Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Front Immunol ; 12: 711637, 2021.
Article in English | MEDLINE | ID: mdl-34408752

ABSTRACT

Children with autoimmune disorders are especially at risk of vaccine-preventable diseases due to their underlying disease and the immunosuppressive treatment often required for a long period. In addition, vaccine coverage remains too low in this vulnerable population. This can be explained by a fear of possible adverse effects of vaccines under immunosuppression, but also a lack of data and clear recommendations, particularly with regard to vaccination with live vaccines. In this review, the latest literature and recommendations on vaccination in immunosuppressed children are discussed in detail, with the aim to provide a set of practical guidelines on vaccination for specialists caring for children suffering from different autoimmune disorders and treated with various immunosuppressive regimens.


Subject(s)
Autoimmune Diseases/immunology , Immunocompromised Host , Immunogenicity, Vaccine , Immunosuppressive Agents/therapeutic use , Vaccination , Antibody Formation/drug effects , Antigen-Presenting Cells/immunology , Attitude of Health Personnel , Autoimmune Diseases/drug therapy , B-Lymphocyte Subsets/immunology , Child , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Parents/psychology , Practice Guidelines as Topic , Rheumatologists/psychology , Risk , T-Lymphocyte Subsets/immunology , Vaccination Hesitancy , Vaccine-Preventable Diseases/prevention & control , Vaccines, Attenuated , Vaccines, Live, Unattenuated/adverse effects
2.
Gastroenterology ; 161(2): 669-680.e0, 2021 08.
Article in English | MEDLINE | ID: mdl-33617891

ABSTRACT

BACKGROUND & AIMS: Patients with inflammatory bowel disease (IBD) may be at increased risk of some vaccine-preventable diseases. The effectiveness and safety of vaccinations may be altered by immunosuppressive therapies or IBD itself. These recommendations developed by the Canadian Association of Gastroenterology and endorsed by the American Gastroenterological Association, aim to provide guidance on immunizations in adult and pediatric patients with IBD. This publication focused on live vaccines. METHODS: Systematic reviews evaluating the efficacy, effectiveness, and safety of vaccines in patients with IBD, other immune-mediated inflammatory diseases, and the general population were performed. Critical outcomes included mortality, vaccine-preventable diseases, and serious adverse events. Immunogenicity was considered a surrogate outcome for vaccine efficacy. Certainty of evidence and strength of recommendations were rated according to the GRADE (Grading of Recommendation Assessment, Development, and Evaluation) approach. Key questions were developed through an iterative process and voted on by a multidisciplinary panel. Recommendations were formulated using the Evidence-to-Decision framework. Strong recommendation means that most patients should receive the recommended course of action, whereas a conditional recommendation means that different choices will be appropriate for different patients. RESULTS: Three good practice statements included reviewing a patient's vaccination status at diagnosis and at regular intervals, giving appropriate vaccinations as soon as possible, and not delaying urgently needed immunosuppressive therapy to provide vaccinations. There are 4 recommendations on the use of live vaccines. Measles, mumps, rubella vaccine is recommended for both adult and pediatric patients with IBD not on immunosuppressive therapy, but not for those using immunosuppressive medications (conditional). Varicella vaccine is recommended for pediatric patients with IBD not on immunosuppressive therapy, but not for those using immunosuppressive medications (conditional). For adults, recommendations are conditionally in favor of varicella vaccine for those not on immunosuppressive therapy, and against for those on therapy. No recommendation was made regarding the use of live vaccines in infants born to mothers using biologics because the desirable and undesirable effects were closely balanced and the evidence was insufficient. CONCLUSIONS: Maintaining appropriate vaccination status in patients with IBD is critical to optimize patient outcomes. In general, live vaccines are recommended in patients not on immunosuppressive therapy, but not for those using immunosuppressive medications. Additional studies are needed to evaluate the safety and efficacy of live vaccines in patients on immunosuppressive therapy.


Subject(s)
Gastroenterology/standards , Immunization/standards , Immunosuppressive Agents/adverse effects , Inflammatory Bowel Diseases/drug therapy , Opportunistic Infections/prevention & control , Vaccines, Live, Unattenuated/administration & dosage , Canada , Consensus , Contraindications, Drug , Evidence-Based Medicine/standards , Humans , Immunization/adverse effects , Immunocompromised Host , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/mortality , Opportunistic Infections/diagnosis , Opportunistic Infections/immunology , Opportunistic Infections/mortality , Patient Safety , Risk Assessment , Risk Factors , Treatment Outcome , Vaccine Efficacy , Vaccines, Live, Unattenuated/adverse effects
3.
BMC Infect Dis ; 21(1): 117, 2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33499826

ABSTRACT

BACKGROUND: Herpes zoster (HZ) infection of hematopoietic stem cell transplant (HSCT) patients is of clinical concern. Vaccination could help restore immunity to varicella zoster virus (VZV); however, temporal changes in immunogenicity and safety of live HZ vaccines after HSCT is still unclear. The aim of this study was to elucidate the temporal immunogenicity and safety of the HZ vaccine according to time since HSCT and to determine optimal timing of vaccination. METHODS: Live HZ vaccine was administered to patients 2-5 years or > 5 years post-HSCT. Control groups comprised patients with a hematologic malignancy who received cytotoxic chemotherapy and healthy volunteers. Humoral and cellular immunogenicity were measured using a glycoprotein enzyme-linked immunosorbent assay (gpELISA) and an interferon-γ (IFN-γ) enzyme-linked immunospot (ELISPOT) assay. Vaccine-related adverse events were also monitored. RESULTS: Fifty-six patients with hematologic malignancy (41 in the HSCT group and 15 in the chemotherapy group) along with 30 healthy volunteers were enrolled. The geometric mean fold rises (GMFRs) in humoral immune responses of the 2-5 year and > 5 year HSCT groups, and the healthy volunteer group, were comparable and significantly higher than that of the chemotherapy group (3.15, 95% CI [1.96-5.07] vs 5.05, 95% CI [2.50-10.20] vs 2.97, 95% CI [2.30-3.83] vs 1.42, 95% CI [1.08-1.86]). The GMFR of cellular immune responses was highest in the HSCT 2-5 year group and lowest in the chemotherapy group. No subject suffered clinically significant adverse events or reactivation of VZV within the follow-up period. CONCLUSION: Our findings demonstrate that a live HZ vaccine is immunogenic and safe when administered 2 years post-HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Herpes Zoster Vaccine , Herpes Zoster/prevention & control , Herpesvirus 3, Human/immunology , Transplant Recipients , Vaccines, Live, Unattenuated , Aged , Antibodies, Viral/immunology , Case-Control Studies , Female , Follow-Up Studies , Hematologic Neoplasms/epidemiology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/statistics & numerical data , Herpes Zoster Vaccine/adverse effects , Herpes Zoster Vaccine/immunology , Humans , Immunogenicity, Vaccine/physiology , Male , Middle Aged , Transplant Recipients/statistics & numerical data , Treatment Outcome , Vaccination/adverse effects , Vaccination/methods , Vaccination/statistics & numerical data , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/immunology
4.
Am J Public Health ; 111(3): 498-503, 2021 03.
Article in English | MEDLINE | ID: mdl-33476239

ABSTRACT

The increased risk of harm from COVID-19 infection in pregnancy highlights the importance of including pregnant people in COVID-19 vaccine development and deployment. Promising vaccines being developed include replication-competent platforms, which are typically contraindicated during pregnancy because of theoretical risk. However, replicating vaccines are administered in and around pregnancy, either inadvertently because of unknown pregnancy status or when recommended.The historical cases of Ebola virus, yellow fever, and rubella demonstrate that contradictory messages around the safety of live vaccines in pregnancy have critical public health costs. First, restricting study or use of replicating vaccines in pregnancy may delay or deny access to the only available protection against deadly diseases. Additionally, not vaccinating pregnant people may slow epidemic control. Finally, uncertainty and worry around the safety of live vaccines may lead to terminations of otherwise desired pregnancies after inadvertent vaccination in pregnancy.If one of the vaccines deployed to combat the current global COVID-19 pandemic is replication competent, historical cases offer important lessons for ethical and effective protection for pregnant populations.


Subject(s)
COVID-19 Vaccines/adverse effects , COVID-19/epidemiology , Drug Development/organization & administration , Pregnancy Complications, Infectious/prevention & control , Pregnant Women/psychology , COVID-19/prevention & control , Drug Development/standards , Female , Humans , Pandemics , Pregnancy , SARS-CoV-2 , Vaccines, Live, Unattenuated/adverse effects
5.
Int J Hematol ; 112(1): 105-114, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32253664

ABSTRACT

This case-control study investigated immune thrombocytopenic purpura (ITP) risk following live, inactivated, and simultaneous vaccination, with a focus on infants aged < 2 years. We matched case patients with ITP to one or two control patients with other diseases by institution, hospital visit timing, sex, and age. We calculated McNemar's pairwise odds ratios (ORs [95% confidence interval]) with 114 case-control pairs. The case group had 27 (44%) males and 22 (35%) infants, and the control group included 49 (43%) males and 42 (37%) infants. For all age groups, the McNemar's OR for ITP occurrence was 1.80 (0.54-6.84, p = 0.64) for all vaccines. Among infants, these were 1.50 (0.17-18.0, p = 0.50) for all vaccines, 2.00 (0.29-22.1, p = 0.67) for live vaccines, and 1.00 (0.01-78.5, p = 0.50) for inactivated vaccines. Sex-adjusted common ORs for simultaneous vaccination were 1.52 (0.45-5.21, p = 0.71) for all vaccines, 1.83 (0.44-7.59, p = 0.40) for inactivated vaccines only, and 1.36 (0.29-6.30, p = 0.69) for mixed live and inactivated vaccines. In infants, these were 1.95 (0.44-8.72, p = 0.38), 1.41 (0.29-6.94, p = 0.67) and 2.85 (0.43-18.9, p = 0.28), respectively. These limited data suggest no significant ITP risk following vaccinations or simultaneous vaccination in any age group, including infants.


Subject(s)
Purpura, Thrombocytopenic, Idiopathic/etiology , Vaccination/adverse effects , Vaccines, Attenuated/adverse effects , Vaccines, Inactivated/adverse effects , Vaccines, Live, Unattenuated/adverse effects , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Humans , Male , Risk , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Inactivated/administration & dosage , Vaccines, Live, Unattenuated/administration & dosage , Young Adult
6.
Vet Microbiol ; 240: 108537, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31902490

ABSTRACT

The safety of the live Escherichia coli vaccine Poulvac® E. coli was tested with a flock (10,000) of layer parents aged 30 weeks. Three and 7 days after vaccination, 60 whole unbroken eggs, the egg white and yolk of 60 eggs and 60 cloacal swabs were enriched in MacConkey broth. At both sampling times, 6 out of 60 whole eggs were found positive for coliform bacteria. None of the enriched samples of yolk + egg white were positive for coliform bacteria. Three and seven days after vaccination 44 and 37, respectively out of 60 swabs were positive for coliform bacteria in MacConkey broth. All coliform isolates collected from whole eggs and cloacal swabs were tested in parallel for growth on minimal agar and blood agar to identify the vaccine strain. Some isolates showed reduced growth on minimal agar compared to blood agar and they were tested further with a PCR for the aroA gene mutation and all were found with the wild type version of the gene. Only two isolates did not grow on minimal agar but grew on blood agar and they were tested both with PCR and PFGE. They also showed the wild type version of the aroA gene and their PFGE profile was different from the vaccine strain of Poulvac® E. coli. In conclusion, the Poulvac® E. coli vaccine strain of E. coli was not identified at the detection limit of one CFU on one egg or in the content of one egg or from a cloacal swab of one hen with at least 95 % probability on flock level. The use of the vaccine is safe for hens in lay with lack of survival of the vaccine strain and lack of negative effects on the hens including egg production.


Subject(s)
Escherichia coli Vaccines/analysis , Escherichia coli/isolation & purification , Vaccination/veterinary , Animals , Chickens/microbiology , Cloaca/microbiology , Culture Media/chemistry , Egg Shell/microbiology , Egg Yolk/microbiology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Vaccines/adverse effects , Female , Food Microbiology , Limit of Detection , Ovum/microbiology , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/analysis , Whole Genome Sequencing
7.
Vet Res ; 50(1): 90, 2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31694705

ABSTRACT

Virus infections possess persistent health challenges in swine industry leading to severe economic losses worldwide. The economic burden caused by virus infections such as Porcine Reproductive and Respiratory Syndrome Virus, Swine influenza virus, Porcine Epidemic Diarrhea Virus, Porcine Circovirus 2, Foot and Mouth Disease Virus and many others are associated with severe morbidity, mortality, loss of production, trade restrictions and investments in control and prevention practices. Pigs can also have a role in zoonotic transmission of some viral infections to humans. Inactivated and modified-live virus vaccines are available against porcine viral infections with variable efficacy under field conditions. Thus, improvements over existing vaccines are necessary to: (1) Increase the breadth of protection against evolving viral strains and subtypes; (2) Control of emerging and re-emerging viruses; (3) Eradicate viruses localized in different geographic areas; and (4) Differentiate infected from vaccinated animals to improve disease control programs. Nanoparticles (NPs) generated from virus-like particles, biodegradable and biocompatible polymers and liposomes offer many advantages as vaccine delivery platform due to their unique physicochemical properties. NPs help in efficient antigen internalization and processing by antigen presenting cells and activate them to elicit innate and adaptive immunity. Some of the NPs-based vaccines could be delivered through both parenteral and mucosal routes to trigger efficient mucosal and systemic immune responses and could be used to target specific immune cells such as mucosal microfold (M) cells and dendritic cells (DCs). In conclusion, NPs-based vaccines can serve as novel candidate vaccines against several porcine viral infections with the potential to enhance the broader protective efficacy under field conditions. This review highlights the recent developments in NPs-based vaccines against porcine viral pathogens and how the NPs-based vaccine delivery system induces innate and adaptive immune responses resulting in varied level of protective efficacy.


Subject(s)
Nanoparticles/analysis , Swine Diseases/prevention & control , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/pharmacology , Virus Diseases/veterinary , Animals , Swine , Swine Diseases/virology , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/pharmacology , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/pharmacology , Viral Vaccines/adverse effects , Virus Diseases/prevention & control , Virus Diseases/virology
9.
Sci Rep ; 9(1): 5297, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30923339

ABSTRACT

TNF plays a critical role in mononuclear cell recruitment during acute Bacillus Calmette-Guérin (BCG) infection leading to an effective immune response with granuloma formation, but may also cause tissue injury mediated by TNFR1 or TNFR2. Here we investigated the role of myeloid and T cell specific TNFR1 and R2 expression, and show that absence of TNFR1 in myeloid cells attenuated liver granuloma formation and liver injury in response to acute BCG infection, while TNFR2 expressed in myeloid cells contributed only to liver injury. TNFR1 was the main receptor controlling cytokine production by liver mononuclear cells after antigenic specific response, modified CD4/CD8 ratio and NK, NKT and regulatory T cell recruitment. Further analysis of CD11b+CD3+ phagocytic cells revealed a TCRαß expressing subpopulation of unknown function, which increased in response to BCG infection dependent of TNFR1 expression on myeloid cells. In conclusion, TNFR1 expressed by myeloid cells plays a critical role in mononuclear cell recruitment and injury of the liver after BCG infection.


Subject(s)
BCG Vaccine/adverse effects , Granuloma/immunology , Hepatitis/immunology , Mycobacterium bovis/pathogenicity , Myeloid Cells/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , BCG Vaccine/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Granuloma/microbiology , Granuloma/pathology , Hepatitis/microbiology , Hepatitis/pathology , Humans , Liver/cytology , Liver/immunology , Liver/pathology , Mice , Mice, Knockout , Mycobacterium bovis/immunology , Myeloid Cells/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Vaccines, Live, Unattenuated/adverse effects
10.
Article in English | MEDLINE | ID: mdl-29661808

ABSTRACT

Worldwide, there are multiple formaldehyde-inactivated and at least two live attenuated hepatitis A vaccines now in clinical use. The impressive immunogenicity of inactivated vaccines is reflected in rapid seroconversion rates, enabling both preexposure and postexposure prophylaxis. Universal childhood vaccination programs targeting young children have led to significant drops in the incidence of hepatitis A both in toddlers and in susceptible nonimmune adults in regions with intermediate endemicity for hepatitis A. Although the safety of inactivated vaccines is well established, further studies are needed concerning the implications of fecal virus shedding by recipients of attenuated vaccines, as well as the long-term persistence of immune memory in children receiving novel immunization schedules consisting of single doses of inactivated vaccines.


Subject(s)
Hepatitis A Vaccines/administration & dosage , Hepatitis A/prevention & control , Vaccines, Inactivated/administration & dosage , Vaccines, Live, Unattenuated/administration & dosage , Global Health , Hepatitis A/epidemiology , Hepatitis A/immunology , Hepatitis A Vaccines/adverse effects , Humans , Immunization Programs/statistics & numerical data , Immunization Schedule , Immunoglobulins/administration & dosage , Post-Exposure Prophylaxis/methods , Pre-Exposure Prophylaxis/methods , Vaccines, Inactivated/adverse effects , Vaccines, Live, Unattenuated/adverse effects
11.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29925653

ABSTRACT

The poliovirus eradication initiative has spawned global immunization infrastructure and dramatically decreased the prevalence of the disease, yet the original virus eradication goal has not been met. The suboptimal properties of the existing vaccines are among the major reasons why the program has repeatedly missed eradication deadlines. Oral live poliovirus vaccine (OPV), while affordable and effective, occasionally causes the disease in the primary recipients, and the attenuated viruses rapidly regain virulence and can cause poliomyelitis outbreaks. Inactivated poliovirus vaccine (IPV) is safe but expensive and does not induce the mucosal immunity necessary to interrupt virus transmission. While the need for a better vaccine is widely recognized, current efforts are focused largely on improvements to the OPV or IPV, which are still beset by the fundamental drawbacks of the original products. Here we demonstrate a different design of an antipoliovirus vaccine based on in situ production of virus-like particles (VLPs). The poliovirus capsid protein precursor, together with a protease required for its processing, are expressed from a Newcastle disease virus (NDV) vector, a negative-strand RNA virus with mucosal tropism. In this system, poliovirus VLPs are produced in the cells of vaccine recipients and are presented to their immune systems in the context of active replication of NDV, which serves as a natural adjuvant. Intranasal administration of the vectored vaccine to guinea pigs induced strong neutralizing systemic and mucosal antibody responses. Thus, the vectored poliovirus vaccine combines the affordability and efficiency of a live vaccine with absolute safety, since no full-length poliovirus genome is present at any stage of the vaccine life cycle.IMPORTANCE A new, safe, and effective vaccine against poliovirus is urgently needed not only to complete the eradication of the virus but also to be used in the future to prevent possible virus reemergence in a postpolio world. Currently, new formulations of the oral vaccine, as well as improvements to the inactivated vaccine, are being explored. In this study, we designed a viral vector with mucosal tropism that expresses poliovirus capsid proteins. Thus, poliovirus VLPs are produced in vivo, in the cells of a vaccine recipient, and are presented to the immune system in the context of vector virus replication, stimulating the development of systemic and mucosal immune responses. Such an approach allows the development of an affordable and safe vaccine that does not rely on the full-length poliovirus genome at any stage.


Subject(s)
Genetic Vectors , Newcastle disease virus/genetics , Poliomyelitis/prevention & control , Poliovirus Vaccines/immunology , Poliovirus/genetics , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Viral/blood , Capsid Proteins/genetics , Capsid Proteins/immunology , Guinea Pigs , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Newcastle disease virus/immunology , Newcastle disease virus/physiology , Poliomyelitis/immunology , Poliomyelitis/virology , Poliovirus/enzymology , Poliovirus/immunology , Poliovirus Vaccine, Inactivated/administration & dosage , Poliovirus Vaccine, Inactivated/adverse effects , Poliovirus Vaccine, Inactivated/genetics , Poliovirus Vaccine, Inactivated/immunology , Poliovirus Vaccines/adverse effects , Poliovirus Vaccines/standards , Vaccination , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/genetics , Vaccines, Live, Unattenuated/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/adverse effects , Vaccines, Virus-Like Particle/genetics
12.
BMC Pediatr ; 18(1): 134, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29642863

ABSTRACT

BACKGROUND: In children with epilepsy, fever and infection can trigger seizures. Immunization can also induce inflammation and fever, which could theoretically trigger a seizure. The risk of seizure after immunization in children with pre-existing epilepsy is not known. The study objective was to determine the risk of medically attended seizure after immunization in children with epilepsy < 7 years of age. METHODS: We conducted a retrospective study of children < 7 years of age with epilepsy in Nova Scotia, Canada from 2010 to 2014. Hospitalizations, emergency visits, unscheduled clinic visits, and telephone calls for seizures were extracted from medical records. Immunization records were obtained from family physicians and Public Health with informed consent. We conducted a risk interval analysis to estimate the relative risk (RR) of seizure during risk periods 0-14, 0-2, and 5-14 days post-immunization versus a control period 21-83 days post-immunization. RESULTS: There were 302 children with epilepsy who were eligible for the study. Immunization records were retrieved on 147 patients (49%), of whom 80 (54%) had one or more immunizations between the epilepsy diagnosis date and age 7 years. These 80 children had 161 immunization visits and 197 medically attended seizures. Children with immunizations had more seizures than either those with no immunizations or those with no records (mean 2.5 versus 0.7 versus 0.9, p < 0.001). The risk of medically attended seizure was not increased 0-14 days after any vaccine (RR = 1.1, 95% confidence interval (CI): 0.5-2.8) or 0-2 days after inactivated vaccines (RR = 0.9, 95% CI: 0.1-7.1) versus 21-83 days post-immunization. No seizure events occurred 5-14 days after live vaccines. CONCLUSIONS: Children with epilepsy do not appear to be at increased risk of medically attended seizure following immunization. These findings suggest that immunization is safe in children with epilepsy, with benefits outweighing risks.


Subject(s)
Epilepsy/complications , Seizures/etiology , Vaccination/adverse effects , Adolescent , Child , Child, Preschool , Female , Fever/etiology , Humans , Infant , Infant, Newborn , Male , Nova Scotia , Retrospective Studies , Risk Assessment , Time Factors , Vaccines, Attenuated/adverse effects , Vaccines, Live, Unattenuated/adverse effects
13.
Internist (Berl) ; 59(2): 205-212, 2018 Feb.
Article in German | MEDLINE | ID: mdl-29230484

ABSTRACT

Infectious diseases are responsible for up to 5% of fatalities even in developed countries. In addition, there is an increasing susceptibility for infections in elderly people due to physiological aging of the immune system. The principles of vaccination are based on a targeted activation of the human immune system. Principally, a distinction is made between passive immunization, i.e. the application of specific antibodies against a pathogen and active immunization. In active immunization, i.e. vaccination, weakened (attenuated) or dead pathogens or components of pathogens (antigens) are administered. After a latency period that depends on the vaccine, complete immune protection is achieved and immunity is maintained for a certain period of time. In contrast to dead vaccines, by the use of live vaccines there is always a risk for infection with the administered vaccine. In passive immunization antibodies are administered. As a rule passive immunization is carried out in persons who have had contact with an infected person and in whom no or uncertain immunity against the corresponding disease is present. Based on the recommendations of the Standing Committee on Vaccination (STIKO), influenza, pneumococcal, herpes zoster, early summer meningoencephalitis (FSME) and travel vaccines are described.


Subject(s)
Communicable Diseases/immunology , Vaccination/methods , Herpes Zoster Vaccine/adverse effects , Herpes Zoster Vaccine/immunology , Humans , Immunity, Active/immunology , Immunization, Passive , Immunocompetence/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Risk Factors , Streptococcus pneumoniae/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/immunology
14.
J Infect ; 72 Suppl: S13-22, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27233121

ABSTRACT

Immunocompromised children have a higher risk of developing infections and associated higher rates of mortality and morbidity. Although this group could benefit the most from vaccine administration, specific considerations regarding immunisations are required. This review is a summary of the vaccines that are relevant to the immunocompromised host, covering both live and non-live vaccines. The burden of disease, safety, immunogenicity/effectiveness and specific recommendations for each vaccine are described as well as specific guidelines from different organisations.


Subject(s)
Immunocompromised Host , Vaccines/adverse effects , Vaccines/immunology , Child , Humans , Immunization/methods , Immunization/standards , Immunogenicity, Vaccine , Infant , Practice Guidelines as Topic , Vaccines/administration & dosage , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology , Vaccines, Inactivated/standards , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/immunology , Vaccines, Live, Unattenuated/standards
15.
Anim Health Res Rev ; 16(1): 40-54, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26050571

ABSTRACT

Bovine viral diarrhea viruses (BVDV) are diverse genetically and antigenically. This diversity impacts both diagnostic testing and vaccination. In North America, there are two BVDV species, 1 and 2 with 3 subgenotypes, BVDV1a, BVDV1b and BVDV2a. Initially, US vaccines contained BVDV1a cytopathic strains. With the reporting of BVDV2 severe disease in Canada and the USA there was focus on protection by BVDV1a vaccines on BVDV2 disease. There was also emphasis of controlling persistently infected (PI) cattle resulted in studies for fetal protection afforded by BVDV1a vaccines. Initially, studies indicated that some BVDV1a vaccines gave less than 100% protection against BVDV2 challenge for fetal infection. Eventually vaccines in North America added BVDV2a to modified live virus (MLV) and killed BVDV1a vaccines. Ideally, vaccines should stimulate complete immunity providing 100% protection against disease, viremias, shedding, and 100% fetal protection in vaccinates when challenged with a range of diverse antigenic viruses (subgenotypes). There should be a long duration of immunity stimulated by vaccines, especially for fetal protection. MLV vaccines should be safe when given according to the label and free of other pathogens. While vaccines have now included BVDV1a and BVDV2a, with the discovery of the predominate subgenotype of BVDV in the USA to be BVDV1b, approximately 75% or greater in prevalence, protection in acute challenge and fetal protection studies became more apparent for BVDV1b. Thus many published studies examined protection by BVDV1a and BVDV2a vaccines against BVDV1b in acute challenge and fetal protection studies. There are no current BVDV1b vaccines in the USA. There are now more regulations on BVDV reproductive effects by the USDA Center for Veterinary Biologics (CVB) regarding label claims for protection against abortion, PI calves, and fetal infections, including expectations for studies regarding those claims. Also, the USDA CVB has a memorandum providing the guidance for exemption of the warning label statement against the use of the MLV BVDV in pregnant cows and calves nursing pregnant cows. In reviews of published studies in the USA, the results of acute challenge and fetal protection studies are described, including subgenotypes in vaccines and challenge strains and the results in vaccinates and the vaccinates' fetuses/newborns. In general, vaccines provide protection against heterologous strains, ranging from 100% to partial but statistically significant protection. In recent studies, the duration of immunity afforded by vaccines was investigated and reported. Issues of contamination remain, especially since fetal bovine serums may be contaminated with noncytopathic BVDV. In addition, the potential for immunosuppression by MLV vaccines exists, and new vaccines will be assessed in the future to prove those MLV components are not immunosuppressive by experimental studies. As new subgenotypes are found, the efficacy of the current vaccines should be evaluated for these new strains.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease , Diarrhea Viruses, Bovine Viral/genetics , Genetic Variation , Vaccination/veterinary , Abortion, Veterinary/etiology , Animals , Antibodies, Viral/immunology , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Diarrhea Viruses, Bovine Viral/classification , North America/epidemiology , Vaccination/methods , Vaccines, Inactivated , Vaccines, Live, Unattenuated/adverse effects
16.
J Infect Dis ; 212(2): 195-201, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25505300

ABSTRACT

BACKGROUND: Infection with influenza A virus (IAV) increases susceptibility to respiratory bacterial infections, resulting in increased bacterial carriage and complications such acute otitis media, pneumonia, bacteremia, and meningitis. Recently, vaccination with live attenuated influenza virus (LAIV) was reported to enhance subclinical bacterial colonization within the nasopharynx, similar to IAV. Although LAIV does not predispose to bacterial pneumonia, whether it may alter bacterial transmigration toward the middle ear, where it could have clinically relevant implications, has not been investigated. METHODS: BALB/c mice received LAIV or phosphate-buffered saline 1 or 7 days before or during pneumococcal colonization with either of 2 clinical isolates, 19F or 7F. Middle ear bacterial titers were monitored daily via in vivo imaging. RESULTS: LAIV increased bacterial transmigration to and persistence within the middle ear. When colonization followed LAIV inoculation, a minimum LAIV incubation period of 4 days was required before bacterial transmigration commenced. CONCLUSIONS: While LAIV vaccination is safe and effective at reducing IAV and coinfection with influenza virus and bacteria, LAIV may increase bacterial transmigration to the middle ear and could thus increase the risk of clinically relevant acute otitis media. These data warrant further investigations into interactions between live attenuated viruses and naturally colonizing bacterial pathogens.


Subject(s)
Influenza Vaccines/adverse effects , Otitis Media/microbiology , Pneumococcal Infections/microbiology , Vaccination , Animals , Bacterial Translocation , Immunity, Innate , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Mice, Inbred BALB C , Otitis Media/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/physiology , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/adverse effects , Vaccines, Live, Unattenuated/immunology
17.
J Allergy Clin Immunol ; 133(4): 961-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24582311

ABSTRACT

The present uncertainty of which live viral or bacterial vaccines can be given to immunodeficient patients and the growing neglect of societal adherence to routine immunizations has prompted the Medical Advisory Committee of the Immune Deficiency Foundation to issue recommendations based on published literature and the collective experience of the committee members. These recommendations address the concern for immunodeficient patients acquiring infections from healthy subjects who have not been immunized or who are shedding live vaccine-derived viral or bacterial organisms. Such transmission of infectious agents can occur within the hospital, clinic, or home or at any public gathering. Collectively, we define this type of transmission as close-contact spread of infectious disease that is particularly relevant in patients with impaired immunity who might have an infection when exposed to subjects carrying vaccine-preventable infectious diseases or who have recently received a live vaccine. Immunodeficient patients who have received therapeutic hematopoietic stem transplantation are also at risk during the time when immune reconstitution is incomplete or while they are receiving immunosuppressive agents to prevent or treat graft-versus-host disease. This review recommends the general education of what is known about vaccine-preventable or vaccine-derived diseases being spread to immunodeficient patients at risk for close-contact spread of infection and describes the relative risks for a child with severe immunodeficiency. The review also recommends a balance between the need to protect vulnerable subjects and their social needs to integrate into society, attend school, and benefit from peer education.


Subject(s)
Bacterial Infections/transmission , Bacterial Vaccines/adverse effects , Immunocompromised Host , Vaccines, Live, Unattenuated/adverse effects , Viral Vaccines/adverse effects , Virus Diseases/transmission , Bacterial Infections/immunology , Bacterial Infections/prevention & control , Bacterial Vaccines/immunology , Child , Child, Preschool , Humans , Immunologic Deficiency Syndromes , Vaccines, Live, Unattenuated/immunology , Viral Vaccines/immunology , Virus Diseases/immunology , Virus Diseases/prevention & control
18.
MMWR Morb Mortal Wkly Rep ; 63(4): 81, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24476980

ABSTRACT

Two live rotavirus oral vaccines, RotaTeq (RV5) (Merck & Co., Inc.) and Rotarix (RV1) (GlaxoSmithKline Biologicals), are approved for prevention of rotavirus gastroenteritis and recommended at ages 2, 4 (RV5/RV1), and 6 (RV5) months by the Advisory Committee on Immunization Practices. Because most childhood vaccines are injectable, vaccination providers might have less experience administering oral vaccines. To assess that hypothesis, CDC searched for reports to the Vaccine Adverse Event Reporting System (VAERS) of rotavirus vaccine administration errors involving injection and eye splashes in the United States during the period January 1, 2006-August 1, 2013. A total of 66 reports were found.


Subject(s)
Medical Errors/statistics & numerical data , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Administration, Oral , Adverse Drug Reaction Reporting Systems , Centers for Disease Control and Prevention, U.S. , Eye Injuries/chemically induced , Humans , Infant , Injections/adverse effects , Injections/statistics & numerical data , Rotavirus Vaccines/adverse effects , United States , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/adverse effects
19.
Obstet Gynecol ; 122(6): 1271-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24201689

ABSTRACT

OBJECTIVE: To characterize maternal and infant outcomes for pregnant women who received live H1N1 influenza vaccine and had no reported adverse events. METHODS: We identified Vaccine Adverse Event Reporting System reports, which described receipt of live H1N1 vaccine during pregnancy without an indication of an adverse event at the time of the report during October 2009 to June 2010. We reviewed the initial reports and obtained pregnancy outcome and infant data through 6 months of age from medical records. We reviewed the numbers and characteristics of pregnancy complications and infant outcomes including major birth defects and medically important infant conditions. Rates of spontaneous abortion, preterm birth, and major birth defects and their 95% confidence intervals were calculated. RESULTS: The Vaccine Adverse Event Reporting System received 113 reports stating receipt of live H1N1 vaccine during pregnancy with no adverse events reported. We obtained follow-up maternal records on 95 of the 113 (84%) live H1N1 reports (40.2% were vaccinated in the first trimester) and found: 87 live births (two twin pregnancies) and no maternal deaths occurred. Number and rates of pregnancy-specific adverse events included: 10 (10.5%, 5.8-18.3) spontaneous abortions; four (4.7%, 1.8-11.4) preterm deliveries at 35-36 weeks of gestation; three (3.4%, 1.2-9.7) infants had one or more major birth defects noted at birth: one cleft palate, one cleft lip, and one microtia (underdeveloped or absent external ear). Seven neonates and infants were hospitalized for medically important conditions. One infant death occurred in a 2.5-month-old boy as a result of pertussis. CONCLUSION: Rates of spontaneous abortion, preterm birth, and major birth defects in pregnant women who received live H1N1 vaccine were similar to or lower than published background rates. No concerning patterns of medical conditions in infants were identified. LEVEL OF EVIDENCE: : III.


Subject(s)
Abortion, Spontaneous/epidemiology , Congenital Abnormalities/epidemiology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Premature Birth/epidemiology , Adolescent , Adult , Female , Humans , Infant , Infant, Newborn , Influenza Vaccines/adverse effects , Live Birth , Male , Pregnancy , United States/epidemiology , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL