Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Vaccine ; 37(43): 6248-6254, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31500964

ABSTRACT

Emerging and re-emerging infectious diseases are an expanding global threat to public health, security, and economies. Increasing populations, urbanization, deforestation, climate change, anti-vaccination movements, war, and international travel are some of the contributing factors to this trend. The recent Ebola, MERS-CoV, and Zika outbreaks demonstrated we are insufficiently prepared to respond with proven safe and effective countermeasures (i.e., vaccines and therapeutics). The State University of New York Upstate Medical University and the Trudeau Institute convened a summit of key opinion and thought leaders in the life sciences and biomedical research and development enterprises to explore global biopreparedness challenges, take an inventory of existing capabilities and capacities related to preparation and response, assess current "gaps," and prospect what could be done to improve our position. Herein we describe the summit proceedings, "Translational Immunology Supporting Biomedical Countermeasure Development for Emerging Vector-borne Viral Diseases," held October 2-3, 2018, at the Trudeau Institute in Saranac Lake, NY.


Subject(s)
Communicable Diseases, Emerging , Disease Vectors , Viral Vaccines/pharmacology , Virus Diseases/prevention & control , Animals , Clinical Trials as Topic , Communicable Diseases, Emerging/prevention & control , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Influenza, Human/etiology , Influenza, Human/prevention & control , Pregnancy , Pregnancy Complications, Infectious/etiology , Translational Research, Biomedical , Viral Vaccines/therapeutic use , Zika Virus Infection/etiology , Zika Virus Infection/prevention & control
2.
Mil Med ; 182(5): e1749-e1756, 2017 05.
Article in English | MEDLINE | ID: mdl-29087920

ABSTRACT

BACKGROUND: Scientific publication in academic literature is a key venue in which the U.S. Department of Defense's Global Emerging Infections Surveillance and Response System (GEIS) program disseminates infectious disease surveillance data. Bibliometric analyses are tools to evaluate scientific productivity and impact of published research, yet are not routinely used for disease surveillance. Our objective was to incorporate bibliometric indicators to measure scientific productivity and impact of GEIS-funded infectious disease surveillance, and assess their utility in the management of the GEIS surveillance program. METHODS: Metrics on GEIS program scientific publications, project funding, and countries of collaborating institutions from project years 2006 to 2012 were abstracted from annual reports and program databases and organized by the six surveillance priority focus areas: respiratory infections, gastrointestinal infections, febrile and vector-borne infections, antimicrobial resistance, sexually transmitted infections, and capacity building and outbreak response. Scientific productivity was defined as the number of scientific publications in peer-reviewed literature derived from GEIS-funded projects. Impact was defined as the number of citations of a GEIS-funded publication by other peer-reviewed publications, and the Thomson Reuters 2-year journal impact factor. Indicators were retrieved from the Web of Science and Journal Citation Report. To determine the global network of international collaborations between GEIS partners, countries were organized by the locations of collaborating institutions. RESULTS: Between 2006 and 2012, GEIS distributed approximately US $330 million to support 921 total projects. On average, GEIS funded 132 projects (range 96-160) with $47 million (range $43 million-$53 million), annually. The predominant surveillance focus areas were respiratory infections with 317 (34.4%) projects and $225 million, and febrile and vector-borne infections with 274 (29.8%) projects and $45 million. The number of annual respiratory infections-related projects peaked in 2006 and 2009. The number of febrile and vector-borne infections projects increased from 29 projects in 2006 to 58 in 2012. There were 651 articles published in 147 different peer-reviewed journals, with an average Thomson Reuters 2-year journal impact factor of 4.2 (range 0.3-53.5). On average, 93 articles were published per year (range 67-117) with $510,000 per publication. Febrile and vector-borne, respiratory, and gastrointestinal infections had 287, 167, and 73 articles published, respectively. Of the 651 articles published, 585 (89.9%) articles were cited at least once (range 1-1,045). Institutions from 90 countries located in all six World Health Organization regions collaborated with surveillance projects. CONCLUSIONS: These findings summarize the GEIS-funded surveillance portfolio between 2006 and 2012, and demonstrate the scientific productivity and impact of the program in each of the six disease surveillance priority focus areas. GEIS might benefit from further financial investment in both the febrile and vector-borne and sexually transmitted infections surveillance priority focus areas and increasing peer-reviewed publications of surveillance data derived from respiratory infections projects. Bibliometric indicators are useful to measure scientific productivity and impact in surveillance systems; and this methodology can be utilized as a management tool to assess future changes to GEIS surveillance priorities. Additional metrics should be developed when peer-reviewed literature is not used to disseminate noteworthy accomplishments.


Subject(s)
Biomedical Research/trends , Civil Defense/standards , Disease Outbreaks , Population Surveillance/methods , Animals , Bibliometrics , Civil Defense/organization & administration , Disease Vectors , Global Health , Humans , Respiratory Tract Infections/epidemiology , World Health Organization/organization & administration
3.
Vaccine ; 35(40): 5366-5372, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28844407

ABSTRACT

Influenza viruses frequently acquire mutations undergoing antigenic drift necessitating annual evaluation of vaccine strains. Highly conserved epitopes have been identified in the hemagglutinin (HA) head and stem regions, however, current influenza vaccines induce only limited responses to these conserved sites. Here, we describe a novel seasonal recombinant HA nanoparticle influenza vaccine (NIV) formulated with a saponin-based adjuvant, Matrix-M™. NIV induced hemagglutination inhibition (HAI) and microneutralizing (MN) antibodies against a broad range of influenza A(H3N2) subtypes. In a comparison of NIV against standard-dose and high-dose inactivated influenza vaccines (IIV and IIV-HD, respectively) in ferrets NIV elicited HAI and MN responses exceeding those induced by IIV-HD against homologous A(H3N2) by 7 fold, A(H1N1) by 26 fold, and B strain viruses by 2 fold. NIV also induced MN responses against all historic A/H3N2 strains tested, spanning more than a decade of viral evolution from the 2000-2017 influenza seasons whereas IIV and IIV-HD induced HAI and MN responses were largely directed against the homologous A(H3N2), A(H1N1), and B virus strains. NIV induced superior protection compared to IIV and IIV-HD in ferrets challenged with a homologous or 10-year drifted influenza A(H3N2) strain. HAI positive and HAI negative neutralizing monoclonal antibodies derived from mice immunized with NIV were active against homologous and drifted influenza A(H3N2) strains. Taken together these observations suggest that NIV can induce responses to one or more highly conserved HA head and stem epitopes and result in highly neutralizing antibodies against both homologous and drift strains.


Subject(s)
Ferrets/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Nanoparticles/chemistry , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutination Inhibition Tests , Hemagglutinins/immunology , Hemagglutinins/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Mice
4.
Vaccine ; 35(33): 4079-4087, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28669616

ABSTRACT

Clostridium difficile is the number one cause of nosocomial antibiotic-associated diarrhea in developed countries. Historically, pathogenesis was attributed two homologous glucosylating toxins, toxin-A (TcdA) and toxin-B (TcdB). Over the past decade, however, highly virulent epidemic strains of C. difficile (B1/NAP1/027) have emerged and are linked to an increase in morbidity and mortality. Increased virulence is attributed to multiple factors including: increased production of A- and B-toxins; production of binary toxin (CDT); and the emergence of more toxic TcdB variants (TcdB(027)). TcdB(027) is more cytotoxicity to cells; causes greater tissue damage and toxicity in animals; and is antigenically distinct from historical TcdB (TcdB(003)). Broadly protective vaccines and therapeutic antibody strategies, therefore, may target TcdA, TcdB variants and CDT. To facilitate the generation of multivalent toxin-based C. difficile vaccines and therapeutic antibodies, we have generated fusion proteins constructed from the receptor binding domains (RBD) of TcdA, TcdB(003), TcdB(027) and CDT. Herein, we describe the development of a trivalent toxin (T-toxin) vaccine (CDTb/TcdB(003)/TcdA) and quadravalent toxin (Q-toxin) vaccine (CDTb/TcB(003)/TcdA/TcdB(027)) fusion proteins that retain the protective toxin neutralizing epitopes. Active immunization of mice or hamsters with T-toxin or Q-toxin fusion protein vaccines elicited the generation of toxin neutralizing antibodies to each of the toxins. Hamsters immunized with the Q-toxin vaccine were broadly protected against spore challenge with historical C. difficile 630 (toxinotype 0/ribotype 003) and epidemic NAP1 (toxinotype III/ribotype 027) strains. Fully human polyclonal antitoxin IgG was produced by immunization of transgenic bovine with these fusion proteins. In passive transfer studies, mice were protected against lethal toxin challenge. Hamsters treated with human antitoxin IgG were completely protected when challenged with historical or epidemic strains of C. difficile. The use of chimeric fusion proteins is an attractive approach to producing multivalent antitoxin vaccines and therapeutic polyclonal antibodies for prevention and treatment of C. difficile infections (CDI).


Subject(s)
Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Cross Protection , Immunotoxins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/therapeutic use , Antitoxins/blood , Antitoxins/therapeutic use , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Clostridioides difficile/genetics , Female , Immunoglobulin G/blood , Immunoglobulin G/therapeutic use , Immunotoxins/genetics , Male , Mesocricetus , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
5.
PLoS Negl Trop Dis ; 10(2): e0004423, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26919472

ABSTRACT

BACKGROUND: A vaccine to prevent infection and disease caused by Plasmodium vivax is needed both to reduce the morbidity caused by this parasite and as a key component in efforts to eradicate malaria worldwide. Vivax malaria protein 1 (VMP001), a novel chimeric protein that incorporates the amino- and carboxy- terminal regions of the circumsporozoite protein (CSP) and a truncated repeat region that contains repeat sequences from both the VK210 (type 1) and the VK247 (type 2) parasites, was developed as a vaccine candidate for global use. METHODS: We conducted a first-in-human Phase 1 dose escalation vaccine study with controlled human malaria infection (CHMI) of VMP001 formulated in the GSK Adjuvant System AS01B. A total of 30 volunteers divided into 3 groups (10 per group) were given 3 intramuscular injections of 15 µg, 30 µg, or 60 µg respectively of VMP001, all formulated in 500 µL of AS01B at each immunization. All vaccinated volunteers participated in a P. vivax CHMI 14 days following the third immunization. Six non-vaccinated subjects served as infectivity controls. RESULTS: The vaccine was shown to be well tolerated and immunogenic. All volunteers generated robust humoral and cellular immune responses to the vaccine antigen. Vaccination did not induce sterile protection; however, a small but significant delay in time to parasitemia was seen in 59% of vaccinated subjects compared to the control group. An association was identified between levels of anti-type 1 repeat antibodies and prepatent period. SIGNIFICANCE: This trial was the first to assess the efficacy of a P. vivax CSP vaccine candidate by CHMI. The association of type 1 repeat-specific antibody responses with delay in the prepatency period suggests that augmenting the immune responses to this domain may improve strain-specific vaccine efficacy. The availability of a P. vivax CHMI model will accelerate the process of P. vivax vaccine development, allowing better selection of candidate vaccines for advancement to field trials.


Subject(s)
Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Antibodies, Protozoan/immunology , Female , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Male , Middle Aged , Protozoan Proteins/administration & dosage , Protozoan Proteins/adverse effects , Vaccination , Young Adult
6.
Clin Microbiol Rev ; 28(3): 743-800, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26085551

ABSTRACT

This comprehensive review outlines the impact of military-relevant respiratory infections, with special attention to recruit training environments, influenza pandemics in 1918 to 1919 and 2009 to 2010, and peacetime operations and conflicts in the past 25 years. Outbreaks and epidemiologic investigations of viral and bacterial infections among high-risk groups are presented, including (i) experience by recruits at training centers, (ii) impact on advanced trainees in special settings, (iii) morbidity sustained by shipboard personnel at sea, and (iv) experience of deployed personnel. Utilizing a pathogen-by-pathogen approach, we examine (i) epidemiology, (ii) impact in terms of morbidity and operational readiness, (iii) clinical presentation and outbreak potential, (iv) diagnostic modalities, (v) treatment approaches, and (vi) vaccine and other control measures. We also outline military-specific initiatives in (i) surveillance, (ii) vaccine development and policy, (iii) novel influenza and coronavirus diagnostic test development and surveillance methods, (iv) influenza virus transmission and severity prediction modeling efforts, and (v) evaluation and implementation of nonvaccine, nonpharmacologic interventions.


Subject(s)
Military Personnel , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control , Disease Outbreaks/statistics & numerical data , Humans , Respiratory Tract Infections/therapy , United States , Vaccination/standards
7.
Mil Med ; 180(2): 145-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25643381

ABSTRACT

During a military public health laboratory symposium held in 1999, concerns were raised that the military health system lacked a standardized antimicrobial resistance (AMR) surveillance system that allowed comparison of data across sites, investigation of trends, and understanding of resistance mechanisms. The purpose of this review was to assess if current AMR activities in the military health system have addressed the aforementioned gaps. It was determined that much progress has already been made within the Department of Defense with respect to monitoring and understanding AMR through initiatives such as the Antimicrobial Resistance Monitoring and Research Program-a strong Department of Defense-wide surveillance program. These surveillance efforts can be made more robust through harmonization of testing and reporting structures across military treatment facilities, and by encouraging military treatment facility participation.


Subject(s)
Drug Resistance, Microbial , Military Medicine/organization & administration , Public Health Surveillance/methods , Humans , Military Personnel , Population Surveillance/methods , Statistics as Topic/methods , Statistics as Topic/standards , United States , Veterans Health
8.
J Epidemiol Glob Health ; 5(1): 23-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25700920

ABSTRACT

In developing countries, public health professionals and scientists need targeted training and practical skills to respond to global emerging infectious disease threats. The Certificate in Emerging Infectious Disease Research was developed in 2008 to aid such professionals to respond to complex emerging disease problems. The short-course was modified slightly in 2013 and renamed the Certificate in One Health. To evaluate the immediate impact of the short-course, an online survey of 176 past participants from both the courses was conducted. The survey tool assessed the program's process, impact, and outcome measures respectively via assessing the courses' perceived strengths and weaknesses, perceived skills gained, and the participants' current position, publication status, funding status, and educational attainment; 85 (48.3%) participants completed the survey. Reported program strengths included the curriculum, expertise of lecturers, and diversity of the training cohort. The principal reported weakness was the compressed academic schedule. The most frequently reported benefits included: epidemiological and biostatistical skills, followed by One-Health knowledge, and research skills. Twenty-eight percent of the survey respondents reported publishing one or more manuscripts since completing the course and 21% reported receiving research funding. The course appears to have had a positive, immediate impact on the students' self-perceived knowledge and capabilities.


Subject(s)
Certification/standards , Communicable Diseases, Emerging/prevention & control , Public Health/education , Research/education , Curriculum/standards , Florida , Humans , Professional Competence/standards , Professional Competence/statistics & numerical data , Students , Universities
9.
Mil Med ; 180(1): 7-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25562850

ABSTRACT

From April 24 to 26, 2013, the Armed Forces Health Surveillance Center and the U.S. Africa Command cosponsored the inaugural meeting of the West Africa Malaria Task Force in Accra, Ghana. The meeting's purpose was to identify common challenges, explore regional and transcontinental collaborations, and to share knowledge about best practices in the fight against malaria in West Africa. Military representatives from Benin, Burkina Faso, Ghana, Liberia, Niger, Nigeria, Senegal, and Togo participated in the Task Force; various U.S. Government agencies were also represented, including the Department of Defense, the Centers for Disease Control and Prevention, and the Agency for International Development. African nation participants presented brief overviews of their military's malaria prevention and control measures, surveillance programs, diagnostic capabilities, and treatment regimens emphasizing gaps within existing programs. Representatives from U.S. agencies discussed activities and capabilities relevant for the region, challenges and lessons learned regarding malaria, and highlighted opportunities for enhanced partnerships to counter malaria in West Africa. This article summarizes the major conclusions of the Task Force meeting, identifies relevant focus areas for future Task Force activities, and outlines opportunities for further inclusion of West African militaries to improve regional malaria surveillance and control efforts.


Subject(s)
Malaria/prevention & control , Military Personnel , Population Surveillance , Africa, Western , Evidence-Based Medicine , Humans , Malaria/diagnosis , Malaria/drug therapy , United States
11.
Vaccine ; 32(19): 2251-9, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24126211

ABSTRACT

BACKGROUND: Novel influenza viruses continue to pose a potential pandemic threat worldwide. In recent years, plants have been used to produce recombinant proteins, including subunit vaccines. A subunit influenza vaccine, HAC1, based on recombinant hemagglutinin from the 2009 pandemic A/California/04/2009 (H1N1) strain of influenza virus, has been manufactured using a plant virus-based transient expression technology in Nicotiana benthamiana plants and demonstrated to be immunogenic and safe in pre-clinical studies (Shoji et al., 2011). METHODS: A first-in-human, Phase 1, single-center, randomized, placebo-controlled, single-blind, dose escalation study was conducted to investigate safety, reactogenicity and immunogenicity of an HAC1 formulation at three escalating dose levels (15 µg, 45 µg and 90 µg) with and without Alhydrogel(®), in healthy adults 18-50 years of age (inclusive). Eighty participants were randomized into six study vaccine groups, a saline placebo group and an approved monovalent H1N1 vaccine group. Recipients received two doses of vaccine or placebo (except for the monovalent H1N1 vaccine cohort, which received a single dose of vaccine, later followed by a dose of placebo). RESULTS: The experimental vaccine was safe and well tolerated, and comparable to placebo and the approved monovalent H1N1 vaccine. Pain and tenderness at the injection site were the only local solicited reactions reported following vaccinations. Nearly all adverse events were mild to moderate in severity. The HAC1 vaccine was also immunogenic, with the highest seroconversion rates, based on serum hemagglutination-inhibition and virus microneutralization antibody titers, in the 90 µg non-adjuvanted HAC1 vaccine group after the second vaccine dose (78% and 100%, respectively). CONCLUSIONS: This is the first study demonstrating the safety and immunogenicity of a plant-produced subunit H1N1 influenza vaccine in healthy adults. The results support further clinical investigation of the HAC1 vaccine as well as demonstrate the feasibility of the plant-based technology for vaccine antigen production.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Adult , Antibodies, Viral/blood , Female , Humans , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Male , Middle Aged , Recombinant Proteins/immunology , Single-Blind Method , Nicotiana , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , Young Adult
12.
Infect Control Hosp Epidemiol ; 32(11): 1116-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22011540

ABSTRACT

We describe the clinical, microbiologic, and molecular features of the first series of qacA/B-containing strains of methicillin-resistant Staphylococcus aureus from infected US patients. All qac-carrying strains were clonally diverse, and qacA strains exhibited increased tolerance to chlorhexidine as measured by minimum inhibitory concentrations, minimum bactericidal concentrations, and postexposure colony counts.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Bacterial Proteins/genetics , Chlorhexidine/analogs & derivatives , Membrane Transport Proteins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcal Infections/epidemiology , Adult , Aged , Chlorhexidine/pharmacology , District of Columbia/epidemiology , Electrophoresis, Gel, Pulsed-Field , Female , Hospitals, Community , Hospitals, Teaching , Humans , Male , Maryland/epidemiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Microbial Sensitivity Tests , Middle Aged , Penicillin-Binding Proteins , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric , Virginia/epidemiology , Young Adult
13.
Expert Rev Vaccines ; 10(5): 589-99, 2011 May.
Article in English | MEDLINE | ID: mdl-21604980

ABSTRACT

Malaria continues to be a worldwide leading cause of morbidity and mortality, and the development of an effective malaria vaccine remains a research imperative. Of the multiple approaches that have been pursued, the RTS,S/AS01 vaccine candidate represents the most developed and clinically validated malaria vaccine formulation. Throughout its development, increasingly more effective adjuvants have been key in improving the potency of the vaccine. RTS,S-based vaccine formulations have been demonstrated to be safe, well tolerated, immunogenic, and to confer partial efficacy in both malaria-naive and -experienced adults as well as children. Further research to optimize and improve vaccine efficacy is ongoing.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria/epidemiology , Malaria/prevention & control , Adjuvants, Immunologic/adverse effects , Humans , Malaria Vaccines/adverse effects , Vaccines
14.
Am J Trop Med Hyg ; 84(5): 825-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21540397

ABSTRACT

A sensitive biomarker of malaria infection would obviate the need for placebo control arms in clinical trials of malaria prophylactic drugs. Antibodies to the 42-kDa fragment of merozoite surface protein-1 (MSP1(42)) have been identified as a potential marker of malaria exposure in individuals receiving prophylaxis with mefloquine. We conducted an open-label trial to determine the sensitivity of seroconversion to MSP1(42), defined as a fourfold rise in enzyme-linked immunosorbant assay (ELISA) titer, among 23 malaria naïve volunteers receiving mefloquine prophylaxis and 6 controls after Plasmodium falciparum sporozoite challenge. All members of the control cohort but none of the mefloquine cohort developed patent parasitemia. Four of six controls but zero of the mefloquine cohort seroconverted to MSP1(42). We conclude that malaria infection during suppressive prophylaxis does not induce antibody response to the blood-stage antigen MSP1(42) in a malaria-naïve study population.


Subject(s)
Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/immunology , Antimalarials/therapeutic use , Malaria, Falciparum/prevention & control , Mefloquine/therapeutic use , Protozoan Proteins/immunology , Adult , Antimalarials/administration & dosage , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mefloquine/administration & dosage
15.
Vaccine ; 28(31): 5135-44, 2010 Jul 12.
Article in English | MEDLINE | ID: mdl-19737527

ABSTRACT

Plasmodium falciparum Liver Stage Antigen 1 (LSA-1) is a pre-erythrocytic stage antigen. Our LSA-1 vaccine candidate is a recombinant protein with full-length C- and N-terminal flanking domains and two of the 17 amino acid repeats from the central repeat region termed "LSA-NRC." We describe the first Phase I/II study of this recombinant LSA-NRC protein formulated with either the AS01 or AS02 adjuvant system. We conducted an open-label Phase I/II study. Thirty-six healthy malaria-naïve adults received one of four formulations by intra-deltoid injection on a 0 and 1 month schedule; low dose (LD) LSA-NRC/AS01:10microg LSA-NRC/0.5ml AS01 (n=5), high dose (HD) LSA-NRC/AS01: 50microg LSA-NRC/0.5ml AS01 (n=13); LD LSA-NRC/AS02: 10microg LSA-NRC/0.5ml AS02 (n=5) and HD LSA-NRC/AS02: 50microg LSA-NRC/0.5ml AS02 (n=13). Two weeks post-second immunization, the high dose vaccinees and 6 non-immunized infectivity controls underwent experimental malaria sporozoite challenge. The vaccines showed a reassuring safety profile but were moderately reactogenic. There were no serious adverse events. All subjects seroconverted after the first immunization. Following the second immunization, LSA-1-specific CD4+ T cells producing two cytokines (IL-2 and IFN-gamma) were found by intra-cellular staining in all subjects in the LD LSA-NRC/AS01B group and in 3 of 5 subjects in the LD LSA-NRC/AS02 group. In contrast, the HD LSA-NRC/AS01 and HD LSA-NRC/AS02 group subjects had fewer LSA-1-specific CD4+ T cells, and minimal to no IFN-gamma responses. There was no increase in LSA-1-specific CD8+ T cells found in any group. Per protocol, 22 high dose vaccinees, but no low dose vaccinees, underwent P. falciparum homologous malaria challenge (3D7 clone). All vaccinees became parasitemic and there was no delay in their pre-patent period versus controls (p=0.95). LSA-NRC/AS01 and LSA-NRC/AS02 elicited antigen-specific antibody and CD4+ T cell responses, but elicited no protective immunity. Although the optimal antigen dose of LSA-NRC may not have been selected for the challenge portion of the protocol, further vaccine development based upon LSA-1 should not be excluded and should include alternative vaccine platforms able to elicit additional effector mechanisms such as CD8+ T cells.


Subject(s)
Antigens, Protozoan/immunology , CD4-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Adjuvants, Immunologic/pharmacology , Adult , Antibodies, Protozoan/blood , Antibody Formation , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunization Schedule , Immunization, Secondary , Interferon-gamma/immunology , Interleukin-2/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria, Falciparum/immunology , Male , Parasitemia/immunology , Plasmodium falciparum/immunology , Recombinant Proteins/immunology , Sporozoites/immunology , Young Adult
16.
PLoS One ; 4(7): e6465, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19649245

ABSTRACT

BACKGROUND: This study advances the clinical development of the RTS,S/AS01B candidate malaria vaccine to malaria endemic populations. As a primary objective it compares the safety and reactogenicity of RTS,S/AS01B to the more extensively evaluated RTS,S/AS02A vaccine. METHODOLOGY: A Phase IIb, single centre, double-blind, controlled trial of 6 months duration with a subsequent 6 month single-blind follow-up conducted in Kisumu West District, Kenya between August 2005 and August 2006. 255 healthy adults aged 18 to 35 years were randomized (1ratio1ratio1) to receive 3 doses of RTS,S/AS02A, RTS,S/AS01B or rabies vaccine (Rabipur; Chiron Behring GmbH) at months 0, 1, 2. The primary objective was the occurrence of severe (grade 3) solicited or unsolicited general (i.e. systemic) adverse events (AEs) during 7 days follow up after each vaccination. PRINCIPAL FINDINGS: Both candidate vaccines had a good safety profile and were well tolerated. One grade 3 systemic AE occurred within 7 days of vaccination (RTS,S/AS01B group). No unsolicited AEs or SAEs were related to vaccine. A marked increase in anti-CS antibody GMTs was observed post Dose 2 of both RTS,S/AS01B (31.6 EU/mL [95% CI: 23.9 to 41.6]) and RTS,S/AS02A (16.7 EU/mL [95% CI: 12.9 to 21.7]). A further increase was observed post Dose 3 in both the RTS,S/AS01B (41.4 EU/mL [95% CI: 31.7 to 54.2]) and RTS,S/AS02A (21.4 EU/mL [95% CI: 16.0 to 28.7]) groups. Anti-CS antibody GMTs were significantly greater with RTS,S/AS01B compared to RTS,S/AS02A at all time points post Dose 2 and Dose 3. Both candidate vaccines produced strong anti-HBs responses. Vaccine efficacy in the RTS,S/AS01B group was 29.5% (95% CI: -15.4 to 56.9, p = 0.164) and in the RTS,S/AS02A group 31.7% (95% CI: -11.6 to 58.2, p = 0.128). CONCLUSIONS: Both candidate malaria vaccines were well tolerated over a 12 month surveillance period. A more favorable immunogenicity profile was observed with RTS,S/AS01B than with RTS,S/AS02A. TRIAL REGISTRATION: Clinicaltrials.gov NCT00197054.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria/prevention & control , Adult , Double-Blind Method , Follow-Up Studies , Humans , Kenya/epidemiology , Malaria/epidemiology , Malaria Vaccines/adverse effects , Single-Blind Method , Treatment Outcome
17.
J Infect Dis ; 200(3): 337-46, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19569965

ABSTRACT

BACKGROUND: To further increase the efficacy of malaria vaccine RTS,S/AS02A, we tested the RTS,S antigen formulated using the AS01B Adjuvant System (GlaxoSmithKline Biologicals). METHODS: In a double-blind, randomized trial, 102 healthy volunteers were evenly allocated to receive RTS,S/AS01B or RTS,S/AS02A vaccine at months 0, 1, and 2 of the study, followed by malaria challenge. Protected vaccine recipients were rechallenged 5 months later. RESULTS: RTS,S/AS01B and RTS,S/AS02A were well tolerated and were safe. The efficacy of RTS,S/AS01B and RTS,S/AS02A was 50% (95% confidence interval [CI], 32.9%-67.1%) and 32% (95% CI, 17.6%-47.6%), respectively. At the time of initial challenge, the RTS,S/AS01B group had greater circumsporozoite protein (CSP)-specific immune responses, including higher immunoglobulin (Ig) G titers, higher numbers of CSP-specific CD4(+) T cells expressing 2 activation markers (interleukin-2, interferon [IFN]-gamma, tumor necrosis factor-alpha, or CD40L), and more ex vivo IFN-gamma enzyme-linked immunospots (ELISPOTs) than did the RTS,S/AS02A group. Protected vaccine recipients had a higher CSP-specific IgG titer (geometric mean titer, 188 vs 73 mug/mL; P < .001), higher numbers of CSP-specific CD4(+) T cells per 10(6) CD4(+) T cells (median, 963 vs 308 CSP-specific CD4(+) T cells/10(6) CD4(+) T cells; P < .001), and higher numbers of ex vivo IFN-gamma ELISPOTs (mean, 212 vs 96 spots/million cells; P < .001). At rechallenge, 4 of 9 vaccine recipients in each group were still completely protected. CONCLUSIONS: The RTS,S/AS01B malaria vaccine warrants comparative field trials with RTS,S/AS02A to determine the best formulation for the protection of children and infants. The association between complete protection and immune responses is a potential tool for further optimization of protection. Trial registration. ClinicalTrials.gov identifier NCT00075049.


Subject(s)
Malaria Vaccines , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Adult , Animals , Antibodies, Protozoan/blood , Cross-Sectional Studies , Double-Blind Method , Follow-Up Studies , Humans , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria Vaccines/standards , Malaria, Falciparum/immunology , Mozambique/epidemiology , Parasitemia , Time Factors
18.
PLoS One ; 4(4): e5254, 2009.
Article in English | MEDLINE | ID: mdl-19390585

ABSTRACT

BACKGROUND: This Phase 1/2a study evaluated the safety, immunogenicity, and efficacy of an experimental malaria vaccine comprised of the recombinant Plasmodium falciparum protein apical membrane antigen-1 (AMA-1) representing the 3D7 allele formulated with either the AS01B or AS02A Adjuvant Systems. METHODOLOGY/PRINCIPAL FINDINGS: After a preliminary safety evaluation of low dose AMA-1/AS01B (10 microg/0.5 mL) in 5 adults, 30 malaria-naïve adults were randomly allocated to receive full dose (50 microg/0.5 mL) of AMA-1/AS01B (n = 15) or AMA-1/AS02A (n = 15), followed by a malaria challenge. All vaccinations were administered intramuscularly on a 0-, 1-, 2-month schedule. All volunteers experienced transient injection site erythema, swelling and pain. Two weeks post-third vaccination, anti-AMA-1 Geometric Mean Antibody Concentrations (GMCs) with 95% Confidence Intervals (CIs) were high: low dose AMA-1/AS01B 196 microg/mL (103-371 microg/mL), full dose AMA-1/AS01B 279 microg/mL (210-369 microg/mL) and full dose AMA-1/AS02A 216 microg/mL (169-276 microg/mL) with no significant difference among the 3 groups. The three vaccine formulations elicited equivalent functional antibody responses, as measured by growth inhibition assay (GIA), against homologous but not against heterologous (FVO) parasites as well as demonstrable interferon-gamma (IFN-gamma) responses. To assess efficacy, volunteers were challenged with P. falciparum-infected mosquitoes, and all became parasitemic, with no significant difference in the prepatent period by either light microscopy or quantitative polymerase chain reaction (qPCR). However, a small but significant reduction of parasitemia in the AMA-1/AS02A group was seen with a statistical model employing qPCR measurements. SIGNIFICANCE: All three vaccine formulations were found to be safe and highly immunogenic. These immune responses did not translate into significant vaccine efficacy in malaria-naïve adults employing a primary sporozoite challenge model, but encouragingly, estimation of parasite growth rates from qPCR data may suggest a partial biological effect of the vaccine. Further evaluation of the immunogenicity and efficacy of the AMA-1/AS02A formulation is ongoing in a malaria-experienced pediatric population in Mali. TRIAL REGISTRATION: www.clinicaltrials.gov NCT00385047.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Protozoan/immunology , Lipid A/analogs & derivatives , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Membrane Proteins/immunology , Protozoan Proteins/immunology , Saponins/administration & dosage , Adjuvants, Immunologic/pharmacology , Adolescent , Adult , Alleles , Animals , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/genetics , Double-Blind Method , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Humans , Lipid A/administration & dosage , Lipid A/pharmacology , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Middle Aged , Plasmodium falciparum/immunology , Plasmodium falciparum/metabolism , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Saponins/pharmacology
19.
Vaccine ; 26(18): 2191-202, 2008 Apr 24.
Article in English | MEDLINE | ID: mdl-18387719

ABSTRACT

BACKGROUND: Immunization with RTS,S/AS02 consistently protects some vaccinees against malaria infection in experimental challenges and in field trials. A brief immunization schedule against falciparum malaria would be compatible with the Expanded Programme on Immunization, or in combination with other prevention measures, interrupt epidemic malaria or protect individuals upon sudden travel to an endemic area. METHODS: We conducted an open label, Phase 2a trial of two different full dose schedules of RTS,S/AS02 in 40 healthy malaria-naïve adults. Cohort 1 (n=20) was immunized on a 0, 1, and 3 month schedule and Cohort 2 (n=20) on a 0, 7, and 28 day schedule. Three weeks later, 38 vaccinees and 12 unimmunized infectivity controls underwent malaria challenge. RESULTS: Both regimens had a good safety and tolerability profile. Peak GMCs of antibody to the circumsporozoite protein (CSP) were similar in Cohort 1 (78 microg/mL; 95% CI: 45-134) and Cohort 2 (65 microg/mL; 95% CI: 40-104). Vaccine efficacy for Cohort 1 was 45% (95% CI: 18-62%) and for Cohort 2, 39% (95% CI: 11-56%). Protected volunteers had a higher GMC of anti-CSP antibody (114 microg/mL) than did volunteers with a 2-day delay (70 microg/mL) or no delay (30 microg/mL) in the time to onset of parasitemia (Kruskal-Wallis, p=0.019). A trend was seen for higher CSP-specific IFN-gamma responses in PBMC from protected volunteers only in Cohort 1, but not in Cohort 2, for ex vivo and for cultured ELISPOT assays. CONCLUSION: In malaria-naïve adults, the efficacy of three-dose RTS,S/AS02 regimens on either a 0, 1, and 3 month schedule or an abbreviated 0, 7, and 28 day schedule was not discernibly different from two previously reported trials of two-dose regimens given at 0, 1 month that conferred 47% (95% CI: -19 to 76%) protection and in another trial 42% (95% CI: 5-63%). A strong association of CSP-specific antibody with protection against malaria challenge is observed and confirms similar observations made in other studies. Subsequent trials of adjuvanted RTS,S in African children and infants on a 0, 1, and 2 month schedule have demonstrated a favorable safety and efficacy profile.


Subject(s)
Immunization Schedule , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Adolescent , Adult , Antibodies, Protozoan/blood , Cells, Cultured , Female , Humans , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Male , Middle Aged , Parasitemia/prevention & control , Protozoan Proteins/immunology
20.
Clin Infect Dis ; 46(1): 101-2, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18171221

ABSTRACT

Persistence of vaccinia at vaccination sites may help determine the risk associated with secondary transmission. Culture, PCR, and antigen detection were performed on serial vaccination site swab specimens. On day 21 after vaccination, 37% of volunteers were culture positive, most of whom had received vaccine for the first time. Vaccinia is detectable at least through day 21 after vaccination.


Subject(s)
Smallpox Vaccine/pharmacokinetics , Smallpox/prevention & control , Vaccinia virus/isolation & purification , Humans , Prospective Studies , Smallpox/virology , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/adverse effects , Vaccinia virus/immunology , Vaccinia virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...