1
|
Yoo R, Jore MM, Julien J. Targeting Bottlenecks in Malaria Transmission: Antibody-Epitope Descriptions Guide the Design of Next-Generation Biomedical Interventions. Immunol Rev 2025; 330:e70001. [PMID: 39907429 PMCID: PMC11796336 DOI: 10.1111/imr.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Malaria continues to pose a significant burden to global health. Thus, a strong need exists for the development of a diverse panel of intervention strategies and modalities to combat malaria and achieve elimination and eradication goals. Deploying interventions that target bottlenecks in the transmission life cycle of the causative agent of malaria, Plasmodium parasites, is an attractive strategy. The development of highly potent antibody-based biologics, including vaccines, can be greatly facilitated by an in-depth molecular understanding of antibody-epitope interactions. Here, we provide an overview of structurally characterized antibodies targeting lead vaccine candidates expressed during the bottlenecks of the Plasmodium life cycle which include the pre-erythrocytic and sexual stages. The repeat region of the circumsporozoite protein (CSP), domain 1 of Pfs230 and domains 1 and 3 of Pfs48/45 are critical Plasmodium regions targeted by the most potent antibodies at the two bottlenecks of transmission, with other promising targets emerging and requiring further characterization.
Collapse
Affiliation(s)
- Randy Yoo
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
| | - Matthijs M. Jore
- Department of Medical MicrobiologyRadboudumcNijmegenThe Netherlands
| | - Jean‐Philippe Julien
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
2
|
El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success-lessons learned and future prospects. Trop Med Health 2023; 51:29. [PMID: 37198702 DOI: 10.1186/s41182-023-00516-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The world has made great strides towards beating malaria, although about half of the world population is still exposed to the risk of contracting malaria. Developing an effective malaria vaccine was a huge challenge for medical science. In 2021 the World Health Organization (WHO) approved the first malaria vaccine, RTS,S/AS01 vaccine (Mosquirix™), for widespread use. This review highlights the history of development, and the different approaches and types of malaria vaccines, and the literature to date. It covers the developmental stages of RTS,S/AS01 and recommends steps for its deployment. The review explores other potential vaccine candidates and their status, and suggests options for their further development. It also recommends future roles for vaccines in eradicating malaria. Questions remain on how RTS,S vaccine will work in widespread use and how it can best be utilized to benefit vulnerable communities. CONCLUSION Malaria vaccines have been in development for almost 60 years. The RTS,S/AS01 vaccine has now been approved, but cannot be a stand-alone solution. Development should continue on promising candidates such as R21, PfSPZ and P. vivax vaccines. Multi-component vaccines may be a useful addition to other malaria control techniques in achieving eradication of malaria.
Collapse
Affiliation(s)
- Amal A El-Moamly
- Department of Medical Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mohamed A El-Sweify
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
3
|
Schneider CG, Fey J, Zou X, Gerbasi V, Savransky T, Batt C, Bergmann-Leitner E, Angov E. Norovirus-VLPs expressing pre-erythrocytic malaria antigens induce functional immunity against sporozoite infection. Vaccine 2022; 40:4270-4280. [PMID: 35697572 DOI: 10.1016/j.vaccine.2022.05.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/27/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
Despite the development of prophylactic anti-malarial drugs and practices to prevent infection, malaria remains a health concern. Preclinical testing of novel malaria vaccine strategies achieved through rational antigen selection and novel particle-based delivery platforms is yielding encouraging results. One such platform, self-assembling virus-like particles (VLP) is safer than attenuated live viruses, and has been approved as a vaccination tool by the FDA. We explore the use of Norovirus sub-viral particles lacking the natural shell (S) domain forming the interior shell but that retain the protruding (P) structures of the native virus as a vaccine vector. Epitope selection and their surface display has the potential to focus antigen specific immune responses to crucial epitopes. Recombinant P-particles displaying epitopes from two malaria antigens, Plasmodium falciparum (Pf) CelTOS and Plasmodium falciparum (Pf) CSP, were evaluated for immunogenicity and their ability to confer protection in a murine challenge model. Immune responses induced in mice resulted either in sterile protection (displaying PfCelTOS epitopes) or in antibodies with functional activity against sporozoites (displaying PfCSP epitopes) in an in vitro liver-stage development assay (ILSDA). These results are encouraging and support further evaluation of this platform as a vaccine delivery system.
Collapse
Affiliation(s)
- Cosette G Schneider
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA.
| | - Julien Fey
- Agave BioSystems, Ithaca, NY 14850, USA.
| | - Xiaoyan Zou
- Naval Medical Research Center, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Vince Gerbasi
- Naval Medical Research Center, Silver Spring, MD 20910, USA.
| | - Tatyana Savransky
- Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; General Dynamics Information Technology, Falls Church, VA 22042, USA.
| | - Carl Batt
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA.
| | - Elke Bergmann-Leitner
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| | - Evelina Angov
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
4
|
Sherrard-Smith E, Ngufor C, Sanou A, Guelbeogo MW, N'Guessan R, Elobolobo E, Saute F, Varela K, Chaccour CJ, Zulliger R, Wagman J, Robertson ML, Rowland M, Donnelly MJ, Gonahasa S, Staedke SG, Kolaczinski J, Churcher TS. Inferring the epidemiological benefit of indoor vector control interventions against malaria from mosquito data. Nat Commun 2022; 13:3862. [PMID: 35790746 PMCID: PMC9256631 DOI: 10.1038/s41467-022-30700-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/11/2022] [Indexed: 12/03/2022] Open
Abstract
The cause of malaria transmission has been known for over a century but it is still unclear whether entomological measures are sufficiently reliable to inform policy decisions in human health. Decision-making on the effectiveness of new insecticide-treated nets (ITNs) and the indoor residual spraying of insecticide (IRS) have been based on epidemiological data, typically collected in cluster-randomised control trials. The number of these trials that can be conducted is limited. Here we use a systematic review to highlight that efficacy estimates of the same intervention may vary substantially between trials. Analyses indicate that mosquito data collected in experimental hut trials can be used to parameterize mechanistic models for Plasmodium falciparum malaria and reliably predict the epidemiological efficacy of quick-acting, neuro-acting ITNs and IRS. Results suggest that for certain types of ITNs and IRS using this framework instead of clinical endpoints could support policy and expedite the widespread use of novel technologies.
Collapse
Affiliation(s)
| | - Corine Ngufor
- Centre de Recherches Entomologiques de Cotonou, Cotonou, Benin
- London School of Hygiene and Tropical Medicine, London, UK
| | - Antoine Sanou
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Moussa W Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Raphael N'Guessan
- London School of Hygiene and Tropical Medicine, London, UK
- Institut Pierre Richet, Bouake, Côte d'Ivoire
| | - Eldo Elobolobo
- Centro de Investigação em Saúde de Manhiça, Manhiça, Mozambique
| | - Francisco Saute
- Centro de Investigação em Saúde de Manhiça, Manhiça, Mozambique
| | | | | | - Rose Zulliger
- US President's Malaria Initiative, USAID, Washington, DC, USA
| | | | | | - Mark Rowland
- London School of Hygiene and Tropical Medicine, London, UK
| | | | | | | | | | | |
Collapse
|
5
|
Hasegawa RB, Small DS. Estimating Malaria Vaccine Efficacy in the Absence of a Gold Standard Case Definition: Mendelian Factorial Design. J Am Stat Assoc 2022. [DOI: 10.1080/01621459.2020.1863222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Raiden B. Hasegawa
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA
| | - Dylan S. Small
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
6
|
Galactionova K, Smith TA, Penny MA. Insights from modelling malaria vaccines for policy decisions: the focus on RTS,S. Malar J 2021; 20:439. [PMID: 34794430 PMCID: PMC8600337 DOI: 10.1186/s12936-021-03973-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 11/04/2021] [Indexed: 11/17/2022] Open
Abstract
Mathematical models are increasingly used to inform decisions throughout product development pathways from pre-clinical studies to country implementation of novel health interventions. This review illustrates the utility of simulation approaches by reviewing the literature on malaria vaccine modelling, with a focus on its link to the development of policy guidance for the first licensed product, RTS,S/AS01. The main contributions of modelling studies have been in inferring the mechanism of action and efficacy profile of RTS,S; to predicting the public health impact; and economic modelling mainly comprising cost-effectiveness analysis. The value of both product-specific and generic modelling of vaccines is highlighted.
Collapse
Affiliation(s)
- Katya Galactionova
- Swiss Tropical and Public Health Institute, 4051, Basel, Switzerland.,University of Basel, 4001, Basel, Switzerland.,European Center of Pharmaceutical Medicine, Brombacherstrasse 5, 4057, Basel, Switzerland
| | - Thomas A Smith
- Swiss Tropical and Public Health Institute, 4051, Basel, Switzerland. .,University of Basel, 4001, Basel, Switzerland.
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, 4051, Basel, Switzerland.,University of Basel, 4001, Basel, Switzerland
| |
Collapse
|
7
|
Duffy PE. Transmission-Blocking Vaccines: Harnessing Herd Immunity for Malaria Elimination. Expert Rev Vaccines 2021; 20:185-198. [PMID: 33478283 PMCID: PMC11127254 DOI: 10.1080/14760584.2021.1878028] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Transmission-blocking vaccines (TBV) prevent community spread of malaria by targeting mosquito sexual stage parasites, a life-cycle bottleneck, and will be used in elimination programs. TBV rely on herd immunity to reduce mosquito infections and thereby new infections in both vaccine recipients and non-recipients, but do not provide protection once an individual receives an infectious mosquito bite which complicates clinical development. AREAS COVERED Here, we describe the concept and biology behind TBV, and we provide an update on clinical development of the leading vaccine candidate antigens. Search terms 'malaria vaccine,' 'sexual stages,' 'transmission blocking vaccine,' 'VIMT' and 'SSM-VIMT' were used for PubMed queries to identify relevant literature. EXPERT OPINION Candidates targeting P. falciparum zygote surface antigen Pfs25, and its P. vivax orthologue Pvs25, induced functional activity in humans that reduced mosquito infection in surrogate assays, but require increased durability to be useful in the field. Candidates targeting gamete surface antigens Pfs230 and Pfs48/45, respectively, are in or nearing clinical trials. Nanoparticle platforms and adjuvants are being explored to enhance immunogenicity. Efficacy trials require special considerations, such as cluster-randomized designs to measure herd immunity that reduces human and mosquito infection rates, while addressing human and mosquito movements as confounding factors.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Shahzamal M, Mans B, de Hoog F, Paini D, Jurdak R. Vaccination strategies on dynamic networks with indirect transmission links and limited contact information. PLoS One 2020; 15:e0241612. [PMID: 33180786 PMCID: PMC7660487 DOI: 10.1371/journal.pone.0241612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/18/2020] [Indexed: 12/04/2022] Open
Abstract
Infectious diseases are still a major global burden for modern society causing 13 million deaths annually. One way to reduce the morbidity and mortality rates from infectious diseases is through pre-emptive or targeted vaccinations. Current theoretical vaccination strategies based on contact networks, however, rely on highly specific individual contact information which is difficult and costly to obtain, in order to identify influential spreading individuals. Current approaches also focus only on direct contacts between individuals for spreading, and disregard indirect transmission where a pathogen can spread between one infected individual and one susceptible individual who visit the same location within a short time-frame without meeting. This paper presents a novel vaccination strategy which relies on coarse-grained contact information, both direct and indirect, that can be easily and efficiently collected. Rather than tracking exact contact degrees of individuals, our strategy uses the types of places people visit to estimate a range of contact degrees for individuals, considering both direct and indirect contacts. We conduct extensive computer simulations to evaluate the performance of our strategy in comparison to state-of-the-art vaccination strategies. Results show that, when considering indirect links, our lower cost vaccination strategy achieves comparable performance to the contact-degree based approach and outperforms other existing strategies without requiring over-detailed information.
Collapse
Affiliation(s)
- Md Shahzamal
- Data61, CSIRO, Brisbane, Australia
- Macquarie University, Sydney, Australia
- * E-mail:
| | | | | | - Dean Paini
- Health and Biosecurity, CSIRO, Canberra, Australia
| | - Raja Jurdak
- Data61, CSIRO, Brisbane, Australia
- Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
9
|
Penny MA, Camponovo F, Chitnis N, Smith TA, Tanner M. Future use-cases of vaccines in malaria control and elimination. Parasite Epidemiol Control 2020; 10:e00145. [PMID: 32435704 PMCID: PMC7229487 DOI: 10.1016/j.parepi.2020.e00145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 02/18/2020] [Accepted: 03/29/2020] [Indexed: 11/29/2022] Open
Abstract
Malaria burden has significantly changed or decreased over the last 20 years, however, it remains an important health problem requiring the rigorous application of existing tools and approaches, as well as the development and use of new interventions. A malaria vaccine has long been considered a possible new intervention to aid malaria burden reduction. However, after decades of development, only one vaccine to protect children has completed phase 3 studies. Before being widely recommended for use, it must further demonstrate safety, impact and feasibility in ongoing pilot implementation studies. Now is an appropriate time to consider the use-cases and health targets of future malaria vaccines. These must be considered in the context of likely innovations in other malaria tools such as vector control, as well as the significant knowledge gaps on the appropriate target antigens, and the immunology of vaccine-induced protection. Here we discuss the history of malaria vaccines and suggest some future use-cases for future malaria vaccines that will support achieving malaria health goals in different settings.
Collapse
Affiliation(s)
| | - Flavia Camponovo
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Nakul Chitnis
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Thomas A. Smith
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Cheung YB, Ma X, Lam KF, Milligan P. Estimation of the primary, secondary and composite effects of malaria vaccines using data on multiple clinical malaria episodes. Vaccine 2020; 38:4964-4969. [PMID: 32536547 DOI: 10.1016/j.vaccine.2020.05.086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/30/2020] [Accepted: 05/29/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND An effective malaria vaccine affects the risk of malaria directly, through the vaccine-induced immune response (the primary effect), and indirectly, as a consequence of reduced exposure to malaria infection and disease, leading to slower acquisition of natural immunity (the secondary effect). The beneficial primary effect may be offset by a negative secondary effect, resulting in a smaller or nil composite effect. Reports of malaria vaccine trials usually present only the composite effect. We aimed to demonstrate how the primary and secondary effects can also be estimated from trial data. METHODS We propose an enhancement to the conditional frailty model for the estimation of primary effect using data on disease episodes. We use the Andersen-Gill model to estimate the composite effect. We consider taking the ratio of the hazard ratios to estimate the secondary effect. We used directed acyclic graphs and data from a randomized trial of the RTS,S/AS02 malaria vaccine to illustrate the problems and solutions. Time-varying effects were estimated by partitioning the follow-up into four time periods. RESULTS The primary effect estimates from our proposed model were consistently stronger than the conditional frailty model in the existing literature. The primary effect of the vaccine was consistently stronger than the composite effect across all time periods. Both the primary and composite effects were stronger in the first three months, with hazard ratios (95% confidence interval) 0.62 (0.49-0.79) and 0.68 (0.54-0.84), respectively; the hazard ratios weakened over time. The secondary effect appeared mild, with hazard ratio 1.09 (1.02-1.16) in the first three months. CONCLUSIONS The proposed analytic strategy facilitates a more comprehensive interpretation of trial data on multiple disease episodes. The RTS,S/AS02 vaccine had modest primary and secondary effects that waned over time, but the composite effect in preventing clinical malaria remained positive up to the end of the study. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT00197041.
Collapse
Affiliation(s)
- Yin Bun Cheung
- Programme in Health Services & Systems Research, Duke-NUS Medical School, 20 College Road, Singapore 169856, Singapore; Centre for Quantitative Medicine, Duke-NUS Medical School, 20 College Road, Singapore 169856, Singapore; Center for Child Health Research, University of Tampere and Tampere University Hospital, Arvo Ylpön katu 34, Tampere 33520, Finland.
| | - Xiangmei Ma
- Centre for Quantitative Medicine, Duke-NUS Medical School, 20 College Road, Singapore 169856, Singapore
| | - K F Lam
- Centre for Quantitative Medicine, Duke-NUS Medical School, 20 College Road, Singapore 169856, Singapore; Department of Statistics and Actuarial Science, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
11
|
Camponovo F, Ockenhouse CF, Lee C, Penny MA. Mass campaigns combining antimalarial drugs and anti-infective vaccines as seasonal interventions for malaria control, elimination and prevention of resurgence: a modelling study. BMC Infect Dis 2019; 19:920. [PMID: 31664924 PMCID: PMC6820916 DOI: 10.1186/s12879-019-4467-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/12/2019] [Indexed: 11/25/2022] Open
Abstract
Background The only licensed malaria vaccine, RTS,S/AS01, has been developed for morbidity-control in young children. The potential impact on transmission of deploying such anti-infective vaccines to wider age ranges, possibly with co-administration of antimalarial treatment, is unknown. Combinations of existing malaria interventions is becoming increasingly important as evidence mounts that progress on reducing malaria incidence is stalling and threatened by resistance. Methods Malaria transmission and intervention dynamics were simulated using OpenMalaria, an individual-based simulation model of malaria transmission, by considering a seasonal transmission setting and by varying epidemiological and setting parameters such as transmission intensity, case management, intervention types and intervention coverages. Chemopreventive drugs and anti-infective vaccine efficacy profiles were based on previous studies in which model parameters were fitted to clinical trial data. These intervention properties were used to evaluate the potential of seasonal mass applications of preventative anti-infective malaria vaccines, alone or in combination with chemoprevention, to reduce malaria transmission, prevent resurgence, and/or reach transmission interruption. Results Deploying a vaccine to all ages on its own is a less effective intervention strategy compared to chemoprevention alone. However, vaccines combined with drugs are likely to achieve dramatic prevalence reductions and in few settings, transmission interruption. The combined mass intervention will result in lower prevalence following the intervention compared to chemoprevention alone and will increase chances of interruption of transmission resulting from a synergistic effect between both interventions. The combination of vaccine and drug increases the time before transmission resurges after mass interventions cease compared to mass treatment alone. Deploying vaccines and drugs together requires fewer rounds of mass intervention and fewer years of intervention to achieve the same public health impact as chemoprevention alone. Conclusions Through simulations we identified a previously unidentified value of deploying vaccines with drugs, namely the greatest benefit will be in preventing and delaying transmission resurgence for longer periods than with other human targeted interventions. This is suggesting a potential role for deploying vaccines alongside drugs in transmission foci as part of surveillance-response strategies.
Collapse
Affiliation(s)
- Flavia Camponovo
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Cynthia Lee
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
12
|
Pasin C, Halloran ME, Gilbert PB, Langevin E, Ochiai RL, Pitisuttithum P, Capeding MR, Carrasquilla G, Frago C, Cortés M, Chambonneau L, Moodie Z. Periods of high dengue transmission defined by rainfall do not impact efficacy of dengue vaccine in regions of endemic disease. PLoS One 2018; 13:e0207878. [PMID: 30543657 PMCID: PMC6292612 DOI: 10.1371/journal.pone.0207878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To evaluate the association of rainy season with overall dengue disease incidence and with the efficacy of the Sanofi Pasteur recombinant, live, attenuated, tetravalent vaccine (CYD-TDV) in two randomized, controlled multicenter phase III clinical trials in Asia and Latin America. METHODS Rainy seasons were defined for each study site using climatological information from the World Meteorological Organization. The dengue attack rate in the placebo group for each study month was calculated as the number of symptomatic, virologically-confirmed dengue events in a given month divided by the number of participants at risk in the same month. Time-dependent Cox proportional hazard models were used to test whether rainy season was associated with dengue disease and whether it modified vaccine efficacy in each of the two trials and in both of the trials combined. FINDINGS Rainy season, country, and age were all significantly associated with dengue disease in both studies. Vaccine efficacy did not change during the rainy season in any of the analyses. CONCLUSIONS Although dengue transmission and exposure are expected to increase during the rainy season, our results indicate that CYD-TDV vaccine efficacy remains constant throughout the year in endemic regions.
Collapse
Affiliation(s)
- Chloé Pasin
- Université de Bordeaux, INSERM U1219 Bordeaux Population Health center, INRIA SISTM, Bordeaux, France
- Vaccine Research Institute, Creteil, France
- ENS Cachan, Université Paris-Saclay, Cachan, France
| | - M. Elizabeth Halloran
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Peter B. Gilbert
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | | | | | - Punnee Pitisuttithum
- Vaccine Trial Centre and Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Nakorn Pratum, Thailand
| | | | | | | | | | | | - Zoe Moodie
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
13
|
Ogwang R, Akena G, Yeka A, Osier F, Idro R. The 2015-2016 malaria epidemic in Northern Uganda; What are the implications for malaria control interventions? Acta Trop 2018; 188:27-33. [PMID: 30145260 PMCID: PMC7116666 DOI: 10.1016/j.actatropica.2018.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 08/17/2018] [Accepted: 08/17/2018] [Indexed: 02/03/2023]
Abstract
Vector control and effective case management are currently the backbone strategies of malaria control. Kitgum district, an area of perennial holoendemic malaria transmission intensity in Northern Uganda, appears to have experienced a malaria epidemic in 2015. This study aimed to describe the malaria trends in Kitgum General Hospital from 2011 to 2017 in relation to climatic factors and the application of population-based malaria control interventions. Hospital records were examined retrospectively to calculate malaria normal channels, malaria cases per 1000 population, test positivity rates (TPR) and to enumerate pregnancy malaria, hospitalizations and deaths. Climatic factors (humidity, temperature and rainfall) and population-based malaria control interventions that had been applied during this period were described. Kitgum district experienced an epidemic between the years 2015 and 2016; the malaria burden rose above the established normal channels. At its peak the number of malaria cases attending KGH was over 20 times above the normal channels. The total number of cases per 1000 population increased from 7 in 2014 to 113 in 2015 and 114 in 2016 (p value for trend <0.0001). Similarly, TPR increased from 10.5% to 54.6% between 2014 and 2016 (p value for trend <0.0001). This trend was also observed for malaria attributable hospitalizations, and malaria in pregnancy. There were no significant changes in any of the climatic factors assessed (p value = 0.92, 0.99, 0.52 for relative humidity, max temperature, and rainfall, respectively). The malaria upsurge occurred in conjunction with a general decline in the use and application of malaria control interventions. Specifically, indoor residual spraying was interrupted in 2014. In response to the epidemic, IRS was reapplied together with mass distribution of long-lasting insecticide treated nets (LLINs) in 2017. Subsequently, there was a decline in all malaria indicators. The epidemic in Kitgum occurred in association with the interruption of IRS and appears to have abated following its re-introduction alongside LLINs. The study suggests that to enable malaria elimination in areas of high malaria transmission intensity, effective control measures may need to be sustained for the long-term.
Collapse
Affiliation(s)
- Rodney Ogwang
- Makerere University College of Health Sciences, Kampala, Uganda; Centre of Tropical Neuroscience, Kitgum Site, Uganda
| | | | - Adoke Yeka
- Makerere University College of Health Sciences, Kampala, Uganda
| | - Faith Osier
- Centre for Infectious Diseases, Parasitology Heidelberg University Hospital, Heidelberg, Germany; KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Richard Idro
- Makerere University College of Health Sciences, Kampala, Uganda; Centre of Tropical Neuroscience, Kitgum Site, Uganda; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
14
|
Tailoring a Plasmodium vivax Vaccine To Enhance Efficacy through a Combination of a CSP Virus-Like Particle and TRAP Viral Vectors. Infect Immun 2018; 86:IAI.00114-18. [PMID: 29986894 PMCID: PMC6105880 DOI: 10.1128/iai.00114-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
Vivax malaria remains one of the most serious and neglected tropical diseases, with 132 to 391 million clinical cases per year and 2.5 billion people at risk of infection. A vaccine against Plasmodium vivax could have more impact than any other intervention, and the use of a vaccine targeting multiple antigens may result in higher efficacy against sporozoite infection than targeting a single antigen. Vivax malaria remains one of the most serious and neglected tropical diseases, with 132 to 391 million clinical cases per year and 2.5 billion people at risk of infection. A vaccine against Plasmodium vivax could have more impact than any other intervention, and the use of a vaccine targeting multiple antigens may result in higher efficacy against sporozoite infection than targeting a single antigen. Here, two leading P. vivax preerythrocytic vaccine candidate antigens, the P. vivax circumsporozoite protein (PvCSP) and the thrombospondin-related adhesion protein (PvTRAP) were delivered as a combined vaccine. This strategy provided a dose-sparing effect, with 100% sterile protection in mice using doses that individually conferred low or no protection, as with the unadjuvanted antigens PvTRAP (0%) and PvCSP (50%), and reached protection similar to that of adjuvanted components. Efficacy against malaria infection was assessed using a new mouse challenge model consisting of a double-transgenic Plasmodium berghei parasite simultaneously expressing PvCSP and PvTRAP used in mice immunized with the virus-like particle (VLP) Rv21 previously reported to induce high efficacy in mice using Matrix-M adjuvant, while PvTRAP was concomitantly administered in chimpanzee adenovirus and modified vaccinia virus Ankara (MVA) vectors (viral-vectored TRAP, or vvTRAP) to support effective induction of T cells. We examined immunity elicited by these vaccines in the context of two adjuvants approved for human use (AddaVax and Matrix-M). Matrix-M supported the highest anti-PvCSP antibody titers when combined with Rv21, and, interestingly, mixing PvCSP Rv21 and PvTRAP viral vectors enhanced immunity to malaria over levels provided by single vaccines.
Collapse
|
15
|
Smith NR, Trauer JM, Gambhir M, Richards JS, Maude RJ, Keith JM, Flegg JA. Agent-based models of malaria transmission: a systematic review. Malar J 2018; 17:299. [PMID: 30119664 PMCID: PMC6098619 DOI: 10.1186/s12936-018-2442-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/04/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Much of the extensive research regarding transmission of malaria is underpinned by mathematical modelling. Compartmental models, which focus on interactions and transitions between population strata, have been a mainstay of such modelling for more than a century. However, modellers are increasingly adopting agent-based approaches, which model hosts, vectors and/or their interactions on an individual level. One reason for the increasing popularity of such models is their potential to provide enhanced realism by allowing system-level behaviours to emerge as a consequence of accumulated individual-level interactions, as occurs in real populations. METHODS A systematic review of 90 articles published between 1998 and May 2018 was performed, characterizing agent-based models (ABMs) relevant to malaria transmission. The review provides an overview of approaches used to date, determines the advantages of these approaches, and proposes ideas for progressing the field. RESULTS The rationale for ABM use over other modelling approaches centres around three points: the need to accurately represent increased stochasticity in low-transmission settings; the benefits of high-resolution spatial simulations; and heterogeneities in drug and vaccine efficacies due to individual patient characteristics. The success of these approaches provides avenues for further exploration of agent-based techniques for modelling malaria transmission. Potential extensions include varying elimination strategies across spatial landscapes, extending the size of spatial models, incorporating human movement dynamics, and developing increasingly comprehensive parameter estimation and optimization techniques. CONCLUSION Collectively, the literature covers an extensive array of topics, including the full spectrum of transmission and intervention regimes. Bringing these elements together under a common framework may enhance knowledge of, and guide policies towards, malaria elimination. However, because of the diversity of available models, endorsing a standardized approach to ABM implementation may not be possible. Instead it is recommended that model frameworks be contextually appropriate and sufficiently described. One key recommendation is to develop enhanced parameter estimation and optimization techniques. Extensions of current techniques will provide the robust results required to enhance current elimination efforts.
Collapse
Affiliation(s)
- Neal R Smith
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| | - James M Trauer
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Manoj Gambhir
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- IBM Research Australia, Melbourne, Australia
| | - Jack S Richards
- Life Sciences, Burnet Institute, Melbourne, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
- Department of Infectious Diseases, Monash University, Melbourne, Australia
| | - Richard J Maude
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Harvard TH Chan School of Public Health, Harvard University, Boston, USA
| | - Jonathan M Keith
- School of Mathematical Sciences, Monash University, Clayton, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, University of Melbourne, Parkville, Australia
| |
Collapse
|
16
|
Collins KA, Snaith R, Cottingham MG, Gilbert SC, Hill AVS. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci Rep 2017; 7:46621. [PMID: 28422178 PMCID: PMC5395940 DOI: 10.1038/srep46621] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
The leading malaria vaccine in development is the circumsporozoite protein (CSP)-based particle vaccine, RTS,S, which targets the pre-erythrocytic stage of Plasmodium falciparum infection. It induces modest levels of protective efficacy, thought to be mediated primarily by CSP-specific antibodies. We aimed to enhance vaccine efficacy by generating a more immunogenic CSP-based particle vaccine and therefore developed a next-generation RTS,S-like vaccine, called R21. The major improvement is that in contrast to RTS,S, R21 particles are formed from a single CSP-hepatitis B surface antigen (HBsAg) fusion protein, and this leads to a vaccine composed of a much higher proportion of CSP than in RTS,S. We demonstrate that in BALB/c mice R21 is immunogenic at very low doses and when administered with the adjuvants Abisco-100 and Matrix-M it elicits sterile protection against transgenic sporozoite challenge. Concurrent induction of potent cellular and humoral immune responses was also achieved by combining R21 with TRAP-based viral vectors and protective efficacy was significantly enhanced. In addition, in contrast to RTS,S, only a minimal antibody response to the HBsAg carrier was induced. These studies identify an anti-sporozoite vaccine component that may improve upon the current leading malaria vaccine RTS,S. R21 is now under evaluation in Phase 1/2a clinical trials.
Collapse
Affiliation(s)
- Katharine A. Collins
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Rebecca Snaith
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Matthew G. Cottingham
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Sarah C. Gilbert
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Adrian V. S. Hill
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
17
|
Smith TA, Chitnis N, Penny M, Tanner M. Malaria Modeling in the Era of Eradication. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a025460. [PMID: 28003280 DOI: 10.1101/cshperspect.a025460] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mathematical models provide the essential basis of rational research and development strategies in malaria, informing the choice of which technologies to target, which deployment strategies to consider, and which populations to focus on. The Internet and remote sensing technologies also enable assembly of ever more relevant field data. Together with supercomputing technology, this has made available timely descriptions of the geography of malaria transmission and disease across the world and made it possible for policy and planning to be informed by detailed simulations of the potential impact of intervention programs. These information technology advances do not replace the basic understanding of the dynamics of malaria transmission that should be embedded in the thinking of anyone planning malaria interventions. The appropriate use of modeling may determine whether we are living in an era of hubris or indeed in an age of eradication.
Collapse
Affiliation(s)
- Thomas A Smith
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel CH 4002, Switzerland; and University of Basel, Basel CH 4001, Switzerland
| | - Nakul Chitnis
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel CH 4002, Switzerland; and University of Basel, Basel CH 4001, Switzerland
| | - Melissa Penny
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel CH 4002, Switzerland; and University of Basel, Basel CH 4001, Switzerland
| | - Marcel Tanner
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel CH 4002, Switzerland; and University of Basel, Basel CH 4001, Switzerland
| |
Collapse
|
18
|
Abstract
This article attempts to draw together current knowledge on the biology of Plasmodium and experience gained from past control campaigns to interpret and guide current efforts to discover and develop exciting new strategies targeting the parasite with the objective of interrupting transmission. Particular note is made of the advantages of targeting often unappreciated small, yet vital, bottleneck populations to enhance both the impact and the useful lifetime of hard-won interventions. A case is made for the standardization of methods to measure transmission blockade to permit the rational comparison of how diverse interventions (drugs, vaccines, insecticides, Genetically Modified technologies) targeting disparate aspects of parasite biology may impact upon the commonly used parameter of parasite prevalence in the human population.
Collapse
Affiliation(s)
- R E Sinden
- The Jenner Institute, Oxford, United Kingdom.
| |
Collapse
|
19
|
Stuckey EM, Miller JM, Littrell M, Chitnis N, Steketee R. Operational strategies of anti-malarial drug campaigns for malaria elimination in Zambia's southern province: a simulation study. Malar J 2016; 15:148. [PMID: 26957364 PMCID: PMC4784285 DOI: 10.1186/s12936-016-1202-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/01/2016] [Indexed: 01/28/2023] Open
Abstract
Background Malaria elimination requires reducing both the potential of mosquitoes to transmit parasites to humans and humans to transmit parasites to mosquitoes. To achieve this goal in Southern province, Zambia a mass test and treat (MTAT) campaign was conducted from 2011–2013 to complement high coverage of long-lasting insecticide-treated nets (LLIN). To identify factors likely to increase campaign effectiveness, a modelling approach was applied to investigate the simulated effect of alternative operational strategies for parasite clearance in southern province. Methods OpenMalaria, a discrete-time, individual-based stochastic model of malaria, was parameterized for the study area to simulate anti-malarial drug administration for interruption of transmission. Simulations were run for scenarios with a range of artemisinin-combination therapies, proportion of the population reached by the campaign, targeted age groups, time between campaign rounds, Plasmodium falciparum test protocols, and the addition of drugs aimed at preventing onward transmission. A sensitivity analysis was conducted to assess uncertainty of simulation results. Scenarios were evaluated based on the reduction in all-age parasite prevalence during the peak transmission month one year following the campaign, compared to the currently-implemented strategy of MTAT 19 % population coverage at pilot and 40 % coverage during the first year of implementation in the presence of 56 % LLIN use and 18 % indoor residual spray coverage. Results Simulation results suggest the most important determinant of success in reducing prevalence is the population coverage achieved in the campaign, which would require more than 1 year of campaign implementation for elimination. The inclusion of single low-dose primaquine, which acts as a gametocytocide, or ivermectin, which acts as an endectocide, to the drug regimen did not further reduce parasite prevalence one year following the campaign compared to the currently-implemented strategy. Simulation results indicate a high proportion of low-density infections were missed by rapid diagnostic tests that would be treated and cleared with mass drug administration (MDA). Conclusions The optimal implementation strategy for MTAT or MDA will vary by background level of prevalence, by rate of infections imported to the area, and by ability to operationally achieve high population coverage. Overall success with new parasite clearance strategies depends on continued coverage of vector control interventions to ensure sustained gains in reduction of disease burden.
Collapse
Affiliation(s)
- Erin M Stuckey
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland. .,Bill & Melinda Gates Foundation, Seattle, WA, USA.
| | | | | | - Nakul Chitnis
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| | | |
Collapse
|
20
|
Tailoring a Combination Preerythrocytic Malaria Vaccine. Infect Immun 2015; 84:622-34. [PMID: 26667840 PMCID: PMC4771343 DOI: 10.1128/iai.01063-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/27/2015] [Indexed: 12/20/2022] Open
Abstract
The leading malaria vaccine candidate, RTS,S, based on the Plasmodium falciparum circumsporozoite protein (CSP), will likely be the first publicly adopted malaria vaccine. However, this and other subunit vaccines, such as virus-vectored thrombospondin-related adhesive protein (TRAP), provide only intermediate to low levels of protection. In this study, the Plasmodium berghei homologues of antigens CSP and TRAP are combined. TRAP is delivered using adenovirus- and vaccinia virus-based vectors in a prime-boost regime. Initially, CSP is also delivered using these viral vectors; however, a reduction of anti-CSP antibodies is seen when combined with virus-vectored TRAP, and the combination is no more protective than either subunit vaccine alone. Using an adenovirus-CSP prime, protein-CSP boost regime, however, increases anti-CSP antibody titers by an order of magnitude, which is maintained when combined with virus-vectored TRAP. This combination regime using protein CSP provided 100% protection in C57BL/6 mice compared to no protection using virus-vectored TRAP alone and 40% protection using adenovirus-CSP prime and protein-CSP boost alone. This suggests that a combination of CSP and TRAP subunit vaccines could enhance protection against malaria.
Collapse
|
21
|
Abstract
In 2013 there were an estimated 584,000 deaths and 198 million clinical illnesses due to malaria, the majority in sub-Saharan Africa. Vaccines would be the ideal addition to the existing armamentarium of anti-malaria tools. However, malaria is caused by parasites, and parasites are much more complex in terms of their biology than the viruses and bacteria for which we have vaccines, passing through multiple stages of development in the human host, each stage expressing hundreds of unique antigens. This complexity makes it more difficult to develop a vaccine for parasites than for viruses and bacteria, since an immune response targeting one stage may not offer protection against a later stage, because different antigens are the targets of protective immunity at different stages. Furthermore, depending on the life cycle stage and whether the parasite is extra- or intra-cellular, antibody and/or cellular immune responses provide protection. It is thus not surprising that there is no vaccine on the market for prevention of malaria, or any human parasitic infection. In fact, no vaccine for any disease with this breadth of targets and immune responses exists. In this limited review, we focus on four approaches to malaria vaccines, (1) a recombinant protein with adjuvant vaccine aimed at Plasmodium falciparum (Pf) pre-erythrocytic stages of the parasite cycle (RTS,S/AS01), (2) whole sporozoite vaccines aimed at Pf pre-erythrocytic stages (PfSPZ Vaccine and PfSPZ-CVac), (3) prime boost vaccines that include recombinant DNA, viruses and bacteria, and protein with adjuvant aimed primarily at Pf pre-erythrocytic, but also asexual erythrocytic stages, and (4) recombinant protein with adjuvant vaccines aimed at Pf and Plasmodium vivax sexual erythrocytic and mosquito stages. We recognize that we are not covering all approaches to malaria vaccine development, or most of the critically important work on development of vaccines against P. vivax, the second most important cause of malaria. Progress during the last few years has been significant, and a first generation malaria candidate vaccine, RTS,S/AS01, is under review by the European Medicines Agency (EMA) for its quality, safety and efficacy under article 58, which allows the EMA to give a scientific opinion about products intended exclusively for markets outside of the European Union. However, much work is in progress to optimize malaria vaccines in regard to magnitude and durability of protective efficacy and the financing and practicality of delivery. Thus, we are hopeful that anti-malaria vaccines will soon be important tools in the battle against malaria.
Collapse
|
22
|
Penny MA, Pemberton-Ross P, Smith TA. The time-course of protection of the RTS,S vaccine against malaria infections and clinical disease. Malar J 2015; 14:437. [PMID: 26537608 PMCID: PMC4634589 DOI: 10.1186/s12936-015-0969-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/27/2015] [Indexed: 01/08/2023] Open
Abstract
Background Recent publications have reported follow-up of the RTS,S/AS01 malaria vaccine candidate Phase III trials at 11 African sites for 32 months (or longer). This includes site- and time-specific estimates of incidence and efficacy against clinical disease with four different vaccination schedules. These data allow estimation of the time-course of protection against infection associated with two different ages of vaccination, both with and without a booster dose. Methods Using an ensemble of individual-based stochastic models, each trial cohort in the Phase III trial was simulated assuming many different hypothetical profiles for the vaccine efficacy against infection in time, for both the primary course and boosting dose and including the potential for either exponential or non-exponential decay. The underlying profile of protection was determined by Bayesian fitting of these model predictions to the site- and time-specific incidence of clinical malaria over 32 months (or longer) of follow-up. Using the same stochastic models, projections of clinical efficacy in each of the sites were modelled and compared to available observed trial data. Results The initial protection of RTS,S immediately following three doses is estimated as providing an efficacy against infection of 65 % (when immunizing infants aged 6–12 weeks old) and 91 % (immunizing children aged 5–17 months old at first vaccination). This protection decays relatively rapidly, with an approximately exponential decay for the 6–12 weeks old cohort (with a half-life of 7.2 months); for the 5–17 months old cohort a biphasic decay with a similar half-life is predicted, with an initial rapid decay followed by a slower decay. The boosting dose was estimated to return protection to an efficacy against infection of 50–55 % for both cohorts. Estimates of clinical efficacy by trial site are consistent with those reported in the trial for all cohorts. Conclusions The site- and time-specific clinical observations from the RTS,S/AS01 trial data allowed a reasonably precise estimation of the underlying vaccine protection against infection which is consistent with common underlying efficacy and decay rates across the trial sites. This calibration suggests that the decay in efficacy against clinical disease is more rapid than that against infection because of age-shifts in the incidence of disease. The dynamical models predict that clinical effectiveness will continue to decay and that likely effects beyond the time-scale of the trial will be small. Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0969-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa A Penny
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, 4051, Basel, Switzerland. .,University of Basel, Petersplatz 1, Basel, Switzerland.
| | - Peter Pemberton-Ross
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, 4051, Basel, Switzerland. .,University of Basel, Petersplatz 1, Basel, Switzerland.
| | - Thomas A Smith
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, 4051, Basel, Switzerland. .,University of Basel, Petersplatz 1, Basel, Switzerland.
| |
Collapse
|
23
|
Birkett AJ. Building an effective malaria vaccine pipeline to address global needs. Vaccine 2015; 33:7538-43. [PMID: 26469721 DOI: 10.1016/j.vaccine.2015.09.111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 02/08/2023]
Abstract
Despite impressive gains over the last 15 years in reducing the mortality associated with malaria, it remains a public health emergency. New interventions, such as vaccines, are needed to ensure that previous gains serve as a foundation for future progress. Vaccines have the potential to prevent severe disease and death in those most vulnerable, and to accelerate elimination and eradication by breaking the cycle of parasite transmission. The pipeline is as healthy as it has ever been, with approaches targeting different stages of the parasite lifecycle using an array of technologies. This article reviews recent progress and reviews key considerations in the quest to develop products that are aligned with the unmet medical need.
Collapse
Affiliation(s)
- Ashley J Birkett
- PATH Malaria Vaccine Initiative (MVI), 455 Massachusetts Avenue NW, Suite 1000, Washington, DC 20001-2621, USA.
| |
Collapse
|
24
|
Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine 2015; 33 Suppl 4:D13-23. [PMID: 26324116 DOI: 10.1016/j.vaccine.2015.07.091] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/25/2015] [Accepted: 07/27/2015] [Indexed: 01/14/2023]
Abstract
In 2013 there were an estimated 584,000 deaths and 198 million clinical illnesses due to malaria, the majority in sub-Saharan Africa. Vaccines would be the ideal addition to the existing armamentarium of anti-malaria tools. However, malaria is caused by parasites, and parasites are much more complex in terms of their biology than the viruses and bacteria for which we have vaccines, passing through multiple stages of development in the human host, each stage expressing hundreds of unique antigens. This complexity makes it more difficult to develop a vaccine for parasites than for viruses and bacteria, since an immune response targeting one stage may not offer protection against a later stage, because different antigens are the targets of protective immunity at different stages. Furthermore, depending on the life cycle stage and whether the parasite is extra- or intra-cellular, antibody and/or cellular immune responses provide protection. It is thus not surprising that there is no vaccine on the market for prevention of malaria, or any human parasitic infection. In fact, no vaccine for any disease with this breadth of targets and immune responses exists. In this limited review, we focus on four approaches to malaria vaccines, (1) a recombinant protein with adjuvant vaccine aimed at Plasmodium falciparum (Pf) pre-erythrocytic stages of the parasite cycle (RTS,S/AS01), (2) whole sporozoite vaccines aimed at Pf pre-erythrocytic stages (PfSPZ Vaccine and PfSPZ-CVac), (3) prime boost vaccines that include recombinant DNA, viruses and bacteria, and protein with adjuvant aimed primarily at Pf pre-erythrocytic, but also asexual erythrocytic stages, and (4) recombinant protein with adjuvant vaccines aimed at Pf and Plasmodium vivax sexual erythrocytic and mosquito stages. We recognize that we are not covering all approaches to malaria vaccine development, or most of the critically important work on development of vaccines against P. vivax, the second most important cause of malaria. Progress during the last few years has been significant, and a first generation malaria candidate vaccine, RTS,S/AS01, is under review by the European Medicines Agency (EMA) for its quality, safety and efficacy under article 58, which allows the EMA to give a scientific opinion about products intended exclusively for markets outside of the European Union. However, much work is in progress to optimize malaria vaccines in regard to magnitude and durability of protective efficacy and the financing and practicality of delivery. Thus, we are hopeful that anti-malaria vaccines will soon be important tools in the battle against malaria.
Collapse
Affiliation(s)
| | | | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
25
|
Penny MA, Galactionova K, Tarantino M, Tanner M, Smith TA. The public health impact of malaria vaccine RTS,S in malaria endemic Africa: country-specific predictions using 18 month follow-up Phase III data and simulation models. BMC Med 2015; 13:170. [PMID: 26219380 PMCID: PMC4518512 DOI: 10.1186/s12916-015-0408-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/25/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The RTS,S/AS01 malaria vaccine candidate recently completed Phase III trials in 11 African sites. Recommendations for its deployment will partly depend on predictions of public health impact in endemic countries. Previous predictions of these used only limited information on underlying vaccine properties and have not considered country-specific contextual data. METHODS Each Phase III trial cohort was simulated explicitly using an ensemble of individual-based stochastic models, and many hypothetical vaccine profiles. The true profile was estimated by Bayesian fitting of these models to the site- and time-specific incidence of clinical malaria in both trial arms over 18 months of follow-up. Health impacts of implementation via two vaccine schedules in 43 endemic sub-Saharan African countries, using country-specific prevalence, access to care, immunisation coverage and demography data, were predicted via weighted averaging over many simulations. RESULTS The efficacy against infection of three doses of vaccine was initially approximately 65 % (when immunising 6-12 week old infants) and 80 % (children 5-17 months old), with a 1 year half-life (exponential decay). Either schedule will avert substantial disease, but predicted impact strongly depends on the decay rate of vaccine effects and average transmission intensity. CONCLUSIONS For the first time Phase III site- and time-specific data were available to estimate both the underlying profile of RTS,S/AS01 and likely country-specific health impacts. Initial efficacy will probably be high, but decay rapidly. Adding RTS,S to existing control programs, assuming continuation of current levels of malaria exposure and of health system performance, will potentially avert 100-580 malaria deaths and 45,000 to 80,000 clinical episodes per 100,000 fully vaccinated children over an initial 10-year phase.
Collapse
Affiliation(s)
- Melissa A Penny
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| | - Katya Galactionova
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| | - Michael Tarantino
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| | - Marcel Tanner
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| | - Thomas A Smith
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
26
|
Pemberton-Ross P, Smith TA, Hodel EM, Kay K, Penny MA. Age-shifting in malaria incidence as a result of induced immunological deficit: a simulation study. Malar J 2015; 14:287. [PMID: 26206255 PMCID: PMC4513612 DOI: 10.1186/s12936-015-0805-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/10/2015] [Indexed: 11/10/2022] Open
Abstract
Effective population-level interventions against Plasmodium falciparum malaria lead to age-shifts, delayed morbidity or rebounds in morbidity and mortality whenever they are deployed in ways that do not permanently interrupt transmission. When long-term intervention programmes target specific age-groups of human hosts, the age-specific morbidity rates ultimately adjust to new steady-states, but it is very difficult to study these rates and the temporal dynamics leading up to them empirically because the changes occur over very long time periods. This study investigates the age and magnitude of age- and time- shifting of incidence induced by either pre-erythrocytic vaccination (PEV) programmes or seasonal malaria chemo-prevention (SMC), using an ensemble of individual-based stochastic simulation models of P. falciparum dynamics. The models made various assumptions about immunity decay, transmission heterogeneity and were parameterized with data on both age-specific infection and disease incidence at different levels of exposure, on the durations of different stages of the parasite life-cycle and on human demography. Effects of transmission intensity, and of levels of access to malaria treatment were considered. While both PEV and SMC programmes are predicted to have overall strongly positive health effects, a shift of morbidity into older children is predicted to be induced by either programme if transmission levels remain static and not reduced by other interventions. Predicted shifting of burden continue into the second decade of the programme. Even if long-term surveillance is maintained it will be difficult to avoid mis-attribution of such long-term changes in age-specific morbidity patterns to other factors. Conversely, short-lived transient changes in incidence measured soon after introduction of a new intervention may give over-positive views of future impacts. Complementary intervention strategies could be designed to specifically protect those age-groups at risk from burden shift.
Collapse
Affiliation(s)
- Peter Pemberton-Ross
- Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.
- Universität Basel, 4003, Basel, Switzerland.
| | - Thomas A Smith
- Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.
- Universität Basel, 4003, Basel, Switzerland.
| | - Eva Maria Hodel
- Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK.
| | - Katherine Kay
- Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK.
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.
- Universität Basel, 4003, Basel, Switzerland.
| |
Collapse
|
27
|
Malaria vaccines: looking back and lessons learnt. Asian Pac J Trop Biomed 2015; 1:74-8. [PMID: 23569729 DOI: 10.1016/s2221-1691(11)60072-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 12/26/2010] [Accepted: 02/28/2011] [Indexed: 11/20/2022] Open
Abstract
The current status of malaria vaccine approaches has the background of a long and arduous path of malaria disease control and vaccine development. Here, we critically review with regard to unilateral interventional approaches and highlight the impact of socioeconomic elements of malaria endemicity. The necessity of re-energizing basic research of malaria life-cycle and Plasmodium developmental biology to provide the basis for promising and cost-effective vaccine approaches and to reach eradication goals is more urgent than previously believed. We closely analyse the flaws of various vaccine approaches, outline future directions and challenges that still face us and conclude that the focus of the field must be shifted to the basic research efforts including findings on the skin stage of infection. We also reflect on economic factors of vaccine development and the impact of public perception when it comes to vaccine uptake.
Collapse
|
28
|
Nunes JK, Woods C, Carter T, Raphael T, Morin MJ, Diallo D, Leboulleux D, Jain S, Loucq C, Kaslow DC, Birkett AJ. Development of a transmission-blocking malaria vaccine: progress, challenges, and the path forward. Vaccine 2014; 32:5531-9. [PMID: 25077422 DOI: 10.1016/j.vaccine.2014.07.030] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/12/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
Abstract
New interventions are needed to reduce morbidity and mortality associated with malaria, as well as to accelerate elimination and eventual eradication. Interventions that can break the cycle of parasite transmission, and prevent its reintroduction, will be of particular importance in achieving the eradication goal. In this regard, vaccines that interrupt malaria transmission (VIMT) have been highlighted as an important intervention, including transmission-blocking vaccines that prevent human-to-mosquito transmission by targeting the sexual, sporogonic, or mosquito stages of the parasite (SSM-VIMT). While the significant potential of this vaccine approach has been appreciated for decades, the development and licensure pathways for vaccines that target transmission and the incidence of infection, as opposed to prevention of clinical malaria disease, remain ill-defined. This article describes the progress made in critical areas since 2010, highlights key challenges that remain, and outlines important next steps to maximize the potential for SSM-VIMTs to contribute to the broader malaria elimination and eradication objectives.
Collapse
Affiliation(s)
- Julia K Nunes
- PATH Malaria Vaccine Initiative, Washington, DC, USA
| | - Colleen Woods
- PATH Malaria Vaccine Initiative, Washington, DC, USA; PATH Malaria Vaccine Initiative, Seattle, WA, USA
| | | | | | | | | | | | - Sanjay Jain
- PATH Malaria Vaccine Initiative, Washington, DC, USA
| | | | - David C Kaslow
- PATH Malaria Vaccine Initiative, Washington, DC, USA; PATH, Seattle, WA, USA
| | | |
Collapse
|
29
|
Birkett AJ, Moorthy VS, Loucq C, Chitnis CE, Kaslow DC. Malaria vaccine R&D in the Decade of Vaccines: breakthroughs, challenges and opportunities. Vaccine 2014; 31 Suppl 2:B233-43. [PMID: 23598488 DOI: 10.1016/j.vaccine.2013.02.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 02/06/2013] [Accepted: 02/25/2013] [Indexed: 01/23/2023]
Abstract
While recent progress has been made in reducing malaria mortality with other interventions, vaccines are still urgently needed to further reduce the incidence of clinical disease, including during pregnancy, and to provide "herd protection" by blocking parasite transmission. The most clinically advanced candidate, RTS,S, is presently undergoing Phase 3 evaluation in young African children across 13 clinical sites in eight African countries. In the 12-month period following vaccination, RTS,S conferred approximately 50% protection from clinical Plasmodium falciparum disease in children aged 5-17 months, and approximately 30% protection in children aged 6-12 weeks when administered in conjunction with Expanded Program for Immunization (EPI) vaccines. The development of more highly efficacious vaccines to prevent clinical disease caused by both P. falciparum and Plasmodium vivax, as well as vaccines to support elimination efforts by inducing immunity that blocks malaria parasite transmission, are priorities. Some key barriers to malaria vaccine development include: a paucity of well-characterized target immunogens and an absence of clear correlates of protection to enable vaccine development targeting all stages of the P. falciparum and P. vivax lifecycles; a limited number of safe and effective delivery systems, including adjuvants, that induce potent, long-lived protective immunity, be it by antibody, CD4+, and/or CD8+ T cell responses; and, for vaccines designed to provide "herd protection" by targeting sexual stage and/or mosquito antigens, the lack of a clear clinical and regulatory pathway to licensure using non-traditional endpoints. Recommendations to overcome these, and other key challenges, are suggested in this document.
Collapse
Affiliation(s)
- Ashley J Birkett
- PATH Malaria Vaccine Initiative, Washington, DC 20001-2621, USA.
| | | | | | | | | |
Collapse
|
30
|
Theisen M, Roeffen W, Singh SK, Andersen G, Amoah L, van de Vegte-Bolmer M, Arens T, Tiendrebeogo RW, Jones S, Bousema T, Adu B, Dziegiel MH, Christiansen M, Sauerwein R. A multi-stage malaria vaccine candidate targeting both transmission and asexual parasite life-cycle stages. Vaccine 2014; 32:2623-30. [DOI: 10.1016/j.vaccine.2014.03.020] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 02/28/2014] [Accepted: 03/05/2014] [Indexed: 11/27/2022]
|
31
|
Perkins TA, Scott TW, Le Menach A, Smith DL. Heterogeneity, mixing, and the spatial scales of mosquito-borne pathogen transmission. PLoS Comput Biol 2013; 9:e1003327. [PMID: 24348223 PMCID: PMC3861021 DOI: 10.1371/journal.pcbi.1003327] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 09/24/2013] [Indexed: 11/18/2022] Open
Abstract
The Ross-Macdonald model has dominated theory for mosquito-borne pathogen transmission dynamics and control for over a century. The model, like many other basic population models, makes the mathematically convenient assumption that populations are well mixed; i.e., that each mosquito is equally likely to bite any vertebrate host. This assumption raises questions about the validity and utility of current theory because it is in conflict with preponderant empirical evidence that transmission is heterogeneous. Here, we propose a new dynamic framework that is realistic enough to describe biological causes of heterogeneous transmission of mosquito-borne pathogens of humans, yet tractable enough to provide a basis for developing and improving general theory. The framework is based on the ecological context of mosquito blood meals and the fine-scale movements of individual mosquitoes and human hosts that give rise to heterogeneous transmission. Using this framework, we describe pathogen dispersion in terms of individual-level analogues of two classical quantities: vectorial capacity and the basic reproductive number, . Importantly, this framework explicitly accounts for three key components of overall heterogeneity in transmission: heterogeneous exposure, poor mixing, and finite host numbers. Using these tools, we propose two ways of characterizing the spatial scales of transmission—pathogen dispersion kernels and the evenness of mixing across scales of aggregation—and demonstrate the consequences of a model's choice of spatial scale for epidemic dynamics and for estimation of , both by a priori model formulas and by inference of the force of infection from time-series data. Pathogens transmitted by mosquitoes, such as malaria and dengue, are notorious for the biological complexity associated with how they are transmitted within local communities. Yet mathematical models for these pathogens, which are critical tools for making recommendations for control policy, are based around concepts originally designed to describe how molecules interact in chemical systems. To provide those interested in mosquito-borne diseases a more appropriate tool for modeling their transmission, we introduce a mathematical framework that is based on the spatial locations where mosquitoes lay eggs and feed on blood and how mosquitoes and hosts move about those locations. Analysis of this framework shows that the transmission contributions of different hosts and locations can be calculated, and that overall potential for transmission in a community depends on three concepts: heterogeneous exposure (some people bitten by mosquitoes more than others), poor mixing (non-random contacts between hosts and mosquitoes), and finite population sizes (each host can contribute at most one new infection towards the population total). Together, these factors determine critical levels of vaccination coverage to eliminate a pathogen and the spatial areas over which transmission should be modeled and studied in the field.
Collapse
Affiliation(s)
- T. Alex Perkins
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Entomology, University of California, Davis, California, United States of America
- * E-mail:
| | - Thomas W. Scott
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Entomology, University of California, Davis, California, United States of America
| | - Arnaud Le Menach
- Center for Disease Dynamics, Economics and Policy, Washington, D.C., United States of America
| | - David L. Smith
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Center for Disease Dynamics, Economics and Policy, Washington, D.C., United States of America
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
32
|
The role of mathematical modelling in guiding the science and economics of malaria elimination. Int Health 2013; 2:239-46. [PMID: 21270957 PMCID: PMC3004029 DOI: 10.1016/j.inhe.2010.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 07/22/2010] [Accepted: 09/13/2010] [Indexed: 11/21/2022] Open
Abstract
Unprecedented efforts are now underway to eliminate malaria from many regions. Despite the enormous financial resources committed, if malaria elimination is perceived as failing it is likely that this funding will not be sustained. It is imperative that methods are developed to use the limited data available to design site-specific, cost-effective elimination programmes. Mathematical modelling is a way of including mechanistic understanding to use available data to make predictions. Different strategies can be evaluated much more rapidly than is possible through trial and error in the field. Mathematical modelling has great potential as a tool to guide and inform current elimination efforts. Economic modelling weighs costs against characterised effects or predicted benefits in order to determine the most cost-efficient strategy but has traditionally used static models of disease not suitable for elimination. Dynamic mathematical modelling and economic modelling techniques need to be combined to contribute most effectively to ongoing policy discussions. We review the role of modelling in previous malaria control efforts as well as the unique nature of elimination and the consequent need for its explicit modelling, and emphasise the importance of good disease surveillance. The difficulties and complexities of economic evaluation of malaria control, particularly the end stages of elimination, are discussed.
Collapse
|
33
|
Nunes JK, Cárdenas V, Loucq C, Maire N, Smith T, Shaffer C, Måseide K, Brooks A. Modeling the public health impact of malaria vaccines for developers and policymakers. BMC Infect Dis 2013; 13:295. [PMID: 23815273 PMCID: PMC3711926 DOI: 10.1186/1471-2334-13-295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 06/17/2013] [Indexed: 11/15/2022] Open
Abstract
Background Efforts to develop malaria vaccines show promise. Mathematical model-based estimates of the potential demand, public health impact, and cost and financing requirements can be used to inform investment and adoption decisions by vaccine developers and policymakers on the use of malaria vaccines as complements to existing interventions. However, the complexity of such models may make their outputs inaccessible to non-modeling specialists. This paper describes a Malaria Vaccine Model (MVM) developed to address the specific needs of developers and policymakers, who need to access sophisticated modeling results and to test various scenarios in a user-friendly interface. The model’s functionality is demonstrated through a hypothetical vaccine. Methods The MVM has three modules: supply and demand forecast; public health impact; and implementation cost and financing requirements. These modules include pre-entered reference data and also allow for user-defined inputs. The model includes an integrated sensitivity analysis function. Model functionality was demonstrated by estimating the public health impact of a hypothetical pre-erythrocytic malaria vaccine with 85% efficacy against uncomplicated disease and a vaccine efficacy decay rate of four years, based on internationally-established targets. Demand for this hypothetical vaccine was estimated based on historical vaccine implementation rates for routine infant immunization in 40 African countries over a 10-year period. Assumed purchase price was $5 per dose and injection equipment and delivery costs were $0.40 per dose. Results The model projects the number of doses needed, uncomplicated and severe cases averted, deaths and disability-adjusted life years (DALYs) averted, and cost to avert each. In the demonstration scenario, based on a projected demand of 532 million doses, the MVM estimated that 150 million uncomplicated cases of malaria and 1.1 million deaths would be averted over 10 years. This is equivalent to 943 uncomplicated cases and 7 deaths averted per 1,000 vaccinees. In discounted 2011 US dollars, this represents $11 per uncomplicated case averted and $1,482 per death averted. If vaccine efficacy were reduced to 75%, the estimated uncomplicated cases and deaths averted over 10 years would decrease by 14% and 19%, respectively. Conclusions The MVM can provide valuable information to assist decision-making by vaccine developers and policymakers, information which will be refined and strengthened as field studies progress allowing further validation of modeling assumptions.
Collapse
|
34
|
Wenger EA, Eckhoff PA. A mathematical model of the impact of present and future malaria vaccines. Malar J 2013; 12:126. [PMID: 23587051 PMCID: PMC3658952 DOI: 10.1186/1475-2875-12-126] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/20/2013] [Indexed: 11/30/2022] Open
Abstract
Background With the encouraging advent of new malaria vaccine candidates, mathematical modelling of expected impacts of present and future vaccines as part of multi-intervention strategies is especially relevant. Methods The impact of potential malaria vaccines is presented utilizing the EMOD model, a comprehensive model of the vector life cycle coupled to a detailed mechanistic representation of intra-host parasite and immune dynamics. Values of baseline transmission and vector feeding behaviour parameters are identified, for which local elimination is enabled by layering pre-erythrocytic vaccines of various efficacies on top of high and sustained insecticide-treated net coverage. The expected reduction in clinical cases is further explored in a scenario that targets children by adding a pre-erythrocytic vaccine to the EPI programme for newborns. Results At high transmission, there is a minimal reduction in clinical disease cases, as the time to infection is only slightly delayed. At lower transmission, there is an accelerating community-level protection that has subtle dependences on heterogeneities in vector behaviour, ecology, and intervention coverage. At very low transmission, the trend reverses as many children are vaccinated to prevent few cases. Conclusions The maximum-impact setting is one in which the impact of increasing bed net coverage has saturated, vector feeding is primarily outdoors, and transmission is just above the threshold where small perturbations from a vaccine intervention result in large community benefits.
Collapse
|
35
|
Eckhoff P. Mathematical models of within-host and transmission dynamics to determine effects of malaria interventions in a variety of transmission settings. Am J Trop Med Hyg 2013; 88:817-827. [PMID: 23589530 PMCID: PMC3752743 DOI: 10.4269/ajtmh.12-0007] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A model for Anopheles population dynamics and malaria transmission is combined with a within-host dynamics microsolver to study baseline transmission, the effects of seasonality, and the impact of interventions. The Garki Project is recreated in simulation of the pre-intervention baseline and the different combinations of interventions deployed. Modifications are introduced, and longer project duration, extension of dry-season spraying, and transmission-blocking vaccines together achieve local elimination in some conditions. A variety of interventions are simulated in transmission settings that vary in transmission intensity and underlying seasonality. Adding vaccines to existing vector control efforts extends the ability to achieve elimination to higher baseline transmission and less favorable vector behavior. If one species of the Anopheles gambiae species complex feeds disproportionately outdoors for a given complex average behavior, vector control impacts are less than for a single species. Non-zero dry-season transmission limits seasonal oscillation in parasite dynamics and impact of wet-season interventions.
Collapse
Affiliation(s)
- Philip Eckhoff
- *Address correspondence to Philip Eckhoff, Intellectual Ventures Laboratory, 1575 132nd Avenue NE, Bellevue, WA 98004. E-mail:
| |
Collapse
|
36
|
Crowell V, Yukich JO, Briët OJT, Ross A, Smith TA. A novel approach for measuring the burden of uncomplicated Plasmodium falciparum malaria: application to data from Zambia. PLoS One 2013; 8:e57297. [PMID: 23468961 PMCID: PMC3585385 DOI: 10.1371/journal.pone.0057297] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 01/21/2013] [Indexed: 11/18/2022] Open
Abstract
Measurement of malaria burden is fraught with complexity, due to the natural history of the disease, delays in seeking treatment or failure of case management. Attempts to establish an appropriate case definition for a malaria episode has often resulted in ambiguities and challenges because of poor information about treatment seeking, patterns of infection, recurrence of fever and asymptomatic infection. While the primary reason for treating malaria is to reduce disease burden, the effects of treatment are generally ignored in estimates of the burden of malaria morbidity, which are usually presented in terms of numbers of clinical cases or episodes, with the main data sources being reports from health facilities and parasite prevalence surveys. The use of burden estimates that do not consider effects of treatment, leads to under-estimation of the impact of improvements in case management. Official estimates of burden very likely massively underestimate the impact of the roll-out of ACT as first-line therapy across Africa. This paper proposes a novel approach for estimating burden of disease based on the point prevalence of malaria attributable disease, or equivalently, the days with malaria fever in unit time. The technique makes use of data available from standard community surveys, analyses of fever patterns in malaria therapy patients, and data on recall bias. Application of this approach to data from Zambia for 2009–2010 gave an estimate of 2.6 (95% credible interval: 1.5–3.7) malaria attributable fever days per child-year. The estimates of recall bias, and of the numbers of days with illness contributing to single illness recalls, could be applied more generally. To obtain valid estimates of the overall malaria burden using these methods, there remains a need for surveys to include the whole range of ages of hosts in the population and for data on seasonality patterns in confirmed case series.
Collapse
Affiliation(s)
- Valerie Crowell
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Joshua O. Yukich
- Department of Global Health Systems and Development, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Olivier J. T. Briët
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Amanda Ross
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Thomas A. Smith
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
37
|
Eckhoff P. P. falciparum infection durations and infectiousness are shaped by antigenic variation and innate and adaptive host immunity in a mathematical model. PLoS One 2012; 7:e44950. [PMID: 23028698 PMCID: PMC3446976 DOI: 10.1371/journal.pone.0044950] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 08/14/2012] [Indexed: 11/29/2022] Open
Abstract
Many questions remain about P. falciparum within-host dynamics, immunity, and transmission–issues that may affect public health campaign planning. These gaps in knowledge concern the distribution of durations of malaria infections, determination of peak parasitemia during acute infection, the relationships among gametocytes and immune responses and infectiousness to mosquitoes, and the effect of antigenic structure on reinfection outcomes. The present model of intra-host dynamics of P. falciparum implements detailed representations of parasite and immune dynamics, with structures based on minimal extrapolations from first-principles biology in its foundations. The model is designed to quickly and readily accommodate gains in mechanistic understanding and to evaluate effects of alternative biological hypothesis through in silico experiments. Simulations follow the parasite from the liver-stage through the detailed asexual cycle to clearance while tracking gametocyte populations. The modeled immune system includes innate inflammatory and specific antibody responses to a repertoire of antigens. The mechanistic focus provides clear explanations for the structure of the distribution of infection durations through the interaction of antigenic variation and innate and adaptive immunity. Infectiousness to mosquitoes appears to be determined not only by the density of gametocytes but also by the level of inflammatory cytokines, which harmonizes an extensive series of study results. Finally, pre-existing immunity can either decrease or increase the duration of infections upon reinfection, depending on the degree of overlap in antigenic repertoires and the strength of the pre-existing immunity.
Collapse
Affiliation(s)
- Philip Eckhoff
- Intellectual Ventures, Bellevue, Washington, United States of America.
| |
Collapse
|
38
|
Smith T, Tediosi F. Simulating the impact of malaria vaccination: what has been learnt? Expert Rev Vaccines 2012; 11:751-3. [DOI: 10.1586/erv.12.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Brooks A, Briët OJT, Hardy D, Steketee R, Smith TA. Simulated impact of RTS,S/AS01 vaccination programs in the context of changing malaria transmission. PLoS One 2012; 7:e32587. [PMID: 22412892 PMCID: PMC3295753 DOI: 10.1371/journal.pone.0032587] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 02/01/2012] [Indexed: 12/29/2022] Open
Abstract
Introduction The RTS,S/AS01 pre-erythrocytic malaria vaccine is in phase III clinical trials. It is critical to anticipate where and how it should be implemented if trials are successful. Such planning may be complicated by changing levels of malaria transmission. Methods/results Computer simulations were used to examine RTS,S/AS01 impact, using a vaccine profile based on phase II trial results, and assuming that protection decays only slowly. Settings were simulated in which baseline transmission (in the absence of vaccine) was fixed or varied between 2 and 20 infectious mosquito bites per person per annum (ibpa) over ten years. Four delivery strategies were studied: routine infant immunization (EPI), EPI plus infant catch-up, EPI plus school-based campaigns, and EPI plus mass campaigns. Impacts in changing transmission settings were similar to those in fixed settings. Assuming a persistent effect of vaccination, at 2 ibpa, the vaccine averted approximately 5–7 deaths per 1000 doses of vaccine when delivered via mass campaigns, but the benefit was less at higher transmission levels. EPI, catch-up and school-based strategies averted 2–3 deaths per 1000 doses in settings with 2 ibpa. In settings where transmission was decreasing or increasing, EPI, catch-up and school-based strategies averted approximately 3–4 deaths per 1000 doses. Discussion Where transmission is changing, it appears to be sufficient to consider simulations of pre-erythrocytic vaccine impact at a range of initial transmission levels. At 2 ibpa, mass campaigns averted the most deaths and reduced transmission, but this requires further study. If delivered via EPI, RTS,S/AS01 could avert approximately 6–11 deaths per 1000 vaccinees in all examined settings, similar to estimates for pneumococcal conjugate vaccine in African infants. These results support RTS,S/AS01 implementation via EPI, for example alongside vector control interventions, providing that the phase III trials provide support for our assumptions about efficacy.
Collapse
Affiliation(s)
- Alan Brooks
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
| | | | | | | | | |
Collapse
|
40
|
Storti-Melo LM, da Costa DR, Souza-Neiras WC, Cassiano GC, Couto VSCD, Póvoa MM, Soares IDS, de Carvalho LH, Arevalo-Herrera M, Herrera S, Rossit ARB, Cordeiro JA, de Mattos LC, Machado RLD. Influence of HLA-DRB-1 alleles on the production of antibody against CSP, MSP-1, AMA-1, and DBP in Brazilian individuals naturally infected with Plasmodium vivax. Acta Trop 2012; 121:152-5. [PMID: 22107686 DOI: 10.1016/j.actatropica.2011.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 09/19/2011] [Accepted: 10/12/2011] [Indexed: 11/16/2022]
Abstract
We evaluated the influence of allelic frequency of the human leukocyte antigen (HLA) -DRB1 on the acquisition of antibody response against malaria sporozoite and merozoite peptides in patients with Plasmodium vivax malaria acquired in endemic areas of Brazil. IgG antibodies were detected by enzyme-linked immunosorbent assay against four peptides of circumsporozoite protein (CSP) (amino, carboxyl, and VK210 and VK247 repeats) and peptides of merozoite surface protein 1 (MSP-1), apical membrane antigen 1 (AMA-1), and Duffy-binding protein (DBP). We found an association between HLA-DR3 and HLA-DR5 alleles and lack of antibody response to CSP amino terminal, as well as an association between HLA-DR3 and the highest antibody response to MSP1 (Pv200L). In conclusion, we suggest a potential regulatory role of the HLA-DRB1 alleles in the production of antibodies to a conserved region of P. vivax CSP and MSP1 in Brazilian population exposed to malaria.
Collapse
Affiliation(s)
- Luciane Moreno Storti-Melo
- Departamento de Biociências, Campus Universitário Prof. Alberto Carvalho, Universidade Federal de Sergipe, Itabaiana, SE, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Importance of factors determining the effective lifetime of a mass, long-lasting, insecticidal net distribution: a sensitivity analysis. Malar J 2012; 11:20. [PMID: 22244509 PMCID: PMC3273435 DOI: 10.1186/1475-2875-11-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 01/13/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Long-lasting insecticidal nets (LLINs) reduce malaria transmission by protecting individuals from infectious bites, and by reducing mosquito survival. In recent years, millions of LLINs have been distributed across sub-Saharan Africa (SSA). Over time, LLINs decay physically and chemically and are destroyed, making repeated interventions necessary to prevent a resurgence of malaria. Because its effects on transmission are important (more so than the effects of individual protection), estimates of the lifetime of mass distribution rounds should be based on the effective length of epidemiological protection. METHODS Simulation models, parameterised using available field data, were used to analyse how the distribution's effective lifetime depends on the transmission setting and on LLIN characteristics. Factors considered were the pre-intervention transmission level, initial coverage, net attrition, and both physical and chemical decay. An ensemble of 14 stochastic individual-based model variants for malaria in humans was used, combined with a deterministic model for malaria in mosquitoes. RESULTS The effective lifetime was most sensitive to the pre-intervention transmission level, with a lifetime of almost 10 years at an entomological inoculation rate of two infectious bites per adult per annum (ibpapa), but of little more than 2 years at 256 ibpapa. The LLIN attrition rate and the insecticide decay rate were the next most important parameters. The lifetime was surprisingly insensitive to physical decay parameters, but this could change as physical integrity gains importance with the emergence and spread of pyrethroid resistance. CONCLUSIONS The strong dependency of the effective lifetime on the pre-intervention transmission level indicated that the required distribution frequency may vary more with the local entomological situation than with LLIN quality or the characteristics of the distribution system. This highlights the need for malaria monitoring both before and during intervention programmes, particularly since there are likely to be strong variations between years and over short distances. The majority of SSA's population falls into exposure categories where the lifetime is relatively long, but because exposure estimates are highly uncertain, it is necessary to consider subsequent interventions before the end of the expected effective lifetime based on an imprecise transmission measure.
Collapse
|
42
|
A periodically-forced mathematical model for the seasonal dynamics of malaria in mosquitoes. Bull Math Biol 2012; 74:1098-124. [PMID: 22218880 PMCID: PMC3339865 DOI: 10.1007/s11538-011-9710-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 11/24/2011] [Indexed: 11/01/2022]
Abstract
We describe and analyze a periodically-forced difference equation model for malaria in mosquitoes that captures the effects of seasonality and allows the mosquitoes to feed on a heterogeneous population of hosts. We numerically show the existence of a unique globally asymptotically stable periodic orbit and calculate periodic orbits of field-measurable quantities that measure malaria transmission. We integrate this model with an individual-based stochastic simulation model for malaria in humans to compare the effects of insecticide-treated nets (ITNs) and indoor residual spraying (IRS) in reducing malaria transmission, prevalence, and incidence. We show that ITNs are more effective than IRS in reducing transmission and prevalence though IRS would achieve its maximal effects within 2 years while ITNs would need two mass distribution campaigns over several years to do so. Furthermore, the combination of both interventions is more effective than either intervention alone. However, although these interventions reduce transmission and prevalence, they can lead to increased clinical malaria; and all three malaria indicators return to preintervention levels within 3 years after the interventions are withdrawn.
Collapse
|
43
|
Smith T, Ross A, Maire N, Chitnis N, Studer A, Hardy D, Brooks A, Penny M, Tanner M. Ensemble modeling of the likely public health impact of a pre-erythrocytic malaria vaccine. PLoS Med 2012; 9:e1001157. [PMID: 22272189 PMCID: PMC3260300 DOI: 10.1371/journal.pmed.1001157] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 11/30/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The RTS,S malaria vaccine may soon be licensed. Models of impact of such vaccines have mainly considered deployment via the World Health Organization's Expanded Programme on Immunization (EPI) in areas of stable endemic transmission of Plasmodium falciparum, and have been calibrated for such settings. Their applicability to low transmission settings is unclear. Evaluations of the efficiency of different deployment strategies in diverse settings should consider uncertainties in model structure. METHODS AND FINDINGS An ensemble of 14 individual-based stochastic simulation models of P. falciparum dynamics, with differing assumptions about immune decay, transmission heterogeneity, and treatment access, was constructed. After fitting to an extensive library of field data, each model was used to predict the likely health benefits of RTS,S deployment, via EPI (with or without catch-up vaccinations), supplementary vaccination of school-age children, or mass vaccination every 5 y. Settings with seasonally varying transmission, with overall pre-intervention entomological inoculation rates (EIRs) of two, 11, and 20 infectious bites per person per annum, were considered. Predicted benefits of EPI vaccination programs over the simulated 14-y time horizon were dependent on duration of protection. Nevertheless, EPI strategies (with an initial catch-up phase) averted the most deaths per dose at the higher EIRs, although model uncertainty increased with EIR. At two infectious bites per person per annum, mass vaccination strategies substantially reduced transmission, leading to much greater health effects per dose, even at modest coverage. CONCLUSIONS In higher transmission settings, EPI strategies will be most efficient, but vaccination additional to the EPI in targeted low transmission settings, even at modest coverage, might be more efficient than national-level vaccination of infants. The feasibility and economics of mass vaccination, and the circumstances under which vaccination will avert epidemics, remain unclear. The approach of using an ensemble of models provides more secure conclusions than a single-model approach, and suggests greater confidence in predictions of health effects for lower transmission settings than for higher ones.
Collapse
Affiliation(s)
- Thomas Smith
- Swiss Tropical and Public Health Institute, Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Maire N, Shillcutt SD, Walker DG, Tediosi F, Smith TA. Cost-effectiveness of the introduction of a pre-erythrocytic malaria vaccine into the expanded program on immunization in sub-Saharan Africa: analysis of uncertainties using a stochastic individual-based simulation model of Plasmodium falciparum malaria. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2011; 14:1028-1038. [PMID: 22152171 DOI: 10.1016/j.jval.2011.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 06/10/2011] [Accepted: 06/13/2011] [Indexed: 05/31/2023]
Abstract
OBJECTIVE To evaluate the cost-effectiveness of introducing the RTS,S malaria vaccine into the Expanded Programme on Immunization (EPI) in Sub-Saharan Africa (SSA), the contributions of different sources of uncertainty, and the associated expected value of perfect information (EVPI). METHODS Vaccination was simulated in populations of 100,000 people at 10 different entomological inoculation rates (EIRs), using an existing stochastic model and a 10-year time horizon. Incremental cost-effectiveness ratios (ICERs) and EVPI were computed from weighted averages of outputs using two different assignments of the EIR distribution in 2007. Uncertainty was evaluated by resampling of epidemiological, vaccination, and health systems model parameters. RESULTS Health benefits were predicted consistently only at low transmission, and program costs always substantially exceeded case management savings. Optimal cost-effectiveness was at EIR of about 10 infectious bites per annum (ibpa). Main contributors to ICER uncertainty were uncertainty in transmission intensity, price per vaccine dose, decay rate of the vaccine effect, degree of homogeneity in host response, and some epidemiological model parameters. Other health system costs were unimportant. With a ceiling ratio of 207 international dollars per disability-adjusted life-year averted, 52.4% of parameterizations predicted cost-effectiveness in the primary analysis. CONCLUSIONS Cost-effectiveness of RTS,S will be maximal in low endemicity settings (EIR 2-20 ibpa). Widespread deployment of other transmission-reducing interventions will thus improve cost-effectiveness, suggesting a selective introduction strategy. EVPI is substantial. Accrual of up-to-date information on local endemicity to guide deployment decisions would be highly efficient.
Collapse
Affiliation(s)
- Nicolas Maire
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | | | | | | |
Collapse
|
45
|
Crowell V, Hardy D, Briët O, Chitnis N, Maire N, Smith T. Can we depend on case management to prevent re-establishment of P. falciparum malaria, after local interruption of transmission? Epidemics 2011; 4:1-8. [PMID: 22325009 DOI: 10.1016/j.epidem.2011.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 09/01/2011] [Accepted: 10/31/2011] [Indexed: 10/15/2022] Open
Abstract
Recent declines in malaria burden in many parts of the world have prompted consideration of how interruption of Plasmodium falciparum transmission could be maintained, if achieved, and notably whether large-scale vector control could be replaced with surveillance. This information is essential for elimination feasibility assessments and planning. The risk of re-establishment of transmission depends mainly on vectorial capacity (receptivity), likely to rebound once vector control is removed, the rate of importation of infections (vulnerability), the capacity to detect and treat infections and the level of immunity in infected individuals. Timely detection and removal of new infections is likely to be critical to prevent re-establishment of transmission. We assess, through mathematical modeling and simulation, which levels of case detection and treatment (case management) are required to prevent re-establishment of transmission of P. falciparum after local interruption of transmission has been achieved, in settings with varying receptivity and vulnerability. We find that, even at rather low levels of receptivity, case management alone cannot reliably prevent re-establishment of P. falciparum malaria transmission in the face of medium to high importation rates. Thus, if vector control is to be discontinued, preventing the importations by controlling transmission in source areas will generally be necessary for preventing reintroduction in such settings, and cannot be substituted by very high levels of case management coverage.
Collapse
Affiliation(s)
- Valerie Crowell
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
| | | | | | | | | | | |
Collapse
|
46
|
Smith TA, Chitnis N, Briët OJ, Tanner M. Uses of mosquito-stage transmission-blocking vaccines against Plasmodium falciparum. Trends Parasitol 2011; 27:190-6. [DOI: 10.1016/j.pt.2010.12.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 12/27/2010] [Accepted: 12/30/2010] [Indexed: 11/29/2022]
|
47
|
Ross A, Maire N, Sicuri E, Smith T, Conteh L. Determinants of the cost-effectiveness of intermittent preventive treatment for malaria in infants and children. PLoS One 2011; 6:e18391. [PMID: 21490967 PMCID: PMC3072385 DOI: 10.1371/journal.pone.0018391] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 02/28/2011] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Trials of intermittent preventive treatment in infants (IPTi) and children (IPTc) have shown promising results in reducing malaria episodes but with varying efficacy and cost-effectiveness. The effects of different intervention and setting characteristics are not well known. We simulate the effects of the different target age groups and delivery channels, seasonal or year-round delivery, transmission intensity, seasonality, proportions of malaria fevers treated and drug characteristics. METHODS We use a dynamic, individual-based simulation model of Plasmodium falciparum malaria epidemiology, antimalarial drug action and case management to simulate DALYs averted and the cost per DALY averted by IPTi and IPTc. IPT cost components were estimated from economic studies alongside trials. RESULTS IPTi and IPTc were predicted to be cost-effective in most of the scenarios modelled. The cost-effectiveness is driven by the impact on DALYs, particularly for IPTc, and the low costs, particularly for IPTi which uses the existing delivery strategy, EPI. Cost-effectiveness was predicted to decrease with low transmission, badly timed seasonal delivery in a seasonal setting, short-acting and more expensive drugs, high frequencies of drug resistance and high levels of treatment of malaria fevers. Seasonal delivery was more cost-effective in seasonal settings, and year-round in constant transmission settings. The difference was more pronounced for IPTc than IPTi due to the different proportions of fixed costs and also different assumed drug spacing during the transmission season. The number of DALYs averted was predicted to decrease as a target five-year age-band for IPTc was shifted from children under 5 years into older ages, except at low transmission intensities. CONCLUSIONS Modelling can extend the information available by predicting impact and cost-effectiveness for scenarios, for outcomes and for multiple strategies where, for practical reasons, trials cannot be carried out. Both IPTi and IPTc are generally cost-effective but could be rendered cost-ineffective by characteristics of the setting, drug or implementation.
Collapse
Affiliation(s)
- Amanda Ross
- Swiss Tropical and Public Health Institute, Basel, Switzerland.
| | | | | | | | | |
Collapse
|
48
|
Abstract
Vaccines could be a crucial component of efforts to eradicate malaria. Current attempts to develop malaria vaccines are primarily focused on Plasmodium falciparum and are directed towards reducing morbidity and mortality. Continued support for these efforts is essential, but if malaria vaccines are to be used as part of a repertoire of tools for elimination or eradication of malaria, they will need to have an impact on malaria transmission. We introduce the concept of "vaccines that interrupt malaria transmission" (VIMT), which includes not only "classical" transmission-blocking vaccines that target the sexual and mosquito stages but also pre-erythrocytic and asexual stage vaccines that have an effect on transmission. VIMT may also include vaccines that target the vector to disrupt parasite development in the mosquito. Importantly, if eradication is to be achieved, malaria vaccine development efforts will need to target other malaria parasite species, especially Plasmodium vivax, where novel therapeutic vaccines against hypnozoites or preventive vaccines with effect against multiple stages could have enormous impact. A target product profile (TPP) for VIMT is proposed and a research agenda to address current knowledge gaps and develop tools necessary for design and development of VIMT is presented.
Collapse
|
49
|
Bejon P, Williams TN, Liljander A, Noor AM, Wambua J, Ogada E, Olotu A, Osier FHA, Hay SI, Färnert A, Marsh K. Stable and unstable malaria hotspots in longitudinal cohort studies in Kenya. PLoS Med 2010; 7:e1000304. [PMID: 20625549 PMCID: PMC2897769 DOI: 10.1371/journal.pmed.1000304] [Citation(s) in RCA: 197] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 05/27/2010] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Infectious diseases often demonstrate heterogeneity of transmission among host populations. This heterogeneity reduces the efficacy of control strategies, but also implies that focusing control strategies on "hotspots" of transmission could be highly effective. METHODS AND FINDINGS In order to identify hotspots of malaria transmission, we analysed longitudinal data on febrile malaria episodes, asymptomatic parasitaemia, and antibody titres over 12 y from 256 homesteads in three study areas in Kilifi District on the Kenyan coast. We examined heterogeneity by homestead, and identified groups of homesteads that formed hotspots using a spatial scan statistic. Two types of statistically significant hotspots were detected; stable hotspots of asymptomatic parasitaemia and unstable hotspots of febrile malaria. The stable hotspots were associated with higher average AMA-1 antibody titres than the unstable clusters (optical density [OD] = 1.24, 95% confidence interval [CI] 1.02-1.47 versus OD = 1.1, 95% CI 0.88-1.33) and lower mean ages of febrile malaria episodes (5.8 y, 95% CI 5.6-6.0 versus 5.91 y, 95% CI 5.7-6.1). A falling gradient of febrile malaria incidence was identified in the penumbrae of both hotspots. Hotspots were associated with AMA-1 titres, but not seroconversion rates. In order to target control measures, homesteads at risk of febrile malaria could be predicted by identifying the 20% of homesteads that experienced an episode of febrile malaria during one month in the dry season. That 20% subsequently experienced 65% of all febrile malaria episodes during the following year. A definition based on remote sensing data was 81% sensitive and 63% specific for the stable hotspots of asymptomatic malaria. CONCLUSIONS Hotspots of asymptomatic parasitaemia are stable over time, but hotspots of febrile malaria are unstable. This finding may be because immunity offsets the high rate of febrile malaria that might otherwise result in stable hotspots, whereas unstable hotspots necessarily affect a population with less prior exposure to malaria.
Collapse
Affiliation(s)
- Philip Bejon
- Kilifi KEMRI-Wellcome Trust Collaborative Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Souza-Neiras WC, Storti-Melo LM, Cassiano GC, Couto VSCA, Couto AARA, Soares IS, Carvalho LH, Cunha MG, Póvoa MM, Herrera S, Herrera MA, Rossit ARM, Carareto CMA, Machado RLD. Plasmodium vivax circumsporozoite genotypes: a limited variation or new subspecies with major biological consequences? Malar J 2010; 9:178. [PMID: 20573199 PMCID: PMC2908638 DOI: 10.1186/1475-2875-9-178] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 06/23/2010] [Indexed: 12/02/2022] Open
Abstract
Background Plasmodium vivax circumsporozoite variants have been identified in several geographical areas. The real implication of the genetic variation in this region of the P. vivax genome has been questioned for a long time. Although previous studies have observed significant association between VK210 and the Duffy blood group, we present here that evidences of this variation are limited to the CSP central portion. Methods The phylogenetic analyses were accomplished starting from the amplification of conserved domains of 18 SSU RNAr and Cyt B. The antibodies responses against the CSP peptides, MSP-1, AMA-1 and DBP were detected by ELISA, in plasma samples of individuals infected with two P. vivax CS genotypes: VK210 and P. vivax-like. Results These analyses of the two markers demonstrate high similarity among the P. vivax CS genotypes and surprisingly showed diversity equal to zero between VK210 and P. vivax-like, positioning these CS genotypes in the same clade. A high frequency IgG antibody against the N- and C-terminal regions of the P. vivax CSP was found as compared to the immune response to the R- and V- repetitive regions (p = 0.0005, Fisher's Exact test). This difference was more pronounced when the P. vivax-like variant was present in the infection (p = 0.003, Fisher's Exact test). A high frequency of antibody response against MSP-1 and AMA-1 peptides was observed for all P. vivax CS genotypes in comparison to the same frequency for DBP. Conclusions This results target that the differences among the P. vivax CS variants are restrict to the central repeated region of the protein, mostly nucleotide variation with important serological consequences.
Collapse
Affiliation(s)
- Wanessa C Souza-Neiras
- Departamento de Biologia, Universidade Estadual Paulista Júlio Mesquita Filho, São José do Rio Preto, São Paulo State, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|