Back to Journals » Clinical Pharmacology: Advances and Applications » Volume 17

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

Authors Sallam M , Al-Khatib AO, Al-Mahzoum KS, Abdelaziz DH, Sallam M

Received 19 December 2024

Accepted for publication 25 March 2025

Published 1 April 2025 Volume 2025:17 Pages 29—47

DOI https://doi.org/10.2147/CPAA.S513282

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Arthur E. Frankel



Malik Sallam,1– 3 Arwa Omar Al-Khatib,4 Kholoud Sultan Al-Mahzoum,5 Doaa H Abdelaziz,6,7 Mohammed Sallam8

1Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan; 2Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan; 3Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden; 4Faculty of Pharmacy, Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; 5Sheikh Jaber Al-Ahmad Al-Sabah Hospital, Ministry of Health, Kuwait City, Kuwait; 6Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia; 7Department of Clinical Pharmacy, the National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt; 8Department of Pharmacy, Mediclinic Parkview Hospital, Mediclinic Middle East, Dubai, United Arab Emirates

Correspondence: Malik Sallam, Email [email protected]

Introduction: Malaria remains a persistent challenge in global health, disproportionately affecting populations in endemic regions (eg, sub-Saharan Africa). Despite decades of international collaborative efforts, malaria continues to claim hundreds of thousands of lives each year, with young children and pregnant women enduring the heaviest burden. This concise review aimed to provide an up-to-date assessment of malaria vaccines progress, challenges, and future directions.
Methods: A PubMed/MEDLINE search (2015– 2024) was conducted to identify studies on malaria vaccine development, implementation barriers, efficacy, and vaccination hesitancy. Clinical trials, reviews, and global health reports were included based on relevance to the review aims. No strict inclusion criteria were applied, and selection was guided by key review themes and policy relevance.
Results: 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. The review identified 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. Promising developments in malaria vaccination, such as next-generation candidates (eg, mRNA-based vaccines), hold the potential to offer improved efficacy, longer-lasting protection, and greater scalability. There is a critical need to integrate malaria vaccination efforts with established malaria control interventions (eg, insecticide-treated bed nets, vector control strategies, and anti-malarial drugs).
Conclusion: 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: malaria control, malaria vaccine, Plasmodium falciparum, vaccine efficacy, immunization programs, public health

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 the availability of malaria rapid diagnostic tests (RDTs).6–8 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 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.15–18

In Africa, malaria control efforts face notable challenges.19–21 These challenges include climate change which shifts transmission dynamics,22,23 poverty which weakens healthcare infrastructure,24 and substandard services in healthcare hindering effective control measures.19 The growing resistance to anti-malarial drugs and insecticides, coupled with the emergence of new vector species and increased outdoor transmission are additional challenges.25–29 Beyond Africa, malaria persists as a serious public health threat. In South America, the Amazon basin remains a focal point, where P. vivax predominates and complicates treatment due to its relapse-prone biology.30–32 Meanwhile, in Southeast Asia’s near-elimination settings, progress is impeded by several challenges.33,34 These include asymptomatic infections, cross-border malaria transmission, resistance to anti-malarial drugs, and the difficulty of pinpointing infection hotspots in remote regions.34–36 These regional complexities highlight the need for innovative surveillance systems, adaptive intervention strategies, and global cooperation to achieve sustained malaria control.

While malaria incidence has declined over the last few decades, progress in malaria prevention and control is still needed.37–39 Challenges in sustaining effective malaria control measures have been exacerbated by the emergence of Plasmodium resistance to various anti-malarial drugs.40–43 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.44

It is also important to point out that the socio-economic toll of malaria remains profound.45,46 This malaria toll perpetuates poverty by disrupting education, workforce productivity, and economic stability, especially in regions with fragile healthcare systems.47–49 Further complicating these challenges that face malaria control is the rising insecticide resistance among the mosquito vector.50,51 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.12,52,53

The quest for an effective malaria vaccine has spanned more than seven decades.54,55 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.56,57 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.58 In both scenarios, coverage was assumed to reach 75% of the diphtheria-tetanus-pertussis (DTP3) vaccination coverage, accounting for the greater difficulty in reaching children at older ages compared to the standard DTP schedule.58 However, the complex life cycle of Plasmodium, involving both human and mosquito hosts, has posed unique challenges for malaria vaccine development.59–61

Early malaria vaccine strategies targeted various stages of the Plasmodium life cycle, including the pre-erythrocytic, blood-stage, and transmission phases.62–67 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).68,69 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.70–72 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.73 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 by incorporating a higher antigen-to-adjuvant ratio to potentially enhance immune response.74–77 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.78–81 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).82–85

Viral-vector malaria vaccines have shown potential in targeting both pre-erythrocytic and sexual stages of P. falciparum.78 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.86,87 Transmission-blocking vaccines (TBVs) using Pfs25-IMX313 demonstrated safety and immunogenicity but had limited transmission-reducing activity, emphasizing the need for further optimization.87,88

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 outline the critical historical milestones and challenges in malaria vaccine development. Third, the review aimed to evaluate the barriers to 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.

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.

Overview of Malaria Burden Worldwide

Malaria remains a formidable global health challenge.19,89,90 This challenge is particularly pronounced in the tropical and subtropical regions, where it disproportionately affects vulnerable populations and perpetuates cycles of poverty.11,19,91 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.92–94 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).95 This system reflects that parasite density decreases significantly between ages 2 and 5, impacting endemicity assessments.95 Despite its limitations, this classification remains a useful framework for prioritizing interventions in are areas with high burden of malaria.92,95,96

The heaviest burden of malaria is observed in SSA, accounting for approximately 95% of malaria cases and deaths globally.19 In 2022, the WHO African region reported an estimated 233 million cases and 580,000 deaths due to malaria.97 Children under five years of age are particularly vulnerable, representing about 80% of all malaria deaths in the African region.98 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).

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, 2022.99 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.

Historical Development of Malaria Vaccines

The quest for a malaria vaccine has been one of the most complex and enduring challenges in tropical medicine.100,101 Unlike many other infectious diseases, malaria is caused by a eukaryotic parasite with a highly complex life cycle involving mosquito and human hosts.102 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.54,59,103 A timeline of the major milestones in malaria vaccine development is illustrated in (Figure 2).

Figure 2 Timeline of major milestones in malaria vaccine development. Sources of data are in.76,104–114

Figure 3 Key challenges in implementing malaria vaccination in resource-limited settings.

Initial attempts to develop a malaria vaccine in the mid-20th century primarily focused on the erythrocytic stage where Plasmodium infects erythrocytes.115 However, these early malaria vaccines faced significant limitations, as they failed to generate strong or lasting immune responses.78,116 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.104,117 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.118–120

The development and approval of RTS,S/AS01 (Mosquirix) by the WHO in 2021 was a landmark achievement.68,105 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.121 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.72 AS01 is a liposome-based adjuvant with 3-O-desacyl-4ʹ-monophosphoryl lipid A (MPL) and QS-21 saponin, which together boost innate and adaptive immunity, significantly increasing the vaccine efficacy against P. falciparum.122

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.123 After these promising results, the WHO recommended large-scale pilot programs in 2015, which were launched in Ghana, Kenya, and Malawi.124 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.125,126

The Oxford R21/Matrix-M malaria vaccine, a promising malaria vaccine, demonstrated high efficacy rates in early-stage trials.75 The efficacy of R21/Matrix-M was reported at up to 80% in African children over a one-year period, particularly in those receiving the higher adjuvant dose.127–129 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.76,130 The smaller dosage and improved immunogenicity profile of R21/Matrix-M, alongside the use of a potent adjuvant (Matrix-M), offer enhanced protection,131 although its potential to outperform RTS,S/AS01 has not been tested yet.75,76

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.106,132,133 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.134,135 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.136

In a recent phase 2b trial conducted in Burkina Faso, RH5.1/Matrix-M showed promising results in children aged 5–17 months.106 The vaccine was well-tolerated with a favorable safety profile and demonstrated robust immunogenicity.106 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.137

Since the start of the new millennium, malaria vaccine research has diversified beyond pre-erythrocytic targets.138–140 Blood-stage vaccines, including candidates such as merozoite surface protein (MSP) and glutamate rich protein (GLURP), have been evaluated for their potential to inhibit erythrocyte invasion and reduce parasitemia.141,142 Among blood-stage vaccine candidates, GMZ2 which comprised conserved domains of GLURP and MSP3, showed modest but statistically significant efficacy in preventing naturally acquired P. falciparum infection in clinical trials.143,144 However, its limited protective effect led to a shift toward identifying novel blood-stage vaccine targets or enhancing efficacy.138

Vaccines composed of attenuated P. falciparum sporozoites (PfSPZ) have also been undergoing clinical trials to test for protection against malaria as reviewed by Richie et al.145 The first-generation PfSPZ vaccine employs gamma irradiation to achieve attenuation, rendering sporozoites non-replicative while preserving immunogenicity.146 Second-generation chemo-attenuated PfSPZ vaccines showed improved efficacy against heterologous strains but require co-administered drugs, presenting safety concerns due to the risk of parasitemia recrudescence.145,147 Ongoing clinical trials continue to evaluate sporozoite immunization across diverse populations, aiming to optimize immunogenicity, dosing regimens, and cross-strain protection.148–150

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.54,78,151 Viral vector vaccines use modified viruses to deliver malaria antigens, enhancing cellular immunity with fewer doses.152–154

Building on coronavirus disease 2019 (COVID-19) vaccine success, mRNA vaccines offer rapid production and adaptability, enabling tailored responses across different Plasmodium stages.155–157 By eliciting robust humoral and cellular immunity, these next-generation malaria vaccines could have the potential to improve individual protection and reduce transmission, supporting malaria elimination goals in high-burden areas.137,158

Malaria Immune Response and Vaccine Mechanisms

The immune response to Plasmodium infection is complex, with the involvement of both innate and acquired immunity.159–163 Early on during malaria infection, innate immune mechanisms are activated with the action of dendritic cells (DCs) among other innate cells to recognize parasite-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs).160,164,165 The major PAMPs identified in P. falciparum include glycosylphosphatidylinositol anchors, hemozoin, and immunostimulatory nucleic acid motifs, which trigger DC maturation and initiate downstream immune signaling.166,167

The acquired immune response follows, with activation of T and B lymphocytes and antibody production.168,169 During the pre-erythrocytic stage, when sporozoites are inoculated into the skin by an infected mosquito, the CSP is a primary target of the host immune response.170,171 Sporozoites spend hours in the dermis, which provides an opportunity for antigen presentation and priming of the immune cells.172,173 Neutralizing antibodies targeting CSP can prevent hepatocyte invasion, while CD4+ T cell responses contribute to protective immunity.161,174,175

The currently approved malaria vaccines, including RTS,S/AS01 and R21/Matrix-M, utilize recombinant CSP antigens to elicit robust anti-sporozoite antibody responses.176,177 However, the RTS,S/AS01 vaccine’s limited efficacy is partly attributed to its narrow immunological focus with minimal induction of CD8+ T cell responses, which are critical for eliminating infected hepatocytes.178 The absence of substantial CD8+ T-cell-mediated immunity, a key component in rodent models of malaria protection, represents a notable limitation of current vaccines.168,179 Overcoming this issue requires novel vaccines capable of eliciting both humoral and cytotoxic responses, including the identification of broadly conserved CD8+ T cell epitopes presented by MHC class I molecules across diverse human populations.180,181 Additionally, genetic variability in CSP among P. falciparum strains may reduce vaccine efficacy against heterologous parasites, highlighting the need for broader antigenic coverage in future vaccine designs.182–184

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

The RTS,S/AS01 and R21/Matrix-M vaccines offer protective immunity against P. falciparum.185 These vaccines provide a remarkable milestone in the ability to control malaria; however, it is important to delineate their efficacy, safety, and cost in relation to successful implementation in endemic regions.

The RTS,S/AS01 vaccine demonstrated a modest efficacy of approximately 33–36% in reducing clinical malaria cases over four years, with waning protection observed without booster doses.186–189 Notably, no significant efficacy against severe malaria was observed in younger infants even with a booster dose.190 Immunogenicity data reveal that the RTS,S/AS01 booster dose increases total IgG levels against vaccine antigens, with notable differences observed in IgG subclasses.191 Additionally, variable efficacy of RTS,S/AS01 has been shown to depend on the genetics of the local P. falciparum population.192

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.186,193,194 Rare febrile seizures were observed within seven days post-vaccination but resolved without long-term complications.186 Importantly, no fatal adverse events were causally linked to the vaccine.195 A phase 3b clinical trial conducted in Ghana further confirmed the RTS,S/AS01 vaccine safety profile, whether administered alone or co-administered with other vaccines such as yellow fever and measles-rubella.196

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.131,197 Its safety profile is favorable, with no significant adverse effects reported during clinical trials.75 Mild reactions, such as localized pain and fever, were the most commonly observed side effects.75,128

Comparatively, the RTS,S/AS01 and R21/Matrix-M vaccines demonstrated similar efficacy and safety profiles in preventing malaria, particularly in children.198 While evidence directly comparing their efficacy is lacking, early analyses suggest that both vaccines would provide a cost-effective approach for malaria prevention.199,200 Despite their potential, the modest efficacy of RTS,S/AS01 and R21/Matrix-M highlights the need for continued efforts needed to achieve more durable, high-impact malaria vaccines essential for eradication efforts.201

Challenges in Malaria Vaccine 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.202 Nevertheless, achieving a malaria vaccine that provides high efficacy and long-lasting immunity remains challenging.203 Additionally, achieving the full potential of malaria vaccines requires addressing complex logistical, operational, and social challenges, particularly in malaria-endemic regions with limited resources.52,204 Specifically, the challenges of implementing malaria vaccination range from infrastructure limitations to community perceptions as highlighted in (Figure 3).

Logistical and Infrastructure Barriers

A major logistical 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.205 In rural areas in SSA where travel to healthcare centers can be difficult, completing this schedule poses a significant challenge.206 Missed doses, common in settings where healthcare access is inconsistent, can compromise the malaria vaccine efficacy.207

Moreover, the malaria vaccine requires a cold chain capacity, which can be problematic in regions with limited electricity and high temperatures.204,208 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 is not maintained.209

Healthcare Workforce Constraints

Healthcare workers constraints can complicate logistical barriers to the successful implementation of malaria vaccination.210 Many malaria-endemic countries, such as those in SSA, already struggle with a shortage of healthcare providers trained to administer and monitor vaccination programs, particularly in under-resourced areas.21,211

Success in implementing RTS,S/AS01 and R21/Matrix-M vaccines 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.212 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.205 Despite initial challenges with the four-dose schedule and resource limitations, this approach can enhance the vaccine impact without overburdening healthcare systems.205

Financial Barriers

Financial issues can also be 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.213 While organizations such as Gavi, the Vaccine Alliance, have committed support for this goal, securing sustainable funding remains essential.214 In SSA, where healthcare 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.24

Vaccine Hesitancy as a Barrier to Successful Malaria Vaccine Implementation

Public acceptance of malaria vaccines is equally important for malaria vaccination program success with evidence pointing to high acceptance in Africa.215–217 Vaccine hesitancy, often rooted in distrust of health care systems or cultural beliefs, can limit malaria vaccine uptake.218 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.219

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.220 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.221

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.222 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.223

In the context of 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.206 This qualitative study revealed that community rumors leading to vaccine refusals emphasized the need for robust, culturally tailored vaccine promotion efforts.206 These findings by Grant et al offered critical insights for scaling RTS,S/AS01E in Ghana and future malaria vaccine rollouts across Africa.206

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%.224 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.224

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.52,225–227 While RTS,S/AS01 and R21/Matrix-M vaccines approval marked historic achievements and can be viewed as important benchmarks to compare with other novel vaccines, their modest efficacy highlights the need for next-generation malaria vaccines with improved efficacy profiles, easier administration, and long-lasting protection.186,228 The future of malaria vaccine development relies on innovative scientific approaches, enhanced global collaboration, and integration of vaccination with other malaria control strategies.229

In addition, other Plasmodium species, such as P. vivax, which can cause severe malaria, have historically received less focus compared to P. falciparum.230 The importance of focus on P. vivax is highlighted by being a predominant Plasmodium species in most non-African endemic countries.230 The view that P. vivax might not be a priority for prevention compared 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.231,232 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.233,234

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.125 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.80,235

The success of mRNA vaccines in the COVID-19 pandemic raised the interest in applying this novel vaccine type to malaria prevention.235 mRNA vaccines have the advantage of rapid scalability, adaptability to new antigens, and the ability to elicit strong immune responses.236 Efforts to develop mRNA vaccines targeting P. falciparum are already underway, with preclinical studies showing promising results.137 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.237–239 These approaches could complement existing malaria vaccines and offer new and enhanced opportunities for malaria prevention.

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.240,241 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.242,243

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.84 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.244

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.245,246 International partnerships between governments, research institutions, pharmaceutical companies, and global health organizations (eg, the WHO, Gavi, and the Bill & Melinda Gates Foundation), are examples of the helpful efforts needed to drive progress in malaria prevention through vaccination.247,248

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.249,250 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.21,24,204,212,251 Additionally, integrating malaria vaccines into national immunization programs with coordinated international support can be viewed as an important step toward reducing the malaria burden and its eventual elimination.252–254

Conclusions

Malaria vaccination has moved from aspiration to reality with the introduction and approval of RTS,S/AS01 and R21/Matrix-M vaccines which are considered two milestones 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 offers valuable insights into the ultimate goal of malaria elimination; however, significant challenges remain and need to be addressed by global collaborative efforts.

Data Sharing Statement

Data supporting this review are available in the reference section. In addition, the analyzed data that were used during the current systematic review are available from the corresponding author on a reasonable request.

Acknowledgment

This paper has been uploaded to Preprints.org as a preprint which can be accessed through the following link: https://www.preprints.org/manuscript/202412.1635/v1.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

We declare that we received no funding nor financial support/grants by any institutional, private, or corporate entity.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Makam P, Matsa R. “Big Three” Infectious Diseases: tuberculosis, Malaria and HIV/AIDS. Curr Top Med Chem. 2021;21(31):2779–2799. doi:10.2174/1568026621666210916170417

2. Murray CJ, Ortblad KF, Guinovart 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. doi:10.1016/s0140-6736(14)60844-8

3. Phillips MA, Burrows JN, Manyando C, van Huijsduijnen RH, Van Voorhis WC, Wells TNC. Malaria. Nature Reviews Disease Primers. 2017;3:17050. doi:10.1038/nrdp.2017.50

4. Balaji SN, Deshmukh R, Trivedi V. Severe malaria: biology, clinical manifestation, pathogenesis and consequences. J Vector Borne Dis. 2020;57:1–13. doi:10.4103/0972-9062.308793

5. Talapko J, Škrlec I, Alebić T, Jukić M, Včev A. Malaria: the Past and the Present. Microorganisms. 2019;7. doi:10.3390/microorganisms7060179.

6. Raghavendra K, Barik TK, Reddy BP, Sharma P, Dash AP. Malaria vector control: from past to future. Parasitol Res. 2011;108:757–779. doi:10.1007/s00436-010-2232-0

7. van Schalkwyk DA. History of Antimalarial Agents. In: eLS; 2015:1–5. doi:10.1002/9780470015902.a0003624.pub3

8. Wongsrichanalai C, Barcus MJ, Muth S, Sutamihardja A, Wernsdorfer WH. A Review of Malaria Diagnostic Tools: microscopy and Rapid Diagnostic Test (RDT). American JTrop Med Hyg. 2007;77:119–127. doi:10.4269/ajtmh.2007.77.119

9. Viana GM, Melo G, Nobrega De Sousa T, Pucca MB. Editorial: challenges for diagnosis, treatment, and elimination of malaria. Fronti Tropical Dis. 2024;5:1394693. doi:10.3389/fitd.2024.1394693

10. Liu J, Modrek S, Gosling RD, Feachem RG. Malaria eradication: is it possible? Is it worth it? Should we do it? Lancet Glob Health. 2013;1(1):e2–3. doi:10.1016/s2214-109x(13)70002-0

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(11b):53–56. doi:10.7196/SAMJ.2019.v109i11b.14250

12. Oladipo HJ, Tajudeen YA, Oladunjoye IO, et al. Increasing challenges of malaria control in sub-Saharan Africa: priorities for public health research and policymakers. Ann Med Surg. 2022;81:104366. doi:10.1016/j.amsu.2022.104366

13. Swann T. “Anarchist technologies”: anarchism, cybernetics and mutual aid in community responses to the COVID-19 crisis. Organization. 2023;30(1):193–209. doi:10.1177/13505084221090632

14. Sarfo JO, Amoadu M, Kordorwu PY, et al. Malaria amongst children under five in sub-Saharan Africa: a scoping review of prevalence, risk factors and preventive interventions. European Journal of Medical Research. 2023;28(1):80. doi:10.1186/s40001-023-01046-1

15. Schantz-Dunn J, Nour NM. Malaria and pregnancy: a global health perspective. Rev Obstet Gynecol. 2009;2:186–192.

16. Cohen C, Karstaedt A, Frean J, et al. Increased prevalence of severe malaria in HIV-infected adults in South Africa. Clin Infect Dis. 2005;41:1631–1637. doi:10.1086/498023

17. Fried M, Duffy PE. Malaria during Pregnancy. Cold Spring Harb Perspect Med. 2017;7:25551. doi:10.1101/cshperspect.a025551

18. Berhe AD, Doritchamou JYA, Duffy PE. Malaria in pregnancy: adverse pregnancy outcomes and the future of prevention. Fronti Tropical Dis. 2023;4:1229735. doi:10.3389/fitd.2023.1229735

19. Li J, Docile HJ, 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. doi:10.1007/s44197-024-00228-2

20. Hamisi MA, Asri NAM, Yassim ASM, Suppian R. A systematic review on malaria and Tuberculosis (TB) vaccine challenges in sub-Saharan African clinical trials. PLoS One. 2025;20:e0317233. doi:10.1371/journal.pone.0317233

21. Olawade DB, Wada OZ, Ezeagu CN, et al. Malaria vaccination in Africa: a mini-review of challenges and opportunities. Medicine. 2024;103:e38565. doi:10.1097/md.0000000000038565

22. Obeagu EI, Obeagu GU. Adapting to the shifting landscape: implications of climate change for malaria control: a review. Medicine. 2024;103:e39010. doi:10.1097/md.0000000000039010

23. Megersa DM, Luo X-S. Effects of Climate Change on Malaria Risk to Human Health: a Review. Atmosphere. 2025;16:71. doi:10.3390/atmos16010071

24. Okumu F, Gyapong M, Casamitjana N, et al. What Africa can do to accelerate and sustain progress against malaria. PLOS Glob Public Health. 2022;2:e0000262. doi:10.1371/journal.pgph.0000262

25. Conrad MD, Rosenthal PJ. Antimalarial drug resistance in Africa: the calm before the storm? Lancet Infect Dis. 2019;19:e338–e351. doi:10.1016/s1473-3099(19)30261-0

26. Rosenthal PJ, Asua V, Conrad MD. Emergence, transmission dynamics and mechanisms of artemisinin partial resistance in malaria parasites in Africa. Nat Rev Microbiol. 2024;22:373–384. doi:10.1038/s41579-024-01008-2

27. Sinha S, Medhi B, Sehgal R. Challenges of drug-resistant malaria. Parasite. 2014;21. doi:10.1051/parasite/2014059.

28. Msugupakulya BJ, Urio NH, Jumanne M, et al. Changes in contributions of different Anopheles vector species to malaria transmission in east and Southern Africa from 2000 to 2022. Parasites Vectors. 2023;16:408. doi:10.1186/s13071-023-06019-1

29. Sougoufara S, Ottih EC, Tripet F. The need for new vector control approaches targeting outdoor biting anopheline malaria vector communities. Parasites Vectors. 2020;13:295. doi:10.1186/s13071-020-04170-7

30. Recht J, Siqueira AM, Monteiro WM, Herrera SM, Herrera S, Lacerda MVG. Malaria in Brazil, Colombia, Peru and Venezuela: current challenges in malaria control and elimination. Malaria j. 2017;16:273. doi:10.1186/s12936-017-1925-6

31. Amaral PST, Garcia KKS, Suárez-Mutis MC, et al. Malaria in areas under mining activity in the Amazon: a review. Rev Soc Bras Med Trop. 2024;57:e002002024. doi:10.1590/0037-8682-0551-2023

32. Sallum MAM, Conn J, Correa M, Grillet ME. Malaria Transmission in South America—Present Status and Prospects for Elimination. In: Manguin S, Dev V, editors. Towards Malaria Elimination - a Leap Forward. Rijeka: IntechOpen; 2018;2018:1. doi:10.5772/intechopen.76964.

33. Mueller I, Vantaux A, Karl S, et al. Asia-Pacific ICEMR: understanding Malaria Transmission to Accelerate Malaria Elimination in the Asia Pacific Region. Am J Trop Med Hyg. 2022;107:131–137. doi:10.4269/ajtmh.21-1336

34. Sa-ngamuang C, Lawpoolsri S, Su Yin M, et al. Assessment of malaria risk in Southeast Asia: a systematic review. Malaria j. 2023;22:339. doi:10.1186/s12936-023-04772-3

35. Pyae Phyo A, Nosten F. The Artemisinin Resistance in Southeast Asia: an Imminent Global Threat to Malaria Elimination. In: Manguin S, Dev V, editors. Towards Malaria Elimination - a Leap Forward. Rijeka: IntechOpen; 2018;2018:1. doi:10.5772/intechopen.76519.

36. Fauziah N, Jati KM, Rinawan FR, Nugraha NF, Alisjahbana B, Hutagalung J. Emerging malaria in Indonesia: an overview of Plasmodium knowlesi infections. Parasite Epidemiol Control. 2025;28:e00405. doi:10.1016/j.parepi.2024.e00405

37. 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. doi:10.1093/jtm/taab046

38. Leal Filho W, May J, May M, Nagy GJ. 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 j. 2023;22:248. doi:10.1186/s12936-023-04682-4

39. El-Moamly AA. How can we get malaria control back on track? BMJ. 2024;385:q1408. doi:10.1136/bmj.q1408

40. Ippolito MM, Moser KA, Kabuya JB, Cunningham C, Juliano JJ. Antimalarial Drug Resistance and Implications for the WHO Global Technical Strategy. Curr Epidemiol Rep. 2021;8:46–62. doi:10.1007/s40471-021-00266-5

41. White NJ. Antimalarial drug resistance. J Clin Invest. 2004;113:1084–1092. doi:10.1172/jci21682

42. Assefa A, Fola AA, 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 j. 2024;23:34. doi:10.1186/s12936-024-04848-8

43. 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. doi:10.3390/tropicalmed9090223

44. Guyant P, Corbel V, Guérin PJ, et al. Past and new challenges for malaria control and elimination: the role of operational research for innovation in designing interventions. Malaria j. 2015;14:279. doi:10.1186/s12936-015-0802-4

45. Sachs J, Malaney P. The economic and social burden of malaria. Nature. 2002;415:680–685. doi:10.1038/415680a

46. Mori AT, Mallange G, Kühl M-J, Okell L. Cost of treating severe malaria in children in Africa: a systematic literature review. Malaria j. 2024;23:334. doi:10.1186/s12936-024-05173-w

47. Ricci F. Social implications of malaria and their relationships with poverty. Mediterr J Hematol Infect Dis. 2012;4:e2012048. doi:10.4084/mjhid.2012.048

48. Andrade MV, Noronha K, Diniz BPC, et al. The economic burden of malaria: a systematic review. Malaria j. 2022;21:283. doi:10.1186/s12936-022-04303-6

49. Wafula ST, Habermann T, Franke MA, et al. What are the pathways between poverty and malaria in sub-Saharan Africa? A systematic review of mediation studies. Infect Diseases Poverty. 2023;12:58. doi:10.1186/s40249-023-01110-2

50. Suh PF, Elanga-Ndille E, Tchouakui M, et al. Impact of insecticide resistance on malaria vector competence: a literature review. Malaria j. 2023;22:19. doi:10.1186/s12936-023-04444-2

51. Nkya TE, Akhouayri I, Poupardin R, et al. Insecticide resistance mechanisms associated with different environments in the malaria vector Anopheles gambiae: a case study in Tanzania. Malaria j. 2014;13:28. doi:10.1186/1475-2875-13-28

52. Sutanto H. Combating Malaria with Vaccines: insights from the One Health Framework. Acta Microbiol Hellenica. 2024;69:153–166. doi:10.3390/amh69030015

53. Cohen JM, Okumu F, Moonen B. The fight against malaria: diminishing gains and growing challenges. Sci Transl Med. 2022;14:eabn3256. doi:10.1126/scitranslmed.abn3256

54. El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success—lessons learned and future prospects. Trop Med Int Health. 2023;51:29. doi:10.1186/s41182-023-00516-w

55. Moorthy VS, Good MF, Hill AV. Malaria vaccine developments. Lancet. 2004;363:150–156. doi:10.1016/s0140-6736(03)15267-1

56. Tizifa TA, Kabaghe AN, McCann RS, van den Berg H, Van Vugt M, Phiri KS. Prevention Efforts for Malaria. Curr Trop Med Rep. 2018;5:41–50. doi:10.1007/s40475-018-0133-y

57. 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. doi:10.1016/j.vaccine.2015.01.059

58. Sauboin CJ, Van Bellinghen LA, 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. doi:10.1186/s12936-015-1046-z

59. Crompton PD, Pierce SK, Miller LH. Advances and challenges in malaria vaccine development. J Clin Invest. 2010;120:4168–4178. doi:10.1172/jci44423

60. Lorenz V, Karanis P. Malaria vaccines: looking back and lessons learnt. Asian Pac J Trop Biomed. 2011;1:74–78. doi:10.1016/s2221-1691(11)60072-5

61. 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. doi:10.3390/ijerph110706791

62. Duffy PE, Sahu T, Akue A, Milman N, Anderson C. Pre-erythrocytic malaria vaccines: identifying the targets. Expert Rev Vaccines. 2012;11:1261–1280. doi:10.1586/erv.12.92

63. Zheng J, Pan H, Gu Y, et al. Prospects for Malaria Vaccines: pre-Erythrocytic Stages, Blood Stages, and Transmission-Blocking Stages. Biomed Res Int. 2019;2019:9751471. doi:10.1155/2019/9751471

64. Richie TL, Billingsley PF, Sim BK, et al. Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine. 2015;33:7452–7461. doi:10.1016/j.vaccine.2015.09.096

65. Tougan T, Ito K, Palacpac NM, Egwang TG, 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. doi:10.1016/j.parint.2016.06.012

66. Varo R, Chaccour C, Bassat Q. Update on malaria. Med Clin. 2020;155:395–402. doi:10.1016/j.medcli.2020.05.010

67. Duffy PE. Transmission-Blocking Vaccines: harnessing Herd Immunity for Malaria Elimination. Expert Rev Vaccines. 2021;20:185–198. doi:10.1080/14760584.2021.1878028

68. Laurens MB. RTS,S/AS01 vaccine (Mosquirix™): an overview. Hum Vaccin Immunother. 2020;16:480–489. doi:10.1080/21645515.2019.1669415

69. Marques-da-Silva C, Peissig K, Kurup SP. Pre-Erythrocytic Vaccines against Malaria. Vaccines. 2020;8:400. doi:10.3390/vaccines8030400

70. Zavala F, Tam JP, Hollingdale MR, et al. Rationale for development of a synthetic vaccine against Plasmodium falciparum malaria. Science. 1985;228:1436–1440. doi:10.1126/science.2409595

71. World Health Organization. WHO recommends groundbreaking malaria vaccine for children at risk. Available from: https://www.who.int/news/item/06-10-2021-who-recommends-groundbreaking-malaria-vaccine-for-children-at-risk. Accessed November 2, 2024.

72. Egbewande OM. The RTS,S malaria vaccine: journey from conception to recommendation. Public Health Pract. 2022;4:100283. doi:10.1016/j.puhip.2022.100283

73. Syed YY. RTS,S/AS01 malaria vaccine (Mosquirix®): a profile of its use. Drugs Therapy Perspect. 2022;38:373–381. doi:10.1007/s40267-022-00937-3

74. Hanboonkunupakarn B, Mukaka M, Jittamala P, et al. A randomised trial of malaria vaccine R21/Matrix-M™ with and without antimalarial drugs in Thai adults. Npj Vaccines. 2024;9:124. doi:10.1038/s41541-024-00920-1

75. Aderinto N, Olatunji G, Kokori E, Sikirullahi S, Aboje JE, Ojabo RE. A perspective on Oxford’s R21/Matrix-M™ malaria vaccine and the future of global eradication efforts. Malar J. 2024;23:16. doi:10.1186/s12936-024-04846-w

76. World Health Organization. R21/Matrix-M malaria vaccine: evidence to recommendations framework, 2023. Available from: https://www.who.int/publications/m/item/r21-matrix-m-malaria-vaccine--evidence-to-recommendations-framework--2023. Accessed November 5, 2024.

77. Moorthy V, Hamel MJ, Smith PG. Malaria vaccines for children: and now there are two. Lancet. 2024;403:504–505. doi:10.1016/s0140-6736(23)02743-5

78. Tsoumani ME, Voyiatzaki C, Efstathiou A. Malaria Vaccines: from the Past towards the mRNA Vaccine Era. Vaccines. 2023;11:1452. doi:10.3390/vaccines11091452

79. Matarazzo L, Bettencourt PJG. mRNA vaccines: a new opportunity for malaria, tuberculosis and HIV. Front Immunol. 2023;14:1172691. doi:10.3389/fimmu.2023.1172691

80. 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. doi:10.1007/s11686-023-00712-y

81. 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. doi:10.3389/fimmu.2022.896958

82. Greenwood B, Dicko A, Sagara I, et al. Seasonal vaccination against malaria: a potential use for an imperfect malaria vaccine. Malar J. 2017;16:182. doi:10.1186/s12936-017-1841-9

83. Ubillos I, Ayestaran A, Nhabomba AJ, 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. doi:10.1186/s12916-018-1186-4

84. Dicko A, Ouedraogo J-B, Zongo I, et al. Seasonal vaccination with RTS,S/AS01E 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. Lancet Infect Dis. 2024;24:75–86. doi:10.1016/S1473-3099(23)00368-7

85. Chandramohan D, Zongo I, Sagara I, et al. Seasonal Malaria Vaccination with or without Seasonal Malaria Chemoprevention. N Engl J Med. 2021;385:1005–1017. doi:10.1056/NEJMoa2026330

86. Mensah VA, Gueye A, Ndiaye M, 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. doi:10.1371/journal.pone.0167951

87. de Graaf H, Payne RO, Taylor I, et al. Safety and Immunogenicity of ChAd63/MVA Pfs25-IMX313 in a Phase I First-in-Human Trial. Front Immunol. 2021;12:694759. doi:10.3389/fimmu.2021.694759

88. Zaric M, Marini A, Nielsen CM, et al. Poor CD4(+) T Cell Immunogenicity Limits Humoral Immunity to P. falciparum Transmission-Blocking Candidate Pfs25 in Humans. Front Immunol. 2021;12:732667. doi:10.3389/fimmu.2021.732667

89. Nasir SMI, Amarasekara S, Wickremasinghe R, Fernando D, Udagama P. Prevention of re-establishment of malaria: historical perspective and future prospects. Malaria j. 2020;19:452. doi:10.1186/s12936-020-03527-8

90. Cibulskis RE, Alonso P, Aponte J, et al. Malaria: global progress 2000 – 2015 and future challenges. Infect Diseases Poverty. 2016;5:61. doi:10.1186/s40249-016-0151-8

91. Parija SC. The persistent challenges of malaria. Trop Parasitol. 2021;11:1–2. doi:10.4103/tp.tp_29_21

92. Hay SI, Smith DL, Snow RW. Measuring malaria endemicity from intense to interrupted transmission. Lancet Infect Dis. 2008;8:369–378. doi:10.1016/s1473-3099(08)70069-0

93. Weiss DJ, Lucas TCD, Nguyen M, 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. doi:10.1016/s0140-6736(19)31097-9

94. Battle KE, Lucas TCD, Nguyen M, 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. doi:10.1016/s0140-6736(19)31096-7

95. World Health Organization. WHO malaria terminology, 2021 update. Available from: https://www.who.int/publications/i/item/9789240038400. Accessed November 2, 2024.

96. Autino B, Noris A, Russo R, Castelli F. Epidemiology of malaria in endemic areas. Mediterr J Hematol Infect Dis. 2012;4:e2012060. doi:10.4084/mjhid.2012.060

97. World Health Organization. Malaria - Key facts. Available from: https://www.who.int/news-room/fact-sheets/detail/malaria. Accessed November 3, 2024.

98. Mbishi JV, Chombo S, Luoga P, et al. Malaria in under-five children: prevalence and multi-factor analysis of high-risk African countries. BMC Public Health. 2024;24:1687. doi:10.1186/s12889-024-19206-1

99. World Health Organization. The Global Health Observatory: malaria burden data: cases and deaths. Available from: https://www.who.int/data/gho/data/themes/topics/topic-details/GHO/malaria-cases-deaths. Accessed November 2, 2024.

100. Hill AV. Vaccines against malaria. Philos Trans R Soc Lond B Biol Sci. 2011;366:2806–2814. doi:10.1098/rstb.2011.0091

101. Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine. 2015;33(4):D13–23. doi:10.1016/j.vaccine.2015.07.091

102. Sato S. Plasmodium-a brief introduction to the parasites causing human malaria and their basic biology. J Physiol Anthropol. 2021;40(1):1. doi:10.1186/s40101-020-00251-9

103. Richie T. High road, low road? Choices and challenges on the pathway to a malaria vaccine. Parasitology. 2006;133(S2):S113–144. doi:10.1017/s0031182006001843

104. Nussenzweig RS, Nussenzweig V. Development of sporozoite vaccines. Philos Trans R Soc Lond B Biol Sci. 1984;307:117–128. doi:10.1098/rstb.1984.0113

105. World Health Organization. WHO recommends groundbreaking malaria vaccine for children at risk. Available from: https://www.who.int/news/item/06-10-2021-who-recommends-groundbreaking-malaria-vaccine-for-children-at-risk. Accessed November 3, 2024.

106. Natama HM, Salkeld J, Somé A, 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. doi:10.1016/s1473-3099(24)00752-7

107. Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature. 1967;216:160–162. doi:10.1038/216160a0

108. Dame JB, Williams JL, McCutchan TF, 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. doi:10.1126/science.6204383

109. Nussenzweig V, Nussenzweig RS. Circumsporozoite proteins of malaria parasites. Cell. 1985;42:401–403. doi:10.1016/0092-8674(85)90093-5

110. Ballou WR, Hoffman SL, Sherwood JA, et al. Safety and efficacy of a recombinant DNA Plasmodium falciparum sporozoite vaccine. Lancet. 1987;1:1277–1281. doi:10.1016/s0140-6736(87)90540-x

111. Stoute JA, Slaoui M, Heppner DG, et al. 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. doi:10.1056/nejm199701093360202

112. Alonso PL, Sacarlal J, Aponte JJ, 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. doi:10.1016/s0140-6736(04)17223-1

113. Rts SCTP. 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. doi:10.1016/s0140-6736(15)60721-8

114. World Health Organization. Malaria vaccine pilot launched in Malawi. Available from: https://www.who.int/news/item/23-04-2019-malaria-vaccine-pilot-launched-in-malawi. Accessed November 3, 2024.

115. Vanderberg JP. Reflections on early malaria vaccine studies, the first successful human malaria vaccination, and beyond. Vaccine. 2009;27(1):2–9. doi:10.1016/j.vaccine.2008.10.028

116. Richie TL, Saul A. Progress and challenges for malaria vaccines. Nature. 2002;415(6872):694–701. doi:10.1038/415694a

117. Young JF, Hockmeyer WT, Gross M, et al. Expression of Plasmodium falciparum Circumsporozoite Proteins in Escherichia coli for Potential Use in a Human Malaria Vaccine. Science. 1985;228(4702):958–962. doi:10.1126/science.2988125

118. Molina-Franky J, Cuy-Chaparro L, Camargo A, et al. Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J. 2020;19(1):56. doi:10.1186/s12936-020-3141-z

119. Coppi A, Natarajan R, Pradel G, et al. 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(2):341–356. doi:10.1084/jem.20101488

120. Campo JJ, Aponte JJ, Skinner J, 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(3):519–531. doi:10.1074/mcp.M114.044677

121. Corradin G, Céspedes N, Verdini A, Kajava AV, Arévalo-Herrera M, Herrera S. Chapter 5 - Malaria Vaccine Development Using Synthetic Peptides as a Technical Platform. In: Melief CJM editor, Advances in Immunology. Vol. 114. Academic Press; 2012:107–149. doi:10.1016/B978-0-12-396548-6.00005-6

122. Roman F, Burny W, Ceregido MA, et al. Adjuvant system AS01: from mode of action to effective vaccines. Expert Rev Vaccines. 2024;23(1):715–729. doi:10.1080/14760584.2024.2382725

123. World Health Organization. Full Evidence Report on the RTS,S/AS01 Malaria Vaccine. Available from: https://cdn.who.int/media/docs/default-source/immunization/mvip/full-evidence-report-on-the-rtss-as01-malaria-vaccine-for-sage-mpag-sept2021.pdf. Accessed November 3, 2024.

124. World Health Organization. RTS,S malaria vaccine reaches more than 650 000 children in Ghana, Kenya and Malawi through groundbreaking pilot programme. Available from: 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 November 3, 2024.

125. Björkman A, Benn CS, Aaby P, Schapira A. RTS,S/AS01 malaria vaccine—proven safe and effective? Lancet Infect Dis. 2023;23(8):e318–e322. doi:10.1016/S1473-3099(23)00126-3

126. Sinnis P, Fidock DA. The RTS,S vaccine-a chance to regain the upper hand against malaria? Cell. 2022;185(5):750–754. doi:10.1016/j.cell.2022.01.028

127. Datoo MS, Natama MH, Somé A, 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(10287):1809–1818. doi:10.1016/s0140-6736(21)00943-0

128. Datoo MS, Dicko A, Tinto H, 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(10426):533–544. doi:10.1016/s0140-6736(23)02511-4

129. Datoo MS, Natama HM, Somé A, 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(12):1728–1736. doi:10.1016/s1473-3099(22)00442-x

130. Mahase E. WHO recommends second vaccine for malaria prevention in children. BMJ. 2023;383:2291. doi:10.1136/bmj.p2291

131. 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;30:140. doi:10.1093/jtm/taad140

132. Palacpac NMQ, Horii T. RH5.1/Matrix-M: highlighting blood-stage malaria vaccines. Lancet Infect Dis. 2024. doi:10.1016/s1473-3099(24)00800-4

133. Silk SE, Kalinga WF, Salkeld J, 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(10):1105–1117. doi:10.1016/s1473-3099(24)00312-8

134. King LDW, Pulido D, Barrett JR, et al. Preclinical development of a stabilized RH5 virus-like particle vaccine that induces improved antimalarial antibodies. Cell Rep Med. 2024;5(7):101654. doi:10.1016/j.xcrm.2024.101654

135. Douglas AD, Williams AR, Illingworth JJ, et al. The blood-stage malaria antigen PfRH5 is susceptible to vaccine-inducible cross-strain neutralizing antibody. Nat Commun. 2011;2:601. doi:10.1038/ncomms1615

136. Ragotte RJ, Higgins MK, Draper SJ. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol. 2020;36(6):545–559. doi:10.1016/j.pt.2020.04.003

137. Laurenson AJ, Laurens MB. A new landscape for malaria vaccine development. PLoS Pathog. 2024;20(6):e1012309. doi:10.1371/journal.ppat.1012309

138. Duffy PE, Patrick Gorres J. Malaria vaccines since 2000: progress, priorities, products. Npj Vaccines. 2020;5(1):48. doi:10.1038/s41541-020-0196-3

139. Duffy PE. Current approaches to malaria vaccines. Curr Opin Microbiol. 2022;70:102227. doi:10.1016/j.mib.2022.102227

140. Hoffman SL, Vekemans J, Richie TL, Duffy PE. The March Toward Malaria Vaccines. Am J Prev Med. 2015;49(6):S319–333. doi:10.1016/j.amepre.2015.09.011

141. Miura K. Progress and prospects for blood-stage malaria vaccines. Expert Rev Vaccines. 2016;15:765–781. doi:10.1586/14760584.2016.1141680

142. Dassah S, Adu B, Tiendrebeogo RW, et al. GMZ2 Vaccine-Induced Antibody Responses, Naturally Acquired Immunity and the Incidence of Malaria in Burkinabe Children. Front Immunol. 2022;13:899223. doi:10.3389/fimmu.2022.899223

143. Sirima SB, Mordmüller B, Milligan P, et al. A phase 2b randomized, controlled trial of the efficacy of the GMZ2 malaria vaccine in African children. Vaccine. 2016;34(38):4536–4542. doi:10.1016/j.vaccine.2016.07.041

144. Theisen M, Adu B, Mordmüller B, Singh S. The GMZ2 malaria vaccine: from concept to efficacy in humans. Expert Rev Vaccines. 2017;16(9):907–917. doi:10.1080/14760584.2017.1355246

145. Richie TL, Church LWP, Murshedkar T, et al. Sporozoite immunization: innovative translational science to support the fight against malaria. Expert Rev Vaccines. 2023;22(1):964–1007. doi:10.1080/14760584.2023.2245890

146. James ER, Matheny S, Overby J, et al. A First for Human Vaccinology: GMP Compliant Radiation Attenuation of Plasmodium falciparum Sporozoites for Production of a Vaccine Against Malaria. Front Immunol. 2022;13:851028. doi:10.3389/fimmu.2022.851028

147. Mwakingwe-Omari A, Healy SA, Lane J, et al. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature. 2021;595(7866):289–294. doi:10.1038/s41586-021-03684-z

148. Berry AA, Richie TL, Church LWP, et al. Safety, tolerability and immunogenicity of a condensed, multi-dose prime regimen of PfSPZ Vaccine for the prevention of Plasmodium falciparum malaria infection. Malaria j. 2025;24(1):88. doi:10.1186/s12936-025-05299-5

149. Nunes-Cabaço H, Moita D, Prudêncio M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front Immunol. 2022;13:977472. doi:10.3389/fimmu.2022.977472

150. Moita D, Rôla C, Nunes-Cabaço H, et al. The effect of dosage on the protective efficacy of whole-sporozoite formulations for immunization against malaria. Npj Vaccines. 2023;8(1):182. doi:10.1038/s41541-023-00778-9

151. Kanoi BN, Maina M, Likhovole C, Kobia FM, Gitaka J. Malaria vaccine approaches leveraging technologies optimized in the COVID-19 era. Fronti Tropical Dis. 2022;3:988665. doi:10.3389/fitd.2022.988665

152. Ewer KJ, Sierra-Davidson K, Salman AM, et al. Progress with viral vectored malaria vaccines: a multi-stage approach involving “unnatural immunity”. Vaccine. 2015;33(52):7444–7451. doi:10.1016/j.vaccine.2015.09.094

153. Reyes‐Sandoval A, Harty JT, Todryk SM. Viral vector vaccines make memory T cells against malaria. Immunology. 2007;121(2):158–165. doi:10.1111/j.1365-2567.2006.02552.x

154. Milicic A, Rollier CS, Tang CK, Longley R, Hill AVS, Reyes-Sandoval A. Adjuvanting a viral vectored vaccine against pre-erythrocytic malaria. Sci Rep. 2017;7(1):7284. doi:10.1038/s41598-017-07246-0

155. Chaudhary N, Weissman D, Whitehead KA. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat Rev Drug Discov. 2021;20:817–838. doi:10.1038/s41573-021-00283-5

156. Scaria PV, Roth N, Schwendt K, et al. mRNA vaccines expressing malaria transmission-blocking antigens Pfs25 and Pfs230D1 induce a functional immune response. Npj Vaccines. 2024;9:9. doi:10.1038/s41541-023-00783-y

157. Makoni M. mRNA vaccine against malaria effective in preclinical model. Lancet Microbe. 2023;4:e970. doi:10.1016/s2666-5247(23)00332-4

158. Laurens MB. Novel malaria vaccines. Hum Vaccin Immunother. 2021;17:4549–4552. doi:10.1080/21645515.2021.1947762

159. Mandala WL, Harawa V, Dzinjalamala F, Tembo D. The role of different components of the immune system against Plasmodium falciparum malaria: possible contribution towards malaria vaccine development. mol Biochem Parasitol. 2021;246:111425. doi:10.1016/j.molbiopara.2021.111425

160. Gowda DC, Wu X. Parasite Recognition and Signaling Mechanisms in Innate Immune Responses to Malaria. Front Immunol. 2018;9:3006. doi:10.3389/fimmu.2018.03006

161. Rochford R, Kazura J. Introduction: immunity to malaria. Immunol Rev. 2020;293:5–7. doi:10.1111/imr.12831

162. López C, Yepes-Pérez Y, Hincapié-Escobar N, Díaz-Arévalo D, Patarroyo MA. What Is Known about the Immune Response Induced by Plasmodium vivax Malaria Vaccine Candidates? Front Immunol. 2017;8:126. doi:10.3389/fimmu.2017.00126

163. Pohl K, Cockburn IA. Innate immunity to malaria: the good, the bad and the unknown. Front Immunol. 2022;13:914598. doi:10.3389/fimmu.2022.914598

164. da Silva HB, Caetano SS, Monteiro I, et al. Early skin immunological disturbance after Plasmodium-infected mosquito bites. Cell Immunol. 2012;277:22–32. doi:10.1016/j.cellimm.2012.06.003

165. Kalantari P. The Emerging Role of Pattern Recognition Receptors in the Pathogenesis of Malaria. Vaccines. 2018;6:13. doi:10.3390/vaccines6010013

166. Cai C, Hu Z, Yu X. Accelerator or Brake: immune Regulators in Malaria. Front Cell Infect Microbiol. 2020;10:610121. doi:10.3389/fcimb.2020.610121

167. Krishnegowda G, Hajjar AM, Zhu J, et al. Induction of proinflammatory responses in macrophages by the glycosylphosphatidylinositols of Plasmodium falciparum: cell signaling receptors, glycosylphosphatidylinositol (GPI) structural requirement, and regulation of GPI activity. J Biol Chem. 2005;280:8606–8616. doi:10.1074/jbc.M413541200

168. Kurup SP, Butler NS, Harty JT. T cell-mediated immunity to malaria. Nat Rev Immunol. 2019;19:457–471. doi:10.1038/s41577-019-0158-z

169. Rogers KJ, Vijay R, Butler NS. Anti-malarial humoral immunity: the long and short of it. Microbes Infect. 2021;23:104807. doi:10.1016/j.micinf.2021.104807

170. Abuga KM, Jones-Warner W, Hafalla JCR. Immune responses to malaria pre-erythrocytic stages: implications for vaccine development. Parasite Immunol. 2021;43:e12795. doi:10.1111/pim.12795

171. Ménard R, Tavares J, Cockburn I, Markus M, Zavala F, Amino R. Looking under the skin: the first steps in malarial infection and immunity. Nat Rev Microbiol. 2013;11:701–712. doi:10.1038/nrmicro3111

172. Yamauchi LM, Coppi A, Snounou G, Sinnis P. Plasmodium sporozoites trickle out of the injection site. Cell Microbiol. 2007;9:1215–1222. doi:10.1111/j.1462-5822.2006.00861.x

173. Sinnis P, Zavala F. The skin: where malaria infection and the host immune response begin. Semin Immunopathol. 2012;34:787–792. doi:10.1007/s00281-012-0345-5

174. Doolan DL, Dobaño C, Baird JK. Acquired immunity to malaria. Clin Microbiol Rev. 2009;22:13–36. doi:10.1128/cmr.00025-08

175. Gonzales SJ, Reyes RA, Braddom AE, Batugedara G, Bol S, Bunnik EM. Naturally Acquired Humoral Immunity Against Plasmodium falciparum Malaria. Front Immunol. 2020;11:594653. doi:10.3389/fimmu.2020.594653

176. Long CA, Zavala F. Malaria vaccines and human immune responses. Curr Opin Microbiol. 2016;32:96–102. doi:10.1016/j.mib.2016.04.006

177. Suscovich TJ, Fallon JK, Das J, et al. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Sci Transl Med. 2020;12. doi:10.1126/scitranslmed.abb4757

178. Stanisic DI, McCall MBB. Correlates of malaria vaccine efficacy. Expert Rev Vaccines. 2021;20:143–161. doi:10.1080/14760584.2021.1882309

179. Van Braeckel-Budimir N, Harty JT. CD8 T-cell-mediated protection against liver-stage malaria: lessons from a mouse model. Front Microbiol. 2014;5:272. doi:10.3389/fmicb.2014.00272

180. Testa JS, Philip R. Role of T-cell epitope-based vaccine in prophylactic and therapeutic applications. Future Virol. 2012;7:1077–1088. doi:10.2217/fvl.12.108

181. Davies DH, Duffy P, Bodmer JL, Felgner PL, Doolan DL. Large screen approaches to identify novel malaria vaccine candidates. Vaccine. 2015;33:7496–7505. doi:10.1016/j.vaccine.2015.09.059

182. Takala SL, Plowe CV. Genetic diversity and malaria vaccine design, testing and efficacy: preventing and overcoming “vaccine resistant malaria”. Parasite Immunol. 2009;31:560–573. doi:10.1111/j.1365-3024.2009.01138.x

183. Ajibola O, Diop MF, Ghansah A, et al. In silico characterisation of putative Plasmodium falciparum vaccine candidates in African malaria populations. Sci Rep. 2021;11:16215. doi:10.1038/s41598-021-95442-4

184. Miotto O, Amambua-Ngwa A, Amenga-Etego LN, et al. Identification of complex Plasmodium falciparum genetic backgrounds circulating in Africa: a multicountry genomic epidemiology analysis. Lancet Microbe. 2024;5:100941. doi:10.1016/j.lanmic.2024.07.004

185. 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 J. 2024;100:877–878. doi:10.1093/postmj/qgae086

186. Ogieuhi IJ, Ajekiigbe VO, Kolo-Manma K, et al. 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. doi:10.1186/s12982-024-00284-w

187. Adjei MR, Okine R, Tweneboah PO, et al. 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 j. 2024;23:305. doi:10.1186/s12936-024-05123-6

188. 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. doi:10.1186/s12916-015-0408-2

189. White MT, Verity R, Griffin JT, 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. doi:10.1016/s1473-3099(15)00239-x

190. Samuels AM, Ansong D, Kariuki SK, 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. doi:10.1016/s1473-3099(22)00273-0

191. Sánchez L, Vidal M, Jairoce C, 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. doi:10.1038/s41541-020-0192-7

192. Neafsey DE, Juraska M, Bedford T, et al. Genetic Diversity and Protective Efficacy of the RTS,S/AS01 Malaria Vaccine. N Engl J Med. 2015;373:2025–2037. doi:10.1056/NEJMoa1505819

193. Yihunie W, Kebede B, Tegegne BA, et al. 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. doi:10.2147/cpaa.S400155

194. Chutiyami M, Saravanakumar P, Bello UM, et al. Malaria vaccine efficacy, safety, and community perception in Africa: a scoping review of recent empirical studies. Infection. 2024;52:2007–2028. doi:10.1007/s15010-024-02196-y

195. Guerra Mendoza Y, Garric E, Leach A, 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. doi:10.1080/21645515.2019.1586040

196. Asante KP, Ansog D, Kaali S, 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. doi:10.1016/j.vaccine.2020.03.014

197. Mahase E. Ghana approves Oxford’s malaria vaccine for children aged 5 to 36 months. BMJ. 2023;381:850. doi:10.1136/bmj.p850

198. Hammershaimb EA, Berry AA. Pre-erythrocytic malaria vaccines: RTS,S, R21, and beyond. Expert Rev Vaccines. 2024;23:49–52. doi:10.1080/14760584.2023.2292204

199. Schmit N, Topazian HM, Natama HM, 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. doi:10.1016/s1473-3099(23)00816-2

200. Seo MK, Baker P, Ngo KN. 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. doi:10.1186/1475-2875-13-66

201. Thellier M, Gemegah AAJ, Tantaoui I. Global Fight against Malaria: goals and Achievements 1900–2022. J Clin Med. 2024;13:5680. doi:10.3390/jcm13195680

202. Matola Y, Chumbi G, Moyo CS, Bwanali A, Lubanga AF. 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. Fronti Tropical Dis. 2024;5:1387078. doi:10.3389/fitd.2024.1387078

203. Beeson JG, Kurtovic L, Dobaño C, et al. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med. 2019;11. doi:10.1126/scitranslmed.aau1458.

204. Okesanya OJ, Atewologun F, Lucero-Prisno DE, et al. Bridging the gap to malaria vaccination in Africa: challenges and opportunities. Journal of Medicine, Surgery, and Public Health. 2024;2:100059. doi:10.1016/j.glmedi.2024.100059

205. Hill J, Bange T, Hoyt J, et al. 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. doi:10.1016/s2214-109x(24)00013-5

206. Grant J, Gyan T, Agbokey F, Webster J, Greenwood B, Asante KP. 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 j. 2022;21:147. doi:10.1186/s12936-022-04168-9

207. Amimo F. Malaria vaccination: hurdles to reach high-risk children. BMC Med. 2024;22:111. doi:10.1186/s12916-024-03321-2

208. Adjei MR, Amponsa-Achiano K, Okine R, et al. Post introduction evaluation of the malaria vaccine implementation programme in Ghana, 2021. BMC Public Health. 2023;23:586. doi:10.1186/s12889-023-15481-6

209. 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 j. 2020;19:202. doi:10.1186/s12936-020-03253-1

210. Adeshina OO, Nyame S, Milner J, Milojevic A, Asante KP. Barriers and facilitators to nationwide implementation of the malaria vaccine in Ghana. Health Policy Plan. 2023;38:28–37. doi:10.1093/heapol/czac077

211. Nkwenkeu SF, Jalloh MF, Walldorf JA, 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. doi:10.1186/s12889-020-8347-z

212. World Health Organization. Tackling malaria in countries hardest hit by the disease: ministerial conference report, Yaoundé, Cameroon, 6, March 2024. Available from: https://www.who.int/publications/i/item/9789240100459. Accessed November 5, 2024.

213. Adamu AA, Jalo RI, Ndwandwe D, Wiysonge CS. 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. 2024;12:111. doi:10.3390/vaccines12020111

214. Zarocostas J. Gavi unveils malaria vaccine plans. Lancet. 2023;401:1485. doi:10.1016/s0140-6736(23)00902-9

215. Sulaiman SK, Musa MS, Tsiga-Ahmed FI, Dayyab FM, ulaiman AK, Bako AT. 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. doi:10.1371/journal.pone.0278224

216. Aremu TO, Ajibola OA, Oluwole OE, Adeyinka KO, Dada SO, Okoro ON. Looking Beyond the Malaria Vaccine Approval to Acceptance and Adoption in Sub-Saharan Africa. Fronti Tropical Dis. 2022;3:857844. doi:10.3389/fitd.2022.857844

217. Kigongo E, Kabunga A, Opollo MS, et al. Community readiness and acceptance for the implementation of a novel malaria vaccine among t-risk children in sub-saharan Africa: a systematic review protocol. Malar J. 2024;23:182. doi:10.1186/s12936-024-04995-y

218. Mumtaz H, Nadeem A, Bilal W, et al. Acceptance, availability, and feasibility of RTS, S/AS01 malaria vaccine: a review. Immun Inflamm Dis. 2023;11:e899. doi:10.1002/iid3.899

219. Bam V, Mohammed A, Kusi-Amponsah A, et al. Caregivers’ perception and acceptance of malaria vaccine for Children. PLoS One. 2023;18:e0288686. doi:10.1371/journal.pone.0288686

220. Ojakaa DI, Jarvis JD, Matilu MI, Thiam S. Acceptance of a malaria vaccine by caregivers of sick children in Kenya. Malar J. 2014;13:172. doi:10.1186/1475-2875-13-172

221. Nyalundja AD, Bugeme PM, Guillaume AS, et al. 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. 2024;12. doi:10.3390/vaccines12040380.

222. Mtenga S, Kimweri A, Romore I, et al. Stakeholders’ opinions and questions regarding the anticipated malaria vaccine in Tanzania. Malar J. 2016;15:189. doi:10.1186/s12936-016-1209-6

223. Amin MA, Afrin S, Bonna AS, Rozars MFK, Nabi MH, Hawlader MDH. 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. doi:10.1111/hex.13862

224. Ansar F, Azzam A, Rauf MS, et al. Global Analysis of RTS, S/AS01 Malaria Vaccine Acceptance Rates and Influencing Factors: a Systematic Review. Cureus. 2024;16:e60678. doi:10.7759/cureus.60678

225. Academia. editorial - ClinicalMedicine. Malaria: (still) a global health priority. EClinicalMedicine. 2021;34:100891. doi:10.1016/j.eclinm.2021.100891

226. Chutiyami M. Recent Trends in Malaria Vaccine Research Globally: a Bibliometric Analysis From 2005 to 2022. J Parasitol Res. 2024;2024:8201097. doi:10.1155/2024/8201097

227. Miura K, Flores-Garcia Y, Long CA, Zavala F. Vaccines and monoclonal antibodies: new tools for malaria control. Clin Microbiol Rev. 2024;37:e0007123. doi:10.1128/cmr.00071-23

228. Locke E, Flores-Garcia Y, Mayer BT, 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. doi:10.1038/s41541-024-00819-x

229. Draper SJ, Sack BK, King CR, et al. Malaria Vaccines: recent Advances and New Horizons. Cell Host Microbe. 2018;24:43–56. doi:10.1016/j.chom.2018.06.008

230. Mueller I, Shakri AR, Chitnis CE. Development of vaccines for Plasmodium vivax malaria. Vaccine. 2015;33:7489–7495. doi:10.1016/j.vaccine.2015.09.060

231. Price RN, Commons RJ, Battle KE, Thriemer K, Mendis K. Plasmodium vivax in the Era of the Shrinking P. falciparum Map. Trends Parasitol. 2020;36:560–570. doi:10.1016/j.pt.2020.03.009

232. Khan N, Daily JP. Update on pathogenesis, management, and control of Plasmodium vivax. Curr Opin Infect Dis. 2022;35:404–409. doi:10.1097/qco.0000000000000867

233. da Veiga GTS, Moriggi MR, Vettorazzi JF, Müller-Santos M, Albrecht L. Plasmodium vivax vaccine: what is the best way to go? Front Immunol. 2022;13:910236. doi:10.3389/fimmu.2022.910236

234. Habtamu K, Petros B, Yan G. Plasmodium vivax: the potential obstacles it presents to malaria elimination and eradication. Tropical Diseases, Travel Med and Vaccines. 2022;8:27. doi:10.1186/s40794-022-00185-3

235. Tajudeen YA, Oladipo HJ, Yusuff SI, et al. A landscape review of malaria vaccine candidates in the pipeline. Tropical Diseases, Travel Med and Vaccines. 2024;10:19. doi:10.1186/s40794-024-00222-3

236. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines — a new era in vaccinology. Nat Rev Drug Discov. 2018;17:261–279. doi:10.1038/nrd.2017.243

237. Patel PN, Tolia N. Structural vaccinology of malaria transmission-blocking vaccines. Expert Rev Vaccines. 2021;20:199–214. doi:10.1080/14760584.2021.1873135

238. Hayashi CTH, Cao Y, Clark LC, et al. mRNA-LNP expressing PfCSP and Pfs25 vaccine candidates targeting infection and transmission of Plasmodium falciparum. NPJ Vaccines. 2022;7:155. doi:10.1038/s41541-022-00577-8

239. 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. doi:10.1186/s12936-024-04972-5

240. Musoke D, Atusingwize E, Namata C, Ndejjo R, Wanyenze RK, Kamya MR. Integrated malaria prevention in low- and middle-income countries: a systematic review. Malaria j. 2023;22:79. doi:10.1186/s12936-023-04500-x

241. von Seidlein L, Hanboonkunupakarn B, Jittamala 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. doi:10.1080/21645515.2019.1643675

242. Duffy PE, orres JP, Healy SA, Fried M. Malaria vaccines: a new era of prevention and control. Nat Rev Microbiol. 2024;2024:1. doi:10.1038/s41579-024-01065-7

243. González-Sanz M, Berzosa P, Norman FF. Updates on Malaria Epidemiology and Prevention Strategies. Curr Infect Dis Rep. 2023;1–9. doi:10.1007/s11908-023-00805-9

244. Tine R, Herrera S, Badji MA, 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. doi:10.1186/s12936-023-04654-8

245. World Health Organization. Global Malaria Programme operational strategy 2024–2030. Available from: https://www.who.int/publications/i/item/9789240090149. Accessed November 5, 2024.

246. Ward CL, 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. doi:10.1177/1556264617742236

247. 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. doi:10.1371/journal.pgph.0002834

248. Osoro CB, Ochodo E, Kwambai TK, et al. 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. doi:10.1136/bmjgh-2023-014719.

249. Chitnis CE, Schellenberg D, Vekemans J, et al. Building momentum for malaria vaccine research and development: key considerations. Malaria j. 2020;19:421. doi:10.1186/s12936-020-03491-3

250. Greenwood B. New tools for malaria control - using them wisely. J Infect. 2017;74(1):S23–s26. doi:10.1016/s0163-4453(17)30187-1

251. Wangdi K, Banwell C, Gatton ML, Kelly GC, Namgay R, Clements AC. Malaria burden and costs of intensified control in Bhutan, 2006–14: an observational study and situation analysis. Lancet Glob Health. 2016;4:e336–343. doi:10.1016/s2214-109x(16)00083-8

252. Healer J, Cowman AF, Kaslow DC, Birkett AJ. Vaccines to Accelerate Malaria Elimination and Eventual Eradication. Cold Spring Harb Perspect Med. 2017;7. doi:10.1101/cshperspect.a025627.

253. 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. doi:10.3389/fpubh.2024.1433213

254. Amani A, Mboussou F, Impouma B, Cabore J, Moeti MR. Introduction and rollout of malaria vaccines in Cameroon and Burkina Faso: early lessons learned. Lancet Glob Health. 2024;12:e740–e741. doi:10.1016/s2214-109x(24)00101-3

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.