Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
BMC Health Serv Res ; 22(1): 1026, 2022 Aug 12.
Article in English | MEDLINE | ID: mdl-35962401

ABSTRACT

BACKGROUND: Perinatal healthcare professionals (PHCPs) provide essential support to all parents in the perinatal period, including young parents aged 16-24, who are at an increased risk of morbidity and mortality. Little is known about the impact of COVID-19 restrictions on the provision of perinatal services, and on perinatal healthcare professionals, caring for young parents in the UK. METHODS: A UK based qualitative study using semi-structured interviews with perinatal healthcare professionals (n = 17). Data were analysed using thematic analysis. RESULTS: Two themes were identified describing perinatal healthcare professionals' perceptions of providing care to young parents during the pandemic. Perinatal healthcare professionals perceived that young parents' needs were amplified by the pandemic and that pandemic-related changes to the service, such as the use of telemedicine to replace face-to-face interactions, did not manage to successfully mitigate the increased feelings of anxiety and isolation experienced by young parents. Concerns were raised by perinatal healthcare professionals that these changes reduced young parent's access to vital support for themselves and their child and may contribute to exacerbating pre-existing inequalities. CONCLUSIONS: This study provides insight into the impact of the COVID-19 pandemic on the provision of perinatal care to young parents. Perinatal mental health professionals felt these negative impacts could be overcome by using a blended approach of technology and face-to-face interactions allowing regular contact with young parents and facilitating the exchange of vital information, while maintaining access to opportunities for social interactions with other parents. Findings from this study could be used to future-proof services against further COVID-19 restrictions.


Subject(s)
COVID-19 , Health Personnel , Perinatal Care , COVID-19/epidemiology , Delivery of Health Care , Female , Health Personnel/psychology , Humans , Pandemics , Pregnancy , Qualitative Research
3.
BMC Psychiatry ; 21(1): 73, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33541304

ABSTRACT

BACKGROUND: Point of Care Testing (POCT) is being increasingly used to augment the delivery of physical health care in a variety of settings, but their use in mental health has been limited. Research into understanding the barriers faced for successful implementation of POCT in these settings is lacking. We aimed to identify factors affecting engagement and implementation of POCT within mental health teams by exploring the attitudes to POCT, and the perceived impact POCT has on the practice of mental healthcare clinicians. METHODS: Alongside a study evaluating the impact of a point of care device in Community Mental Health Teams (CMHTs), qualitative interviews were carried out with CMHT clinicians using POCT as part of annual physical checks for patients with severe and enduring mental illness. Data were collected using semi-structured interviews and analysed using thematic analysis. RESULTS: Fifteen clinicians were interviewed across a range of professional backgrounds. Clinicians identified usability of the technology, positive impact on their patient's experience and improved self-efficacy as drivers for successful implementation of POCT into their clinical practice. Issues with device functioning and the potential for a negative effect on the therapeutic relationship with their patients were identified as barriers. Level of physical heath training was not found to be a barrier by mental health professionals to using POCT. CONCLUSIONS: Understanding barriers and drivers for engagement is important to allow co-production of POCT and guidelines to facilitate introduction of POCT into routine clinical practice.


Subject(s)
Mental Health Services , Point-of-Care Testing , Health Personnel , Humans , Point-of-Care Systems , Qualitative Research
4.
BMC Fam Pract ; 21(1): 243, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33243139

ABSTRACT

BACKGROUND: People with severe mental illness experience physical health significantly worse than the general population. Physical health monitoring is shared between primary care and secondary mental healthcare services, though there is debate whether mental health teams should provide more physical healthcare. The views of mental health clinicians and patients with mental illness towards physical healthcare provision are unclear. AIMS: To explore the attitudes of Community Mental Health Team (CMHT) clinicians and patients experiencing severe mental illness towards physical healthcare and its provision. DESIGN AND SETTING: Qualitative study in a CMHT setting. METHODS: Interviews were carried out with CMHT clinicians and patients with severe mental illness. Data were collected using semi-structured interviews and analysed using thematic analysis. RESULTS: There were 14 patients and 15 clinicians recruited. Patients varied in their awareness of the association between physical and mental health, but were engaged in physical health monitoring. Clinicians were aware of the importance of physical healthcare but reported barriers to provision, including lack of training, resource constraints and uncertainty in their role. There was no consensus in either group regarding how physical healthcare should be provided, with diverse attitudes expressed for why CMHTs should and shouldn't provide more physical healthcare. CONCLUSIONS: Increasing physical healthcare provision from mental health teams requires healthcare-related barriers be addressed, but it remains unclear whether CMHT clinicians or patients believe this to be a solution.


Subject(s)
Mental Disorders , Mental Health , Attitude , Delivery of Health Care , Humans , Mental Disorders/therapy , Qualitative Research
5.
BJPsych Open ; 6(6): e127, 2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33107428

ABSTRACT

BACKGROUND: Physical health outcomes in severe mental illness are worse than in the general population. Routine physical health check completion in this group is poor. AIMS: To quantitatively and qualitatively evaluate the impact of point of care (POC) blood testing on physical health check completion in community mental health services. METHOD: In a prospective cohort design, we equipped an early intervention service (EIS) and a community mental health team (CMHT) with a POC blood testing device for 6 months. We compared rates of blood test and full physical health check completion in the intervention teams with a matched EIS and CMHT, historically and during the intervention. We explored attitudes to POC testing using thematic analysis of semi-structured interviews with patients and clinicians. RESULTS: Although the CMHT scarcely used the POC device and saw no change in outcomes, direct comparison of testing rates in the intervention period showed increased physical health check completion in the EIS with the device (rate ratio RR = 5.18; 95% CI 2.54-12.44; P < 0.001) compared with usual care. The rate was consistent with the EIS's increasing rate of testing over time (RR = 0.45; 95% 0.09-2.08; P = 0.32). Similar trends were seen in blood test completion. POC testing was acceptable to patients but clinicians reported usability, provision and impact on the therapeutic relationship as barriers to uptake. CONCLUSIONS: POC testing was beneficial and acceptable to patients and may increase physical health check uptake. Further research, accounting for clinician barriers, is needed to evaluate its clinical and cost-effectiveness.

6.
Cochrane Database Syst Rev ; 5: CD008268, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32356360

ABSTRACT

BACKGROUND: Sore throat is a common condition associated with a high rate of antibiotic prescriptions, despite limited evidence for the effectiveness of antibiotics. Corticosteroids may improve symptoms of sore throat by reducing inflammation of the upper respiratory tract. This review is an update to our review published in 2012. OBJECTIVES: To assess the clinical benefit and safety of corticosteroids in reducing the symptoms of sore throat in adults and children. SEARCH METHODS: We searched CENTRAL (Issue 4, 2019), MEDLINE (1966 to 14 May 2019), Embase (1974 to 14 May 2019), the Database of Abstracts of Reviews of Effects (DARE, 2002 to 2015), and the NHS Economic Evaluation Database (inception to 2015). We also searched the World Health Organization International Clinical Trials Registry Platform (WHO ICTRP) and ClinicalTrials.gov. SELECTION CRITERIA: We included randomised controlled trials (RCTs) that compared steroids to either placebo or standard care in adults and children (aged over three years) with sore throat. We excluded studies of hospitalised participants, those with infectious mononucleosis (glandular fever), sore throat following tonsillectomy or intubation, or peritonsillar abscess. DATA COLLECTION AND ANALYSIS: We used standard methodological procedures expected by Cochrane. MAIN RESULTS: We included one new RCT in this update, for a total of nine trials involving 1319 participants (369 children and 950 adults). In eight trials, participants in both corticosteroid and placebo groups received antibiotics; one trial offered delayed prescription of antibiotics based on clinical assessment. Only two trials reported funding sources (government and a university foundation). In addition to any effect of antibiotics and analgesia, corticosteroids increased the likelihood of complete resolution of pain at 24 hours by 2.40 times (risk ratio (RR) 2.4, 95% confidence interval (CI) 1.29 to 4.47; P = 0.006; I² = 67%; high-certainty evidence) and at 48 hours by 1.5 times (RR 1.50, 95% CI 1.27 to 1.76; P < 0.001; I² = 0%; high-certainty evidence). Five people need to be treated to prevent one person continuing to experience pain at 24 hours. Corticosteroids also reduced the mean time to onset of pain relief and the mean time to complete resolution of pain by 6 and 11.6 hours, respectively, although significant heterogeneity was present (moderate-certainty evidence). At 24 hours, pain (assessed by visual analogue scales) was reduced by an additional 10.6% by corticosteroids (moderate-certainty evidence). No differences were reported in recurrence/relapse rates, days missed from work or school, or adverse events for participants taking corticosteroids compared to placebo. However, the reporting of adverse events was poor, and only two trials included children or reported days missed from work or school. The included studies were assessed as moderate quality evidence, but the small number of included studies has the potential to increase the uncertainty, particularly in terms of applying these results to children. AUTHORS' CONCLUSIONS: Oral or intramuscular corticosteroids, in addition to antibiotics, moderately increased the likelihood of both resolution and improvement of pain in participants with sore throat. Given the limited benefit, further research into the harms and benefits of short courses of steroids is needed to permit informed decision-making.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Anti-Bacterial Agents/administration & dosage , Pharyngitis/drug therapy , Adolescent , Adult , Child , Child, Preschool , Drug Therapy, Combination/methods , Humans , Middle Aged , Randomized Controlled Trials as Topic , Time Factors , Tonsillitis/drug therapy , Treatment Outcome , Young Adult
7.
Front Immunol ; 10: 1254, 2019.
Article in English | MEDLINE | ID: mdl-31214195

ABSTRACT

The malaria genome encodes over 5,000 proteins and many of these have also been proposed to be potential vaccine candidates, although few of these have been tested clinically. RH5 is one of the leading blood-stage Plasmodium falciparum malaria vaccine antigens and Phase I/II clinical trials of vaccines containing this antigen are currently underway. Its likely mechanism of action is to elicit antibodies that can neutralize merozoites by blocking their invasion of red blood cells (RBC). However, many other antigens could also elicit neutralizing antibodies against the merozoite, and most of these have never been compared directly to RH5. The objective of this study was to compare a range of blood-stage antigens to RH5, to identify any antigens that outperform or synergize with anti-RH5 antibodies. We selected 55 gene products, covering 15 candidate antigens that have been described in the literature and 40 genes selected on the basis of bioinformatics functional prediction. We were able to make 20 protein-in-adjuvant vaccines from the original selection. Of these, S-antigen and CyRPA robustly elicited antibodies with neutralizing properties. Anti-CyRPA IgG generally showed additive GIA with anti-RH5 IgG, although high levels of anti-CyRPA-specific rabbit polyclonal IgG were required to achieve 50% GIA. Our data suggest that further vaccine antigen screening efforts are required to identify a second merozoite target with similar antibody-susceptibility to RH5.


Subject(s)
Erythrocytes/immunology , Erythrocytes/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Merozoites/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Antibody Specificity/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Female , Humans , Immunoglobulin G/immunology , Malaria, Falciparum/prevention & control , Mice , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Polymorphism, Genetic , Protozoan Proteins/genetics , Protozoan Proteins/immunology
8.
Sci Rep ; 8(1): 3390, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29467399

ABSTRACT

Heterologous prime-boost vaccination with viral vectors simian adenovirus 63 (ChAd63) and Modified Vaccinia Ankara (MVA) induces potent T cell and antibody responses in humans. The 8-week regimen demonstrates significant efficacy against malaria when expressing the pre-erythrocytic malaria antigen Thrombospondin-Related Adhesion Protein fused to a multiple epitope string (ME-TRAP). We tested these vaccines in 7 new 4- and 8- week interval schedules to evaluate safety and immunogenicity of multiple ChAd63 ME-TRAP priming vaccinations (denoted A), multiple MVA ME-TRAP boosts (denoted M) and alternating vectors. All regimens exhibited acceptable reactogenicity and CD8+ T cell immunogenicity was enhanced with a 4-week interval (AM) and with incorporation of additional ChAd63 ME-TRAP vaccination at 4- or 8-weeks (AAM or A_A_M). Induction of TRAP antibodies was comparable between schedules. T cell immunity against the ChAd63 hexon did not affect T cell responses to the vaccine insert, however pre-vaccination ChAd63-specific T cells correlated with reduced TRAP antibodies. Vaccine-induced antibodies against MVA did not affect TRAP antibody induction, and correlated positively with ME-TRAP-specific T cells. This study identifies potentially more effective immunisation regimens to assess in Phase IIa trials and demonstrates a degree of flexibility with the timing of vectored vaccine administration, aiding incorporation into existing vaccination programmes.


Subject(s)
Epitopes/immunology , Genetic Vectors/immunology , Liver/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Adenoviruses, Simian/immunology , Adolescent , Adult , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunization, Secondary/methods , Male , Middle Aged , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccination/methods , Vaccinia/immunology , Vaccinia virus/immunology , Young Adult
9.
JCI Insight ; 2(12)2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28614791

ABSTRACT

BACKGROUND: Plasmodium vivax is the most widespread human malaria geographically; however, no effective vaccine exists. Red blood cell invasion by the P. vivax merozoite depends on an interaction between the Duffy antigen receptor for chemokines (DARC) and region II of the parasite's Duffy-binding protein (PvDBP_RII). Naturally acquired binding-inhibitory antibodies against this interaction associate with clinical immunity, but it is unknown whether these responses can be induced by human vaccination. METHODS: Safety and immunogenicity of replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and modified vaccinia virus Ankara (MVA) viral vectored vaccines targeting PvDBP_RII (Salvador I strain) were assessed in an open-label dose-escalation phase Ia study in 24 healthy UK adults. Vaccines were delivered by the intramuscular route in a ChAd63-MVA heterologous prime-boost regimen using an 8-week interval. RESULTS: Both vaccines were well tolerated and demonstrated a favorable safety profile in malaria-naive adults. PvDBP_RII-specific ex-vivo IFN-γ T cell, antibody-secreting cell, memory B cell, and serum IgG responses were observed after the MVA boost immunization. Vaccine-induced antibodies inhibited the binding of vaccine homologous and heterologous variants of recombinant PvDBP_RII to the DARC receptor, with median 50% binding-inhibition titers greater than 1:100. CONCLUSION: We have demonstrated for the first time to our knowledge that strain-transcending antibodies can be induced against the PvDBP_RII antigen by vaccination in humans. These vaccine candidates warrant further clinical evaluation of efficacy against the blood-stage P. vivax parasite. TRIAL REGISTRATION: Clinicaltrials.gov NCT01816113. FUNDING: Support was provided by the UK Medical Research Council, UK National Institute of Health Research Oxford Biomedical Research Centre, and the Wellcome Trust.

10.
J Immunol ; 197(4): 1242-51, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27412417

ABSTRACT

There is great interest in the development of Ab-inducing subunit vaccines targeting infections, including HIV, malaria, and Ebola. We previously reported that adenovirus vectored vaccines are potent in priming Ab responses, but uncertainty remains regarding the optimal approach for induction of humoral immune responses. In this study, using OVA as a model Ag, we assessed the magnitude of the primary and anamnestic Ag-specific IgG responses of mice to four clinically relevant vaccine formulations: replication-deficient adenovirus; modified vaccinia Ankara (a poxvirus); protein with alum; and protein in the squalene oil-in-water adjuvant Addavax. We then used flow cytometric assays capable of measuring total and Ag-specific germinal center (GC) B cell and follicular Th cell responses to compare the induction of these responses by the different formulations. We report that adenovirus vectored vaccines induce Ag insert-specific GC B cell and Ab responses of a magnitude comparable to those induced by a potent protein/squalene oil-in-water formulation whereas-despite a robust overall GC response-the insert-specific GC B cell and Ab responses induced by modified vaccinia Ankara were extremely weak. Ag-specific follicular Th cell responses to adenovirus vectored vaccines exceeded those induced by other platforms at day 7 after immunization. We found little evidence that innate immune activation by adenovirus may act as an adjuvant in such a manner that the humoral response to a recombinant protein may be enhanced by coadministering with an adenovirus lacking a transgene of interest. Overall, these studies provide further support for the use of replication-deficient adenoviruses to induce humoral responses.


Subject(s)
Adenoviridae/immunology , Adjuvants, Immunologic/pharmacology , B-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , Viral Vaccines/immunology , Animals , Antigens/immunology , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Genetic Vectors , Germinal Center/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Ovalbumin/immunology
11.
Sci Rep ; 6: 18848, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26743316

ABSTRACT

Transmission-blocking vaccines (TBV) target the sexual-stages of the malaria parasite in the mosquito midgut and are widely considered to be an essential tool for malaria elimination. High-titer functional antibodies are required against target antigens to achieve effective transmission-blocking activity. We have fused Pfs25, the leading malaria TBV candidate antigen to IMX313, a molecular adjuvant and expressed it both in ChAd63 and MVA viral vectors and as a secreted protein-nanoparticle. Pfs25-IMX313 expressed from viral vectors or as a protein-nanoparticle is significantly more immunogenic and gives significantly better transmission-reducing activity than monomeric Pfs25. In addition, we demonstrate that the Pfs25-IMX313 protein-nanoparticle leads to a qualitatively improved antibody response in comparison to soluble Pfs25, as well as to significantly higher germinal centre (GC) responses. These results demonstrate that antigen multimerization using IMX313 is a very promising strategy to enhance antibody responses against Pfs25, and that Pfs25-IMX313 is a highly promising TBV candidate vaccine.


Subject(s)
Adjuvants, Immunologic/genetics , Antibodies, Protozoan/biosynthesis , Immunogenicity, Vaccine , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Protozoan Proteins/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Culicidae/drug effects , Culicidae/parasitology , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/immunology , Germinal Center/drug effects , Germinal Center/immunology , Humans , Insect Vectors/drug effects , Insect Vectors/parasitology , Life Cycle Stages/drug effects , Life Cycle Stages/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Mice, Inbred BALB C , Pichia/genetics , Pichia/metabolism , Plasmids/chemistry , Plasmids/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccination , Vaccines, Synthetic
12.
Front Immunol ; 6: 348, 2015.
Article in English | MEDLINE | ID: mdl-26217340

ABSTRACT

Malaria vaccine development has largely focused on Plasmodium falciparum; however, a reawakening to the importance of Plasmodium vivax has spurred efforts to develop vaccines against this difficult to treat and at times severe form of relapsing malaria, which constitutes a significant proportion of human malaria cases worldwide. The almost complete dependence of P. vivax red blood cell invasion on the interaction of the P. vivax Duffy-binding protein region II (PvDBP_RII) with the human Duffy antigen receptor for chemokines (DARC) makes this antigen an attractive vaccine candidate against blood-stage P. vivax. Here, we generated both preclinical and clinically compatible adenoviral and poxviral vectored vaccine candidates expressing the Salvador I allele of PvDBP_RII - including human adenovirus serotype 5 (HAdV5), chimpanzee adenovirus serotype 63 (ChAd63), and modified vaccinia virus Ankara (MVA) vectors. We report on the antibody and T cell immunogenicity of these vaccines in mice or rabbits, either used alone in a viral vectored prime-boost regime or in "mixed-modality" adenovirus prime - protein-in--adjuvant boost regimes (using a recombinant PvDBP_RII protein antigen formulated in Montanide(®)ISA720 or Abisco(®)100 adjuvants). Antibodies induced by these regimes were found to bind to native parasite antigen from P. vivax infected Thai patients and were capable of inhibiting the binding of PvDBP_RII to its receptor DARC using an in vitro binding inhibition assay. In recent years, recombinant ChAd63 and MVA vectors have been quickly translated into human clinical trials for numerous antigens from P. falciparum as well as a growing number of other pathogens. The vectors reported here are immunogenic in small animals, elicit antibodies against PvDBP_RII, and have recently entered clinical trials, which will provide the first assessment of the safety and immunogenicity of the PvDBP_RII antigen in humans.

13.
J Infect Dis ; 211(7): 1076-86, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25336730

ABSTRACT

BACKGROUND: Circumsporozoite protein (CS) is the antigenic target for RTS,S, the most advanced malaria vaccine to date. Heterologous prime-boost with the viral vectors simian adenovirus 63 (ChAd63)-modified vaccinia virus Ankara (MVA) is the most potent inducer of T-cells in humans, demonstrating significant efficacy when expressing the preerythrocytic antigen insert multiple epitope-thrombospondin-related adhesion protein (ME-TRAP). We hypothesized that ChAd63-MVA containing CS may result in a significant clinical protective efficacy. METHODS: We conducted an open-label, 2-site, partially randomized Plasmodium falciparum sporozoite controlled human malaria infection (CHMI) study to compare the clinical efficacy of ChAd63-MVA CS with ChAd63-MVA ME-TRAP. RESULTS: One of 15 vaccinees (7%) receiving ChAd63-MVA CS and 2 of 15 (13%) receiving ChAd63-MVA ME-TRAP achieved sterile protection after CHMI. Three of 15 vaccinees (20%) receiving ChAd63-MVA CS and 5 of 15 (33%) receiving ChAd63-MVA ME-TRAP demonstrated a delay in time to treatment, compared with unvaccinated controls. In quantitative polymerase chain reaction analyses, ChAd63-MVA CS was estimated to reduce the liver parasite burden by 69%-79%, compared with 79%-84% for ChAd63-MVA ME-TRAP. CONCLUSIONS: ChAd63-MVA CS does reduce the liver parasite burden, but ChAd63-MVA ME-TRAP remains the most promising antigenic insert for a vectored liver-stage vaccine. Detailed analyses of parasite kinetics may allow detection of smaller but biologically important differences in vaccine efficacy that can influence future vaccine development. CLINICAL TRIALS REGISTRATION: NCT01623557.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adenoviruses, Simian/genetics , Adenoviruses, Simian/immunology , Adolescent , Adult , Antibodies, Protozoan/biosynthesis , Epitopes/immunology , Female , Genetic Vectors , Humans , Interferon-gamma/immunology , Liver/virology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Middle Aged , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Young Adult
14.
PLoS One ; 9(9): e107903, 2014.
Article in English | MEDLINE | ID: mdl-25254500

ABSTRACT

The development of protective vaccines against many difficult infectious pathogens will necessitate the induction of effective antibody responses. Here we assess humoral immune responses against two antigens from the blood-stage merozoite of the Plasmodium falciparum human malaria parasite--MSP1 and AMA1. These antigens were delivered to healthy malaria-naïve adult volunteers in Phase Ia clinical trials using recombinant replication-deficient viral vectors--ChAd63 to prime the immune response and MVA to boost. In subsequent Phase IIa clinical trials, immunized volunteers underwent controlled human malaria infection (CHMI) with P. falciparum to assess vaccine efficacy, whereby all but one volunteer developed low-density blood-stage parasitemia. Here we assess serum antibody responses against both the MSP1 and AMA1 antigens following i) ChAd63-MVA immunization, ii) immunization and CHMI, and iii) primary malaria exposure in the context of CHMI in unimmunized control volunteers. Responses were also assessed in a cohort of naturally-immune Kenyan adults to provide comparison with those induced by a lifetime of natural malaria exposure. Serum antibody responses against MSP1 and AMA1 were characterized in terms of i) total IgG responses before and after CHMI, ii) responses to allelic variants of MSP1 and AMA1, iii) functional growth inhibitory activity (GIA), iv) IgG avidity, and v) isotype responses (IgG1-4, IgA and IgM). These data provide the first in-depth assessment of the quality of adenovirus-MVA vaccine-induced antibody responses in humans, along with assessment of how these responses are modulated by subsequent low-density parasite exposure. Notable differences were observed in qualitative aspects of the human antibody responses against these malaria antigens depending on the means of their induction and/or exposure of the host to the malaria parasite. Given the continued clinical development of viral vectored vaccines for malaria and a range of other diseases targets, these data should help to guide further immuno-monitoring studies of vaccine-induced human antibody responses.


Subject(s)
Adenoviridae/immunology , Antigens, Protozoan/immunology , Immunity, Humoral/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Vaccination/methods , Vaccinia virus/immunology , Adenoviridae/genetics , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Blood/parasitology , Environmental Exposure/adverse effects , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Malaria Vaccines/genetics , Malaria, Falciparum/blood , Malaria, Falciparum/immunology , Pan troglodytes , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology , Species Specificity , Vaccinia virus/genetics
15.
Mol Ther ; 22(12): 2142-2154, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25156127

ABSTRACT

The development of effective vaccines against difficult disease targets will require the identification of new subunit vaccination strategies that can induce and maintain effective immune responses in humans. Here we report on a phase 1a clinical trial using the AMA1 antigen from the blood-stage Plasmodium falciparum malaria parasite delivered either as recombinant protein formulated with Alhydrogel adjuvant with and without CPG 7909, or using recombinant vectored vaccines--chimpanzee adenovirus ChAd63 and the orthopoxvirus MVA. A variety of promising "mixed-modality" regimens were tested. All volunteers were primed with ChAd63, and then subsequently boosted with MVA and/or protein-in-adjuvant using either an 8- or 16-week prime-boost interval. We report on the safety of these regimens, as well as the T cell, B cell, and serum antibody responses. Notably, IgG antibody responses primed by ChAd63 were comparably boosted by AMA1 protein vaccine, irrespective of whether CPG 7909 was included in the Alhydrogel adjuvant. The ability to improve the potency of a relatively weak aluminium-based adjuvant in humans, by previously priming with an adenoviral vaccine vector encoding the same antigen, thus offers a novel vaccination strategy for difficult or neglected disease targets when access to more potent adjuvants is not possible.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Protozoan/administration & dosage , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Adenoviruses, Simian/genetics , Adult , Aluminum Hydroxide/administration & dosage , Antigens, Protozoan/immunology , Combined Modality Therapy , Genetic Vectors/administration & dosage , Humans , Immunization, Secondary , Male , Middle Aged , Oligodeoxyribonucleotides/administration & dosage , Orthopoxvirus/genetics , Vaccination , Young Adult
16.
Immunology ; 141(4): 628-44, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24303947

ABSTRACT

Acquisition of non-sterilizing natural immunity to Plasmodium falciparum malaria has been shown in low transmission areas following multiple exposures. However, conflicting data from endemic areas suggest that the parasite may interfere with the induction of effective B-cell responses. To date, the impact of blood-stage parasite exposure on antigen-specific B cells has not been reported following controlled human malaria infection (CHMI). Here we analysed human B-cell responses in a series of Phase I/IIa clinical trials, which include CHMI, using candidate virus-vectored vaccines encoding two blood-stage antigens: merozoite surface protein 1 (MSP1) and apical membrane antigen 1 (AMA1). Previously vaccinated volunteers show boosting of pre-existing antigen-specific memory B-cell (mBC) responses following CHMI. In contrast, unvaccinated malaria-naive control volunteers developed an mBC response against MSP1 but not AMA1. Serum IgG correlated with the mBC response after booster vaccination but this relationship was less well maintained following CHMI. A significant reduction in peripheral MSP1-specific mBC was observed at the point of diagnosis of blood-stage infection. This was coincident with a reduction in peripheral blood B-cell subsets expressing CXCR3 and elevated serum levels of interferon-γ and CXCL9, suggesting migration away from the periphery. These CHMI data confirm that mBC and antibody responses can be induced and boosted by blood-stage parasite exposure, in support of epidemiological studies on low-level parasite exposure.


Subject(s)
Adenoviridae/genetics , Antigens, Protozoan/administration & dosage , B-Lymphocytes/drug effects , Immunization , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Membrane Proteins/administration & dosage , Merozoite Surface Protein 1/administration & dosage , Plasmodium falciparum/drug effects , Protozoan Proteins/administration & dosage , Vaccinia virus/genetics , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/parasitology , Chemokine CXCL9/blood , Genetic Vectors , Humans , Immunization Schedule , Immunization, Secondary , Immunoglobulin G/blood , Immunologic Memory , Interferon-gamma/blood , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/blood , Malaria, Falciparum/diagnosis , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Membrane Proteins/genetics , Membrane Proteins/immunology , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Receptors, CXCR3/blood , Time Factors
17.
J Leukoc Biol ; 95(2): 369-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24163420

ABSTRACT

New tools are required to expedite the development of an effective vaccine against the blood-stage infection with the human malaria parasite Plasmodium falciparum. This work describes the assessment of the ADRB assay in a mouse model, characterizing the functional interaction between antimalarial serum antibodies and FcRs upon neutrophils. We describe a reproducible, antigen-specific assay, dependent on functional FcR signaling, and show that ADRB activity is induced equally by IgG1 and IgG2a isotypes and is modulated by blocking FcR function. However, following immunization of mice with the blood-stage vaccine candidate antigen MSP142, no measurable ADRB activity was induced against PEMS and neither was vaccine efficacy modulated against Plasmodium yoelii blood-stage challenge in γ(-/-) mice compared with WT mice. In contrast, following a primary, nonlethal P. yoelii parasite challenge, serum from vaccinated mice and nonimmunized controls showed anti-PEMS ADRB activity. Upon secondary challenge, nonimmunized γ(-/-) mice showed a reduced ability to control blood-stage parasitemia compared with immunized γ(-/-) mice; however, WT mice, depleted of their neutrophils, did not lose their ability to control infection. Thus, whereas neutrophil-induced ADRB against PEMS does not appear to play a role in protection against P. yoelii rodent malaria, induction of ADRB activity after challenge suggests that antigen targets of anti-PEMS ADRB activity remain to be established, as well as further supporting the observation that ADRB activity to P. falciparum arises following repeated natural exposure.


Subject(s)
Antibodies, Protozoan/immunology , Immunoassay/methods , Malaria/immunology , Malaria/parasitology , Neutrophils/immunology , Plasmodium yoelii/immunology , Respiratory Burst/immunology , Animals , Antigens, Protozoan/immunology , Female , Humans , Immunoglobulin G/metabolism , Mice , Parasites/immunology , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Treatment Outcome
18.
Sci Rep ; 3: 1706, 2013.
Article in English | MEDLINE | ID: mdl-23609325

ABSTRACT

Rodent malaria species Plasmodium yoelii and P. chabaudi have been widely used to validate vaccine approaches targeting blood-stage merozoite antigens. However, increasing data suggest the P. berghei rodent malaria may be able to circumvent vaccine-induced anti-merozoite responses. Here we confirm a failure to protect against P. berghei, despite successful antibody induction against leading merozoite antigens using protein-in-adjuvant or viral vectored vaccine delivery. No subunit vaccine approach showed efficacy in mice following immunization and challenge with the wild-type P. berghei strains ANKA or NK65, or against a chimeric parasite line encoding a merozoite antigen from P. falciparum. Protection was not improved in knockout mice lacking the inhibitory Fc receptor CD32b, nor against a Δsmac P. berghei parasite line with a non-sequestering phenotype. An improved understanding of the mechanisms responsible for protection, or failure of protection, against P. berghei merozoites could guide the development of an efficacious vaccine against P. falciparum.


Subject(s)
Antibody Formation/immunology , Antimalarials/immunology , Malaria Vaccines/immunology , Malaria/immunology , Merozoites/immunology , Plasmodium berghei/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cell Line , Female , HEK293 Cells , Humans , Immunization/methods , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Rabbits , Receptors, IgG/immunology , Rodentia/immunology
19.
Expert Rev Vaccines ; 12(4): 365-78, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23560918

ABSTRACT

Viral vectored vaccine delivery platforms have traditionally been used for the induction of cellular rather than humoral immunity. However, in recent years, recombinant adenoviral and poxviral vectored vaccines have been optimized to induce B-cell responses, resulting in the demonstration of high-titer antibody responses in a wide variety of animal species. These approaches have now been translated, confirming the induction of substantial levels of antigen-specific IgG in a series of Phase I human clinical trials targeting HIV-1 and Plasmodium falciparum malaria. To further improve the induction of antibodies, mixed-modality regimens based on recombinant viral and protein/adjuvant vaccines are now being assessed. However, limited data exist about the underlying mechanisms mediating the induction of B-cell responses by these subunit vaccines and their ability to influence the qualitative aspects of vaccine-induced B-cell populations and immunoglobulin. Future studies in this area are needed to guide the rational design of antibody-inducing subunit vaccine strategies.


Subject(s)
Adenoviridae/genetics , Drug Carriers/administration & dosage , Drug Delivery Systems , Poxviridae/genetics , Vaccination/methods , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Antibodies, Protozoan/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , Clinical Trials, Phase I as Topic , HIV Infections/prevention & control , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control
20.
PLoS One ; 7(9): e44943, 2012.
Article in English | MEDLINE | ID: mdl-22984589

ABSTRACT

Viral vectored vaccines have been shown to induce both T cell and antibody responses in animals and humans. However, the induction of even higher level T cell responses may be crucial in achieving vaccine efficacy against difficult disease targets, especially in humans. Here we investigate the oligomerization domain of the α-chain of C4b-binding protein (C4 bp) as a candidate T cell "molecular adjuvant" when fused to malaria antigens expressed by human adenovirus serotype 5 (AdHu5) vectored vaccines in BALB/c mice. We demonstrate that i) C-terminal fusion of an oligomerization domain can enhance the quantity of antigen-specific CD4(+) and CD8(+) T cell responses induced in mice after only a single immunization of recombinant AdHu5, and that the T cells maintain similar functional cytokine profiles; ii) an adjuvant effect is observed for AdHu5 vectors expressing either the 42 kDa C-terminal domain of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(42)) or the 83 kDa ectodomain of P. falciparum strain 3D7 apical membrane antigen 1 (PfAMA1), but not a candidate 128kDa P. falciparum MSP1 biallelic fusion antigen; iii) following two homologous immunizations of AdHu5 vaccines, antigen-specific T cell responses are further enhanced, however, in both BALB/c mice and New Zealand White rabbits no enhancement of functional antibody responses is observed; and iv) that the T cell adjuvant activity of C4 bp is not dependent on a functional Fc-receptor γ-chain in the host, but is associated with the oligomerization of small (<80 kDa) antigens expressed by recombinant AdHu5. The oligomerization domain of C4 bp can thus adjuvant T cell responses induced by AdHu5 vectors against selected antigens and its clinical utility as well as mechanism of action warrant further investigation.


Subject(s)
Adenoviridae/genetics , Complement C4b-Binding Protein/metabolism , Malaria/prevention & control , T-Lymphocytes/cytology , Adjuvants, Immunologic/genetics , Animals , Antigens, Protozoan/genetics , Female , Genetic Vectors , Malaria Vaccines/genetics , Merozoite Surface Protein 1/genetics , Mice , Mice, Inbred BALB C , Plasmodium falciparum/genetics , Plasmodium yoelii/genetics , Protein Structure, Tertiary , Rabbits , Receptors, IgG/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL