Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Clin Microbiol Rev ; 34(2)2021 03 17.
Article in English | MEDLINE | ID: mdl-33568554

ABSTRACT

Influenza poses a significant burden on society and health care systems. Although antivirals are an integral tool in effective influenza management, the potential for the emergence of antiviral-resistant viruses can lead to uncertainty and hesitation among front-line prescribers and policy makers. Here, we provide an overview of influenza antiviral resistance in context, exploring the key concepts underlying its development and clinical impact. Due to the acute nature of influenza in immunocompetent patients, resistant viruses that develop during antiviral treatment of a single patient ("treatment-emergent resistance") are usually cleared in a relatively short time, with no impact on future antiviral efficacy. In addition, although available data are limited by small numbers of patients, they show that antiviral treatment still provides clinical benefit to the patient within whom resistance emerges. In contrast, the sustained community transmission of resistant variants in the absence of treatment ("acquired resistance") is of greater concern and can potentially render front-line antivirals ineffective. Importantly, however, resistant viruses are usually associated with reduced fitness such that their widespread transmission is relatively rare. Influenza antivirals are an essential part of effective influenza management due to their ability to reduce the risk of complications and death in infected patients. Although antiviral resistance should be taken seriously and requires continuous careful monitoring, it is not comparable to antibiotic resistance in bacteria, which can become permanent and widespread, with far-reaching medical consequences. The benefits of antiviral treatment far outweigh concerns of potential resistance, which in the vast majority of cases does not have a significant clinical impact.


Subject(s)
Antiviral Agents , Influenza, Human , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral , Humans , Influenza, Human/drug therapy
2.
Hum Vaccin Immunother ; 16(9): 2051-2055, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32783768

ABSTRACT

In 1914, the concept of a prophylactic vaccine, administered to a person before the disease had been contracted, was still controversial. Nevertheless, Almroth Wright tested new pneumococcus vaccines in South Africa, where the incidence of bacterial pneumonia was high amongst workers in the gold mines. He established the use of clinical trials, using around ten thousand workers, both in vaccinated and unvaccinated groups. The two groups were not matched to modern standards. Also, of course, those workers in the control unvaccinated group could not be protected: but some considered a prophylactic vaccine would exacerbate the disease. The vaccines were manufactured to contain a range of pneumococci from different clinical samples, in a serious attempt to match the microbes in the vaccine to the field bacteria. Deaths were averted by the vaccine; and side effects were noted to be minimal. Reexamination of pathology samples from the Spanish Influenza Pandemic showed quite clearly the contribution of pneumococci and streptococci to the mortality of over fifty million people in 1918-1919. The microbe causing this Pandemic was isolated in 1933, and was shown to be a true virus; this finding initiated a huge expanse and interest in influenza virus vaccines, both killed and live. A chance discovery allowed the purification of Influenza M and NP proteins then permitted the production of experimental vaccines. These vaccines were formulated to induce and B and/or T cell responses to the internal proteins. Several of these Universal Influenza Vaccines have been tested in quarantine, and have now reached Phase III trials in the community.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , Influenza, Human/prevention & control , Male , Pandemics , Quarantine , South Africa
3.
Hum Vaccin Immunother ; 15(9): 2009-2012, 2019.
Article in English | MEDLINE | ID: mdl-31121112

ABSTRACT

When we reconsider the virology and history of the Spanish Influenza Pandemic, the science of 2018 provides us with tools which did not exist at the time. Two such tools come to mind. The first lies in the field of 'gain of function' experiments. A potential pandemic virus, such as influenza A (H5N1), can be deliberately mutated in the laboratory in order to change its virulence and spreadability. Key mutations can then be identified. A second tool lies in phylogenetics, combined with molecular clock analysis. It shows that the 1918 pandemic virus first emerged in the years 1915-1916. We have revisited the literature published in Europe and the United States, and the notes left by physicians who lived at the time. In this, we have followed the words of the late Alfred Crosby: who wrote that "contemporary documentary evidence from qualified physicians" is the key to understanding where and how the first outbreaks occurred. In our view, the scientists working in Europe fulfill Crosby's requirement for contemporary evidence of origin. Elsewhere, Crosby also suggested that "the physicians of 1918 were participants in the greatest failure of medical science in the twentieth century". Ours is a different approach. We point to individual pathologists in the United States and in France, who strove to construct the first universal vaccines against influenza. Their efforts were not misdirected, because the ultimate cause of death in nearly all cases flowed from superinfections with respiratory bacteria.


Subject(s)
Bacterial Infections/mortality , Coinfection/mortality , Coinfection/prevention & control , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Military Personnel , Bacterial Infections/epidemiology , Bacterial Infections/prevention & control , Coinfection/microbiology , Europe/epidemiology , Health Personnel , History, 20th Century , Humans , Influenza, Human/history , Pandemics/history , Superinfection/epidemiology , Superinfection/microbiology , Superinfection/prevention & control , United States/epidemiology , Virology
4.
Lancet Infect Dis ; 18(11): e348-e354, 2018 11.
Article in English | MEDLINE | ID: mdl-29935779

ABSTRACT

The influenza epidemic of 1918 represented the greatest failure of medical science in the 20th century. Fortunately, research throughout subsequent years has been making amends. Some studies have applied RT-PCR to the tissue samples from that time, whereas others have reconstructed the pathogen in its virulent state. But the resurrection of the 1918 influenza virus leaves questions unanswered: although more virulent than contemporary H1N1 epidemic viruses in animal models, this increased virulence of the 1918 influenza virus is not sufficient to have been the sole cause of the high mortality rates recorded in humans during the epidemic. Thus, other hypotheses have been investigated. The immune history of the different age groups exposed at the time to the pandemic virus could be a factor, and the notion of original antigenic sin provides an explanation for the unusual pattern of deaths. The presence, or absence, of a cytokine storm in the lungs of young adults might also be involved. The time and location that the 1918 influenza pandemic first emerged from its avian reservoir is contentious, with arguments for China, Europe, and the USA, at various dates. Novel vaccines were tested during 1918, which are the precursors of the universal influenza vaccines that might offer protection in a future pandemic.


Subject(s)
Global Health/history , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza Pandemic, 1918-1919/history , Influenza Pandemic, 1918-1919/mortality , History, 20th Century , Humans , Influenza A Virus, H1N1 Subtype/immunology , Virulence
5.
PLoS One ; 11(12): e0163089, 2016.
Article in English | MEDLINE | ID: mdl-28005959

ABSTRACT

INTRODUCTION: A Proteosome-adjuvanted trivalent inactivated influenza vaccine (P-TIV) administered intra-nasally was shown to be safe, well tolerated and immunogenic in both systemic and mucosal compartments, and effective at preventing illness associated with evidence of influenza infection. METHODS: In two separate studies using the human viral challenge model, subjects were selected to be immunologically naive to A/Panama/2007/1999 (H3N2) virus and then dosed via nasal spray with one of three regimens of P-TIV or placebo. One or two doses, 15 µg or 30 µg, were given either once only or twice 14 days apart (1 x 30 µg, 2 x 30 µg, 2 x 15 µg) and subjects were challenged with A/Panama/2007/1999 (H3N2) virus. Immune responses to the vaccine antigens were measured by haemagglutination inhibition assay (HAI) and nasal wash secretory IgA (sIgA) antibodies. RESULTS: Vaccine reactogenicity was mild, predictable and generally consistent with earlier Phase I studies with this vaccine. Seroconversion to A/Panama/2007/1999 (H3N2), following vaccination but prior to challenge, occurred in 57% to 77% of subjects in active dosing groups and 2% of placebo subjects. The greatest relative rise in sIgA, following vaccination but prior to challenge, was observed in groups that received 2 doses. CONCLUSION: Intranasal vaccination significantly protected against influenza (as defined by influenza symptoms combined with A/Panama seroconversion) following challenge with A/Panama/2007/1999 (H3N2). When data were pooled from both studies, efficacy ranged from 58% to 82% in active dosing groups for any influenza symptoms with seroconversion, 67% to 85% for systemic or lower respiratory illness and seroconversion, and 65% to 100% for febrile illness and seroconversion. The two dose regimen was found to be superior to the single dose regimen. In this study, protection against illness associated with evidence of influenza infection (evidence determined by seroconversion) following challenge with virus, significantly correlated with pre-challenge HAI titres (p = 0.0003) and mucosal sIgA (p≤0.0001) individually, and HAI (p = 0.028) and sIgA (p = 0.0014) together. HAI and sIgA levels were inversely related to rates of illness. TRIAL REGISTRATION: ClinicalTrials.gov NCT02522754.


Subject(s)
Immunoglobulin A, Secretory/analysis , Immunoglobulin G/blood , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Adjuvants, Immunologic , Administration, Intranasal , Adult , Antibodies, Viral/blood , Antibody Formation , Female , Hemagglutination Inhibition Tests , Humans , Influenza Vaccines/administration & dosage , Male , Placebo Effect , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Young Adult
6.
PLoS One ; 9(5): e93761, 2014.
Article in English | MEDLINE | ID: mdl-24850536

ABSTRACT

We investigated the protective efficacy of two intranasal chitosan (CSN and TM-CSN) adjuvanted H5N1 Influenza vaccines against highly pathogenic avian Influenza (HPAI) intratracheal and intranasal challenge in a ferret model. Six groups of 6 ferrets were intranasally vaccinated twice, 21 days apart, with either placebo, antigen alone, CSN adjuvanted antigen, or TM-CSN adjuvanted antigen. Homologous and intra-subtypic antibody cross-reacting responses were assessed. Ferrets were inoculated intratracheally (all treatments) or intranasally (CSN adjuvanted and placebo treatments only) with clade 1 HPAI A/Vietnam/1194/2004 (H5N1) virus 28 days after the second vaccination and subsequently monitored for morbidity and mortality outcomes. Clinical signs were assessed and nasal as well as throat swabs were taken daily for virology. Samples of lung tissue, nasal turbinates, brain, and olfactory bulb were analysed for the presence of virus and examined for histolopathological findings. In contrast to animals vaccinated with antigen alone, the CSN and TM-CSN adjuvanted vaccines induced high levels of antibodies, protected ferrets from death, reduced viral replication and abrogated disease after intratracheal challenge, and in the case of CSN after intranasal challenge. In particular, the TM-CSN adjuvanted vaccine was highly effective at eliciting protective immunity from intratracheal challenge; serologically, protective titres were demonstrable after one vaccination. The 2-dose schedule with TM-CSN vaccine also induced cross-reactive antibodies to clade 2.1 and 2.2 H5N1 viruses. Furthermore ferrets immunised with TM-CSN had no detectable virus in the respiratory tract or brain, whereas there were signs of virus in the throat and lungs, albeit at significantly reduced levels, in CSN vaccinated animals. This study demonstrated for the first time that CSN and in particular TM-CSN adjuvanted intranasal vaccines have the potential to protect against significant mortality and morbidity arising from infection with HPAI H5N1 virus.


Subject(s)
Chitosan/analogs & derivatives , Chitosan/administration & dosage , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Vaccination , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Animals, Outbred Strains , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dogs , Ferrets , Humans , Influenza, Human/blood , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Male , Nose/immunology , Nose/virology , Trachea/immunology , Trachea/virology , Vaccine Potency , Viral Load
7.
PLoS One ; 8(8): e71376, 2013.
Article in English | MEDLINE | ID: mdl-23951151

ABSTRACT

Influenza is a major cause of morbidity and mortality. Despite vaccination, many elderly recipients do not develop a protective antibody response. To determine whether Human Leukocyte Antigen (HLA) alleles modulate seroprotection to influenza, a cohort of HLA class II-typed high-risk vaccine recipients was investigated. Haemagglutinin inhibition (HAI) titres were measured 14-40 days post-subunit vaccination. Seroprotection was defined as HAI titres reaching 40 or greater for all three vaccine strains. HLA-DRB1*04∶01 and HLA-DPB1*04∶01 alleles were detected at higher frequencies in seroprotected compared with non-seroprotected individuals. Thus, the presence of certain HLA class II alleles may determine the magnitude of antibody responses to influenza vaccination.


Subject(s)
Antibodies, Viral/biosynthesis , HLA-DP beta-Chains/genetics , HLA-DRB1 Chains/genetics , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Orthomyxoviridae/immunology , Vaccination , Aged , Aged, 80 and over , Alleles , Antibodies, Viral/blood , Female , Gene Expression , Gene Frequency , HLA-DP beta-Chains/immunology , HLA-DRB1 Chains/immunology , Hemagglutination Inhibition Tests , Humans , Immunity, Active , Influenza Vaccines/administration & dosage , Influenza, Human/blood , Influenza, Human/genetics , Influenza, Human/immunology , Male , Vaccines, Subunit
8.
Antivir Ther ; 18(6): 785-91, 2013.
Article in English | MEDLINE | ID: mdl-23714753

ABSTRACT

BACKGROUND: Antivirals reduce influenza viral replication and illness measures, particularly if initiated early, within 48 h of symptom onset. Whether experimental antivirals that reduce respiratory syncytial virus (RSV) load would also reduce disease is unknown. This study compares viral and disease dynamics in humans experimentally infected with influenza or RSV. METHODS: Clinical strains of RSV-A and influenza A were inoculated intranasally into 20 and 17 healthy volunteers, respectively, on day 0. Symptom scores and nasal washes were performed twice daily, and daily mucus weights were collected. Viral loads in nasal washes were quantified by culture (plaque assay in HEp-2 cells for RSV and by end point dilution in Madin-Darby canine kidney cells for influenza). RESULTS: After influenza inoculation, influenza viral load and illness markers increased simultaneously until day 2. Within individual subjects, peak influenza load occurred 0.4 days (95% CI -0.4, 1.3) before peak symptoms. Influenza viral load and disease declined thereafter. After RSV inoculation, a longer incubation period occurred prior to viral detection and symptom onset. RSV load and disease increased together until day 5. Within individual subjects, peak RSV loads occurred 0.2 days (95% CI -0.7, 1.05) before peak symptoms, after which both illness measures and viral load declined together. CONCLUSIONS: Viral and disease dynamics in experimental human infections suggest that reducing RSV load, if timed similarly to clinically-effective influenza antivirals, might be expected to have a similar or greater window of opportunity for reducing clinical RSV disease.


Subject(s)
Influenza A virus/physiology , Influenza, Human/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , Adolescent , Adult , Antiviral Agents/therapeutic use , Humans , Middle Aged , Respiratory Syncytial Virus Infections/drug therapy , Treatment Outcome , Viral Load , Virus Replication , Young Adult
9.
Br J Clin Pharmacol ; 76(2): 210-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23617282

ABSTRACT

There are now more than 5 experimental vaccine formulations which induce T and B cell immunity towards the internally situated virus proteins matrix (M1 and M2e) and nucleoprotein (NP), and towards stem and stalk regions of the HA which have a shared antigenic structure amongst many of the 17 influenza A virus sub types. Such 'universal vaccines' could be used, at least in theory, as a prophylactic stockpile vaccine for newly emerged epidemic and novel pandemic influenza A viruses or as a supplement to conventional HA/NA vaccines. My own laboratory has approached the problem from the clinical viewpoint by identifying CD4(+) cells which are present in influenza infected volunteers who resist influenza infection. We have established precisely which peptides in M and NP proteins react with these immune CD4 cells. These experimental vaccines induce immunity in animal models but with a single exception no data have been published on protection against influenza virus infection in humans. The efficacy of the latter vaccine is based on vaccinia virus (MVA) as a carrier and was analyzed in a quarantine unit. Given the absence of induced HI antibody in the new universal vaccines a possible licensing strategy is a virus challenge model in quarantine whereby healthy volunteers can be immunized with the new vaccine and thereafter deliberately infected and clinical signs recorded alongside quantities of virus excreted and compared with unvaccinated controls.


Subject(s)
Drug Approval/methods , Influenza Vaccines/immunology , Influenza, Human/immunology , Drug Approval/legislation & jurisprudence , Humans , Immunity/immunology , Immunization/methods , Influenza, Human/prevention & control , Quarantine/methods , Research Subjects , Time Factors , Vaccination/methods
10.
Expert Rev Vaccines ; 11(8): 929-37, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23002974

ABSTRACT

From the time of the Spanish influenza pandemic in 1918 to the present seclusion of volunteers in quarantine units, either modified hotels, Phase I units or specially constructed clinics, have been key in investigating new vaccines and antivirals. Carefully selected healthy, young volunteers undergo a 10-12-day sojourn under intense medical supervision. Clinical sampling includes nasal and throat washes for virus recovery, blood for clinical chemistry, analysis of B- and T-cell response and, more recently, analysis of human genes responding to infection. The authors' studies are resulting in new developments of universal influenza vaccines that could stimulate and prime CD4 and CD8 cells to shared epitopes in all influenza A viruses. Ethnographic study has noted an absence of quarantine stress in the volunteers for the first time.


Subject(s)
Anthropology, Cultural/methods , Biomedical Research/methods , Influenza A virus/pathogenicity , Influenza, Human/prevention & control , Biomedical Research/organization & administration , CD4-Positive T-Lymphocytes/immunology , Clinical Trials as Topic , Disease Transmission, Infectious/prevention & control , Hemagglutination Tests/methods , Hospitals, Isolation , Humans , Infection Control/methods , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/transmission , Quarantine , Signal Transduction
12.
Viral Immunol ; 21(1): 83-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18355126

ABSTRACT

There are reports that not all individuals exposed to HIV-1 become infected and the possibility exists that some individuals may be completely resistant to infection with this virus. This study aims to investigate, in vitro, whether certain peripheral blood mononuclear cells (PBMCs) are completely resistant to HIV-1 and HIV-2 infection. PBMCs obtained from 130 unrelated healthy HIV-1- and HIV-2-seronegative volunteers were infected with four different isolates of HIV-1 (H995 and MN) and HIV-2 (CBL-20 and ROD) using several multiplicities of infection. Cultures were maintained for 21 d. Virus replication was measured using the viral p24 core antigen levels in the case of HIV-1, and by reverse transcriptase (RT) activity in the case of HIV-2, at 5, 14, and 21 d post-infection. Marked variations were observed among PBMCs from individual donors with regard to replication rates for HIV-1 and HIV-2. None of the PBMCs from any single donor was shown to have zero viral replication rates for all four HIV isolates tested. However, PBMCs from some individuals were shown to have either very low or very high viral replication rates when infected with one or more virus isolates. Our results clearly distinguished three groups of PBMCs with varying degrees of viral replication for both HIV-1 and HIV-2 infection in vitro: (a) those with high viral replication rates, (b) those with moderate viral replication rates, and (c) those with low viral replication rates. Our data indicate that although none of the PBMCs tested were shown to be completely resistant to in vitro HIV-1 and HIV-2 infection, partial resistance to infection was seen for some donor samples.


Subject(s)
HIV-1/growth & development , HIV-2/growth & development , Leukocytes, Mononuclear/virology , Adolescent , Adult , Cells, Cultured , Cytopathogenic Effect, Viral , Female , Giant Cells/virology , HIV Core Protein p24/biosynthesis , HIV Reverse Transcriptase/biosynthesis , Humans , Male
13.
Influenza Other Respir Viruses ; 2(1): 33-40, 2008 Jan.
Article in English | MEDLINE | ID: mdl-19453491

ABSTRACT

BACKGROUND: Influenza A viruses are of major concern for public health, causing worldwide epidemics associated with high morbidity and mortality. Vaccines are critical for protection against influenza, but given the recent emergence of new strains with pandemic potential, and some limitations of the current production systems, there is a need for new approaches for vaccine development. OBJECTIVE: To demonstrate the immunogenicity and protective efficacy of plant-produced influenza antigens. Method We engineered, using influenza A/Wyoming/3/03 (H3N2) as a model virus, the stem and globular domains of hemagglutinin (HA) produced in plants as fusions to a carrier protein and used purified antigens with and without adjuvant for ferret immunization. RESULTS: These plant-produced antigens were highly immunogenic and conferred complete protection against infection in the ferret challenge model. The addition of plant-produced neuraminidase was shown to enhance the immune response in ferrets. CONCLUSIONS: Plants can be used as a production vehicle for vaccine development against influenza. Domains of HA can generate protective immune responses in ferrets.


Subject(s)
Hemagglutinins, Viral/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Animals , Antibodies, Viral/blood , Body Temperature , Body Weight , Enzyme-Linked Immunosorbent Assay , Female , Ferrets , Hemagglutination Inhibition Tests , Hemagglutinins, Viral/genetics , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/genetics , Plants, Genetically Modified , Severity of Illness Index , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Virus Shedding
14.
Am J Ther ; 14(5): 455-61, 2007.
Article in English | MEDLINE | ID: mdl-17890935

ABSTRACT

Prophylaxis against influenza is difficult, and current approaches against pandemics may be ineffective because of shortages of the two proven classes of antivirals in the face of a large-scale infection. Herbal/natural products may represent an effective alternative to conventional attempts to protect against infection by avian influenza virus. QR-435, an all-natural compound of green tea extract and other agents, has been developed to provide protection against a wide range of viral infections. The antiviral activities of several QR-435 preparations as well as QR-435 (1) green tea extract were tested against A/Sydney/5/97 and A/Panama-Resvir 17 strains of avian influenza virus H3N2 by means of an assay based on Madin-Darby canine kidney cells. Toxic effects of QR-435 formulations on these cells were also evaluated as were the virucidal properties of a commercially available mask impregnated with QR-435. The efficacy of a QR-435/mask combination was compared with that of the QR control/mask combination, an untreated mask, and no mask. QR-435 had significant in vitro activity against H3N2 at concentrations that were not associated with significant cellular toxic effects. The antiviral activity of QR-435 (1) was similar to that of QR-435. Masks impregnated with QR-435 were highly effective in blocking the passage of live H3N2 virus. These preclinical results warrant further evaluation of the prophylactic use of QR-435 against viral infection in humans.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Plant Extracts/pharmacology , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/toxicity , Combined Modality Therapy , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Infection Control/methods , Influenza, Human/prevention & control , Kidney/cytology , Masks , Paper , Plant Extracts/administration & dosage , Plant Extracts/toxicity , Tea/chemistry , Toxicity Tests
15.
Am J Ther ; 14(5): 462-8, 2007.
Article in English | MEDLINE | ID: mdl-17890936

ABSTRACT

BACKGROUND: Prophylaxis against influenza infection can take several forms, none of which is totally effective at preventing the spread of the disease. QR-435, an all-natural compound of green-tea extract and other agents, has been developed to protect against a range of viral infections, including the influenza subtype H3N2. METHODS: Several different QR-435 formulations were tested against the two influenza A H3N2 viruses (A/Sydney/5/97 and A/Panama/2007/99) in the ferret model. Most experiments included negative (phosphate-buffered saline) and positive (oseltamivir 5 mg/kg, twice daily) controls. QR-435 and the control were administered 5 minutes after intranasal delivery of the virus as prophylaxis against infection resulting from exposure to infected but untreated ferrets and for prevention of transmission from infected and treated ferrets to untreated animals. Effects of QR-435 on seroconversion, virus shedding, and systemic sequelae of infection (weight loss, fever, reduced activity) were evaluated. RESULTS: QR-435 prevented transmission and provided prophylaxis against influenza virus H3N2. Prophylaxis with QR-435 was significantly more than with oseltamivir in these experiments. Optimal in vivo efficacy of QR-435 requires a horseradish concentration of at least 50% of that in the original formulation, and the benefits of this preparation appear to be dose dependent. CONCLUSIONS: QR-435 is effective for both prevention of H3N2 viral transmission and prophylaxis. These preclinical results warrant further evaluation of its prophylactic properties against avian influenza virus infection in humans.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H3N2 Subtype/drug effects , Orthomyxoviridae Infections/prevention & control , Plant Extracts/therapeutic use , Administration, Intranasal , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Armoracia/chemistry , Disease Models, Animal , Ferrets , Fever/drug therapy , Fever/etiology , Infection Control/methods , Motor Activity/drug effects , Orthomyxoviridae Infections/transmission , Oseltamivir/therapeutic use , Plant Extracts/administration & dosage , Tea/chemistry , Virus Shedding/drug effects , Weight Loss/drug effects
16.
Influenza Other Respir Viruses ; 1(1): 27-34, 2007 Jan.
Article in English | MEDLINE | ID: mdl-19453477

ABSTRACT

Influenza is a highly contagious and debilitating disease that imposes an excess burden of complications and mortality. Antiviral therapy is the primary intervention for treatment and post-exposure prophylaxis (PEP) of influenza. Amantadine and rimantadine are members of the M2 class of antiviral agents and are moderately effective in influenza management. However, their utility is compromised by high levels of resistance, tolerability concerns and a lack of efficacy against influenza B. An alternative class of agents, the neuraminidase inhibitors (NIs), represent the most advanced form of antiviral therapy available, and act by specifically inhibiting the neuraminidase enzymes that are present on all influenza subtypes. Two NIs, oseltamivir and zanamivir, are currently available for clinical use. Oseltamivir, the most widely used NI, is administered orally as a prodrug (oseltamivir carboxylate) and systemically distributed to all potential infection sites. Zanamivir, a second NI, is administered by inhalation via a disk inhaler and deposited primarily in the respiratory tract. When administered within 48 hours of symptom onset, both agents significantly reduce illness duration and symptom severity, and decrease the rate of influenza-associated complications. With oseltamivir, greater benefits are detected with earlier treatment initiation (<12 hours). In PEP, both NIs effectively protect the close contacts of index cases from symptomatic influenza. Oseltamivir and zanamivir are generally well tolerated and associated with a low level of resistance. Emerging evidence supports the activity of both NIs against the H5N1avian influenza infection, which is a pandemic candidate. However, the WHO currently recommends the use of oseltamivir for the management of suspected cases, given the systemic nature of the H5N1 challenge. Ongoing studies are exploring the effectiveness of oseltamivir, zanamivir and other NIs for pandemic management.


Subject(s)
Antiviral Agents/therapeutic use , Chemoprevention/methods , Disease Outbreaks/prevention & control , Influenza, Human/drug therapy , Influenza, Human/prevention & control , Amantadine/therapeutic use , Drug Resistance, Viral , Humans , Influenza, Human/epidemiology , Oseltamivir/therapeutic use , Rimantadine/therapeutic use , Zanamivir/therapeutic use
17.
Virus Res ; 122(1-2): 11-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16904219

ABSTRACT

The influenza pandemic of 1918 caused unprecedented levels of morbidity and mortality in its 12-month period of circulation around the globe. The haemagglutinin molecule has been shown to affect the pathogenicity of some subtypes of influenza A viruses. Using a recombinant vaccinia system that allowed expression of the 1918 influenza haemagglutinin, we performed functional assays to assess the glycoprotein's involvement in determining the high pathogenicity of the 1918 virus. We show that in respect of expression levels, proteolytic processing, receptor-binding, membrane fusion and antigenic properties, the haemagglutinin of the 1918 virus is unremarkable when compared with the haemagglutinins of other 'early' H1 influenza viruses. This suggests that whilst the 1918 haemagglutinin, as a new/novel antigen in the human population, was responsible for the influenza pandemic its functions per se were not responsible for the high mortality and acute symptoms experienced by patients infected with the 1918 influenza virus.


Subject(s)
Antigens, Viral/immunology , Antigens, Viral/physiology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/physiology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Amino Acid Sequence , Animals , Cell Line , Gene Expression , Genetic Vectors , Haplorhini , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Molecular Sequence Data , Vaccinia virus/genetics , Virulence Factors/immunology , Virulence Factors/physiology , Virus Attachment , Virus Internalization
18.
Antivir Chem Chemother ; 16(2): 129-34, 2005.
Article in English | MEDLINE | ID: mdl-15889535

ABSTRACT

A potent virucidal mixture containing amyl metacresol and dichlorobenzyl alcohol at low pH inactivated enveloped respiratory viruses influenza A, respiratory synctial virus (RSV) and severe acute respiratory syndrome coronavirus (SARS-CoV) but not viruses with icosahedral symmetry, such as adenoviruses or rhinoviruses. A titre of approximately 3.5 log10 TCID50 was reduced to below the level of detection within two minutes. Electron microscopy of purified influenza A virus showed extensive clumping and morphological changes in spike configuration after contact with the virucidal mixture, but no overt destruction of the viral membrane. We conclude that, formulated as a lozenge, the mixture could have significant effects in reducing the infectivity of certain infectious viruses in the throat and presumably in cough droplets, thus reducing, theoretically, opportunities for person-to-person transmission.


Subject(s)
Antiviral Agents/pharmacology , Benzyl Alcohols/pharmacology , Cresols/pharmacology , Influenza A virus/drug effects , Respiratory Syncytial Viruses/drug effects , Severe acute respiratory syndrome-related coronavirus/drug effects , Adenoviridae/drug effects , Dosage Forms , Drug Combinations , Humans , Rhinovirus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...