Preprint
Review

This version is not peer-reviewed.

Current Developments in Malaria Vaccination: A Concise Review on Implementation, Challenges, and Future Directions

A peer-reviewed article of this preprint also exists.

Submitted:

16 December 2024

Posted:

19 December 2024

You are already at the latest version

Abstract

Malaria remains a persistent challenge in global health, disproportionately affecting populations in endemic regions (e.g., sub-Saharan Africa). Despite decades of international collaborative efforts, this parasitic disease continues to claim hundreds of thousands of lives each year, with young children and pregnant women enduring the heaviest burden of malaria. The introduction of pre-erythrocytic malaria vaccines (RTS,S/AS01 and R21/Matrix-M), represents an important milestone in malaria control efforts with promising results from the erythrocytic vaccine RH5.1/Matrix-M in recent clinical trials. However, the approval of these vaccines is accompanied by significant challenges such as the limited efficacy, the complexity of multi-dose regimens, and numerous barriers to widespread implementation in resource-limited settings. This concise review provides an overview of the historical development and current status of malaria vaccines, tracking their milestones from initial scientific breakthroughs to the deployment of first-generation vaccines. The review also examines the complex challenges to broad malaria vaccination coverage, including logistical barriers, healthcare infrastructure effect, financial limitations, malaria vaccine hesitancy, among other obstacles in malaria-endemic regions. Additionally, we explore promising developments in malaria vaccination, such as next-generation candidates (e.g., mRNA-based vaccines), that hold the potential to offer improved efficacy, longer-lasting protection, and greater scalability. Finally, we emphasize the critical need to integrate malaria vaccination efforts with established malaria control interventions (e.g., insecticide-treated bed nets, vector control strategies, and anti-malarial drugs). Based on this review, we concluded that achieving sustained control of malaria morbidity and mortality will require strong global collaboration, sufficient funding, and continuous efforts to address inequities in access and delivery of malaria control measures including the malaria vaccines.

Keywords: 
;  ;  ;  ;  ;  

1. Introduction

Malaria is one of the big three infectious diseases along with human immunodeficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS) and tuberculosis [1,2]. This life-threatening infectious disease is caused by Plasmodium parasites, primarily transmitted to humans through the bites of infected Anopheles mosquitoes [3]. Of the five Plasmodium species capable of infecting humans (P. falciparum, P. malariae, P. ovale, P. vivax and P. knowlesi), P. falciparum is responsible for the majority of severe malaria cases and fatalities [4].
Despite the ancient origins of malaria, it remains a significant global health threat [5]. Advances in understanding the life cycle of Plasmodium in the 19th and early 20th centuries laid the foundations for vector control strategies, the development of anti-malarial drugs, and availability of malaria rapid diagnostic tests (RDTs) [6,7,8]. However, the Plasmodium ability to develop and select resistance to treatment and the mosquito’s adaptation to control measures have made malaria eradication a challenging task [9,10].
Malaria control and prevention remain important priorities, as this tropical infectious disease continues to exert a substantial burden on global health, particularly across the tropical and sub-tropical regions [11,12]. The World Health Organization (WHO) World Malaria Report 2023 indicated that in the year 2022 there were an estimated 249 million malaria cases in 85 endemic countries, marking an increase of 5 million cases from 2021 [13]. Children under five years of age remain the most vulnerable group to malaria burden, accounting for the majority of malaria-related deaths especially in sub-Saharan Africa (SSA) [14]. Pregnant women and immunocompromised patients are also considered as other malaria at-risk groups [15,16,17].
While malaria incidence has declined over the last few decades, progress in malaria prevention and control is still needed [18,19,20]. Challenges in sustaining effective malaria control measures have been exacerbated by the emergence of Plasmodium resistance to various anti-malarial drugs [21,22,23,24]. Beyond the issue of drug resistance, sustaining malaria control requires community engagement, enhanced resistance surveillance, anti-malarial quality monitoring, and building capacity to track mosquito behavior changes as illustrated recently by Guyant et al. [25].
It is also important to point out that the socio-economic toll of malaria remains profound [26,27]. This malaria toll perpetuate poverty by disrupting education, workforce productivity, and economic stability, especially in regions with fragile health care systems [28,29,30]. Further complicating these challenges that face the control of malaria is the rising insecticide resistance among the mosquito vector [31,32]. Taken together, the negative impacts of malaria and the challenges faced in its control highlight the continuous need for sustainable, innovative control measures such as the implementation of vaccination [12,33,34].
The quest for an effective malaria vaccine have spanned more than seven decades [35,36]. These efforts were primarily driven by the need for a long-term cost-effective measure to complement other malaria control measures such as insecticide-treated bed nets and anti-malarial drugs [37,38]. For example, an early study by Sauboin et al. estimated that malaria vaccination at 6, 10, and 14 weeks could prevent over 5 million clinical cases and 31,000 deaths in 42 countries over 10 years, while vaccination at 6, 7.5, and 9 months could avert 12.5 million cases and 65,400 deaths with 75% DTP3 coverage [39]. However, the complex life cycle of Plasmodium, involving both human and mosquito hosts, has posed unique challenges for malaria vaccine development [40,41,42].
Early malaria vaccine strategies targeted various stages of the Plasmodium life cycle, including the pre-erythrocytic, blood-stage, and transmission phases [43,44,45,46,47]. The first-generation malaria vaccine, RTS,S/AS01 (Mosquirix), targets the pre-erythrocytic stage of Plasmodium by generating an immune response against the P. falciparum immunodominant circumsporozoite protein (CSP) [48,49]. Following decades of research and extensive clinical trials, the WHO recommended RTS,S/AS01 in 2021 for children in areas of moderate to high transmission [50,51,52]. Data showed over 47% RTS,S/AS01 vaccine efficacy against clinical malaria and hospitalizations within 12 months post-third dose, though efficacy declines to 34% at 30 months without a booster dose [53]. Another promising development was the approval of the Oxford R21/Matrix M vaccine, which also targets the pre-erythrocytic stage and builds upon the RTS,S/AS01 platform by incorporating a higher antigen-to-adjuvant ratio to potentially enhance immune response [54,55,56,57]. Additional malaria vaccines, including those based on next-generation technologies like mRNA, are currently in development, offering hope for more efficacious malaria control measures in the near future [58,59,60,61]. An important issue that should be considered in malaria vaccination with the efficacies reported for the approved vaccines is the rapid waning of protection despite high initial efficacy. This limitation, likely due to insufficient immunological memory in individuals with prior malaria exposure, could be addressed through annual mass vaccination campaigns timed before seasonal transmission peaks (seasonal vaccination) [62,63,64,65].
Viral-vector malaria vaccines have shown potential in targeting both pre-erythrocytic and sexual stages of P. falciparum [58]. The ChAd63-MVA prime-boost regimen encoding multiple epitope–thrombospondin-related adhesion protein (ME-TRAP) induced strong CD8+ T-cell responses and reduced infection risk in endemic settings [66,67]. Transmission-blocking vaccines (TBVs) using Pfs25-IMX313 demonstrated safety and immunogenicity but had limited transmission-reducing activity, emphasizing the need for further optimization [67,68].
A deeper understanding of vaccine implementation challenges, particularly in resource-limited settings, is critical. Lessons from the rollout of COVID-19 vaccines underscore the importance of robust global partnerships, equitable access strategies, and community engagement to overcome logistical and sociopolitical barriers. Malaria vaccine initiatives must build on these insights to tackle similar obstacles, including vaccine hesitancy, funding gaps, and infrastructural limitations.
The current review aimed to concisely address the following objectives. First, the review aimed to describe the latest estimates on the global burden of malaria, highlighting its persistent impact on health and socioeconomic development, particularly in endemic regions. Second, the review aimed to outlines the critical historical milestones and challenges in malaria vaccine development. Third, the review aimed to evaluate the barriers in malaria vaccination implementation, with a focus on the issue of vaccine hesitancy. Finally, the review explored future directions, including the potential of next-generation vaccine candidates, the integration of vaccination with other malaria control measures, and the importance of global collaboration and sustained funding. The ultimate aim of the current review was to offer insights that can help to strengthen global malaria control efforts.

2. Materials and Methods

An ad hoc literature search was conducted in PubMed/Medline to identify recent and relevant studies on malaria vaccination, guided by the key objectives of this review. These objectives included description of the global burden of malaria and the key milestones in malaria vaccine development; evaluation of the malaria vaccine implementation barriers, including vaccine hesitancy; and investigation of the future directions in vaccine candidates, integration with other control measures, and the role of global collaboration.
The exact search strategy was: (("malaria"[Title/Abstract] AND "vaccine"[Title/Abstract]) AND (("RTS,S"[Title/Abstract] OR "R21/Matrix-M"[Title/Abstract]) OR ("efficacy"[Title/Abstract] OR "safety"[Title/Abstract] OR "vaccine hesitancy"[Title/Abstract] OR "implementation"[Title/Abstract] OR "challenges"[Title/Abstract] OR "next-generation vaccines"[Title/Abstract]))) OR ("Malaria Vaccines"[MeSH Terms] AND "Vaccination"[MeSH Terms]) AND (2015:2024[Date - Publication]) AND ("English"[Language]) AND ("Africa"[ MeSH Terms] OR "Asia, Southeastern"[ MeSH Terms]) AND (clinical trial[Publication Type] OR review[Publication Type]) which yielded 96 records. Articles published between 1 January 2015, and 1 December 2024, were included.
Given the narrative nature of this review, rather than being a systematic review, no strict inclusion or exclusion criteria were applied for study selection. Instead, two independent authors (the first and senior authors) conducted the literature review for the retrieved records guided by the study objectives. Articles were considered for inclusion based on their relevance to key review themes, including the global burden of malaria, milestones in vaccine development (notably RTS,S/AS01 and R21/Matrix-M), real-world implementation data, vaccine efficacy, challenges in vaccination coverage, and the socio-political factors influencing vaccine uptake. Special emphasis was placed on published studies that addressed barriers to malaria vaccine implementation, such as malaria vaccine hesitancy/resistance. Reports and policy documents from global health organizations, particularly the WHO, were also included to ensure the inclusion of the most current and policy-relevant evidence on malaria vaccination.

3. Overview of Malaria Burden Worldwide

Malaria remains a formidable global health challenge [69,70,71]. This challenge is particularly pronounced in the tropical and subtropical regions, where it disproportionately affects vulnerable populations and perpetuates cycles of poverty [11,70,72]. As stated in the introductory section of this review, and according to the WHO World Malaria Report 2023, there were an estimated 249 million malaria cases worldwide in 2022, with an increasing trend of 5 million cases compared to 2021 across 85 endemic countries [13].
Malaria endemicity classification helps determine transmission intensity, though no single measure is entirely satisfactory [73,74,75]. Traditionally, parasite rate or spleen rate in children aged 2–9 years have been used to define levels of endemicity as follows: hypo-endemic (0–10%), meso-endemic (10–50%), hyper-endemic (>50%), and holo-endemic (≥75% consistently, with low adult spleen rates) [76]. This system reflects that parasite density decreases significantly between ages 2 and 5, impacting endemicity assessments [76]. Despite its limitations, this classification remains a useful framework for prioritizing interventions in are areas with high burden of malaria [73,76,77].
The heaviest burden of malaria is observed in SSA, accounting for approximately 95% of malaria cases and deaths globally [70]. In 2022, the WHO African region reported an estimated 233 million cases and 580,000 deaths due to malaria [78]. Children under five years of age are particularly vulnerable, representing about 80% of all malaria deaths in the African region [79]. For example, Nigeria alone accounted for 27% of global malaria deaths, followed by the Democratic Republic of the Congo (DRC), Tanzania, and Niger [12]. The global distribution of malaria incidence in 2022 and mortality rates in 2021 based on the WHO data are shown in (Figure 1).

4. Historical Development of Malaria Vaccines

The quest for a malaria vaccine has been one of the most complex and enduring challenges in tropical medicine [81,82]. Unlike many other infectious diseases, malaria is caused by a eukaryotic parasite with a highly complex life cycle involving both mosquito and human hosts [83]. This life complexity, besides the ability of Plasmodium to evade the human immune system through antigenic variation, has historically made the development of an effective vaccine particularly challenging [35,40,84]. A timeline of the major milestones in malaria vaccine development is illustrated in (Figure 2).
Initial attempts to develop a malaria vaccine in the mid-20th century primarily focused on the erythrocytic stage where Plasmodium infects erythrocytes [96]. However, these early malaria vaccines faced significant limitations, as they failed to generate strong or lasting immune responses [58,97]. It was not until the identification of key antigens involved in the sporozoite stage—specifically the CSP of P. falciparum—that a major breakthrough occurred [88,98]. The CSP, expressed on the surface of the sporozoite stage of Plasmodium, became the target for a pre-erythrocytic vaccines designed to prevent the parasite from reaching the liver, where it matures and multiplies [99,100,101].
The development and approval of RTS,S/AS01 (Mosquirix) by the WHO in 2021 was a landmark achievement [48,94]. RTS,S is a recombinant protein vaccine based on the CSP of P. falciparum, fused with the hepatitis B surface antigen (HBsAg), and formulated with the AS01 adjuvant to enhance the immune response [102]. The name RTS,S/AS01 reflects the vaccine composition: RTS,S includes repeated T-cell epitopes from P. falciparum CSP and HBsAg to enhance immune response [52]. AS01 is a liposome-based adjuvant with MPL and QS-21 saponin, which together boost innate and adaptive immunity, significantly increasing the vaccine efficacy against P. falciparum [103].
Based on data from the RTS,S/AS01 Malaria Vaccine Implementation Programme report, the RTS,S/AS01 vaccine, evaluated in Phase 3 trials in SSA, showed an efficacy of around 36% in reducing clinical malaria cases in young children over four years, with a favorable safety profile [104]. After these promising results, the WHO recommended large-scale pilot programs in 2015, which were launched in Ghana, Kenya, and Malawi [105]. In 2021, following positive pilot outcomes, WHO recommended RTS,S for broader use in children in high-transmission regions, despite challenges like modest efficacy, a four-dose schedule, and waning immunity [106,107].
The Oxford R21/Matrix-M malaria vaccine, a promising malaria vaccine, demonstrated high efficacy rates in early-stage trials [108]. The efficacy of R21/Matrix-M was reported of up to 80% in African children over a one-year period, particularly in those receiving the higher adjuvant dose [109,110,111]. This led to its approval by the WHO in 2023 the second malaria vaccine for malaria prevention in children after reviewing its safety and efficacy [56,112]. The smaller dosage and improved immunogenicity profile of R21/Matrix-M, alongside the use of a potent adjuvant (Matrix-M), offer an enhanced protection [113], although its potential to outperform RTS,S/AS01 has not been tested yet [56,108].
A significant breakthrough in malaria vaccine research is the reticulocyte-binding protein homolog 5 (RH5) RH5.1/Matrix-M vaccine, a blood-stage P. falciparum vaccine candidate [95,114,115]. The RH5.1/Matrix-M vaccine is specifically designed to target the parasite during its erythrocytic phase, after exiting the hepatocytes while entering the bloodstream [116,117]. This different vaccination approach addresses a critical gap left by pre-erythrocytic vaccines such as RTS,S/AS01 and R21/Matrix-M, which are unable to protect against blood-stage parasites [118].
In a recent phase 2b trial conducted in Burkina Faso, RH5.1/Matrix-M showed promising results in children aged 5–17 months [95]. The vaccine was well-tolerated with a favorable safety profile and demonstrated robust immunogenicity [95]. The encouraging results suggest that RH5.1/Matrix-M vaccine could serve as a valuable addition to the malaria vaccine arsenal, complementing the existing approved vaccines by offering protection against the erythrocytic stage of P. falciparum [119].
New malaria vaccine candidates, including viral vector and mRNA-based vaccines, aim to provide stronger, longer-lasting immunity compared to current options like RTS,S [35,58,120]. Viral vector vaccines use modified viruses to deliver malaria antigens, enhancing cellular immunity with fewer doses [121,122,123].
Building on coronavirus disease 2019 (COVID-19) vaccine success, mRNA vaccines offer rapid production and adaptability, enabling tailored responses across different Plasmodium stages [124,125,126]. By eliciting robust humoral and cellular immunity, these next-generation malaria vaccines could have the potential to improve individual protection and to reduce transmission, supporting malaria elimination goals in high-burden areas [119,127].

5. Vaccine Efficacy and Safety of RTS,S/AS01 and R21/Matrix-M Malaria Vaccines

The RTS,S/AS01 and R21/Matrix-M vaccines represent two landmark achievements in malaria control, offering targeted immunity against P. falciparum [128]. These vaccines provide a new leap in the ability to control malaria; however, they also need further understanding of their efficacy, safety, and cost to ensure their successful implementation in endemic regions.
The RTS,S/AS01 malaria vaccine demonstrated a modest efficacy of approximately 33–36% in reducing clinical malaria cases over four years, with waning protection observed without booster doses [129,130,131,132]. Notably, no significant efficacy against severe malaria was observed in younger infants even with a booster dose [133]. Immunogenicity data reveal that the RTS,S/AS01 booster dose increases total IgG levels against vaccine antigens, with notable differences observed in IgG subclasses [134]. Additionally, variable efficacy of RTS,S/AS01 has been shown to depend on the genetics of the local P. falciparum population [135].
Post-approval safety evaluations confirmed a favorable safety profile for RTS,S/AS01, with mild injection site reactions and transient fever as the most common adverse events [129,136,137]. Rare febrile seizures were observed within seven days post-vaccination but resolved without long-term complications [129]. Importantly, no fatal adverse events were causally linked to the vaccine [138]. A phase 3b clinical trial conducted in Ghana further validated the RTS,S/AS01 vaccine safety profile, whether administered alone or co-administered with other vaccines such as yellow fever and measles-rubella [139].
The Oxford R21/Matrix-M vaccine, approved in 2023, demonstrated promising efficacy, achieving up to 75% protection in children aged 5–17 months, particularly when a booster dose is administered one year after the initial three-dose regimen [113,140]. Its safety profile is favorable, with no significant adverse effects reported during clinical trials [108]. Mild reactions, such as localized pain and fever, were the most commonly observed side effects [108,110].
Comparatively, the RTS,S/AS01 and R21/Matrix-M vaccines demonstrated similar efficacy and safety profiles in preventing malaria, particularly in children [141]. While evidence directly comparing their efficacy is lacking, early analyses suggest that both vaccines would provide a cost-effective approach for malaria prevention [142,143]. Despite the promise that both RTS,S/AS01 and R21/Matrix-M vaccines offer, the modest efficacy of these vaccines highlight the critical need for continued efforts in malaria vaccine research. Achieving higher, longer-lasting protection remains essential in the fight against malaria and to progress toward its eradication efforts [144].

6. Challenges in Malaria Vaccination Implementation

There is no doubt that the introduction and approval of the RTS,S/AS01 and R21/Matrix-M malaria vaccines offered a new hope in the fight against malaria [145]. Nevertheless, achieving a malaria vaccine that provides high efficacy and long-lasting immunity remains challenging [146]. Additionally, achieving the full potential of malaria vaccines requires addressing complex logistical, operational, and social challenges, particularly in malaria-endemic regions with limited resources [33,147].
Specifically, the challenges of implementing malaria vaccination range from infrastructure limitations to community perceptions as highlighted in (Figure 3).
For example, a major challenge is the RTS,S/AS01 vaccine four-dose schedule: the first three doses are given at months 0, 1, and 2, with a crucial booster at 18 months [148]. In rural areas in SSA where travel to health care centers can be difficult, completing this schedule poses a significant challenge [149]. Missed doses, common in settings where health care access is inconsistent, can compromise the malaria vaccine efficacy [150].
Moreover, the malaria vaccine requires a cold chain capacity, which can be problematic in regions with limited electricity and high temperatures [147,151]. Transportation difficulties and inconsistent cold storage infrastructure can also disrupt vaccine delivery in remote areas, risking loss of efficacy if the malaria vaccine temperature range was not maintained [152].
Health care workers constraints can compound these logistical barriers to successful implementation of malaria vaccination [153]. Many malaria-endemic countries, such as those in SSA, already struggle with a shortage of health care providers trained to administer and monitor vaccination programs, particularly in under-resourced areas [154,155].
Success in implementing RTS,S/AS01 can be enhanced by integrating it effectively into routine immunization schedules without overburdening health systems that are already strained by the demands of child immunizations and maternal care [156]. A study by Hill et al. in Kenya highlighted that integrating RTS,S/AS01 malaria vaccine into the Essential Programme on Immunisation could streamline its delivery and strengthen uptake [148]. Despite initial challenges with the four-dose schedule and resource limitations, this approach can enhance the vaccine impact without overburdening health care systems [148].
Financial barriers also play a critical barrier in implementing malaria vaccination with the procurement, storage, and delivery of RTS,S/AS01 requiring substantial investment, a challenge for many low-income countries where malaria is endemic [157]. While organizations such as Gavi, the Vaccine Alliance, have committed support for this goal, securing sustainable funding remains essential [158]. In SSA, where health care budgets are constrained, the cost of maintaining large-scale malaria vaccine campaigns, along with ongoing malaria control efforts, requires long-term commitments from global health stakeholders [159].

6.1. Vaccine Hesitancy as a Barrier to Successful Malaria Vaccine Implementation

Public acceptance of RTS,S/AS01 is equally important for malaria vaccination program success with evidence pointing to high acceptance in Africa [160,161,162]. Vaccine hesitancy, often rooted in distrust of health care systems or cultural beliefs, can limit malaria vaccine uptake [163]. In a study by Bam et al., caregivers in Ghana showed generally positive perceptions of the RTS,S/AS01 malaria vaccine for children, citing benefits like reduced hospital visits and cost savings, although concerns about potential side effects highlighted the need for targeted health education to address vaccine hesitancy and promote broader malaria vaccine uptake [164].
In an early study by Ojakaa et al., 88% of caregivers in Kenya expressed willingness to accept a malaria vaccine for children, with acceptance highest in malaria-endemic areas [165]. More recently, in a study by Nyalundja et al., only 7.26% of adults in eastern DRC were aware of the malaria vaccine, though 52.6% were willing to vaccinate their under-five children, with higher acceptance associated with factors such as middle income and semi-rural residence [166].
In another study by Mtenga et al., 84.2% of Tanzanian mothers expressed strong acceptance of a malaria vaccine for their children, with additional support from community stakeholders who saw the vaccine as a valuable complement to existing prevention strategies, though they raised questions about efficacy, side effects, and eligibility [167]. Outside Africa, in a study by Amin et al., 70% of parents in malaria-endemic areas of Bangladesh indicated willingness to vaccinate their under-five children against malaria, with higher acceptance associated with residence, education, income, and family size [168].
In the context these challenges, an important study by Grant et al., highlighted key implementation challenges of malaria vaccination in Ghana, including issues with the dosing schedule, eligibility criteria, and logistical support, such as cold-chain and transport limitations [149]. This qualitative study revealed that community rumors leading to vaccine refusals emphasized the need for robust, culturally tailored vaccine promotion efforts [149]. These findings by Grant et al. offered critical insights for scaling RTS,S/AS01E in Ghana and future malaria vaccine rollouts across Africa [149].
A recent systematic review by Ansar et al. analyzed 18 studies involving 18,561 participants, revealing an overall RTS,S/AS01 malaria vaccine acceptance rate of 87.5%, with rates ranging from 32.3% to 99.3% [169]. Countries like Ghana and Nigeria showed particularly high vaccine acceptance, driven by factors such as prior vaccination experiences, knowledge about malaria, and community engagement in prevention behaviors [169].

7. Future Directions

As malaria continues to claim hundreds of thousands of lives annually, particularly in SSA, the development of more effective malaria vaccines remains an urgent global health priority [33,170,171,172]. While RTS,S/AS01 and R21/Matrix-M vaccines approval marked historic achievements and can be viewed as important benchmark to compare with other novel vaccines, their modest efficacy highlight the need for next-generation malaria vaccines with improved efficacy profiles, easier administration, and long-lasting protection [129,173]. The future of malaria vaccine development relies on innovative scientific approaches, enhanced global collaboration, and integration of vaccination with other malaria control strategies [174].
In addition, other Plasmodium species, such as P. vivax, which can cause severe malaria, have historically received less focus compared to P. falciparum [175]. The importance of focus on P. vivax is highlighted by being a predominant Plasmodium species in most non-African endemic countries [175]. The view that P. vivax might not be a priority for prevention as opposed to P. falciparum can be challenged as follows. Dormant P. vivax hypnozoites, capable of reactivating months after initial infection, high transmission potential, coupled with asymptomatic carriers and outdoor-biting mosquito vectors, the ability of P. vivax to cause severe disease justify the quest for effective vaccines targeting this species [176,177]. Vaccines targeting P. vivax are essential to prevent relapses, reduce disease burden, and disrupt transmission, addressing critical gaps in the global effort to eliminate malaria [178,179].

7.1. Next-Generation Vaccine Candidates

The limitations of RTS,S/AS01 and R21/Matrix-M vaccines, particularly its modest efficacy, emphasize the need for vaccines that provide more robust and longer-lasting immunity [106]. Therefore, new vaccine types are being explored in the context of malaria prevention, including mRNA-based vaccines and vaccines targeting different stages of the Plasmodium life cycle [60,180].
The success of mRNA vaccines in the COVID-19 pandemic raised the interest in applying this novel vaccine type to malaria prevention [180]. mRNA vaccines have the advantage of rapid scalability, adaptability to new antigens, and the ability to elicit strong immune responses [181]. Efforts to develop mRNA vaccines targeting P. falciparum are already underway, with preclinical studies showing promising results [119]. Similarly, mRNA-based transmission-blocking vaccines are under development, aiming to interrupt the mosquito-to-human transmission cycle by targeting the sexual stages of the parasite within the mosquito [182,183,184]. These approaches could complement existing malaria vaccines and offer new enhanced opportunities for malaria prevention.

7.2. Integration with other Malaria Control Measures

Malaria vaccines must be viewed as part of a comprehensive approach to malaria control rather than as a stand-alone solution [185,186]. Future malaria control strategies require the incorporation of malaria vaccination with established effective measures such as insecticide-treated bed nets, indoor residual spraying, RDTs, and effective anti-malarial drugs [187,188].
Combination strategies, such as using malaria vaccines alongside seasonal malaria chemoprevention (SMC), have already shown potential for synergistic effects, particularly in high-transmission areas as shown in a recent study by Dicko et al. [64]. Operational research can also be critical to understand the best approach for malaria vaccines’ deployment within specific epidemiological contexts, to ensure they complement existing interventions and maximize the public health impact of malaria vaccination [189].

7.3. Global Collaboration and Funding

To address malaria challenges, it is essential to stress that collaborative concerted global efforts are needed to ensure that malaria vaccines are successfully developed, manufactured, and distributed [190,191]. International partnerships between governments, research institutions, pharmaceutical companies, and the global health organizations (e.g., the WHO, Gavi, and the Bill & Melinda Gates Foundation), are examples of the helpful efforts needed to drive progress in malaria prevention through vaccination [192,193].
Future collaboration would be critical to secure the necessary funding for ongoing research, in order to ensure equitable access to malaria vaccines, and to maintain the momentum toward malaria elimination [194,195]. These efforts are recommended to help establish cold chain infrastructure, ensure equitable vaccine delivery, and support community engagement to build public trust in malaria vaccination in the endemic regions [147,155,156,159,196]. Additionally, integrating malaria vaccines into national immunization programs with coordinated international support, can be viewed as an important step toward reducing malaria burden and its eventual elimination [197,198,199].

8. Conclusions

Malaria vaccination has moved from aspiration to reality with the introduction and approval of RTS,S/AS01 and R21/Matrix-M vaccines that are considered as two milestones in in the protection of young children in high-endemic regions from severe malaria. In addition, the recent phase 2b efficacy data for the RH5.1/Matrix-M vaccine represents another critical development, targeting the erythrocytic stage of P. falciparum and filling gaps left by pre-erythrocytic vaccines. While the approved malaria vaccines have limitations in efficacy and require a multi-dose schedule, the approval of these vaccines highlights both the promise and the challenges of malaria vaccination. Next-generation malaria vaccine candidates, including the emerging mRNA-based vaccines, bring renewed hope to the area of malaria control and prevention with the potential for higher efficacy and long-lasting immunity. However, real progress will depend on combining vaccination efforts with proven effective interventions to reduce malaria burden such as vector control and anti-malarial drugs. These measures should be sustained by global collaboration and dedicated funding. So far, the progress that has been made in malaria vaccination efforts offer valuable insights for the ultimate goal of malaria elimination; however, significant challenges remain and need to be addressed by global collaborative efforts.

Author Contributions

Conceptualization, Malik Sallam; methodology, Malik Sallam, Arwa Omar Al-Khatib, Kholoud Al-Mahzoum, Doaa H. Abdelaziz and Mohammed Sallam; software, Malik Sallam; investigation, Malik Sallam, Arwa Omar Al-Khatib, Kholoud Al-Mahzoum, Doaa H. Abdelaziz and Mohammed Sallam; resources, Malik Sallam; data curation, Malik Sallam, Arwa Omar Al-Khatib, Kholoud Al-Mahzoum, Doaa H. Abdelaziz and Mohammed Sallam; writing—original draft preparation, Malik Sallam; writing—review and editing, Malik Sallam, Arwa Omar Al-Khatib, Kholoud Al-Mahzoum, Doaa H. Abdelaziz and Mohammed Sallam; visualization, Malik Sallam; supervision, Malik Sallam; project administration, Malik Sallam. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AE Adverse event
AIDS Acquired immunodeficiency syndrome
ChAd63 Chimpanzee adenovirus 63
COVID-19 Coronavirus disease 2019
CSP Circumsporozoite protein
DRC the Democratic Republic of the Congo
DTP3 Diphtheria tetanus toxoid and pertussis
HBsAg Hepatitis B surface antigen
HIV Human immunodeficiency virus
ME-TRAP Multiple epitope–thrombospondin-related adhesion protein
MVA modified Vaccinia Ankara
P. Plasmodium
RDT Rapid diagnostic test
RH5 Reticulocyte-binding protein homolog 5
SMC Seasonal malaria chemoprevention
SSA sub-Saharan Africa
TBV Transmission-blocking vaccine
WHO The World Health Organization

References

  1. Makam, P.; Matsa, R. "Big Three" Infectious Diseases: Tuberculosis, Malaria and HIV/AIDS. Curr Top Med Chem 2021, 21, 2779–2799. [Google Scholar] [CrossRef] [PubMed]
  2. Murray, C.J.; Ortblad, K.F.; Guinovart, C.; Lim, S.S.; Wolock, T.M.; Roberts, D.A.; Dansereau, E.A.; Graetz, N.; Barber, R.M.; Brown, J.C.; et al. Global, regional, and national incidence and mortality for HIV, tuberculosis, and malaria during 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014, 384, 1005–1070. [Google Scholar] [CrossRef] [PubMed]
  3. Phillips, M.A.; Burrows, J.N.; Manyando, C.; van Huijsduijnen, R.H.; Van Voorhis, W.C.; Wells, T.N.C. Malaria. Nature Reviews Disease Primers 2017, 3, 17050. [Google Scholar] [CrossRef] [PubMed]
  4. Balaji, S.N.; Deshmukh, R.; Trivedi, V. Severe malaria: Biology, clinical manifestation, pathogenesis and consequences. J Vector Borne Dis 2020, 57, 1–13. [Google Scholar] [CrossRef] [PubMed]
  5. Talapko, J.; Škrlec, I.; Alebić, T.; Jukić, M.; Včev, A. Malaria: The Past and the Present. Microorganisms 2019, 7. [Google Scholar] [CrossRef] [PubMed]
  6. Raghavendra, K.; Barik, T.K.; Reddy, B.P.; Sharma, P.; Dash, A.P. Malaria vector control: from past to future. Parasitol Res 2011, 108, 757–779. [Google Scholar] [CrossRef]
  7. van Schalkwyk, D.A. History of Antimalarial Agents. In eLS; 2015; pp. 1–5. [CrossRef]
  8. Wongsrichanalai, C.; Barcus, M.J.; Muth, S.; Sutamihardja, A.; Wernsdorfer, W.H. A Review of Malaria Diagnostic Tools: Microscopy and Rapid Diagnostic Test (RDT). The American Journal of Tropical Medicine and Hygiene 2007, 77, 119–127. [Google Scholar] [CrossRef] [PubMed]
  9. Viana, G.M.; Melo, G.; Nobrega De Sousa, T.; Pucca, M.B. Editorial: Challenges for diagnosis, treatment, and elimination of malaria. Frontiers in Tropical Diseases 2024, 5, 1394693. [Google Scholar] [CrossRef]
  10. Liu, J.; Modrek, S.; Gosling, R.D.; Feachem, R.G. Malaria eradication: is it possible? Is it worth it? Should we do it? Lancet Glob Health 2013, 1, e2–3. [Google Scholar] [CrossRef] [PubMed]
  11. Maharaj, R.; Kissoon, S.; Lakan, V.; Kheswa, N. Rolling back malaria in Africa - challenges and opportunities to winning the elimination battle. S Afr Med J 2019, 109, 53–56. [Google Scholar] [CrossRef]
  12. Oladipo, H.J.; Tajudeen, Y.A.; Oladunjoye, I.O.; Yusuff, S.I.; Yusuf, R.O.; Oluwaseyi, E.M.; AbdulBasit, M.O.; Adebisi, Y.A.; El-Sherbini, M.S. Increasing challenges of malaria control in sub-Saharan Africa: Priorities for public health research and policymakers. Ann Med Surg (Lond) 2022, 81, 104366. [Google Scholar] [CrossRef]
  13. World Health Organization. World malaria report 2023; World Health Organization, 2023. [Google Scholar]
  14. Sarfo, J.O.; Amoadu, M.; Kordorwu, P.Y.; Adams, A.K.; Gyan, T.B.; Osman, A.G.; Asiedu, I.; Ansah, E.W. Malaria amongst children under five in sub-Saharan Africa: a scoping review of prevalence, risk factors and preventive interventions. Eur J Med Res 2023, 28, 80. [Google Scholar] [CrossRef] [PubMed]
  15. Schantz-Dunn, J.; Nour, N.M. Malaria and pregnancy: a global health perspective. Rev Obstet Gynecol 2009, 2, 186–192. [Google Scholar]
  16. Cohen, C.; Karstaedt, A.; Frean, J.; Thomas, J.; Govender, N.; Prentice, E.; Dini, L.; Galpin, J.; Crewe-Brown, H. Increased prevalence of severe malaria in HIV-infected adults in South Africa. Clin Infect Dis 2005, 41, 1631–1637. [Google Scholar] [CrossRef]
  17. Fried, M.; Duffy, P.E. Malaria during Pregnancy. Cold Spring Harb Perspect Med 2017, 7. [Google Scholar] [CrossRef]
  18. Liu, Q.; Jing, W.; Kang, L.; Liu, J.; Liu, M. Trends of the global, regional and national incidence of malaria in 204 countries from 1990 to 2019 and implications for malaria prevention. J Travel Med 2021, 28. [Google Scholar] [CrossRef] [PubMed]
  19. Leal Filho, W.; May, J.; May, M.; Nagy, G.J. Climate change and malaria: some recent trends of malaria incidence rates and average annual temperature in selected sub-Saharan African countries from 2000 to 2018. Malaria Journal 2023, 22, 248. [Google Scholar] [CrossRef] [PubMed]
  20. El-Moamly, A.A. How can we get malaria control back on track? Bmj 2024, 385, q1408. [Google Scholar] [CrossRef] [PubMed]
  21. Ippolito, M.M.; Moser, K.A.; Kabuya, J.B.; Cunningham, C.; Juliano, J.J. Antimalarial Drug Resistance and Implications for the WHO Global Technical Strategy. Curr Epidemiol Rep 2021, 8, 46–62. [Google Scholar] [CrossRef] [PubMed]
  22. White, N.J. Antimalarial drug resistance. J Clin Invest 2004, 113, 1084–1092. [Google Scholar] [CrossRef] [PubMed]
  23. Assefa, A.; Fola, A.A.; Tasew, G. Emergence of Plasmodium falciparum strains with artemisinin partial resistance in East Africa and the Horn of Africa: is there a need to panic? Malaria Journal 2024, 23, 34. [Google Scholar] [CrossRef]
  24. Zheng, D.; Liu, T.; Yu, S.; Liu, Z.; Wang, J.; Wang, Y. Antimalarial Mechanisms and Resistance Status of Artemisinin and Its Derivatives. Trop Med Infect Dis 2024, 9, 223. [Google Scholar] [CrossRef] [PubMed]
  25. Guyant, P.; Corbel, V.; Guérin, P.J.; Lautissier, A.; Nosten, F.; Boyer, S.; Coosemans, M.; Dondorp, A.M.; Sinou, V.; Yeung, S.; et al. Past and new challenges for malaria control and elimination: the role of operational research for innovation in designing interventions. Malaria Journal 2015, 14, 279. [Google Scholar] [CrossRef]
  26. Sachs, J.; Malaney, P. The economic and social burden of malaria. Nature 2002, 415, 680–685. [Google Scholar] [CrossRef] [PubMed]
  27. Mori, A.T.; Mallange, G.; Kühl, M.-J.; Okell, L. Cost of treating severe malaria in children in Africa: a systematic literature review. Malaria Journal 2024, 23, 334. [Google Scholar] [CrossRef]
  28. Ricci, F. Social implications of malaria and their relationships with poverty. Mediterr J Hematol Infect Dis 2012, 4, e2012048. [Google Scholar] [CrossRef] [PubMed]
  29. Andrade, M.V.; Noronha, K.; Diniz, B.P.C.; Guedes, G.; Carvalho, L.R.; Silva, V.A.; Calazans, J.A.; Santos, A.S.; Silva, D.N.; Castro, M.C. The economic burden of malaria: a systematic review. Malaria Journal 2022, 21, 283. [Google Scholar] [CrossRef] [PubMed]
  30. Wafula, S.T.; Habermann, T.; Franke, M.A.; May, J.; Puradiredja, D.I.; Lorenz, E.; Brinkel, J. What are the pathways between poverty and malaria in sub-Saharan Africa? A systematic review of mediation studies. Infectious Diseases of Poverty 2023, 12, 58. [Google Scholar] [CrossRef]
  31. Suh, P.F.; Elanga-Ndille, E.; Tchouakui, M.; Sandeu, M.M.; Tagne, D.; Wondji, C.; Ndo, C. Impact of insecticide resistance on malaria vector competence: a literature review. Malaria Journal 2023, 22, 19. [Google Scholar] [CrossRef]
  32. Nkya, T.E.; Akhouayri, I.; Poupardin, R.; Batengana, B.; Mosha, F.; Magesa, S.; Kisinza, W.; David, J.-P. Insecticide resistance mechanisms associated with different environments in the malaria vector Anopheles gambiae: a case study in Tanzania. Malaria Journal 2014, 13, 28. [Google Scholar] [CrossRef]
  33. Sutanto, H. Combating Malaria with Vaccines: Insights from the One Health Framework. Acta Microbiologica Hellenica 2024, 69, 153–166. [Google Scholar] [CrossRef]
  34. Cohen, J.M.; Okumu, F.; Moonen, B. The fight against malaria: Diminishing gains and growing challenges. Sci Transl Med 2022, 14, eabn3256. [Google Scholar] [CrossRef] [PubMed]
  35. El-Moamly, A.A.; El-Sweify, M.A. Malaria vaccines: the 60-year journey of hope and final success—lessons learned and future prospects. Tropical Medicine and Health 2023, 51, 29. [Google Scholar] [CrossRef] [PubMed]
  36. Moorthy, V.S.; Good, M.F.; Hill, A.V. Malaria vaccine developments. Lancet 2004, 363, 150–156. [Google Scholar] [CrossRef] [PubMed]
  37. Tizifa, T.A.; Kabaghe, A.N.; McCann, R.S.; van den Berg, H.; Van Vugt, M.; Phiri, K.S. Prevention Efforts for Malaria. Current Tropical Medicine Reports 2018, 5, 41–50. [Google Scholar] [CrossRef] [PubMed]
  38. Constenla, D. Assessing the economic benefits of vaccines based on the health investment life course framework: a review of a broader approach to evaluate malaria vaccination. Vaccine 2015, 33, 1527–1540. [Google Scholar] [CrossRef] [PubMed]
  39. Sauboin, C.J.; Van Bellinghen, L.A.; Van De Velde, N.; Van Vlaenderen, I. Potential public health impact of RTS,S malaria candidate vaccine in sub-Saharan Africa: a modelling study. Malar J 2015, 14, 524. [Google Scholar] [CrossRef]
  40. Crompton, P.D.; Pierce, S.K.; Miller, L.H. Advances and challenges in malaria vaccine development. J Clin Invest 2010, 120, 4168–4178. [Google Scholar] [CrossRef]
  41. Lorenz, V.; Karanis, P. Malaria vaccines: looking back and lessons learnt. Asian Pac J Trop Biomed 2011, 1, 74–78. [Google Scholar] [CrossRef]
  42. Lorenz, V.; Karanis, G.; Karanis, P. Malaria vaccine development and how external forces shape it: an overview. Int J Environ Res Public Health 2014, 11, 6791–6807. [Google Scholar] [CrossRef]
  43. Duffy, P.E.; Sahu, T.; Akue, A.; Milman, N.; Anderson, C. Pre-erythrocytic malaria vaccines: identifying the targets. Expert Rev Vaccines 2012, 11, 1261–1280. [Google Scholar] [CrossRef] [PubMed]
  44. Zheng, J.; Pan, H.; Gu, Y.; Zuo, X.; Ran, N.; Yuan, Y.; Zhang, C.; Wang, F. Prospects for Malaria Vaccines: Pre-Erythrocytic Stages, Blood Stages, and Transmission-Blocking Stages. Biomed Res Int 2019, 2019, 9751471. [Google Scholar] [CrossRef]
  45. Richie, T.L.; Billingsley, P.F.; Sim, B.K.; James, E.R.; Chakravarty, S.; Epstein, J.E.; Lyke, K.E.; Mordmüller, B.; Alonso, P.; Duffy, P.E.; et al. Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine 2015, 33, 7452–7461. [Google Scholar] [CrossRef]
  46. Tougan, T.; Ito, K.; Palacpac, N.M.; Egwang, T.G.; Horii, T. Immunogenicity and protection from malaria infection in BK-SE36 vaccinated volunteers in Uganda is not influenced by HLA-DRB1 alleles. Parasitol Int 2016, 65, 455–458. [Google Scholar] [CrossRef] [PubMed]
  47. Varo, R.; Chaccour, C.; Bassat, Q. Update on malaria. Med Clin (Barc) 2020, 155, 395–402. [Google Scholar] [CrossRef] [PubMed]
  48. Laurens, M.B. RTS,S/AS01 vaccine (Mosquirix™): an overview. Hum Vaccin Immunother 2020, 16, 480–489. [Google Scholar] [CrossRef] [PubMed]
  49. Marques-da-Silva, C.; Peissig, K.; Kurup, S.P. Pre-Erythrocytic Vaccines against Malaria. Vaccines (Basel) 2020, 8, 400. [Google Scholar] [CrossRef]
  50. Zavala, F.; Tam, J.P.; Hollingdale, M.R.; Cochrane, A.H.; Quakyi, I.; Nussenzweig, R.S.; Nussenzweig, V. Rationale for development of a synthetic vaccine against Plasmodium falciparum malaria. Science 1985, 228, 1436–1440. [Google Scholar] [CrossRef] [PubMed]
  51. World Health Organization. WHO recommends groundbreaking malaria vaccine for children at risk. Available online: https://www.who.int/news/item/06-10-2021-who-recommends-groundbreaking-malaria-vaccine-for-children-at-risk (accessed on 2 November 2024).
  52. Egbewande, O.M. The RTS,S malaria vaccine: Journey from conception to recommendation. Public Health Pract (Oxf) 2022, 4, 100283. [Google Scholar] [CrossRef]
  53. Syed, Y.Y. RTS,S/AS01 malaria vaccine (Mosquirix®): a profile of its use. Drugs & Therapy Perspectives 2022, 38, 373–381. [Google Scholar] [CrossRef]
  54. Hanboonkunupakarn, B.; Mukaka, M.; Jittamala, P.; Poovorawan, K.; Pongsuwan, P.; Stockdale, L.; Provstgaard-Morys, S.; Chotivanich, K.; Tarning, J.; Hoglund, R.M.; et al. A randomised trial of malaria vaccine R21/Matrix-M™ with and without antimalarial drugs in Thai adults. npj Vaccines 2024, 9, 124. [Google Scholar] [CrossRef] [PubMed]
  55. Aderinto, N.; Olatunji, G.; Kokori, E.; Sikirullahi, S.; Aboje, J.E.; Ojabo, R.E. A perspective on Oxford's R21/Matrix-M™ malaria vaccine and the future of global eradication efforts. Malar J 2024, 23, 16. [Google Scholar] [CrossRef]
  56. World Health Organization. R21/Matrix-M malaria vaccine: Evidence to recommendations framework, 2023. Available online: https://www.who.int/publications/m/item/r21-matrix-m-malaria-vaccine--evidence-to-recommendations-framework--2023 (accessed on 5 November 2024).
  57. Moorthy, V.; Hamel, M.J.; Smith, P.G. Malaria vaccines for children: and now there are two. Lancet 2024, 403, 504–505. [Google Scholar] [CrossRef]
  58. Tsoumani, M.E.; Voyiatzaki, C.; Efstathiou, A. Malaria Vaccines: From the Past towards the mRNA Vaccine Era. Vaccines (Basel) 2023, 11, 1452. [Google Scholar] [CrossRef] [PubMed]
  59. Matarazzo, L.; Bettencourt, P.J.G. mRNA vaccines: a new opportunity for malaria, tuberculosis and HIV. Front Immunol 2023, 14, 1172691. [Google Scholar] [CrossRef] [PubMed]
  60. Borkens, Y. Malaria & mRNA Vaccines: A Possible Salvation from One of the Most Relevant Infectious Diseases of the Global South. Acta Parasitologica 2023, 68, 916–928. [Google Scholar] [CrossRef] [PubMed]
  61. Yang, L.; Tang, L.; Zhang, M.; Liu, C. Recent Advances in the Molecular Design and Delivery Technology of mRNA for Vaccination Against Infectious Diseases. Front Immunol 2022, 13, 896958. [Google Scholar] [CrossRef] [PubMed]
  62. Greenwood, B.; Dicko, A.; Sagara, I.; Zongo, I.; Tinto, H.; Cairns, M.; Kuepfer, I.; Milligan, P.; Ouedraogo, J.B.; Doumbo, O.; et al. Seasonal vaccination against malaria: a potential use for an imperfect malaria vaccine. Malar J 2017, 16, 182. [Google Scholar] [CrossRef]
  63. Ubillos, I.; Ayestaran, A.; Nhabomba, A.J.; Dosoo, D.; Vidal, M.; Jiménez, A.; Jairoce, C.; Sanz, H.; Aguilar, R.; Williams, N.A.; et al. Baseline exposure, antibody subclass, and hepatitis B response differentially affect malaria protective immunity following RTS,S/AS01E vaccination in African children. BMC Med 2018, 16, 197. [Google Scholar] [CrossRef] [PubMed]
  64. Dicko, A.; Ouedraogo, J.-B.; Zongo, I.; Sagara, I.; Cairns, M.; Yerbanga, R.S.; Issiaka, D.; Zoungrana, C.; Sidibe, Y.; Tapily, A.; et al. Seasonal vaccination with RTS,S/AS01<sub>E</sub> vaccine with or without seasonal malaria chemoprevention in children up to the age of 5 years in Burkina Faso and Mali: a double-blind, randomised, controlled, phase 3 trial. The Lancet Infectious Diseases 2024, 24, 75–86. [Google Scholar] [CrossRef] [PubMed]
  65. Chandramohan, D.; Zongo, I.; Sagara, I.; Cairns, M.; Yerbanga, R.S.; Diarra, M.; Nikièma, F.; Tapily, A.; Sompougdou, F.; Issiaka, D.; et al. Seasonal Malaria Vaccination with or without Seasonal Malaria Chemoprevention. N Engl J Med 2021, 385, 1005–1017. [Google Scholar] [CrossRef]
  66. Mensah, V.A.; Gueye, A.; Ndiaye, M.; Edwards, N.J.; Wright, D.; Anagnostou, N.A.; Syll, M.; Ndaw, A.; Abiola, A.; Bliss, C.; et al. Safety, Immunogenicity and Efficacy of Prime-Boost Vaccination with ChAd63 and MVA Encoding ME-TRAP against Plasmodium falciparum Infection in Adults in Senegal. PLoS One 2016, 11, e0167951. [Google Scholar] [CrossRef] [PubMed]
  67. de Graaf, H.; Payne, R.O.; Taylor, I.; Miura, K.; Long, C.A.; Elias, S.C.; Zaric, M.; Minassian, A.M.; Silk, S.E.; Li, L.; et al. Safety and Immunogenicity of ChAd63/MVA Pfs25-IMX313 in a Phase I First-in-Human Trial. Front Immunol 2021, 12, 694759. [Google Scholar] [CrossRef]
  68. Zaric, M.; Marini, A.; Nielsen, C.M.; Gupta, G.; Mekhaiel, D.; Pham, T.P.; Elias, S.C.; Taylor, I.J.; de Graaf, H.; Payne, R.O.; et al. Poor CD4(+) T Cell Immunogenicity Limits Humoral Immunity to P. falciparum Transmission-Blocking Candidate Pfs25 in Humans. Front Immunol 2021, 12, 732667. [Google Scholar] [CrossRef] [PubMed]
  69. Nasir, S.M.I.; Amarasekara, S.; Wickremasinghe, R.; Fernando, D.; Udagama, P. Prevention of re-establishment of malaria: historical perspective and future prospects. Malaria Journal 2020, 19, 452. [Google Scholar] [CrossRef] [PubMed]
  70. Li, J.; Docile, H.J.; Fisher, D.; Pronyuk, K.; Zhao, L. Current Status of Malaria Control and Elimination in Africa: Epidemiology, Diagnosis, Treatment, Progress and Challenges. Journal of Epidemiology and Global Health 2024, 14, 561–579. [Google Scholar] [CrossRef] [PubMed]
  71. Cibulskis, R.E.; Alonso, P.; Aponte, J.; Aregawi, M.; Barrette, A.; Bergeron, L.; Fergus, C.A.; Knox, T.; Lynch, M.; Patouillard, E.; et al. Malaria: Global progress 2000 – 2015 and future challenges. Infectious Diseases of Poverty 2016, 5, 61. [Google Scholar] [CrossRef] [PubMed]
  72. Parija, S.C. The persistent challenges of malaria. Trop Parasitol 2021, 11, 1–2. [Google Scholar] [CrossRef]
  73. Hay, S.I.; Smith, D.L.; Snow, R.W. Measuring malaria endemicity from intense to interrupted transmission. Lancet Infect Dis 2008, 8, 369–378. [Google Scholar] [CrossRef]
  74. Weiss, D.J.; Lucas, T.C.D.; Nguyen, M.; Nandi, A.K.; Bisanzio, D.; Battle, K.E.; Cameron, E.; Twohig, K.A.; Pfeffer, D.A.; Rozier, J.A.; et al. Mapping the global prevalence, incidence, and mortality of Plasmodium falciparum, 2000-17: a spatial and temporal modelling study. Lancet 2019, 394, 322–331. [Google Scholar] [CrossRef] [PubMed]
  75. Battle, K.E.; Lucas, T.C.D.; Nguyen, M.; Howes, R.E.; Nandi, A.K.; Twohig, K.A.; Pfeffer, D.A.; Cameron, E.; Rao, P.C.; Casey, D.; et al. Mapping the global endemicity and clinical burden of Plasmodium vivax, 2000-17: a spatial and temporal modelling study. Lancet 2019, 394, 332–343. [Google Scholar] [CrossRef] [PubMed]
  76. World Health Organization. WHO malaria terminology, 2021 update. Available online: https://www.who.int/publications/i/item/9789240038400 (accessed on 3 November 2024).
  77. Autino, B.; Noris, A.; Russo, R.; Castelli, F. Epidemiology of malaria in endemic areas. Mediterr J Hematol Infect Dis 2012, 4, e2012060. [Google Scholar] [CrossRef] [PubMed]
  78. World Health Organization. Malaria - Key facts. Available online: https://www.who.int/news-room/fact-sheets/detail/malaria (accessed on 3 November 2024).
  79. Mbishi, J.V.; Chombo, S.; Luoga, P.; Omary, H.J.; Paulo, H.A.; Andrew, J.; Addo, I.Y. Malaria in under-five children: prevalence and multi-factor analysis of high-risk African countries. BMC Public Health 2024, 24, 1687. [Google Scholar] [CrossRef] [PubMed]
  80. World Health Organization. The Global Health Observatory: Malaria burden data: cases and deaths. Available online: https://www.who.int/data/gho/data/themes/topics/topic-details/GHO/malaria-cases-deaths (accessed on 2 November 2024).
  81. Hill, A.V. Vaccines against malaria. Philos Trans R Soc Lond B Biol Sci 2011, 366, 2806–2814. [Google Scholar] [CrossRef]
  82. Hoffman, S.L.; Vekemans, J.; Richie, T.L.; Duffy, P.E. The march toward malaria vaccines. Vaccine 2015, 33 Suppl 4, D13–23. [Google Scholar] [CrossRef]
  83. Sato, S. Plasmodium-a brief introduction to the parasites causing human malaria and their basic biology. J Physiol Anthropol 2021, 40, 1. [Google Scholar] [CrossRef] [PubMed]
  84. Richie, T. High road, low road? Choices and challenges on the pathway to a malaria vaccine. Parasitology 2006, 133 Suppl, S113–144. [Google Scholar] [CrossRef]
  85. Nussenzweig, R.S.; Vanderberg, J.; Most, H.; Orton, C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature 1967, 216, 160–162. [Google Scholar] [CrossRef] [PubMed]
  86. Dame, J.B.; Williams, J.L.; McCutchan, T.F.; Weber, J.L.; Wirtz, R.A.; Hockmeyer, W.T.; Maloy, W.L.; Haynes, J.D.; Schneider, I.; Roberts, D.; et al. Structure of the gene encoding the immunodominant surface antigen on the sporozoite of the human malaria parasite Plasmodium falciparum. Science 1984, 225, 593–599. [Google Scholar] [CrossRef] [PubMed]
  87. Nussenzweig, V.; Nussenzweig, R.S. Circumsporozoite proteins of malaria parasites. Cell 1985, 42, 401–403. [Google Scholar] [CrossRef]
  88. Nussenzweig, R.S.; Nussenzweig, V. Development of sporozoite vaccines. Philos Trans R Soc Lond B Biol Sci 1984, 307, 117–128. [Google Scholar] [CrossRef]
  89. Ballou, W.R.; Hoffman, S.L.; Sherwood, J.A.; Hollingdale, M.R.; Neva, F.A.; Hockmeyer, W.T.; Gordon, D.M.; Schneider, I.; Wirtz, R.A.; Young, J.F.; et al. Safety and efficacy of a recombinant DNA Plasmodium falciparum sporozoite vaccine. Lancet 1987, 1, 1277–1281. [Google Scholar] [CrossRef]
  90. Stoute, J.A.; Slaoui, M.; Heppner, D.G.; Momin, P.; Kester, K.E.; Desmons, P.; Wellde, B.T.; Garçon, N.; Krzych, U.; Marchand, M. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med 1997, 336, 86–91. [Google Scholar] [CrossRef]
  91. Alonso, P.L.; Sacarlal, J.; Aponte, J.J.; Leach, A.; Macete, E.; Milman, J.; Mandomando, I.; Spiessens, B.; Guinovart, C.; Espasa, M.; et al. Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial. Lancet 2004, 364, 1411–1420. [Google Scholar] [CrossRef]
  92. RTS, S.C.T.P. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet 2015, 386, 31–45. [Google Scholar] [CrossRef]
  93. World Health Organization. Malaria vaccine pilot launched in Malawi. Available online: https://www.who.int/news/item/23-04-2019-malaria-vaccine-pilot-launched-in-malawi (accessed on 3 November 2024).
  94. World Health Organization. WHO recommends groundbreaking malaria vaccine for children at risk. Available online: https://www.who.int/news/item/06-10-2021-who-recommends-groundbreaking-malaria-vaccine-for-children-at-risk (accessed on 3 November 2024).
  95. Natama, H.M.; Salkeld, J.; Somé, A.; Soremekun, S.; Diallo, S.; Traoré, O.; Rouamba, T.; Ouédraogo, F.; Ouédraogo, E.; Daboné, K.C.S.; et al. Safety and efficacy of the blood-stage malaria vaccine RH5.1/Matrix-M in Burkina Faso: interim results of a double-blind, randomised, controlled, phase 2b trial in children. Lancet Infect Dis 2024. [Google Scholar] [CrossRef] [PubMed]
  96. Vanderberg, J.P. Reflections on early malaria vaccine studies, the first successful human malaria vaccination, and beyond. Vaccine 2009, 27, 2–9. [Google Scholar] [CrossRef] [PubMed]
  97. Richie, T.L.; Saul, A. Progress and challenges for malaria vaccines. Nature 2002, 415, 694–701. [Google Scholar] [CrossRef] [PubMed]
  98. Young, J.F.; Hockmeyer, W.T.; Gross, M.; Ballou, W.R.; Wirtz, R.A.; Trosper, J.H.; Beaudoin, R.L.; Hollingdale, M.R.; Miller, L.H.; Diggs, C.L.; et al. Expression of Plasmodium falciparum circumsporozoite proteins in Escherichia coli for potential use in a human malaria vaccine. Science 1985, 228, 958–962. [Google Scholar] [CrossRef]
  99. Molina-Franky, J.; Cuy-Chaparro, L.; Camargo, A.; Reyes, C.; Gómez, M.; Salamanca, D.R.; Patarroyo, M.A.; Patarroyo, M.E. Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J 2020, 19, 56. [Google Scholar] [CrossRef] [PubMed]
  100. Coppi, A.; Natarajan, R.; Pradel, G.; Bennett, B.L.; James, E.R.; Roggero, M.A.; Corradin, G.; Persson, C.; Tewari, R.; Sinnis, P. The malaria circumsporozoite protein has two functional domains, each with distinct roles as sporozoites journey from mosquito to mammalian host. J Exp Med 2011, 208, 341–356. [Google Scholar] [CrossRef] [PubMed]
  101. Campo, J.J.; Aponte, J.J.; Skinner, J.; Nakajima, R.; Molina, D.M.; Liang, L.; Sacarlal, J.; Alonso, P.L.; Crompton, P.D.; Felgner, P.L.; et al. RTS,S vaccination is associated with serologic evidence of decreased exposure to Plasmodium falciparum liver- and blood-stage parasites. Mol Cell Proteomics 2015, 14, 519–531. [Google Scholar] [CrossRef] [PubMed]
  102. Corradin, G.; Céspedes, N.; Verdini, A.; Kajava, A.V.; Arévalo-Herrera, M.; Herrera, S. Chapter 5 - Malaria Vaccine Development Using Synthetic Peptides as a Technical Platform. In Advances in Immunology, Melief, C.J.M., Ed.; Academic Press: 2012; Volume 114, pp. 107–149. [CrossRef]
  103. Roman, F.; Burny, W.; Ceregido, M.A.; Laupèze, B.; Temmerman, S.T.; Warter, L.; Coccia, M. Adjuvant system AS01: from mode of action to effective vaccines. Expert Rev Vaccines 2024, 23, 715–729. [Google Scholar] [CrossRef] [PubMed]
  104. World Health Organization. Full Evidence Report on the RTS,S/AS01 Malaria Vaccine. Available online: https://cdn.who.int/media/docs/default-source/immunization/mvip/full-evidence-report-on-the-rtss-as01-malaria-vaccine-for-sage-mpag- (accessed on day month year).
  105. World Health Organization. RTS,S malaria vaccine reaches more than 650 000 children in Ghana, Kenya and Malawi through groundbreaking pilot programme. Available online: https://www.who.int/news/item/20-04-2021-rts-s-malaria-vaccine-reaches-more-than-650-000-children-in-ghana-kenya-and-malawi-through-groundbreaking-pilot-programme (accessed on 3 November 2024).
  106. Björkman, A.; Benn, C.S.; Aaby, P.; Schapira, A. RTS,S/AS01 malaria vaccine—proven safe and effective? The Lancet Infectious Diseases 2023, 23, e318–e322. [Google Scholar] [CrossRef]
  107. Sinnis, P.; Fidock, D.A. The RTS,S vaccine—a chance to regain the upper hand against malaria? Cell 2022, 185, 750–754. [Google Scholar] [CrossRef]
  108. Aderinto, N.; Olatunji, G.; Kokori, E.; Sikirullahi, S.; Aboje, J.E.; Ojabo, R.E. A perspective on Oxford’s R21/Matrix-M™ malaria vaccine and the future of global eradication efforts. Malaria Journal 2024, 23, 16. [Google Scholar] [CrossRef] [PubMed]
  109. Datoo, M.S.; Natama, M.H.; Somé, A.; Traoré, O.; Rouamba, T.; Bellamy, D.; Yameogo, P.; Valia, D.; Tegneri, M.; Ouedraogo, F.; et al. Efficacy of a low-dose candidate malaria vaccine, R21 in adjuvant Matrix-M, with seasonal administration to children in Burkina Faso: a randomised controlled trial. Lancet 2021, 397, 1809–1818. [Google Scholar] [CrossRef]
  110. Datoo, M.S.; Dicko, A.; Tinto, H.; Ouédraogo, J.B.; Hamaluba, M.; Olotu, A.; Beaumont, E.; Ramos Lopez, F.; Natama, H.M.; Weston, S.; et al. Safety and efficacy of malaria vaccine candidate R21/Matrix-M in African children: a multicentre, double-blind, randomised, phase 3 trial. Lancet 2024, 403, 533–544. [Google Scholar] [CrossRef] [PubMed]
  111. Datoo, M.S.; Natama, H.M.; Somé, A.; Bellamy, D.; Traoré, O.; Rouamba, T.; Tahita, M.C.; Ido, N.F.A.; Yameogo, P.; Valia, D.; et al. Efficacy and immunogenicity of R21/Matrix-M vaccine against clinical malaria after 2 years' follow-up in children in Burkina Faso: a phase 1/2b randomised controlled trial. Lancet Infect Dis 2022, 22, 1728–1736. [Google Scholar] [CrossRef]
  112. Mahase, E. WHO recommends second vaccine for malaria prevention in children. BMJ 2023, 383, p2291. [Google Scholar] [CrossRef]
  113. Genton, B. R21/Matrix-M™ malaria vaccine: a new tool to achieve WHO's goal to eliminate malaria in 30 countries by 2030? J Travel Med 2023, 30. [Google Scholar] [CrossRef] [PubMed]
  114. Palacpac, N.M.Q.; Horii, T. RH5.1/Matrix-M: highlighting blood-stage malaria vaccines. Lancet Infect Dis 2024. [Google Scholar] [CrossRef] [PubMed]
  115. Silk, S.E.; Kalinga, W.F.; Salkeld, J.; Mtaka, I.M.; Ahmed, S.; Milando, F.; Diouf, A.; Bundi, C.K.; Balige, N.; Hassan, O.; et al. Blood-stage malaria vaccine candidate RH5.1/Matrix-M in healthy Tanzanian adults and children; an open-label, non-randomised, first-in-human, single-centre, phase 1b trial. Lancet Infect Dis 2024, 24, 1105–1117. [Google Scholar] [CrossRef] [PubMed]
  116. King, L.D.W.; Pulido, D.; Barrett, J.R.; Davies, H.; Quinkert, D.; Lias, A.M.; Silk, S.E.; Pattinson, D.J.; Diouf, A.; Williams, B.G.; et al. Preclinical development of a stabilized RH5 virus-like particle vaccine that induces improved antimalarial antibodies. Cell Rep Med 2024, 5, 101654. [Google Scholar] [CrossRef]
  117. Douglas, A.D.; Williams, A.R.; Illingworth, J.J.; Kamuyu, G.; Biswas, S.; Goodman, A.L.; Wyllie, D.H.; Crosnier, C.; Miura, K.; Wright, G.J.; et al. The blood-stage malaria antigen PfRH5 is susceptible to vaccine-inducible cross-strain neutralizing antibody. Nat Commun 2011, 2, 601. [Google Scholar] [CrossRef] [PubMed]
  118. Ragotte, R.J.; Higgins, M.K.; Draper, S.J. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol 2020, 36, 545–559. [Google Scholar] [CrossRef]
  119. Laurenson, A.J.; Laurens, M.B. A new landscape for malaria vaccine development. PLoS Pathog 2024, 20, e1012309. [Google Scholar] [CrossRef] [PubMed]
  120. Kanoi, B.N.; Maina, M.; Likhovole, C.; Kobia, F.M.; Gitaka, J. Malaria vaccine approaches leveraging technologies optimized in the COVID-19 era. Frontiers in Tropical Diseases 2022, 3, 988665. [Google Scholar] [CrossRef]
  121. Ewer, K.J.; Sierra-Davidson, K.; Salman, A.M.; Illingworth, J.J.; Draper, S.J.; Biswas, S.; Hill, A.V.S. Progress with viral vectored malaria vaccines: A multi-stage approach involving “unnatural immunity”. Vaccine 2015, 33, 7444–7451. [Google Scholar] [CrossRef]
  122. Reyes-Sandoval, A.; Harty, J.T.; Todryk, S.M. Viral vector vaccines make memory T cells against malaria. Immunology 2007, 121, 158–165. [Google Scholar] [CrossRef]
  123. Milicic, A.; Rollier, C.S.; Tang, C.K.; Longley, R.; Hill, A.V.S.; Reyes-Sandoval, A. Adjuvanting a viral vectored vaccine against pre-erythrocytic malaria. Scientific Reports 2017, 7, 7284. [Google Scholar] [CrossRef]
  124. Chaudhary, N.; Weissman, D.; Whitehead, K.A. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat Rev Drug Discov 2021, 20, 817–838. [Google Scholar] [CrossRef]
  125. Scaria, P.V.; Roth, N.; Schwendt, K.; Muratova, O.V.; Alani, N.; Lambert, L.E.; Barnafo, E.K.; Rowe, C.G.; Zaidi, I.U.; Rausch, K.M.; et al. mRNA vaccines expressing malaria transmission-blocking antigens Pfs25 and Pfs230D1 induce a functional immune response. npj Vaccines 2024, 9, 9. [Google Scholar] [CrossRef]
  126. Makoni, M. mRNA vaccine against malaria effective in preclinical model. Lancet Microbe 2023, 4, e970. [Google Scholar] [CrossRef]
  127. Laurens, M.B. Novel malaria vaccines. Hum Vaccin Immunother 2021, 17, 4549–4552. [Google Scholar] [CrossRef] [PubMed]
  128. Al-obeidee, M.; Al-obeidee, E. A new era in malaria prevention: a comparative look at RTS,S/AS01 and R21/Matrix-M vaccines. Postgraduate Medical Journal 2024, 100, 877–878. [Google Scholar] [CrossRef] [PubMed]
  129. Ogieuhi, I.J.; Ajekiigbe, V.O.; Kolo-Manma, K.; Akingbola, A.; Odeniyi, T.A.; Soyemi, T.S.; Ayomide, J.H.; Thiyagarajan, B.; Awolola, B.D. A narrative review of the RTS S AS01 malaria vaccine and its implementation in Africa to reduce the global malaria burden. Discover Public Health 2024, 21, 152. [Google Scholar] [CrossRef]
  130. Adjei, M.R.; Okine, R.; Tweneboah, P.O.; Baafi, J.V.; Afriyie, N.A.; Teviu, E.A.A.; Nyuzaghl, J.A.-I.; Dzotsi, E.K.; Ohene, S.-A.; Grobusch, M.P. The impact of the RTS,S malaria vaccine on uncomplicated malaria: evidence from the phase IV study districts, Upper East Region, Ghana, 2020–2022. Malaria Journal 2024, 23, 305. [Google Scholar] [CrossRef] [PubMed]
  131. Penny, M.A.; Galactionova, K.; Tarantino, M.; Tanner, M.; Smith, T.A. 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. [Google Scholar] [CrossRef]
  132. White, M.T.; Verity, R.; Griffin, J.T.; Asante, K.P.; Owusu-Agyei, S.; Greenwood, B.; Drakeley, C.; Gesase, S.; Lusingu, J.; Ansong, D.; et al. Immunogenicity of the RTS,S/AS01 malaria vaccine and implications for duration of vaccine efficacy: secondary analysis of data from a phase 3 randomised controlled trial. Lancet Infect Dis 2015, 15, 1450–1458. [Google Scholar] [CrossRef]
  133. Samuels, A.M.; Ansong, D.; Kariuki, S.K.; Adjei, S.; Bollaerts, A.; Ockenhouse, C.; Westercamp, N.; Lee, C.K.; Schuerman, L.; Bii, D.K.; et al. Efficacy of RTS,S/AS01(E) malaria vaccine administered according to different full, fractional, and delayed third or early fourth dose regimens in children aged 5-17 months in Ghana and Kenya: an open-label, phase 2b, randomised controlled trial. Lancet Infect Dis 2022, 22, 1329–1342. [Google Scholar] [CrossRef]
  134. Sánchez, L.; Vidal, M.; Jairoce, C.; Aguilar, R.; Ubillos, I.; Cuamba, I.; Nhabomba, A.J.; Williams, N.A.; Díez-Padrisa, N.; Cavanagh, D.; et al. Antibody responses to the RTS,S/AS01E vaccine and Plasmodium falciparum antigens after a booster dose within the phase 3 trial in Mozambique. npj Vaccines 2020, 5, 46. [Google Scholar] [CrossRef]
  135. Neafsey, D.E.; Juraska, M.; Bedford, T.; Benkeser, D.; Valim, C.; Griggs, A.; Lievens, M.; Abdulla, S.; Adjei, S.; Agbenyega, T.; et al. Genetic Diversity and Protective Efficacy of the RTS,S/AS01 Malaria Vaccine. N Engl J Med 2015, 373, 2025–2037. [Google Scholar] [CrossRef] [PubMed]
  136. Yihunie, W.; Kebede, B.; Tegegne, B.A.; Getachew, M.; Abebe, D.; Aschale, Y.; Belew, H.; Bahiru, B. Systematic Review of Safety of RTS,S with AS01 and AS02 Adjuvant Systems Using Data from Randomized Controlled Trials in Infants, Children, and Adults. Clin Pharmacol 2023, 15, 21–32. [Google Scholar] [CrossRef] [PubMed]
  137. Chutiyami, M.; Saravanakumar, P.; Bello, U.M.; Salihu, D.; Adeleye, K.; Kolo, M.A.; Dawa, K.K.; Hamina, D.; Bhandari, P.; Sulaiman, S.K.; et al. Malaria vaccine efficacy, safety, and community perception in Africa: a scoping review of recent empirical studies. Infection 2024, 52, 2007–2028. [Google Scholar] [CrossRef]
  138. Guerra Mendoza, Y.; Garric, E.; Leach, A.; Lievens, M.; Ofori-Anyinam, O.; Pirçon, J.Y.; Stegmann, J.U.; Vandoolaeghe, P.; Otieno, L.; Otieno, W.; et al. Safety profile of the RTS,S/AS01 malaria vaccine in infants and children: additional data from a phase III randomized controlled trial in sub-Saharan Africa. Hum Vaccin Immunother 2019, 15, 2386–2398. [Google Scholar] [CrossRef]
  139. Asante, K.P.; Ansong, D.; Kaali, S.; Adjei, S.; Lievens, M.; Nana Badu, L.; Agyapong Darko, P.; Boakye Yiadom Buabeng, P.; Boahen, O.; Maria Rettig, T.; et al. Immunogenicity and safety of the RTS,S/AS01 malaria vaccine co-administered with measles, rubella and yellow fever vaccines in Ghanaian children: A phase IIIb, multi-center, non-inferiority, randomized, open, controlled trial. Vaccine 2020, 38, 3411–3421. [Google Scholar] [CrossRef]
  140. Mahase, E. Ghana approves Oxford's malaria vaccine for children aged 5 to 36 months. Bmj 2023, 381, 850. [Google Scholar] [CrossRef] [PubMed]
  141. Hammershaimb, E.A.; Berry, A.A. Pre-erythrocytic malaria vaccines: RTS,S, R21, and beyond. Expert Rev Vaccines 2024, 23, 49–52. [Google Scholar] [CrossRef] [PubMed]
  142. Schmit, N.; Topazian, H.M.; Natama, H.M.; Bellamy, D.; Traoré, O.; Somé, M.A.; Rouamba, T.; Tahita, M.C.; Bonko, M.D.A.; Sourabié, A.; et al. The public health impact and cost-effectiveness of the R21/Matrix-M malaria vaccine: a mathematical modelling study. Lancet Infect Dis 2024, 24, 465–475. [Google Scholar] [CrossRef] [PubMed]
  143. Seo, M.K.; Baker, P.; Ngo, K.N. Cost-effectiveness analysis of vaccinating children in Malawi with RTS,S vaccines in comparison with long-lasting insecticide-treated nets. Malar J 2014, 13, 66. [Google Scholar] [CrossRef] [PubMed]
  144. Thellier, M.; Gemegah, A.A.J.; Tantaoui, I. Global Fight against Malaria: Goals and Achievements 1900-2022. J Clin Med 2024, 13, 5680. [Google Scholar] [CrossRef] [PubMed]
  145. Matola, Y.; Chumbi, G.; Moyo, C.S.; Bwanali, A.; Lubanga, A.F. Beyond RTS,S malaria vaccine piloting to adoption and historic introduction in sub-Saharan Africa: a new hope in the fight against the vector-borne disease. Frontiers in Tropical Diseases 2024, 5, 1387078. [Google Scholar] [CrossRef]
  146. Beeson, J.G.; Kurtovic, L.; Dobaño, C.; Opi, D.H.; Chan, J.A.; Feng, G.; Good, M.F.; Reiling, L.; Boyle, M.J. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med 2019, 11. [Google Scholar] [CrossRef]
  147. Okesanya, O.J.; Atewologun, F.; Lucero-Prisno, D.E.; Adigun, O.A.; Oso, T.A.; Manirambona, E.; Olabode, N.O.; Eshun, G.; Agboola, A.O.; Okon, I.I. Bridging the gap to malaria vaccination in Africa: Challenges and opportunities. Journal of Medicine, Surgery, and Public Health 2024, 2, 100059. [Google Scholar] [CrossRef]
  148. Hill, J.; Bange, T.; Hoyt, J.; Kariuki, S.; Jalloh, M.F.; Webster, J.; Okello, G. Integration of the RTS,S/AS01 malaria vaccine into the Essential Programme on Immunisation in western Kenya: a qualitative longitudinal study from the health system perspective. Lancet Glob Health 2024, 12, e672–e684. [Google Scholar] [CrossRef]
  149. Grant, J.; Gyan, T.; Agbokey, F.; Webster, J.; Greenwood, B.; Asante, K.P. Challenges and lessons learned during the planning and early implementation of the RTS,S/AS01E malaria vaccine in three regions of Ghana: a qualitative study. Malaria Journal 2022, 21, 147. [Google Scholar] [CrossRef]
  150. Amimo, F. Malaria vaccination: hurdles to reach high-risk children. BMC Med 2024, 22, 111. [Google Scholar] [CrossRef]
  151. Adjei, M.R.; Amponsa-Achiano, K.; Okine, R.; Tweneboah, P.O.; Sally, E.T.; Dadzie, J.F.; Osei-Sarpong, F.; Adjabeng, M.J.; Bawa, J.T.; Bonsu, G.; et al. Post introduction evaluation of the malaria vaccine implementation programme in Ghana, 2021. BMC Public Health 2023, 23, 586. [Google Scholar] [CrossRef]
  152. Fortpied, J.; Collignon, S.; Moniotte, N.; Renaud, F.; Bayat, B.; Lemoine, D. The thermostability of the RTS,S/AS01 malaria vaccine can be increased by co-lyophilizing RTS,S and AS01. Malaria Journal 2020, 19, 202. [Google Scholar] [CrossRef]
  153. Adeshina, O.O.; Nyame, S.; Milner, J.; Milojevic, A.; Asante, K.P. Barriers and facilitators to nationwide implementation of the malaria vaccine in Ghana. Health Policy Plan 2023, 38, 28–37. [Google Scholar] [CrossRef]
  154. Nkwenkeu, S.F.; Jalloh, M.F.; Walldorf, J.A.; Zoma, R.L.; Tarbangdo, F.; Fall, S.; Hien, S.; Combassere, R.; Ky, C.; Kambou, L.; et al. Health workers' perceptions and challenges in implementing meningococcal serogroup a conjugate vaccine in the routine childhood immunization schedule in Burkina Faso. BMC Public Health 2020, 20, 254. [Google Scholar] [CrossRef] [PubMed]
  155. Olawade, D.B.; Wada, O.Z.; Ezeagu, C.N.; Aderinto, N.; Balogun, M.A.; Asaolu, F.T.; David-Olawade, A.C. Malaria vaccination in Africa: A mini-review of challenges and opportunities. Medicine (Baltimore) 2024, 103, e38565. [Google Scholar] [CrossRef]
  156. World Health Organization. Tackling malaria in countries hardest hit by the disease: ministerial conference report, Yaoundé, Cameroon, 6 March 2024. Available online: https://www.who.int/publications/i/item/9789240100459 (accessed on 5 November 2024).
  157. Adamu, A.A.; Jalo, R.I.; Ndwandwe, D.; Wiysonge, C.S. Assessing the Implementation Determinants of Pilot Malaria Vaccination Programs in Ghana, Kenya, and Malawi through a Complexity Lens: A Rapid Review Using a Consolidated Framework for Implementation Research. Vaccines (Basel) 2024, 12, 111. [Google Scholar] [CrossRef]
  158. Zarocostas, J. Gavi unveils malaria vaccine plans. Lancet 2023, 401, 1485. [Google Scholar] [CrossRef]
  159. Okumu, F.; Gyapong, M.; Casamitjana, N.; Castro, M.C.; Itoe, M.A.; Okonofua, F.; Tanner, M. What Africa can do to accelerate and sustain progress against malaria. PLOS Glob Public Health 2022, 2, e0000262. [Google Scholar] [CrossRef]
  160. Sulaiman, S.K.; Musa, M.S.; Tsiga-Ahmed, F.I.; Dayyab, F.M.; Sulaiman, A.K.; Bako, A.T. A systematic review and meta-analysis of the prevalence of caregiver acceptance of malaria vaccine for under-five children in low-income and middle-income countries (LMICs). PLoS One 2022, 17, e0278224. [Google Scholar] [CrossRef]
  161. Aremu, T.O.; Ajibola, O.A.; Oluwole, O.E.; Adeyinka, K.O.; Dada, S.O.; Okoro, O.N. Looking Beyond the Malaria Vaccine Approval to Acceptance and Adoption in Sub-Saharan Africa. Frontiers in Tropical Diseases 2022, 3, 857844. [Google Scholar] [CrossRef]
  162. Kigongo, E.; Kabunga, A.; Opollo, M.S.; Tumwesigye, R.; Musinguzi, M.; Akello, A.R.; Nabaziwa, J.; Hardido, T.G.; Puleh, S.S. Community readiness and acceptance for the implementation of a novel malaria vaccine among at-risk children in sub-saharan Africa: a systematic review protocol. Malar J 2024, 23, 182. [Google Scholar] [CrossRef]
  163. Mumtaz, H.; Nadeem, A.; Bilal, W.; Ansar, F.; Saleem, S.; Khan, Q.A.; Tango, T.; Farkouh, C.; Belay, N.F.; Verma, R.; et al. Acceptance, availability, and feasibility of RTS, S/AS01 malaria vaccine: A review. Immun Inflamm Dis 2023, 11, e899. [Google Scholar] [CrossRef]
  164. Bam, V.; Mohammed, A.; Kusi-Amponsah, A.; Armah, J.; Lomotey, A.Y.; Budu, H.I.; Atta Poku, C.; Kyei-Dompim, J.; Dwumfour, C. Caregivers' perception and acceptance of malaria vaccine for Children. PLoS One 2023, 18, e0288686. [Google Scholar] [CrossRef] [PubMed]
  165. Ojakaa, D.I.; Jarvis, J.D.; Matilu, M.I.; Thiam, S. Acceptance of a malaria vaccine by caregivers of sick children in Kenya. Malar J 2014, 13, 172. [Google Scholar] [CrossRef] [PubMed]
  166. Nyalundja, A.D.; Bugeme, P.M.; Guillaume, A.S.; Ntaboba, A.B.; Hatu'm, V.U.; Tamuzi, J.L.; Ndwandwe, D.; Iwu-Jaja, C.; Wiysonge, C.S.; Katoto, P. Socio-Demographic Factors Influencing Malaria Vaccine Acceptance for Under-Five Children in a Malaria-Endemic Region: A Community-Based Study in the Democratic Republic of Congo. Vaccines (Basel) 2024, 12. [Google Scholar] [CrossRef]
  167. Mtenga, S.; Kimweri, A.; Romore, I.; Ali, A.; Exavery, A.; Sicuri, E.; Tanner, M.; Abdulla, S.; Lusingu, J.; Kafuruki, S. Stakeholders' opinions and questions regarding the anticipated malaria vaccine in Tanzania. Malar J 2016, 15, 189. [Google Scholar] [CrossRef]
  168. Amin, M.A.; Afrin, S.; Bonna, A.S.; Rozars, M.F.K.; Nabi, M.H.; Hawlader, M.D.H. Knowledge and acceptance of malaria vaccine among parents of under-five children of malaria endemic areas in Bangladesh: A cross-sectional study. Health Expect 2023, 26, 2630–2643. [Google Scholar] [CrossRef]
  169. Ansar, F.; Azzam, A.; Rauf, M.S.; Ajmal, Z.; Asad Ullah, G.; Rauf, S.; Akram, R.; Ghauri, F.K.; Chudhary, F.; Iftikhar, H.; et al. Global Analysis of RTS, S/AS01 Malaria Vaccine Acceptance Rates and Influencing Factors: A Systematic Review. Cureus 2024, 16, e60678. [Google Scholar] [CrossRef]
  170. editorial - ClinicalMedicine. Malaria: (still) a global health priority. EClinicalMedicine 2021, 34, 100891. [Google Scholar] [CrossRef]
  171. Chutiyami, M. Recent Trends in Malaria Vaccine Research Globally: A Bibliometric Analysis From 2005 to 2022. J Parasitol Res 2024, 2024, 8201097. [Google Scholar] [CrossRef]
  172. Miura, K.; Flores-Garcia, Y.; Long, C.A.; Zavala, F. Vaccines and monoclonal antibodies: new tools for malaria control. Clin Microbiol Rev 2024, 37, e0007123. [Google Scholar] [CrossRef]
  173. Locke, E.; Flores-Garcia, Y.; Mayer, B.T.; MacGill, R.S.; Borate, B.; Salgado-Jimenez, B.; Gerber, M.W.; Mathis-Torres, S.; Shapiro, S.; King, C.R.; et al. Establishing RTS,S/AS01 as a benchmark for comparison to next-generation malaria vaccines in a mouse model. npj Vaccines 2024, 9, 29. [Google Scholar] [CrossRef] [PubMed]
  174. Draper, S.J.; Sack, B.K.; King, C.R.; Nielsen, C.M.; Rayner, J.C.; Higgins, M.K.; Long, C.A.; Seder, R.A. Malaria Vaccines: Recent Advances and New Horizons. Cell Host Microbe 2018, 24, 43–56. [Google Scholar] [CrossRef]
  175. Mueller, I.; Shakri, A.R.; Chitnis, C.E. Development of vaccines for Plasmodium vivax malaria. Vaccine 2015, 33, 7489–7495. [Google Scholar] [CrossRef]
  176. Price, R.N.; Commons, R.J.; Battle, K.E.; Thriemer, K.; Mendis, K. Plasmodium vivax in the Era of the Shrinking P. falciparum Map. Trends Parasitol 2020, 36, 560–570. [Google Scholar] [CrossRef] [PubMed]
  177. Khan, N.; Daily, J.P. Update on pathogenesis, management, and control of Plasmodium vivax. Curr Opin Infect Dis 2022, 35, 404–409. [Google Scholar] [CrossRef] [PubMed]
  178. da Veiga, G.T.S.; Moriggi, M.R.; Vettorazzi, J.F.; Müller-Santos, M.; Albrecht, L. Plasmodium vivax vaccine: What is the best way to go? Front Immunol 2022, 13, 910236. [Google Scholar] [CrossRef]
  179. Habtamu, K.; Petros, B.; Yan, G. Plasmodium vivax: the potential obstacles it presents to malaria elimination and eradication. Tropical Diseases, Travel Medicine and Vaccines 2022, 8, 27. [Google Scholar] [CrossRef]
  180. Tajudeen, Y.A.; Oladipo, H.J.; Yusuff, S.I.; Abimbola, S.O.; Abdulkadir, M.; Oladunjoye, I.O.; Omotosho, A.O.; Egbewande, O.M.; Shittu, H.D.; Yusuf, R.O.; et al. A landscape review of malaria vaccine candidates in the pipeline. Tropical Diseases, Travel Medicine and Vaccines 2024, 10, 19. [Google Scholar] [CrossRef] [PubMed]
  181. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines — a new era in vaccinology. Nature Reviews Drug Discovery 2018, 17, 261–279. [Google Scholar] [CrossRef]
  182. Patel, P.N.; Tolia, N. Structural vaccinology of malaria transmission-blocking vaccines. Expert Rev Vaccines 2021, 20, 199–214. [Google Scholar] [CrossRef] [PubMed]
  183. Hayashi, C.T.H.; Cao, Y.; Clark, L.C.; Tripathi, A.K.; Zavala, F.; Dwivedi, G.; Knox, J.; Alameh, M.G.; Lin, P.J.C.; Tam, Y.K.; et al. mRNA-LNP expressing PfCSP and Pfs25 vaccine candidates targeting infection and transmission of Plasmodium falciparum. NPJ Vaccines 2022, 7, 155. [Google Scholar] [CrossRef] [PubMed]
  184. Amimo, F. Leveraging malaria vaccines and mRNA technology to tackle the global inequity in pharmaceutical research and production towards disease elimination. Malar J 2024, 23, 136. [Google Scholar] [CrossRef] [PubMed]
  185. Musoke, D.; Atusingwize, E.; Namata, C.; Ndejjo, R.; Wanyenze, R.K.; Kamya, M.R. Integrated malaria prevention in low- and middle-income countries: a systematic review. Malaria Journal 2023, 22, 79. [Google Scholar] [CrossRef] [PubMed]
  186. von Seidlein, L.; Hanboonkunupakarn, B.; Jittamala, P.; Pongsuwan, P.; Chotivanich, K.; Tarning, J.; Hoglund, R.M.; Winterberg, M.; Mukaka, M.; Peerawaranun, P.; et al. Combining antimalarial drugs and vaccine for malaria elimination campaigns: a randomized safety and immunogenicity trial of RTS,S/AS01 administered with dihydroartemisinin, piperaquine, and primaquine in healthy Thai adult volunteers. Hum Vaccin Immunother 2020, 16, 33–41. [Google Scholar] [CrossRef] [PubMed]
  187. Duffy, P.E.; Gorres, J.P.; Healy, S.A.; Fried, M. Malaria vaccines: a new era of prevention and control. Nature Reviews Microbiology 2024. [Google Scholar] [CrossRef] [PubMed]
  188. González-Sanz, M.; Berzosa, P.; Norman, F.F. Updates on Malaria Epidemiology and Prevention Strategies. Curr Infect Dis Rep 2023, 1–9. [Google Scholar] [CrossRef] [PubMed]
  189. Tine, R.; Herrera, S.; Badji, M.A.; Daniels, K.; Ndiaye, P.; Smith Gueye, C.; Tairou, F.; Slutsker, L.; Hwang, J.; Ansah, E.; et al. Defining operational research priorities to improve malaria control and elimination in sub-Saharan Africa: results from a country-driven research prioritization setting process. Malar J 2023, 22, 219. [Google Scholar] [CrossRef] [PubMed]
  190. World Health Organization. Global Malaria Programme operational strategy 2024-2030. Available online: https://www.who.int/publications/i/item/9789240090149 (accessed on 5 November 2024).
  191. Ward, C.L.; Shaw, D.; Anane-Sarpong, E.; Sankoh, O.; Tanner, M.; Elger, B. The Ethics of Health Care Delivery in a Pediatric Malaria Vaccine Trial: The Perspectives of Stakeholders From Ghana and Tanzania. J Empir Res Hum Res Ethics 2018, 13, 26–41. [Google Scholar] [CrossRef] [PubMed]
  192. Nunes, C.; McKee, M.; Howard, N. The role of global health partnerships in vaccine equity: A scoping review. PLOS Glob Public Health 2024, 4, e0002834. [Google Scholar] [CrossRef] [PubMed]
  193. Osoro, C.B.; Ochodo, E.; Kwambai, T.K.; Otieno, J.A.; Were, L.; Sagam, C.K.; Owino, E.J.; Kariuki, S.; Ter Kuile, F.O.; Hill, J. Policy uptake and implementation of the RTS,S/AS01 malaria vaccine in sub-Saharan African countries: status 2 years following the WHO recommendation. BMJ Glob Health 2024, 9. [Google Scholar] [CrossRef]
  194. Chitnis, C.E.; Schellenberg, D.; Vekemans, J.; Asturias, E.J.; Bejon, P.; Collins, K.A.; Crabb, B.S.; Herrera, S.; Laufer, M.; Rabinovich, N.R.; et al. Building momentum for malaria vaccine research and development: key considerations. Malaria Journal 2020, 19, 421. [Google Scholar] [CrossRef]
  195. Greenwood, B. New tools for malaria control - using them wisely. J Infect 2017, 74 Suppl 1, S23–s26. [Google Scholar] [CrossRef]
  196. Wangdi, K.; Banwell, C.; Gatton, M.L.; Kelly, G.C.; Namgay, R.; Clements, A.C. Malaria burden and costs of intensified control in Bhutan, 2006-14: an observational study and situation analysis. Lancet Glob Health 2016, 4, e336–343. [Google Scholar] [CrossRef]
  197. Healer, J.; Cowman, A.F.; Kaslow, D.C.; Birkett, A.J. Vaccines to Accelerate Malaria Elimination and Eventual Eradication. Cold Spring Harb Perspect Med 2017, 7. [Google Scholar] [CrossRef] [PubMed]
  198. Dev, V.; Wangdi, K. Editorial: World Malaria Day 2023 - ending malaria transmission: reaching the last mile to zero malaria. Front Public Health 2024, 12, 1433213. [Google Scholar] [CrossRef] [PubMed]
  199. Amani, A.; Mboussou, F.; Impouma, B.; Cabore, J.; Moeti, M.R. Introduction and rollout of malaria vaccines in Cameroon and Burkina Faso: early lessons learned. Lancet Glob Health 2024, 12, e740–e741. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Global distribution of malaria incidence and mortality rates. (A) Estimated malaria incidence per 100,000 population at risk in 2022. (B) Estimated malaria mortality rate per 100,000 population in 2021. Data source: World Health Organization Global Health Observatory, 2022 [80]. The maps were generated in Microsoft Excel, powered by Bing, using geospatial data from the Australian Bureau of Statistics, GeoNames, Microsoft, Navinfo, TomTom, and Wikipedia. We remain neutral regarding jurisdictional claims depicted in the maps.
Figure 1. Global distribution of malaria incidence and mortality rates. (A) Estimated malaria incidence per 100,000 population at risk in 2022. (B) Estimated malaria mortality rate per 100,000 population in 2021. Data source: World Health Organization Global Health Observatory, 2022 [80]. The maps were generated in Microsoft Excel, powered by Bing, using geospatial data from the Australian Bureau of Statistics, GeoNames, Microsoft, Navinfo, TomTom, and Wikipedia. We remain neutral regarding jurisdictional claims depicted in the maps.
Preprints 143031 g001
Figure 2. Timeline of major milestones in malaria vaccine development. Sources of data are in [56,85,86,87,88,89,90,91,92,93,94,95].
Figure 2. Timeline of major milestones in malaria vaccine development. Sources of data are in [56,85,86,87,88,89,90,91,92,93,94,95].
Preprints 143031 g002
Figure 3. Key challenges in implementing malaria vaccination in resource-limited settings.
Figure 3. Key challenges in implementing malaria vaccination in resource-limited settings.
Preprints 143031 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated