Skip to main content
Discoveries logoLink to Discoveries
. 2024 Jun 30;12(2):e187. doi: 10.15190/d.2024.6

Recent Vaccines against Emerging and Tropical Infectious Diseases

Ismail Mazhar 1,*, Mir Muhammad Rai 1, Abdullah Ahmad 1, Natasha Nadeem 1, Aamir Shahid Javed 1, Hassan Mumtaz 2
PMCID: PMC11910014  PMID: 40093847

Abstract

Emerging diseases, re-emerging diseases and tropical diseases are a slowly progressing problem globally. This may in part be the result of shifting population, growing poverty, inadequate distribution of resources, or even complacency against personal hygiene. As a result of the low income and low standards of health in developing countries, they provide the perfect breeding grounds for the pathogens and parasites that are the root cause of Neglected Tropical diseases (NTDs). In the case of emerging diseases, most are of zoonotic origin and the recent COVID-19 pandemic is a key example. However, it is not just new diseases but re-emerging diseases such as Influenza that highlight the relentless nature of these infections. Vaccines represent the ultimate safety net against these diseases by bolstering immune systems and lowering subsequent mortality and morbidity of these conditions. In fact, against diseases with high mortalities such as AIDS, Hepatitis, and Malaria, vaccine development has markedly reduced mortality and prolonged life expectancy of those afflicted with these conditions. However, this research highlights the importance of enhancing vaccine efficacy and response. The review further underscores the necessity of research, the timing of vaccine administration, effective resource management by governments, and the perception of the population. Therefore, the review offers valuable insights for the medical community and the pharmaceutical industry in improving research and management to maximize the potential of vaccines.

Keywords: Vaccines, emerging, tropical, diseases.

SUMMARY

1. Introduction

2. Vaccines, usage, and their effects

3. Vaccines against Neglected Tropical Diseases

4. Vaccines against helminthic NTDs

  • 4.1. Soil-transmitted helminthiases - Hookworm, ascariasis, trichuriasis, strongyloidiasis

  • 4.2. Schistosomiasis:

  • 4.3. Lymphatic filariasis/podoconiosis

  • 4.4. Onchocerciasis

  • 4.5. Dracunculiasis

  • 4.6. Taeniasis/cysticercosis

  • 4.7. Echinococcosis

  • 4.8. Food - borne Trematodiases - Fascioliasis, clonorchiasis, opisthorchiasis, paragonimiasis

5. Vaccines against protozoal NTDs

  • 5.1. Leishmaniasis

  • 5.2. Human African trypanosomiasis

  • 5.3. American trypanosomiasis

  • 5.4. Scabies and other ectoparasites

6. Vaccines against bacterial NTDs

  • 6.1. Leprosy

  • 6.2. Buruli ulcer

  • 6.3. Trachoma

  • 6.4. Yaws/other endemic trepenomatoses

7. Vaccines against fungal NTDs

8. Vaccines against emerging diseases

9. EIDs with vaccines undergoing human trials

10. EIDs with vaccines in pre-clinical stage

11. Improvement in policies, practices implementation

12. Conclusion

1. Introduction

The emergence and reemergence of old and new infectious diseases are accompanied by the epidemic and pandemic challenges. The emergence and reemergence of diseases are accelerated by factors like rapid human development, and changes in demographics, populations, and the environment. This is also linked to zoonoses in the changing human-animal ecosystem and is affected by a growing globalized society whose geopolitical borders are unrecognized by pathogens. Infectious diseases affect over 1.6 billion people annually, and vaccines are the best prophylactic tool against them1. Neglected Tropical Diseases [NTDs] are a group of parasitic and related infectious diseases such as amebiasis, Chagas disease, Cysticercosis, Echinococcosis, Hookworm, Leishmaniasis, and Schistosomiasis, etc 2. These diseases are common in low and middle-income countries, living below the World Bank poverty line in conditions prevalent with Human Immunodeficiency Virus (HIV) and Malaria. Additionally, these diseases are highly prevalent in tropical and subtropical regions and are closely associated with poverty, wars, stigma, and marginalized populations who have no access to drinkable, clean water3. Despite their global public health importance, progress in developing vaccines for NTD pathogens has lagged due to technical and financial incentives2. However, the lack of vaccines against NTDs and EIDs [Emerging Infectious Diseases] is not shocking. Numerous difficulties such as the complex life cycle of some pathogens, low investment in research and lack of industry interest negatively impact the development of vaccines. Another problem is the recent rise of the anti-vaccination movement, which not only impacts public health and vaccination advances but also social and political stability1.

Elaborating further, NTDs and EIDs have been documented as being given a lack of attention, giving rise to lack of treatment options and deficient prevention. Tropical diseases impact poverty demographic, representing an unrecognized and major impact of these diseases globally, and are a major hurdle to efforts to enhance human health and reduce poverty.

2. Vaccines, usage, and their effects

NTDs are a major threat. These have not been sufficiently discussed or examined regarding their distinctive characteristics. It is important to identify emerging and reemerging neglected tropical diseases. NTDs and emerging and reemerging infectious diseases include HIV, Dengue, Ebola, Chagas disease, Malaria, Leishmaniasis, Zika, and Chikungunya1.

EIDs are an additional category that have a major impact on global health. However, EIDs are a result of recently discovered pathogens that have a zoonotic origin while in contrast NTDs comprise mainly of ancient diseases. The theory of coevolution between hosts and pathogens states that eventually, the pathogens develop capabilities to move onto new hosts hence highlighting their sheer unpredictability and the consequent inability to respond to them4. This classification first came to global notice in the 1960s with the appearance of viral hemorrhagic fevers, such as Ebola and Crimean-Congo hemorrhagic fever.

Along with NTDs, EIDs also threaten global public health, as they can cause a rise in unpredictable pandemics. Vaccination is one of the most efficient ways to control and prevent infectious diseases. Although technological development has been on the rise in the last few decades, the development of a vaccine that induces a protective and safe immune response is a rather difficult task, especially against most of the NTDs and EIDs5. However, the recent advances in vaccine development have allowed the development and licensing of new vaccines that can directly target these diseases6.

3. Vaccines against Neglected Tropical Diseases

Vaccine development for NTDs has faced many obstacles, the primary challenge remaining the chronic and debilitating nature of the conditions which affect the economic productivity of the region in addition to long-term consequences on human health. As NTDs nearly exclusively occur in the world’s population living in extreme poverty - nearly 700 to 800 million, the complexity of their management “traps” populations in a vicious cycle of poverty7. Other barriers in the slow progress of vaccine development have been cited to be greater interest in multi-drug administration programs, lack of funding in Personal Development Planning [PDPs], antigen/adjuvant access discoveries a growing “anti-vax” movement in the world, lack of effective product development and especially, reduced industry interest to invest7. Geopolitical challenges are also detrimental, as any initiation of clinical development is impeded by endemic barriers, as there is limited access to standardized equipment, health literacy, human resources, and laboratories in the affected tropics2.

Initially, NTD vaccines in the 20th century comprised live attenuated or killed organisms. The difficulty in maintaining living organisms and the costs required, however, has resulted in the favoring of expanding R&D efforts through the use of genomes and proteomes for NTD pathogens. Currently, the stages of vaccine development for NTDs (as classified by the WHO)8 have been summarized in Table 1. Only three NTDs have licensed vaccines (dengue, yellow fever, and rabies), while vaccines for the remaining are either in various stages of clinical testing or have had no progress in vaccine development efforts - the “neglected” vaccine development for these remaining NTDs is evident and is further discussed.

Table 1. NTDs, Stages of Vaccine Development and Type of Vaccines; Adapted and modified from2,7,8.

NTD Stage of vaccine development Type of Vaccine
Helminths
Hookworm infection Phase 1 – 2 Recombinant protein Vaccine
Ascariasis Preclinical RNA Vaccine
Trichuriasis Preclinical Recombinant protein Vaccine
Strongyloidiasis
Schistosomiasis Phase 1 – 2 Protein Subunit Vaccine
Lymphatic filariasis/Podoconiosis
Onchocerciasis Preclinical Protein Subunit Vaccine
Dracunculiasis
Taeniasis/cysticercosis Veterinary transmission blocking vaccine Protein Subunit Vaccine
Echinococcosis Veterinary transmission blocking vaccine Protein Subunit Vaccine
Foodborne trematodiases Preclinical Protein Subunit Vaccine
Protozoa
Leishmaniasis Phase 1 – 2 DNA Vaccine
Human African trypanosomiasis
American trypanosomiasis Preclinical Protein Subunit Vaccine
Scabies and other ectoparasites Preclinical Recombinant protein Vaccine
Bacteria
Leprosy Phase 1 Protein Subunit Vaccine
Buruli ulcer Preclinical Protein Subunit Vaccine
Trachoma Phase 1 Protein Subunit Vaccine
Yaws/other endemic treponematoses
Viruses
Yellow Fever Licensed Live attenuated vaccine
Rabies Licensed Inactivated vaccine
Dengue and Chikungunya Licensed vaccine and additional candidates in clinical development Live attenuated vaccine
Fungi
Mycetoma
Chromoblastomycosis and other deep mycoses

4. Vaccines against helminthic NTDs

4.1. Soil-transmitted helminthiases - Hookworm, Aascariasis, Trichuriasis, Strongyloidiasis

Compared to other diseases, vaccination efforts in soil-transmitted helminthiases face greater challenges due to their complex life cycles in the environment, avoiding adverse side-effects of vaccines, difficulty in choosing appropriate animal models, and the suitable route of administration of vaccine needed for the immune response9. The helminths include Hookworm, Ascariasis, Trichuriasis, and Srongyloidiasis, all in various stages of vaccine development10.

4.2. Schistosomiasis

The nature of the adult schistosomes is the primary obstacle to vaccine development because of their ability to evade human immune systems, added to by the complex multi-stage cycle and by the potential to elicit allergic reactions11,12. For the development of a vaccine, Sm-p80 has been considered the most promising candidate It is postulated that the induction of a balanced immune response by a vaccine candidate would be ideal13. However, the resource-limited nature of endemic regions and the aforementioned immunological complexity continue to pose challenges in vaccine development14.

4.3. Lymphatic filariasis/podoconiosis

There have been no human clinical trials for lymphatic filariasis vaccination underway yet, however, several vaccine candidates have been identified, with the understanding that a vaccine combined with targeted chemotherapy is an optimal approach for the elimination of this disease. Compared to the ease of vaccine studies in animal models in other studies, the life cycle of Wuchereria bancrofti makes it difficult to maintain in rodent models under laboratory conditions15,16. Vaccine development attempts include cocktail vaccines, multisubunit vaccines, multi-epitope vaccines, chimeric vaccines, and multivalent vaccines17. The nature of the re-emergence of this disease makes it crucial to develop a prophylactic vaccine to eliminate its spread18.

4.4. Onchocerciasis

Although the global prevalence of onchocerciasis has reduced through mass drug administration (MDA) of Ivermectin, the infection is still difficult to control because Ivermectin cannot be administered in endemic areas co-endemic with loiasis due to the risk of serious adverse effects. Also, ivermectin is not given to children under the age of five, which makes them both vulnerable to infection as well as major reservoirs of transmission19. Further, the elimination of onchocerciasis is complicated by the emerging drug resistance to ivermectin20. A vaccine is essential to reduce the global burden of onchocerciasis. The Excretory/Secretory products (ESPs) of Onchocerca volvulus have revealed potential vaccine candidates in the form of functional proteins21. Thus far, two adjuvanted recombinant antigens, Ov-103 and Ov-RAL-2, have been selected for the development of bivalent vaccines based on consistent results of the induction of protective immunity in mice. The bivalent vaccine is currently in clinical trials in naturally infected cattle and requires further testing in animal models before being forwarded to clinical trials in humans. The utilization of such a vaccine would be vaccination in children under the age of five, which will both prevent infection in children who cannot be given ivermectin, reduce the use of drugs, delay drug resistance, and ultimately, reduce the disease burden as it would reduce adult worm loads.

4.5. Dracunculiasis

As of 2022, efforts in Dracunculiasis eradication are only centered around prevention through interventions like prevention of water contamination, and there is no vaccine nor any medication available for the disease22.

4.6. Taeniasis/cysticercosis

Currently, only a veterinary transmission-blocking vaccine exists in the form of porcine vaccines23. There has been no progress for a human vaccine, as animal vaccines remain cheaper. However, calreticulin, a tegument protein of the parasite, has been explored as a potential vaccine candidate, which has demonstrated reduced worm burden in hamster models24.

4.7. Echinococcosis

A veterinary transmission-blocking vaccine is present for echinococcus. For the development of human vaccines, DNA vaccines with antigen B and recombinant protein vaccines have shown protective immune responses25. Additionally, multi-epitope proteins are helpful for immunity against E. granulosus through immunoinformatics approaches. A recombinant leucine aminopeptidase vaccine has been discovered as a potential vaccine antigen of E. multilocularis26.

4.8. Food-borne Trematodiases - Fascioliasis, Clonorchiasis, Opisthorchiasis, Paragonimiasis

Cathepsin 1 (CL1) is considered the antigen of choice for the development of a diagnostic tool and as a potential vaccine candidate27. Studies have been conducted to assess the efficacy of this antigen in F. hepatica and F. Gigantica for the development of a vaccine through trials conducted on animal models in sheep, cows, and goats28-30. Further, a chimeric vaccine antigen prepared from FhCatL1 and leucine aminopeptidase (FhLAP) also was demonstrated to elicit an immune response in both rabbit and sheep models31. As for F. Gigantica, a study showed the protective efficacy of a combined recombinant vaccine in mice comprising recominant® pro-proteins of cathepsin L1H and B3 (rproFgCatL1H and reproFgCatB3)32. A multi-epitope subunit vaccine was also found to be suitable for further investigation for the development of a vaccine against Fasciola Gigantica33. However, a meta-analysis found the overall pooled efficacy for all vaccine candidates to be non-significant and indicated further need for testing in animal models34.

Candidates for the serodiagnosis of Clonorchiasis have been identified35. These can be further studied to test for immune response in animal models. Also, significant worm reduction rates were observed in rats vaccinated orally Bacillus subtilis spores expressing C. sinensis proteins, which has suggested the possibility of preventing clonorchiasis transmission through an oral vaccine in addition to killing cercariea/metacercariae, which are the second intermediate host35,36.

There is no current vaccine for Opisthorchis, either. However, successful vaccination has been reported in hamster models using exosome-like extracellular vesicles, recombinant proteins, and chimeric subunit vaccines37-39.

Only one study has compared the in-vitro and in-vivo excretory-secretory products for any Paragonimus species (lung fluke), which may indicate targets for the development of vaccines40.

5. Vaccines against protozoal NTDs

5.1. Leishmaniasis

Due to the complicated host-agent interaction, the development of a vaccine is difficult, however, vaccination has promising potential due to the life-long immunity induced by recovery from natural infection41,42.

For cutaneous leishmaniasis, five live attenuated vaccines have been explored43, and in general, there are Phase 1 and Phase 2 clinical trials underway for the development of a vaccine. Challenges to vaccine development have been stated to be the lack of appropriate adjuvants, which have been discussed in previous literature44,45. Progress has also been evident on the exploration of nano-vaccines in previous reviews46.

5.2. Human African trypanosomiasis

Human African trypanosomiasis has a complex nature which makes it difficult to develop a vaccine. It is documented that Trypanosoma brucei causes loss of immunological memory by disrupting B-cell response and can also evade human immune systems47. There is, hence, sparse literature on the development of a vaccine but a multi-epitope vaccine has been formulated48.

5.3. American trypanosomiasis

A successful vaccine against the parasite is planned to be able to induce type 1 cytokines, cytotoxic T lymphocytes and a lytic antibody response Most studies have tested, in murine models, the efficacy of genes/proteins as prophylactic vaccines49,50. The efficacy of α-Gal epitope, Galα13Galβ14GlcNAc bound to human serum albumin (HAS) was examined. C57BL/6 mice immunized with this vaccine elicited anti-α-Gal antibody-mediated humoral reaction and were protected from lethal challenge infection with T. cruzi Y strain (1 × 105 parasite inoculum). A decline was observed in T-cell permeation in tissues, cardiac inflammation, necrotic myocytes and parasite burden in the heart of vaccinated mice51. In a recent study, Bivona et al. investigated the 80 kDa prolyl oligopeptidase (Tc80) as a novel immunogen for Chagas vaccine. Mice were immunized with recombinant Salmonella encoding Tc80 (STc80) and recombinant Tc80 protein in a prime-boost approach and displayed that immunized mouse elicited splenic production of Th1 cytokines, such as IFN-γ, IL-2 and TNF-α, Tc80-specific, complement-dependent trypanolytic antibodies, as well as polyfunctional CD4+ T cells and cytotoxic T lymphocytes associated with significant protection from challenge infection and chronic pathology52. For the mice infected with T. cruzi Tulahuen cl2 strain, rather than the individual treatments with the vaccine or Bz alone the combined treatment with the Bz and the vaccinenhad a more positive effect on the course of heart disease51. For example, delivery of DNA vaccine via the intradermal/electroporation route (vs. intramuscular route) was most effective in generating protective immunity to challenge infection and this vaccine was the simplest in design49. This is the first case of the immunogenicity of T. cruzi-derived recombinant antigens formulated as an emulsion with a TLR4 agonist in a non-human primate model. Our results strongly support the need for further exploration of the effect of the vaccine in a therapeutic model of naturally-infected Chagasic non-human primates and evaluation of the preventive efficacy of this type of vaccine, which would strengthen the rationale for the clinical development as a human vaccine against Chagas disease53.

5.4. Scabies and other ectoparasites

The transcriptomic profile of the parasite and its interactions with a person’s immune system has been studied, therefore helping to increase understanding to support research for vaccine production54,55. Currently, an anti-mite vaccine has been tested in mice and a subunit cocktail vaccine has been demonstrated to reduce transmission in rabbits, with more trials needed before the vaccine candidates proceed to clinical stages56,57.

6. Vaccines against bacterial NTDs

6.1. Leprosy

Immunoprophylaxis with the Bacillus Calmette-Guérin [BCG] vaccine is currently the most effective intervention to prevent leprosy58. However, the efficacy rates of the BCG vaccine, and with BCG revaccination in combination with chemoprophylaxis not showing very encouraging results for the prevention of leprosy transmission, a Phase 1 clinical trial for the development of a leprosy vaccine is underway comprising a recombinant antigen of Mycobacterium leprae59.

6.2. Buruli ulcer

Current treatment includes antibiotics, surgery, and the use of BCG vaccine. However, the BCG vaccine’s effects are short-term, and mouse models have been utilized to demonstrate the efficacy of recombinant BCG which expresses the antigenic M. ulcerans proteins60. Other proteins like Ag85A have been assessed for potential use in human clinical trials61, in addition to work on a multi-epitope vaccine utilizing the PE-PGRS protein through an integrated vaccinomics approach62. A study also identified a particular strain of M. ulcerans which could be used to set up immunization studies to test the efficacy of vaccine candidates63. Vaccine development remains in pre-clinical stages, with additional R&D required for the development of both diagnostic and preventive tools64.

6.3. Trachoma

Due to the complex life cycle of Chlamydia, research has extensively explored its molecular pathogenesis to identify potential stages of intervention and proteins of immunogenicity65,66. More studies are needed, however, to explore the importance of interferon-gamma production and how it correlates to protection from infection67. MOMP is a surface antigen most expressed in Chlamydia, and has been explored as an oral vaccine, with efficacy demonstrated in mice68. The most recent progress has been the development of a multivalent vaccine called CTH522 which consists of MOMP proteins and is the only vaccine candidate undergoing Phase 1 clinical trials in humans69.

6.4. Yaws/other endemic trepenomatoses

The Outer Membrane Proteins [OMPs] of Treponema subspecies can be further studied along with identifying B-cell epitopes to support vaccine development efforts70. It is recommended that the vaccine candidate tprL should be studied for vaccination development, and the ability of phase variation resulting in antigenic diversity should be considered71.

7. Vaccines against fungal NTDs

There has been no vaccine development against fungal infections, let alone fungal NTDs. The use of immunoinformatics tools, and in silico approaches have been recommended to be useful for the development of an efficient fungal vaccine72. Given that any development would be directed towards more prevalent fungal infections, it is likely the fungal NTDs will remain neglected for a while yet.

8. Vaccines against emerging diseases

Due to the “emerging” nature of this disease, the historical vaccine efforts for these diseases have been hampered by lack of funds and poor market potential, leaving the Global health community vulnerable in times of an outbreak, as seen with the COVID-19 pandemic and Ebola Epidemic73. To ameliorate this oversight and bolster global response to future pandemics and epidemics, WHO has released an R&D blueprint for high-priority pathogens that can cause an epidemic74. Together with Coalition for Epidemic Preparedness Innovations (CEPI), these efforts have done much to improve the vaccine development for these EIDs, however, it is sobering to realize that the preventive pipeline for this disease still lags behind the efforts for NTDs75. The vaccine stages can be seen in Table 2.

Table 2. EIDs, Stages of Vaccine Development and Type of Vaccines.

EID Stage of vaccine development Type of Vaccine
Crimean-Congo Hemorrhagic Fever Mostly animal models being tested (a Bulgarian vaccine present but it's unlikely to gain international approval) DNA virus vaccine the most promising in animal models (Bulgarian vaccine is live attenuated)
Ebola Virus Licensed rVSV based vaccine
Lassa fever Phase 1 DNA vaccine
Nipah Virus Fever Only animal models tested. Vaccine available for horses (Equivac HeV). Multiple models
Rift Valley Fever Phase 2 Adenovirus based
Covid-19 Licensed Multiple licensed

9. EIDs with vaccines undergoing human trials

After the coronavirus, Ebola virus, Marburg Virus and Lassa Virus have the vaccines furthest along in development.

Ebola virus is included in WHO’s blueprint for high-priority diseases due to its high mortality rate and the likelihood of person-to-person contact. Before the 2013 outbreak, several vaccine candidates showed promise in small rodent models, but only a few went into the more advanced stages of development. These efforts were also hampered by a dire lack of funds, low market potential, and low cross-protection between viral species73. Nonetheless, the 2013 epidemic resulted in increased interest in an efficient vaccine, and many Phase I/II trials were initiated for many different types of vaccines such as a recombinant adenovirus vector (consisting of Ad5.EBOV GP)76, replication incompetent modified vaccinia Ankara (MVA) vector77 and DNA vaccines78.

Recombinant vesicular stomatitis virus (rVSV) showed the most promise with its 2015 Phase III trial giving critical evidence of the utility of a ringed vaccination strategy in an Ebola outbreak79. Indeed, even WHO saw VSV as a good candidate platform for vaccines for an outbreak80. Subsequently, a rVSV-based vaccine (Ervebo) was FDA-approved and licensed as the first-ever Ebola Vaccine81.

Lassa Virus (LAV) vaccine development has seen a similar trajectory as the Ebola virus vaccine. Included in WHO’s high-priority pathogen list due to high mortality rate, lack of therapeutic options and struggles encountered while controlling it during an outbreak79, its vaccine efforts have been impeded by its low commercial prospects not offsetting the high cost of vaccine development82 and high genetic and biodiversity shown by the virus83. Nonetheless, when compared to other EIDs, LAV has one of the most robust development pipelines with many vaccine candidates showing success in animal models and potential for human testing75.

WHO has released a rigorous profile for an ideal LAV vaccine which includes elements such as affordability, long-term stability [negating needs for cold chain facilities], protection for special populations [HIV patients, pregnant women, children], efficacy and protection that lasts 3 years[with an option to extend them via boosters] and an uncomplicated method of administration84 Several vaccines candidates such as a Measles virus-based vaccine85 and an rVSV vaccine86, fulfill several of these objectives. From all of the available, a DNA vaccine for Lassa virus was the first to enter Phase 1 of human clinical trials [NCT03805984], with more predicated to follow based on the robust pipeline of Lassa virus and the fact that CEPI has guaranteed funding until phase 2 clinical trials75.

Rift Valley Fever Virus (RVFV) is another pathogen added to the WHO blueprint due to its high risk of geographical expansion and high morbidity associated with it87. The majority of the vaccine efforts for RVFV have been aimed at Veterinary use88. Development of human vaccines has been limited by the strict regulations surrounding human vaccine approval and the need for efficacy studies (which can be difficult for diseases such as RVFV which has intermittent occurrence). Two vaccines have been evaluated for humans; a live attenuated one (MP-12)89and a formalin-inactivated (TSI-GSD-200)90. Unfortunately, the licensing of these vaccines is limited by safety concerns (MP-12 has shown to be a teratogen in sheep)91 and practicality concerns (TSI-GSD-200 needs multiple to be efficacious)90. An adenovirus vaccine (ChAdOx1) has shown great potential in animal studies and is expected to enter Phase 1 human trials as well92. Considering these ground realities and the fact that RVFV outbreaks in animals precede human epidemics, the current best way to deal with RFVF is to centre your vaccination strategy on animals88.

10. EIDs with vaccines in pre-clinical stage

Nipah Virus (NiV) is a zoonotic virus that causes respiratory and neurological symptoms in humans (its mortality going up to 80%, such as in the case of the Bangladeshi variant)93 causing outbreaks almost annually. Passive immunization options include a recombinant human monoclonal antibody (m102.4) that was found to be safe and well tolerated in a Phase 1 trial94. For active immunization, current vaccine vectors, consisting of some of the same vectors as Ebola and Lassa Virus, such as rVSV, Measles, and Adenovirus are under development along with sub-unit vaccines and mRNA vaccines95. While many of them have shown great promise in animal trials, none of them have reached the stage of clinical trials in humans, due in part to the complexity of running such trials on a rare disease like Nipah fever96. Nonetheless, CEPI now supports several NiV human vaccine candidates, and the recent extensive body of research data can help in the creation of a vaccine that can be licensed in an outbreak. There are also options such as the Equivac® horse vaccine that can help break transmission of the virus to people.

Crimean-Congo hemorrhagic fever virus (CCHFV) is another high-priority pathogen with a global case fatality rate of 40% with higher rates in the developing world97. In addition to the problems faced by other EIDs, CCHFV vaccine development is further complicated by the fact that the virus is asymptomatic in most animals. Newborn mice do show signs of the disease, but they are poor models for research because of their immature immune systems98. This has been mitigated by the development of humanized mouse models99 and cynomolgus macaque models100 and they have been a boon in finding suitable vaccine candidates. Just like other EIDs, multiple different vaccine candidates, such as virus vector vaccines, mRNA, DNA, and sub-unit vaccines have shown success in animal models101. DNA vaccines have been the first to show results in primates and as such give credence for their use in human trials102. Do note that there is a live-attenuated vaccine that has been in use in Bulgaria since the 1960s103 but the data on it has been limited and it’s unlikely that this vaccine will ever get global approval due to safety concerns.

The genetic variability of CCHFV (which results in difficulty creating a single vaccine that could tackle the different virus strains) and the over-reliance on prototype IbAr10200 CCHFV strain (common in ticks but has an unknown virulence status in humans) in most vaccine studies are still huge concerns that must be addressed. The use of heterologous vaccine studies could be one solution98. Presently, the stages of vaccine development for EIDs have been summarized in Table 2 and Figure 1.

Figure 1. Stages of Vaccine Development.

Figure 1

11. Improvement in policies, practices implementation

Neglected tropical diseases (NTDs), have been granted inadequate research due to limited resource allocation, and lead to few interventions. However, new challenges have risen despite the amazing progress made in the fight against neglected tropical diseases. Better drugs, innovative diagnostics and new insecticides are often identified as the priority; however, access to these new tools may not be sufficient to attain and maintain disease elimination, if certain challenges and priorities are not considered. Systemic healthcare policies are needed to eliminate NTDs globally notably in India, China, Russia, Brazil, and South Africa. However, the present policies, practices, and research on them do not meet the need. In terms of policies, attention has been continuously focused on biomedical sciences and excluded ecological, social sciences and interdisciplinary approaches104.

Similarly,international collaborations and public-private partnerships have not been focused on systematically. The call from Tungiasis articulated the need a globally scalable collaboration amongst the stakeholders of endemic countries and to develop culturally appropriate communication techniques105. Despite marginal improvements in the vaccine and drugs for NTDs, the insufficiency in terms of disease surveillance, control, treatment, prevention, and elimination of NTDs is unmet106. A study in Ethiopia provided productive results on implementing an intervention to integrate the diagnosis, reporting, management and prevention of four common NTDs into Ethiopia’s primary healthcare system. The intervention consisted of providing health workers with supportive supervision, adapted job aids, and improved medical and diagnostic supplies. It was implemented for six months, and acceptability and cost-effectiveness were evaluated. Results indicate improvement in detecting, recording and managing target NTDs107. Additionally, five steps are suggested for effective policymaking, implementation, and eradication of NTDs which include Community engagement and formalization of community health workers’ role and shift of financial support from disease-oriented programs to availability of donated drugs in health care structures, disease-integrated interventions and improved access to international guidelines for primary health care staff106.

In the case of Emerging infectious diseases (EIDs), the problem that further complicates any development of preventative measures is the unpredictability of the pathogens. Moreover, in recent studies, it has been found that the increasingly erratic climate change has led to the increased redistribution of animal species which increases the subsequent risk to the public in those areas. The Intergovernmental Panel on Climate Change (IPCC) has further elaborated on this risk in terms of climate-related hazards and vulnerability of human and ecological systems. To meet this evolving challenge of EIDs, it has become vital to form a unified approach that addresses human, environmental, and animal health together and not the previous single-aim approach that barely yielded results. Hence the aptly coined “One Health Approach”27. The ‘One Health Approach’ was first named in 2003-2004 and was associated with the 2003 emergence of SARS and the emergence of a particularly virulent strain of avian flu. To elaborate, the One Health Approach focuses on the response and action at the human-animal-ecological interface, especially keeping in light the zoonotic diseases. Additionally, the core of this approach is interdisciplinary collaboration and is envisioned by numerous organizations such as WHO and UNICEF to also include other fields such as biodiversity, social sciences, and ecology28.

Advances in vaccine development have resulted in a variety of new vaccines being introduced into recommended immunization schedules. Armenia introduced the the pneumococcal conjugate vaccine (PCV) and rotavirus vaccine (RV). Multilevel logistic regression models were used to evaluate community- and individual-level factors associated with uptake. When developing strategies for new vaccine implementation, characteristics of the patient such as residence, distance to health clinics, age and siblings must be considered. Further exploration of cluster differences may provide better evaluation based on the increased uptake of these and other new vaccines16.

A recurring issue is the unpredictability of the outbreaks of any emerging infectious disease. Consequently, this makes it difficult to stockpile vaccines for extended amounts of time as they have short shelf lives and even if some can be preserved by methods such as cold chains, the required infrastructure is often extremely challenging to manage in developing countries where these diseases are most prone to emerge. However, the development of mRNA vaccines has rendered these concerns obsolete, owing to how quickly they can be manufactured, and the costs subsequently lowered by employing self-amplifying RNA. This solution was especially proven during the COVID-19 pandemic where rapid development of mRNA vaccines provided the safety blanket required by the global population to return to normalcy once again. Thus, the development of such types of vaccines may prove to be the solution for further rapid responses to any new EID29.

12. Conclusion

It is concluded that vaccines are undeniably effective in providing a crucial safety net against emerging, reemerging, and tropical diseases by strengthening immune systems, reducing mortality, and extending the life expectancy of affected individuals. However, the effectiveness and response of the vaccines depend on the extent of research, the timing of administration, governmental resource management, and public perception. Therefore, it is further concluded that reform and proper management are essential to fully realize the potential of vaccines. The review is likely to benefit medical community and pharmaceutical industry in aligning to latest research and management of resources to maximize the benefits of vaccines.

Acknowledgments

CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan. This study did not receive any funding in any form.

Footnotes

Conflict of interests: The authors would like to disclose no conflict of interest related to the publication of the article.

Abbreviations: Neglected Tropical Diseases (NTDs); Emerging Tropical Diseases (EIDs); World Health Organisation (WHO); Acquired Immunodeficiency Syndrome (AIDS); Human Immunodeficiency Virus (HIV); Research and Development (R and D); Major Outer Membrane Protein (MOMP); Outer Membrane Protein (OMP).

DISCOVERIES is a peer-reviewed, open access, online, multidisciplinary and integrative journal, publishing high impact and innovative manuscripts from all areas related to MEDICINE, BIOLOGY and CHEMISTRY

References

  • 1.Vaccines against Emerging and Neglected Infectious Diseases: An Overview. Vuitika Larissa, Prates-Syed Wasim A, Silva Jaqueline Dinis Queiros, Crema Karin P, Côrtes Nelson, Lira Aline, Lima Julia Beatriz Menuci, Camara Niels Olsen Saraiva, Schimke Lena F, Cabral-Marques Otavio, Sadraeian Mohammad, Chaves Lorena C S, Cabral-Miranda Gustavo. Vaccines. 2022;10(9) doi: 10.3390/vaccines10091385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Vaccines to combat the neglected tropical diseases. Bethony Jeffrey M, Cole Rhea N, Guo Xiaoti, Kamhawi Shaden, Lightowlers Marshall W, Loukas Alex, Petri William, Reed Steven, Valenzuela Jesus G, Hotez Peter J. Immunological reviews. 2011;239(1):237–70. doi: 10.1111/j.1600-065X.2010.00976.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Neglected tropical diseases: an effective global response to local poverty-related disease priorities. Engels Dirk, Zhou Xiao-Nong. Infectious diseases of poverty. 2020;9(1):10. doi: 10.1186/s40249-020-0630-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Emerging infectious disease: An underappreciated area of strategic concern for food security. Brooks Daniel R, Hoberg Eric P, Boeger Walter A, Trivellone Valeria. Transboundary and emerging diseases. 2022;69(2):254–267. doi: 10.1111/tbed.14009. [DOI] [PubMed] [Google Scholar]
  • 5.Emerging Concepts and Technologies in Vaccine Development. Brisse Morgan, Vrba Sophia M, Kirk Natalie, Liang Yuying, Ly Hinh. Frontiers in immunology. 2020;11:583077. doi: 10.3389/fimmu.2020.583077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.The Many Faces of Emerging and Reemerging Infectious Disease. Lessler Justin, Orenstein Walter A. Epidemiologic reviews. 2019;41(1):1–5. doi: 10.1093/epirev/mxz011. [DOI] [PubMed] [Google Scholar]
  • 7.The global fight to develop antipoverty vaccines in the anti-vaccine era. Hotez Peter J. Human vaccines & immunotherapeutics. 2018;14(9):2128–2131. doi: 10.1080/21645515.2018.1430542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Neglected tropical diseases. Geneva: World Health Organization. https://www.who.int/health-topics/neglected-tropical-diseases#tab=tab_1. https://www.who.int/health-topics/neglected-tropical-diseases#tab=tab_1.
  • 9.New Routes and Opportunities for Modular Construction of Particulate Vaccines: Stick, Click, and Glue. Brune Karl D, Howarth Mark. Frontiers in immunology. 2018;9:1432. doi: 10.3389/fimmu.2018.01432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Soil-Transmitted Helminth Vaccines: Are We Getting Closer? Zawawi Ayat, Else Kathryn J. Frontiers in immunology. 2020;11:576748. doi: 10.3389/fimmu.2020.576748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Immunological Considerations for Schistosoma Vaccine Development: Transitioning to Endemic Settings. Driciru Emmanuella, Koopman Jan Pieter R, Cose Stephen, Siddiqui Afzal A, Yazdanbakhsh Maria, Elliott Alison M, Roestenberg Meta. Frontiers in immunology. 2021;12:635985. doi: 10.3389/fimmu.2021.635985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fifteen Years of Sm-p80-Based Vaccine Trials in Nonhuman Primates: Antibodies From Vaccinated Baboons Confer Protection in vivo and in vitro From Schistosoma mansoni and Identification of Putative Correlative Markers of Protection. Zhang Weidong, Le Loc, Ahmad Gul, Molehin Adebayo J, Siddiqui Arif J, Torben Workineh, Karmakar Souvik, Rojo Juan U, Sennoune Souad, Lazarus Samara, Khatoon Sabiha, Freeborn Jasmin, Sudduth Justin, Rezk Ashraf F, Carey David, Wolf Roman F, Papin James F, Damian Ray, Gray Sean A, Marks Florian, Carter Darrick, Siddiqui Afzal A. Frontiers in immunology. 2020;11:1246. doi: 10.3389/fimmu.2020.01246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Schistosomiasis-from immunopathology to vaccines. McManus Donald P, Bergquist Robert, Cai Pengfei, Ranasinghe Shiwanthi, Tebeje Biniam Mathewos, You Hong. Seminars in immunopathology. 2020;42(3):355–371. doi: 10.1007/s00281-020-00789-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.A comprehensive and critical overview of schistosomiasis vaccine candidates. Al-Naseri Aya, Al-Absi Samar, El Ridi Rashika, Mahana Noha. Journal of parasitic diseases : official organ of the Indian Society for Parasitology. 2021;45(2):557–580. doi: 10.1007/s12639-021-01387-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lymphatic filariasis vaccine development: neglected for how long? Chavda Vivek P, Pandya Anjali, Pulakkat Sreeranjini, Soniwala Moinuddin, Patravale Vandana. Expert review of vaccines. 2021;20(11):1471–1482. doi: 10.1080/14760584.2021.1990760. [DOI] [PubMed] [Google Scholar]
  • 16.In-silico design of a multi-epitope vaccine candidate against onchocerciasis and related filarial diseases. Shey Robert Adamu, Ghogomu Stephen Mbigha, Esoh Kevin Kum, Nebangwa Neba Derrick, Shintouo Cabirou Mounchili, Nongley Nkemngo Francis, Asa Bertha Fru, Ngale Ferdinand Njume, Vanhamme Luc, Souopgui Jacob. Scientific reports. 2019;9(1):4409. doi: 10.1038/s41598-019-40833-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Evaluating the Vaccine Potential of a Tetravalent Fusion Protein (rBmHAXT) Vaccine Antigen Against Lymphatic Filariasis in a Mouse Model. Chauhan Nikhil, Khatri Vishal, Banerjee Priyankana, Kalyanasundaram Ramaswamy. Frontiers in immunology. 2018;9:1520. doi: 10.3389/fimmu.2018.01520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Next Step Lymphatic Filariasis Eradication: Current Status in the Development of a Vaccine Against Lymphatic Filariasis. Kalyanasundaram Ramaswamy. Lymphatic Filariasis. 2018:33-46. [Google Scholar]
  • 19.From river blindness to river epilepsy: Implications for onchocerciasis elimination programmes. Colebunders Robert, Siewe Fodjo Joseph Nelson, Hopkins Adrian, Hotterbeekx An, Lakwo Thomson L, Kalinga Akili, Logora Makoy Yibi, Basáñez Maria-Gloria. PLoS neglected tropical diseases. 2019;13(7):e0007407. doi: 10.1371/journal.pntd.0007407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Advancing a Human Onchocerciasis Vaccine From Antigen Discovery to Efficacy Studies Against Natural Infection of Cattle With Onchocerca ochengi. Zhan Bin, Bottazzi Maria Elena, Hotez Peter J, Lustigman Sara. Frontiers in cellular and infection microbiology. 2022;12:869039. doi: 10.3389/fcimb.2022.869039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.The Functional Parasitic Worm Secretome: Mapping the Place of Onchocerca volvulus Excretory Secretory Products. Vanhamme Luc, Souopgui Jacob, Ghogomu Stephen, Ngale Njume Ferdinand. Pathogens (Basel, Switzerland) 2020;9(11) doi: 10.3390/pathogens9110975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Progress Toward Global Eradication of Dracunculiasis - Worldwide, January 2021-June 2022. Hopkins Donald R, Weiss Adam J, Yerian Sarah, Sapp Sarah G H, Cama Vitaliano A. MMWR. Morbidity and mortality weekly report. 2022;71(47):1496–1502. doi: 10.15585/mmwr.mm7147a2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vaccine for a neglected tropical disease Taenia solium cysticercosis: fight for eradication against all odds. Kaur Rimanpreet, Arora Naina, Rawat Suraj S, Keshri Anand Kumar, Sharma Shubha Rani, Mishra Amit, Singh Gagandeep, Prasad Amit. Expert review of vaccines. 2021;20(11):1447–1458. doi: 10.1080/14760584.2021.1967750. [DOI] [PubMed] [Google Scholar]
  • 24.Insights into the diagnosis, vaccines, and control of Taenia solium, a zoonotic, neglected parasite. Hossain Md Shahadat, Shabir Shafqat, Toye Philip, Thomas Lian F, Falcone Franco H. Parasites & vectors. 2023;16(1):380. doi: 10.1186/s13071-023-05989-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Current situation and future prospects of <i>Echinococcus granulosus</i> vaccine candidates: A systematic review. Anvari Davood, Rezaei Fatemeh, Ashouri Alireza, Rezaei Saeed, Majidiani Hamidreza, Pagheh Abdol Sattar, Rezaei Fatemeh, Shariatzadeh Seyyed Ali, Fotovati Amir, Siyadatpanah Abolghasem, Gholami Shirzad, Ahmadpour Ehsan. Transboundary and Emerging Diseases. 2020;68(3):1080-1096. doi: 10.1111/tbed.13772. [DOI] [PubMed] [Google Scholar]
  • 26.A multi-method and structure-based in silico vaccine designing against Echinococcus granulosus through investigating enolase protein. Pourseif Mohammad Mostafa, Yousefpour Mitra, Aminianfar Mohammad, Moghaddam Gholamali, Nematollahi Ahmad. BioImpacts : BI. 2019;9(3):131–144. doi: 10.15171/bi.2019.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.High relatedness of bioinformatic data and realistic experimental works on the potentials of Fasciola hepatica and F. gigantica cathepsin L1 as a diagnostic and vaccine antigen. Fereig Ragab M, Metwally Samy, El-Alfy El-Sayed, Abdelbaky Hanan H, Shanab Obeid, Omar Mosaab A, Alsayeqh Abdullah F. Frontiers in public health. 2022;10:1054502. doi: 10.3389/fpubh.2022.1054502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Phage display-based vaccine with cathepsin L and excretory-secretory products mimotopes of Fasciola hepatica induces protective cellular and humoral immune responses in sheep. Villa-Mancera Abel, Olivares-Pérez Jaime, Olmedo-Juárez Agustín, Reynoso-Palomar Alejandro. Veterinary parasitology. 2021;289:109340. doi: 10.1016/j.vetpar.2020.109340. [DOI] [PubMed] [Google Scholar]
  • 29.Antibody recognition of cathepsin L1-derived peptides in Fasciola hepatica-infected and/or vaccinated cattle and identification of protective linear B-cell epitopes. Garza-Cuartero Laura, Geurden Thomas, Mahan Suman M., Hardham John M., Dalton John P., Mulcahy Grace. Vaccine. 2018;36(7):958-968. doi: 10.1016/j.vaccine.2018.01.020. [DOI] [PubMed] [Google Scholar]
  • 30.Vaccination with cathepsin L mimotopes of Fasciola hepatica in goats reduces worm burden, morphometric measurements, and reproductive structures. Villa-Mancera Abel, Alcalá-Canto Yazmín, Olivares-Pérez Jaime, Molina-Mendoza Pedro, Hernández-Guzmán Karina, Utrera-Quintana Fernando, Carreón-Luna Lorenzo, Olmedo-Juárez Agustín, Reynoso-Palomar Alejandro. Microbial Pathogenesis. 2021;155:104859. doi: 10.1016/j.micpath.2021.104859. [DOI] [PubMed] [Google Scholar]
  • 31.Moderate protection is induced by a chimeric protein composed of leucine aminopeptidase and cathepsin L1 against Fasciola hepatica challenge in sheep. Ortega-Vargas S, Espitia C, Sahagún-Ruiz A, Parada C, Balderas-Loaeza A, Villa-Mancera A, Quiroz-Romero H. Vaccine. 2019;37(24):3234–3240. doi: 10.1016/j.vaccine.2019.04.067. [DOI] [PubMed] [Google Scholar]
  • 32.The combined recombinant cathepsin L1H and cathepsin B3 vaccine against Fasciola gigantica infection. Kueakhai Pornanan, Changklungmoa Narin, Cheukamud Werachon, Osotprasit Supawadee, Chantree Pathanin, Preyavichyapugdee Narin, Sobhon Prasert, Meemon Krai. Parasitology international. 2021;83:102353. doi: 10.1016/j.parint.2021.102353. [DOI] [PubMed] [Google Scholar]
  • 33.Fasciola gigantica vaccine construct: an in silico approach towards identification and design of a multi-epitope subunit vaccine using calcium binding EF-hand proteins. Das Kanhu Charan, Konhar Ruchishree, Biswal Devendra Kumar. BMC immunology. 2023;24(1):1. doi: 10.1186/s12865-022-00535-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Protective efficacy of liver fluke DNA vaccines: A systematic review and meta-analysis: Guiding novel vaccine development. Jayaraj Rama, Kumarasamy Chellan, Norbury Luke, Piedrafita David, Smooker Peter. Veterinary parasitology. 2019;267:90–98. doi: 10.1016/j.vetpar.2019.01.010. [DOI] [PubMed] [Google Scholar]
  • 35.Clonorchis sinensis and clonorchiasis. Na Byoung-Kuk, Pak Jhang Ho, Hong Sung-Jong. Acta tropica. 2020;203:105309. doi: 10.1016/j.actatropica.2019.105309. [DOI] [PubMed] [Google Scholar]
  • 36.Recent Progress in the Development of Liver Fluke and Blood Fluke Vaccines. McManus Donald P. Vaccines. 2020;8(3) doi: 10.3390/vaccines8030553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vaccination of hamsters with Opisthorchis viverrini extracellular vesicles and vesicle-derived recombinant tetraspanins induces antibodies that block vesicle uptake by cholangiocytes and reduce parasite burden after challenge infection. Chaiyadet Sujittra, Sotillo Javier, Krueajampa Watchara, Thongsen Sophita, Brindley Paul J, Sripa Banchob, Loukas Alex, Laha Thewarach. PLoS neglected tropical diseases. 2019;13(5):e0007450. doi: 10.1371/journal.pntd.0007450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Recombinant Opisthorchis viverrini tetraspanin expressed in Pichia pastoris as a potential vaccine candidate for opisthorchiasis. Phung Luyen Thi, Chaiyadet Sujittra, Hongsrichan Nuttanan, Sotillo Javier, Dieu Hang Dinh Thi, Tran Canh Quang, Brindley Paul J, Loukas Alex, Laha Thewarach. Parasitology research. 2019;118(12):3419–3427. doi: 10.1007/s00436-019-06488-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Partial protection with a chimeric tetraspanin-leucine aminopeptidase subunit vaccine against Opisthorchis viverrini infection in hamsters. Thi Phung Luyen, Chaiyadet Sujittra, Hongsrichan Nuttanan, Sotillo Javier, Dinh Thi Dieu Hang, Quang Tran Canh, Brindley Paul J, Loukas Alex, Laha Thewarach. Acta tropica. 2020;204:105355. doi: 10.1016/j.actatropica.2020.105355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Comparative proteomics of adult Paragonimus kellicotti excretion/secretion products released in vitro or present in the lung cyst nodule. Di Maggio Lucia S, Curtis Kurt C, Erdmann-Gilmore Petra, Sprung Robert S W, Townsend R Reid, Weil Gary J, Fischer Peter U. PLoS neglected tropical diseases. 2022;16(8):e0010679. doi: 10.1371/journal.pntd.0010679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Leishmaniasis. Geneva: World Health Organization. https://www.who.int/news-room/fact-sheets/detail/leishmaniasis https://www.who.int/news-room/fact-sheets/detail/leishmaniasis
  • 42.Cutaneous Leishmaniasis: A 2022 Updated Narrative Review into Diagnosis and Management Developments. de Vries Henry J C, Schallig Henk D. American journal of clinical dermatology. 2022;23(6):823–840. doi: 10.1007/s40257-022-00726-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.The potential of live attenuated vaccines against Cutaneous Leishmaniasis. Zabala-Peñafiel A, Todd D, Daneshvar H, Burchmore R. Experimental parasitology. 2020;210:107849. doi: 10.1016/j.exppara.2020.107849. [DOI] [PubMed] [Google Scholar]
  • 44.Particulate and non-particle adjuvants in Leishmaniasis vaccine designs: A review. Kelleci Kübra, Allahverdiyev Adil, Bağirova Melahat, Ihlamur Murat, Abamor Emrah Ş. Journal of vector borne diseases. 2023;60(2):125–141. doi: 10.4103/0972-9062.361179. [DOI] [PubMed] [Google Scholar]
  • 45.Visceral leishmaniasis: An overview of vaccine adjuvants and their applications. Ratnapriya Sneha, Keerti K, Sahasrabuddhe Amogh A, Dube Anuradha. Vaccine. 2019;37(27):3505–3519. doi: 10.1016/j.vaccine.2019.04.092. [DOI] [PubMed] [Google Scholar]
  • 46.From infection to vaccination: reviewing the global burden, history of vaccine development, and recurring challenges in global leishmaniasis protection. Volpedo Greta, Huston Ryan H, Holcomb Erin A, Pacheco-Fernandez Thalia, Gannavaram Sreenivas, Bhattacharya Parna, Nakhasi Hira L, Satoskar Abhay R. Expert review of vaccines. 2021;20(11):1431–1446. doi: 10.1080/14760584.2021.1969231. [DOI] [PubMed] [Google Scholar]
  • 47.Detrimental Effect of Trypanosoma brucei brucei Infection on Memory B Cells and Host Ability to Recall Protective B-cell Responses. Moon Sangphil, Janssens Ibo, Kim Kyung Hyun, Stijlemans Benoit, Magez Stefan, Radwanska Magdalena. The Journal of infectious diseases. 2022;226(3):528–540. doi: 10.1093/infdis/jiac112. [DOI] [PubMed] [Google Scholar]
  • 48.Immunoinformatic design of a putative multi-epitope vaccine candidate against Trypanosoma brucei gambiense. Danazumi Ammar Usman, Iliyasu Gital Salahuddin, Idris Salisu, Bs Dibba Lamin, Balogun Emmanuel Oluwadare, Górna Maria Wiktoria. Computational and structural biotechnology journal. 2022;20:5574–5585. doi: 10.1016/j.csbj.2022.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Immunity and vaccine development efforts against Trypanosoma cruzi. Rios Lizette E, Vázquez-Chagoyán Juan Carlos, Pacheco Antonio Ortega, Zago M Paola, Garg Nisha J. Acta tropica. 2019;200:105168. doi: 10.1016/j.actatropica.2019.105168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.A therapeutic vaccine prototype induces protective immunity and reduces cardiac fibrosis in a mouse model of chronic Trypanosoma cruzi infection. Barry Meagan A, Versteeg Leroy, Wang Qian, Pollet Jeroen, Zhan Bin, Gusovsky Fabian, Bottazzi Maria Elena, Hotez Peter J, Jones Kathryn M. PLoS neglected tropical diseases. 2019;13(5):e0007413. doi: 10.1371/journal.pntd.0007413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.A prophylactic α-Gal-based glycovaccine effectively protects against murine acute Chagas disease. Portillo Susana, Zepeda Brenda G, Iniguez Eva, Olivas Janet J, Karimi Nasim H, Moreira Otacilio C, Marques Alexandre F, Michael Katja, Maldonado Rosa A, Almeida Igor C. NPJ vaccines. 2019;4:13. doi: 10.1038/s41541-019-0107-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Chagas disease vaccine design: the search for an efficient Trypanosoma cruzi immune-mediated control. Bivona Augusto E, Alberti Andrés Sánchez, Cerny Natacha, Trinitario Sebastián N, Malchiodi Emilio L. Biochimica et biophysica acta. Molecular basis of disease. 2020;1866(5):165658. doi: 10.1016/j.bbadis.2019.165658. [DOI] [PubMed] [Google Scholar]
  • 53.Safety and immunogenicity of a recombinant vaccine against Trypanosoma cruzi in Rhesus macaques. Dumonteil Eric, Herrera Claudia, Tu Weihong, Goff Kelly, Fahlberg Marissa, Haupt Erin, Kaur Amitinder, Marx Preston A, Ortega-Lopez Jaime, Hotez Peter J, Bottazzi Maria Elena. Vaccine. 2020;38(29):4584–4591. doi: 10.1016/j.vaccine.2020.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ectoparasites: Scabies. Thomas Cristina, Coates Sarah J, Engelman Daniel, Chosidow Olivier, Chang Aileen Y. Journal of the American Academy of Dermatology. 2020;82(3):533–548. doi: 10.1016/j.jaad.2019.05.109. [DOI] [PubMed] [Google Scholar]
  • 55.Transcriptome Analysis of Host Inflammatory Responses to the Ectoparasitic Mite Sarcoptes scabiei var. hominis. Shehwana Huma, Ijaz Sadaf, Fatima Abeera, Walton Shelley, Sheikh Zafar Iqbal, Haider Waseem, Naz Shumaila. Frontiers in immunology. 2021;12:778840. doi: 10.3389/fimmu.2021.778840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Designing the next generation of proton-exchange membrane fuel cells. Jiao Kui, Xuan Jin, Du Qing, Bao Zhiming, Xie Biao, Wang Bowen, Zhao Yan, Fan Linhao, Wang Huizhi, Hou Zhongjun, Huo Sen, Brandon Nigel P, Yin Yan, Guiver Michael D. Nature. 2021;595(7867):361–369. doi: 10.1038/s41586-021-03482-7. [DOI] [PubMed] [Google Scholar]
  • 57.Vertical MoS2 transistors with sub-1-nm gate lengths. Wu Fan, Tian He, Shen Yang, Hou Zhan, Ren Jie, Gou Guangyang, Sun Yabin, Yang Yi, Ren Tian-Ling. Nature. 2022;603(7900):259–264. doi: 10.1038/s41586-021-04323-3. [DOI] [PubMed] [Google Scholar]
  • 58.BCG: a vaccine with multiple faces. Yamazaki-Nakashimada Marco Antonio, Unzueta Alberto, Berenise Gámez-González Luisa, González-Saldaña Napoleón, Sorensen Ricardo U. Human vaccines & immunotherapeutics. 2020;16(8):1841–1850. doi: 10.1080/21645515.2019.1706930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Prevention of transmission of leprosy: The current scenario. Palit Aparna, Kar Hemanta Kumar. Indian journal of dermatology, venereology and leprology. 2020;86(2):115–123. doi: 10.4103/ijdvl.IJDVL_326_19. [DOI] [PubMed] [Google Scholar]
  • 60.Systematic review of M. Bovis BCG and other candidate vaccines for Buruli ulcer prophylaxis. Muhi Stephen, Stinear Timothy P. Vaccine. 2021;39(50):7238–7252. doi: 10.1016/j.vaccine.2021.05.092. [DOI] [PubMed] [Google Scholar]
  • 61.Vaccination efforts for Buruli ulcer. Chavda Vivek P, Haritopoulou-Sinanidou Melina, Bezbaruah Rajashri, Apostolopoulos Vasso. Expert review of vaccines. 2022;21(10):1419–1428. doi: 10.1080/14760584.2022.2113514. [DOI] [PubMed] [Google Scholar]
  • 62.Mycobacterium ulcerans Mouse Model Refinement for Pre-Clinical Profiling of Vaccine Candidates. Bénard Angèle, Sala Claudia, Pluschke Gerd. PloS one. 2016;11(11):e0167059. doi: 10.1371/journal.pone.0167059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Structural basis and designing of peptide vaccine using PE-PGRS family protein of Mycobacterium ulcerans-An integrated vaccinomics approach. Nain Zulkar, Karim Mohammad Minnatul, Sen Monokesh Kumer, Adhikari Utpal Kumar. Molecular immunology. 2020;120:146–163. doi: 10.1016/j.molimm.2020.02.009. [DOI] [PubMed] [Google Scholar]
  • 64.The search for a Buruli Ulcer vaccine and the effectiveness of the Bacillus Calmette-Guérin vaccine. Ishwarlall Tamara Z, Okpeku Moses, Adeniyi Adebayo A, Adeleke Matthew A. Acta tropica. 2022;228:106323. doi: 10.1016/j.actatropica.2022.106323. [DOI] [PubMed] [Google Scholar]
  • 65.Chlamydia trachomatis: quest for an eye-opening vaccine breakthrough. Chavda Vivek P, Pandya Anjali, Kypreos Erica, Patravale Vandana, Apostolopoulos Vasso. Expert review of vaccines. 2022;21(6):771–781. doi: 10.1080/14760584.2022.2061461. [DOI] [PubMed] [Google Scholar]
  • 66.Corrigendum: Molecular pathogenesis of Chlamydia trachomatis. Jury Brittany, Fleming Charlotte, Huston Wilhelmina M, Luu Laurence Don Wai. Frontiers in cellular and infection microbiology. 2023;13:1358553. doi: 10.3389/fcimb.2023.1281823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chlamydia trachomatis: Cell biology, immunology and vaccination. Murray Sam M, McKay Paul F. Vaccine. 2021;39(22):2965–2975. doi: 10.1016/j.vaccine.2021.03.043. [DOI] [PubMed] [Google Scholar]
  • 68.Status of vaccine research and development of vaccines for Chlamydia trachomatis infection. Poston Taylor B, Gottlieb Sami L, Darville Toni. Vaccine. 2019;37(50):7289–7294. doi: 10.1016/j.vaccine.2017.01.023. [DOI] [PubMed] [Google Scholar]
  • 69.Safety and immunogenicity of the chlamydia vaccine candidate CTH522 adjuvanted with CAF01 liposomes or aluminium hydroxide: a first-in-human, randomised, double-blind, placebo-controlled, phase 1 trial. Abraham Sonya, Juel Helene B, Bang Peter, Cheeseman Hannah M, Dohn Rebecca B, Cole Tom, Kristiansen Max P, Korsholm Karen S, Lewis David, Olsen Anja W, McFarlane Leon R, Day Suzanne, Knudsen Sara, Moen Kjersti, Ruhwald Morten, Kromann Ingrid, Andersen Peter, Shattock Robin J, Follmann Frank. The Lancet. Infectious diseases. 2019;19(10):1091–1100. doi: 10.1016/S1473-3099(19)30279-8. [DOI] [PubMed] [Google Scholar]
  • 70.B-Cell Epitope Mapping of TprC and TprD Variants of Treponema pallidum Subspecies Informs Vaccine Development for Human Treponematoses. Molini Barbara, Fernandez Mark C, Godornes Charmie, Vorobieva Anastassia, Lukehart Sheila A, Giacani Lorenzo. Frontiers in immunology. 2022;13:862491. doi: 10.3389/fimmu.2022.862491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Transcriptional and immunological analysis of the putative outer membrane protein and vaccine candidate TprL of Treponema pallidum. Haynes Austin M, Fernandez Mark, Romeis Emily, Mitjà Oriol, Konda Kelika A, Vargas Silver K, Eguiluz Maria, Caceres Carlos F, Klausner Jeffrey D, Giacani Lorenzo. PLoS neglected tropical diseases. 2021;15(1):e0008812. doi: 10.1371/journal.pntd.0008812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fungal Vaccine Development: State of the Art and Perspectives Using Immunoinformatics. Inácio Moisés Morais, Moreira André Luís Elias, Cruz-Leite Vanessa Rafaela Milhomem, Mattos Karine, Silva Lana O'Hara Souza, Venturini James, Ruiz Orville Hernandez, Ribeiro-Dias Fátima, Weber Simone Schneider, Soares Célia Maria de Almeida, Borges Clayton Luiz. Journal of fungi (Basel, Switzerland) 2023;9(6) doi: 10.3390/jof9060633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Vaccines against Ebola virus and Marburg virus: recent advances and promising candidates. Suschak John J, Schmaljohn Connie S. Human vaccines & immunotherapeutics. 2019;15(10):2359–2377. doi: 10.1080/21645515.2019.1651140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.WHO's Blueprint List of Priority Diseases. Friedrich M J. JAMA. 2018;319(19):1973. doi: 10.1001/jama.2018.5712. [DOI] [PubMed] [Google Scholar]
  • 75.A review of Lassa fever vaccine candidates. Salami Kolawole, Gouglas Dimitrios, Schmaljohn Connie, Saville Melanie, Tornieporth Nadia. Current opinion in virology. 2019;37:105–111. doi: 10.1016/j.coviro.2019.07.006. [DOI] [PubMed] [Google Scholar]
  • 76.A replication defective recombinant Ad5 vaccine expressing Ebola virus GP is safe and immunogenic in healthy adults. Ledgerwood J E, Costner P, Desai N, Holman L, Enama M E, Yamshchikov G, Mulangu S, Hu Z, Andrews C A, Sheets R A, Koup R A, Roederer M, Bailer R, Mascola J R, Pau M G, Sullivan N J, Goudsmit J, Nabel G J, Graham B S. Vaccine. 2010;29(2):304–13. doi: 10.1016/j.vaccine.2010.10.037. [DOI] [PubMed] [Google Scholar]
  • 77.Safety and Immunogenicity of a Heterologous Prime-Boost Ebola Virus Vaccine Regimen in Healthy Adults in the United Kingdom and Senegal. Venkatraman Navin, Ndiaye Birahim Pierre, Bowyer Georgina, Wade Djibril, Sridhar Saranya, Wright Daniel, Powlson Jonathan, Ndiaye Ibrahima, Dièye Siry, Thompson Craig, Bakhoum Momar, Morter Richard, Capone Stefania, Del Sorbo Mariarosaria, Jamieson Sophie, Rampling Tommy, Datoo Mehreen, Roberts Rachel, Poulton Ian, Griffiths Oliver, Ballou W Ripley, Roman François, Lewis David J M, Lawrie Alison, Imoukhuede Egeruan, Gilbert Sarah C, Dieye Tandakha N, Ewer Katie J, Mboup Souleymane, Hill Adrian V S. The Journal of infectious diseases. 2019;219(8):1187–1197. doi: 10.1093/infdis/jiy639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.A DNA vaccine for Ebola virus is safe and immunogenic in a phase I clinical trial. Martin Julie E, Sullivan Nancy J, Enama Mary E, Gordon Ingelise J, Roederer Mario, Koup Richard A, Bailer Robert T, Chakrabarti Bimal K, Bailey Michael A, Gomez Phillip L, Andrews Charla A, Moodie Zoe, Gu Lin, Stein Judith A, Nabel Gary J, Graham Barney S. Clinical and vaccine immunology : CVI. 2006;13(11):1267–77. doi: 10.1128/CVI.00162-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Henao-Restrepo Ana Maria, Camacho Anton, Longini Ira M, Watson Conall H, Edmunds W John, Egger Matthias, Carroll Miles W, Dean Natalie E, Diatta Ibrahima, Doumbia Moussa, Draguez Bertrand, Duraffour Sophie, Enwere Godwin, Grais Rebecca, Gunther Stephan, Gsell Pierre-Stéphane, Hossmann Stefanie, Watle Sara Viksmoen, Kondé Mandy Kader, Kéïta Sakoba, Kone Souleymane, Kuisma Eewa, Levine Myron M, Mandal Sema, Mauget Thomas, Norheim Gunnstein, Riveros Ximena, Soumah Aboubacar, Trelle Sven, Vicari Andrea S, Røttingen John-Arne, Kieny Marie-Paule. Lancet (London, England) 2017;389(10068):505–518. doi: 10.1016/S0140-6736(16)32621-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Fathi Anahita, Dahlke Christine, Addo Marylyn M. Human vaccines & immunotherapeutics. 2019;15(10):2269–2285. doi: 10.1080/21645515.2019.1649532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ervebo (Ebola Zaire Vaccine, Live/rVSVΔG-ZEBOV-GP): The First Licensed Vaccine for the Prevention of Ebola Virus Disease. Piszczatoski Christopher R., Gums John G. Journal of Pharmacy Technology. 2020;36(6):243-250. [Google Scholar]
  • 82.The complexity and cost of vaccine manufacturing - An overview. Plotkin Stanley, Robinson James M, Cunningham Gerard, Iqbal Robyn, Larsen Shannon. Vaccine. 2017;35(33):4064–4071. doi: 10.1016/j.vaccine.2017.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Vaccine platforms for the prevention of Lassa fever. Purushotham Jyothi, Lambe Teresa, Gilbert Sarah C. Immunology letters. 2019;215:1–11. doi: 10.1016/j.imlet.2019.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.WHO Target Product Profile for Lassa virus Vaccine. Geneva: World Health Organization. https://www.who.int/publications/m/item/who-target-product-profile-for-lassa-virus-vaccine https://www.who.int/publications/m/item/who-target-product-profile-for-lassa-virus-vaccine
  • 85.Vaccine platforms to control Lassa fever. Lukashevich Igor S, Pushko Peter. Expert review of vaccines. 2016;15(9):1135–50. doi: 10.1080/14760584.2016.1184575. [DOI] [PubMed] [Google Scholar]
  • 86.Advanced vaccine candidates for Lassa fever. Lukashevich Igor S. Viruses. 2012;4(11):2514–57. doi: 10.3390/v4112514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.JMM Profile: Rift Valley fever: a zoonotic viral haemorrhagic disease. Lean Fabian Z X, Johnson Nicholas. Journal of medical microbiology. 2022;72(12) doi: 10.1099/jmm.0.001619. [DOI] [PubMed] [Google Scholar]
  • 88.An Overview of Rift Valley Fever Vaccine Development Strategies. Kitandwe Paul Kato, McKay Paul F, Kaleebu Pontiano, Shattock Robin J. Vaccines. 2022;10(11) doi: 10.3390/vaccines10111794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rift Valley fever MP-12 vaccine Phase 2 clinical trial: Safety, immunogenicity, and genetic characterization of virus isolates. Pittman Phillip R, Norris Sarah L, Brown Elizabeth S, Ranadive Manmohan V, Schibly Barbara A, Bettinger George E, Lokugamage Nandadeva, Korman Lawrence, Morrill John C, Peters Clarence J. Vaccine. 2016;34(4):523–530. doi: 10.1016/j.vaccine.2015.11.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Immunogenicity and safety of an inactivated Rift Valley fever vaccine in a 19-year study. Rusnak Janice M, Gibbs Paul, Boudreau Ellen, Clizbe Denise P, Pittman Phillip. Vaccine. 2011;29(17):3222–9. doi: 10.1016/j.vaccine.2011.02.037. [DOI] [PubMed] [Google Scholar]
  • 91.Teratogenicity of a mutagenised Rift Valley fever virus (MVP 12) in sheep. Hunter P, Erasmus B J, Vorster J H. The Onderstepoort journal of veterinary research. 2002;69(1):95–8. [PubMed] [Google Scholar]
  • 92.Safety and efficacy of ChAdOx1 RVF vaccine against Rift Valley fever in pregnant sheep and goats. Stedman Anna, Wright Daniel, Wichgers Schreur Paul J, Clark Madeleine H A, Hill Adrian V S, Gilbert Sarah C, Francis Michael J, van Keulen Lucien, Kortekaas Jeroen, Charleston Bryan, Warimwe George M. NPJ vaccines. 2019;4:44. doi: 10.1038/s41541-019-0138-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Use of Single-Injection Recombinant Vesicular Stomatitis Virus Vaccine to Protect Nonhuman Primates Against Lethal Nipah Virus Disease. Mire Chad E, Geisbert Joan B, Agans Krystle N, Versteeg Krista M, Deer Daniel J, Satterfield Benjamin A, Fenton Karla A, Geisbert Thomas W. Emerging infectious diseases. 2019;25(6):1144–1152. doi: 10.3201/eid2506.181620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Safety, tolerability, pharmacokinetics, and immunogenicity of a human monoclonal antibody targeting the G glycoprotein of henipaviruses in healthy adults: a first-in-human, randomised, controlled, phase 1 study. Playford Elliott Geoffrey, Munro Trent, Mahler Stephen M, Elliott Suzanne, Gerometta Michael, Hoger Kym L, Jones Martina L, Griffin Paul, Lynch Kathleen D, Carroll Heidi, El Saadi Debra, Gilmour Margaret E, Hughes Benjamin, Hughes Karen, Huang Edwin, de Bakker Christopher, Klein Reuben, Scher Mark G, Smith Ina L, Wang Lin-Fa, Lambert Stephen B, Dimitrov Dimiter S, Gray Peter P, Broder Christopher C. The Lancet. Infectious diseases. 2020;20(4):445–454. doi: 10.1016/S1473-3099(19)30634-6. [DOI] [PubMed] [Google Scholar]
  • 95.Vaccines to Emerging Viruses: Nipah and Hendra. Amaya Moushimi, Broder Christopher C. Annual review of virology. 2020;7(1):447–473. doi: 10.1146/annurev-virology-021920-113833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Advances in diagnostics, vaccines and therapeutics for Nipah virus. Thakur Nazia, Bailey Dalan. Microbes and infection. 2019;21(7):278–286. doi: 10.1016/j.micinf.2019.02.002. [DOI] [PubMed] [Google Scholar]
  • 97.Epidemiological Aspects of Crimean-Congo Hemorrhagic Fever in Western Europe: What about the Future? Portillo Aránzazu, Palomar Ana M, Santibáñez Paula, Oteo José A. Microorganisms. 2021;9(3) doi: 10.3390/microorganisms9030649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pathogenesis and immune response of Crimean-Congo hemorrhagic fever virus in a STAT-1 knockout mouse model. Bente Dennis A, Alimonti Judie B, Shieh Wun-Ju, Camus Gaëlle, Ströher Ute, Zaki Sherif, Jones Steven M. Journal of virology. 2010;84(21):11089–100. doi: 10.1128/JVI.01383-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Crimean-Congo Hemorrhagic Fever Virus for Clinicians-Diagnosis, Clinical Management, and Therapeutics. Frank Maria G, Weaver Gretchen, Raabe Vanessa. Emerging infectious diseases. 2024;30(5):864–873. doi: 10.3201/eid3005.231648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.A cynomolgus macaque model for Crimean-Congo haemorrhagic fever. Haddock Elaine, Feldmann Friederike, Hawman David W, Zivcec Marko, Hanley Patrick W, Saturday Greg, Scott Dana P, Thomas Tina, Korva Miša, Avšič-Županc Tatjana, Safronetz David, Feldmann Heinz. Nature microbiology. 2018;3(5):556–562. doi: 10.1038/s41564-018-0141-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Crimean-Congo Hemorrhagic Fever Virus: Progress in Vaccine Development. Ozdarendeli Aykut. Diagnostics (Basel, Switzerland) 2023;13(16) doi: 10.3390/diagnostics13162708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.A DNA-based vaccine protects against Crimean-Congo haemorrhagic fever virus disease in a Cynomolgus macaque model. Hawman David W, Ahlén Gustaf, Appelberg K Sofia, Meade-White Kimberly, Hanley Patrick W, Scott Dana, Monteil Vanessa, Devignot Stephanie, Okumura Atsushi, Weber Friedemann, Feldmann Heinz, Sällberg Matti, Mirazimi Ali. Nature microbiology. 2021;6(2):187–195. doi: 10.1038/s41564-020-00815-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Healthy individuals' immune response to the Bulgarian Crimean-Congo hemorrhagic fever virus vaccine. Mousavi-Jazi Mehrdad, Karlberg Helen, Papa Anna, Christova Iva, Mirazimi Ali. Vaccine. 2012;30(44):6225–9. doi: 10.1016/j.vaccine.2012.08.003. [DOI] [PubMed] [Google Scholar]
  • 104.Healthcare Policies to Eliminate Neglected Tropical Diseases (NTDs) in India: A Roadmap. Chandra Ajay, Sreeganga S D, Rath Nibedita, Ramaprasad Arkalgud. International journal of environmental research and public health. 2023;20(19) doi: 10.3390/ijerph20196842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Tungiasis: a neglected epidermal parasitic skin disease of marginalized populations--a call for global science and policy. Karunamoorthi Kaliyaperumal. Parasitology research. 2013;112(10):3635–43. doi: 10.1007/s00436-013-3551-8. [DOI] [PubMed] [Google Scholar]
  • 106.Neglected tropical diseases: exploring long term practical approaches to achieve sustainable disease elimination and beyond. Ortu Giuseppina, Williams Oliver. Infectious diseases of poverty. 2017;6(1):147. doi: 10.1186/s40249-017-0361-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Improving neglected tropical disease services and integration into primary healthcare in Southern Nations, Nationalities and People’s Region (SNNPR), Ethiopia: results from a mixed methods intervention evaluation. Donovan Laura, Habte Tedila, Batisso Esey, Getachew Dawit, Stratil Ann-Sophie, Tekalegne Agonafer, Seife Fikre, Mariam Damen, Baker Kevin. 2023 doi: 10.1371/journal.pntd.0011718. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Discoveries are provided here courtesy of Applied Systems

RESOURCES