Skip to main content

Systems biology of malaria explored with nonhuman primates

Abstract

“The Primate Malarias” book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host–Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.

Introduction

Malaria continues to be an intractable disease, ravishing communities in about 100 countries [1]. The cause of this disease—Plasmodium parasites transmitted by Anopheles mosquitoes—has been known since the late nineteenth century [2]. The World Health Organization’s Global Malaria Eradication Programme’s strategic use of the insecticide dichlorodiphenyltrichloroethane between 1959 and 1965 was halted, between costs concerns and as experts recognized that a multi-pronged approach would be required to achieve the goal of malaria eradication (reviewed in [3]). Once molecular biological methods took hold in the 1970s, attempts to make a malaria vaccine became the envisioned panacea (reviewed in [4]). These efforts continue today, with the full recognition that making and introducing an effective malaria vaccine(s) is no small task, especially since the ultimate protection worldwide will require vaccines that have long-lasting immunity and are effective against multiple species of Plasmodium with their ever-changing genetic variation (reviewed in [57]). Many factors must be considered to ensure the effectiveness of vaccines in malaria endemic communities [8]. Meanwhile, a panoply of rapid diagnostic tests has come to the forefront [9], complementing the traditional gold standard microscopy detection of parasites in blood smears, and drug development advancements continue, leading to new treatments and combination therapies to overcome rampant drug resistance [10]. Research has advanced incrementally, leading to an understanding of the life cycle of malaria parasites in their vertebrate and invertebrate hosts, the multiple species and strains of parasites and mosquitoes involved, as well as the pathogenesis, immune responses, and epidemiology attributed to each species in human populations. The rise of the Internet in the 1990s became critical in this long fight, enabling communication and progress among scientists and communities in ways not imagined in the past. Lately, the era of systems biology has emerged along with controlled human malarial infection (CHMI) studies, promising more in-depth understanding of malaria, host–parasite interactions and immunity.

This article highlights malaria research from early and recent understandings gained from malariotherapy and modern CHMI studies to NHP infection studies and their current utility in support of today’s malaria eradication goals, with emphasis on the value of longitudinal infections and systems biological approaches. This article also pays tribute to the authors of "The Primate Malarias” [11, 12], as this publication has been instrumental for scientists who have followed in their paths.

Malaria parasite life cycles, within Anopheles mosquitoes and vertebrate hosts

Malaria parasites must successfully develop sequentially in the midgut and salivary glands of female Anopheles mosquito hosts and then be injected into the skin and survive in the liver and blood of humans, NHPs, or other vertebrate hosts. Some parasites must also transform within the blood of the vertebrate host into sexual-stage male and female gametocytes that are infectious for newly biting female Anopheles mosquitoes. The malaria parasite life cycles pose many challenges for both the parasites and hosts in their joint struggle to survive and propagate (Fig. 1). Throughout their transformative growth and development journey, the parasites must divide and multiply manifold, in the mosquito midgut, in hepatocytes, and inside red blood cells (RBCs). Importantly, throughout the process they must be able to overcome both innate and adaptive immune response barriers. Relapsing malaria parasite species, such as Plasmodium vivax (represented in Fig. 1, and discussed further below), have the added complexity of latent hypnozoite stages of the parasite in the liver and their possible activation that causes relapse parasitaemias in the blood, including the production of infectious gametocytes. Furthermore, P. vivax merozoites are restricted to young CD71+ reticulocyte host RBCs [13]. It is humbling to recognize the complexity of malaria parasites and their life cycles. Their size alone is large compared to viruses that are notoriously extremely difficult to control. For example, Plasmodium parasite genomes (discussed further below) comprise between 5000 and 7000 genes [1416] versus 16 genes coded by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus [17].

Fig. 1
figure 1

Schematic of the life cycle of Plasmodium vivax and comparable sibling simian species, depicted to represent the unique biological features of these species in the life cycle of primate malaria species and the importance of clinical and experimental interventions. The figure represents neotropical NHP models of P. vivax or macaque NHP models of Plasmodium cynomolgi and other simian parasite species that serve as surrogates for P. vivax (reviewed in [1820]). The purple and green icons indicate where natural events and experimental manipulations can take place. The green mosquito icons refer to the natural inoculation of sporozoites through biting and the purple mosquito icons refer to the natural biting and infection of Anopheles ssp. mosquitoes by drawing in gametocyte-infected blood. The green medical symbol and syringe denoting the inoculation of sporozoites into the human and NHP hosts, respectively, refer to the possibility of challenging these hosts after immunization with a vaccine candidate to determine if protection can be induced. The purple medical symbol and syringe denote the collection of blood for testing involving human and NHPs, respectively. The purple syringe also signifies the specific collection of blood containing gametocytes from NHPs to artificially feed and infect Anopheles mosquitoes for supporting experiments on host–parasite biology within the vector host, transmission blocking vaccines, and access of sporozoites for in vivo or in vitro infection experiments. The unique biological features of P. vivax and comparable species depicted are the hypnozoite, the preferential invasion of merozoites into reticulocytes, the production of caveolae vesicle complexes (CVCs), represented as a mottled appearance of the infected RBCs, and the early and rapid development and circulation of gametocytes. Red arrows refer to processes relating to features that are currently in need of special research emphasis, answering questions like: (1) What is the make-up of hypnozoites and how are they activated? (2) What are the similarities and differences in primary and relapsing liver-stage schizonts and is their biology with merosome release in the blood stream comparable to rodent Plasmodium species where these were discovered? [21] (3) Which critical factors are required for reticulocyte host cell selection, invasion, and growth in these cells? And (4) what factors determine the development and circulation of gametocytes, potentially permitting transmission from the early stages of a blood-stage infection? “Reprinted from [19], with permission from Elsevier”. The artwork was created by Nagib Haque

Malariotherapy and controlled human malarial infections (CHMIs)

From 1917 until 1976, malariotherapy was performed to treat patients with neurosyphilis and progressive paralysis (also known as general paralysis of the insane), as malarial infections caused fever, which can activate immune responses (reviewed in [22]). Over a period of 50 years (1923–1973), 17,000 patients were treated at the Mott Clinic for Malaria Therapy in England and numerous others underwent such therapy in Romania and the United States (reviewed in [2326]). Aside from the great benefits to patients who recovered from severe neurosyphilis manifestations, malariotherapy provided the means to study Plasmodium infections longitudinally in humans, including the collection of parasitaemia datasets, thereby creating significant knowledge regarding clinical immunity and parasitological features of the disease, albeit in some cases with grave illness and deaths recorded as a result—from Plasmodium falciparum, P. vivax, and Plasmodium malariae infections. Up to 15 malarial paroxysms were observed and monitored in malariotherapy patients. Snounou and Pérignon provide an extraordinary overview of the literature from that time and the numerous retrospective analyses that have followed [23]. Their book chapter, titled “Malariotherapy—Insanity at the Service of Malariology,” is riveting. Indeed, the authors are correct to caution readers that the voluminous literature backing their chapter could become addictive! Malariotherapy was considered unnecessary once penicillin was discovered in the 1940s as a cure for syphilis. Subsequently, such human treatment measures—and the associated collection of experimental data—were deemed unethical and the practice was progressively stopped over the next 30 years. In the latter part of this period, many malaria research studies were instead carried out with prison inmates [2730], and while ethical considerations were not addressed—to a similar extent as required today, the longitudinal infection data provided a basic understanding of malarial infections and disease progression. With gratitude shown for the participants, "The Primate Malarias” book is dedicated in part to the inmates at the United States Penitentiary in Atlanta, Georgia [28], who volunteered to accept infection with human and simian malaria parasites.

Many lessons were learned at the time, and since through the ongoing analysis of malariotherapy data (reviewed in [23]) and (also see [3134]). These studies also ignited interest and establishment of mosquito insectaries for rearing and infecting Anopheles mosquitoes, from which enormous parasite-vector knowledge has been gained. Infection with P. vivax was most common. Retrospectively, many overarching conclusions still stand, and these can now be further explored taking into consideration the many since-discovered biological complexities of each parasite species, their Anopheles mosquito vectors, their epidemiology, ecological factors, strain diversity, parasite and host genetics, and the immune systems of either naïve or primed individuals, male or female, young and old (reviewed in [25, 3538]). Some of the most prominent early discoveries beyond basic clinical and parasitaemic infection data include the demonstration of small exo-erythrocytic forms of P. vivax in liver-biopsies from a patient [39], now appreciated to likely have been hypnozoites (discussed below), the defining of the Duffy-blood group and its relationship with P. vivax susceptibility [40, 41], the dynamics of anaemia during P. vivax and P. falciparum infections and the conclusion that anaemia in acute cases is primarily caused by the removal of uninfected erythrocytes [31, 4244], a process that has also been recognized as a “bystander effect” imposed on a majority of RBCs when a much smaller number of erythrocytes are actually infected (reviewed in [45]).

Today, CHMI studies involving volunteers infected with sporozoites have become a modern—and important—means to test anti-malarial drugs and vaccines and gather natural infection data from individuals over a series of experimental time points until blood-stage parasitaemia is detected and subjects require treatment (reviewed in [4648]). In contrast to malariotherapy of the past, sporozoite-initiated CHMI studies have developed ethical standards that require treatment of individuals upon confirmation of parasites in the blood. Volunteers are monitored carefully and administered a full course of anti-malarial treatment that eliminates the parasites and the exacerbation of symptomology. As such, CHMI experiments have been very beneficial for studying Plasmodium infections from baseline periods to the time of parasitaemia, and helpful for conducting clinical vaccine trials that include sporozoite challenges (reviewed in [8]). Notably, correlates of protection are beginning to surface [49]. CHMI experiments initiated with sporozoites have also helped reveal specific protective immunological responses occurring during the early “pre-patent” period of infection, particularly in malaria-naïve hosts [50, 51], and new strategies for studying and intervening with gametocytaemia [5256]. The time series data collected in CHMI studies are dynamic compared to cross-sectional data, traditionally generated for decades from patient samples in malaria endemic areas, and they have uncovered heterogeneous immune responses among individuals (reviewed in [57]). Additionally, of great benefit, piggybacked projects associated with CHMI experiments have provided insights into the epigenetic and gene expression characteristics of the resulting blood-stage parasites and their impact on immune cells [5861].

CHMI studies have been mostly performed with P. falciparum, and close to a hundred such studies have been performed to date. Following suit, CHMI studies using P. vivax sporozoites have begun to emerge (reviewed in [62, 63]), including with vaccine trials [6466]. CHMI research with P. vivax was initially stymied due to additional, unique challenges of attaining infectious P. vivax sporozoites [6770] and the ability of hypnozoites to hide undetected in the liver and cause relapsing parasitaemias at some future point in time. Recent advances in the concentration of P. vivax gametocytes from infected volunteers [55] are helping to overcome the first of these hurdles, making it possible to generate P. vivax sporozoites from feeding Anopheles mosquitoes on well-characterized gametocyte-containing stocks and involving human volunteers with reliable infection records and medical histories. Thus, it may be possible in the future to perform such investigations routinely. At any rate, CHMI studies initiated with iRBCs are also now a viable option for investigations of P. falciparum [7174], P. vivax [53, 55, 7476] and P. malariae [77, 78]. Such CHMI studies involve low-dose blood-stage inoculations, with very clear ethical guidelines in place to ensure the safety of volunteers.

While CHMI experiments are generally safe and have demonstrated clear advantages for scientific advancements in understanding malaria, the viewpoints and concerns of participants and their communities and other stakeholders remain important to understand and take into consideration [7982].

NHP infection models (NHPIMs)

NHP infection models (NHPIMs) have been vital to research on malaria and over 70 human infectious diseases, including many caused by viruses, bacteria and other parasites [83, 84], and notably, recently for Ebola [85], Zika [86] and SARS-CoV-2 infection and coronavirus disease-2019 (Covid-19) [87]. With rapidly advancing technologies, NHP species chosen to best suit specific experimental questions and requirements increasingly hold much potential for complementary research on malaria, malarial immunity, and pathogenesis, as demonstrated in recent longitudinal experiments (discussed below). Given the close evolutionary relationship of humans and macaque species, many basic elements of the immune system are shared among them, making Old World monkey macaque species generally the most widely used NHP animal models for infectious disease research. Just as in humans, there is diversity in genes that comprise components of NHP immune systems. In turn, many reagents developed for the study of immune responses in mice or humans will not necessarily crossreact and be functional in studies of the NHP immune responses. However, various mouse, human, and NHP reagents have been confirmed to crossreact with conserved epitopes and others are under development and testing; e.g., see the NHP Reagent Resource web page [88].

Malaria with its myriad of host–parasite interactions throughout the parasite’s life cycle (Fig. 1), which are only beginning to be understood at the molecular level, is overwhelmingly complex. The complexity is compounded by the biological differences between species of Plasmodium, as elegantly detailed in “The Primate Malarias” [11, 12]. As also acknowledged by others [89], the authors of “The Primate Malarias” had the foresight to study and capture ‘now classic’ iRBC illustrations and basic morphological information regarding various human and NHP malaria parasite species and their life cycles in primates and Anopheles mosquito hosts, documenting nuances that distinguish species and strains, and experimental NHP and mosquito host infections. This all helped to set the stage for the use of different host–parasite combinations as model systems for research—whether for drug or vaccine candidate testing, immune response, or pathogenesis studies. The authors described infections of apes and Old World and New World monkeys, the most studied today being macaque (Old World) and Saimiri and Aotus (New World/neotropical) species. Numerous experiments had been performed to determine whether infections initiated by sporozoites of various species of Plasmodium infect and thrive in the liver and blood, or only the liver, and whether gametocytes were observed. Blood infections were studied including the level of infectivity of the NHP blood to mosquitoes. Numerous publications have followed since, by these authors and others, continuing to shine light on the utility of these model parasite species and the importance of NHP models for malaria research (reviewed in [1820, 9095]). The Plasmodium-NHP infection models have been and can continue to be useful for addressing specific hypotheses that may be challenging or not possible with humans, including when considering confounding factors among individuals within different populations and epidemiological and ecological situations worldwide (discussed in [25, 35, 96, 97]).

Differences in Plasmodium infections in splenectomized animals has also been documented in “The Primate Malarias” [11, 12] and is important to recognize. Humans and NHPs are more susceptible to higher parasitaemic blood-stage infections when lacking a spleen, and various experiments over time have been performed with parasites that have been ‘passaged’ through splenectomized animals and then frozen as iRBC stabilates for future use. From the 1960s until the turn of the twenty-first century, splenectomized chimpanzees had in fact been a source of occasional infectious samples to study the main four human malaria parasite species (P. falciparum, P. vivax, P. malariae and P. ovale) [98102] (reviewed in [20]), but these types of experiments are no longer permitted, given ethical concerns and restrictions [103].

Importantly, while splenectomized NHPs have yielded higher parasitaemias of different species for various experimental purposes, and their use has been helpful to ensure the availability of long-term frozen infected RBC (iRBC) stocks for future use, such stocks and splenectomized animals are not advised for immunological and pathology studies seeking to understand the normal course of host–parasite interactions and disease ramifications. Aside from the spleen’s prominent role in removal of damaged RBCs, the spleen has been associated with the expression of parasite virulence factors, namely the var/SICAvar variant antigen genes and proteins at the surface of Plasmodium-iRBCs, specifically for P. falciparum, P. coatneyi, and P. knowlesi, and likely P. fragile (reviewed in [92]). Plasmodium knowlesi-iRBCs were shown to become less virulent after passage through splenectomized animals [104, 105], resulting in the downregulation of the expression of this gene family [106]. Splenic influence on antigenic variation has also been reported for P. falciparum-iRBCs in Saimiri sciureus monkeys [107] and in humans [108]. A study based on Aotus monkey infections has also indicated that the spleen influenced transcription of 67 P. vivax genes [109]. Moreover, the spleen has become recognized as a niche for a subpopulation of P. vivax iRBCs, possibly to promote their invasion and replication in host reticulocytes (reviewed in [110]). Interestingly in this regard, P. cynomolgi infections became attenuated when passaged in splenectomized rhesus [111]. The spleen may also be a niche for P. cynomolgi. While this remains to be explored, a computational model revealed that during infection of Macaca mulatta (rhesus monkeys), a subpopulation of P. cynomolgi-iRBCs becomes ‘concealed’ from the circulation [112]. In summary, the role of the spleen may prove to be distinctive for species that express var/SICAvar genes and proteins, compared to others that do not possess comparable genes and proteins. These distinctions are important as they relate to virulence and cytoadhesion and sequestration of iRBCs, or concealment of iRBCs, respectively.

Interestingly, the preface to “The Primate Malarias” noted that this book was timely in 1971 given the recent awareness that monkey malaria parasites can also infect humans [11, 12]. This comment well preceded the landmark studies of Singh et al. showing that human cases diagnosed in Malaysian Borneo as P. malariae were in fact P. knowlesi zoonoses [113]. Since that time, P. knowlesi has been documented as a zoonosis throughout most of Southeast Asia and as the main type of human-causing malarial disease in Malaysia [114]. More recently, P. cynomolgi has become recognized in the same geographical area as an additional zoonotic parasite of public health concern [115, 116] (reviewed in [89, 117]). Recently, adding to these concerns, a few cases of P. coatneyi, P. inui, Plasmodium inui-like, and P. simiovale malaria were identified in Malaysia when using molecular methods [118]. Geographical studies have been performed mapping Macaca fascicularis and the less-accessible macaque species Macaca nemestrina and Macaca leonina, highlighting their likelihood as well as possible hosts and sources of zoonotic P. knowlesi infections [119], and it is noteworthy that Macaca arctoides was recently identified as another macaque NHP species that can harbour this simian malaria parasite and be a possible source of zoonotic infections [120]. New World monkey infections and zoonoses involving P. simium, which is genetically and biologically similar to P. vivax [121], and P. brasilianum, which is genetically and biologically similar to P. malariae [89, 122], and P. knowlesi zoonotic infections are the subjects of other papers in this thematic issue.

Many basic biological and clinically relevant questions can be asked in NHP infection experiments (see Box 1). Longitudinal experiments can be designed without the urgent need to treat, and with repeat collection of multiple immunologically relevant tissues, thus capturing data not typically possible in CHMI studies. A holistic view of the immune response is possible with NHPIMs, especially with the Old World macaque models, including the collection and analysis of key tissues in addition to peripheral blood (e.g., skin, bone marrow, lymph nodes, liver, spleen) and integrated analyses of multiple data types during infections and from necropsies. Parenchymal tissues, secondary lymphoid tissues and the lymphatic/humoral circulation all hold relevant information. Over the past few decades, preclinical vaccine trials using NHP models have helped to test the immunogenicity of vaccine constructs, dosing, delivery platforms, and sometimes protection with parasite challenges. Such experiments have been cost-effective in the vaccine development pipeline. Today, these can be perfected to maximize these benefits.

Box 1 Questions that can be addressed with NHP models

Parasite biology

• What molecular processes support hypnozoite development, maintain their dormancy, and trigger their activation?

• How do hypnozoites manipulate hepatocytes to suppress innate immune mechanisms from inside the cell and preserve them until activation?

• How do malaria parasites enter and remodel the infected RBC in vivo to suit their prime directive of multiplying and releasing new merozoite progeny?

• What is the molecular genesis of gametocytogenesis of non-falciparum malaria parasites?

Immunology

• What are the mechanisms that thwart the formation of long-lived serological anti-parasite antibody responses, while memory cells remain persistent and protective?

• What are the host–parasite mechanisms that enable antigenic variation and immune evasion?

• What is the role of the spleen in these processes?

• How does pre-existing immunity against malaria parasites impact vaccine-generated responses and subsequent infections with genetically heterologous parasites?

• How do immune responses that are detected in the blood stream differ from what is occurring in various tissues and the systemic development of NAI and VCI?

• What are key identifiable immune correlates of protection for NAI and VCI?

• What immune responses and host molecules contribute to the development of anaemia?

Pathogenesis

• What systemic ramifications may occur due to the presence of LSFs in hepatocytes?

• Can peripheral blood metabolites become diagnostic for hypnozoites?

• What factors cause disease pathogenesis, inflammation, and pathology in various tissues?

• What roles do extracellular vesicles play, or the microbiota, and in relation to immunity?

• What factors contribute to loss of uninfected RBCs and bone marrow dysfunction?

• What host-directed therapies can be developed to reverse adverse manifestations of malaria?

Vector-parasite biology and transmission

• The field is wide open for the application of the latest technologies to study mosquito infectivity, transmission, and subsequent host–parasite expression and interactions in NHPs

Old World monkeys—macaque species

Old World monkey species are valuable for researching the dynamic mechanistic intricacies of malarial naturally acquired and vaccine candidate-induced immunity (NAI and VCI), as well as gametocytogenesis and transmission. They also hold much potential for studying malarial pathogenesis and immune evasion mechanisms, and they are the preferred NHP species for longitudinal systems biological infection experiments. Significantly, the four known human malaria species (P. falciparum, P. vivax, P. malariae and P. ovale) do not develop in the blood of macaques. Simian malaria parasites, with comparable biology to the human malaria parasite species, are therefore relied upon when using these animal models. However, it is worth noting that while P. falciparum blood-stage parasitaemia does not develop in rhesus macaques, rhesus can be infected with P. falciparum up to the liver stage [123, 124]. Based on infection studies in M. mulatta, P. coatneyi has become recognized as a model for P. falciparum, P. cynomolgi as a model for P. vivax, and P. knowlesi as a model for both, depending on the research question. The history and prospects for using these species were reviewed recently for P. coatneyi [95], for P. cynomolgi [20], and for P. knowlesi in general terms [94] and with particular focus on antigenic variation [92].

The macaque NHP animal models develop a spectrum of malarial disease manifestations that closely resemble the human disease states caused by either human or simian malaria parasites. For example, P. coatneyi infections in rhesus macaques recapitulate malaria complications associated with pregnancy caused by P. falciparum, including with placental pathology and iRBCs localized in this tissue [125128]. Plasmodium coatneyi infected rhesus also experience anaemia [129131], with evidence of infected RBC sequestration and possible cerebral involvement [132134]. Thus, this is a useful model for studying various aspects of P. falciparum pathogenesis (reviewed in [95]). In contrast, P. cynomolgi in rhesus macaques mirrors P. vivax relapse phenotypes [135138], and this host–parasite pair has become recognized for studying primary and relapsing malaria parasite pathogenesis, with benefits understanding asexual and sexual stage immune responses and anaemia [139, 140]. Experimental studies with rhesus have also confirmed that P. cynomolgi causes placental pathology during pregnancy, which can result in severe pregnancy outcomes [141144]. Plasmodium knowlesi infection of olive baboons (Papio anubis) has also been shown to be a model for studying malarial pathology including cerebral malaria [145] and malaria during pregnancy, with confirmed sequestration of iRBCs in the placenta [146]. Recently, Japanese macaques (Macaca fuscata) have been shown to be an additional viable NHP model for relapsing cynomolgi malaria [147]. Juvenile and adult macaques used for malaria research generally range from about 5–15 kg, allowing for adequate blood and bone marrow sample access at experimental time points and other tissues of interest selectively through biopsies or during necropsy.

Historically, malaria research involving macaque species has been carried out in the United States, the United Kingdom, The Netherlands, China, Thailand, India, Sri Lanka, and other parts of Southeast Asia. Research with these species is currently prominent in The Netherlands, Thailand, Japan, China, and the United States, where different species of macaques are routinely available through domestic breeding programmes or importation. In the United States, National Primate Research Centers (NPRCs), in addition to governmental laboratories, academic institutions, and private businesses, breed and/or may acquire macaque species for malaria research. Macaque species have been generally available as required for malaria research experiments, although at the time of this writing, their availability has been diminished due to their widespread use in SARS-CoV-2/Covid-19 experiments. This situation is likely to reverse, thanks to the development and testing of effective drugs and vaccines to fight the SARS-CoV-2/Covid-19 pandemic. Currently, as a result, severe Covid-19 illness has been subsiding [148].

New World monkeys— Saimiri and Aotus species

New World monkeys of the genus Aotus and Saimiri have been critical and will remain important for P. falciparum and P. vivax vaccine candidate trials, drug studies, and research into immune responses and pathogenesis caused by these parasite species, as well as basic research involving simian malaria parasite species (reviewed in [1820, 26, 90, 91, 149, 150]). These monkey species have also been used to study the infectivity of the human parasite species P. malariae [102] and P. ovale, with the latter only shown to develop liver-stage parasites but not blood-stage parasites [101]. New World monkey systems-based research limitations include repeated tissue sampling and volume limitations imposed by their small size (~ 1 kg). In addition, specialized knowledge and husbandry are required to work with these animals, and there is currently a dearth of reactive biological reagents available for studying these species in systems-scale analyses [88]. Nevertheless, each of these models remain highly relevant to understand some aspects of immunity and pathogenesis. Data from each can improve upon interpretations of cross-sectional data from human studies and thus support intervention-based studies, and when ethical, support the collection of tissues to evaluate temporal changes that lead to specific clinical complications and can reveal hidden parasite niches (recent examples for P. vivax in Saimiri boliviensis and Aotus lemurinus include [151, 152]). Also, a recent insightful example of IgG, IgM, and IgA antibody isotype cross-reactivity relationships and lessons learned between P. falciparum-challenged human and Aotus nancymaae responses was shown using protein microarray technology [153155]. Critically, as discussed below, the continued use of these animal models for generating infectious gametocytes for P. vivax research is paramount, particularly to research P. vivax hypnozoites and relapse biology. Historically valuable P. vivax isolates (over two dozen) that had been adapted to grow in New Word monkeys and stored as frozen iRBC stabilates are highly relevant today. These show individualized characteristics ranging from frequent early relapsing phenotypes (e.g., the Chesson strain) to late-relapsing phenotypes (North Korean strain) [1820].

Aotus and Saimiri monkey species are currently available from the Keeling Center for Comparative Medicine and Research at The University of Texas MD Anderson Cancer Center, Texas, USA, which has maintained U.S. National Institute of Health (NIH)-supported New World monkey breeding colonies for medical research purposes, including malaria research. Opportunities for malaria research with New World monkey species also exist in Peru, Panama, Brazil, and Colombia.

Twenty-first century—turning point in malaria research

Within this article, following an overview of relevant background information, systems biological studies and experimental frameworks are highlighted involving longitudinal NHP infections initiated with P. coatneyi, P. cynomolgi, and P. knowlesi sporozoites, from which basic biological knowledge has and continues to be gained regarding host infections initiated with each of these species. This research, conducted by the Malaria Host–Pathogen Interaction Center (MaHPIC), has involved longitudinal Plasmodium infections using macaque species, the most common being M. mulatta (rhesus monkeys) and M. fascicularis (kra monkeys, also known as long-tailed macaques, or cynomolgus monkeys).

NHP research in the modern era of malaria systems biology and malaria eradication

It is well accepted that the host and parasite influence each other, and host–parasite interactions can set off cascades of molecular events and pathways, both locally and systemically, as each thrives to survive. The simian malaria parasites and their NHP model systems continue to provide unique research opportunities for in-depth investigations, aimed to ultimately inform translational solutions to prevent or treat malaria caused by any of the various Plasmodium parasite species infecting people. NHPIMs cannot recapitulate all health conditions and situations pertinent to humans, but they can begin to take them on, one or a few at a time; tease them apart, deconvolve large datasets, and reveal hidden biological information. It is becoming increasingly possible to gain a multi-dimensional view of the immune system in action, which is critical for determining: (a) what works to the host’s advantage, resulting in reduction or elimination of parasites, inflammation, and pathology, (b) what works to the host’s detriment, resulting in thriving parasites, illness, and pathology, and ultimately, and (c) what interventions may help favour the host. These three areas of inquiry remain at the forefront today.

Malaria eradication goal raises the bar to tackle all species of Plasmodium and prioritize vaccines

In 2007, a new concerted effort to eradicate malaria began with a declaration by Bill and Melinda Gates to eradicate this disease [156]. Much debate followed [157] and continues today [158] on the feasibility of malaria eradication, and the challenges and bottlenecks faced [159, 160]. With the return of malaria eradication as a global goal, new projects were initiated to better document malaria cases and intervention methods being used regionally and globally. Gaps in knowledge were evaluated, and new research agendas developed to help fill those gaps, including for P. vivax which became recognized as a widespread but important and neglected species [161, 162]. These steps led to improved information about the prevalence, morbidity and mortality of both most predominant human malaria species, P. falciparum [163] and P. vivax [164, 165], as well as public recognition that global eradication will also require the elimination of the lesser prevalent human malaria species P. malariae and P. ovale, as well as simian malaria zoonotic species (reviewed in [89, 117, 118, 166, 167]). Three of these zoonotic species in particular, P. cynomolgi, P. knowlesi, and P. coatneyi, have been critical for studying malaria in vivo with NHP infections (reviewed in [92, 94, 95, 168]). Notwithstanding, P. falciparum research continues to predominate, given its well-known potential for serious illness and lethal outcomes, though P. vivax has gained recognition and research support as a parasite species that is widespread, debilitating, and which can be fatal (reviewed in [169]).

Researchers continue to question how to make the next big leap and meet the demands of malaria eradication [158, 160, 170, 171]. The successful evolution of host–parasite interactions favours the survival of both the parasite and its vertebrate and invertebrate hosts. The world’s challenge to break the cycle of malaria parasite transmission between—and development within—mosquitoes and humans is huge, yet dedicated researchers over the decades have remained committed to seeking sufficient understanding to interrupt transmission, foremost, with new vector control solutions, diagnostics, drugs, and vaccines [160]. Yet, much remains to be learned about the disease caused by the different Plasmodium species and their transmission dynamics, including ‘within-host’ and involving external factors, operating in a multitude of environments. Indeed, the enormous complexity of this overall system has led to new efforts utilizing mathematical and computational modelling strategies to study the problems and trajectories [172174]. Top goals to achieve malaria eradication include treatment of infected individuals, whether they are symptomatic or not [175], with the goal of getting rid of all parasite subpopulations (circulating or not), including gametocytes [176] that transmit the disease to Anopheles mosquitoes where fertilization occurs and a new brood of parasites forms.

Vaccination is considered necessary to ultimately prevent infections and reduce (or eliminate) the global burden of malarial disease in humans (reviewed in [177]). While challenging to achieve, in-depth knowledge of malaria caused by each Plasmodium species, NAI, VCI, and malarial disease processes is critical for the development and testing of broadly effective vaccine strategies. Immunological comparisons between malaria naïve and chronically infected individuals are also important, particularly with regards to predicting and understanding the outcomes of candidate vaccine testing in malaria endemic areas with different transmission patterns and intensities, and when a large percentage of individuals may be repeatedly or chronically infected with malaria parasites and have significant levels of NAI against malaria.

Raising the profile of Plasmodium vivax and its close relative, Plasmodium cynomolgi

Especially in light of the current malaria eradication agenda, P. vivax has gained widespread special attention as a predominant malaria parasite species—with raised public health concerns [161, 162, 178184]—along with the closely related simian malaria species P. cynomolgi [185, 186]. Each of these species and a few others including P. ovale produce dormant parasite-infected cells in the liver that can stay quiescent for weeks, months or years and then become activated to cause relapsing infections in the blood in the absence of new mosquito infections. Consistent with raised attention on this problem, review articles on hypnozoites and relapses have become frequent [19, 20, 26, 187194].

The biology of the P. vivax, P. ovale and P. cynomolgi latent parasites, called hypnozoites [195] (Fig. 1), is largely unknown. Hypnozoites comprise a large hidden subpopulation of the total P. vivax parasite biomass [196], and treatment options to kill them are currently limited to primaquine and tafenoquine, both with contraindications for pregnant individuals and anyone with glucose-6-phosphate-dehydrogenase deficiency; thus neither are ideal drugs [10, 166, 197]. Hypnozoites have low metabolic activity and are few in number in the liver, and therefore they have been a challenge to study and in turn discover essential biologically pathways to help advance non-haemolytic radical curative treatments. As such, they are a major barrier for malaria elimination and eradication campaigns [182, 184, 198200]. The lack of robust in vitro parasite culture systems in the past has been considered an impediment to basic research on hypnozoites, but research with each of these species has remained viable over the past few decades thanks to the availability of ex vivo clinical samples from patients and NHP infections.

What causes these species to go dormant in hepatocytes—seemingly unaffected by the immune system; and what causes the dormant infected cells to reactivate and produce broods of thriving infectious parasites once again capable of causing blood-stage infections? While hypnozoites were discovered almost 40 years ago—in rhesus macaques (infected with P. cynomolgi) and chimpanzees (infected with P. vivax) [201203], they were not researched with rigor until recently. Given recent investments and the persistence of scientists, hepatocyte culture systems have been established over the last several years for P. vivax [204, 205] and P. cynomolgi [206208]. Robust P. cynomolgi in vitro blood-stage cultures [209] have also become a reality and these are enabling experiments that complement in vivo testing of hypotheses [210, 211]. Hepatocyte cultures are enabling hypnozoite transcriptomic data on P. vivax [204, 212214] and P. cynomolgi [208, 215]. Recently, such in vitro cultures with P. vivax revealed parasite-derived membraneous networks in both schizonts and hypnozoites and possible functions of aquaporin-3 [216]. This type of research comes with many challenges, given the need to distinguish quiescent infected cells that have low metabolic activity from developing or newly activated forms [217, 218]. Today, such culture systems, single-cell -omic technologies [219], and cellular imaging advances [220, 221] are paving the way for using these model systems to understand P. vivax sporozoites [222] and host–parasite interactions in infected hepatocytes, including dormant and activated hypnozoites [223].

NHP models have become an important if not critical resource for enabling ongoing research with hepatocyte cultures. First, they have been vital for establishing gametocytaemia and feeding mosquitoes to generate P. cynomolgi and P. vivax sporozoites to initiate hepatocyte culture infections. Notably the first hepatocyte culture system to show results involved M. fascicularis and P. cynomolgi. P. cynomolgi blood-stage parasites were grown in macaques to generate gametocytes to feed mosquitoes to develop sporozoites to infect M. fascicularis hepatocyte cultures [207]. More in-depth studies can now be performed with NHPIMs, accessing P. cynomolgi or P. vivax gametocytes from infected NHP blood for mosquito feeds, and conducting ex vivo experiments with liver tissue samples.

Importantly, as the number of P. vivax clinical cases declines in endemic areas [1], the demand for NHP resources for investigating hypnozoites is expected to rise. Specifically, during the last decade, institutions in Thailand, Cambodia and India developed clinical capabilities and insectary operations to infect mosquitoes with human blood to attain sporozoites to infect hepatocyte culture systems [204, 205, 224]. These feats have been groundbreaking, but—if P. vivax clinical cases continue to decline, as hoped—logistical challenges will be compounded that include having sufficient availability of patient infected-blood donors, mosquito insectary operations, and experts for establishing, infecting, and analyzing the data coming from hepatocyte cultures. Meanwhile, there is much yet to be learned about the biology of hypnozoites to effectively identify targets for drug intervention, and multi-omic approaches that can distinguish host and parasite targets will be essential [218, 223]. With such knowledge and systems biological analyses, vaccination to eliminate hypnozoites, or at a minimum delay or reduce the number of relapses [225], may one day also become a reality, in addition to targeting the universal parasite developmental life cycle forms or stages known across all species as liver-stage forms (LSFs, or exoerythrocytic stages) and blood-stage forms (BSFs, or erythrocytic stages) (Fig. 1).

Relapsing malaria

Relapses are the ‘strong suit’ of P. vivax malaria and potentially P. ovale—for which less data is available [226228]. P. vivax relapses are responsible for over 50% and potentially as high as 95% of blood-stage infections caused by these species [184, 198, 200, 229]. Yet, little to nothing is known specifically about these infections and immune responses generated against them. Malarial disease results from primary acute blood-stage infections with Plasmodium parasites, and recurrent infections. Studying relapsing malaria, caused by the activation of dormant P. vivax hypnozoites, is especially challenging, impractical—and arguably not possible—in human populations, where a main challenge is the difficulty of distinguishing relapse infections from new mosquito-borne infections. There are a few praise-worthy exceptions on the feasibility questions. Two major studies used molecular methods including whole genome sequencing and protein microarray analyses to distinguish new infections from relapses [230, 231]. Others involved the relocation of volunteers from malaria endemic areas of Cambodia to ‘non-transmission’ regions to ensure that any newly detected parasites in the blood were bona fide relapses [232, 233].

Fortunately, P. vivax relapses can also be studied via controlled experimental infections of New World monkeys, thus overcoming the challenge of discerning relapses from new infections in human studies. Monkey-adapted isolates of P. vivax can be capitalized upon that exhibit different relapse patterns (reviewed in [1820, 26]). Aside from P. vivax, P. cynomolgi in macaques remains a strong model for in-depth investigations of relapses, driven by the fact that macaques are most closely related to humans and P. cynomolgi is a closely related sister species to P. vivax (reviewed in [20, 190]). Moreover, as noted above, P. cynomolgi is a zoonotic species in Southeast Asia and thus has direct relevance as an infectious disease agent for humans [115117]. Critically, longitudinal studies of P. cynomolgi allow for controlled investigations that can distinguish one or more “bona fide relapses” from new infections or the rise or “recrudescence” of persisting low-level blood-stage parasites. Experimentally controlled relapses were first studied by Schmidt and colleagues using P. cynomolgi in rhesus macaques [135137] and subsequently by others ([138, 139] and unpublished data) (see MaHPIC experiments below, called E04, E23, E24 and E25). Experimental infection of M. mulatta and other macaque species, initiated with P. cynomolgi sporozoites, can be designed to mimic primary, relapsing, and new infection occurrences, and such experiments are particularly well suited for studying host immune responses that develop during each of these distinctive blood-stage infections, including with parasite challenges involving homologous and heterologous strains of P. cynomolgi. As discussed below, systems biological approaches are being applied by the MaHPIC to assess the cascade of immune cell types, niches, and memory recall responses that result in naturally acquired protection against P. cynomolgi, while predictably enabling ongoing transmission to Anopheles mosquitoes ([139, 140] and unpublished data).

Gametocytes—the link to ongoing new infections

Circulating gametocytes remain undetected if an infected person is asymptomatic and not seeking treatment and there is no active case detection program in place in their community to identify and treat such individuals. This problem is especially challenging for primary infections with P. vivax, as the P. vivax gametocytes circulate soon during infection of the peripheral blood where they may be accessible to biting mosquitoes before individuals feel sick and seek medical attention [162, 235, 236]. Infectious gametocytes have also been confirmed in asymptomatic, chronic P. falciparum, P. vivax, and P. malariae infections, and they are, therefore, likely present as well in P. ovale infections [235, 237242]. Further, they have been observed in P. falciparum CHMI studies soon after drug treatment [54, 243, 244]. Thus, for multiple and possibly all species, infectious gametocytes can be ingested by mosquitoes before anti-malarial drugs are taken or the immune system is effective, safeguarding their viability and ability to mate in the mosquito and propagate the disease. Today, longitudinal infections in NHPs provide opportunities to effectively study gametocytes and parasite transmission from a systems biological perspective under a variety of experimental conditions and scenarios [131, 139, 245, 246, 234].

Tissue niches: bone marrow and spleen

Cytoadhesion of P. falciparum-trophozoite and schizont-iRBCs to endothelial receptors and their concomitant sequestration in the deep vascular tissues have been known related phenomena for over 50 years, which lead to organ pathology, and explain why only the young P. falciparum ring-stage iRBCs are typically seen in peripheral blood smears (reviewed in [247, 248]). This is the case as well for the P. coatneyi and P. fragile simian malaria parasites with comparable ‘knobby’ iRBC surface structures and var/SICAvar genes and exposed adhesive variant proteins [249254], predicted to be comparable to the adhesive P. falciparum Erythrocyte Membrane Protein-1 (PfEMP-1), which has domains known to adhere to various endothelial receptors [255, 256]. While cytoadhesion and sequestration have been proposed as evolutionary adaptations to avoid iRBC passage and destruction in the spleen, this is not fitting for the majority of Plasmodium species for which all developmental iRBC forms circulate. Questions remain regarding what other functions the adhesive PfEMP-1 and related proteins may serve for the parasites [257, 277].

Recently, an alternative concept called ‘concealment’ was put forth for species including P. vivax and P. cynomolgi to account for non-circulating iRBCs that do not express EMP-1 related proteins [24, 97, 112]. In these cases, other types of host–parasite interactions must account for the removal of subpopulations from circulation and possible associated disease manifestations. Above all, what has become clear, is that for all Plasmodium species, the circulating iRBCs represented in blood smear parasitaemias, do not necessarily represent the entire parasite load, or parasite burden in infected individuals (reviewed in [24, 97, 110, 196]). First, there can be multiple broods of parasites in an individual, each in different stages of the life cycle. Second, there can be an indeterminable number of parasites hidden in various tissues.

In the early stages of infection, P. falciparum gametocytes have been found in the bone marrow, where their maturation takes place [258261], and the bone marrow has also been recognized as a development niche or reservoir for P. vivax gametocytes [262]. Surprisingly, the spleen has also become recognized as a biologically relevant tissue site for P. vivax iRBCs in humans [263265], presumptively to support reticulocyte host cell invasion and the asexual blood-stage development cycle (reviewed in [110]). Aotus and Saimiri monkey NHP infection studies have demonstrated and quantified the burden of P. vivax iRBCs in these tissues relative to others [151, 152, 266]. Further studies using these animal models are now warranted, focused on local and systemic relationships of the infected bone marrow and spleen. Based on longitudinal parasitaemic data from P. cynomolgi infected M. mulatta, examined to better understand P. vivax infections and anaemia, a computational model was used and concluded that a subpopulation of iRBCs was in fact concealed in the tissues [112]. As with hypnozoites, these hidden parasite populations present bottlenecks for malaria eradication efforts.

Plasmodium knowlesi iRBCs have been identified in the tissues of experimentally infected M. fascicularis and M. mulatta [267, 268, 277]. In addition to vital lung, liver and kidney organs, among others, showing high levels of P. knowlesi parasitization, tissues of the gastrointestinal tract stood out, consistent with gastrointestinal complications in patients with P. knowlesi [268, 277]. P. knowlesi infection of baboons has also resulted in the localization of parasites in vital organs, including the brain and placenta [145, 146].

Malaria vaccines, from darkness into the light

Throughout the 1980s, once a few malaria parasite genes had been characterized, a significant R & D focus developed towards a vaccine mimicking immunogenic regions of the parasite’s immunodominant circumsporozoite protein (CSP), hypothesizing that such a vaccine could prevent sporozoite infection of hepatocytes (reviewed in [4]). However, antibodies against this protein proved insufficient to inhibit infections completely, and once the parasites multiplied in even one liver cell, tens of thousands of invasive merozoites were subsequently released into the bloodstream. The need to raise effective B cell, T cell and other cell-mediated immune responses, and optimize adjuvants and delivery systems, took hold, resulting in vaccine renditions that included, among others, the CSP-based P. falciparum RTS,S candidate vaccine (reviewed in [4, 269282]), and alternative CSP-based multi-valent vaccine plans for P. vivax [63, 65, 283, 284]. Recently, the P. falciparum RTS,S/AS01 vaccine product (Mosquirix™) was approved for widespread use in Africa after showing roughly 30% efficacy in preventing severe malaria in children after four sequential injections [285, 286]. This malaria vaccine will be the first to be widely implemented, and thus, it will allow the research community to observe, for the first time, the possible benefits as well as effects on local parasite genetics and dynamics, disease prevalence, and transmission that can result from the introduction of a subunit malaria vaccine based on an immunodominant protein representing a single species and strain [287, 288]. This achievement has and will continue to pave the way for testing of other currently available and future-generation vaccine candidates [280, 281, 289293].

As it stands, the world is still far from being close to having one or more malaria vaccines that can completely prevent malaria from one species, let alone all four human Plasmodium species and zoonotic infections. Uncounted lessons have been learned during the roughly 30 years of RTS,S development, testing, delivery/administration, and monitoring phases. While sterilizing immunity—killing all parasites—has yet to be reached in large clinical trials, it must remain the goal as malaria vaccinology work continues alongside research to understand each parasite species’ biology, diversity, antigenic variation, and evasion strategies, as well as host immune responses, including those that are protective and those that may be detrimental and cause pathology. Moreover, differences in responsiveness between malaria naïve volunteers and people living in different malaria endemic areas must be considered (reviewed in [8, 294]). Ultimately, it is hoped that useful predictive immune signatures and correlates of protection will be identified, and that future vaccine candidates will be tested with much more relevant knowledge known about the likely, and requisite, host responses necessary to achieve protection, if not sterility (i.e., the complete elimination of parasites in an individual).

Since the 1990s, multipronged approaches have been increasingly pursued against multiple parasite targets and life cycle developmental stages, including the sexual stages (Fig. 1), in an attempt to block successful transmission and development of parasites within malaria endemic mosquito populations [295, 296]. Major investments over many decades have advanced malaria vaccine development as a top priority, among government agencies, industries, and foundations, including the Bill and Melinda Gates Foundation (BMGF). The European Malaria Vaccine Initiative (EMVI), launched in 1998, complemented several European malaria vaccine research initiatives [297]. The US-based Malaria Vaccine Initiative (MVI) was launched in 1999 with the benefit of $50 million in start-up support from the then newly developing BMGF, and the MVI leadership team scouted out and supported projects and approaches around the world that they deemed promising, mostly towards P. falciparum vaccines [298], but also including the P. vivax Duffy Binding Protein (PvDBP) as a one-shot key prospect immunogen to prevent entry of P. vivax merozoites into Duffy glycoprotein-positive host RBCs (reviewed in [299301]). These DBP vaccine efforts follow a body of research showing the importance of the Duffy glycoprotein on the surface of RBCs [41] as a receptor for the P. vivax DBP, which is located at the front (apical) end of the merozoite and is known to specifically attach to the cognate RBC receptor prior to invading the host RBC [299, 302304]. Complicating the situation, in the ensuing years, through research in multiple locales, it has become apparent that P. vivax can also invade Duffy negative RBCs by utilizing alternate invasion mechanisms (reviewed in [305, 306]).

Discovery of candidate vaccine proteins and testing them continues, predominantly to develop P. falciparum vaccines [279, 282, 284, 290, 295, 300, 307, 308]. Nonetheless, P. vivax research has been progressing at a competitive pace, even prior to the genome sequencing era, greatly aided by the use of both Aotus and Saimiri monkey infections and short-term in vitro blood-stage cultures permitting studies on P. vivax from human infected blood samples [303, 309, 310]. Over the span of several decades, these animal models have been critical for obtaining P. vivax parasites for genetic and biological analyses (reviewed in [19, 20, 311]), especially in light of the absence of a reliable and robust long-term blood-stage culture system for P. vivax [312314], as has been available for P. falciparum for decades [315]. From a vaccine development standpoint, such research—and the use of the related P. cynomolgi and P. knowlesi parasites for fundamental basic biological studies and to generate crossreactive antibody reagents and perform basic experiments—has been critical for the identification of immunogenic merozoite surface and apically located proteins, which have been regarded as vaccine candidates (reviewed in [19, 25, 161, 299, 301, 311]). These include the PvDBP and the Reticulocyte Binding Protein (PvRBP) family members that have been shown to adhere to reticulocytes [300, 316320]. Specifically, young CD71+ and CD98+ reticulocytes were identified as the host cells for P. vivax [13, 318, 321, 322] (reviewed in [323]). Importantly, the PvRBPs are related to the subsequently discovered P. falciparum Reticulocyte Binding Protein homologues (Rh) [324326] and the Rh5 family member [327] has progressed from pre-clinical trials in A. nancymaae monkeys to clinical vaccine trials in humans [328331]. Several of the above-named P. vivax proteins have been investigated as prospective vaccine candidates in field immunogenicity studies [332, 333], and several of these protein transcripts, along with others, were selectively upregulated according to comparative transcriptomic analyses of P. vivax infections in S. boliviensis and A. nancymaae monkeys, thereby pointing to roles in typical or alternative invasion pathways [334]. Identifying possible effective vaccine targets, for killing of parasites, remains a major goal.

An overarching challenge in the malaria vaccine development pipeline has been to express recombinant proteins or make peptides that can be representative of high-priority target proteins, structurally and antigenically, and on a population scale. While ongoing studies continue to establish a basis for new directions for malaria vaccine development, they are just the beginning. For example, a recent study assessed the global diversity and population structure of high priority P. falciparum vaccine candidate antigens [7]. This analysis included data from over 2600 parasite genomes from 15 malaria endemic countries and evaluated and compared the target haplotypes and 3-dimensional structures. Such analyses will help to optimize vaccine constructs to achieve broader efficacy in populations. Furthermore, protein microarray technologies have been used strategically and effectively to identify immunogenic parasite proteins from human and NHP samples [155, 290, 335], and creative genetic tools and parasite screenings are bearing new fruit [336338]. The post-genome sequencing era with ‘omics’ experiments and ‘big data’ analyses continue to promise incomparably richer and more comprehensive understandings of parasite genes, transcripts, proteins, and metabolites, which together will hopefully reveal new promising high-priority vaccine targets. NHPs are important in this effort.

Whole organism vaccine strategies

By the turn of the twentieth century, the prospects of whole organism malaria vaccines took root with both irradiated and genetically attenuated sporozoite immunization approaches, followed by chemoprophylactic regimens in combination with sporozoite immunogens (reviewed in [282, 308, 339341]). These whole parasite vaccine approaches flourished despite many challenges and much to the credit of the company Sanaria™ successfully establishing necessary feasibility and ethical arguments [278]. As would be expected, whole parasite vaccines give broader immunity than subunit antigen or epitope-based vaccines [342], and P. falciparum sporozoite immunogens have shown homologous protection in malaria naïve individuals [50, 123]. However, the general efficacy of pre-erythrocytic vaccines in individuals in endemic areas has remained low (reviewed in [8]). Thus, the prospects for their widespread use in malaria endemic areas have been in question, raising new challenges to be overcome, largely around the need to better understand NAI and VCI responses in individuals with prior and likely repeat exposure to Plasmodium infections and malaria. By contrast, P. falciparum sporozoite immunizations along with prophylactic drugs have shown homologous and heterologous efficacy and have been suggested for use by travelers to Africa and potentially widespread use in Africa [343345]. Comparable research on P. vivax sporozoite vaccines has yet to be reported. This research has been hampered by the unmet challenge of reliably generating sporozoite immunogens in the absence of blood-stage culture systems that can reliably produce infectious gametocytes for infecting Anopheles mosquitoes. A few studies with sporozoite immunizations concurrent with blood-stage drug cover have been carried out with P. knowlesi [342, 346] and P. cynomolgi in M. mulatta ([347] and Joyner et al. pers. commun.), providing groundwork for future experiments where NHP model systems can delve into mechanisms at work in the face of these strategies.

Major impediments to immune intervention, challenges and opportunities

Now, in the twenty-first century, the lack of detailed understanding of NAI against malaria caused by any species and its limited effectiveness such that chronic parasitaemia ensues rather than sterilizing immunity, and the general lack of reliable malaria vaccine correlates of protection, have been recognized as major impediments to the elimination and eradication of the disease through vaccines or other immunologically based interventions (reviewed in [6, 8, 35, 96, 348]). It is also unclear how chronic exposure and immune activation to any species of Plasmodium impacts the establishment of new immunity against specific antigens represented in vaccine candidates. Specifically, chronic exposure to Plasmodium infection may interfere with inducing efficacious protection, as evidenced recently in malaria endemic field-based vaccine trials (reviewed in [8]). Moreover, economic and cultural factors may contribute to poor nutrition and baseline poor health with anaemia being common in malaria endemic areas, or incomplete intake of costly anti-malarial regimens can result in chronic infections and the potential for ongoing transmission. With progressive understanding of immunological mechanisms, networks, and cascades, novel vaccines and other immunological-based interventions may be envisioned that, ideally, will more successfully target and eliminate the infectious parasite load, thereby supporting host resilience and recovery in malaria endemic areas. Some ideas in this direction have been discussed in relation to the finding that pre-immunization inflammation was associated with malaria vaccine protection [49].

Innate and adaptive immunity

In brief, innate immune signaling pathways are activated by parasite-derived materials such as DNA, RNA, glycosylphosphatidylinositol (GPI), and haemozoin, otherwise known as Pathogen Associated Molecular Patterns (PAMPs) or Danger Associated Molecular Patterns (DAMPs) (reviewed in [349352]). Subsequent adaptive responses target infected RBCs and help suppress clinical disease. Much remains to be understood about the innate responses attributed to various cells including γδ T cells, natural killer cells, NKT cells, dendritic cells, monocytes/macrophages, neutrophils, and platelets [353355], and adaptive B cell and T cell responses, including memory responses, and the relative importance and host dependence upon each [356]. Likewise, the importance of various immunoglobulin isotypes remains to be fully understood [357, 358]. Importantly, IgM has become recognized in human clinical studies as relevant for protection against disease caused by P. falciparum during both acute and chronic infections, being produced initially upon infection as well as through memory B cell (MBC) responses [359363]. Long-lived IgM responses were detected against 15 antigens in recent P. vivax immunogenicity field studies, raising questions on the origin and role of IgM compared to IgG [364, 365]. Also of note, a recent transcriptomic study found that genes and networks associated with ubiquitin-proteasomal proteolysis, which are important for innate and adaptive immunity, were disrupted in children with severe malaria [366].

CHMI experiments have served as an alternative to traditional field studies by attempting to explain the immune response to malaria parasites over time in malaria naïve and non-malaria naïve individuals. For example, one noteworthy observation is that epigenetic imprinting occurs in innate immune cells after exposure to blood-stage parasites in vivo [367]. This is interesting given recent advances generally on the potential importance of innate immune memory, including against Plasmodium [368, 369], and “trained immunity”, defined as heterologous immune responses elicited by live vaccines [370]. It remains unclear how much trained immunity responses are relevant to NAI against malaria. Also of consideration, during CHMI studies initiated with sporozoites, treatment is given when parasitaemia is first detected (i.e., when low), and therefore, the ‘natural’ parasite antigen load and inflammatory responses will be reduced, thus altering the natural course of immunity.

Disease, tolerance and recovery

A hallmark of acute malaria is an array of inflammatory processes that can cause fever and other symptoms and signs of illness, which result in possible mild to severe damage to the body’s tissues and organs. Inflammatory processes can aid a host in its anti-malarial fight and recovery, but they can also overwhelm the host [371]. Numerous known and yet-ill-characterized immunological mechanisms contribute to disease tolerance, for example, involving control of monocyte/macrophage activation [369] and peripheral blood mononuclear cell (PBMC) regulatory [372] responses, and enhanced p53 expression in monocytes that was shown to attenuate inflammation and predict protection from fever [373]. Future research is warranted to better understand these and other such processes, which can suggest possible host-directed and adjunctive therapies that may support host wellness and survival.

Asymptomatic infections, chronicity, and anaemia

The majority of human Plasmodium infections are asymptomatic with chronic long-lasting infections that provide a blood-borne reservoir of parasites for transmission to Anopheles mosquitoes and propagation of the disease [374, 375]. Unless active case detection measures are in place, these asymptomatic infections generally go unnoticed and remain a major barrier for malaria elimination and eradication efforts. This fact raises a key question: Why is the immune system unable to rid the body of all parasites? Future research should include focused efforts to identify the parasite’s immune evasion targets and mechanisms. Additionally, the expansion of so-called atypical memory B cells (AtMBCs) has been reported in people chronically exposed to malaria parasites [376379]. These cells are characterized by the expression of inhibitory markers, and they can be activated by inflammatory signals, such as IFNγ and TLR-9 activation by parasite DNA (reviewed in [380]). Recent data suggests that AtMBCs or IgM producing MBCs may be part of a normal memory immune response in chronically exposed or vaccinated individuals and various questions remain relating to their development and functionality [359, 363, 380382]. In in vitro co-cultures, AtMBCs increase and classical memory B cells contract in the presence of parasites [383].

Future studies using NHPIMs can help advance this critical line of research by determining from which B cell tissue compartments these cells originate, the extent of their persistence, to what cytokines they respond, and to the extent they help or hinder the development of immunological memory responses against the parasites [384]. Delineating their contributions to parasite neutralization as well as pathogenesis is critical in part since AtMBCs are known to produce anti-erythrocyte antibodies that can contribute to the destruction of both infected and uninfected erythrocytes [385387]. These data expand upon prior knowledge relating to autoimmune or specific anti-erythrocyte antibodies in the pathogenesis of malarial anaemia [388393]. Future goals should include tracing these responses between the tissues and the peripheral blood, especially to enhance understanding of immune memory provided by T and B cells, but also since a preponderance of immune activity occurs in the tissues and the peripheral blood provides a limited view of the systemic activities.

Immunosuppression

In 2013, Mueller and colleagues reviewed known information relating to Plasmodium infections causing immunosuppression, including the influence of regulatory T cells, B cell populations, and antigen presenting cells [25]. This topic is of high interest and was expanded upon in 2021 by Calle and colleagues [394]. As immunosuppression is evident during acute infections and afterwards, and can persist during chronic infections, they have stressed the critical points that: (1) malaria impacts the overall health of the immune system, and (2) it is important to conduct research aimed towards reestablishing normal functions. This is generally important for public health but also to support the development of effective immune responses to vaccines.

Route to interventions

Decades of immune response studies converge on similar conclusions with regards to the need to understand much more about the intricate host–parasite relationships and immune responses that allow for sustained parasitism [19, 4648]. Such knowledge has the potential to reveal new immunological targets of intervention, whether against the parasite, or in the form of host-directed or adjunctive therapies, including repurposed drugs [395, 396]. A greater understanding of the NAI responses that lead to partial (or possibly complete) protection is needed, as well as in-depth knowledge about the host and parasite receptor-ligand target molecules required for successful host cell invasion and parasitism, the immune cell types, factors, and pathways that become activated to ward off an infection, the parasite’s immune evasion tactics to overcome such immune activity, and the cascade of pathological consequences that follow.

Normal physiology is affected by malarial infections and tissues can become damaged; in severe cases organ damage may be irreparable. These facts are also evidenced from rare cases of irreversible severe illness caused by P. coatneyi when modelling P. falciparum infection [129] and P. cynomolgi modelling P. vivax infection [397]. Key questions include why individuals living in malaria endemic countries do not develop sterilizing immunity after one or a few infections, and why premunition is not long lasting. An overarching question is why immune memory is insufficient to completely ward off or eliminate new infections. As discussed above, the human host does not completely lack immune cell memory recall responses, but that they are complex and multi-faceted, with many unknowns. Humans also face diverse infecting parasite species and strains, which makes infection scenarios much more complex yet nonetheless akin to what the world has been experiencing with the spread of SARS-CoV-2 variants [398, 399]. The lack of complete immunity is also predictably due to evolutionary safeguards to ensure parasitism, including immune evasion strategies, such as antigenic variation (reviewed in [92, 400]) and escape mechanisms that ensure gametocytogenesis (reviewed in [400402]). Specifically, while infected hosts mount primary and memory recall responses, they tend to favour (or are restricted to achieve) a reduction in parasitaemia and illness over elimination of all parasites, therewith supporting the retention of sexual stage gametocytes and prospects for transmission to mosquitoes.

Systems-based studies of the immune response and cellular dynamics during longitudinal infections will continue to be informative to help address long-standing questions and arrive at new interventions. While clinical immunity can be generated, resulting in asymptomatic infections, chronic sub-patent blood-stage infections remain the norm in endemic areas. This situation naturally serves the parasite well, allowing for transmission of gametocyte progeny to mosquitoes, but remains a health concern for infected individuals as well as the population at large that presents targets for future bites by mosquitoes and propagation of the disease. If natural inoculation of the parasite itself does not lead to sterilizing immunity, what are the chances that a subunit or whole parasite vaccine would? The often-presumed answer is that sterilizing immunity is unlikely, or an extremely elusive goal [96], regardless, and this conclusion has so far been born out in vaccine challenge studies in malaria endemic areas (reviewed in [8]). Aside from vaccines leading to specific protein-targeted immune responses and in general perhaps overcoming parasite strain diversity and antigenic variation, they may need to conquer strain-transcending immunity, although this is still ill-defined [35, 403].

Malaria research advances with systems approaches and nonhuman primate infections

Host–parasite interactions: systems biology, immunology, and vaccinology

A complete understanding of malaria parasite species, their biology, host–parasite interactions, and population dynamics that are essential for their survival, is still lacking. In 1971, at the time of the publication of “The Primate Malarias”, host–parasite interaction(s) was not common terminology [404, 405]. This terminology has taken hold in the post-genome sequencing era, over the past 20 years, where “functional genomics” and the integration of big data sets have become the tools of computational biology research teams [406, 407]. Still today, as was the case 50 years ago, scientists often stand at the cusp of entering deep into the abyss of the unknown, whether aiming to reveal host–parasite receptors, intra-cellular development and growth mechanisms, parasite evasion mechanisms, or to gain a solid understanding of the cascades of molecular and cellular events that result in both disease pathogenesis and immunity. This is palpable when using systems biology, systems immunology, or systems vaccinology approaches (reviewed in [8, 4648, 294, 407411]). Systems biological and immunological approaches are now being used to help interpret and integrate genomics and associated multi-omic data, with the goal of enabling a more holistic if not comprehensive understanding—and predictive capabilities—of the multitude of host–parasite interactions and biological pathways that have evolved to allow successful parasitism: with parasite infection and evasion strategies functioning alongside host immune defense mechanisms. Likewise, systems vaccinology aims to incorporate and visualize high-dimensional data to understand VCI responses and outcomes [412].

Whole blood transcriptomics has been a starting point for numerous functional genomics projects. Transcriptomic models—especially across multiple time points from in vivo infection—gain multi-dimensional complexity when epigenetic interactions and time-dependent biological and immunological phenomena or the impact of the microbiota or co-infections are considered as well (reviewed in [4648, 410]). By integrating transcriptomics and various datatypes, Tran and colleagues identified multiple immune signatures including p53 upregulation in children in Mali that associated with their tolerance to blood-stage infection and fever [373]. Transcriptomics and metabolomics data were integrated by Gardinassi et al. from a P. vivax CHMI trial to define specific immune responses [484], and by Cordy et al. to define metabolic signatures associated with acute and chronic disease caused by P. coatneyi and P. falciparum [131]. Gupta et al. recently used systems approaches, including transcriptomic and metabolic modelling, to show distinctive relationships between longitudinal P. knowlesi infections in M. mulatta and M. fascicularis, which highlight mechanisms of resilience in the latter species [396, 413]. Systems vaccinology approaches were used to study an RTS,S candidate vaccine, demonstrating NK cell responses as correlates of protection associated with RTS,S vaccination [414]. Recently, systems-analysis of multiple malaria vaccine trials has shown that pre-immunization inflammatory responses correlate with protection [49]. Baseline data have also been modelled and integrated to provide predictive information regarding the immune response, as shown for example with an influenza vaccination study that evaluated the frequencies of PBMC subpopulations, the results of which the authors suggest may in part reflect influence by the microbiota or other infectious agents [415]. Pre-immunization correlates of protection have in fact been determined for multiple and diverse vaccine types: against influenza, hepatitis B, yellow fever, and malaria (reviewed in [49]). These examples and many others [46] raise the prospects for discoveries that may improve efficacy of vaccines, by stimulating innate responses predicted to confer protection. However, human trials for malaria vaccines and other diseases have shown that not only internal responses but external environmental factors including endemicity and prior exposure to diseases greatly impact immunity and the ability to predict outcomes of vaccines based on testing in naïve individuals alone (reviewed in [8, 57, 294]). Thus, major challenges remain, at many levels. These include the importance of understanding NAI and VCI responses in children compared to adults, and with different levels of previous exposure to malaria. Such basic research is crucially important, as recognized for many infectious diseases, including SARS-Cov2/Covid-19 [416, 417].

The predictive capabilities, based on mathematical models and the integration of diverse datasets and metadata, support the identification of signature biomarker profiles that may be prognostic or diagnostic, validation experiments that may include perturbations, and in silico screening and testing of candidate drugs and vaccines. Traditional ELISPOT tests, ELISAs, isotype analyses, protein microarrays, and various functional assays (e.g., phagocytosis assays and parasite inhibition of invasion tests) continue to serve a purpose, to validate findings and develop new hypotheses and research directions to delve deeper into the biology of specific cell types and immune networks. Meanwhile, bioinformatic analysis of large and multi-omic datasets, dynamic modelling and spatial imaging technologies are becoming critical for advancing such research, whether on single cells or tissue samples (reviewed in [219, 221]). For an overview of the processes that comprise systems biology, from big data analysis to its visualization to building of complex models and reductionist experimentation to validate the models, various articles are recommended [406, 407].

The integration of multiple-omic and other data types is a key goal for designing quasi-realistic computational models for biological processes. In the simplest case, these are static representations of networks that show which components interact with each other. Dynamic modelling raises the bar further by also addressing questions pertaining to the constant changes in biological systems, such as a host cell or organism, whether these occur within seconds, minutes, a day or longer [418, 419]. Generically, it is hoped that systems approaches will flesh out knowledge gaps in key areas of host–parasite interactions. Pursuing this avenue, new basic, preclinical, and clinical research needs are being defined and prioritized with a burgeoning number of concerted efforts involving multidisciplinary and transdisciplinary teams of researchers. All these modelling efforts require very large quantities of data, and these are beginning to emerge in public databases that offer access to large, system-wide datasets for reuse and iterative analysis (reviewed in [8]). A prominent example for malaria research is PlasmoDB [420, 421], which is associated with the MaHPIC’s longitudinal NHP experiments, described below. While mathematical modelers in the past lacked data, now there is an overload. This has created the need for biologists and mathematical and computational experts to interact, partner, and rely upon each other.

Genomics data are available for key host and parasite species

Genomic and post-genomic technologies have greatly advanced knowledge of malaria parasites of a number of species in various stages of their life cycles. The genome sequences of P. falciparum and three rodent malaria parasite species were tackled first, at the turn of the century [14, 422, 423], followed by others (mentioned below) and recently six Great Ape Laverania malaria parasite species [424] plus a fourth rodent malaria parasite species [425]. This species priority list was largely based on P. falciparum causing the most cases of malaria throughout Sub-Saharan Africa, including severe cases and death, and because P. falciparum and corresponding rodent species are recognized as being most widely amenable to research work in laboratories around the world. However, the in vitro culture environment is very different from the in vivo host environment, with all its complex organ physiology, biology, biochemistry, and immune system, and rodent models—while valuable—do not suffice to reveal the host–parasite interactions that occur in humans or NHPs [426, 427]. Particularly given the availability of the first and additional P. falciparum genomes and robust culture systems, functional genomics studies have become plentiful for P. falciparum for the different parasite development stages throughout the parasite’s life cycle [176, 410, 428].

Plasmodium vivax and P. knowlesi genome sequences were published in 2008 [429, 430], followed by additional P. vivax [431433] and P. knowlesi [434437] genome sequences including the use of advanced technologies and clinical isolates. Plasmodium vivax transcriptomes have since become available representing sporozoites [438, 439], specific liver-stage forms including hypnozoites [212, 213] and blood-stage forms [440448], and proteomes have been developed representing stage-specific blood stages and clinical biomarkers [449454]. Likewise, transcriptomes have been reported for P. knowlesi, importantly showing differences in the parasite’s gene expression between the in vivo NHP environment and in vitro cultures [455] as well as differences in M. mulatta and M. fascicularis host responses [396]. Plasmodium knowlesi ex vivo stage-specific blood-stage proteomes are under analysis, helping to flesh out the biology of this species (Anderson et al. pers. commun.). Plasmodium cynomolgi genome sequences were first published in 2010 [186], followed by an improved genome [168] and transcriptomes representing stage-specific LSFs [208, 214]. Finally, the P. coatneyi genome sequence was published in 2016 [254] and in 2016 and 2017 genome sequences were reported for the remaining two human malaria parasites, P. ovale and P. malariae [15, 456]. Human and NHP (M. mulatta, M. fascicularis and Aotus and Saimiri species) genome sequences have also been published [457463]. So, the genomic puzzle pieces are available to study the interactions of several key parasite species in the context of their human or NHP hosts and, in some instances, with host cells in ex vivo or in vitro experiments. This is in addition to the completion of the genomes of Anopheles vector species and ongoing advancements for studying Plasmodium genomics data [464].

Still and all, the biological functions of most Plasmodium proteins remain undefined, but expedited progress with biological discoveries are likely given the immense amounts of genomic and functional -omic data that continues to be developed and analysed. Technological tools and capabilities have advanced considerably to obtain genomic sequences with improved quality and much more readily from unknown parasite strains, as well as -omic information on host tissues and single cells [465], and when accessible, spatial data providing the interactive physiological tissue and cellular context of both [221]. Today, with advances in big data technologies that enable the generation and analysis of -omic and other large-scale complementary clinical datasets from small blood volumes (100 µl or less, and even from dried blood spots), human blood samples have become amenable to systems biological research that can complement basic in vitro studies and research involving rodents or NHPs.

Deciphering malaria with the support of animal models

The malaria research field and its vision for what’s important and possible have advanced considerably with the recent benefits of CHMI and systems biological and immunological approaches, generating new levels of understanding and numerous new hypotheses. Still today, underlying biological and molecular mechanisms of NAI, VCI and pathogenesis are largely unknown, leaving a hazy picture of the immune response under various scenarios with many gaps and open questions [46, 47]. However, immunological tools and technologies are advancing to make new discovery in these areas possible, and now tantamount, even with chronic infections [375]. Future research is bound to expose the inner workings of many critical biological, immunological, and pathogenic mechanisms and pathways, including immune evasion strategies, and both rodent and NHP animal models can support such endeavors. To the extent possible, it is important to know infection history, lifetime exposure, and how long an individual has been infected. Huge strides cataloguing such essential data have been made by a few research groups performing clinical studies with longitudinal sampling that includes repeat sampling from the same individuals [232, 233, 373, 466468]. Unfortunately, these types of studies are labour intensive and arduous, and not typically possible to complete on large scales.

Numerous investigations for decades have relied upon rodent malaria models to delve into immune response and pathogenesis questions (reviewed in [426, 427]). Apart from expensive model systems such as humanized mice, rodent models are economical and practical, including affordable per diem rates to house and feed the animals and the ready availability of technical support for working with these animals. Moreover, the ability to mimic major clinical manifestations of malaria such as anaemia, cerebral malaria, acute kidney injury, and chronic infection using selected rodent malaria parasite species in conjunction with selected host genetic backgrounds and genetically manipulated parasites have made investigations using rodent model systems efficient and reproducible, alongside studies to decipher the function of the many uncharacterized parasite proteins expressed at different stages of the life cycle (reviewed in [422, 425, 469]). Collaborative cross mice [470, 471] may also serve to identify genetic backgrounds that support the development of chronic, asymptomatic parasitaemia, but significant differences compared to humans and NHPs regarding their immune systems and physiological responses remain, as do other differences and associated limitations. Thus, the trade-off for the utility and reproducibility using rodent models will remain a loss of the complexities and intricacies of malaria pathogenesis that is clear from observations on malaria in humans and NHPs.

NHP infection models (NHPIMs) allow for the in-depth investigation of both broad overarching and specific hypotheses, providing opportunities to enhance what can be achieved using rodents and through clinical studies with humans. Ideally, research using NHPs will continue to complement or validate studies involving rodents, where putative mechanisms can be investigated in greater depth with the benefit of genetic tools and much larger experimental cohorts. Still, rodents can develop exceedingly high parasitaemias, well over 10% in most cases, which undoubtedly must affect immune responses. Such a high level of parasitaemia is rarely seen in human infections or carefully monitored and controlled NHP infections. Moreover, relapse biology and associated immune responses currently cannot be studied using rodent malarial infection models (particularly given the lack of relapses in rodents, except in humanized mice [472, 473]), and as discussed above research on relapses is formidable with humans [231233], making the NHP models for studying relapses preferable if not essential.

In many cases, immune cell subsets between humans and macaques are comparable. Peripheral blood B cell subsets and T-cells can be phenotyped and classified similarly between humans and macaques [139, 474, 475]. Other cell types can also be purified and studied, whether singly or as populations. These strengths of NHP macaque models are further enforced by the ability to access tissues longitudinally or in terminal studies along with peripheral blood. In rodent model systems, tissue samples can also be obtained, but only very small blood volumes are available, limiting determinations on how immune responses in the tissues relate to changes detected in the peripheral blood. Coupling the strengths of macaque models with the many available species and strains of simian malaria parasites makes macaques especially well-suited for immunological investigations, and particularly for systems-based longitudinal studies because of the reasonable amount of informative sample materials that can be obtained over time, covering the entire course of the disease from pre- to post-infection scenarios, and possible re-infection or co-infection situations [476480].

The time is opportune to maximize NHPIM efforts, building upon experience gained from longitudinal NHP experiments by many investigators over many decades (described above), and recently by the MaHPIC, described below. Incidentally, the MaHPIC was established soon after the publication of a review article, titled “Acquired Immunity to Malaria”, where the authors, Doolan, Dobaño and Baird wrote: “One likely starting point for more firmly establishing age-dependent, strain-transcending immunity in malaria and for beginning to sort through the myriad possible mechanisms at work may be a nonhuman primate model” [35]. Similarly, Mueller et al. concluded: “In addition, a more thorough use of the existing non-human primate models as well as experimental infection in human volunteers will be essential for advancing our understanding of the basic processes and specific antigens involved in the establishment of NAI to P. vivax [25].” As noted by Doolan and colleagues, by 1920, the essential elements of NAI had been described. In essence, natural immunity is: “(1) effective in adults after uninterrupted lifelong heavy exposure, (2) lost upon cessation of exposure, (3) species specific, (4) somewhat stage specific, and (5) acquired at a rate which was dependent upon the degree of exposure” [35]. Also, Lefebvre and Harty have emphasized the importance of NHP infections for studying protective immune responses in the liver [96], also indicating that needle aspirate techniques may support such efforts [481]. Silva-Fila and colleagues further recognized the value of both CHMI and NHP research to address immunity and pathogenesis questions, with emphasis on the importance of accessing bone marrow samples [97].

Establishing academic environments for NHP research with complete life cycles

Between 1999 and 2019, the Yerkes NPRC (YNPRC) at Emory University in Atlanta, Georgia (in 2022, renamed as the Emory National Primate Research Center) became recognized as the main NPRC (in the USA) with malaria as a major area of research emphasis, and with a recent history of systemic longitudinal malarial infection studies and an on-site insectary facility. Other academic institutions offer the possibility for NHP studies, or mosquito work, but rarely are these animal and mosquito resources found together in one location, so that NHPs can become experimentally infected via mosquito bites or the inoculation of freshly dissected sporozoites from the salivary glands of mosquitoes. The Yerkes Insectary for Malaria Research (YIFMR) was designed in 2015 and opened in 2017, in response to the Centers for Disease Control and Prevention (CDC, Atlanta, GA) plans to eliminate malaria research projects that involved NHPs. Thus, there was the need to fill this gap, building upon the CDC’s renowned basic infection and evolutionary studies using a variety of primate malaria species and NHP hosts to document host–parasite compatibilities, vaccine trials, and the routine production of gametocytes for infecting female Anopheles mosquitoes to obtain infectious sporozoites (reviewed in [11, 12]). Up until 2017, the CDC was the source of mosquito salivary gland-derived infectious sporozoites for Emory’s malaria research team, including for the MaHPIC, in support of a series of published and unpublished longitudinal infection experiments (described below). The YIFMR was designed for rearing multiple species of Anopheles mosquitoes—for infections with various Plasmodium species of interest (i.e., Anopheles dirus, Anopheles gambiae, Anopheles freeborni, Anopheles stephensi)—and infecting them through direct blood meal feedings on NHPs.

Comparable setups have since been established at two other academic institutions in the United States: the University of Georgia (Athens, GA, currently directed by Chester J. Joyner) with New World and Old World monkey resources and the Oregon NPRC (Portland, Oregon, currently directed by Brandon Wilder) with Old World monkey resources. The expansion of such research is important, especially now while there is potential for in-depth exploration of the life cycle, immunity and pathogenesis using systems biology, immunology, and vaccinology approaches.

The Malaria Host–Pathogen Interaction Center (MaHPIC): building infrastructure for addressing malaria in a systemic manner

Malaria is so complex that systemic analyses have become critical, and these require large transdisciplinary teams that encompass many scientific disciplines. The Malaria Host–Pathogen Interaction Center (MaHPIC) is an example of such a large team effort, which has been focused primarily on NHPIMs and using systems approaches to develop and integrate multi-omic, clinical and parasitological datasets from longitudinal infections. As of the time of this writing, no comparable research program has been noted in the literature, and as discussed above, these studies complement CHMI investigations.

The MaHPIC was established in 2012 at the YNPRC of Emory University [482] and conducted a series of longitudinal NHP infection experiments through September 2017 with USA contract support from the NIH’s National Institute of Allergy and Infectious Diseases (NIAID). The MaHPIC’s founding team comprised investigators from Emory University, the University of Georgia in Athens, the Georgia Institute of Technology, and the CDC, as well as collaborators involved in clinical studies from around the world ([131, 483, 484], and Cordy pers. commun.). The team included malariologists, biologists, biochemists, immunologists, pathologists, clinicians, veterinarians, and mathematical, computational and informatics experts. By September 2017, about 14 terabytes of MaHPIC data had been generated and the majority was released to public websites [482, 485, 486]. Between 2016 and 2019, the team’s work was complemented by the U. S. Defense Advanced Research Projects Agency (DARPA) with funding for a project called “THoR’s HAMMER” (Technology for Host Resistance, Host Acute Models to study Experimental Resistance). Here, the main goal was to reveal characteristics of malarial resilience, by comparing Plasmodium infections in multiple host species, which showed either severe or resilient outcomes (see experiments called E06, E07, E30, E33 and E35 below).

The MaHPIC team put forth the overarching hypothesis that “Nonhuman primate host interactions with Plasmodium pathogens as model systems will provide insights into mechanisms as well as indicators for human malarial disease conditions”. The MaHPIC’s work has been built upon three underlying scientific premises: (1) that the host environment is critical for studying malaria parasite biology, infections, immunity, pathogenesis, and pathology, (2) that studying the dynamics of longitudinal infections is critical, and (3) that the integration and modelling of parasitological, clinical, and multi-omic datasets will reveal novel host–parasite interactions, pathways, and networks.

The MaHPIC’s primary mandate from the NIAID was to run longitudinal infection experiments in macaques, collect peripheral blood and bone marrow samples at specific time points, and generate clinical, parasitological, and multi-omic datasets, which could then be analysed and integrated—initially by the MaHPIC team, and subsequently by others. Representative MaHPIC longitudinal infection experiments and key results are summarized below. Parasites used in the MaHPIC’s experiments included P. coatneyi (Hackeri strain) as a model of P. falciparum, P. cynomolgi (M/B and Ceylon strains) as a model of P. vivax, and P. knowlesi (Malayan strain) to better understand malaria caused by this zoonotic species in its natural (human and Macaca fascicularis) and the traditional experimental (M. mulatta) hosts. Several infection experiments were also carried out with P. vivax in A. nancymaae and S. boliviensis monkeys. Each major longitudinal NHP infection was initiated with sporozoites and allowed to progress for over one month, and in many instances three or more months, when repeat homologous or heterologous sporozoite-initiated infections, relapsing malaria, or recrudescing malaria and chronic disease were being explored. Typically, capillary blood samples were obtained daily as warranted for parasitaemia readings and reticulocyte count analyses, blood chemistry readings using an iSTAT System (Abbot Labs), and targeted and/or untargeted plasma-based metabolomics experiments. Designated time points for venous blood draw samples were determined and selectively utilized as required to generate various large-scale-omic datasets (transcriptomics, metabolomics, lipidomics and/or proteomics), cytokine analyses, serology, adaptive and innate immune profiles, complete blood count haematology measurements, and the enumeration of haemozoin-containing leukocytes. The details of sample collections, methods, and data generated are being compiled elsewhere (DeBarry et al. manuscripts in preparation), in addition to related details already reported with MaHPIC’s public database depositions and in the team’s original research publications (listed at [482]). This body of work represents the ‘tip of the iceberg’ when considering what is possible.

The MaHPIC’s original mandate did not include investigations of the immune response in depth, but the team increasingly built the foundation for doing so given the immune response’s prime relevance to infection and disease. At the start of the MaHPIC, a few basic innate and adaptive immune profiling flow cytometry panels were included in the project’s experimental design, to enable initial comparative analyses of the dynamics of cell types that are activated in the course of infection by different malaria parasite species. The group’s foray into malarial immunity and pathogenesis gained increasing attention, and the design of major infection experiments shifted to address specific outstanding biological, immunobiological, and pathogenesis questions of importance to the field of malaria research. As a result, NIAID also awarded the team supplementary funding to design and implement a systems vaccinology trial experiment, specifically to examine the immune response in M. mulatta to P. cynomolgi sporozoite immunizations in the face of blood-stage drug cover and the outcome from challenging the animals with infectious sporozoites (Joyner et al. pers. commun.).

In total, the MaHPIC successfully optimized an extensive number of flow cytometry panels and gating strategies to track B cells, T cells, monocytes, dendritic cells, neutrophils, and NK cells, as well as cell activation, co-stimulation and trafficking functional markers when relevant. RBC flow cytometry panels and procedures were also developed to monitor erythrocyte progenitor cells, different stages of RBC development from nucleated RBCs to reticulocytes and normocytes, and to distinguish uninfected and infected RBCs [92]. 45 cytokines were identified using Luminex assays and 1300 proteins including among them cytokines were quantitatively measured using SOMAscan—an aptamer-based targeted proteomic technology (SomaLogic, Inc.; Boulder, CO, USA) [487]. Fundamental work was designed to assess the various immune cell populations and specific subsets in the peripheral blood and bone marrow at baseline time points and at specific later time points throughout the course of each longitudinal infection experiment with P. cynomolgi, P. coatneyi or P. knowlesi. Special flow cytometry panels were developed to track and evaluate specific cell types in A. nancymaae and S. boliviensis involved in the immune response to P. vivax. As a result, numerous fluorescently labelled antibodies were identified, tested, and validated in MaHPIC experiments (DeBarry et al. manuscripts in preparation).

Selected longitudinal infection experiments, performed by the MaHPIC

Iterative longitudinal infection experiments were executed by the MaHPIC. A few representative examples are summarized here to demonstrate the types and results of these experiments. The experimental numbers reflect those incorporated in the group’s Laboratory Information Management System.

Experiment 3 (E03): Plasmodium coatneyi infection of M. mulatta

The MaHPIC’s initially designed longitudinal infection experiment (called E03), involved repeat infections of four malaria naïve M. mulatta with 100 P. coatneyi sporozoites [131]. This was designed to build upon knowledge gained in a blood-stage longitudinal experiment focused on anaemia [130]. Then, it was confirmed that the malaria naïve M. mulatta developed severe anaemia, coagulopathy, renal impairment, and a generalized metabolic dysfunction comparable to what is often seen in severe human cases. Major turnover of erythrocytes was demonstrated and attributed to dysfunctional bone marrow. Disease severity turned out to be less once the animals were semi-immune and re-infected [130].

Specifically, E03 [131] showed that rhesus macaques infected with P. coatneyi develop chronic infections after receiving subcurative artemether treatment and that they recapitulate the decrease in haemoglobin levels characteristic of persistent infection in humans [45, 488]. A recent study with kra monkeys infected with P. knowlesi showed evidence of a similar phenotype, but without the administration of subcurative anti-malarial treatment (see E07 below) [268]. Both situations—drug treated or not—are highly relevant for performing comparative systems-level analyses of host–parasite interactions. The E03 study showed specific transcriptomic and metabolic changes associated with acute and chronic infections [131]. Furthermore, distinct metabolic profiles were identified in M. mulatta that were comparable to those observed in human P. falciparum cases. E03 data are still being analysed by the MaHPIC team, for instance, with respect to the integration of transcriptomic and immunological data. Of particular importance has been an analysis of the development of specific antibody responses over time, as chronic infections are established, and the dynamic production and function of different isotypes. A main question relates to how these antibody responses may result in a balance of protective responses with the killing of infected RBCs and adverse responses that result in the elimination of a majority of uninfected bystander RBCs, as determined by Fonseca et al. using a mathematical model [489].

Experiments 04, 23, 24 and 25: Plasmodium cynomolgi infection of M. mulatta

These four iterative experiments adopted the P. cynomolgi-rhesus macaque model of relapsing malaria to delve into specific immunobiological questions and begin the process of using systems biological approaches to understand relapse biology and anaemia as it relates to P. vivax in humans [490]. The course of infection and relapse patterns were studied in experiments involving 11 naive rhesus monkeys, which had been inoculated with 2,000 P. cynomolgi M/B strain sporozoites that had been freshly isolated from Anopheles mosquito salivary glands [139, 140]. The time to blood-stage infection was comparable in all animals, with patency reached within 10–12 days, and all animals relapsed in the course of the study (set for 100 days; initially with a group of five animals designated for E04, and then with six animals designated for E23, followed by repeat parasite inoculations in consecutive experiments called E24 [139] and E25 (Joyner et al. pers. commun); one animal from E04 needed to be euthanized due to severe malaria, as it was not responding to treatment [397].

Based on data from E04, Tang et al. used Weighted Gene Coexpression Network Analysis (WGCNA) to integrate flow cytometry, RNA-sequencing, and systemic cytokine measurements [491]. Despite anaemia and an increase in erythropoietin levels during the acute infection, the bone marrow did not respond appropriately to compensate for blood losses [491]. The acute infections showed ongoing inflammatory responses with Type I and Type II interferon transcriptional signatures, which were associated with intermediate and non-classical monocytes. The complete analyses suggest that, mechanistically, the observed insufficient erythropoiesis may be due to monocyte-associated disruption of GATA1/GATA2 regulation. In summary, monocyte-associated inflammation in the bone marrow influenced the disruption of GATA1/GATA2 transcriptional networks that are required for the differentiation of erythrocytes [491]. In contrast, anaemia was not observed during relapses and the bone marrow samples at that time did not show transcriptomic perturbations reflecting inflammatory responses and dysregulation of erythroid progenitor cells [491]. This is the first systems-level analysis based on NHP longitudinal infections that integrated multiple data types to identify a molecular mechanism that contributed to bone marrow dysfunction, and it provides direction for further study with a focus on longitudinal bone marrow sampling. Shortly thereafter, Brito and colleagues published experiments using bone marrow aspirates from naturally infected P. vivax patients and confirmed a similar mechanism in humans [447]. After decades of realizing that the bone marrow is affected by malaria parasites [492], the mechanisms are now becoming accessible. Additionally, MaHPIC members integrated transcriptomics, metabolomics, and lipidomics data from E04 peripheral blood and bone marrow samples, and synthesized them with immunophenotyping data using a unique and intuitive mutual information-based network analysis approach. This effort distinguished uninfected NHPs, acute infections and relapse infections, in an unsupervised manner, revealing information not possible with a single -omic data type [493].

Systems analyses are showing that macaques are particularly well-suited for investigating changes in the bone marrow and possible relationships to systemic changes impacting not only anaemia, but also the development of long-lived serological immunity. E23 and E24 demonstrated that—as with humans (shown with malariotherapy studies; reviewed above)—clinical immunity can form after a single sporozoite-initiated blood-stage infection and prevent illness; this was the case both during E23 relapses and E24 homologous strain reinfections [139]. For this study, along with clinical and parasitological data, data were integrated from whole blood RNA-sequencing transcriptomics, flow cytometry distinguishing B-cell subpopulations, P. cynomolgi-antigen-specific ELISAs, and opsonic phagocytosis assays to support the functionality of P. cynomolgi antigen-specific antibodies. Together, the various data types and validation experiments demonstrated that immunity after the primary parasitaemia and a bout of clinical illness was associated with a rapid expansion of memory B cells (MBCs) in a recall response and the production of anti-parasite IgG1 that was able to mediate clearance of parasitized RBCs. Antibodies were also detected that specifically reacted with uninfected RBCs, consistent with them being a causal factor in the observed anaemia. Specific B-cell subpopulations and immunoglobulin isotypes were identified among the peripheral blood mononuclear cells. As in humans, the B cell subpopulations identified included: naive (IgD+CD27−), unswitched memory (USM: IgD+CD27+), switched memory (SM: IgD−CD27+), and double-negative B cells (DN: IgD−CD27−). The USM and SM B cells dramatically and rapidly expanded during relapses, including IgG+SM B cells, and IgM+SM B cells though fewer in number; in contrast, only IgG+SM B cells increased during the homologous reinfections, however, IgM recognizing both infected and uninfected RBCs was significantly increased. Overall, the reduction of infected RBCs in the peripheral blood by phagocytosis coincided with specific MBC and antibody responses and the lack of clinically detectable illness in both relapses and homologous reinfections. Having generated insights into the B-cell dynamics involved in the generation of strain-specific immune memory, E23 and E24 informed new possible directions to understand B-cell dynamics and functions in future experiments.

Experiment 25 was subsequently performed to test the hypothesis that protective recall responses projected (and then observed) for E24 would not function with a heterologous challenge. The six rhesus monkeys that had been infected in E23 and then re-infected in E24 with the M/B strain of P. cynomolgi, were inoculated a few months later with 2,000 freshly isolated P. cynomolgi sporozoites of the Ceylon strain; the analysis and integration of the results from this project are in progress, alongside validation experiments (Joyner et al. pers. commun.). For this series of experiments, rectal swabs were also acquired and microbiome and multi-omic integrative analyses are underway to understand host–parasite–microbiome relationships (Cordy et al. pers. commun.).

This series of longitudinal experiments nicely demonstrates the value of this NHP model for exploring immunity to relapsing malaria species, beyond what can typically be done with humans, especially because in the animal model the sporozoite inoculations, treatment of LSFs, and monitoring of blood-stage infections can be rigorously controlled. In a broad sense, these longitudinal macaque infections with P. cynomolgi mirror clinical manifestations and immunity results observed in humans with P. vivax (reviewed in [25]). In recent years, this NHP model has permitted, for the first time, a close look into the dynamic NAI adaptive responses that curtail clinical exacerbation of disease and pathology, as well as the B cell phenotypes and dynamics that define recall responses responsible for reducing parasitaemia [139]. In future longitudinal P. cynomolgi infection experiments, MBC subpopulations and their functionality could be studied in greater depth, including with the sequencing of the B-cell receptors to gain information on their specificity against specific antigens.

Importantly, E23 also capitalized on the opportunity to evaluate the reduction in relapse asexual-stage compared to sexual-stage parasitaemia—assessed by blood-smear counting of gametocytes and transcriptome analysis of gametocyte transcripts [139]. The reduction in parasitaemia during relapses coincided with a reduction in the number of circulating gametocytes, however, the cumulative proportion of gametocytes compared to asexual-stage parasites increased during relapses. These results raise questions as to whether and to what degree relapse infections in humans are clinically silent but infectious by providing a gametocyte reservoir for biting Anopheles mosquitoes [139]. It is remarkable—as deduced from the NHP model experiments—that the parasite and host seem to have co-evolved to survive together with non-sterilizing protective host immunity that enables transmission of gametocytes to mosquitoes and thus the propagation of the parasite species. Transmission experiments are now warranted to clarify and understand the combined NHP host–parasite–vector relationships in light of possible intervention strategies to prevent relapsing parasitaemias that can support transmission. Strategically controlled and monitored sequential mosquito feedings (on infected animals or their infected blood, at multiple time points) can confirm levels of infectivity, and, in concert with systems immunological approaches can lead to an improved understanding of how the primed immune system permits relapsing infections and how the parasites sufficiently evade memory recall responses in order to transmit gametocytes to mosquitoes. Future studies can also be envisioned that delve into gametocyte biology and immunity against the sexual stages, providing possible new insights pertinent to understanding transmission reduction or enhancement activities that can be related to P. vivax and P. falciparum infections (reviewed in [296]).

Experiments 06, 07, E30, E33, E35: Plasmodium knowlesi infection of M. mulatta and M. fascicularis

Peterson et al. delved into the pathogenesis of P. knowlesi infections that had been initiated with Malayan strain sporozoites in M. fascicularis of Mauritius origin and demonstrated that these natural macaque hosts developed clinical illness [268]. However, the animals recovered without anti-malarial intervention. One of the most striking findings was that these animals—unlike M. mulatta [277]—did not display evidence of dyserythropoiesis, and in fact, their bone marrow responded when erythropoietin levels were elevated. This response is in direct contrast to what occurs in rhesus macaques infected with P. coatneyi or P. cynomolgi [130, 131, 139, 140]. One can envision future comparative systems biological studies assessing multiple macaque hosts with the goal of understanding the molecular networks that are affected and the factors that contribute to bone marrow disruption. Indeed, comparisons of peripheral blood transcriptional responses from P. knowlesi infected M. mulatta and M. fascicularis, analysed temporally, show dramatic differences at the onset of the acute infection that seem to support the recovery of M. fascicularis, whereas M. mulatta continues to show inflammatory and anti-inflammatory processes [396]. Perhaps these distinct responses reflect in part the differences observed in the bone marrow. Furthermore, M. fascicularis responded to the parasite as early as 3 days after inoculation with sporozoites, unlike M. mulatta, showing early cytokine signaling along with interferon responses while the parasites were still in the liver. Among other findings, Gupta and colleagues also report pre-infection differences in neutrophils and naïve CD4+T cells that lead to differences in Ca2+ homeostasis, which ultimately balances inflammation and cell proliferation during the log phase of parasitaemia expansion [413]. These P. knowlesi infection experiments also incorporated unique telemetry measurements generated from implanted devices that continuously tracked temperature, blood pressure, heart rate and activity levels (Brady et al. pers. commun.).

Comparisons across experiments based on different NHP host–parasite models

The MaHPIC established that the development of anaemia in rhesus macaques infected with P. coatneyi, P. cynomolgi, or P. knowlesi replicates the hemoglobin kinetics of humans with P. falciparum or P. vivax [130, 131, 139, 140, 277]. During the acute phase of infection, rhesus macaques show evidence of inefficient erythropoiesis, which is further complicated by the removal of uninfected RBCs. Interestingly, the most severe phase of anaemia sets in when erythropoiesis is restored, implying that removal of uninfected RBCs may be the largest contributor to malarial anaemia, as hypothesized in 1999 [44]. Indeed, mathematical modelling of longitudinal infection data by the MaHPIC has supported this hypothesis [112, 489, 494, 495]. These findings suggest that searches are warranted for an adjunctive therapy for severe malarial anaemia that would prevent the removal of uninfected RBCs. Further systems-based studies detailing in vivo mechanisms that cause the removal of uninfected RBCs could be informative, particularly with the advancement of single-cell technologies that allow the fluorescent labelling of erythrocytes with the goal of isolating the phagocytes that take them up in different tissues. In addition, proteomic and other experiments could be applied to identify possible targets on the uninfected RBC surfaces that could be blocked by future antibody-based therapies to prevent their removal by phagocytes.

Data from E03 and E04 were also studied together to determine what common changes may occur in the host due to Plasmodium infection regardless of the infecting species. In one example, Tang et al. interpreted peripheral blood transcriptomics data from E03 and E04 by use of a dynamic model of purine metabolism and showed how gene expression associated with purine metabolism from each of these experiments was reflected in specific downstream alterations in metabolomic signatures [496]. Specifically, a main finding was a pattern of flux rearrangement within the purine pathway system that increased production and excretion of inosine, hypoxanthine, and xanthine. More pronounced changes in the flux patterns were associated with higher parasitaemias and the possible relevance of changes in purine metabolism with regards to inflammation and parasite proliferation was discussed. A similar analysis involving human data showed consistent trends in the flux patterns [496]. Other examples show how transcriptomics and metabolomics data from MaHPIC’s NHP experiments are reflected in data from human studies [131, 483].

Conclusions

The future of malaria immunology and vaccine development—if we can go to the moon…?!

In 2019 many celebrated the 50th anniversary of the first landing of humans on the moon, and another USA-led trip to the moon was projected for 2024. A year later, the SARS-CoV-2/Covid-19 pandemic struck hard and inspired the development, testing, approval, production and distribution of multiple SARS-CoV-2/Covid-19 vaccines in a matter of months [497] and such research continues with intensity to prevent future pandemics [498]. If scientists and supporting professionals can achieve such ‘unrealistic’ feats during a deadly pandemic, why can’t they make and deliver effective malaria vaccines that are direly required for the eradication of a disease that kills hundreds of thousands every year? Or can they? Is the eradication of malaria simply a matter of will, persistence, and enough funding? Parasitic infections are arguably more complex than viral infections, but the hope persists that scientists will discover an Achille’s heel(s) of the parasite, which then can be exploited as a vaccine target(s), and possibly for the future development of host-directed therapies that will save millions of lives worldwide and greatly increase the chances for malaria eradication. How such elusive targets might be found is of course unknown, but, no doubt, ample global research funding and resources must be maintained to achieve such goals [499501] (see Box 2).

Box 2 Millennials, generation Z and beyond!

The future will soon rely on Millennials (born between 1981 and 1996) and Generation Z (born from 1997 onward) [502]. Is it possible today to speed up research so that the next several decades, led by these new generations of scientists, yield exponential growth in understanding the full, multi-faceted extent of malaria? Can we envision an equivalent of ‘going to the moon’ within this time frame? If so, what will that take? Modern methods of systems immunology and vaccinology hold great promise, if they are supported by computational modelling capabilities that allow efficient analysis, integration, and interpretation of large diverse datasets, and offer the hope of revealing insights not possible otherwise. For sure, the field needs patience, time, and resolve, to realize its potential. Funding bodies, as well as passionate wealthy individuals and philanthropically inclined companies, need to recognize and embrace the prospect of a long haul, combined with an innovative, risk-taking mindset of ‘tackling this challenge,’ ‘doing things differently,’ and ‘not being deterred.’ Such research—including studies involving NHPs—has so far advanced in fits and starts. Funding comes, and funding goes. Capable dedicated research teams form, specialized training occurs, projects gain momentum, and then agendas veer off. This constant ebb and flow of research funding makes it difficult to maintain strong active research teams. If the moon-shot goal of malaria research is disease eradication, the future of today’s post-genomics era will require an expansion of research capabilities and talents, with individuals and teams able to analyse and decipher meaning from the vast amounts of genomic, epigenomic, and other data types that can be generated, for instance, in the course of longitudinal infections. To advance malaria research with the use of NHPs, NHP and malariology expertise must be sustained and nurtured, and this expertise must be augmented with a deepened knowledge of the concepts and approaches of immunology, vaccinology, computational biology, and systems biology. The training and cultivation of junior scientists as capable leaders in this research area is essential—now. This sentiment was expressed over 12 years ago by the CDC’s preeminent malaria parasite NHP infection expert and entomologist William Collins [503], a co-author of “The Primate Malarias”. The same is now expressed by this author, as she nears retirement!

Malaria research interests and agendas have become all-encompassing with consideration of today’s malaria eradication goals. The pursuit of effective vaccines continues alongside epidemiological tracking of parasites, species and infections, and focused attention on the need to detect and treat relapse and chronic asymptomatic cases of malaria. Improved diagnostics, drugs and issues relating to insecticide availability and resistance are also at the forefront. New efforts include rapid species-specific detection of infections using non-invasive procedures [504, 505], the identification of biomarkers of immunity and disease [51, 131, 506509], and genetically altered mosquitoes that do not transmit the disease [270]. The need continues for new drugs and drug combination therapies in the face of drug resistance, or contraindications, as with primaquine or tafenoquine use in pregnant and G6PD deficient individuals [10], as well as insecticide-treated nets, other vector inhibitory tools, and procedures for the monitoring and evaluation of epidemiological studies and clinical trials. In addition, the nationwide and regional epidemiological, ecological, socioeconomic, and cultural factors must be considered in the 100 or so countries with malaria.

As a result of the recent SARS-CoV-2/Covid-19 pandemic, communities and their malaria control/elimination/eradication agendas have been disrupted in countless unexpected ways [271] and many questions have been raised on evaluating, managing, and treating syndemics such as the one between malaria and SARS-CoV-2/Covid-19 [272]. With signs that the SARS-CoV-2/Covid-19 pandemic is coming under control, it is time to reassess research directions to keep advancing the goals of malaria eradication. As has been learned with the SARS-CoV-2/Covid-19 pandemic experience, it is wise to stay ahead of predictable epidemics – and pandemics. Scientific and public health experts must keep working together, mastering and advancing key knowledge about malaria and devising ways forward to prevent, treat, and eliminate the disease.

Today with the benefits of cellular imaging, -omic technologies, and computational methods, scientists can visualize biological functions and dynamics, in essence beginning to gain traction on understanding complex in vivo dynamical systems. The advent of systems biological approaches offers hope that it is possible to capitalize on NHP model systems hand-in-hand with CHMI studies. Notwithstanding, despite advances recognized in this review, the use of NHPs for malaria research has arguably been quite limited to date, especially compared to other infectious diseases of widespread importance. Any reluctance is unfortunate because clear evidence is mounting that NHPs can be an effective tool to tease out the details of NAI, VCI and pathogenesis in controlled longitudinal infection studies. As mechanistic depth is required to address many hypotheses regarding the molecular basis of host–parasite interactions, necessitating rigorous sampling of blood, bone marrow, or other tissues, NHPs may be the preferred host of the future.

It is especially timely now for more in-depth studies to better understand NAI to P. vivax primary infections, relapses, and repeat infections using robust Old World macaque-P. cynomolgi infection models [168, 190], along with or followed by confirmatory/validation experiments with P. vivax in the New World monkeys [20]. Likewise, Old World macaque-P. coatneyi and -P. knowlesi infection models are excellent for studying acute and chronic immune responses and pathogenesis, as well as antigenic variation based on the large Schizont Infected Cell Agglutination variant antigens (SICAvar) multigene family that corresponds to the var gene family in P. falciparum [92, 94]. Malaria during pregnancy has been studied using P. coatneyi, P. cynomolgi and P. knowlesi in rhesus monkeys and P. knowlesi in baboons (reviewed in [273]), and future studies on immunity and pathogenesis would also be of value with these models.

Comparative studies with different parasite species, strains, and co-infections, possibly involving animals of different age groups and sexes (infants, juveniles, adults), could also be fruitful toward understanding NAI and VCI in these populations. Such studies may also lead to insights supporting the development of vaccines that provide species- or strain-transcending immunity, comparable to multiple natural exposures in malaria endemic areas to homologous or heterologous parasites that can result in the building of NAI, with a lack of symptomology. While this level of immunity would be a major achievement toward eradication, the problem of asymptomatic infections and chronicity must also be addressed. Otherwise, transmission will remain a likely fact, and the chances for eradication will be much reduced. Interventions focused on NAI goals must include both anti-disease and anti-parasite immunity, as well as the elimination of infectious gametocytes.

A caveat for studying the cellular dynamics within the immune system while relying solely on peripheral blood or bone marrow is that not all immune cell types of interest, or their specific activated phenotypes, are necessarily circulating in the blood or present in the bone marrow. Moreover, a subset of iRBCs can become sequestered or concealed in various tissues and organs [24, 112, 151, 152, 267, 268, 274, 277], so that peripheral blood parasitaemia counts do not necessarily reflect the true parasite load in the host (reviewed in [24, 97, 110, 196]). Tissue samples other than peripheral blood can seldom be obtained from humans. However, various tissues can be accessed from infected NHPs with biopsy procedures or during necropsies in terminal studies. Future experiments are warranted to assess the different cell type niches for Plasmodium and their functional attributes in NHP infections. It will be informative to assess these at several time points during infections, when possible, from sequential biopsies on individual animals, or from a cohort of animals with staggered terminal analyses, such that the cell types within different tissues could be examined. These studies can be performed, for example, during the liver stage and acute and chronic stages of infections, post treatment, and during relapses in the case of P. vivax or P. cynomolgi. With such analyses, a holistic picture of the dynamics of infection, immunity and disease will take shape. Similarly, future iterations of the types of experiments carried out by the MaHPIC will permit the pursuit of hypothesis-driven questions that will gradually become more specific. Thus, within the limits imposed by ethical regulations, more information will be gained from a specific period of an infection or from a specific tissue or cell type and targeted studies will yield insights into the living tissue architecture and the parasite’s environment.

The ever-elusive efficacious malaria vaccine, which can be counted on for complete protection against one or all species, strains and variants of Plasmodium, may not be in the realm of realistic possibilities—today. However, with greater fundamental understanding of the workings of the immune system and host–parasite interactions, strategic, fortuitous—or serendipitous—discoveries will pave the way forward in ways unbeknown today. All things considered, from the hundreds of millions of Plasmodium infections and clinical cases of malaria that still occur annually in close to 100 countries [1], the goals of regional elimination and global eradication [160], and the potential today for incrementally deep exploration into host–pathogen interactions and the identification of new intervention targets and strategies (parasite- or host-directed), it is important to capitalize on the value of multiple animal models for bringing critical knowledge to the fight against malaria. In this vein, Zuck and colleagues boldly concluded that “the study of host–pathogen interactions presents an unmatched opportunity for the field of systems biology, just as the approach of systems biology presents an unmatched opportunity for the eradication of malaria [275].”

Fifty years ago, the authors of “The Primate Malarias” demonstrated much curiosity and dedication, thereby consciously or not paving the way for decades of malaria research involving NHPs to follow their lead. Once again, the malaria research field is at an important juncture, and the question is whether society has the determination to utilize all available resources and the cornucopia of amazing new technologies to advance our understanding of malaria, immunity, and pathogenesis for the benefit of translational research that will ultimately benefit communities affected by this disease worldwide. The catch phrase “use them or lose them” needs to be taken to heart when it comes to NHP model systems. The discussion of advances based on NHP models described here is intended to inspire curiosity and the pursuit of new avenues toward knowledge regarding the multi-faceted system that encompasses malaria. The necessary tools are emerging. They are powerful and unprecedented, but they will only be efficacious if they are used and relentlessly refined and improved by new cohorts of well-trained malariologists. This article’s goal—while honoring leaders from the past—is thus a call to action. The longitudinal infection experiments performed by the MaHPIC are an example of the benefits reaped from decades of prior research with NHPs. It was timely in 2012 for the MaHPIC team to begin developing the systems biological underpinnings for longitudinal NHP infection research, and their work added new dimensions of understanding to key aspects of the disease, such as relapses, chronic infections, anaemia, and immune memory. However, much is yet to be learned, venturing into uncharted territory. If technologies continue to advance as expected, and the tenacity for new research using NHPIMs remains strong, insights into malaria in these models will continue to provide a multi-dimensional holistic view of parasitism at the cellular, tissue and organ levels, and spawn a wider, and at the same time sharper focus onto possible targets for interventions and disease eradication. Terabytes of data from MaHPIC experiments have been deposited in the NIAID-supported Bioinformatics Resource Centers, PlasmoDB [420, 276] and other public databases for the benefit of the global research community. Many opportunities now exist for the analysis and re-analysis of data across MaHPIC’s clinical and NHP experiments [482].

Availability of data and materials

Not applicable.

Abbreviations

atMBC:

Atypical memory B cells

CDC:

Centers for Disease Control and Prevention

CHMI:

Controlled human malaria infection

CSP:

Circumsporozoite protein

iRBC:

Infected red blood cell

MaHPIC:

Malaria Host–Pathogen Interaction Center

MBC:

Memory B cell

NAI:

Naturally acquired immunity

NHP:

Nonhuman primate

NHPIM:

Nonhuman primate infection model

NPRC:

National Primate Research Center

PBMCs:

Peripheral blood mononuclear cells

RBC:

Red blood cell

SICA:

Schizont-infected agglutination antigen

SICAvar :

Schizont-infected agglutination variant antigen gene

VCI:

Vaccine candidate-induced immunity

var :

Variant antigen gene

YNPRC:

Yerkes National Primate Research Center

References

  1. WHO. World malaria report 2021. Geneva: World Health Organization; 2021.

    Google Scholar 

  2. Cox FE. History of the discovery of the malaria parasites and their vectors. Parasit Vectors. 2010;3:5.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Najera JA, Gonzalez-Silva M, Alonso PL. Some lessons for the future from the Global Malaria Eradication Programme (1955–1969). PLoS Med. 2011;8: e1000412.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Vanderberg JP. Reflections on early malaria vaccine studies, the first successful human malaria vaccination, and beyond. Vaccine. 2009;27:2–9.

    Article  CAS  PubMed  Google Scholar 

  5. Crompton PD, Pierce SK, Miller LH. Advances and challenges in malaria vaccine development. J Clin Invest. 2010;120:4168–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Beeson JG, Kurtovic L, Dobano C, Opi DH, Chan JA, Feng G, et al. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med. 2019;11:eaau1458.

    Article  CAS  PubMed  Google Scholar 

  7. Naung MT, Martin E, Munro J, Mehra S, Guy AJ, Laman M, et al. Global diversity and balancing selection of 23 leading Plasmodium falciparum candidate vaccine antigens. PLoS Comput Biol. 2022;18: e1009801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mo AXY, Pesce J, Augustine AD, Bodmer JL, Breen J, Leitner W, et al. Understanding vaccine-elicited protective immunity against pre-erythrocytic stage malaria in endemic regions. Vaccine. 2020;38:7569–77.

    Article  PubMed  Google Scholar 

  9. Kavanaugh MJ, Azzam SE, Rockabrand DM. Malaria rapid diagnostic tests: literary review and recommendation for a quality assurance, quality control algorithm. Diagnostics. 2021;11:768.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hanboonkunupakarn B, White NJ. Advances and roadblocks in the treatment of malaria. Br J Clin Pharmacol. 2020;88:374–82.

    Article  PubMed  Google Scholar 

  11. Coatney GR, Collins WE, Warren M, Contacos PG. The primate malarias. Washington, DC: U.S Department of Health, Education and Welfare; 1971.

    Google Scholar 

  12. Coatney GR, Collins WE, Warren M, Contacos PG. The primate malarias, e-book. Atlanta: Division of Parasitic Diseases; 2003. p. 1–381 (original book published in 1971).

    Google Scholar 

  13. Malleret B, Li A, Zhang R, Tan KS, Suwanarusk R, Claser C, et al. Plasmodium vivax: restricted tropism and rapid remodeling of CD71-positive reticulocytes. Blood. 2015;125:1314–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gardner MJ, Hall N, Fung E, White O, Berriman M, Hyman RW, et al. Genome sequence of the human malaria parasite Plasmodium falciparum. Nature. 2002;419:498–511.

    Article  CAS  PubMed  Google Scholar 

  15. Ansari HR, Templeton TJ, Subudhi AK, Ramaprasad A, Tang J, Lu F, et al. Genome-scale comparison of expanded gene families in Plasmodium ovale wallikeri and Plasmodium ovale curtisi with Plasmodium malariae and with other Plasmodium species. Int J Parasitol. 2016;46:685–96.

    Article  CAS  PubMed  Google Scholar 

  16. Garrido-Cardenas JA, Gonzalez-Ceron L, Manzano-Agugliaro F, Mesa-Valle C. Plasmodium genomics: an approach for learning about and ending human malaria. Parasitol Res. 2019;118:1–27.

    Article  PubMed  Google Scholar 

  17. Nelson CW, Ardern Z, Goldberg TL, Meng C, Kuo CH, Ludwig C, et al. Dynamically evolving novel overlapping gene as a factor in the SARS-CoV-2 pandemic. Elife. 2020;9: e59633.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Galinski MR, Barnwell JW. Non-human primate models for human malaria research. In: Abee CR, Mansfield K, Tardif SD, Morris T, editors. Nonhuman primates in biomedical research. 2nd ed. Amsterdam: Elsevier; 2012. p. 299–323.

    Chapter  Google Scholar 

  19. Galinski MR, Meyer EV, Barnwell JW. Plasmodium vivax: modern strategies to study a persistent parasite’s life cycle. Adv Parasitol. 2013;81:1–26.

    Article  PubMed  Google Scholar 

  20. Pasini EM, Kocken CHM. Parasite-host interaction and pathophysiology studies of the human relapsing malarias Plasmodium vivax and Plasmodium ovale infections in non-human primates. Front Cell Infect Microbiol. 2020;10: 614122.

    Article  CAS  PubMed  Google Scholar 

  21. Baer K, Klotz C, Kappe SH, Schnieder T, Frevert U. Release of hepatic Plasmodium yoelii merozoites into the pulmonary microvasculature. PLoS Pathog. 2007;3: e171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Evans SS, Repasky EA, Fisher DT. Fever and the thermal regulation of immunity: the immune system feels the heat. Nat Rev Immunol. 2015;15:335–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Snounou G, Perignon JL. Malariotherapy—insanity at the service of malariology. Adv Parasitol. 2013;81:223–55.

    Article  PubMed  Google Scholar 

  24. Baird JK. Evidence and implications of mortality associated with acute Plasmodium vivax malaria. Clin Microbiol Rev. 2013;26:36–57.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mueller I, Galinski MR, Tsuboi T, Arevalo-Herrera M, Collins WE, King CL. Natural acquisition of immunity to Plasmodium vivax: epidemiological observations and potential targets. Adv Parasitol. 2013;81:77–131.

    Article  PubMed  Google Scholar 

  26. White NJ. Determinants of relapse periodicity in Plasmodium vivax malaria. Malar J. 2011;10:297.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Clyde DF, Most H, McCarthy VC, Vanderberg JP. Immunization of man against sporozite-induced falciparum malaria. Am J Med Sci. 1973;266:169–77.

    Article  CAS  PubMed  Google Scholar 

  28. Coatney GR, Cooper WC, Ruhe DS. Studies in human malaria; the organization of a program for testing potential antimalarial drugs in prisoner volunteers. Am J Hyg. 1948;47:113–9.

    CAS  PubMed  Google Scholar 

  29. Rieckmann KH, McNamara JV, Kass L, Powell RD. Gametocytocidal and sporontocidal effects of primaquine upon two strains of Plasmodium falciparum. Mil Med. 1969;134:802–19.

    Article  CAS  PubMed  Google Scholar 

  30. Rieckmann KH, McNamara JV, Powell RD. Effects of quinine and of chloroquine in vitro against a strain of chloroquine-resistant Plasmodium falciparum that displays a relative resistance to quinine in vivo. Mil Med. 1969;134:795–801.

    Article  CAS  PubMed  Google Scholar 

  31. Collins WE, Jeffery GM, Roberts JM. A retrospective examination of anemia during infection of humans with Plasmodium vivax. Am J Trop Med Hyg. 2003;68:410–2.

    Article  PubMed  Google Scholar 

  32. Sama W, Dietz K, Smith T. Distribution of survival times of deliberate Plasmodium falciparum infections in tertiary syphilis patients. Trans R Soc Trop Med Hyg. 2006;100:811–6.

    Article  CAS  PubMed  Google Scholar 

  33. Bretscher MT, Maire N, Chitnis N, Felger I, Owusu-Agyei S, Smith T. The distribution of Plasmodium falciparum infection durations. Epidemics. 2011;3:109–18.

    Article  PubMed  Google Scholar 

  34. Childs LM, Buckee CO. Dissecting the determinants of malaria chronicity: why within-host models struggle to reproduce infection dynamics. J R Soc Interface. 2015;12:20141379.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Doolan DL, Dobano C, Baird JK. Acquired immunity to malaria. Clin Microbiol Rev. 2009;22:13–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Moormann AM. How might infant and paediatric immune responses influence malaria vaccine efficacy? Parasite Immunol. 2009;31:547–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Flanagan KL, Fink AL, Plebanski M, Klein SL. Sex and gender differences in the outcomes of vaccination over the life course. Annu Rev Cell Dev Biol. 2017;33:577–99.

    Article  CAS  PubMed  Google Scholar 

  38. Moormann AM, Nixon CE, Forconi CS. Immune effector mechanisms in malaria: an update focusing on human immunity. Parasite Immunol. 2019;41: e12628.

    Article  PubMed  Google Scholar 

  39. Shortt HE, Garnham PC, et al. The pre-erythrocytic stage of human malaria, Plasmodium vivax. Br Med J. 1948;1:547.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Boyd MF. A review of studies on immunity to vivax malaria. J Natl Malar Soc. 1947;6:12–31.

    CAS  PubMed  Google Scholar 

  41. Miller LH, Mason SJ, Clyde DF, McGinniss MH. The resistance factor to Plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy. N Engl J Med. 1976;295:302–4.

    Article  CAS  PubMed  Google Scholar 

  42. Kitchen SF. The infection of reticulocytes by Plasmodium vivax. Am J Trop Med Hyg. 1938. https://doi.org/10.4269/ajtmh.1938.s1-18.347.

    Article  Google Scholar 

  43. Kitchen S. Symptomology: general considerations. Philadelphia: Saunders; 1949.

    Google Scholar 

  44. Jakeman GN, Saul A, Hogarth WL, Collins WE. Anaemia of acute malaria infections in non-immune patients primarily results from destruction of uninfected erythrocytes. Parasitology. 1999;119(Pt 2):127–33.

    Article  PubMed  Google Scholar 

  45. White NJ. Anaemia and malaria. Malar J. 2018;17:371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Loiseau C, Cooper MM, Doolan DL. Deciphering host immunity to malaria using systems immunology. Immunol Rev. 2020;293:115–43.

    Article  CAS  PubMed  Google Scholar 

  47. Tran TM, Crompton PD. Decoding the complexities of human malaria through systems immunology. Immunol Rev. 2020;293:144–62.

    Article  CAS  PubMed  Google Scholar 

  48. Tran TM, Samal B, Kirkness E, Crompton PD. Systems immunology of human malaria. Trends Parasitol. 2012;28:248–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Neal ML, Duffy FJ, Du Y, Aitchison JD, Stuart KD. Preimmunization correlates of protection shared across malaria vaccine trials in adults. NPJ Vaccines. 2022;7:5.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361:468–77.

    Article  CAS  PubMed  Google Scholar 

  51. Obiero JM, Campo JJ, Scholzen A, Randall A, Bijker EM, Roestenberg M, et al. Antibody biomarkers associated with sterile protection induced by controlled human malaria infection under chloroquine prophylaxis. mSphere. 2019;4:2–17.

    Article  Google Scholar 

  52. Vallejo AF, Rubiano K, Amado A, Krystosik AR, Herrera S, Arevalo-Herrera M. Optimization of a membrane feeding assay for Plasmodium vivax infection in Anopheles albimanus. PLoS Negl Trop Dis. 2016;10: e0004807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Griffin P, Pasay C, Elliott S, Sekuloski S, Sikulu M, Hugo L, et al. Safety and reproducibility of a clinical trial system using induced blood stage Plasmodium vivax infection and its potential as a model to evaluate malaria transmission. PLoS Negl Trop Dis. 2016;10: e0005139.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Reuling IJ, van de Schans LA, Coffeng LE, Lanke K, Meerstein-Kessel L, Graumans W, et al. A randomized feasibility trial comparing four antimalarial drug regimens to induce Plasmodium falciparum gametocytemia in the controlled human malaria infection model. Elife. 2018;7: e31549.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Collins KA, Wang CY, Adams M, Mitchell H, Robinson GJ, Rampton M, et al. A Plasmodium vivax experimental human infection model for evaluating efficacy of interventions. J Clin Invest. 2020;130:2920–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Shibeshi W, Bagchus W, Yalkinoglu O, Tappert A, Engidawork E, Oeuvray C. Reproducibility of malaria sporozoite challenge model in humans for evaluating efficacy of vaccines and drugs: a systematic review. BMC Infect Dis. 2021;21:1274.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Yap XZ, McCall MBB, Sauerwein RW. Fast and fierce versus slow and smooth: heterogeneity in immune responses to Plasmodium in the controlled human malaria infection model. Immunol Rev. 2020;293:253–69.

    Article  CAS  PubMed  Google Scholar 

  58. Lavstsen T, Magistrado P, Hermsen CC, Salanti A, Jensen AT, Sauerwein R, et al. Expression of Plasmodium falciparum erythrocyte membrane protein 1 in experimentally infected humans. Malar J. 2005;4:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Abdi AI, Hodgson SH, Muthui MK, Kivisi CA, Kamuyu G, Kimani D, et al. Plasmodium falciparum malaria parasite var gene expression is modified by host antibodies: longitudinal evidence from controlled infections of Kenyan adults with varying natural exposure. BMC Infect Dis. 2017;17:585.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bachmann A, Bruske E, Krumkamp R, Turner L, Wichers JS, Petter M, et al. Controlled human malaria infection with Plasmodium falciparum demonstrates impact of naturally acquired immunity on virulence gene expression. PLoS Pathog. 2019;15: e1007906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pickford AK, Michel-Todo L, Dupuy F, Mayor A, Alonso PL, Lavazec C, et al. Expression patterns of Plasmodium falciparum clonally variant genes at the onset of a blood infection in malaria-naive humans. mBio. 2021;12:e0163621.

    Article  PubMed  Google Scholar 

  62. Payne RO, Griffin PM, McCarthy JS, Draper SJ. Plasmodium vivax controlled human malaria infection—progress and prospects. Trends Parasitol. 2017;33:141–50.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Reyes-Sandoval A. Plasmodium vivax pre-erythrocytic vaccines. Parasitol Int. 2021;84: 102411.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Arevalo-Herrera M, Vasquez-Jimenez JM, Lopez-Perez M, Vallejo AF, Amado-Garavito AB, Cespedes N, et al. Protective efficacy of Plasmodium vivax radiation-attenuated sporozoites in Colombian volunteers: a randomized controlled trial. PLoS Negl Trop Dis. 2016;10: e0005070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bennett JW, Yadava A, Tosh D, Sattabongkot J, Komisar J, Ware LA, et al. Phase 1/2a trial of Plasmodium vivax malaria vaccine candidate VMP001/AS01B in malaria-naive adults: safety, immunogenicity, and efficacy. PLoS Negl Trop Dis. 2016;10: e0004423.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Arevalo-Herrera M, Gaitan X, Larmat-Delgado M, Caicedo MA, Herrera SM, Henao-Giraldo J, et al. Randomized clinical trial to assess the protective efficacy of a Plasmodium vivax CS synthetic vaccine. Nat Commun. 2022;13:1603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Herrera S, Fernandez O, Manzano MR, Murrain B, Vergara J, Blanco P, et al. Successful sporozoite challenge model in human volunteers with Plasmodium vivax strain derived from human donors. Am J Trop Med Hyg. 2009;81:740–6.

    Article  PubMed  Google Scholar 

  68. Solarte Y, Manzano MR, Rocha L, Hurtado H, James MA, Arevalo-Herrera M, et al. Plasmodium vivax sporozoite production in Anopheles albimanus mosquitoes for vaccine clinical trials. Am J Trop Med Hyg. 2011;84:28–34.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Herrera S, Solarte Y, Jordan-Villegas A, Echavarria JF, Rocha L, Palacios R, et al. Consistent safety and infectivity in sporozoite challenge model of Plasmodium vivax in malaria-naive human volunteers. Am J Trop Med Hyg. 2011;84:4–11.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Arevalo-Herrera M, Forero-Pena DA, Rubiano K, Gomez-Hincapie J, Martinez NL, Lopez-Perez M, et al. Plasmodium vivax sporozoite challenge in malaria-naive and semi-immune Colombian volunteers. PLoS ONE. 2014;9: e99754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Payne RO, Milne KH, Elias SC, Edwards NJ, Douglas AD, Brown RE, et al. Demonstration of the blood-stage Plasmodium falciparum controlled human malaria infection model to assess efficacy of the P. falciparum Apical membrane antigen 1 vaccine, FMP2.1/AS01. J Infect Dis. 2016;213:1743–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Dejon-Agobe JC, Ateba-Ngoa U, Lalremruata A, Homoet A, Engelhorn J, Nouatin OP, et al. Controlled human malaria infection of healthy adults with lifelong malaria exposure to assess safety, immunogenicity, and efficacy of the asexual blood stage malaria vaccine candidate GMZ2. Clin Infect Dis. 2019;69:1377–84.

    Article  CAS  PubMed  Google Scholar 

  73. Milne K, Ivens A, Reid AJ, Lotkowska ME, O’Toole A, Sankaranarayanan G, et al. Mapping immune variation and var gene switching in naive hosts infected with Plasmodium falciparum. Elife. 2021. https://doi.org/10.7554/eLife.62800.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Woolley SD, Marquart L, Woodford J, Chalon S, Moehrle JJ, McCarthy JS, et al. Haematological response in experimental human Plasmodium falciparum and Plasmodium vivax malaria. Malar J. 2021;20:470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. McCarthy JS, Griffin PM, Sekuloski S, Bright AT, Rockett R, Looke D, et al. Experimentally induced blood-stage Plasmodium vivax infection in healthy volunteers. J Infect Dis. 2013;208:1688–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Minassian AM, Themistocleous Y, Silk SE, Barrett JR, Kemp A, Quinkert D, et al. Controlled human malaria infection with a clone of Plasmodium vivax with high quality genome assembly. JCI Insight. 2021;6: e152465.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Woodford J, Collins KA, Odedra A, Wang C, Jang IK, Domingo GJ, et al. An experimental human blood-stage model for studying Plasmodium malariae infection. J Infect Dis. 2020;221:948–55.

    PubMed  Google Scholar 

  78. Chavatte JM, Snounou G. Controlled human malaria infection-maker and breaker of dogma. PLoS Med. 2021;18: e1003591.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Njue M, Njuguna P, Kapulu MC, Sanga G, Bejon P, Marsh V, et al. Ethical considerations in controlled human malaria infection studies in low resource settings: experiences and perceptions of study participants in a malaria challenge study in Kenya. Wellcome Open Res. 2018;3:39.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Kraft SA, Duenas DM, Kublin JG, Shipman KJ, Murphy SC, Shah SK. Exploring ethical conscerns about human challenge studies: a qualitative study of controlled human malaria infection study participants’ motivations and attitudes. J Empir Res Hum Res Ethics. 2019;14:49–60.

    Article  PubMed  Google Scholar 

  81. Jao I, Marsh V, Che Chi P, Kapulu M, Hamaluba M, Molyneux S, et al. Deliberately infecting healthy volunteers with malaria parasites: perceptions and experiences of participants and other stakeholders in a Kenyan-based malaria infection study. Bioethics. 2020;34:819–32.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Chi PC, Owino EA, Jao I, Olewe F, Ogutu B, Bejon P, et al. Understanding the benefits and burdens associated with a malaria human infection study in Kenya: experiences of study volunteers and other stakeholders. Trials. 2021;22:494.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Gardner MB, Luciw PA. Macaque models of human infectious disease. ILAR J. 2008;49:220–55.

    Article  CAS  PubMed  Google Scholar 

  84. Di Santo JP, Apetrei C. Animal models for viral diseases: non-human primate and humanized mouse models for viral infections. Curr Opin Virol. 2017;25:v–vii.

    Article  PubMed  Google Scholar 

  85. Niemuth NA, Fallacara D, Triplett CA, Tamrakar SM, Rajbhandari A, Florence C, et al. Natural history of disease in cynomolgus monkeys exposed to Ebola virus Kikwit strain demonstrates the reliability of this non-human primate model for Ebola virus disease. PLoS ONE. 2021;16: e0252874.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Dudley DM, Aliota MT, Mohr EL, Newman CM, Golos TG, Friedrich TC, et al. Using macaques to address critical questions in Zika virus research. Annu Rev Virol. 2019;6:481–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Chang MC, Hild S, Grieder F. Nonhuman primate models for SARS-CoV-2 research: consider alternatives to macaques. Lab Anim. 2021;50:113–4.

    Article  Google Scholar 

  88. Nonhuman Primate Reagent Resource. https://www.nhpreagents.org/.

  89. Antinori S, Bonazzetti C, Giacomelli A, Corbellino M, Galli M, Parravicini C, et al. Non-human primate and human malaria: past, present and future. J Travel Med. 2021. https://doi.org/10.1093/jtm/taab036.

    Article  PubMed  Google Scholar 

  90. Collins WE. The owl monkey as a model for malaria. San Diego: Academic Press; 1994.

    Book  Google Scholar 

  91. Herrera S, Perlaza BL, Bonelo A, Arevalo-Herrera M. Aotus monkeys: their great value for anti-malaria vaccines and drug testing. Int J Parasitol. 2002;32:1625–35.

    Article  PubMed  Google Scholar 

  92. Galinski MR, Lapp SA, Peterson MS, Ay F, Joyner CJ, LeRoch KG, et al. Plasmodium knowlesi: a superb in vivo nonhuman primate model of antigenic variation in malaria. Parasitol. 2018;145:85–100.

    Article  CAS  Google Scholar 

  93. Martinelli A, Culleton R. Non-human primate malaria parasites: out of the forest and into the laboratory. Parasitol. 2018;145:41–54.

    Article  Google Scholar 

  94. Pasini EM, Zeeman AM, Voorberg-VAN DER Wel A, Kocken CHM. Plasmodium knowlesi: a relevant, versatile experimental malaria model. Parasitol. 2018;145:56–70.

    Article  Google Scholar 

  95. Lombardini ED, Gettayacamin M, Turner GD, Brown AE. A review of Plasmodium coatneyi-macaque models of severe malaria. Vet Pathol. 2015;52:998–1011.

    Article  CAS  PubMed  Google Scholar 

  96. Lefebvre MN, Harty JT. You shall not pass: memory CD8 T cells in liver-stage malaria. Trends Parasitol. 2020;36:147–57.

    Article  PubMed  Google Scholar 

  97. Silva-Filho JL, Lacerda MVG, Recker M, Wassmer SC, Marti M, Costa FTM. Plasmodium vivax in hematopoietic niches: hidden and dangerous. Trends in Parasitol. 2020;36:447–58.

    Article  Google Scholar 

  98. Bruce-Chwatt LJ, Garnham PC, Shute PG, Draper CC. Induced double infection with Plasmodium vivax and P. falciparum in a splenectomized chimpanzee. Trans R Soc Trop Med Hyg. 1970;64:2.

    CAS  PubMed  Google Scholar 

  99. Ponnudurai T, Lensen AH, van Gemert GJ, Bolmer M, van Belkum A, van Eerd P, et al. Large-scale production of Plasmodium vivax sporozoites. Parasitol. 1990;101(Pt 3):317–20.

    Article  Google Scholar 

  100. Sullivan JS, Strobert E, Yang C, Morris CL, Galland GG, Richardson BB, et al. Adaptation of a strain of Plasmodium vivax from India to New World monkeys, chimpanzees, and anopheline mosquitoes. J Parasitol. 2001;87:1398–403.

    Article  CAS  PubMed  Google Scholar 

  101. Collins WE, Jeffery GM. Plasmodium ovale: parasite and disease. Clin Microbiol Rev. 2005;18:570–81.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Collins WE, Jeffery GM. Plasmodium malariae: parasite and disease. Clin Microbiol Rev. 2007;20:579–92.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Altevogt BM, Pankevich DE, Pope AM, Kahn JP. Research agenda. Guiding limited use of chimpanzees in research. Science. 2012;335:41–42.

  104. Barnwell JW, Howard RJ, Coon HG, Miller LH. Splenic requirement for antigenic variation and expression of the variant antigen on the erythrocyte membrane in cloned Plasmodium knowlesi malaria. Infect Immun. 1983;40:985–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Barnwell JW, Howard RJ, Miller LH. Influence of the spleen on the expression of surface antigens on parasitized erythrocytes. Ciba Found Symp. 1983;94:117–36.

    CAS  PubMed  Google Scholar 

  106. Lapp SA, Korir-Morrison C, Jiang J, Bai Y, Corredor V, Galinski MR. Spleen-dependent regulation of antigenic variation in malaria parasites: Plasmodium knowlesi SICAvar expression profiles in splenic and asplenic hosts. PLoS ONE. 2013;8: e78014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Hommel M, David PH, Oligino LD. Surface alterations of erythrocytes in Plasmodium falciparum malaria. Antigenic variation, antigenic diversity, and the role of the spleen. J Exp Med. 1983;157:1137–48.

    Article  CAS  PubMed  Google Scholar 

  108. Bachmann A, Esser C, Petter M, Predehl S, von Kalckreuth V, Schmiedel S, et al. Absence of erythrocyte sequestration and lack of multicopy gene family expression in Plasmodium falciparum from a splenectomized malaria patient. PLoS ONE. 2009;4: e7459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Fernandez-Becerra C, Bernabeu M, Castellanos A, Correa BR, Obadia T, Ramirez M, et al. Plasmodium vivax spleen-dependent genes encode antigens associated with cytoadhesion and clinical protection. Proc Natl Acad Sci USA. 2020;117:13056–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Fernandez-Becerra C, Aparici-Herraiz I, Del Portillo HA. Cryptic erythrocytic infections in Plasmodium vivax, another challenge to its elimination. Parasitol Int. 2022;87: 102527.

    Article  PubMed  Google Scholar 

  111. Schmidt LH, Fradkin R, Sesler C, Squires W, Zeyen P. Attenuation of the virulence of the M strain of Plasmodium cynomolgi during prolonged multiplication in splenectomized rhesus monkeys. Am J Trop Med Hyg. 1987;37:460–90.

    Article  CAS  PubMed  Google Scholar 

  112. Fonseca LL, Joyner CJ, MaHPIC-Consortium, Galinski MR, Voit EO. A model of Plasmodium vivax concealment based on Plasmodium cynomolgi infections in Macaca mulatta. Malar J. 2017;16:1–16.

    Article  Google Scholar 

  113. Singh B, Kim Sung L, Matusop A, Radhakrishnan A, Shamsul SS, Cox-Singh J, et al. A large focus of naturally acquired Plasmodium knowlesi infections in human beings. Lancet. 2004;363:1017–24.

    Article  PubMed  Google Scholar 

  114. Cooper DJ, Rajahram GS, William T, Jelip J, Mohammad R, Benedict J, et al. Plasmodium knowlesi malaria in Sabah, Malaysia, 2015–2017: ongoing increase in incidence despite near-elimination of the human-only Plasmodium species. Clin Infect Dis. 2020;70:361–7.

    Article  PubMed  Google Scholar 

  115. Ta TH, Hisam S, Lanza M, Jiram AI, Ismail N, Rubio JM. First case of a naturally acquired human infection with Plasmodium cynomolgi. Malar J. 2014;13:68.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Imwong M, Madmanee W, Suwannasin K, Kunasol C, Peto TJ, Tripura R, et al. Asymptomatic natural human infections with the simian malaria parasites Plasmodium cynomolgi and Plasmodium knowlesi. J Infect Dis. 2019;219:695–702.

    Article  CAS  PubMed  Google Scholar 

  117. Raja TN, Hu TH, Kadir KA, Mohamad DSA, Rosli N, Wong LL, et al. Naturally acquired human Plasmodium cynomolgi and P. knowlesi infections, Malaysian Borneo. Emerg Infect Dis. 2020;26:1801–9.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Yap NJ, Hossain H, Nada-Raja T, Ngui R, Muslim A, Hoh BP, et al. Natural human infections with Plasmodium cynomolgi, P. inui, and 4 other simian malaria parasites, Malaysia. Emerg Infect Dis. 2021;27:2187–91.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Moyes CL, Shearer FM, Huang Z, Wiebe A, Gibson HS, Nijman V, et al. Predicting the geographical distributions of the macaque hosts and mosquito vectors of Plasmodium knowlesi malaria in forested and non-forested areas. Parasit Vectors. 2016;9:242.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Fungfuang W, Udom C, Tongthainan D, Kadir KA, Singh B. Malaria parasites in macaques in Thailand: stump-tailed macaques (Macaca arctoides) are new natural hosts for Plasmodium knowlesi, Plasmodium inui, Plasmodium coatneyi and Plasmodium fieldi. Malar J. 2020;19:350.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Mourier T, de Alvarenga DAM, Kaushik A, de Pina-Costa A, Douvropoulou O, Guan Q, et al. The genome of the zoonotic malaria parasite Plasmodium simium reveals adaptations to host switching. BMC Biol. 2021;19:219.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Lalremruata A, Magris M, Vivas-Martinez S, Koehler M, Esen M, Kempaiah P, et al. Natural infection of Plasmodium brasilianum in humans: man and monkey share quartan malaria parasites in the Venezuelan Amazon. EBioMedicine. 2015;2:1186–92.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science. 2013;341:1359–65.

    Article  CAS  PubMed  Google Scholar 

  124. Shears MJ, Seilie AM, Sim KLB, Hoffman SL, Murphy SC. Quantification of Plasmodium knowlesi versus Plasmodium falciparum in the rhesus liver: implications for malaria vaccine studies in rhesus models. Malar J. 2020;19:313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Davison BB, Cogswell FB, Baskin GB, Falkenstein KP, Henson EW, Krogstad DJ. Placental changes associated with fetal outcome in the Plasmodium coatneyi/rhesus monkey model of malaria in pregnancy. Am J Trop Med Hyg. 2000;63:158–73.

    Article  CAS  PubMed  Google Scholar 

  126. Davison BB, Cogswell FB, Baskin GB, Falkenstein KP, Henson EW, Tarantal AF, et al. Plasmodium coatneyi in the rhesus monkey (Macaca mulatta) as a model of malaria in pregnancy. Am J Trop Med Hyg. 1998;59:189–201.

    Article  CAS  PubMed  Google Scholar 

  127. Davison BB, Kaack MB, Rogers LB, Rasmussen KK, Rasmussen T, Henson EW, et al. Alterations in the profile of blood cell types during malaria in previously unexposed primigravid monkeys. J Infect Dis. 2005;191:1940–52.

    Article  PubMed  Google Scholar 

  128. Davison BB, Kaack MB, Rogers LB, Rasmussen KK, Rasmussen TA, Henson EW, et al. The role of soluble tumor necrosis factor receptor types I and II and tumor necrosis factor-alpha in malaria during pregnancy. J Infect Dis. 2006;194:123–32.

    Article  CAS  PubMed  Google Scholar 

  129. Moreno A, Garcia A, Cabrera-Mora M, Strobert E, Galinski MR. Disseminated intravascular coagulation complicated by peripheral gangrene in a rhesus macaque (Macaca mulatta) experimentally infected with Plasmodium coatneyi. Am J Trop Med Hyg. 2007;76:648–54.

    Article  CAS  PubMed  Google Scholar 

  130. Moreno A, Cabrera-Mora M, Garcia A, Orkin J, Strobert E, Barnwell JW, et al. Plasmodium coatneyi in rhesus macaques replicates the multisystemic dysfunction of severe malaria in humans. Infect Imm. 2013;81:1889–904.

    Article  CAS  Google Scholar 

  131. Cordy RJ, Patrapuvich R, Lili LN, Cabrera-Mora M, Chien J-T, Tharp GK, et al. Distinct amino acid and lipid perturbations characterize acute versus chronic malaria. JCI Insight. 2019;4:1–21.

    Article  Google Scholar 

  132. Aikawa M, Brown A, Smith CD, Tegoshi T, Howard RJ, Hasler TH, et al. A primate model for human cerebral malaria: Plasmodium coatneyi-infected rhesus monkeys. Am J Trop Med Hyg. 1992;46:391–7.

    Article  CAS  PubMed  Google Scholar 

  133. Kawai S, Aikawa M, Kano S, Suzuki M. A primate model for severe human malaria with cerebral involvement: Plasmodium coatneyi-infected Macaca fuscata. Am J Trop Med Hyg. 1993;48:630–6.

    Article  CAS  PubMed  Google Scholar 

  134. Kawai S, Sugiyama M. Imaging analysis of the brain in a primate model of cerebral malaria. Acta Trop. 2010;114:152–6.

    Article  PubMed  Google Scholar 

  135. Schmidt LH. Plasmodium cynomolgi infections in the rhesus monkey. Am J Trop Med Hyg. 1982;31:609–11.

    Article  CAS  PubMed  Google Scholar 

  136. Schmidt LH. Compatibility of relapse patterns of Plasmodium cynomolgi infections in rhesus monkeys with continuous cyclical development and hypnozoite concepts of relapse. Am J Trop Med Hyg. 1986;35:1077–99.

    Article  CAS  PubMed  Google Scholar 

  137. Schmidt LH, Cramer DV, Rossan RN, Harrison J. The characteristics of Plasmodium cynomolgi infections in various old world primates. Am J Trop Med Hyg. 1977;26:356–72.

    Article  CAS  PubMed  Google Scholar 

  138. Deye GA, Gettayacamin M, Hansukjariya P, Im-erbsin R, Sattabongkot J, Rothstein Y, et al. Use of a rhesus Plasmodium cynomolgi model to screen for anti-hypnozoite activity of pharmaceutical substances. Am J Trop Med Hyg. 2012;86:931–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Joyner CJ, Brito CFA, Saney CL, Joice Cordy R, Smith ML, Lapp SA, et al. Humoral immunity prevents clinical malaria during Plasmodium relapses without eliminating gametocytes. PLoS Pathog. 2019;15: e1007974.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Joyner CJ, Moreno A, Meyer EVS, Cabrera-Mora M, Malaria-Consortium, Kissinger JC, et al. Plasmodium cynomolgi infections in rhesus macaques display clinical and parasitological features pertinent to modelling vivax malaria pathology and relapse infections. Malar J. 2016;15:1–18.

    Article  CAS  Google Scholar 

  141. Kamboj K, Dutta GP. Severity of blood-induced Plasmodium cynomolgi B and Plasmodium cynomolgi cynomolgi infection in pregnant rhesus monkeys. Indian J Malariol. 1983;20:1–5.

    Google Scholar 

  142. Saxena N, Murthy PS. Hydrolytic enzyme activity in rhesus monkey placenta during early gestational malaria: histochemical studies. J Vector Borne Dis. 2005;42:135–40.

    PubMed  Google Scholar 

  143. Saxena N, Murthy PS. Oxidoreductases in early gestational monkey placenta during maternal malarial infection: histochemical localisation. J Vector Borne Dis. 2007;44:116–21.

    CAS  PubMed  Google Scholar 

  144. Saxena N, Pandey VC, Saxena PN, Upadhyay S. Hydrolytic enzymes of rhesus placenta during Plasmodium cynomolgi infection: ultrastructural and biochemical studies. Indian J Exp Biol. 1993;31:54–6.

    CAS  PubMed  Google Scholar 

  145. Ozwara H, Langermans JA, Maamun J, Farah IO, Yole DS, Mwenda JM, et al. Experimental infection of the olive baboon (Paplio anubis) with Plasmodium knowlesi: severe disease accompanied by cerebral involvement. Am J Trop Med Hyg. 2003;69:188–94.

    Article  PubMed  Google Scholar 

  146. Onditi FI, Nyamongo OW, Omwandho CO, Maina NW, Maloba F, Farah IO, et al. Parasite accumulation in placenta of non-immune baboons during Plasmodium knowlesi infection. Malar J. 2015;14:118.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Kawai S, Annoura T, Araki T, Shiogama Y, Soma S, Takano JI, et al. Development of an effective alternative model for in vivo hypnozoite-induced relapse infection: a Japanese macaque (Macaca fuscata) model experimentally infected with Plasmodium cynomolgi. Parasitol Int. 2020;76: 102096.

    Article  CAS  PubMed  Google Scholar 

  148. Mallapaty S, Callaway E, Kozlov M, Ledford H, Pickrell J, Van Noorden R. How COVID vaccines shaped 2021 in eight powerful charts. Nature. 2021;600:580–3.

    Article  CAS  PubMed  Google Scholar 

  149. Stewart VA. Plasmodium vivax under the microscope: the Aotus model. Trends Parasitol. 2003;19:589–94.

    Article  PubMed  Google Scholar 

  150. Ward JM, Vallender EJ. The resurgence and genetic implications of New World primates in biomedical research. Trends Genet. 2012;28:586–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Obaldia N 3rd, Meibalan E, Sa JM, Ma S, Clark MA, Mejia P, et al. Bone marrow is a major parasite reservoir in Plasmodium vivax infection. MBio. 2018;9:e00625-18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Peterson MS, Joyner CJ, Cordy RJ, Salinas JL, Machiah D, Lapp SA, et al. Plasmodium vivax parasite load is associated with histopathology in Saimiri boliviensis with findings comparable to P. vivax pathogenesis in humans. Open Forum Infect Dis. 2019;6:1–9.

    Article  CAS  Google Scholar 

  153. Laurens MB, Billingsley P, Richman A, Eappen AG, Adams M, Li T, et al. Successful human infection with P. falciparum using three aseptic Anopheles stephensi mosquitoes: a new model for controlled human malaria infection. PLoS ONE. 2013;8: e68969.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Burns JM Jr, Miura K, Sullivan J, Long CA, Barnwell JW. Immunogenicity of a chimeric Plasmodium falciparum merozoite surface protein vaccine in Aotus monkeys. Malar J. 2016;15:159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Taghavian O, Jain A, Joyner CJ, Ketchum S, Nakajima R, Jasinskas A, et al. Antibody; rofiling by proteome microarray with multiplex isotype detection reveals overlap between human and Aotus nancymaae controlled malaria infections. Proteomics. 2018;18: e1870115.

    Article  CAS  PubMed  Google Scholar 

  156. Gates B, Gates M. Malaria forum keynote address. Seattle: Bill & Melinda Gates Foundation; 2007.

    Google Scholar 

  157. Roberts L, Enserink M. Malaria. Did they really say ... eradication? Science. 2007;318:1544–5.

    Article  CAS  PubMed  Google Scholar 

  158. Enserink M. Eradication goal splits malaria community. Science. 2019;365:847–8.

    Article  CAS  PubMed  Google Scholar 

  159. Alonso PL, Brown G, Arevalo-Herrera M, Binka F, Chitnis C, Collins F, et al. A research agenda to underpin malaria eradication. PLoS Med. 2011;8: e1000406.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Rabinovich RN, Drakeley C, Djimde AA, Hall BF, Hay SI, Hemingway J, et al. malERA: an updated research agenda for malaria elimination and eradication. PLoS Med. 2017;14: e1002456.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Galinski MR, Barnwell JW. Plasmodium vivax: who cares? Malar J. 2008;7:1–18.

    Article  CAS  Google Scholar 

  162. Mueller I, Galinski MR, Baird JK, Carlton JM, Kochar DK, Alonso PL, et al. Key gaps in the knowledge of Plasmodium vivax, a neglected human malaria parasite. Lancet Infect Dis. 2009;9:555–66.

    Article  CAS  PubMed  Google Scholar 

  163. Weiss DJ, Lucas TCD, Nguyen M, Nandi AK, Bisanzio D, Battle KE, et al. Mapping the global prevalence, incidence, and mortality of Plasmodium falciparum, 2000–17: a spatial and temporal modelling study. Lancet. 2019;394:322–31.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Battle KE, Lucas TCD, Nguyen M, Howes RE, Nandi AK, Twohig KA, et al. Mapping the global endemicity and clinical burden of Plasmodium vivax, 2000–17: a spatial and temporal modelling study. Lancet. 2019;394:332–43.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Twohig KA, Pfeffer DA, Baird JK, Price RN, Zimmerman PA, Hay SI, et al. Growing evidence of Plasmodium vivax across malaria-endemic Africa. PLoS Negl Trop Dis. 2019;13: e0007140.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Lover AA, Baird JK, Gosling R, Price RN. Malaria elimination: time to target all species. Am J Trop Med Hyg. 2018;99:17–23.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Hang JW, Tukijan F, Lee EQ, Abdeen SR, Aniweh Y, Malleret B. Zoonotic malaria: non-Laverania Plasmodium biology and invasion mechanisms. Pathogens. 2021. https://doi.org/10.3390/pathogens10070889.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Pasini EM, Bohme U, Rutledge GG, Voorberg-Van der Wel A, Sanders M, Berriman M, et al. An improved Plasmodium cynomolgi genome assembly reveals an unexpected methyltransferase gene expansion. Wellcome Open Res. 2017;2:42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Baird JK. “Lively” invasive Plasmodium vivax causes severe and complicated malaria. Travel Med Infect Dis. 2019;30:7–8.

    Article  PubMed  Google Scholar 

  170. Keeling PJ, Rayner JC. The origins of malaria: there are more things in heaven and earth. Parasitol. 2015;142(Suppl 1):S16-25.

    Article  Google Scholar 

  171. Muller O, Lu G, Jahn A, Razum O. Malaria eradication. Lancet. 2020;395: e68.

    Article  PubMed  Google Scholar 

  172. Mandal S, Sarkar RR, Sinha S. Mathematical models of malaria—a review. Malar J. 2011;10:202.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Eckhoff P. Mathematical models of within-host and transmission dynamics to determine effects of malaria interventions in a variety of transmission settings. Am J Trop Med Hyg. 2013;88:817–27.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Gutierrez JB, Galinski MR, Cantrell S, Voit EO. From within host dynamics to the epidemiology of infectious disease: scientific overview and challenges. Math Biosci. 2015;270:143–55.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Chen I, Clarke SE, Gosling R, Hamainza B, Killeen G, Magill A, et al. “Asymptomatic” malaria: a chronic and debilitating infection that should be treated. PLoS Med. 2016;13: e1001942.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Chawla J, Oberstaller J, Adams JH. Targeting gametocytes of the malaria parasite Plasmodium falciparum in a functional genomics era: next steps. Pathogens. 2021. https://doi.org/10.3390/pathogens10030346.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Duffy PE, Patrick GJ. Malaria vaccines since 2000: progress, priorities, products. NPJ Vaccines. 2020;5:48.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Mendis K, Sina BJ, Marchesini P, Carter R. The neglected burden of Plasmodium vivax malaria. Am J Trop Med Hyg. 2001;64:97–106.

    Article  CAS  PubMed  Google Scholar 

  179. Baird JK. Neglect of Plasmodium vivax malaria. Trends Parasitol. 2007;23:533–9.

    Article  PubMed  Google Scholar 

  180. Price RN, Tjitra E, Guerra CA, Yeung S, White NJ, Anstey NM. Vivax malaria: neglected and not benign. Am J Trop Med Hyg. 2007;77:79–87.

    Article  PubMed  Google Scholar 

  181. Price RN, Commons RJ, Battle KE, Thriemer K, Mendis K. Plasmodium vivax in the era of the shrinking P. falciparum map. Trends Parasitol. 2020;36:560–70.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Battle KE, Baird JK. The global burden of Plasmodium vivax malaria is obscure and insidious. PLoS Med. 2021;18: e1003799.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Commons RJ, Simpson JA, Thriemer K, Hossain MS, Douglas NM, Humphreys GS, et al. Risk of Plasmodium vivax parasitaemia after Plasmodium falciparum infection: a systematic review and meta-analysis. Lancet Infect Dis. 2019;19:91–101.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Commons RJ, Simpson JA, Watson J, White NJ, Price RN. Estimating the proportion of Plasmodium vivax recurrences caused by relapse: a systematic review and meta-analysis. Am J Trop Med Hyg. 2020;103:1094–9.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Waters AP, Higgins DG, McCutchan TF. Evolutionary relatedness of some primate models of Plasmodium. Mol Biol Evol. 1993;10:914–23.

    CAS  PubMed  Google Scholar 

  186. Tachibana S, Sullivan SA, Kawai S, Nakamura S, Kim HR, Goto N, et al. Plasmodium cynomolgi genome sequences provide insight into Plasmodium vivax and the monkey malaria clade. Nat Genet. 2012;44:1051–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Krotoski WA. The hypnozoite and malarial relapse. Prog Clin Parasitol. 1989;1:1–19.

    CAS  PubMed  Google Scholar 

  188. Cogswell FB. The hypnozoite and relapse in primate malaria. Clin Microbiol Rev. 1992;5:26–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Wells TN, Burrows JN, Baird JK. Targeting the hypnozoite reservoir of Plasmodium vivax: the hidden obstacle to malaria elimination. Trends Parasitol. 2010;26:145–51.

    Article  PubMed  Google Scholar 

  190. Joyner C, Barnwell JW, Galinski MR. No more monkeying around: primate malaria model systems are key to understanding Plasmodium vivax liver-stage biology, hypnozoites, and relapses. Front Microbiol. 2015;6:1–8.

    Article  Google Scholar 

  191. Markus MB. Transition from plasmodial hypnozoite to schizont demonstrated. Trends Parasitol. 2020;36:407–8.

    Article  PubMed  Google Scholar 

  192. Merrick CJ. Hypnozoites in Plasmodium: do parasites parallel plants? Trends Parasitol. 2021;37:273–82.

    Article  CAS  PubMed  Google Scholar 

  193. Zanghi G, Vaughan AM. Plasmodium vivax pre-erythrocytic stages and the latent hypnozoite. Parasitol Int. 2021;85: 102447.

    Article  PubMed  Google Scholar 

  194. Schafer C, Zanghi G, Vaughan AM, Kappe SHI. Plasmodium vivax latent liver stage infection and relapse: biological insights and new experimental tools. Annu Rev Microbiol. 2021;75:87–106.

    Article  CAS  PubMed  Google Scholar 

  195. Markus MB. Dormancy in mammalian malaria. Trends Parasitol. 2012;28:39–45.

    Article  PubMed  Google Scholar 

  196. Baird JK. Basic research of Plasmodium vivax biology enabling its management as a clinical and public health problem. Front Cell Infect Microbiol. 2021;11: 696598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Markus MB. Safety and efficacy of tafenoquine for Plasmodium vivax malaria prophylaxis and radical cure: overview and perspectives. Ther Clin Risk Manag. 2021;17:989–99.

    Article  PubMed  PubMed Central  Google Scholar 

  198. White MT, Karl S, Battle KE, Hay SI, Mueller I, Ghani AC. Modelling the contribution of the hypnozoite reservoir to Plasmodium vivax transmission. Elife. 2014. https://doi.org/10.7554/eLife.04692.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Robinson LJ, Wampfler R, Betuela I, Karl S, White MT, Li Wai Suen CS, et al. Strategies for understanding and reducing the Plasmodium vivax and Plasmodium ovale hypnozoite reservoir in Papua New Guinean children: a randomised placebo-controlled trial and mathematical model. PLoS Med. 2015;12: e1001891.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Adekunle AI, Pinkevych M, McGready R, Luxemburger C, White LJ, Nosten F, et al. Modeling the dynamics of Plasmodium vivax infection and hypnozoite reactivation in vivo. PLoS Negl Trop Dis. 2015;9: e0003595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Krotoski WA, Bray RS, Garnham PC, Gwadz RW, Killick-Kendrick R, Draper CC, et al. Observations on early and late post-sporozoite tissue stages in primate malaria. II. The hypnozoite of Plasmodium cynomolgi bastianellii from 3 to 105 days after infection, and detection of 36- to 40-hour pre-erythrocytic forms. Am J Trop Med Hyg. 1982;31:211–25.

    Article  CAS  PubMed  Google Scholar 

  202. Krotoski WA, Collins WE, Bray RS, Garnham PC, Cogswell FB, Gwadz RW, et al. Demonstration of hypnozoites in sporozoite-transmitted Plasmodium vivax infection. Am J Trop Med Hyg. 1982;31:1291–3.

    Article  CAS  PubMed  Google Scholar 

  203. Krotoski WA, Garnham PC, Bray RS, Krotoski DM, Killick-Kendrick R, Draper CC, et al. Observations on early and late post-sporozoite tissue stages in primate malaria. I. Discovery of a new latent form of Plasmodium cynomolgi (the hypnozoite), and failure to detect hepatic forms within the first 24 hours after infection. Am J Trop Med Hyg. 1982;31:24–35.

    Article  CAS  PubMed  Google Scholar 

  204. Roth A, Maher SP, Conway AJ, Ubalee R, Chaumeau V, Andolina C, et al. A comprehensive model for assessment of liver stage therapies targeting Plasmodium vivax and Plasmodium falciparum. Nat Commun. 2018;9:1837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Posfai D, Maher SP, Roesch C, Vantaux A, Sylvester K, Peneau J, et al. Plasmodium vivax liver and blood stages recruit the druggable host membrane channel aquaporin-3. Cell Chem Biol. 2020;27(719–727): e715.

    Google Scholar 

  206. Dembélé L, Franetich J-F, Lorthiois A, Gego A, Snounou G, Mazier D. Long-term co-culture of primary hepatocytes and GFP-HepaRG cells for investigations on Plasmodium hypnozoites. Protoc Exch. 2014. https://doi.org/10.1038/protex.2014.003.

    Article  Google Scholar 

  207. Dembele L, Gego A, Zeeman AM, Franetich JF, Silvie O, Rametti A, et al. Towards an in vitro model of Plasmodium hypnozoites suitable for drug discovery. PLoS ONE. 2011;6: e18162.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Voorberg-van der Wel A, Roma G, Gupta DK, Schuierer S, Nigsch F, Carbone W, et al. A comparative transcriptomic analysis of replicating and dormant liver stages of the relapsing malaria parasite Plasmodium cynomolgi. Elife. 2017. https://doi.org/10.7554/eLife.29605.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Chua ACY, Ong JJY, Malleret B, Suwanarusk R, Kosaisavee V, Zeeman AM, et al. Robust continuous in vitro culture of the Plasmodium cynomolgi erythrocytic stages. Nat Commun. 2019;10:3635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Ong JJY, Russell B, Han JH. Plasmodium cynomolgi Berok growth inhibition assay by thiol-reactive probe based flow cytometric measurement. Bio Protoc. 2021;11: e4147.

    Article  CAS  PubMed  Google Scholar 

  211. Shen FH, Ong JJY, Sun YF, Lei Y, Chu RL, Kassegne K, et al. A Chimeric Plasmodium vivax merozoite surface protein antibody recognizes and blocks erythrocytic P. cynomolgi Berok merozoites in vitro. Infect Immun. 2021. https://doi.org/10.1128/IAI.00645-20.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Gural N, Mancio-Silva L, Miller AB, Galstian A, Butty VL, Levine SS, et al. In vitro culture, drug sensitivity, and transcriptome of Plasmodium vivax hypnozoites. Cell Host Microbe. 2018;23(395–406): e394.

    Google Scholar 

  213. Cubi R, Vembar SS, Biton A, Franetich JF, Bordessoulles M, Sossau D, et al. Laser capture microdissection enables transcriptomic analysis of dividing and quiescent liver stages of Plasmodium relapsing species. Cell Microbiol. 2017. https://doi.org/10.1111/cmi.12735.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Bertschi NL, Voorberg-van der Wel A, Zeeman AM, Schuierer S, Nigsch F, Carbone W, et al. Transcriptomic analysis reveals reduced transcriptional activity in the malaria parasite Plasmodium cynomolgi during progression into dormancy. Elife. 2018. https://doi.org/10.7554/eLife.41081.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Dembele L, Franetich JF, Lorthiois A, Gego A, Zeeman AM, Kocken CH, et al. Persistence and activation of malaria hypnozoites in long-term primary hepatocyte cultures. Nat Med. 2014;20:307–12.

    Article  CAS  PubMed  Google Scholar 

  216. Sylvester K, Maher SP, Posfai D, Tran MK, Crawford MC, Vantaux A, et al. Characterization of the tubovesicular network in Plasmodium vivax liver stage hypnozoites and schizonts. Front Cell Infect Microbiol. 2021;11: 687019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Gupta DK, Dembele L, Voorberg- Wel A, Roma G, Yip A, Chuenchob V, et al. The Plasmodium liver-specific protein 2 (LISP2) is an early marker of liver stage development. Elife. 2019. https://doi.org/10.7554/eLife.43362.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Dembele L, Gupta DK, Dutta B, Chua ACY, Sze SK, Bifani P. Quantitative proteomic analysis of simian primary hepatocytes reveals candidate molecular markers for permissiveness to relapsing malaria Plasmodium cynomolgi. Proteomics. 2019. https://doi.org/10.1002/pmic.201900021.

    Article  PubMed  Google Scholar 

  219. Stuart T, Satija R. Integrative single-cell analysis. Nat Rev Genet. 2019;20:257–72.

    Article  CAS  PubMed  Google Scholar 

  220. Afriat A, Zuzarte-Luís V, Bahar Halpern K, Buchauer L, Marques S, Lahree A, et al. A spatiotemporally resolved single cell atlas of the Plasmodium liver stage. BioRxiv. 2021. https://doi.org/10.1101/2021.12.03.471111.

    Article  Google Scholar 

  221. Eisenstein M. Companies seek slice of spatial imaging market. Nat Biotechnol. 2019;37:490–1.

    Article  CAS  PubMed  Google Scholar 

  222. Ruberto AA, Bourke C, Vantaux A, Maher SP, Jex A, Witkowski B, et al. Single-cell RNA sequencing of Plasmodium vivax sporozoites reveals stage- and species-specific transcriptomic signatures. BioRxiv. 2021. https://doi.org/10.1101/2021.11.24.469176.

    Article  Google Scholar 

  223. Ruberto AA, Maher SP, Vantaux A, Joyner CJ, Bourke C, Balan B, et al. Single-cell RNA profiling of Plasmodium vivax liver stages reveals parasite- and host specific transcriptomic signatures and drug targets. BioRxiv. 2022. https://doi.org/10.1101/2022.02.01.478648.

    Article  Google Scholar 

  224. Subramani PA, Vartak-Sharma N, Sreekumar S, Mathur P, Nayer B, Dakhore S, et al. Plasmodium vivax liver stage assay platforms using Indian clinical isolates. Malar J. 2020;19:214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. White M, Amino R, Mueller I. Theoretical implications of a pre-erythrocytic Plasmodium vivax r for preventing elapses. Trends Parasitol. 2017;33:260–3.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Groger M, Fischer HS, Veletzky L, Lalremruata A, Ramharter M. A systematic review of the clinical presentation, treatment and relapse characteristics of human Plasmodium ovale malaria. Malar J. 2017;16:112.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Veletzky L, Groger M, Lagler H, Walochnik J, Auer H, Fuehrer HP, et al. Molecular evidence for relapse of an imported Plasmodium ovale wallikeri infection. Malar J. 2018;17:78.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Wangdahl A, Sonden K, Wyss K, Stenstrom C, Bjorklund D, Zhang J, et al. Relapse of Plasmodium vivax and Plasmodium ovale malaria with and without primaquine treatment in a non-endemic area. Clin Infect Dis. 2021;74(7):1199–207.

    Article  CAS  PubMed Central  Google Scholar 

  229. Betuela I, Rosanas-Urgell A, Kiniboro B, Stanisic DI, Samol L, de Lazzari E, et al. Relapses contribute significantly to the risk of Plasmodium vivax infection and disease in Papua New Guinean children 1–5 years of age. J Infect Dis. 2012;206:1771–80.

    Article  PubMed  Google Scholar 

  230. Chuquiyauri R, Molina DM, Moss EL, Wang R, Gardner MJ, Brouwer KC, et al. Genome-scale protein microarray comparison of human antibody responses in Plasmodium vivax relapse and reinfection. Am J Trop Med Hyg. 2015;93:801–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Cowell AN, Valdivia HO, Bishop DK, Winzeler EA. Exploration of Plasmodium vivax transmission dynamics and recurrent infections in the Peruvian Amazon using whole genome sequencing. Genome Med. 2018;10:52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Popovici J, Friedrich LR, Kim S, Bin S, Run V, Lek D, et al. Genomic analyses reveal the common occurrence and complexity of Plasmodium vivax relapses in Cambodia. mBio. 2018. https://doi.org/10.1128/mBio.01888-17.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Popovici J, Pierce-Friedrich L, Kim S, Bin S, Run V, Lek D, et al. Recrudescence, reinfection, or relapse? A more rigorous framework to assess chloroquine efficacy for Plasmodium vivax malaria. J Infect Dis. 2019;219:315–22.

    Article  CAS  PubMed  Google Scholar 

  234. Jones DP. Sequencing the exposome: a call to action. Toxicol Rep. 2016;3:29–45.

    Article  CAS  PubMed  Google Scholar 

  235. Bousema T, Drakeley C. Epidemiology and infectivity of Plasmodium falciparum and Plasmodium vivax gametocytes in relation to malaria control and elimination. Clin Microbiol Rev. 2011;24:377–410.

    Article  PubMed  PubMed Central  Google Scholar 

  236. Adapa SR, Taylor RA, Wang C, Thomson-Luque R, Johnson LR, Jiang RHY. Plasmodium vivax readiness to transmit: implication for malaria eradication. BMC Syst Biol. 2019;13:5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Nguitragool W, Mueller I, Kumpitak C, Saeseu T, Bantuchai S, Yorsaeng R, et al. Very high carriage of gametocytes in asymptomatic low-density Plasmodium falciparum and P. vivax infections in western Thailand. Parasit Vectors. 2017;10:512.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Chaumeau V, Kajeechiwa L, Fustec B, Landier J, Naw Nyo S, Nay Hsel S, et al. Contribution of asymptomatic Plasmodium infections to the transmission of malaria in Kayin State, Myanmar. J Infect Dis. 2019;219:1499–509.

    Article  PubMed  Google Scholar 

  239. Pinilla YT, Boussougou-Sambe ST, Grassle S, Ngossanga B, Doumba-Ndalembouly AG, Weierich A, et al. Experimental transmission of Plasmodium malariae to Anopheles gambiae. J Infect Dis. 2021;223:522–6.

    Article  PubMed  Google Scholar 

  240. Yman V, Wandell G, Mutemi DD, Miglar A, Asghar M, Hammar U, et al. Persistent transmission of Plasmodium malariae and Plasmodium ovale species in an area of declining Plasmodium falciparum transmission in eastern Tanzania. PLoS Negl Trop Dis. 2019;13: e0007414.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Barry A, Bradley J, Stone W, Guelbeogo MW, Lanke K, Ouedraogo A, et al. Higher gametocyte production and mosquito infectivity in chronic compared to incident Plasmodium falciparum infections. Nat Commun. 2021;12:2443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Ahmad A, Prom A, Bradley J, Ndiath M, Etoketim B, Bah M, et al. Gametocyte carriage after seasonal malaria chemoprevention in Plasmodium falciparum infected asymptomatic children. Malar J. 2021;20:169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Collins KA, Wang CY, Adams M, Mitchell H, Rampton M, Elliott S, et al. A controlled human malaria infection model enabling evaluation of transmission-blocking interventions. J Clin Invest. 2018;128:1551–62.

    Article  PubMed  PubMed Central  Google Scholar 

  244. Collins KA, Ruckle T, Elliott S, Marquart L, Ballard E, Chalon S, et al. DSM265 at 400 milligrams clears asexual stage parasites but not mature gametocytes from the blood of healthy subjects experimentally infected with Plasmodium falciparum. Antimicrob Agents Chemother. 2019. https://doi.org/10.1128/AAC.01837-18.

    Article  PubMed  PubMed Central  Google Scholar 

  245. Raja AI, Brickley EB, Taaffe J, Ton T, Zhao Z, Bock KW, et al. A primate model of severe malarial anaemia: a comparative pathogenesis study. Sci Rep. 2019;9:18965.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Kim YC, Dema B, Rodriguez-Garcia R, Lopez-Camacho C, Leoratti FMS, Lall A, et al. Evaluation of chimpanzee adenovirus and MVA expressing TRAP and CSP from Plasmodium cynomolgi to prevent malaria relapse in nonhuman primates. Vaccines. 2020. https://doi.org/10.3390/vaccines8030363.

    Article  PubMed  PubMed Central  Google Scholar 

  247. Smith JD, Rowe JA, Higgins MK, Lavstsen T. Malaria’s deadly grip: cytoadhesion of Plasmodium falciparum-infected erythrocytes. Cell Microbiol. 2013;15:1976–83.

    Article  CAS  PubMed  Google Scholar 

  248. Lee WC, Russell B, Renia L. Sticking for a cause: the falciparum malaria parasites cytoadherence paradigm. Front Immunol. 2019;10:1444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Garnham PCC. Plasmodium knowlesi and subspecies, Plasmodium coatneyi and Plasmodium fragile. In malaria parasites and other haemosporidia. Oxford: Blackwell Scientific Publications; 1966. p. 323–56.

    Google Scholar 

  250. Fremount HN, Miller LH. Deep vascular schizogony in Plasmodium fragile: organ distribution and ultrastructure of erythrocytes adherent to vascular endothelium. Am J Trop Med Hyg. 1975;24:1–8.

    Article  CAS  PubMed  Google Scholar 

  251. Handunnetti SM, Mendis KN, David PH. Antigenic variation of cloned Plasmodium fragile in its natural host Macaca sinica. Sequential appearance of successive variant antigenic types. J Exp Med. 1987;165:1269–83.

    Article  CAS  PubMed  Google Scholar 

  252. Nakano Y, Fujioka H, Luc KD, Rabbege JR, Todd GD, et al. A correlation of the sequestration rate of Plasmodium coatneyi-infected erythrocytes in cerebral and subcutaneous tissues of a rhesus monkey. Am J Trop Med Hyg. 1996;55:311–4.

    Article  CAS  PubMed  Google Scholar 

  253. Collins WE, Warren M, Sullivan JS, Galland GG, Strobert E, Nace D, et al. Studies on sporozoite-induced and chronic infections with Plasmodium fragile in Macaca mulatta and New World monkeys. J Parasitol. 2006;92:1019–26.

    Article  PubMed  Google Scholar 

  254. Chien JT, Pakala SB, Geraldo JA, Lapp SA, Humphrey JC, Barnwell JW, et al. High-quality genome assembly and annotation for Plasmodium coatneyi, generated using Single-molecule real-time PacBio technology. Genome Announc. 2016. https://doi.org/10.1128/genomeA.00883-16.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Leech JH, Barnwell JW, Miller LH, Howard RJ. Identification of a strain-specific malarial antigen exposed on the surface of Plasmodium falciparum-infected erythrocytes. J Exp Med. 1984;159:1567–75.

    Article  CAS  PubMed  Google Scholar 

  256. Korir CC, Galinski MR. Proteomic studies of Plasmodium knowlesi SICA variant antigens demonstrate their relationship with P. falciparum EMP1. Infect Genet Evol. 2006;6:75–9.

    Article  CAS  PubMed  Google Scholar 

  257. Howard RJ. Antigenic variation of bloodstage malaria parasites. Philos Trans R Soc Lond B Biol Sci. 1984;307:141–58.

    Article  CAS  PubMed  Google Scholar 

  258. Farfour E, Charlotte F, Settegrana C, Miyara M, Buffet P. The extravascular compartment of the bone marrow: a niche for Plasmodium falciparum gametocyte maturation? Malar J. 2012;11:285.

    Article  PubMed  PubMed Central  Google Scholar 

  259. Aguilar R, Magallon-Tejada A, Achtman AH, Moraleda C, Joice R, Cistero P, et al. Molecular evidence for the localization of Plasmodium falciparum immature gametocytes in bone marrow. Blood. 2014;123:959–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Joice R, Nilsson SK, Montgomery J, Dankwa S, Egan E, Morahan B, et al. Plasmodium falciparum transmission stages accumulate in the human bone marrow. Sci Transl Med. 2014;6:244re245.

  261. Pelle KG, Oh K, Buchholz K, Narasimhan V, Joice R, Milner DA, et al. Transcriptional profiling defines dynamics of parasite tissue sequestration during malaria infection. Genome Med. 2015;7:19.

    Article  PubMed  PubMed Central  Google Scholar 

  262. Baro B, Deroost K, Raiol T, Brito M, Almeida AC, de Menezes-Neto A, et al. Plasmodium vivax gametocytes in the bone marrow of an acute malaria patient and changes in the erythroid miRNA profile. PLoS Negl Trop Dis. 2017;11: e0005365.

    Article  PubMed  PubMed Central  Google Scholar 

  263. Machado Siqueira A, Lopes Magalhaes BM, Cardoso Melo G, Ferrer M, Castillo P, Martin-Jaular L, et al. Spleen rupture in a case of untreated Plasmodium vivax infection. PLoS Negl Trop Dis. 2012;6: e1934.

    Article  PubMed  PubMed Central  Google Scholar 

  264. Kho S, Qotrunnada L, Leonardo L, Andries B, Wardani PAI, Fricot A, et al. Evaluation of splenic accumulation and colocalization of immature reticulocytes and Plasmodium vivax in asymptomatic malaria: a prospective human splenectomy study. PLoS Med. 2021;18: e1003632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Kho S, Qotrunnada L, Leonardo L, Andries B, Wardani PAI, Fricot A, et al. Hidden biomass of intact malaria parasites in the human spleen. N Engl J Med. 2021;384:2067–9.

    Article  PubMed  Google Scholar 

  266. Fremount HN, Rossan RN. Anatomical distribution of developing trophozoites and schizonts of Plasmodium vivax in Aotus lemurinus lemurinus and Saimiri sciureus. J Parasitol. 1990;76:428–30.

    Article  CAS  PubMed  Google Scholar 

  267. Miller LH, Fremount HN, Luse SA. Deep vascular schizogony of Plasmodium knowlesi in Macaca mulatta. Distribution in organs and ultrastructure of parasitized red cells. Am J Trop Med Hyg. 1971;20:816–24.

    Article  CAS  PubMed  Google Scholar 

  268. Peterson MS, Joyner CJ, Brady JA, Wood JS, Cabrera-Mora M, Saney CL, et al. Clinical recovery of Macaca fascicularis infected with Plasmodium knowlesi. Malar J. 2021;20:486.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Ballou WR. The development of the RTS, S malaria vaccine candidate: challenges and lessons. Parasite Immunol. 2009;31:492–500.

    Article  CAS  PubMed  Google Scholar 

  270. Nakkazi E. Fighting malaria with genetically modified mosquitoes. BMJ. 2020;370: m2172.

    Article  PubMed  Google Scholar 

  271. Rogerson SJ, Beeson JG, Laman M, Poespoprodjo JR, William T, Simpson JA, et al. Identifying and combating the impacts of COVID-19 on malaria. BMC Med. 2020;18:239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Hussein MIH, Albashir AAD, Elawad O, Homeida A. Malaria and COVID-19: unmasking their ties. Malar J. 2020;19:457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Doritchamou J, Teo A, Fried M, Duffy PE. Malaria in pregnancy: the relevance of animal models for vaccine development. Lab Anim. 2017;46:388–98.

    Article  Google Scholar 

  274. Toda H, Diaz-Varela M, Segui-Barber J, Roobsoong W, Baro B, Garcia-Silva S, et al. Plasma-derived extracellular vesicles from Plasmodium vivax patients signal spleen fibroblasts via NF-kB facilitating parasite cytoadherence. Nat Commun. 2020;11:2761.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Zuck M, Austin LS, Danziger SA, Aitchison JD, Kaushansky A. The promise of systems biology approaches for revealing host pathogen interactions in malaria. Front Microbiol. 2017;8:2183.

    Article  PubMed  PubMed Central  Google Scholar 

  276. Amos B, Aurrecoechea C, Barba M, Barreto A, Basenko EY, Bazant W, et al. VEuPathDB: the eukaryotic pathogen, vector and host bioinformatics resource center. Nucleic Acids Res. 2022;50:D898–911.

    Article  CAS  PubMed  Google Scholar 

  277. Peterson MS, Joyner JC, Lapp SA, Brady JA, Wood JS, Cabrera-Mora M, et al. Plasmodium knowlesi cytoadhesion involves SICA variant proteins. Front Cell Infect Microbiol. 2022;12: 888496.

    Article  Google Scholar 

  278. Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine. 2015;33(Suppl 4):D13-23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Laurens MB. The promise of a malaria vaccine—are we closer? Annu Rev Microbiol. 2018;72:273–92.

    Article  CAS  PubMed  Google Scholar 

  280. Chitnis CE, Schellenberg D, Vekemans J, Asturias EJ, Bejon P, Collins KA, et al. Building momentum for malaria vaccine research and development: key considerations. Malar J. 2020;19:421.

    Article  PubMed  PubMed Central  Google Scholar 

  281. Arora N, Anbalagan LC, Pannu AK. Towards eradication of malaria: is the WHO’s RTS, S/AS01 vaccination effective enough? Risk Manag Healthc Policy. 2021;14:1033–9.

    Article  PubMed  PubMed Central  Google Scholar 

  282. Ferluga J, Singh I, Rout S, Al-Qahtani A, Yasmin H, Kishore U. Immune responses in malaria and vaccine strategies. In: Kishore U, editor. Microbial pathogenesis: infection and immunity. Cham: Springer; 2021. p. 273–91.

    Chapter  Google Scholar 

  283. Arevalo-Herrera M, Chitnis C, Herrera S. Current status of Plasmodium vivax vaccine. Hum Vaccin. 2010;6:124–32.

    Article  CAS  PubMed  Google Scholar 

  284. Reyes-Sandoval A, Bachmann MF. Plasmodium vivax malaria vaccines: why are we where we are? Hum Vaccin Immunother. 2013;9:2558–65.

    Article  PubMed  PubMed Central  Google Scholar 

  285. Hogan AB, Winskill P, Ghani AC. Estimated impact of RTS, S/AS01 malaria vaccine allocation strategies in sub-Saharan Africa: a modelling study. PLoS Med. 2020;17: e1003377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Maxmen A. Scientists hail historic malaria vaccine approval—but point to challenges ahead. Nature. 2021. https://doi.org/10.1038/d41586-021-02755-5.

    Article  PubMed  Google Scholar 

  287. Pringle JC, Carpi G, Almagro-Garcia J, Zhu SJ, Kobayashi T, Mulenga M, et al. RTS, S/AS01 malaria vaccine mismatch observed among Plasmodium falciparum isolates from southern and central Africa and globally. Sci Rep. 2018;8:6622.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Diallo MA, L’Ollivier C, Diongue K, Badiane AS, Kodio A, Tall ML, et al. Spatiotemporal dynamic of the RTS, S/AS01 malaria vaccine target antigens in Senegal. Am J Trop Med Hyg. 2021;105:1738–46.

    Article  PubMed  Google Scholar 

  289. Collins KA, Snaith R, Cottingham MG, Gilbert SC, Hill AVS. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci Rep. 2017;7:46621.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Hviid L. Looking for needles in the plasmodial haystack. mSphere. 2019. https://doi.org/10.1128/mSphere.00146-19.

    Article  PubMed  PubMed Central  Google Scholar 

  291. Lozano JM, Rodriguez Parra Z, Hernandez-Martinez S, Yasnot-Acosta MF, Rojas AP, Marin-Waldo LS, et al. The search of a malaria vaccine: the time for modified immuno-potentiating probes. Vaccines. 2021. https://doi.org/10.3390/vaccines9020115.

    Article  PubMed  PubMed Central  Google Scholar 

  292. Mallory KL, Taylor JA, Zou X, Waghela IN, Schneider CG, Sibilo MQ, et al. Messenger RNA expressing PfCSP induces functional, protective immune responses against malaria in mice. NPJ Vaccines. 2021;6:84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Schneider CG, Taylor JA, Sibilo MQ, Miura K, Mallory KL, Mann C, et al. Orientation of antigen display on self-assembling protein nanoparticles influences immunogenicity. Vaccines. 2021. https://doi.org/10.3390/vaccines9020103.

    Article  PubMed  PubMed Central  Google Scholar 

  294. Pulendran B. Systems vaccinology: probing humanity’s diverse immune systems with vaccines. Proc Natl Acad Sci USA. 2014;111:12300–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. Bustamante LY, Powell GT, Lin YC, Macklin MD, Cross N, Kemp A, et al. Synergistic malaria vaccine combinations identified by systematic antigen screening. Proc Natl Acad Sci USA. 2017;114:12045–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. de Jong RM, Tebeje SK, Meerstein-Kessel L, Tadesse FG, Jore MM, Stone W, et al. Immunity against sexual stage Plasmodium falciparum and Plasmodium vivax parasites. Immunol Rev. 2020;293:190–215.

    Article  CAS  PubMed  Google Scholar 

  297. Birmingham K, Wunsch H. Grants will determine European malaria vaccine future. Nat Med. 1999;5:969.

    Article  CAS  PubMed  Google Scholar 

  298. Dubovsky F, Moree M, Rabinovich R. Malaria vaccines: orphans in transition. Dev Biol. 2002;110:47–53.

    CAS  Google Scholar 

  299. Chitnis CE, Sharma A. Targeting the Plasmodium vivax Duffy-binding protein. Trends Parasitol. 2008;24:29–34.

    Article  CAS  PubMed  Google Scholar 

  300. Tham WH, Beeson JG, Rayner JC. Plasmodium vivax vaccine research—we’ve only just begun. Int J Parasitol. 2017;47:111–8.

    Article  PubMed  Google Scholar 

  301. De SL, Ntumngia FB, Nicholas J, Adams JH. Progress towards the development of a P. vivax vaccine. Expert Rev Vaccines. 2021;20:97–112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  302. Wertheimer SP, Barnwell JW. Plasmodium vivax interaction with the human Duffy blood group glycoprotein: identification of a parasite receptor-like protein. Exp Parasitol. 1989;69:340–50.

    Article  CAS  PubMed  Google Scholar 

  303. Barnwell JW, Nichols ME, Rubinstein P. In vitro evaluation of the role of the Duffy blood group in erythrocyte invasion by Plasmodium vivax. J Exp Med. 1989;169:1795–802.

    Article  CAS  PubMed  Google Scholar 

  304. Fang XD, Kaslow DC, Adams JH, Miller LH. Cloning of the Plasmodium vivax Duffy receptor. Mol Biochem Parasitol. 1991;44:125–32.

    Article  CAS  PubMed  Google Scholar 

  305. Zimmerman PA. Plasmodium vivax infection in Duffy-negative people in Africa. Am J Trop Med Hyg. 2017;97:636–8.

    Article  PubMed  PubMed Central  Google Scholar 

  306. Kepple D, Pestana K, Tomida J, Abebe A, Golassa L, Lo E. Alternative invasion mechanisms and host immune response to Plasmodium vivax malaria: trends and future directions. Microorganisms. 2020. https://doi.org/10.3390/microorganisms9010015.

    Article  PubMed  PubMed Central  Google Scholar 

  307. Illingworth JJ, Alanine DG, Brown R, Marshall JM, Bartlett HE, Silk SE, et al. Functional comparison of blood-stage Plasmodium falciparum malaria vaccine candidate antigens. Front Immunol. 2019;10:1254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  308. Draper SJ, Sack BK, King CR, Nielsen CM, Rayner JC, Higgins MK, et al. Malaria vaccines: recent advances and new horizons. Cell Host Microbe. 2018;24:43–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  309. Russell B, Suwanarusk R, Malleret B, Costa FT, Snounou G, Baird JK, et al. Human ex vivo studies on asexual Plasmodium vivax: the best way forward. Int J Parasitol. 2012;42:1063–70.

    Article  PubMed  Google Scholar 

  310. Ramos GQ, Baia-da-Silva DC, Lacerda MVG, Monteiro WM, Lopes SCP. Viability and infectivity of Plasmodium vivax gametocytes in short-term culture. Front Cell Infect Microbiol. 2021;11: 676276.

    Article  PubMed  PubMed Central  Google Scholar 

  311. Galinski MR, Barnwell JW. Plasmodium vivax: merozoites, invasion of reticulocytes and considerations for malaria vaccine development. Parasitol Today. 1996;12:20–9.

    Article  CAS  PubMed  Google Scholar 

  312. Noulin F, Borlon C, Van Den Abbeele J, D’Alessandro U, Erhart A. 1912–2012: a century of research on Plasmodium vivax in vitro culture. Trends Parasitol. 2013;29:286–94.

    Article  PubMed  Google Scholar 

  313. Bermudez M, Moreno-Perez DA, Arevalo-Pinzon G, Curtidor H, Patarroyo MA. Plasmodium vivax in vitro continuous culture: the spoke in the wheel. Malar J. 2018;17:301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  314. Gunalan K, Rowley EH, Miller LH. A way forward for culturing Plasmodium vivax. Trends Parasitol. 2020;36:512–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  315. Trager W, Jensen JB. Human malaria parasites in continuous culture. Science. 1976;193:673–5.

    Article  CAS  PubMed  Google Scholar 

  316. Galinski MR, Medina CC, Ingravallo P, Barnwell JW. A reticulocyte-binding protein complex of Plasmodium vivax merozoites. Cell. 1992;69:1213–26.

    Article  CAS  PubMed  Google Scholar 

  317. Han JH, Lee SK, Wang B, Muh F, Nyunt MH, Na S, et al. Identification of a reticulocyte-specific binding domain of Plasmodium vivax reticulocyte-binding protein 1 that is homologous to the PfRh4 erythrocyte-binding domain. Sci Rep. 2016;6:26993.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  318. Gruszczyk J, Kanjee U, Chan LJ, Menant S, Malleret B, Lim NTY, et al. Transferrin receptor 1 is a reticulocyte-specific receptor for Plasmodium vivax. Science. 2018;359:48–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Chan LJ, Dietrich MH, Nguitragool W, Tham WH. Plasmodium vivax reticulocyte binding proteins for invasion into reticulocytes. Cell Microbiol. 2020;22: e13110.

    Article  CAS  PubMed  Google Scholar 

  320. Chim-Ong A, Surit T, Chainarin S, Roobsoong W, Sattabongkot J, Cui L, et al. The blood stage antigen RBP2-P1 of Plasmodium vivax binds reticulocytes and is a target of naturally acquired immunity. Infect Immun. 2020. https://doi.org/10.1128/IAI.00616-19.

    Article  PubMed  PubMed Central  Google Scholar 

  321. Cho JS, Russell B, Kosaisavee V, Zhang R, Colin Y, Bertrand O, et al. Unambiguous determination of Plasmodium vivax reticulocyte invasion by flow cytometry. Int J Parasitol. 2016;46:31–9.

    Article  PubMed  Google Scholar 

  322. Malleret B, El Sahili A, Tay MZ, Carissimo G, Ong ASM, Novera W, et al. Plasmodium vivax binds host CD98hc (SLC3A2) to enter immature red blood cells. Nat Microbiol. 2021;6:991–9.

    Article  CAS  PubMed  Google Scholar 

  323. Thomson-Luque R, Bautista JM. Home sweet home: Plasmodium vivax-infected reticulocytes—the younger the better? Front Cell Infect Microbiol. 2021;11: 675156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  324. Rayner JC, Galinski MR, Ingravallo P, Barnwell JW. Two Plasmodium falciparum genes express merozoite proteins that are related to Plasmodium vivax and Plasmodium yoelii adhesive proteins involved in host cell selection and invasion. Proc Natl Acad Sci USA. 2000;97:9648–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  325. Rayner JC, Vargas-Serrato E, Huber CS, Galinski MR, Barnwell JW. A Plasmodium falciparum homologue of Plasmodium vivax reticulocyte binding protein (PvRBP1) defines a trypsin-resistant erythrocyte invasion pathway. J Exp Med. 2001;194:1571–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  326. Triglia T, Thompson J, Caruana SR, Delorenzi M, Speed T, Cowman AF. Identification of proteins from Plasmodium falciparum that are homologous to reticulocyte binding proteins in Plasmodium vivax. Infect Immun. 2001;69:1084–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  327. Crosnier C, Bustamante LY, Bartholdson SJ, Bei AK, Theron M, Uchikawa M, et al. Basigin is a receptor essential for erythrocyte invasion by Plasmodium falciparum. Nature. 2011;480:534–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  328. Douglas AD, Baldeviano GC, Lucas CM, Lugo-Roman LA, Crosnier C, Bartholdson SJ, et al. A PfRH5-based vaccine is efficacious against heterologous strain blood-stage Plasmodium falciparum infection in aotus monkeys. Cell Host Microbe. 2015;17:130–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  329. Payne RO, Silk SE, Elias SC, Miura K, Diouf A, Galaway F, et al. Human vaccination against RH5 induces neutralizing antimalarial antibodies that inhibit RH5 invasion complex interactions. JCI Insight. 2017. https://doi.org/10.1172/jci.insight.96381.

    Article  PubMed  PubMed Central  Google Scholar 

  330. Douglas AD, Baldeviano GC, Jin J, Miura K, Diouf A, Zenonos ZA, et al. A defined mechanistic correlate of protection against Plasmodium falciparum malaria in non-human primates. Nat Commun. 2019;10:1953.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  331. Minassian AM, Silk SE, Barrett JR, Nielsen CM, Miura K, Diouf A, et al. Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination. Med. 2021;2(701–719): e719.

    Google Scholar 

  332. Lopez C, Yepes-Perez Y, Hincapie-Escobar N, Diaz-Arevalo D, Patarroyo MA. What Is Known about the immune response induced by Plasmodium vivax malaria vaccine candidates? Front Immunol. 2017;8:126.

    Article  PubMed  PubMed Central  Google Scholar 

  333. Franca CT, White MT, He WQ, Hostetler JB, Brewster J, Frato G, et al. Identification of highly-protective combinations of Plasmodium vivax recombinant proteins for vaccine development. Elife. 2017. https://doi.org/10.7554/eLife.28673.

    Article  PubMed  PubMed Central  Google Scholar 

  334. Gunalan K, Sa JM, Moraes Barros RR, Anzick SL, Caleon RL, Mershon JP, et al. Transcriptome profiling of Plasmodium vivax in Saimiri monkeys identifies potential ligands for invasion. Proc Natl Acad Sci USA. 2019;116:7053–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  335. Molina DM, Finney OC, Arevalo-Herrera M, Herrera S, Felgner PL, Gardner MJ, et al. Plasmodium vivax pre-erythrocytic-stage antigen discovery: exploiting naturally acquired humoral responses. Am J Trop Med Hyg. 2012;87:460–9.

    Article  PubMed  PubMed Central  Google Scholar 

  336. Hostetler JB, Sharma S, Bartholdson SJ, Wright GJ, Fairhurst RM, Rayner JC. A library of Plasmodium vivax recombinant merozoite proteins reveals new vaccine candidates and protein-protein interactions. PLoS Negl Trop Dis. 2015;9: e0004264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  337. Ndegwa DN, Hostetler JB, Marin-Menendez A, Sanderson T, Mwikali K, Verzier LH, et al. Using Plasmodium knowlesi as a model for screening Plasmodium vivax blood-stage malaria vaccine targets reveals new candidates. BioRxiv. 2020. https://doi.org/10.1371/journal.ppat.1008864.

    Article  Google Scholar 

  338. Aparici-Herraiz I, Gualdron-Lopez M, Castro-Cavadia CJ, Carmona-Fonseca J, Yasnot MF, Fernandez-Becerra C, et al. Antigen discovery in circulating extracellular vesicles from Plasmodium vivax patients. Front Cell Infect Microbiol. 2021;11: 811390.

    Article  CAS  PubMed  Google Scholar 

  339. Richie TL, Billingsley PF, Sim BK, James ER, Chakravarty S, Epstein JE, et al. Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine. 2015;33:7452–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  340. Vaughan AM, Kappe SHI. Genetically attenuated malaria parasites as vaccines. Expert Rev Vaccines. 2017;16:765–7.

    Article  CAS  PubMed  Google Scholar 

  341. Epstein JE, Richie TL. The whole parasite, pre-erythrocytic stage approach to malaria vaccine development: a review. Curr Opin Infect Dis. 2013;26:420–8.

    Article  CAS  PubMed  Google Scholar 

  342. Weiss WR, Jiang CG. Protective CD8+ T lymphocytes in primates immunized with malaria sporozoites. PLoS ONE. 2012;7: e31247.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  343. Sulyok Z, Fendel R, Eder B, Lorenz FR, Kc N, Karnahl M, et al. Heterologous protection against malaria by a simple chemoattenuated PfSPZ vaccine regimen in a randomized trial. Nat Commun. 2021;12:2518.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  344. Mwakingwe-Omari A, Healy SA, Lane J, Cook DM, Kalhori S, Wyatt C, et al. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature. 2021;595:289–94.

    Article  CAS  PubMed  Google Scholar 

  345. Minkah NK, Kappe SHI. Malaria vaccine gets a parasite boost in the liver. Nature. 2021;95:173–4.

    Article  CAS  Google Scholar 

  346. Pichyangkul S, Spring MD, Yongvanitchit K, Kum-Arb U, Limsalakpetch A, Im-Erbsin R, et al. Chemoprophylaxis with sporozoite immunization in P. knowlesi rhesus monkeys confers protection and elicits sporozoite-specific memory T cells in the liver. PLoS ONE. 2017;12: e0171826.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  347. Wijayalath W, Cheesman S, Tanabe K, Handunnetti S, Carter R, Pathirana S. Strain-specific protective effect of the immunity induced by live malarial sporozoites under chloroquine cover. PLoS ONE. 2012;7: e45861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  348. Moreno A, Joyner C. Malaria vaccine clinical trials: what’s on the horizon. Curr Opin Immunol. 2015;35:98–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  349. Gowda DC, Wu X. Parasite recognition and signaling mechanisms in innate immune responses to malaria. Front Immunol. 2018;9:3006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  350. Bucsan AN, Williamson KC. Setting the stage: the initial immune response to blood-stage parasites. Virulence. 2020;11:88–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  351. Dobbs KR, Crabtree JN, Dent AE. Innate immunity to malaria - the role of monocytes. Immunol Rev. 2020;293:8–24.

    Article  CAS  PubMed  Google Scholar 

  352. Antonelli LR, Junqueira C, Vinetz JM, Golenbock DT, Ferreira MU, Gazzinelli RT. The immunology of Plasmodium vivax malaria. Immunol Rev. 2020;293:163–89.

    Article  CAS  PubMed  Google Scholar 

  353. Kho S, Barber BE, Johar E, Andries B, Poespoprodjo JR, Kenangalem E, et al. Platelets kill circulating parasites of all major Plasmodium species in human malaria. Blood. 2018;132:1332–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  354. Kho S, Minigo G, Andries B, Leonardo L, Prayoga P, Poespoprodjo JR, et al. Circulating neutrophil extracellular traps and neutrophil activation are increased in proportion to disease severity in human malaria. J Infect Dis. 2019;219:1994–2004.

    Article  CAS  PubMed  Google Scholar 

  355. Fernandez D, Segura C, Arman M, McGill S, Burchmore R, Lopera-Mesa T. Uncomplicated Plasmodium vivax malaria: mapping the proteome from circulating platelets. Clin Proteomics. 2022;19:1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  356. Netea M, Schlitzer A, Placek K, Joosten LAB, Schultze JL. Innate and adaptive immune memory: an evolutionary continuum in the host’s response to pathogens. Cell Host Microbe. 2019;25:13–26.

    Article  CAS  PubMed  Google Scholar 

  357. Vidal M, Aguilar R, Campo JJ, Dobano C. Development of quantitative suspension array assays for six immunoglobulin isotypes and subclasses to multiple Plasmodium falciparum antigens. J Immunol Methods. 2018;455:41–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  358. Rogier E, Nace D, Dimbu PR, Wakeman B, Pohl J, Beeson JG, et al. Framework for characterizing longitudinal antibody response in children after Plasmodium falciparum infection. Front Immunol. 2021;12: 617951.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  359. Krishnamurty AT, Thouvenel CD, Portugal S, Keitany GJ, Kim KS, Holder A, et al. Somatically hypermutated Plasmodium-specific IgM(+) memory B cells are rapid, plastic, early responders upon malaria rechallenge. Immunity. 2016;45:402–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  360. Harms Pritchard G, Pepper M. Memory B cell heterogeneity: remembrance of things past. J Leukoc Biol. 2018;103:269–74.

    Article  CAS  PubMed  Google Scholar 

  361. Boyle MJ, Chan JA, Handayuni I, Reiling L, Feng G, Hilton A, et al. IgM in human immunity to Plasmodium falciparum malaria. Sci Adv. 2019;5:eaax4489.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  362. Walker MR, Knudsen AS, Partey FD, Bassi MR, Frank AM, Castberg FC, et al. Acquisition and decay of IgM and IgG responses to merozoite antigens after Plasmodium falciparum malaria in Ghanaian children. PLoS ONE. 2020;15: e0243943.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  363. Hopp CS, Sekar P, Diouf A, Miura K, Boswell K, Skinner J, et al. Plasmodium falciparum-specific IgM B cells dominate in children, expand with malaria, and produce functional IgM. J Exp Med. 2021;218: e20200901.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  364. Bohannon C, Powers R, Satyabhama L, Cui A, Tipton C, Michaeli M, et al. Long-lived antigen-induced IgM plasma cells demonstrate somatic mutations and contribute to long-term protection. Nat Commun. 2016;7:11826.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  365. Liu ZS, Sattabongkot J, White M, Chotirat S, Kumpitak C, Takashima E, et al. Naturally acquired antibody kinetics against Plasmodium vivax antigens in people from a low malaria transmission region in western Thailand. BMC Med. 2022;20:89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  366. Anyona SB, Raballah E, Cheng Q, Hurwitz I, Ndege C, Munde E, et al. Differential gene expression in host ubiquitination processes in childhood malarial anemia. Front Genet. 2021;12: 764759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  367. Walk J, Keramati F, de Bree LCJ, Arts RJW, Blok B, Netea MG, et al. Controlled human malaria infection induces long-term functional changes in monocytes. Front Mol Biosci. 2020;7: 604553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  368. Schrum JE, Crabtree JN, Dobbs KR, Kiritsy MC, Reed GW, Gazzinelli RT, et al. Cutting edge: Plasmodium falciparum induces trained innate immunity. J Immunol. 2018;200:1243–8.

    Article  CAS  PubMed  Google Scholar 

  369. Guha R, Mathioudaki A, Doumbo S, Doumtabe D, Skinner J, Arora G, et al. Plasmodium falciparum malaria drives epigenetic reprogramming of human monocytes toward a regulatory phenotype. PLoS Pathog. 2021;17: e1009430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  370. Gyssens IC, Netea MG. Heterologous effects of vaccination and trained immunity. Clin Microbiol Infect. 2019;25:1457–8.

    Article  CAS  PubMed  Google Scholar 

  371. Nahrendorf W, Ivens A, Spence PJ. Inducible mechanisms of disease tolerance provide an alternative strategy of acquired immunity to malaria. Elife. 2021. https://doi.org/10.7554/eLife.63838.

    Article  PubMed  PubMed Central  Google Scholar 

  372. Portugal S, Moebius J, Skinner J, Doumbo S, Doumtabe D, Kone Y, et al. Exposure-dependent control of malaria-induced inflammation in children. PLoS Pathog. 2014;10: e1004079.

    Article  PubMed  PubMed Central  Google Scholar 

  373. Tran TM, Guha R, Portugal S, Skinner J, Ongoiba A, Bhardwaj J, et al. A molecular signature in blood reveals a role for p53 in regulating malaria-induced inflammation. Immunity. 2019;51(750–765): e710.

    Google Scholar 

  374. Bousema T, Okell L, Felger I, Drakeley C. Asymptomatic malaria infections: detectability, transmissibility and public health relevance. Nat Rev Microbiol. 2014;12:833–40.

    Article  CAS  PubMed  Google Scholar 

  375. Nyarko PB, Claessens A. Understanding host-pathogen-vector interactions with chronic asymptomatic malaria infections. Trends Parasitol. 2021;37:195–204.

    Article  PubMed  Google Scholar 

  376. Weiss GE, Crompton PD, Li S, Walsh LA, Moir S, Traore B, et al. Atypical memory B cells are greatly expanded in individuals living in a malaria-endemic area. J Immunol. 2009;183:2176–82.

    Article  CAS  PubMed  Google Scholar 

  377. Weiss GE, Traore B, Kayentao K, Ongoiba A, Doumbo S, Doumtabe D, et al. The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog. 2010;6: e1000912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  378. Weiss GE, Clark EH, Li S, Traore B, Kayentao K, Ongoiba A, et al. A positive correlation between atypical memory B cells and Plasmodium falciparum transmission intensity in cross-sectional studies in Peru and Mali. PLoS ONE. 2011;6: e15983.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  379. Portugal S, Obeng-Adjei N, Moir S, Crompton PD, Pierce SK. Atypical memory B cells in human chronic infectious diseases: an interim report. Cell Immunol. 2017;321:18–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  380. Braddom AE, Batugedara G, Bol S, Bunnik EM. Potential functions of atypical memory B cells in Plasmodium-exposed individuals. Int J Parasitol. 2020;50:1033–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  381. Sutton HJ, Aye R, Idris AH, Vistein R, Nduati E, Kai O, et al. Atypical B cells are part of an alternative lineage of B cells that participates in responses to vaccination and infection in humans. Cell Rep. 2021;34: 108684.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  382. Braddom AE, Bol S, Gonzales SJ, Reyes RA, Musinguzi K, Nankya F, et al. B cell receptor repertoire analysis in malaria-naive and malaria-experienced individuals reveals unique characteristics of atypical memory B cells. mSphere. 2021;6: e0072621.

    Article  PubMed  Google Scholar 

  383. Reddy SB, Nagy N, Ronnberg C, Chiodi F, Lugaajju A, Heuts F, et al. Direct contact between Plasmodium falciparum and human B-cells in a novel co-culture increases parasite growth and affects B-cell growth. Malar J. 2021;20:303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  384. Perez-Mazliah D, Ndungu FM, Aye R, Langhorne J. B-cell memory in malaria: myths and realities. Immunol Rev. 2020;293:57–69.

    Article  CAS  PubMed  Google Scholar 

  385. Rivera-Correa J, Mackroth MS, Jacobs T, Schulze Zur Wiesch J, Rolling T, Rodriguez A. Atypical memory B-cells are associated with Plasmodium falciparum anemia through anti-phosphatidylserine antibodies. Elife. 2019. https://doi.org/10.7554/eLife.48309.

    Article  PubMed  PubMed Central  Google Scholar 

  386. Rivera-Correa J, Yasnot-Acosta MF, Tovar NC, Velasco-Pareja MC, Easton A, Rodriguez A. Atypical memory B-cells and autoantibodies correlate with anemia during Plasmodium vivax complicated infections. PLoS Negl Trop Dis. 2020;14: e0008466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  387. Rivera-Correa J, Rodriguez A. Autoimmune anemia in malaria. Trends Parasitol. 2020;36:91–7.

    Article  CAS  PubMed  Google Scholar 

  388. Daniel-Ribeiro CT. Is there a role for autoimmunity in immune protection against malaria? Mem Inst Oswaldo Cruz. 2000;95:199–207.

    Article  CAS  PubMed  Google Scholar 

  389. Daniel-Ribeiro CT, Zanini G. Autoimmunity and malaria: what are they doing together? Acta Trop. 2000;76:205–21.

    Article  CAS  PubMed  Google Scholar 

  390. Mourao LC, Roma PM, da Silva Sultane Aboobacar J, Medeiros CM, de Almeida ZB, Fontes CJ, et al. Anti-erythrocyte antibodies may contribute to anaemia in Plasmodium vivax malaria by decreasing red blood cell deformability and increasing erythrophagocytosis. Malar J. 2016;15:397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  391. Mourao LC, Baptista RP, de Almeida ZB, Grynberg P, Pucci MM, Castro-Gomes T, et al. Anti-band 3 and anti-spectrin antibodies are increased in Plasmodium vivax infection and are associated with anemia. Sci Rep. 2018;8:8762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  392. Barber BE, Grigg MJ, Piera K, Amante FH, William T, Boyle MJ, et al. Antiphosphatidylserine immunoglobulin M and immunoglobulin G antibodies are higher in vivax than falciparum malaria, and associated with early anemia in both species. J Infect Dis. 2019;220:1435–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  393. Mourao LC, Medeiros CMP, Cardoso-Oliveira GP, Roma P, Aboobacar J, Rodrigues BCM, et al. Effects of IgG and IgM autoantibodies on non-infected erythrocytes is related to ABO blood group in Plasmodium vivax malaria and is associated with anemia. Microbes Infect. 2020;22:379–83.

    Article  CAS  PubMed  Google Scholar 

  394. Calle CL, Mordmuller B, Singh A. Immunosuppression in malaria: do Plasmodium falciparum parasites hijack the host? Pathogens. 2021. https://doi.org/10.3390/pathogens10101277.

    Article  PubMed  PubMed Central  Google Scholar 

  395. Wei L, Adderley J, Leroy D, Drewry DH, Wilson DW, Kaushansky A, et al. Host-directed therapy, an untapped opportunity for antimalarial intervention. Cell Rep Med. 2021;2: 100423.

    Article  PubMed  PubMed Central  Google Scholar 

  396. Gupta A, Styczynski MP, Galinski MR, Voit EO, Fonseca LL. Dramatic transcriptomic differences in Macaca mulatta and Macaca fascicularis with Plasmodium knowlesi infections. Sci Rep. 2021;11:19519.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  397. Joyner CJ, MaHPIC-Consortium, Wood JS, Moreno A, Garcia A, Galinski MR. Case report: severe and complicated cynomolgi malaria in a rhesus macaque resulted in similar histopathological changes as those seen in human malaria. Am J Trop Med Hyg. 2017;97:548–55.

    Article  CAS  Google Scholar 

  398. Phillips N. The coronavirus is here to stay—here’s what that means. Nature. 2021;590:382–4.

    Article  CAS  PubMed  Google Scholar 

  399. Callaway E. Fast-spreading COVID variant can elude immune responses. Nature. 2021;589:500–1.

    Article  CAS  PubMed  Google Scholar 

  400. Duraisingh MT, Skillman KM. Epigenetic variation and regulation in malaria parasites. Annu Rev Microbiol. 2018;72:355–75.

    Article  CAS  PubMed  Google Scholar 

  401. Josling GA, Williamson KC, Llinas M. Regulation of sexual commitment and gametocytogenesis in malaria parasites. Annu Rev Microbiol. 2018;72:501–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  402. Usui M, Williamson KC. Stressed out about Plasmodium falciparum gametocytogenesis. Front Cell Infect Microbiol. 2021;11: 790067.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  403. Brown KN, Brown IN. Immunity to malaria: antigenic variation in chronic infections of Plasmodium knowlesi. Nature. 1965;208:1286–8.

    Article  CAS  PubMed  Google Scholar 

  404. Dentinger RM. Patterns of infection and patterns of evolution: how a malaria parasite brought “monkeys and man” closer together in the 1960s. J Hist Biol. 2016;49:359–95.

    Article  Google Scholar 

  405. Coatney GR. Simian malaria. Its importance to worldwide eradication of malaria. JAMA. 1963;184:876–7.

    Article  CAS  PubMed  Google Scholar 

  406. Eicher T, Kinnebrew G, Patt A, Spencer K, Ying K, Ma Q, et al. Metabolomics and multi-omics integration: a survey of computational methods and resources. Metabolites. 2020. https://doi.org/10.3390/metabo10050202.

    Article  PubMed  PubMed Central  Google Scholar 

  407. Vodovotz Y, Xia A, Read EL, Bassaganya-Riera J, Hafler DA, Sontag E, et al. Solving immunology? Trends Immunol. 2017;38:116–27.

    Article  CAS  PubMed  Google Scholar 

  408. Winzeler EA. Applied systems biology and malaria. Nat Rev Microbiol. 2006;4:145–51.

    Article  CAS  PubMed  Google Scholar 

  409. De Sousa KP, Doolan DL. Immunomics: a 21st century approach to vaccine development for complex pathogens. Parasitol. 2016;143:236–44.

    Article  Google Scholar 

  410. Lee HJ, Georgiadou A, Otto TD, Levin M, Coin LJ, Conway DJ, et al. Transcriptomic studies of malaria: a paradigm for investigation of systemic host-pathogen interactions. Microbiol Mol Biol Rev. 2018. https://doi.org/10.1128/MMBR.00071-17.

    Article  PubMed  PubMed Central  Google Scholar 

  411. Smith ML, Styczynski MP. Systems biology-based investigation of host-Plasmodium interactions. Trends Parasitol. 2018;34:617–32.

    Article  PubMed  PubMed Central  Google Scholar 

  412. Tsang JS. Utilizing population variation, vaccination, and systems biology to study human immunology. Trends Immunol. 2015;36:479–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  413. Gupta A, Galinski MR, Voit EO. Dynamic control balancing cell proliferation and inflammation is crucial for an effective immune response to malaria. Front Mol Biosci. 2022;15(8): 800721.

    Article  CAS  Google Scholar 

  414. Kazmin D, Nakaya HI, Lee EK, Johnson MJ, van der Most R, van den Berg RA, et al. Systems analysis of protective immune responses to RTS, S malaria vaccination in humans. Proc Natl Acad Sci USA. 2017;114:2425–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  415. Tsang JS, Schwartzberg PL, Kotliarov Y, Biancotto A, Xie Z, Germain RN, et al. Global analyses of human immune variation reveal baseline predictors of postvaccination responses. Cell. 2014;157:499–513.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  416. Netea MG, Li Y. Immune memory in individuals with COVID-19. Nat Cell Biol. 2021;23:582–4.

    Article  CAS  PubMed  Google Scholar 

  417. Chumakov K, Avidan MS, Benn CS, Bertozzi SM, Blatt L, Chang AY, et al. Old vaccines for new infections: exploiting innate immunity to control COVID-19 and prevent future pandemics. Proc Natl Acad Sci USA. 2021. https://doi.org/10.1073/pnas.2101718118.

    Article  PubMed  PubMed Central  Google Scholar 

  418. Voit EO. Models-of-data and models-of-processes in the post-genomic era. Math Biosci. 2002;180:263–74.

    Article  PubMed  Google Scholar 

  419. Voit EO. A first course in systems biology. 2nd ed. New York: Garland Science; 2017.

    Book  Google Scholar 

  420. Aurrecoechea C, Brestelli J, Brunk BP, Dommer J, Fischer S, Gajria B, et al. PlasmoDB: a functional genomic database for malaria parasites. Nucleic Acids Res. 2009;37:D539-543.

    Article  CAS  PubMed  Google Scholar 

  421. Bahl A, Brunk B, Crabtree J, Fraunholz MJ, Gajria B, Grant GR, et al. PlasmoDB: the Plasmodium genome resource. A database integrating experimental and computational data. Nucleic Acids Res. 2003;31:212–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  422. Otto TD, Bohme U, Jackson AP, Hunt M, Franke-Fayard B, Hoeijmakers WA, et al. A comprehensive evaluation of rodent malaria parasite genomes and gene expression. BMC Biol. 2014;12:86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  423. Bohme U, Otto TD, Sanders M, Newbold CI, Berriman M. Progression of the canonical reference malaria parasite genome from 2002–2019. Wellcome Open Res. 2019;4:58.

    Article  PubMed  PubMed Central  Google Scholar 

  424. Otto TD, Gilabert A, Crellen T, Bohme U, Arnathau C, Sanders M, et al. Genomes of all known members of a Plasmodium subgenus reveal paths to virulent human malaria. Nat Microbiol. 2018;3:687–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  425. Ramaprasad A, Klaus S, Douvropoulou O, Culleton R, Pain A. Plasmodium vinckei genomes provide insights into the pan-genome and evolution of rodent malaria parasites. BMC Biol. 2021;19:69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  426. Langhorne J, Buffet P, Galinski M, Good M, Harty J, Leroy D, et al. The relevance of non-human primate and rodent malaria models for humans. Malar J. 2011;10:23.

    Article  PubMed  PubMed Central  Google Scholar 

  427. Craig AG, Grau GE, Janse C, Kazura JW, Milner D, Barnwell JW, et al. The role of animal models for research on severe malaria. PLoS Pathog. 2012;8: e1002401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  428. Chappell L, Ross P, Orchard L, Russell TJ, Otto TD, Berriman M, et al. Refining the transcriptome of the human malaria parasite Plasmodium falciparum using amplification-free RNA-seq. BMC Genomics. 2020;21:395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  429. Carlton JM, Adams JH, Silva JC, Bidwell SL, Lorenzi H, Caler E, et al. Comparative genomics of the neglected human malaria parasite Plasmodium vivax. Nature. 2008;455:757–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  430. Pain A, Bohme U, Berry AE, Mungall K, Finn RD, Jackson AP, et al. The genome of the simian and human malaria parasite Plasmodium knowlesi. Nature. 2008;455:799–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  431. Auburn S, Bohme U, Steinbiss S, Trimarsanto H, Hostetler J, Sanders M, et al. A new Plasmodium vivax reference sequence with improved assembly of the subtelomeres reveals an abundance of pir genes. Wellcome Open Res. 2016;1:4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  432. Ford A, Kepple D, Abagero BR, Connors J, Pearson R, Auburn S, et al. Whole genome sequencing of Plasmodium vivax isolates reveals frequent sequence and structural polymorphisms in erythrocyte binding genes. PLoS Negl Trop Dis. 2020;14: e0008234.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  433. Brashear AM, Huckaby AC, Fan Q, Dillard LJ, Hu Y, Li Y, et al. New Plasmodium vivax genomes from the China–Myanmar border. Front Microbiol. 2020;11:1930.

    Article  PubMed  PubMed Central  Google Scholar 

  434. Lapp SA, Geraldo JA, Chien JT, Ay F, Pakala SB, Batugedara G, et al. PacBio assembly of a Plasmodium knowlesi genome sequence with Hi-C correction and manual annotation of the SICAvar gene family. Parasitol. 2018;145:71–84.

    Article  CAS  Google Scholar 

  435. Benavente ED, de Sessions PF, Moon RW, Grainger M, Holder AA, Blackman MJ, et al. A reference genome and methylome for the Plasmodium knowlesi A1–H.1 line. Int J Parasitol. 2018;48:191–6.

    Article  CAS  PubMed  Google Scholar 

  436. Pinheiro MM, Ahmed MA, Millar SB, Sanderson T, Otto TD, Lu WC. Plasmodium knowlesi genome sequences from clinical isolates reveal extensive genomic dimorphism. PLoS ONE. 2015;10: e0121303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  437. Oresegun DR, Daneshvar C, Cox-Singh J. Plasmodium knowlesi - clinical isolate genome sequencing to inform translational same-species model system for severe malaria. Front Cell Infect Microbiol. 2021;11: 607686.

    Article  PubMed  PubMed Central  Google Scholar 

  438. Roth A, Adapa SR, Zhang M, Liao X, Saxena V, Goffe R, et al. Unraveling the Plasmodium vivax sporozoite transcriptional journey from mosquito vector to human host. Sci Rep. 2018;8:12183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  439. Vivax SC. Transcriptome and histone epigenome of Plasmodium vivax salivary-gland sporozoites point to tight regulatory control and mechanisms for liver-stage differentiation in relapsing malaria. Int J Parasitol. 2019;49:501–13.

    Article  Google Scholar 

  440. Bozdech Z, Mok S, Hu G, Imwong M, Jaidee A, Russell B, et al. The transcriptome of Plasmodium vivax reveals divergence and diversity of transcriptional regulation in malaria parasites. Proc Natl Acad Sci USA. 2008;105:16290–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  441. Boopathi PA, Subudhi AK, Garg S, Middha S, Acharya J, Pakalapati D, et al. Revealing natural antisense transcripts from Plasmodium vivax isolates: evidence of genome regulation in complicated malaria. Infect Genet Evol. 2013;20:428–43.

    Article  CAS  PubMed  Google Scholar 

  442. Zhu L, Mok S, Imwong M, Jaidee A, Russell B, Nosten F, et al. New insights into the Plasmodium vivax transcriptome using RNA-Seq. Sci Rep. 2016;6:20498.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  443. Kim A, Popovici J, Vantaux A, Samreth R, Bin S, Kim S, et al. Characterization of P. vivax blood stage transcriptomes from field isolates reveals similarities among infections and complex gene isoforms. Sci Rep. 2017;7:7761.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  444. Kim A, Popovici J, Menard D, Serre D. Plasmodium vivax transcriptomes reveal stage-specific chloroquine response and differential regulation of male and female gametocytes. Nat Commun. 2019;10:371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  445. Siegel SV, Chappell L, Hostetler JB, Amaratunga C, Suon S, Bohme U, et al. Analysis of Plasmodium vivax schizont transcriptomes from field isolates reveals heterogeneity of expression of genes involved in host–parasite interactions. Sci Rep. 2020;10:16667.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  446. Sa JM, Cannon MV, Caleon RL, Wellems TE, Serre D. Single-cell transcription analysis of Plasmodium vivax blood-stage parasites identifies stage- and species-specific profiles of expression. PLoS Biol. 2020;18: e3000711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  447. Brito MAM, Baro B, Raiol TC, Ayllon-Hermida A, Safe IP, Deroost K, et al. Morphological and transcriptional changes in human bone marrow during natural Plasmodium vivax malaria infections. J Infect Dis. 2020;225:1274–83.

    Article  CAS  PubMed Central  Google Scholar 

  448. Bourgard C, Lopes SCP, Lacerda MVG, Albrecht L, Costa FTM. A suitable RNA preparation methodology for whole transcriptome shotgun sequencing harvested from Plasmodium vivax-infected patients. Sci Rep. 2021;11:5089.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  449. Acharya P, Pallavi R, Chandran S, Dandavate V, Sayeed SK, Rochani A, et al. Clinical proteomics of the neglected human malarial parasite Plasmodium vivax. PLoS ONE. 2011;6: e26623.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  450. Anderson DC, Lapp SA, Akinyi S, Meyer EV, Barnwell JW, Korir-Morrison C, et al. Plasmodium vivax trophozoite-stage proteomes. J Proteomics. 2015;115:157–76.

    Article  CAS  PubMed  Google Scholar 

  451. Anderson DC, Lapp SA, Barnwell JW, Galinski MR. A large scale Plasmodium vivax-Saimiri boliviensis trophozoite-schizont transition proteome. PLoS ONE. 2017;12: e0182561.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  452. Venkatesh A, Patel SK, Ray S, Shastri J, Chatterjee G, Kochar SK, et al. Proteomics of Plasmodium vivax malaria: new insights, progress and potential. Expert Rev Proteomics. 2016;13:771–82.

    Article  CAS  PubMed  Google Scholar 

  453. Venkatesh A, Lahiri A, Reddy PJ, Shastri J, Bankar S, Patankar S, et al. Identification of highly expressed Plasmodium Vivax proteins from clinical isolates using proteomics. Proteomics Clin Appl. 2018;12: e1700046.

    Article  CAS  PubMed  Google Scholar 

  454. Venkatesh A, Aggarwal S, Kumar S, Rajyaguru S, Kumar V, Bankar S, et al. Comprehensive proteomics investigation of P. vivax-infected human plasma and parasite isolates. BMC Infect Dis. 2020;20:188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  455. Lapp SA, Mok S, Zhu L, Wu H, Preiser PR, Bozdech Z, et al. Plasmodium knowlesi gene expression differs in ex vivo compared to in vitro blood-stage cultures. Malar J. 2015;14:1–16.

    Article  CAS  Google Scholar 

  456. Rutledge GG, Bohme U, Sanders M, Reid AJ, Cotton JA, Maiga-Ascofare O, et al. Plasmodium malariae and P. ovale genomes provide insights into malaria parasite evolution. Nature. 2017;542:101–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  457. International Human Genome Sequencing C. Finishing the euchromatic sequence of the human genome. Nature. 2004;431:931–45.

    Article  CAS  Google Scholar 

  458. Warren WC, Harris RA, Haukness M, Fiddes IT, Murali SC, Fernandes J, et al. Sequence diversity analyses of an improved rhesus macaque genome enhance its biomedical utility. Science. 2020. https://doi.org/10.1126/science.abc6617.

    Article  PubMed  PubMed Central  Google Scholar 

  459. Zimin AV, Cornish AS, Maudhoo MD, Gibbs RM, Zhang X, Pandey S, et al. A new rhesus macaque assembly and annotation for next-generation sequencing analyses. Biol Direct. 2014;9:1–15.

    Article  Google Scholar 

  460. Thomas GWC, Wang RJ, Puri A, Harris RA, Raveendran M, Hughes DST, et al. Reproductive longevity predicts mutation rates in primates. Curr Biol. 2018;28(3193–3197): e3195.

    Google Scholar 

  461. Baker JN, Walker JA, Vanchiere JA, Phillippe KR, St Romain CP, Gonzalez-Quiroga P. Evolution of Alu subfamily structure in the Saimiri lineage of New World monkeys. Genome Biol Evol. 2017;9:2365–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  462. Reardon S. A complete human genome sequence is close: how scientists filled in the gaps. Nature. 2021;594:158–9.

    Article  CAS  PubMed  Google Scholar 

  463. Nurk S, Koren S, Rhie A, Rautiainen M, Bzikadze AV, Mikheenko A, Vollger MR, Altemose N, Uralsky L, Gershman A, et al. The complete sequence of a human genome. Science. 2022;376:44–53.

    Article  CAS  PubMed  Google Scholar 

  464. Neafsey DE, Taylor AR, MacInnis BL. Advances and opportunities in malaria population genomics. Nat Rev Genet. 2021;22:502–17.

    Article  CAS  PubMed  Google Scholar 

  465. Dia A, Cheeseman IH. Single-cell genome sequencing of protozoan parasites. Trends Parasitol. 2021;37:803–14.

    Article  CAS  PubMed  Google Scholar 

  466. Tran TM, Li S, Doumbo S, Doumtabe D, Huang CY, Dia S, et al. An intensive longitudinal cohort study of Malian children and adults reveals no evidence of acquired immunity to Plasmodium falciparum infection. Clin Infect Dis. 2013;57:40–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  467. Tran TM, Jones MB, Ongoiba A, Bijker EM, Schats R, Venepally P, et al. Transcriptomic evidence for modulation of host inflammatory responses during febrile Plasmodium falciparum malaria. Sci Rep. 2016;6:31291.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  468. Portugal S, Tran TM, Ongoiba A, Bathily A, Li S, Doumbo S, et al. Treatment of chronic asymptomatic Plasmodium falciparum infection does not increase the risk of clinical malaria upon reinfection. Clin Infect Dis. 2017;64:645–53.

    Article  CAS  PubMed  Google Scholar 

  469. De Niz M, Heussler VT. Rodent malaria models: insights into human disease and parasite biology. Curr Opin Microbiol. 2018;46:93–101.

    Article  PubMed  Google Scholar 

  470. Threadgill DW, Miller DR, Churchill GA, de Villena FP. The collaborative cross: a recombinant inbred mouse population for the systems genetic era. ILAR J. 2011;52:24–31.

    Article  CAS  PubMed  Google Scholar 

  471. Abu Toamih Atamni H, Nashef A, Iraqi FA. The collaborative cross mouse model for dissecting genetic susceptibility to infectious diseases. Mamm Genome. 2018;29:471–87.

    Article  CAS  PubMed  Google Scholar 

  472. Mikolajczak SA, Vaughan AM, Kangwanrangsan N, Roobsoong W, Fishbaugher M, Yimamnuaychok N, et al. Plasmodium vivax liver stage development and hypnozoite persistence in human liver-chimeric mice. Cell Host Microbe. 2015;17:526–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  473. Schafer C, Dambrauskas N, Reynolds LM, Trakhimets O, Raappana A, Flannery EL. Partial protection against P. vivax infection diminishes hypnozoite burden and blood-stage relapses. Cell Host Microbe. 2021;29:752-756 e754.

    Article  CAS  PubMed  Google Scholar 

  474. Vaccari M, Franchini G. Memory T cells in rhesus macaques. Adv Exp Med Biol. 2010;684:126–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  475. Joyner CJ, Ley AM, Nguyen DC, Ali M, Corrado A, Tipton C, et al. Generation of human long-lived plasma cells by developmentally regulated epigenetic imprinting. Life Sci Alliance. 2022. https://doi.org/10.26508/lsa.202101285.

    Article  PubMed  Google Scholar 

  476. van Duivenvoorde LM, Voorberg-van der Wel A, van der Werff NM, Braskamp G, Remarque EJ, Kondova I, et al. Suppression of Plasmodium cynomolgi in rhesus macaques by coinfection with Babesia microti. Infect Immun. 2010;8:1032–9.

    Article  CAS  Google Scholar 

  477. Koehler JW, Bolton M, Rollins A, Snook K, deHaro E, Henson E, et al. Altered immune responses in rhesus macaques co-infected with SIV and Plasmodium cynomolgi: an animal model for coincident AIDS and relapsing malaria. PLoS ONE. 2009;4: e7139.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  478. Semenya AA, Sullivan JS, Barnwell JW, Secor WE. Schistosoma mansoni infection impairs antimalaria treatment and immune responses of rhesus macaques infected with mosquito-borne Plasmodium coatneyi. Infect Immun. 2012;80:3821–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  479. Liu G, Li Y, Qin L, Yan Y, Ye Y, Chen Y, et al. SIV infection aggravates malaria in a Chinese rhesus monkey coinfection model. BMC Infect Dis. 2019;19:965.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  480. Nyakundi RK, Hau J, Ogongo P, Nyamongo O, Jeneby M, Akinyi M, et al. Acquired clinical immunity to malaria in nonhuman primates coinfected with Schistosoma and Plasmodium parasites. Infect Immun. 2022;90: e0046421.

    Article  PubMed  Google Scholar 

  481. Gill US, Pallett LJ, Thomas N, Burton AR, Patel AA, Yona S, et al. Fine needle aspirates comprehensively sample intrahepatic immunity. Gut. 2019;68:1493–503.

    Article  CAS  PubMed  Google Scholar 

  482. Malaria Host–Pathogen Interaction Center. http://MaHPIC.emory.edu; www.systemsbiology.emory.edu.

  483. Gardinassi LG, Arevalo-Herrera M, Herrera S, Cordy RJ, Tran V, Smith MR, et al. Integrative metabolomics and transcriptomics signatures of clinical tolerance to Plasmodium vivax reveal activation of innate cell immunity and T cell signaling. Redox Biol. 2018;17:158–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  484. Gardinassi LG, Cordy RJ, Lacerda MVG, Salinas JL, Monteiro WM, Melo GC, et al. Metabolome-wide association study of peripheral parasitemia in Plasmodium vivax malaria. Int J Med Microbiol. 2017. https://doi.org/10.1016/j.ijmm.2017.09.002.

    Article  PubMed  PubMed Central  Google Scholar 

  485. DeBarry JD, Kissinger JC, Nural MV, Pakala SB, Humphrey JC, Meyer EVS, et al. Practical recommendations for supporting a systems biology cyberinfrastructure. Data Sci J. 2020;19:1–12.

    Article  Google Scholar 

  486. Access Data from MaHPIC—The Malaria Host–Pathogen Interaction Center. https://plasmodb.org/plasmo/app/static-content/PlasmoDB/mahpic.html.

  487. Candia J, Cheung F, Kotliarov Y, Fantoni G, Sellers B, Griesman T, et al. Assessment of variability in the SOMAscan assay. Sci Rep. 2017;7:14248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  488. Fogang B, Biabi MF, Megnekou R, Maloba FM, Essangui E, Donkeu C, et al. High prevalence of asymptomatic malarial anemia and association with early conversion from asymptomatic to symptomatic infection in a Plasmodium falciparum hyperendemic setting in Cameroon. Am J Trop Med Hyg. 2021;106:293–302.

    Article  PubMed  Google Scholar 

  489. Fonseca LL, Alezi HS, Moreno A, Barnwell JW, Galinski MR, Voit EO. Quantifying the removal of red blood cells in Macaca mulatta during a Plasmodium coatneyi infection. Malar J. 2016;15:410.

    Article  PubMed  PubMed Central  Google Scholar 

  490. Douglas NM, Anstey NM, Buffet PA, Poespoprodjo JR, Yeo TW, White NJ, et al. The anaemia of Plasmodium vivax malaria. Malar J. 2012;11:135.

    Article  PubMed  PubMed Central  Google Scholar 

  491. Tang Y, Joyner CJ, Cabrera-Mora M, Lapp SA, Nural MV, Pakala SB, et al. Integrative analysis associates monocytes with insufficient erythropoiesis during acute Plasmodium cynomolgi malaria in rhesus macaques. Malar J. 2017;16:1–16.

    CAS  Google Scholar 

  492. Wickramasinghe SN, Abdalla SH. Blood and bone marrow changes in malaria. Baillieres Best Pract Res Clin Haematol. 2000;13:277–99.

    Article  CAS  PubMed  Google Scholar 

  493. Tang Y, Joyner CJ, Cordy RJ, MaHPIC-Consortium, Galinski MR, Lamb TJ, et al. Multi-omics integrative analysis of acute and relapsing malaria in a nonhuman primate model of P. vivax infection. BioRxiv. 2019. https://doi.org/10.1101/564195.

    Article  Google Scholar 

  494. Fonseca LL, Joyner CJ, Saney CL, MaHPIC-Consortium, Moreno A, Barnwell JW, et al. Analysis of erythrocyte dynamics in rhesus macaque monkeys during infection with Plasmodium cynomolgi. Malar J. 2018;17:410.

    Article  PubMed  PubMed Central  Google Scholar 

  495. Fonseca LL, Voit EO. Comparison of mathematical frameworks for modeling erythropoiesis in the context of malaria infection. Math Biosci. 2015;270:224–36.

    Article  PubMed  PubMed Central  Google Scholar 

  496. Tang A, Gupta A, Garimalla S, MaHPIC-Consortium, Galinski MR, Styczynski MP, Fonseca LL, et al. Metabolic modeling helps interpret transcriptomic changes during malaria. Biochim Biophys Acta Mol Basis Dis. 2017;1864:2329–40.

    Article  CAS  PubMed  Google Scholar 

  497. Ball P. The lightning-fast quest for COVID vaccines—and what it means for other diseases. Nature. 2021;589:16–8.

    Article  CAS  PubMed  Google Scholar 

  498. DeGrace MM, Ghedin E, Frieman MB, Krammer F, Grifoni A, Alisoltani A, et al. Defining the risk of SARS-CoV-2 variants on immune protection. Nature. 2022. https://doi.org/10.1038/s41586-022-04690-5.

    Article  PubMed  Google Scholar 

  499. Thogersen RL, Holder AA, Hill AV, Arnot DE, Imoukhuede EB, Leroy O. Comparative decline in funding of European Commission malaria vaccine projects: what next for the European scientists working in this field? Malar J. 2011;10:255.

    Article  PubMed  PubMed Central  Google Scholar 

  500. Gancberg D, Hoeveler A, Martini A, Draghia-Akli R. Opportunities for vaccine research in Europe. Hum Vaccin Immunother. 2015;11:1917–20.

    Article  PubMed  PubMed Central  Google Scholar 

  501. Head MG, Brown RJ, Newell ML, Scott JAG, Batchelor J, Atun R. The allocation of US$105 billion in global funding from G20 countries for infectious disease research between 2000 and 2017: a content analysis of investments. Lancet Glob Health. 2020;8:e1295–304.

    Article  PubMed  PubMed Central  Google Scholar 

  502. Defining generations: where millennials end and generation Z begins. https://www.pewresearch.org/fact-tank/2019/01/17/where-millennials-end-and-generation-z-begins/.

  503. Collins WE. Interview with the expert: William E. Collins, Ph.D. interviewed by Vicki Glaser. Vector Borne Zoonotic Dis. 2009;9:569–72.

    Article  PubMed  Google Scholar 

  504. Tao D, McGill B, Hamerly T, Kobayashi T, Khare P, Dziedzic A, et al. A saliva-based rapid test to quantify the infectious subclinical malaria parasite reservoir. Sci Transl Med. 2019. https://doi.org/10.1126/scitranslmed.aan4479.

    Article  PubMed  PubMed Central  Google Scholar 

  505. Gimenez AM, Marques RF, Regiart M, Bargieri DY. Diagnostic methods for non-falciparum malaria. Front Cell Infect Microbiol. 2021;11: 681063.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  506. Burel JG, Apte SH, Groves PL, Boyle MJ, Langer C, Beeson JG, et al. Dichotomous miR expression and immune responses following primary blood-stage malaria. JCI Insight. 2017. https://doi.org/10.1172/jci.insight.93434.

    Article  PubMed  PubMed Central  Google Scholar 

  507. Berna AZ, McCarthy JS, Wang XR, Michie M, Bravo FG, Cassells J, et al. Diurnal variation in expired breath volatiles in malaria-infected and healthy volunteers. J Breath Res. 2018;12: 046014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  508. Colvin HN, Joice CR. Insights into malaria pathogenesis gained from host metabolomics. PLoS Pathog. 2020;16: e1008930.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  509. Miglar A, Reuling IJ, Yap XZ, Farnert A, Sauerwein RW, Asghar M. Biomarkers of cellular aging during a controlled human malaria infection. Sci Rep. 2021;11:18733.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Special thanks are extended to Eberhard O. Voit for his critical reading of an initial draft of this paper, and providing editorial feedback and suggestions, and to Chester J. Joyner for contributing his ideas and ongoing insights and discussion relating to malaria, immunity, pathogenesis, and research using nonhuman primates. The MaHPIC team is also acknowledged for 10 years of dedication and discussion towards improved understanding of malaria using nonhuman primates and systems biological approaches.

Funding

The MaHPIC work reviewed here was funded in part by the National Institute of Allergy and Infectious Diseases; National Institutes of Health, Department of Health and Human Services, which established the MaHPIC [Contract No. HHSN272201200031C; MG], the NIH Office of Research Infrastructure Programs/OD P51OD011132, the Defense Advanced Research Program Agency and the US Army Research Office via a cooperative agreement [Contract No. W911NF16C0008; MG], which funded the Technologies for Host Resilience-Host Acute Models of Malaria to study Experimental Resilience (THoR’s HAMMER) consortium. The funding bodies did not participate in the development of this article.

Author information

Authors and Affiliations

Authors

Contributions

MRG developed the content and wrote the manuscript. The author read and approved the final manuscript.

Authors' information

M.R.G has investigated primate malaria parasites and infections for over 30 years. This review highlights research progress during this span of time. She is retiring this year and looking forward to following the current and next generation’s advancements utilizing these resources.

Corresponding author

Correspondence to Mary R. Galinski.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Galinski, M.R. Systems biology of malaria explored with nonhuman primates. Malar J 21, 177 (2022). https://doi.org/10.1186/s12936-022-04199-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-022-04199-2

Keywords