing antibody titres calculated using the Kärber formula. There was good correlation between the two neutralization assays (R(2)=0.88). Inter-assay variation for FRNT titres was 2-fold, compared to a 3-fold variation for PRNT titres. From the distribution of results, a positive cut-off for FRNT was defined as 1:4. In conclusion, FRNT has similar sensitivity to the PRNT and offers the advantage of speed (2 days vs. 7 days), reduced sample volume (40 microL vs. 150 microL), and the possibility of automation using 96-well plates. FRNT appears to be a good substitute for PRNT for characterising the immune response to mumps and for vaccine efficacy studies.


Subject(s)
Antibodies, Neutralizing/blood , Mumps virus/immunology , Mumps/immunology , Neutralization Tests/methods , Animals , Cell Line , Chlorocebus aethiops , Humans , Mumps/blood , Mumps/virology , Vero Cells/immunology , Vero Cells/virology , Viral Plaque Assay
13.
Clin Exp Immunol ; 153(3): 439-47, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18505424

ABSTRACT

Clostridium difficile induces mucosal inflammation via secreted toxins A and B and initial interactions between the toxins and intestinal epithelial cells (which lead to loss of barrier function) are believed to be important in disease pathogenesis. Secreted toxin-specific antibodies may inhibit such interactions. Using the Caco-2 epithelial cell line, we have investigated the use of an anti-toxin A monoclonal antibody (ATAA) in providing protection against toxin A-mediated disruption of epithelial barrier function (assessed by measurement of transepithelial electrical resistance and luminal to basolateral flux of labelled dextran). In contrast to free antibody, ATAA conjugated to sepharose beads was more effective in neutralizing the activity of purified toxin A. Sepharose bead-conjugated ATAA was subsequently used to investigate the contribution of toxin A in epithelial injury mediated by C. difficile supernatant samples (containing toxins A, B and other products). Loss of barrier function mediated by apical application of supernatant samples of reference and epidemic 027 strains of C. difficile was abrogated by neutralization of toxin A. However, this was not the case when the supernatant samples were applied to the basal surface of epithelial monolayers. In conclusion, our studies have shown that (i) sepharose bead-conjugated ATAA is more effective in neutralizing toxin A than free antibody and (ii) when the apical (luminal) surface of epithelial monolayers is exposed to the secretory products of reference and 027 strains of C. difficile, toxin A is required for the initial injury that leads to loss of barrier function.


Subject(s)
Antibodies, Monoclonal/pharmacology , Clostridioides difficile/pathogenicity , Enterotoxins/toxicity , Epithelial Cells/drug effects , Immunotoxins/pharmacology , Intestines/drug effects , Animals , Bacterial Toxins , Caco-2 Cells , Chlorocebus aethiops , Humans , Intestinal Mucosa/drug effects , Sepharose/pharmacology , Vero Cells/immunology
14.
Cancer Gene Ther ; 9(3): 289-95, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11896446

ABSTRACT

BACKGROUND: Systemic IL-2 has shown some activity in metastatic melanoma, but its use is severely limited by toxicity. TG2001 is a product in which the human IL-2 cDNA was incorporated into the genome of Vero cells, a monkey fibroblast cell line. The goal of this intratumorally applied therapy was to create an antitumor immune response stimulated by xeno-antigens and local production of IL-2 in the close vicinity of tumor-specific antigens. TG2001 was reported to have a good safety profile in two previous dose-escalating phase I studies performed in 18 patients with various solid tumors, with encouraging clinical responses in three patients. The objectives of this study were to evaluate the tolerance and incidence of tumor regression in patients with metastatic melanoma, following repeated administration of Vero-IL-2 cells. PATIENTS AND METHODS: This was on open-label, randomized phase II study comparing two doses of Vero-IL-2, 5x10(5) and 5x10(6) cells. Twenty-eight patients with metastatic melanoma were enrolled in the study, 14 in each treatment group. Patients received TG2001 by intratumoral injection on days 1, 3, and 5 every 4 weeks for four cycles, and every 8 weeks thereafter, until evidence of progressive disease (PD). Criteria for patient selection included histologically proven metastatic melanoma, with one tumor accessible for product administration, and at least another tumor site for response assessment. Evaluation included tumor measurements, humoral and T cell-mediated local and systemic immune response, humoral response to Vero cells, adverse events and standard laboratory parameters. RESULTS: None of the patients achieved a confirmed objective response. Stable disease (SD) was seen in six (43%) and eight patients (57%) at the 5x10(5) and the 5x10(6) dose level, respectively. Two patients, one in each group, died during the study (i.e., within 1 month after the last injection) due to PD. Three patients exhibited antibody responses to Vero cells. T-cell immunity, serum cytokine levels and cytokine mRNA expression in tumor biopsies did not show meaningful alterations after therapy, except for a trend toward an increase in intratumoral TH2 cytokine (IL-4 and/or IL-10) levels. The study drug was well tolerated at both dose levels and side effects mainly consisted of injection site pain and erythema, and pyrexia. CONCLUSION: The intratumoral administration of TG2001 was generally well tolerated in patients with metastatic melanoma, and transient disease stabilization was observed in 50% of patients.


Subject(s)
Immunotherapy/methods , Interleukin-2/biosynthesis , Melanoma/therapy , Neoplasms/therapy , Vero Cells/metabolism , Adult , Aged , Animals , Chlorocebus aethiops , Dose-Response Relationship, Drug , Female , Humans , Immunity , Injections, Intralesional , Interleukin-2/genetics , Male , Melanoma/diagnostic imaging , Melanoma/secondary , Middle Aged , RNA, Messenger/metabolism , Radionuclide Imaging , T-Lymphocytes/immunology , Transplantation, Heterologous , Treatment Outcome , Vero Cells/immunology
15.
Rev Latinoam Microbiol ; 43(3): 123-9, 2001.
Article in English | MEDLINE | ID: mdl-17061498

ABSTRACT

Mitogenic and antigenic lymphocyte stimulation was examined in pigs that had been immunized with 2 inactivated vaccines which had been prepared with Aujesky's disease virus (ADV), strain RC/79. One vaccine was partially purified by ultra-centrifugation (Vaccine A) the other one was not (Vaccine B). A second dose of vaccine had no influence over the blastogenic response when the lymphocytes were stimulated with phytohemoagglutinin (PHA). Lymphocyte response to the ADV antigen in the immunized pigs was significantly higher at day 30 post inoculation than at day 0 indicating that it was highly specific. Cellular antigens contained in the viral cultures produced a slight non-specific response as shown by a low increase in the levels of lymphocyte blastic transformation (LBT) in the control group at day 30 p.i., this group only received a non infected Vero cell suspension. This was the case in pigs that received vaccine A as well as in those that were vaccinated with vaccine B. Vaccine B contains a greater quantity of contaminating cellular antigens, since it is an impure vaccine. Such antigens could act as non-specific immunomodulators, potentiating cell-mediated immunity (CMI). This assay demonstrated that inactivated vaccines produced with VPR-RC/79, partially purified and unpurified are capable of inducing a humoral immune response. The blastogenic reaction of the peripheral blood lymphocytes to antigens of ADV strain RC/79, indicated that the employed immunogens also induced the CMI. Results indicate that the analyzed immunogens could be considered for the possible implementation of epidemiological measures, which imply the use of vaccines to prevent pseudo-rabies in Argentina.


Subject(s)
Herpesvirus 1, Suid/immunology , Lymphocyte Activation , Pseudorabies/prevention & control , Sus scrofa/immunology , Swine Diseases/prevention & control , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibody Formation , Antigens, Surface/immunology , Antigens, Viral/immunology , Chlorocebus aethiops , Female , Immunity, Cellular , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Phytohemagglutinins/pharmacology , Vaccines, Inactivated/immunology , Vaccines, Inactivated/isolation & purification , Vero Cells/immunology , Viral Vaccines/isolation & purification
16.
J Immunol ; 165(8): 4528-36, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035093

ABSTRACT

The envelope surface glycoprotein C (gC) of HSV-1 interferes with the complement cascade by binding C3 and activation products C3b, iC3b, and C3c, and by blocking the interaction of C5 and properdin with C3b. Wild-type HSV-1 is resistant to Ab-independent complement neutralization; however, HSV-1 mutant virus lacking gC is highly susceptible to complement resulting in > or =100-fold reduction in virus titer. We evaluated the mechanisms by which complement inhibits HSV-1 gC null virus to better understand how gC protects against complement-mediated neutralization. C8-depleted serum prepared from an HSV-1 and -2 Ab-negative donor neutralized gC null virus comparable to complement-intact serum, indicating that C8 and terminal lytic activity are not required. In contrast, C5-depleted serum from the same donor failed to neutralize gC null virus, supporting a requirement for C5. EDTA-treated serum did not neutralize gC null virus, indicating that complement activation is required. Factor D-depleted and C6-depleted sera neutralized virus, suggesting that the alternative complement pathway and complement components beyond C5 are not required. Complement did not aggregate virus or block attachment to cells. However, complement inhibited infection before early viral gene expression, indicating that complement affects one or more of the following steps in virus replication: virus entry, uncoating, DNA transport to the nucleus, or immediate early gene expression. Therefore, in the absence of gC, HSV-1 is readily inhibited by complement by a C5-dependent mechanism that does not require viral lysis, aggregation, or blocking virus attachment.


Subject(s)
Antibodies, Viral/physiology , Herpesvirus 1, Human/immunology , Adult , Animals , Antibodies, Viral/blood , Chlorocebus aethiops , Complement C5/physiology , Complement C8/physiology , Complement Pathway, Alternative/immunology , Disaccharides/immunology , Gene Expression Regulation, Viral/immunology , Genes, Immediate-Early/immunology , HeLa Cells/immunology , HeLa Cells/metabolism , HeLa Cells/virology , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/ultrastructure , Humans , Neutralization Tests , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/immunology , Vero Cells/immunology , Vero Cells/metabolism , Vero Cells/virology , Viral Envelope Proteins/deficiency , Viral Envelope Proteins/genetics
17.
Cancer Immunol Immunother ; 48(6): 321-30, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10473807

ABSTRACT

Eleven patients with advanced cancer were treated in a clinical gene therapy trial by repeated intra- tumoral injections with different doses of xenogenic fibroblasts secreting high amounts of human interleukin-2 (Vero-IL2). Treatments in a total of 14 courses were well tolerated and resulted in clinical responses and measurable biological effects. Together with increases in serum interleukin-2 (IL-2), modifications of the V-beta T cell receptor repertoire and induction of intratumoral T-cell infiltration were observed. When the intratumoral expression of endogenous cytokine genes and the persistence of the IL-2 transgene at the application site and in peripheral blood were investigated, rapid disappearance of the transgene at the application site appeared to be the most prominent biological effect. Tests detecting a single Vero-IL2 cell against a background of 10(5) non-transfected cells were not able to demonstrate significant expression of exogenous IL-2 (i.e. the transgene or transgene-carrying cells) in tumor biopsies or blood at different times. Therefore, further studies were performed to evaluate the mechanism(s) involved in the rapid disappearance of xenogenic carrier cells in more detail. We show here that significant in vitro cytotoxicity against transgene-carrying Vero cells can be observed in peripheral blood of all the patients before treatment as well as in healthy controls. "Cold" target inhibition shows that significant killing of Vero-IL2 cells is mediated by natural killer (NK) cells. This was confirmed by showing that established CD3(-)/CD16(+)/CD56(+) peripheral blood NK cell clones kill both K562 and Vero-IL2 target cells. The failure of other mechanisms (complement, antibody-dependent cell cytotoxicity or cytotoxic T lymphocytes) to destroy xenogenic, histoincompatible Vero cells in vitro suggests that NK cells also might be responsible for the killing of Vero-IL2 in vivo and for the failure to detect the transgene at the application site. These results might also be of importance for some aspects of the current discussion of xenotransplantation.


Subject(s)
Genetic Therapy , Interleukin-2/genetics , Neoplasms/therapy , Transplantation, Heterologous , Vero Cells/transplantation , Adult , Aged , Animals , Chlorocebus aethiops , Female , Gene Expression , Graft Rejection/immunology , Humans , Injections, Intralesional , Interleukin-2/blood , Interleukin-2/metabolism , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Male , Middle Aged , Neoplasms/immunology , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/metabolism , Safety , Transfection , Transplantation, Heterologous/immunology , Treatment Outcome , Vero Cells/immunology , Vero Cells/metabolism
18.
Mem. Inst. Oswaldo Cruz ; 94(suppl.1): 135-7, Sept. 1999. graf
Article in English | LILACS | ID: lil-245604

ABSTRACT

Through its life cycle from the insect vector to mammalian hosts Trypanosoma cruzi has developed clever strategies to reach the intracellular milieu where it grows sheltered from the hosts' immune system. We have been interested in several aspects of in vitro interactions of different infective forms of the parasite with cultured mammalian cells. We have observed that not only the classically infective trypomastigotes but also amastigotes, originated from the extracellular differentiation of trypomastigotes, can infect cultured cells. Interestingly, the process of invasion of different parasite infective forms is remarkably distinct and also highly dependent on the host cell type.


Subject(s)
Animals , Mammals/immunology , Trypanosoma cruzi/immunology , Coxiella burnetii , Epitopes , Gelsolin , HeLa Cells/immunology , Trypanosoma cruzi/cytology , Vero Cells/immunology
19.
Immunol Lett ; 63(2): 121-3, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9761374

ABSTRACT

The K-562 cell line is widely known and used as a NK cell target. In this study we report that VERO (African green monkey kidney epithelial cell line) is an excellent target of the human NK cell cytotoxicity. Considerable cytotoxicity was observed in a 4 h 51Cr release assay with nonadherent and immunomagnetically separated CD56+ NK cells from PBMC. On the contrary, adding K-562 cells as cold target to the assay the cytotoxicity significantly decreased. Using a standard chromium-release assay the NK cell activity (NKCA) against VERO cells was investigated in a population of healthy volunteers (mean value of cytotoxicity was 26.6%) and compared with the values of cytotoxicity against K-562 target cells (32.6%). The difference was not significant (P > 0.05). The suppressive effect of PTX on in vitro NK cell activity was observed at concentration of 100 microg/ml using VERO target cells as well as K-562 cells. Our studies provide the first evidence that the NK cell activity is suppressed in vitro by PTX using VERO cells as NK target cells.


Subject(s)
Immunosuppressive Agents/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Pentoxifylline/pharmacology , Vero Cells/immunology , Adult , Animals , Chlorocebus aethiops , Cytotoxicity, Immunologic/drug effects , Female , Humans , Male
20.
Nat Biotechnol ; 16(3): 292-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9528012

ABSTRACT

Transgenic potatoes were engineered to synthesize a cholera toxin B subunit (CTB) pentamer with affinity for GMI-ganglioside. Both serum and intestinal CTB-specific antibodies were induced in orally immunized mice. Mucosal antibody titers declined gradually after the last immunization but were restored following an oral booster of transgenic potato. The cytopathic effect of cholera holotoxin (CT) on Vero cells was neutralized by serum from mice immunized with transgenic potato tissues. Following intraileal injection with CT, the plant-immunized mice showed up to a 60% reduction in diarrheal fluid accumulation in the small intestine. Protection against CT was based on inhibition of enterotoxin binding to the cell-surface receptor GMI-ganglioside. These results demonstrate the ability of transgenic food plants to generate protective immunity in mice against a bacterial enterotoxin.


Subject(s)
Cholera Toxin/genetics , Cholera Toxin/immunology , Plants, Genetically Modified/genetics , Vaccines/pharmacology , Animals , Antibodies/blood , Chlorocebus aethiops , Cholera/complications , Cholera/immunology , Diarrhea/complications , Diarrhea/immunology , Diarrhea/therapy , Female , G(M1) Ganglioside/metabolism , Immune Sera , Mice , Mice, Inbred Strains , Mucous Membrane/immunology , Solanum tuberosum/genetics , Vaccines/administration & dosage , Vaccines/genetics , Vero Cells/immunology , Vero Cells/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...