Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Article
Brief Report
Case Report
Commentary
Community Case Study
Editorial
Image
Images
Letter to Editor
Letter to the Editor
Media & News
Mini Review
Obituary
Original Article
Perspective
Review Article
Reviewers; List
Short Communication
Task Force Report
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Article
Brief Report
Case Report
Commentary
Community Case Study
Editorial
Image
Images
Letter to Editor
Letter to the Editor
Media & News
Mini Review
Obituary
Original Article
Perspective
Review Article
Reviewers; List
Short Communication
Task Force Report
View/Download PDF

Translate this page into:

Task Force Report
61 (
4
); 394-436
doi:
10.25259/ANAMS_TFR_15_2024

NAMS task force report on vector borne diseases

Former Director General – Indian Council of Medical Research
ICMR-National Institute of Malaria Research
Department of Emerging Infectious Diseases, Duke-NUS GMS, Singapore
University of Colombo, Sri Lanka
Department of Disease Control at the London School of Hygiene & Tropical Medicine
National Center for Vector Borne Diseases Control, India
ICGEB New Delhi, India
Department of Communicable Diseases at World Health Organization South-East Asia Region
AIPH University, Ex-Vice Chancellor, Central university of Tamil Nadu, India
ICMR-National Institute of Malaria Research, New Delhi, India
National Center for Vector Borne Diseases Control
ICMR-Rajendra Memorial Research Institute of Medical Sciences
Saveetha University Adjunct Professor, Georgetown University, Washington, USA
ICMR-National Institute of Malaria Research
Department of Virology, Karolinska Institutet, Stockholm, Sweden
Sir Ganga Ram Hospital, New Delhi, India
Department of Clinical Virology, Institute of Liver & Biliary Sciences, New Delhi, India
Freelancer
ICMR-National Institute of Malaria Research*
Report approved by DGHS & Ministry of Health and Family Welfare, Government of India.

*Corresponding author: Prof. Nirmal Kumar Ganguly, Sir Ganga Ram Hospital, Delhi, India. ganguly1nk@gmail.com

Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Ganguly NK, Anvikar AR, Gubler DJ, Mandis K, Cameron M, Dhariwal AC, et al. NAMS task force report on vector borne diseases. Ann Natl Acad Med Sci (India). 2025;61:394-436. doi: 10.25259/ANAMS_TFR_15_2024

Abstract

EXECUTIVE SUMMARY

Vector-borne diseases (VBDs) in India, which include viral, parasitic, and bacterial infections, present significant public health challenges. These diseases, transmitted by various vectors such as mosquitoes, ticks, and fleas, contribute to high morbidity and mortality rates, particularly in vulnerable populations. Despite existing control measures, these diseases continue to affect millions, driven by factors such as climate change, urbanization, and inadequate disease surveillance. necessitating urgent attention to fill the gaps in various surveillance, treatment, and prevention strategies.

There is a critical need for a comprehensive understanding of the epidemiology, transmission dynamics, vector behavior, and environmental factors influencing disease spread. The knowledge of vector biology and ecology, resistance to insecticides, and environmental adaptations are crucial for the development of targeted control strategies. Existing diagnostic tools for VBDs are often inadequate, particularly in resource-limited settings. There is a pressing need for rapid, cost-effective diagnostic methods and novel treatments to address drug resistance. Post-infection syndromes and chronic conditions associated with diseases like dengue and chikungunya contribute to the long-term health impacts of VBDs. There is an urgent need for the development and implementation of vaccines for diseases like dengue and malaria. Continued investment in vaccine research is essential to provide long-term solutions to VBDs along with current vector control measures. An integrated approach that combines biological control, environmental management, and community involvement is necessary to enhance the sustainability and effectiveness of control strategies. Enhancing public awareness about VBDs, their transmission, and preventive measures is vital. Education campaigns should target high-risk populations, focusing on behavioral changes to reduce vector exposure.

Promoting interdisciplinary research collaborations among academia, governmental, and Non-Governmental Organizations (NGO) can lead to more holistic approaches in addressing VBDs and strengthening the integration of research findings into public health policy and practice to ensure that evidence-based strategies are implemented at all levels. Establishing sustainable funding mechanisms for research and intervention programs is crucial to maintaining momentum in the fight against VBDs.

In summary, addressing the research gaps, enhancing surveillance, and improving treatment and prevention strategies for VBDs in India are imperative for mitigating their impact. By prioritizing these areas, India can develop a more effective and sustainable response to the growing burden of VBDs, ultimately improving public health outcomes including and improving the quality of life.

Keywords

Vector borne diseases
India
Dengue
Chiungunia
Zika
Japanese encephaltis
Kyasanur forest disease
Chandipura virus
Malaria
Leishmaniasis
Lymphatic filariasis

Introduction

Emerging and re-emerging vector borne disease (VBDs) are among the major public health concerns across the world. They are not directly transmissible among humans in most cases but are mainly spread under suitable conditions as a result of the interaction of vectors, hosts, climate/environment, pathogens, and vulnerable human populations. Their impact has a significant toll on economies and restricts both rural and urban development.

The burdens of several climate-sensitive VBDs are projected to increase due to climate change unless additional adaptation efforts are undertaken by countries. Many infectious agents, vectors, non-human reservoir hosts, and pathogen replication rates can be sensitive to ambient climatic conditions. Elevated proliferation and reproduction rates at higher temperatures, longer transmission seasons, changes in ecology, and climate-related migration of vectors, reservoir hosts, or human populations contribute to this climate sensitivity.1,2

India is vulnerable to VBDs due to its eco-sociodemographic characteristics, diverse climate, and geographic conditions.3 The diverse climatic and geographical conditions create ideal environments for the breeding of various vectors, leading to a significant public health challenge. Malaria continues to be a persistent health issue in several states, while dengue and chikungunya outbreaks are becoming increasingly frequent in urban areas due to the distinct habitats of different vectors.

There is growing evidence that vector capacity has increased for dengue fever, malaria and other mosquito-borne diseases and that higher global average temperatures are making wider geographic areas more suitable for transmission. Transmission rates of malaria are directly influenced by climatic and weather variables such as temperature, with non-climatic socioeconomic factors and health system responses counteracting climatic drivers.

The National Center for Vector Borne Disease Control (NCVBDC) has implemented various control measures to combat VBDs, including integrated vector management, surveillance, and public health education. Interventions such as long-lasting insecticidal nets (LLINS), indoor residual spraying, and vaccination campaigns (e.g., for Japanese encephalitis) have been employed to reduce disease transmission and control VBDs.

VBDs in India can be classified based on the type of causal organism (pathogen) responsible for the disease:

Viral infections

  • Dengue: It is caused by the dengue virus (DENV) and transmitted by Aedes mosquitoes. It is one of the fast-emerging, outbreak-prone, and mosquito-borne viral fevers. In 2023, the reported morbidity and mortality due to dengue were 289235 and 485 respectively. The states with a high number of dengue cases were Bihar, Karnataka, Kerala, Maharashtra, Odisha, Punjab, Rajasthan, Uttar Pradesh, West Bengal, and Delhi.4

  • Chikungunya: Chikungunya, also known as chikungunya virus disease or chikungunya fever, is caused by the chikungunya virus (CHIKV). It is a debilitating but non-fatal viral illness that is spread by the bite of infected Aedes mosquitoes. The number of suspected cases and confirmed cases in 2023 were 200064 and 11477, respectively.5

  • Zika: Zika is a non-fatal but concerning disease due to its association with microcephaly, or reduced head size, in babies born to pregnant women who are infected. It is caused by the Zika virus, and it is also transmitted by Aedes mosquitoes. It has been reported in several Indian states since 2016, including Gujarat, Tamil Nadu, Madhya Pradesh, Rajasthan, Kerala, Maharashtra, Uttar Pradesh, Delhi, and Karnataka.6

  • Japanese encephalitis (JE): JE is zoonotic viral disease which is caused by the JE virus. JE is endemic in a few areas of India and transmitted by the Vishnui group of Culex mosquitos. The case fatality rate (CFR) of this disease is high, and those who survive may suffer with various degrees of neurological sequelae. JE continues to pose a significant public health problem in India. Since 2023, JE and acute encephalitis syndrome (AES) have been reported in 355 districts across 24 states/union territories (UTs) in India.

  • Kyasanur forest disease (KFD): KFD is a tick-borne hemorrhagic fever caused by the kyasanur forest disease virus (KFDV). KFD is transmitted by hard ticks, Haemaphysalis spinigera.

  • Chandipura virus (CHPV): CHPV is known to cause sporadic cases and outbreaks of AES in western, central, and southern India, particularly during the monsoon season. It is transmitted by vectors such as sandflies, mosquitoes, and ticks. Phlebotomus papatasi, a species of sandfly, has been identified as the vector of CHPV in Gujarat. The disease can result in high-CFRs, ranging from 56% to 75%. It primarily affects children under the age of 15 and typically presents as a febrile illness that may progress to severe cases or death.

Protozoan infections

  • Malaria: Malaria is a public health problem in several parts of the country. About 95% of the country’s population resides in malaria-endemic areas, and 80% of malaria reported in the country is confined to areas consisting of 20% of population. It is caused by the Plasmodium parasite, namely Plasmodium viviax (P. vivax), Plasmodium falciparum (P. falciparum), Plasmodium malariae (P. malariae), and Plasmodium ovale (P. ovale) and transmitted by Anopheles mosquitoes elation residing in tribal, hilly, difficult, and inaccessible areas. In 2023, India reported approximately 227564 cases of malaria which led to 83 deaths.7,8

  • Leishmaniasis: It is caused by Leishmania species, and sandflies are the vector for human transmission. Kala-azar is a slow-progressing indigenous disease caused by a protozoan parasite of the genus Leishmania. In India Leishmania donovani is the only parasite causing this disease. In 2023, 520 cases and 4 deaths were reported in India due to Kala-azar.9

Helminthic infections

  • Lymphatic filariasis (elephantiasis): Filariasis has been a major public health problem in India next only to malaria. The disease is caused by the nematode worm, either Wuchereria bancrofti or Brugia malayi, and transmitted by ubiquitous mosquito species Culex quinquefasciatus and Mansonia annulifera/M.uniformis, respectively. In 2023, 6.19 lakh lymphedema and 1.26 lakh hydrocele cases were reported.10 Filariasis cases have been recorded from Andhra Pradesh, Assam, Bihar, Chhattisgarh, Goa, Jharkhand, Karnataka, Gujarat, Kerala, Madhya Pradesh, Maharashtra, Orissa, Tamil Nadu, Uttar Pradesh, West Bengal, Pondicherry, Andaman & Nicobar Islands, Daman & Diu, Dadra & Nagar Haveli, and Lakshadweep.

India is one of the mosquito biogeographic hotspots in the world. In terms of mosquito biodiversity, India comes fifth, behind Brazil, Indonesia, Malaysia, and Thailand. Since there is no reliable collection of data on the field, it is exceedingly impossible to estimate the number of mosquito species and subspecies that exist in India. Most commonly found mosquito species in India include– Anopheles, Aedes, Culex, and Mansonia.11

VECTOR-BORNE VIRAL DISEASES IN INDIA

Background

Over the past decade, the landscape of vector-borne viral infections in India has undergone dynamic changes influenced by climatic shifts, increasing urbanization, and the heightened global movement facilitated by international tourism. Arthropod-borne viruses (arboviruses), primarily ribonucleic acid (RNA) viruses, predominantly belonging to the Flaviviridae family, have emerged or re-emerged as significant health problems. These infections, including dengue virus infection, JE, and chikungunya, exhibit a diverse range of symptoms, from mild influenza-like manifestations to severe clinical conditions, such as hemorrhagic fevers and encephalitis, resulting in considerable morbidity and mortality. India, contending with the widespread presence of vector reservoirs for these infections, faces an annual burden of significant morbidity.12 Challenges persist in early diagnosis, effective management, awareness promotion, and implementation of preventive measures such as vaccination and vector control. This white paper addresses these challenges comprehensively, proposing a focused examination of the existing problems associated with each viral infection.13,14 The paper emphasizes the urgency for an integrated approach, incorporating research and innovation, climate resilience, and international collaboration, to effectively combat the expanding geographical range and impact of vector-borne viral diseases in India.

1. Dengue

DENV is an RNA virus transmitted by Aedes aegypti (A. aegypti) and Aedes albopictus (A. albopictus) families of mosquitoes and causes dengue. There exist four serotypes of the virus (DENV 1-4), each comprised of various genotypes.15 The disease, with its varying degrees of clinical presentation, poses a significant and recurrent health threat in India.

Epidemiology

Over the last 20 years, the global incidence of dengue fever has significantly risen, spreading across 129 countries. It is endemic in over 100 nations within the World Health Organization (WHO), including regions of Africa, the Americas, the Eastern Mediterranean, South-East Asia, and the Western Pacific, leading to an estimated 100–400 million infections annually.16 The surge in dengue cases has been striking, increasing eightfold from year 2000 (505,430 cases) to year 2019 (5.2 million cases).17 India is a major contributor to the global dengue burden, grappling with a high prevalence of the disease. The country harbors multiple co-circulating serotypes as well as many genotypes.15,18-20 Dengue has become hyper-endemic in the country.20,21 According to the report by NCVBDC, as of September 17, 2023, 94,198 cases of dengue infection were reported in India, with 91 deaths.22 Kerala reported the highest number of cases (9770), more than double compared to the numbers seen in the last five years.22,23 The escalation of dengue cases can be attributed to various factors such as enhanced global trade, increased travel, rapid urbanization, population growth, and climate variability. These elements create favorable conditions for the multiplication of Aedes mosquitoes as well as the survival of the virus.24,25 Periodic outbreaks are now common in India, especially during the monsoon and post-monsoon seasons.24,26

Clinical symptoms

Clinical signs and symptoms of dengue infection are usually asymptomatic or range from mild fever to severe bleeding, shock, and organ dysfunction. The incubation period varies from 4-6 (range 3-14) days. There are three clinical phases of dengue infection. The febrile phase is characterized by a high-grade fever (>38.5°C), which may be biphasic. This phase usually lasts for 2-7 days and may be associated with non-specific symptoms like vomiting, headache, myalgia, arthralgia, and maculopapular rash. Depending upon the severity of the disease, bleeding manifestations may occur during this phase. A few infected patients (5-10%) may progress to the critical phase. This is commonly reported among those with a positive previous history of dengue infection. This phase may last for 24-48 hours and is characterized by vasculopathy and coagulopathy, leading to bleeding, shock, and organ dysfunction. In the end, there is a convalescent-phase where the patient starts recovering. This usually lasts for 2-3 days. The clinical presentation in this phase is influenced by factors such as the infected person’s immunity, previous dengue infection, and the viral genotype. This highlights the importance of accurate categorization of patients into different phases of infection for further medical intervention. Symptomatic dengue infection could be either dengue without warning signs (mild dengue) or dengue with warning signs (moderate and severe dengue). Mild dengue is characterized by mild fever, though some individuals may remain asymptomatic. Moderate dengue encompasses additional symptoms such as abdominal tenderness, hepatomegaly, persistent vomiting, pleural effusion, ascites, positive tourniquet test, hypovolemia, hypoproteinemia, and signs of shock. Severe dengue, the most critical form, is marked by significant bleeding, profound shock, severe metabolic disorder, and organ involvement, including hepatic impairment and central nervous system (CNS) manifestations.15

Diagnosis and management

Accurate diagnostic techniques are pivotal in effectively identifying the infection, aiding in early detection, confirming cases, and distinguishing it from other infections. At present, various laboratory-based methods such as serological detection of non-structural protein 1 (NS1) Antigen, IgM antibodies, and nucleic acid amplification test are available.27,28 The tourniquet test procedure has also been employed as a clinical method for dengue infection, though its utility is debatable.29 Conventional diagnosis of active dengue infection is mostly done by laboratory-based tests. For the identification of infection during the acute phase (from day 1 of fever to 9th day of infection), NS1 antigen detection is most commonly used. IgM antibody-based assays are also commonly utilized but only after five days of fever. Sometimes, IgM antibodies might persist beyond 1 to 2 months of infection, therefore, antigen-based assays are more specific for the identification of acute ongoing infection. Polymerase chain reaction (PCR) based assays are used only for epidemiological purposes or for serotyping/genotyping and should be done during the acute phase of infection, which is less than five days of fever. Many commercial enzyme-linked immunosorbent assays (ELISA), as well as rapid card-based assays for both NS1 antigen as well as IgM antibody detection, are available in the market with varying sensitivity and specificity30,31. In fact, there is limited availability of good rapid card tests for the combined detection of NS1 antigen and IgM antibody, which are needed for the rapid diagnosis and decentralization of testing for dengue infections in public health settings.32-34 IgG antibody-based assays are no longer used for the diagnosis of infections. Clinical management of dengue is mostly supportive care, such as fluid replacement and the use of analgesics, with few requiring hospitalization in case of severe dengue infection to prevent further complications.35,36 Till date, there are no clinically approved antivirals present for the treatment of dengue infection.37 A variety of antiviral drug candidates have been tried in the past, but most of the studies were either in-silico or in-vitro analyses; in-vivo studies using animal models are a major limitation in the development of any suitable candidate with good antiviral properties for dengue infection.38 Several herbal preparations in the recent years have also been tried for the treatment of dengue infection, but none has been approved so far.38,39

Preventive measures

In the absence of specific antiviral treatment, prevention is the only recourse to control this infection. Dengvaxia [chimeric yellow fever/dengue tetravalent dengue], a live attenuated vaccine (LAV), is the sole licensed vaccine at present available for dengue, but it is recommended for individuals with prior exposure to dengue infection.40,41 Encouragingly, there are ongoing clinical trials for several new candidates for the dengue vaccine. Among these, QDenga (TAK-003), which is also an LAV vaccine candidate has shown encouraging results in both safety and efficacy against symptomatic dengue.42 It is the second vaccine to get pre-qualified by WHO (https://www.who.int/news/item/15-05-2024-who-prequalifies-new-dengue-vaccine). Three leading Indian vaccine producers — Serum Institute, Panacea Biotec, and Biological E—have obtained non-exclusive licenses for the clinical development and commercialization of TetraVax-DV, a dengue vaccine developed by the US National Institutes of Health.43 Furthermore, Sun Pharma, in partnership with the Indian Council for Genetic Engineering and Biotechnology, has developed and acquired a license for the DSV4 vaccine. DSV4, a synthetic subunit vaccine incorporating dengue virus antigens, has shown effectiveness in stimulating both humoral and cellular immunity.44

Bio-control measures

Various bio-control measures have been implemented to curb the proliferation of Aedes mosquitoes in India. An effective approach involves eradicating breeding sites of A. aegypti within human habitats.45 Additionally, the introduction of larvivorous fish has proven successful in controlling the mosquito population by consuming mosquito larvae.45 Biological control strategies such as herbal remedies and the release of sterile male Aedes have also been deployed.46 India has also actively pursued the implementation of Wolbachia as a biocontrol strategy to manage mosquito breeding.47 The efficacy of Wolbachia release programs depends on the adaptability of Wolbachia strains in their natural environment. The Indian Council for Medical Research (ICMR) has been pivotal in this effort, establishing two colonies of A. aegypti mosquitoes infected with the wMel and wAlbB Wolbachia strains. These colonies, designed to assess the impact of heat stress on Wolbachia, have shown promising outcomes.48 To refine our understanding of the most efficient method for mosquito control as per our countries’ weather conditions, additional field studies are needed.

Challenges

India faces significant challenges in combating DENV infections, with the absence of specific antiviral treatments or an approved vaccine despite the disease’s growing status as a critical public health problem.26 Moreover, there’s a struggle in utilizing the approved dengue vaccine, Dengvaxia, in the country. One major hurdle lies in the dynamic nature of dengue serotypes in India. Since 2000, all four dengue serotypes (DENV-1, DENV-2, DENV-3, and DENV-4) have been co-circulating, causing cyclical outbreaks every 3-4 years.20,49 The circulating serotypes keep changing yearly and from region to region across different states in the country.20 Before 2012, DENV-1 and DENV-3 were dominant, but after 2012, DENV-2 became the most prevalent serotype in most of the regions, but southern states in India saw the emergence of the DENV-4 serotype.49 Few studies from North India highlighted DENV-2 as the most prevalent serotype in circulation, followed by DENV-3 and DENV-1, with DENV-4 being detected sporadically.50-52

The currently available vaccine against dengue infection, Dengvaxia exhibited a skewed response towards protection against DENV-2 and DENV-4 serotypes, and this limited its use to subjects with prior exposure to dengue infection.53,54 Moreover, information about its role in preventing severe outcomes like hemorrhagic manifestation was lacking.43 Therefore, the current immunization program in the country does not support the implementation of the dengue vaccine. More research and extensive efforts should be done for the development of tailor-made vaccines suitable for the epidemiological needs of our country.

Additionally, the number of underdiagnosed cases contributes to the underestimated burden of DENV infection. Most of the studies are hospital-based assessments of infection. Therefore, large-scale multicentre studies combining prevalence, clinical spectra, and follow-ups should be executed to find out the actual state-wise disease burden. There is a need to initiate community-based cohort studies representing different geographic regions as well as different age groups to generate dengue seroprevalence data in the country. Challenges of dengue control in India involve periodic outbreaks and poor vector control.26 These challenges have significant socioeconomic impacts during each outbreak, demanding efficient disease surveillance, early diagnosis, and accurate management to mitigate the illness’s severity and clinical consequences.55,56 Therefore, effective disease surveillance, early detection, and proper management are critical to curb dengue’s impact in India.

2. Chikungunya

Chikungunya is an arboviral disease caused by the CHIKV. It has been identified as a priority pathogen by the coalition for epidemics preparedness innovations.57 The CHIKV belongs to the Alphavirus genus in the Togaviridae family. Its name is derived from the kimakonde language, signifying “to become contorted” or “bent over in pain”.58 This viral illness is primarily transmitted to humans through the bite of infected A. aegypti and A. albopictus mosquitoes, which are also capable of transmitting Dengue and Zika viruses, are primarily active during daylight hours and tend to lay eggs in stagnant water. Both species prefer outdoor feeding, but A. aegypti is known to feed indoors as well.40,58

Epidemiology

The CHIKV, initially identified in Tanzania during the 1950s, is endemic in various global regions. It’s categorized into three main genotypes: West African, East Central South African (ECSA), and Asian. The virus is predominantly found in tropical and subtropical areas, notably in Africa, Southeast Asia, the Indian subcontinent, and the Pacific Region.59 The ECSA genotype of CHIKV migrated from Africa to the Indian Ocean islands in 2004, causing a substantial epidemic.59 Similarly, the Asian genotype has been prevalent in several Asian countries and the ECSA genotype triggered a widespread outbreak in Malaysia in 2008.60,61 As of November 30, 2023, approximately 460,000 CHIKV cases and over 360 deaths have been reported globally.62 CHIKV garnered attention due to its swift global spread. In India, CHIKV outbreaks are traced back to the early 20th century, with the first reported outbreak in Kolkata (Calcutta), West Bengal, in 1963.63 After a long hiatus, CHIKV re-emerged in India in December 2005, affecting nearly 1.4 million people across 13 states, leading to significant economic losses.64 The infection transcended state borders, reaching 28 out of 29 suspected states by 2018.65 In 2019, although the number of affected states with CHIKV infection declined to 25, suspected cases increased from 57,813 to 81,914, and confirmed cases rose from the year 2018 (9756) to 2019 (12,205).65 As per the NCVBDC published data in 2023 (till September 17), there were 93,455 suspected cases spread across 28 states, with only 3,711 confirmed cases in 25 states. The states with the highest number of confirmed cases were Karnataka, Maharashtra, and Tamil Nadu. Additionally, Andhra Pradesh, Gujarat, Haryana, Jharkhand, Punjab, Rajasthan, and Telangana reported a significant number of confirmed cases.65

Clinical symptoms

Most common clinical features of CHIKV infection include fever with joint pain. Other symptoms are rash, headache, and muscle pain. Some individuals may experience debilitating joint pain that persists for months or even years. Treatment includes rest, fluids, and over-the-counter medications for pain and fever. Sometimes it can also cause severe disease and prolonged health problems, particularly in elderly patients and in patients with underlying medical conditions. India faces the challenge of overlapping seasons for chikungunya, dengue, and malaria, all of which are now endemic. Chikungunya and dengue share a common vector and exhibit similar clinical presentations. Therefore, differential diagnosis by clinicians becomes crucial, relying on varied clinical presentations and laboratory methods to initiate appropriate treatment and prevent complications like hemorrhage, acute respiratory distress syndrome renal failure, etc.66

Diagnosis and management

Laboratory diagnosis for CHIKV involves serological methods such as CHIKV-specific IgM ELISA post five days of infection, and IgG antibody ELISA for sero-diagnosis using paired acute and convalescent phase serum samples. Cross-reactivity with other arboviruses affects the sensitivity of the serological assays.67,68 PCR-based techniques, specifically reverse transcription PCR (RT-PCR) and virus isolation by culture-based techniques, are primarily used for epidemiological or research purposes.69,70 After confirmation, symptomatic treatment remains the primary approach for management. Preventive methods include mosquito control measures. Despite the global availability of many vaccine candidates, that are in the pipeline, there are currently no ongoing trials for any of the vaccine in India.71,72

Preventive measures

Multiple CHIKV vaccines are in development; the ground-breaking approval of the Ixchiq vaccine by the US Food and Drug Administration in November 2023 marks a significant milestone in combating this disease. This vaccine is a LAV developed by a French specialty vaccine company Valneva, in Saint-Herblain. The intended use of this vaccine is for individuals aged 18 years and above and at higher risk of exposure. The vaccine targets all three genotypes of the CHIKV.71,73 Clinical trials have shown remarkable efficacy of this vaccine, with 98.9% of recipients meeting the zero response threshold in contrast to 0% in the placebo arm. This demonstrates a robust immune response triggered by the vaccine.74 This approval signifies a breakthrough in combating chikungunya, offering a promising preventive measures against this debilitating illness.

Challenges

Despite being self-limiting and associated with low mortality, CHIKV poses a challenge due to the painful illness course and long-term sequelae, which could impact the quality of life. New diagnostic and clinical protocols to differentiate CHIKV from other similar illnesses like dengue and Zika virus are the need of the hour. In addition, any adaptive changes in the viral genome that could impact several intervention and control measures should be investigated. The NCVBDC data reveals that Karnataka consistently leads in confirmed cases of Chikungunya, followed by Maharashtra and Punjab.65,75 A lack of specific case definitions to differentiate between co-existing arboviruses like dengue, Zika, etc., as well as a low proportion of lab-confirmed cases could contribute to either under- or over-reporting of cases. Well co-ordinated surveillance activities should be done for all the VBDs in the country. The absence of specific antiviral treatment and effective vaccination adds to the complexity of addressing the challenges of Chikungunya in the country.71 Although developing a CHIKV vaccine is technically less challenging than for dengue,72 factors such as genetic diversity, limited funding and resources, difficulties in performing vaccine efficacy studies due to unpredictable outbreak patterns, and a lack of long-term surveillance programs and corresponding data in Chikungunya-affected regions with sporadic transmission pose a significant challenge.57,73,75 These challenges emphasize the critical need to address the rising prevalence of Chikungunya in India by enhancing public awareness, strengthening healthcare infrastructure, and mitigating the economic impact of the disease.

3. Japanese encephalitis

Japanese encephalitis (JE) is a viral neurological disorder caused by the Japanese encephalitis virus (JEV) which is primarily a neurotropic arbovirus affecting the CNS. JEV belongs to the genus Flavivirus of the family Flaviviridae and is transmitted by the infective bites of a female mosquito belonging to Culex tritaeniorhynchus, Culex vishnui, or Culex pseudovishnui group. JE is the most important cause of AES.76

Within the ecosystem, the virus thrives in animals, particularly wild birds, specifically those within the Ardeidae family (such as cattle egrets, pond herons, etc.), and pigs, serving as natural hosts. Pigs and wild birds play a pivotal role in the transmission cycle as reservoirs of infection, often termed ‘amplifier hosts.’ In this cycle, humans and horses are considered the ‘dead-end hosts,’ indicating that they don’t produce sufficiently high virus levels to transmit the infection to feeding mosquitoes. It’s important to note that the virus does not induce any disease among its natural hosts.77

Epidemiology

JE traces its first isolation back to 1935, when it was identified in the brain of a person with fatal encephalitis in Japan.78 In India, the first case of JE occurred in 1955 in Vellore, Tamil Nadu, with a major outbreak in 1973 reported in the Burdwan district of West Bengal. Since then, JE/AES occurrences have been documented across 327 districts in 24 states.79 A substantial outbreak in the eastern UP in 2005 resulted in over 6,000 cases and 1,500 deaths.77 Globally, estimations of JE cases and fatalities are varying. WHO’s 2018 surveillance report, referring to Campbell’s 2011 document, suggested around 68,000 clinical cases yearly, leading to approximately 13,600 to 20,400 deaths.76,80 However, a 2023 Stat Pearls publication cited figures between 30,000 to 50,000 JE cases annually,81 conflicting with WHO’s year-wise reported cases, which indicated 1,877 cases globally in 2022 and 3,392 in 2011.82 This discrepancy raises questions regarding the surveillance or reporting accuracy and possible overestimation in disease estimations.

JE’s epidemiology varies among countries. WHO identified 24 countries in the South-East Asia and Western Pacific regions with JEV transmission risk, encompassing more than 3 billion individuals.83 Predominantly reported cases stemmed from Asian countries, with India and China accounting for 95% of them84 India, an endemic nation for JE, has witnessed outbreaks in 327 districts across 24 states, primarily in the north-eastern and northern regions.85 The highest recorded cases were 6,000 in 2005 and 4,017 in 2006,76,86 followed by 2,545 in 2019. The year 2022 recorded 1109 cases with 130 deaths. By September 30, 2023, NCVBDC documented 828 cases and 48 deaths due to AES/JE.82,87 Specific regions in India, like eastern Uttar Pradesh, Karnataka, Andhra Pradesh, Uttar Pradesh, and West Bengal, are particularly susceptible to JE due to their terrain and annual flooding.85

Clinical symptoms

Following a mosquito bite carrying the JE virus, an incubation period of 5-15 days begins. Most individuals infected with the JE virus either experience no symptoms or have mild symptoms such as fever, headache, and vomiting. However, a small percentage develops encephalitis, characterized by prodromal fever, headache, nausea, diarrhea, vomiting, weakness, and myalgia, lasting a few days. Less than 1% of those infected with the JE virus develop severe neurological illness, and approximately 1 in 4 of these cases result in fatalities. Although certain symptoms may improve following the acute phase, 30%-50% of individuals who survive encephalitis continue to endure neurological, cognitive, or psychiatric issues.76,88

Diagnosis and management

The surveillance for fever and AES helps to identify suspected cases of JE, which can then be confirmed using laboratory tests. Confirmation of JE involves several diagnostic criteria: the presence of IgM antibodies in serum and/or cerebrospinal fluid, a four-fold difference in IgG antibody titer in paired sera, virus isolation from brain tissue, antigen detection through immunofluorescence, and nucleic acid detection via PCR. There is no specific anti-viral treatment available for the infection, and clinical management is only symptomatic.77

Preventive measures

The JEV has five distinct genotypes (I, II, III, IV, and V). In India, the available JE vaccines primarily target the GT-III. JE vaccines available in India are the LAV, cell culture-derived SA 14-14-2, inactivated SA-14-14-2 vaccine (IC51) (IXIARO® by Intercel & JEEV® by Biological Evans India Ltd.), and inactivated vero cell culture-derived Kolar strain, 821564XY, JE vaccine (JENVAC® by Bharat Biotech). Vaccination is not recommended for general use but specifically for individuals residing in endemic areas.89,90 LAV, Cell culture-derived SA 14-14-2 was introduced in the routine immunization under Universal immunization program (UIP) of government in the 297 endemic districts of 24 states of India.79,91-93 All children above the age of 9 months must be offered the vaccine. This vaccination schedule involves two doses: one given alongside measles at 9 months and the second with the DPT booster at 16-24 months. Recently, the Government of India announced the introduction of one dose of JE vaccine for adults in 31 endemic districts.79,92

Challenges

Addressing the challenges associated with JE requires a multifaceted approach encompassing heightened awareness, enhanced surveillance, improved rural healthcare accessibility, and continuous research efforts. Efforts should be directed towards increasing public awareness of JE, as the lack of understanding among the general population can result in delayed diagnosis and appropriate management. Concurrently, addressing the limitations in JE surveillance in India is crucial for accurately tracking the incidence and prevalence of the disease. In rural areas, where JE is more prevalent, there is a pressing need to expand access to healthcare facilities. Limited healthcare accessibility can impede the timely diagnosis and treatment of JE, underscoring the importance of bolstering healthcare infrastructure in these regions. Furthermore, ongoing research is paramount to evaluating the effectiveness of current vaccines against the evolving genotypes of the virus. The emergence of new genotypes, such as GT-I and GT-V, raises questions about the potential impact on existing vaccines.93,94 Similarly, continuous monitoring of the genetic and antigenic variations among circulating JEV strains is essential, especially considering the expansion of GT-I JEV into different parts of India. Staying updated on ongoing research and recommendations is crucial for adapting vaccination strategies to the evolving genotypes of the virus. While JE vaccination is integrated into the UIP in India, the coverage remains limited. Many individuals in endemic areas remain unvaccinated, emphasizing the need for an expanded and inclusive vaccination strategy. Efforts should be intensified to overcome these challenges and ensure comprehensive protection against JE in India.89,94,95 To conclude, enhanced surveillance, better vaccination access, and strengthened vector control measures are key to mitigating the impact of JE in India.

4. Zika virus infection

Zika virus, a member of the Flavivirus genus within the Flaviviridae family, shares its viral lineage with other significant viruses like dengue, yellow fever, West Nile, and JEVs.96,97 This ssRNA virus is primarily transmitted by Aedes species mosquitoes, with A. aegypti and A. albopictus being the main carriers.90,96 The virus exhibits two major geographically distinct lineages: Asian and African.97 Notably, Asian lineage strains have been associated with heightened transmission rates and more severe disease outcomes, including congenital abnormalities like microcephaly and neurological disorders such as Guillain-Barré syndrome.98,99 In contrast, African lineage strains have not shown a similar association with severe outcomes.100

Epidemiology

Zika virus was initially identified in 1947 in Uganda, originating from a Rhesus macaque. Since then, it has spread across 89 countries and territories, triggering outbreaks in Africa, the Americas, Asia, and the Pacific regions.90 In India, three confirmed cases of Zika virus disease were recorded on May 15, 2017, in the Bapu Nagar area of Ahmedabad, Gujarat.101 Subsequently, Zika virus presence has been documented in 16 states/UTs of India.6 The most recent cases were reported in Kerala and Maharashtra in July 2021.25 Notably, the genetic sequences of the Zika virus obtained from cases in Ahmedabad showcased a close relationship to Asian strains, underlining its lineage in India.96

Clinical symptoms

Zika virus primarily spreads through the bite of infected Aedes mosquitoes, but it’s also known to transmit from pregnant women to their fetuses or through sexual contact, blood transfusion, organ transplantation, or even laboratory exposure.102,103 The incubation period of the disease typically ranges from 3 to 14 days. Interestingly, most infected individuals are asymptomatic. When symptoms do arise, they’re generally mild, characterized by fever, rash, conjunctivitis, muscle and joint pain, malaise, and headache, lasting for around 2 to 7 days.

Pregnant women infected with the Zika virus face severe risks as it can cause microcephaly and other birth defects in the developing fetus and newborn. Additionally, it leads to complications like fetal loss, stillbirth, and preterm birth. Furthermore, Zika virus infection is associated with triggering Guillain-Barré syndrome, neuropathy, and myelitis, particularly in adults and older children.104

Diagnosis and management

In India, Zika virus is typically diagnosed using PCR and virus isolation from blood samples, primarily conducted at the National Centre for Disease Control in Delhi and the National Institute of Virology in Pune. However, serological diagnosis can be challenging due to potential cross-reactivity with other flaviviruses like West Nile and Yellow fever.103,105

As of now, there isn’t a specific treatment available for Zika virus infection or disease. Treatment mainly focuses on managing symptoms, using antipyretics and/or analgesics. It’s crucial to avoid nonsteroidal anti-inflammatory drugs until DENV infections are ruled out, to minimize the risk of bleeding. Pregnant women residing in areas with Zika transmission should promptly seek medical attention for laboratory testing, guidance, counselling, and appropriate clinical care. Though numerous potential vaccines for the Zika virus have successfully undergone either phase I or phase II clinical trials; currently, no vaccine is currently available.106

Challenges

The challenges posed by the Zika virus in India are extensive and multifaceted, encompassing several critical aspects. First, the 2021 outbreak of the Zika virus underscores its circulation in the SouthEast Asian region, indicating the potential for new outbreaks in the future.25 The omnipresence of the Aedes mosquito vector in India poses a formidable challenge in curbing the virus’s spread. A study conducted in Jaipur, India, detailed the first Zika Virus outbreak, analyzing its clinico-epidemiological and genomic profile. The study emphasized the urgent requirement for enhanced surveillance and control measures. The spread of the Zika virus to multiple states in India highlights the pressing need for improved surveillance, particularly targeting pregnant women. Additionally, public health education is imperative to mitigate risks during pregnancy.102 The other major challenges are co-infections and cross-reactivity with other viruses. Zika virus shares similarities with other mosquito-borne diseases like dengue and chikungunya, leading to concerns about co-infections and cross-reactivity. This similarity complicates case detection and surveillance, primarily due to the limited availability of diagnostic tests and the challenges in interpreting serologic test results owing to the known cross-reactivity with related flaviviruses, notably DENV.107

5. Kyasanur forest disease virus (KFDV) infection

Kyasanur forest disease (KFD) is a tick-borne viral hemorrhagic fever induced by the KFD virus (KFDV), belonging to the Flaviviridae virus family.108 The KFDV exhibits infectivity in various species, including humans, monkeys, rodents, bats, and small mammals.109 Hard ticks, specifically Hemaphysalis spinigera, serve as reservoirs for the KFD virus, maintaining infection for their entire lifespan. Once infected ticks bite rodents, shrews, or monkeys, these animals become common hosts for the KFDV.109

Epidemiology

KFD was discovered in 1957 after a sick monkey in Karnataka’s Kyasanur forest tested positive.104 Initially confined to specific districts in Karnataka, recent years have seen reported cases emerging in Kerala, Goa, and Maharashtra.108 This geographical expansion beyond the endemic regions has triggered significant public health concerns over the past 15 years.110 KFD’s fatality rate ranges from 3% to 10%, affecting approximately 400 to 500 individuals annually.111 Its peak prevalence occurs from January to May, particularly in forested areas, heightening the risk of transmission during this period. Those regularly exposed to forest environments, such as forest guards, shepherds, and hunters, face a higher vulnerability to contracting KFD, which is transmitted through bites of infected hard ticks.112

Symptoms

The clinical manifestation of KFD typically follows a biphasic pattern, although it’s occasionally described in four stages. In the initial phase, patients commonly exhibit abrupt onset symptoms, including fever, chills, severe muscle pain, headache, vomiting, gastrointestinal issues, and bleeding complications. If the second phase occurs, which is observed in around 10-20% of patients, it emerges at the onset of the third week, termed the biphasic presentation. During this phase, patients may experience another wave of symptoms, featuring fever and neurological manifestations like severe headaches, mental disturbances, tremors, and vision deficits.112

Diagnosis and management

During the early stages of illness, molecular detection via Real time-PCR, Nested PCR, or qPCR, along with virus isolation from blood, can be utilized for diagnosis. Subsequently, serologic testing using ELISA to detect anti-KFD IgM and IgG antibodies becomes feasible. Timely diagnosis of KFD holds immense significance for initiating prompt treatment and managing the disease. While there’s no specific treatment available, early hospitalization coupled with supportive therapy remains crucial.108

Preventive measures

An existing KFD vaccine, developed in the 1960s in India, comprises a formalin-inactivated tissue culture vaccine.106 However, concerns have been raised about its efficacy, given reported cases of the disease in vaccinated individuals. This might be attributed to virus variants in circulation or low vaccine coverage.113,114 To enhance the prevention and control of KFD, further research is imperative, aiming to develop more efficacious vaccines.111,113

Challenges

Most cases of KFD remain underreported due to misdiagnosis or lack of reporting, attributed partly to its similarity to other diseases.115 Additionally, limited access to serological testing and challenges in implementing Real time-PCR-based diagnostic tools in remote areas contribute to this underreporting.115 The absence of comprehensive surveillance and screening at the community level hampers accurate disease burden estimation, affecting policy formulation by stakeholders and policymakers. There is a scarcity of national data surveillance strategies for this disease in our country. Deforestation and habitat loss pose significant challenges,115 as they push human habitation closer to the natural reservoirs of the virus found in infected monkeys and small mammals. This disruption to the ecosystem alters the behavior and distribution of tick vectors and their hosts, influencing virus transmission dynamics. The current vaccination strategy, limited to a 5 km radius around reported KFD areas, may need a relook, as cases have appeared beyond this range. Phylogenetic studies have traced the virus’s dispersal from Karnataka to Goa and Maharashtra, highlighting the need for improved vaccination and surveillance strategies.116 Further research on KFDV’s genotyping and molecular epidemiology is necessary for a deeper understanding of its transmission dynamics.117,118 Novel control strategies, like the development of edible vaccines, have been proposed as preventive measures for KFD.111 Increasing awareness of emerging zoonotic infections, enhancing laboratory capacities, and implementing One Health programs are crucial aspects of public health intervention. Addressing these challenges requires comprehensive measures, including improved access to serologic/molecular testing in remote areas, bolstered surveillance and communication systems, and interdisciplinary collaboration to enhance KFD diagnosis and management. Beyond individual health impacts, these diseases strain healthcare facilities, reduce productivity due to illness, and disproportionately affect marginalized communities, incurring substantial economic burdens from healthcare costs and lost productivity.

6. Chandipura virus (CHPV)

First isolated in 1965 from Chandipura village in Maharashtra State, India, CHPV is an emerging vector-borne pathogen belonging to the family Rhabdoviridae. CHPV carries its genome as negative sense, ssRNA of ∼11kb, expressing five different proteins. It causes sporadic cases and frequent outbreaks of acute encephalitis, influenza-like illness, and neurologic dysfunctions. primarily affecting infants and children (under 15 years) in tropical regions of western, central, and southern parts of India.119,120

Transmission

The proposed vectors of the virus are female sandflies, mosquitoes, and ticks. Phlebotomus papatasi (a sandfly) is reported to be the vector of CHPV disease in Gujarat. Human-to-human transmission without vector involvement is not known to occur, Treatment and prevention: No specific treatment or vaccine is available for the virus. The transmission is supportive to control any complication.120

Chandipura outbreaks in India

In 2003, southern India (Andhra Pradesh) experienced a CHPV outbreak in which ∼350 children developed AES and ∼200 died. Since then, CHPV has been frequently reported in adjoining states in central India and thus poses a serious threat of an epidemic outbreak and significant public health concerns in the Indian subcontinent. Since sandfly vectors are abundant in South-East Asia and African regions, CHPV may be present in other countries of Asia and Africa. The most serious concern about CHPV is that it can cause high case-fatality ratios, ranging from 56% to 75% as reported during previous outbreaks in India. The disease manifests with a febrile illness that may progress to convulsions and coma and can lead to high mortality within 48 to 72 hours of the onset of symptoms typically represented by acute encephalitis. Given these forewarnings, it is of paramount importance that CHPV biology can be understood comprehensively. A lack of vaccine or effective therapy stresses the immediate and urgent need for antiviral therapeutics.121

The Ministry of Health and Family Welfare of the Government of India, reported about 245 cases of AES including 82 deaths with 64 confirmed cases of CHPV infections between early June and 15 August 2024. The CHPV outbreak of 2024 is estimated to be the largest in the past 20 years and calls for early access to care and intensive supportive care of patients for better survival. The recommendations from WHO include vector control and protection against sandfly, mosquito, and tick bites to prevent the further spread of CHPV. It is a big relief, though, that human-to-human transmission of CHPV has not been observed, yet the recent outbreak poses several challenges in preparedness and response against possible future CHPV outbreaks, which include

  • i.

    Lack of treatment: There is no specific treatment, vaccine, or antiviral therapy for CHPV.

  • ii.

    Lack of diagnosis: We need better diagnostic tests/kits for rapid (on-site) detection of CHPV besides PCR based-tests

  • iii.

    More research on CHPV: CHPV receptors on the host cell surfaces remains unknown. Knowledge of such receptor (s) may be instrumental in designing much needed antivirals.

    Likewise, better knowledge of the CHPV life cycle may help design/develop specific antiviral drugs.

  • iv.

    Need for the expansion of the healthcare system and awareness among the people

7. Crimean-congo hemorrhagic fever

Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne viral disease that emerged in India in 2011. It is caused by the Nairovirus of the Bunyaviridae family and transmitted primarily through the bite of Hyalomma ticks or contact with infected livestock and humans. It causes severe hemorrhagic fever with a high mortality rate. Due to the large livestock population and farming industry, the risk of transmission in certain regions is significant, especially among individuals working in animal husbandry or healthcare.122

Epidemiology

The first confirmed case of CCHF in India occurred in January 2011 during a nosocomial outbreak in Ahmedabad, Gujarat. Since then, outbreaks and sporadic cases have been reported, predominantly in Gujarat and Rajasthan. One case was detected in Kerala in 2016 and traced to a slaughterhouse worker who had traveled from Oman. Seroprevalence studies in Gujarat revealed that a substantial proportion of domestic animals, especially cattle, sheep, and goats, had been exposed to CCHF virus. Males are more commonly affected, and proximity to a confirmed CCHF case increases the likelihood of contracting the disease.123

Clinical symptoms

CCHF presents with sudden onset symptoms, including high fever, muscle aches, dizziness, headache, neck pain, and sensitivity to light. Gastrointestinal symptoms such as nausea, vomiting, and abdominal pain are common, followed by mood changes and confusion. Hemorrhagic symptoms, including bleeding from the gums, petechiae, and larger bruises (ecchymoses), may develop, leading to multi-organ failure. The disease can be fatal, with a mortality rate of 30–50%, and most deaths occur within the second week of illness. Survivors usually begin to recover after the 9th or 10th day.122,123

Diagnosis and management

Clinical diagnosing CCHF is challenging because its symptoms mimic those of other hemorrhagic fevers, such as dengue and leptospirosis. Laboratory tests such as ELISA and RT-PCR are used for diagnosis. There is no specific treatment for CCHF. Supportive care, including fluid replacement and treatment of symptoms is the primary approach. Ribavirin, an antiviral drug, has shown some effectiveness in reducing the severity of the disease. In the absence of a vaccine, prevention strategies focus on controlling tick populations and reducing human contact with infected animals and patients.122,124

Challenges

There are several challenges in controlling CCHF. Due to the absence of a vaccine for both animals and humans, disease control relies on vector control and public awareness. Healthcare workers are at a significant risk of contracting the virus through contact with infected patients, and nosocomial outbreaks remain a concern. Diagnosing CCHF can be problematic due to the similarity of its symptoms to other viral hemorrhagic fevers. Improving diagnostic capacity, especially in regions where the disease is endemic, is crucial for timely identification and containment of outbreaks.125

National government initiatives on current preventive measures

  • a)

    Vector control programs: These initiatives concentrate on diminishing mosquito breeding sites, employing insecticides, and fostering community engagement.

  • b)

    Public awareness and education: Empowering communities with knowledge regarding preventive measures and early symptom recognition is a pivotal component of the strategy.

  • c)

    Development of guidelines and operational manuals: Technical guidance is provided to states and other stakeholders for the effective implementation of programs. All guidelines are accessible on the NVBDCP website.114

  • d)

    Establishment of sentinel surveillance hospitals (SSH): Hospitals equipped with laboratory support have been established to enhance diagnostic facilities for diseases like dengue in endemic states since 2007. The number of these hospitals has increased to 805 in 2023. All these hospitals are linked with 17 apex referral laboratories (ARL), which offer advanced diagnostic support.114

  • e)

    Ensuring functional diagnostic facilities: The responsibility for maintaining functional diagnostic facilities (SSH/ARL) and ensuring the availability of kits lies with the respective State Programme Officers of NCVBDC. The government has provided technical guidelines for prevention and control, case management, and effective community participation to the states for implementation.114

VECTOR-BORNE PARASITIC DISEASES IN INDIA

1. Malaria

Malaria is a life-threatening infectious disease that continues to impose a significant burden on global health, particularly in tropical and subtropical regions. Caused by Plasmodium parasites transmitted through the bites of infected Anopheles mosquitoes, malaria’s impact extends far beyond mere health implications.126 The WHO South-East Asia Region had nine malaria-endemic countries in 2022, accounting for 5.2 million cases and contributing to 2% of the burden of malaria cases globally. In 2022, India accounted for about 65.7% of all malaria cases in the region. Almost 46% of all cases in the region were due to P. vivax.127 According to World Malaria Report 2023, Between 2020 and 2022, all countries in the region in which malaria deaths occurred reported an increase in the malaria mortality rate, except for India.127 The disease disproportionately affects low-income countries, where it contributes to a substantial burden of morbidity and mortality, hindering economic development and perpetuating cycles of poverty.128 Common symptoms include fever, chills, and flu-like illness, with severe cases leading to complications such as anemia and organ failure if left untreated. Geographical distribution is influenced by environmental factors like temperature, rainfall, and humidity, dictating the disease’s boundaries, seasonality, and intensity.129 Efforts to control malaria encompass a wide array of preventive measures, including insecticide-treated bed nets, indoor residual spraying, and prophylactic antimalarial drugs.127 However, challenges persist, such as the emergence of drug-resistant parasite strains and insecticide-resistant mosquitoes, along with barriers to healthcare access in affected regions. These challenges underscore the need for continued research and collaborative interventions aimed at combating malaria’s multifaceted complexities. By addressing environmental, social, and medical factors, global initiatives strive to reduce the burden of malaria, improve healthcare delivery, and ultimately, work towards the elimination of this devastating disease.

Malaria burden and trends of malaria reduction

In 2022, malaria remained a significant global health challenge, with an estimated 249 million cases reported across 85 malaria-endemic countries and territories, including French Guiana. This marked an increase of 5 million cases compared to 2021.130 Within the WHO South-East Asia Region, which comprised nine malaria-endemic countries, there were 5.2 million estimated cases, contributing to 2% of the global malaria burden. Notably, India alone accounted for approximately 65.7% of all malaria cases in the region. Of the total cases reported in the region, nearly 46% were attributed to Plasmodium vivax infection.127 Despite significant progress, malaria remains a substantial global health concern, as highlighted by the World Malaria Report 2021, with an estimated 241 million cases and 627,000 deaths in 2020, with African countries carrying a majority of the burden. Over the past two decades, the South-East Asia Region (SEAR) has witnessed a remarkable decline in malaria incidence, dropping from an estimated 23 million cases in 2000 to 5 million in 2020.131 India, with the greatest absolute decline in the region from around 20 million cases in 2000 to approximately 4.1 million in 2020, still accounted for 83% of malaria cases and 82% of fatalities in SEAR.132 The period between 2001 and 2015 marked a significant global reduction in malaria incidence by 30%, along with a 47% decrease in the fatality rate, preventing an estimated 4.3 million deaths.133 This achievement prompted the WHO to adopt the global technical strategy (GTS) for Malaria 2016–2030 in 2015, aiming to provide technical support for scaling up malaria responses and working toward the ambitious goal of a 90% reduction in the global malaria burden and interrupting transmission in at least 35 countries by 2030.133

Despite these efforts, the context of malaria reduction highlights the ongoing challenges, with SEAR and India playing pivotal roles in the global malaria landscape. Continuous commitment to interventions, innovative strategies, and international collaboration remains crucial to achieving the GTS targets and ultimately eliminating malaria as a public health threat.

Tools for malaria control

Diagnostics: Enhanced diagnostics are imperative for monitoring changes in malaria infection rates, ensuring the quality of medicines and insecticides, and assessing factors influencing treatment decisions.134,135 Current rapid diagnostic tests (RDTs) are effective for diagnosing symptomatic malaria with higher parasite densities, expanding access beyond health facilities. However, a critical gap persists as these tests cannot detect low-level blood-stage infections of any malaria species or dormant liver stages of Plasmodium vivax and P. ovale.136 There’s a pressing need for a highly sensitive point-of-care field test capable of rapidly detecting low-density parasitemia and identifying all infected individuals for immediate treatment.

Point-of-care G6PD deficiency test: To safely treat P. vivax, detection of G6PD enzyme deficiency is essential due to the risk of acute hemolytic anemia with primaquine.137 Currently limited to specific facilities, rapid testing for enzyme deficiency, coupled with easy data access, will facilitate the broader implementation of effective P. vivax treatments to reduce malaria transmission.

Malaria microscopy: Malaria microscopy, a cornerstone in the diagnosis of the disease, is not without its challenges, which encompass technical, logistical, and operational aspects. One primary challenge lies in the need for highly skilled microscopists capable of accurately identifying and differentiating various Plasmodium species and their different life stages.138 Achieving proficiency in malaria microscopy demands extensive training and experience, and maintaining a cadre of skilled microscopists can be a logistical challenge in resource-constrained settings. Furthermore, the requirement for microscopic examination of blood smears is time-consuming and labor-intensive, posing operational challenges, particularly in regions facing high caseloads or during disease outbreaks. Additionally, the reliance on microscopy limits access to accurate diagnosis in remote or underserved areas, where skilled professionals and laboratory infrastructure may be lacking. To address these challenges and enhance the effectiveness of malaria microscopy, several solutions are being implemented. Technological advancements have led to the development of automated or semi-automated microscopy systems that assist in parasite detection and species identification, reducing the dependency on manual examination.126 Additionally, the use of quality assurance programs and proficiency testing helps ensure the ongoing competency of microscopists.139 Efforts are being made to strengthen laboratory infrastructure and capacity in malaria-endemic regions, including the recruitment and training of laboratory personnel. Moreover, innovative strategies such as telemedicine and remote consultation are being explored to connect microscopists in remote areas with experienced experts for real-time guidance and quality assurance. In the quest for improved malaria diagnostics, molecular techniques, like PCR, are being integrated into diagnostic algorithms to complement microscopy, especially in detecting low-density infections and differentiating between parasite species.140,141 ICMR-National Institute of Malaria Research has developed India’s first malaria slide bank to strengthen the diagnostic capability and training programs in India.139,140,142 Emphasizing a comprehensive approach that combines technological innovation, capacity building, and strategic partnerships can overcome the challenges associated with malaria microscopy, ensuring accurate and timely diagnosis in the global effort to combat malaria.

Molecular methods: As countries move towards malaria elimination, the rise in low-density and asymptomatic infections necessitates new reference standards. Molecular methods like loop-mediated isothermal amplification and PCR play a crucial role in detecting low-density parasitemia, with PCR distinguishing between local and imported cases.140 While their field applicability is expanding, decreasing costs and complexity enhance their potential for wider adoption.

Insecticide quantification kits: To ensure the quality of insecticide formulations and assess the effectiveness of indoor residual spraying (IRS), straightforward point-of-use kits have been developed. These kits serve as practical alternatives to insecticide bioassay methods, which, although recognized as the gold standard, are often impractical in resource-limited public health systems and are rarely employed in operational programs.7

Insecticide bioassay methods involve exposing live mosquitoes to treated surfaces and measuring the knockdown and mortality rates to determine insecticide efficacy. While accurate, this approach is resource-intensive, time-consuming, and requires specialized skills and facilities, making it challenging for routine use in operational settings. The development of point-of-use kits addresses these limitations. These kits are designed to provide a more practical and accessible means of assessing insecticide quality and IRS efficacy. They offer a simplified, user-friendly approach that can be implemented in diverse settings, including those with limited resources and infrastructure.143 By replacing the traditional bioassay methods with these kits, public health programs can more feasibly and routinely monitor the quality of insecticide formulations and the performance of IRS interventions. This shift towards user-friendly tools enhances the practicality and efficiency of quality assurance measures, contributing to the overall success of malaria control and prevention efforts.

Health education and awareness: Public awareness campaigns are conducted to educate communities about the importance of preventive measures such as allowing house spraying, using bed nets, wearing protective clothing, and seeking timely diagnosis and treatment for fever.144

Community engagement: Engaging communities and involving them in malaria control efforts can enhance the effectiveness and sustainability of interventions.145,146 However, this is not easy. This can include training community health workers, involving local leaders, and mobilizing community participation in vector control activities. Continued commitment from the government, sustained funding, and innovative approaches are essential to further progress in malaria control in India.145

Integration with other health programs

Malaria control efforts are often integrated with other health programs, such as maternal and child health, as well as with broader development initiatives addressing poverty, sanitation, and access to clean water.147

Research and innovation

Research initiatives focus on developing new tools for malaria control, including better diagnostics, drugs, and vaccines. Collaboration with international partners and research institutions would help in leveraging expertise and resources and build partnerships within the country and from overseas.148

Current challenges

Antimalarial drug resistance: Various tools are employed to assess antimalarial drug resistance, with genetic changes linked to reduced drug sensitivity serving as key indicators. For instance, specific mutations in the P. falciparum Kelch13 (PfKelch13) gene are associated with delayed parasite clearance following artemisinin-based treatments.149 Monitoring surveys of these mutations offer insights into the emergence and dissemination of artemisinin partial resistance, characterized by delayed clearance post-treatment. Sulfadoxine-pyrimethamine, utilized as an artemisinin-based combination therapy partner and for chemoprevention, has its resistance monitored through the identification of mutations in the dihydrofolate reductase (dhfr) and dihydropteroate synthase (dhps) genes of P. falciparum. Mefloquine resistance is associated with an upsurge in Pfmdr1 copy numbers, while piperaquine resistance is linked to increased Pf plasmepsin 2/3 copy numbers and mutations in the P. falciparum chloroquine resistance transporter (PfCRT). It is noteworthy that certain mutations are validated as resistance markers solely in parasite strains from specific geographic regions. These genetic assessments play a crucial role in monitoring and understanding antimalarial drug resistance, informing strategies for effective malaria treatment and control.150-152

Diagnostic: Accurate diagnosis plays a pivotal role in achieving the goal of malaria elimination. While P. falciparum and P. vivax contribute to most cases among the five Plasmodium species, the varied distribution of both mono-infection and mixed infections complicates the diagnostic process. Microscopy, traditionally considered the gold standard, requires highly skilled microscopists well-versed in recognizing different Plasmodium species stages capable of detecting even low-density parasitemia. Meeting such requirements in rural India poses a significant challenge, resulting in more than a quarter of malaria cases being overlooked by microscopy.7,135 In areas where microscopy is impractical, RDTs are employed. P. falciparum histidine-rich protein 2 (PfHRP2) antigen is a prominent target in over 90% of malaria RDTs.153 Nevertheless, challenges such as deletions in the Pfhrp2 gene, fluctuations in plasmodium lactic dehydrogenase (pLDH) expression levels, and the prozone phenomenon pose significant hurdles, leading to inaccurate diagnoses of Plasmodium species. Addressing these challenges is imperative for ensuring the reliability of malaria diagnoses, particularly in regions where microscopy may not be feasible

The problem of insecticide resistance in malaria vector: Between 2010 and 2020, out of the 88 countries providing insecticide resistance monitoring data to WHO, 78 countries confirmed resistance to at least one insecticide in a malaria vector species at a specific mosquito collection site. Among these, 29 countries reported resistance to four insecticide classes—pyrethroids, organophosphates, carbamates, and organochlorines—in at least one malaria vector species across different locations within the country.127,143 Within this group, 19 countries identified at least one site where resistance was confirmed for all four classes in a local vector. Globally, resistance to pyrethroids was observed in 87% of countries and 68% of sites, to organochlorines in 82% of countries and 64% of sites, to carbamates in 69% of countries and 34% of sites, and to organophosphates in 60% of countries and 28% of sites. Resistance to these four insecticide classes was found in all WHO regions, although the geographical distribution of resistance varied widely among regions. In India and various other locations, insecticides have been a primary component of vector control strategies. Presently, 83 vector control products have received prequalification from the WHO. In India, a range of insecticides, including dichloro-diphenyl-trichloroethane (DDT), malathion, and various pyrethroids, have been deployed for adult malaria vector control. Additionally, larval control involves the use of temephos, bacterial pesticides, and insect growth regulators, while space spray includes malathion and formulations of synthetic pyrethroids like deltamethrin and cyphenothrin, as well as natural pyrethrum extract.127

Proper utilization of LLINs as vector control tool: LLINs today stand as the globally favored tools for vector control. Ensuring their sustained impact and timely replacement due to wear and ineffectiveness requires diligent monitoring and evaluation. In mid-2017, on distribution by the NCVBDC in high malaria-endemic districts of Odisha, the LLINs demonstrated efficacy, achieving 100% mortality in Anopheles jeyporiens is mosquitoes 30 months post-distribution.131 LLINs are a cornerstone in malaria vector control, primarily distributed through national malaria control programs worldwide, including India’s NVBDCP. Despite being a primary channel for net distribution, challenges persist in achieving optimal coverage, attributed to factors like population growth, timely procurement, replacement delays, emergencies (such as floods), and logistical constraints. To enhance access, India must explore complementary avenues, such as making nets available in the private sector at affordable prices. Although barriers like affordability, economic viability for manufacturers, and regulatory issues exist, they can be pragmatically addressed. The commercialization of nets in the private market is imperative for India to achieve its malaria elimination goal by 2030.127,148

Climate changes: The WHO has identified climate change as the most significant health threat to humanity. This impact is disproportionately felt by populations in low-income countries, even though they contribute minimally to the causes of climate change. Many of these nations also grapple with malaria as a significant public health concern. The implications of climate change extend beyond direct environmental consequences and pose a substantial risk to global health progress. These risks include disruptions to livelihoods, increased exposure to harmful particulates, pathogens, and diseases, strain on health systems, and the exacerbation of existing social and economic inequalities. Therefore, climate change is not merely an isolated threat but a substantial amplifier of various health-related challenges. Malaria transmission is significantly influenced by the distinct and interconnected impacts of temperature, rainfall, and humidity. These elements not only delineate the geographical boundaries of the disease but also govern its seasonal patterns and intensity within those boundaries. As a result, these factors shape the epidemiology of malaria, determine the disease burden, and impact the effectiveness of various interventions. These considerations form the foundation for the development of national malaria strategic plans.126

Modern day developments and malaria: Several modern developments have contributed to fostering malaria transmission. Rapid urbanization, especially in cities like Mumbai, Delhi, and Bangalore, has outpaced the development of proper infrastructure. Poorly planned construction projects, slums with inadequate drainage systems, and stagnant water in construction sites create ideal breeding grounds for Anopheles mosquitoes. High levels of migration from high endemic areas for labor, particularly from rural, malaria-endemic states, contribute to the spread of malaria. Large-scale irrigation projects and dam construction, such as those on the Narmada and Godavari rivers, have also increased malaria risk in nearby areas. The creation of stagnant water in reservoirs, canals, and irrigation systems provides ideal breeding conditions for mosquitoes, leading to higher transmission in surrounding villages and towns.154,155

Zoonotic malaria

Until recently, four human malaria parasites, with P. falciparum and P. vivax being the most prevalent, were recognized. However, a fifth parasite, P. knowlesi, has emerged as a significant challenge for malaria control, particularly in South-East Asia. Initially identified in monkeys, this zoonotic parasite poses a substantial threat, characterized by a human fatality rate of 1–2% and the rapid onset of severe disease. First detected in Malaysia in 2004, P. knowlesi has since spread extensively across Southeast Asia and globally through travel and tourism, except for Timor-Leste. Despite a global decline of 24.2% in P. knowlesi cases in 2022 (2768 cases), notable increases occurred in Indonesia and Thailand. Alarmingly, P. knowlesi was responsible for all malaria-related deaths in Malaysia and Thailand that year. The spread of P. knowlesi complicates malaria elimination efforts and impacts the WHO’s certification process for malaria-free status, which traditionally focused on four human malaria parasite species. In response to the rise of P. knowlesi, WHO is reassessing the criteria for certification.127,156

Future perspectives in malaria elimination

Prospects in malaria research in India involve a multidisciplinary approach aimed at addressing various challenges associated with malaria control and elimination. Here are some potential areas of focus for future research:

Development of novel control tools: Research efforts can focus on developing innovative vector control tools such as new insecticides, repellents, and traps. Emphasis should be placed on environmentally friendly and sustainable interventions to combat insecticide resistance.8,157

Drug discovery and resistance management: Continued research into antimalarial drug discovery is crucial, including the development of new compounds and strategies to overcome drug resistance. Understanding the mechanisms of drug resistance and developing combination therapies can help prolong the efficacy of existing drugs.158,159

Vaccine development: Despite progress, an effective malaria vaccine remains elusive. Research in India can contribute to vaccine development efforts by identifying suitable vaccine candidates, optimizing vaccine delivery strategies, and conducting clinical trials to assess vaccine safety and efficacy.160,161

Genomic studies: Genomic studies of malaria parasites and mosquito vectors can provide valuable insights into their biology, evolution, and transmission dynamics. This knowledge can inform the development of targeted interventions and help track the spread of drug-resistant parasites and insecticide-resistant mosquitoes.

Health systems research: Research on health systems and delivery mechanisms is essential for optimizing the implementation of malaria control interventions. This includes assessing the effectiveness of community health worker programs, strengthening surveillance systems, and improving access to diagnosis and treatment services, particularly in remote and underserved areas.146,162

Climate change and environmental factors: Climate change can influence the distribution and transmission of malaria. Research on the impact of climate change on malaria transmission patterns, as well as the role of environmental factors such as deforestation and urbanization, can inform adaptive strategies for malaria control and elimination.127,162

Behavioral and socioeconomic research: Understanding the social and behavioral determinants of malaria transmission is crucial for designing effective interventions suiting the local conditions. Research can explore factors influencing mosquito breeding habitats, community perceptions of malaria, health-seeking behaviors, and adherence to preventive measures.163

Integration with other health programs: Research can investigate the potential synergies between malaria control efforts and other health programs such as maternal and child health, nutrition, and water, sanitation, and hygiene (WASH). Integrated approaches can maximize health impact and resource efficiency.164

Partnerships and collaboration: Collaboration between researchers, government agencies, NGOs, and international partners is essential for advancing malaria research and translating findings into policy and practice. Collaborative research networks can facilitate knowledge sharing, capacity building, and the mobilization of resources.165

Summary

Despite these challenges, India has made significant progress in malaria control through various interventions, including vector control measures, prompt diagnosis and treatment, community engagement, and research initiatives. The government, along with partners at the national and international levels, continues to prioritize malaria control and elimination efforts to further reduce the burden of this disease in India.

2. Leishmania and leishmaniasis

Historical aspects

The recognition of Leishmania parasites as the causative agents of leishmaniasis began in the 20th century, a disease transmitted by sandflies (vector). Historical evidence of leishmaniasis dates to 2,500 B.C., with primitive descriptions found in ancient archaeological materials and writings. Molecular findings confirm the presence of Leishmania in ancient Egyptian and Christian Nubian mummies dating back to 2,000 B.C. In northern Sudan, Leishmanial DNA was reported in 1,500 B.C., and in Peru, a 6-year-old girl mummy infected with Leishmania was recorded in 800 B.C. In the 18th century, Russell (1756) provided the first detailed clinical examination of leishmaniasis.166,167

Kala-azar symptoms were first described by Indian physicians in the 17th century, with sand flies playing a role in disease transmission. In 1900, William Boog Leishman identified ovoid bodies in splenic smears,168 leading to the term “Dum-dum fever.” Charles Donovan and Ronald Ross later identified a new parasite. Further research proved sandfly as the vector after discovering Leishmania promastigotes in the sandfly gut and its various species were reported.169,170

Epidemiology of Leishmaniasis

Regarding the burden of leishmaniasis, six countries are regarded as important ones. More than 90% of visceral leishmaniasis (VL) cases are reported from these nations: Bangladesh, Brazil, Ethiopia, India, South Sudan, and Sudan. Ten nations make up the majority of cutaneous leishmaniasis (CL) cases, with more than 70% of them involving CL. These nations, which account for more than 70% of cases, are the Islamic Republic of Iran, Ethiopia, Algeria, Brazil, Colombia, Costa Rica, Peru, Sudan, and the Syrian Arab Republic. According to the WHO, the populations of these endemic countries are 431 million and 616 million vulnerable to CL and VL, respectively. According to the WHO (2015), there were 1.3 million leishmaniasis cases reported in 2015. These instances included a million cases of CL, 300,000 cases of VL, and around 20,000–50,000 cases of fatal VL.

Geographical distribution of vl

Visceral leishmaniasis is endemic in 75 countries of African, Asian, and American continents, of which 95% of VL cases occur in 10 developing countries. VL predominantly affects five countries: Brazil, East Africa, Bangladesh, Nepal, and India, with approximately 50,000 deaths reported annually to the WHO. VL remains a significant global health concern due to migration, insufficient control measures, and HIV-VL co-infection, which contributes to its increasing incidence. In 2019, ten countries, including Brazil, Eritrea, Ethiopia, India, Iraq, Kenya, Nepal, Somalia, South Sudan, and Sudan, accounted for over 90% of new VL cases (WHO). Globally, 0.7 to 1 million new cases of VL and more than 1 million new cases of CL are reported worldwide. India, Bangladesh, and Nepal bear the 50% burden of all VL cases.171 In India, 54 districts spread over four nearby states namely Bihar, Jharkhand, West Bengal, and Uttar Pradesh. Bihar alone is responsible for 80-90% of all VL cases reported in India and 40% of the global burden.172 Bihar has consistently been the center of VL infections over a century; concurrently, the rate of new HIV infections is rising.173 Sporadic cases of VL are also now reported from other Indian states. Factors such as crowded housing, which facilitates sandfly breeding and resting sites, malnutrition, and population mobility, increase the risk of VL transmission.

Epidemiology of Post-kala-azar dermal leishmaniasis (PKDL) in the Indian Subcontinent

PKDL is endemic in regions where VL is/was prevalent, such as the Indian and sub-Saharan subcontinent. It typically occurs months to years after the apparent cure of VL or without the earlier treatment of VL cases, making surveillance and early detection essential for effective control strategies.174 In the Indian subcontinent, PKDL predominantly affects children and young adults, with a male predominance noted in some studies. The incidence of PKDL varies geographically within endemic regions, with hotspots identified in certain districts and states.175 PKDL, especially (mixed infection) papulo-nodular form serves as a reservoir for Leishmania parasites, contributing to the persistence and transmission of the disease. The parasite load in PKDL lesions is lower than in VL, but patients remain infectious to sandfly vectors, thus perpetuating the transmission cycle.

Leishmaniasis

Leishmaniasis is a parasitic VBD transmitted by the infected female sand flies. It is not a simple disease but a disease complex. The three primary types of leishmaniasis are PKDL, mucocutaneous, and VL. Other names for VL include “Kala-azar, poor man’s disease, and black fever”.176 It is one of the top 20 neglected tropical illnesses as determined by the WHO and a significant global public health concern. Fever usually lasts more than 2 weeks; weight loss, hepatosplenomegaly, and pancytopenia are its defining symptoms.

Leishmaniasis disease types and their symptoms

There are different forms of leishmaniasis based on their occurrences and affected organs in humans. Since the sand fly inoculates these parasites only in the skin while taking their blood meal, the disease manifestation varies depending on parasite nature.

Cutaneous Leishmaniasis

CL primarily impacts the skin, where lesions manifest as a direct consequence of the infection. The skin lesions associated with CL can lead to considerable discomfort and, if left untreated, may result in disfigurement and complications. In India, districts of Rajasthan (Bikaner) and Punjab have experienced the occurrence of CL, a disease attributed to the parasites L. tropica and L. Major.177

Mucocutaneous Leishmaniasis

In Mucocutaneous Leishmaniasis, the infection spreads to the mucous membranes of the nose and oral cavity, resulting in the formation of metastatic lesions that can partially or completely destroy these membranes. If left untreated, the disease can lead to severe disfigurement or even death, primarily due to malnutrition or subsequent bacterial infections.178,179 This is primarily caused by parasite species such as L. braziliensis, L. panamensis, and L. guyanensis, which are commonly found in Central and South America.

Visceral leishmaniasis

VL, commonly known as kala-azar or black fever in the Indian sub-continent, primarily affects the liver, spleen, and bone marrow, leading to symptoms such as irregular fever, weight loss, splenomegaly, hepatomegaly, impaired renal function, and anemia. Greyish or blackish discoloration of the skin is observed in affected individuals.180 Kala-azar is often associated with socio-economic factors and poverty, exacerbated by poor sanitary and housing conditions, as well as malnutrition. The earliest documented cases of kala-azar in India date back to an outbreak of fever (jawar vikar) in Jessore (now in Bangladesh) in 1825, which resulted in approximately 750,000 deaths over three years.181 Periodic outbreaks have been reported in states like Assam, Bihar, Jharkhand, West Bengal, and Uttar Pradesh, with inter-epidemic periods lasting 10–15 years.

The prevalence of kala-azar saw a significant decrease with the implementation of IRS of DDT under the national malaria eradication program in 1950180 due to the reduced population of Phlebotomus argentipes. However, a resurgence occurred in the 1970s when DDT spraying was gradually discontinued due to concerns about its bio-magnification in the environment.182 The removal of insecticide spraying resulted in the occurrence of PKDL, a skin manifestation of kala-azar sequelae and a potential reservoir of VL.

HIV – VL coinfection

Leishmania-HIV co-infection has been recognized since the late 1980s and is reported in over 35 countries worldwide.183 This co-infection compromises immune responses to VL in patients, resulting in higher rates of treatment failure and relapse.184 In Bihar, India, VL-HIV co-infection rates range from 2% to 7% at the village level.185 HIV infection significantly influences the occurrence of VL, with approximately 5-6% of VL cases worldwide occurring as co-infections with HIV.186 In the Mediterranean area, 5-6% of VL cases involve HIV co-infection, while in Ethiopia, approximately 35% of leishmaniasis patients have VL-HIV co-infection.187 In Brazil, the incidence of VL-HIV co-infection rose from 0.7% in 2001 to 8.5% in 2012.188 Both HIV and Leishmania share dendritic cells (DCs) and macrophages, promoting replication and acceleration of VL and HIV. The suppressed immune response in HIV patients is characterized by reduced CD4+ T-cell counts, exacerbating VL manifestation and reactivation.189 Current prevalence rates show HIV infection in VL patients at 9%.9 Socio-economic factors, including migration from rural to urban areas, contribute to the prevalence of VL-HIV co-infection.

Post-Kala-azar Dermal Leishmaniasis (PKDL)

PKDL was reported as a cutaneous consequence of VL.190 PKDL often appears in individuals either concurrently with or more commonly after apparent recovery from VL.191,192 However, there are instances of VL cases where no prior history of PKDL is evident.193

Characteristic features of PKDL include the growth of macules, papules, and nodules, initially around the face but potentially spreading throughout the body if left untreated.194 The time interval between completion of VL therapy and onset of PKDL can vary, occurring during or up to 2-3 years after treatment. PKDL patients may serve as significant sources of infection for the spread of VL.195

Pathogenesis

Establishing any parasitic infection in a host, involves a series of molecular and cellular interactions and events.194,196-200 This also applies to leishmanial infection, where the parasite’s ability to evade killing, enter host macrophages, and survive and multiply inside the cell determines the host’s initial susceptibility to leishmanial challenge.194,196 Leishmania must withstand the activation of the host’s complement system199 and the effects of natural killer cells upon entering the host.201

While attaching to DCs the leishmania parasites typically adhere to host macrophages. Although promastigotes of distinct leishmanial species preferentially attach to macrophages using the polar flagellum, the initial adhesion of Leishmania may be mediated by the pole of the organism.199 Factors such as the parasite surface charge and surface free energy may impact Leishmania’s binding to macrophages.202 It is likely that the initial attachment of promastigotes is mediated by a small ligand, causing clustering of gp63 or lipophosphoglycan (LPG) molecules at a few locations on the parasite’s surface, ultimately leading to the parasite’s engulfment through ligand-receptor contact.203 The ability of L. mexicana mutants lacking either gp63 or LPG to infect macrophages suggests the presence of other ligands.203,204 Numerous receptors, including the mannose-fucose receptor, fibronectin receptors, C-reactive protein receptors, and receptors recognizing specific amino acid sequences like asp-gly-asp (RGD sequence) or integrin type receptors, are involved in parasite binding to the host cell surface.204 Complement receptors (CR1 and CR3) are particularly significant under physiological settings.205

Following complement-dependent adhesion, promastigotes are engulfed by coiled or tubular pseudopods resembling funnels.206,207 Upon ingestion, the parasitophorous vacuole (PV) or phagolysosome 200 forms when promastigote-containing phagosomes fuse with endocytic organelles. The ability to thrive in the acidic environment of the PV, withstand destruction by lysosomal enzymes and other metabolic products, and reproduce are crucial for parasite intracellular survival.208,209 The development of the PV may be delayed by the parasite’s LPG molecules, allowing promastigotes to convert into amastigotes, which are better suited to the enzymes and acidic pH of the PV.205 A tightly packed glycocalyx, primarily composed of low molecular weight glycoinositolphospholipids (GIPLs) and glycosphingolipids derived from the host, covers the surface of Leishmania amastigotes.210 This layer may protect the parasite surface from the hostile environment of the PV. Several enzymes involved in the synthesis of glycocalyx components appear crucial for parasite survival or act as potent virulence factors in amastigotes.211,212 The parasite inside the PV is shielded from intra-phagolysosomal destruction by the proteolytic activity of gp63.213.214

Several protective factors, including cysteine proteases, acid phosphatase,215 magnesium-dependent adenosine triphosphatase (ATPase),215 and others, are released by parasites in soluble form and are associated with LPG.216 Amastigotes multiply within infected cells before escaping to infect additional host cells. The non-filamentous proteophosphoglycan (aPPG) secreted by amastigotes is believed to protect the parasite from complement-mediated destruction and facilitate its entry into macrophages.217 Amastigotes may employ receptors distinct from those used by promastigotes for their entry into host macrophages.218-221

The survival of intracellular parasites within the host cell depends on factors controlling the biochemistry and physiology of the host cell. These factors likely determine the host cell’s ability to eliminate intracellular parasites through respiratory bursts and the production of degradative enzymes or reactive metabolites such as superoxide anion (O2-), H2O2, and nitric oxide (NO). Inducible nitric oxide synthase (iNOS or NOS2) catalyzes the synthesis of NO from L-arginine and molecular oxygen in macrophages, while the release of reactive oxygen intermediates is mediated by the nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase complex.194,222,223 The ability of the host cell to generate reactive oxygen and nitrogen intermediates is diminished when murine or human macrophages are infected with L. donovani.194,224,225

Similar outcomes were observed with pure parasite surface elements such as GIPL, LPG, or gp63, which may contribute to the parasite’s survival inside macrophages. Leishmanial GIPL administered to cells before stimulation with gamma interferon (IFNγ) or lipopolysaccharides inhibited the activation of iNOS in murine macrophages.226-228 LPG or GIPL, on the other hand, synergize with IFNγ to induce iNOS.227

Promastigotes of L. donovani inhibit the phosphorylation and activation of c-Jun-N-terminal kinase (JNK), p38, and p42/44 MAPK, as well as mitogen-activated protein kinases (MAPKs) in host cells. LPG mutant parasite strains are significantly less capable of these actions.197 Early in the leishmanial infection phase, these events result in the inhibition of the iNOS pathway and NO production.205

Survival of the parasite under stress

To avoid reactive oxygen and nitrogen species, parasites have developed defenses against toxic metabolites. Trypanothione and polyamines, which provide thiols, play a vital role in some of these pathways.214,226,228-232 L. donovani reduces macrophage protein kinase C (PKC) activity, prevents phagosome-lysosome fusion, and inhibits IFNγ-induced tyrosine kinase activation, impairing signaling via interleukin 12 (IL-12) and NO generation. IL-1 and tumor necrosis factor-alpha (TNF-alpha) production are downregulated in macrophages infected with L. donovani.227,232,233 Leishmanial infection increases the production of deactivating cytokines such as IL-10 and transforming growth factor-beta (TGFβ).232-236 Some parasite proteins, such as EL-1α and members of the lysosomal cysteine proteinase B family, are transported to the macrophage’s cytoplasm where they interact with host signaling pathways.237 Amastigotes internalize and degrade class II major histocompatibility complex molecules, suppressing macrophages’ ability to present antigens.238 All of these effects and signals differentially influence macrophage activity and its interaction with other cells, contributing to parasite survival in vivo.

Proteome changes in L. donovaniis associated with oxidative and nitrosative stress: A study on the proteome changes associated with L. donovani promastigotes’ adaptation to oxidative and nitrosative stress conditions revealed that the parasite must effectively counteract these stresses through adaptive changes in its proteome. Key proteins involved in stress response and adaptation were identified, including the up-regulation of antioxidant enzymes in response to oxidative stress and alterations in the expression of proteins associated with the detoxification of NO and repair of damaged biomolecules. The identified changes in the expression of metabolic enzymes and transport proteins suggest a reprogramming of cellular metabolism to meet stress adaptation demands and maintain cellular homeostasis under adverse conditions. These findings provide valuable insights into the molecular mechanisms underlying L. donovani adaptation and suggest potential targets for therapeutic intervention against leishmaniasis.239

Metabolic reconfiguration of central glucose metabolism in L. donovaniis crucial for survival during oxidative stress: The Leishmania parasite adapts its central glucose metabolism to counteract oxidative stress, ensuring its survival. This involves glycolysis, the pentose phosphate pathway (PPP), and the tricarboxylic acid (TCA) cycle. During oxidative stress, the parasite upregulates the PPP to generate NADPH, essential for reducing equivalents and antioxidant defenses. The TCA cycle also undergoes modulation to meet increased energy demands and support antioxidant defense mechanisms. The reconfiguration of central glucose metabolism is tightly regulated at multiple levels, with transcription factors like cAMP-responsive element-binding protein playing a crucial role in this adaptation.240

Glucose-6-phosphate dehydrogenase and trypanothione reductase interaction protects leishmania donovani from metalloid-mediated oxidative stress: Protozoan parasite L. donovani faces oxidative stress induced by metalloids during host invasion. The interaction between glucose-6-phosphate dehydrogenase (G6PD) and trypanothione reductase (TR) is crucial in sustaining the antioxidant defense deployed by the parasite against oxidative stress. G6PD catalyzes the PPP, producing NADPH, while TR reduces oxidized trypanothione, a key antioxidant molecule in Leishmania parasites. Metalloid exposure induces oxidative stress, leading to reactive oxygen species (ROS) and oxidative damage. The G6PD-TR complex facilitates the efficient transfer of reducing equivalents between NADPH and trypanothione pools, enhancing the parasite’s ability to neutralize ROS and maintain redox homeostasis. The interaction is tightly regulated to optimize antioxidant activity in L. donovani, providing insights into parasite adaptation to environmental stress and potential therapeutic strategies against leishmaniasis.241

Phosphorylation of Translation Initiation Factor 2-Alpha plays a role in Leishmania donovani’s survival under stress: Under stress conditions, phosphorylation of eIF2α inhibits global protein synthesis, allowing cells to conserve energy and redirect resources towards stress response mechanisms. The fact was investigated that how phosphorylation of eIF2α influences protein synthesis and cellular responses in L. donovani under stress. By inhibiting translation initiation, phosphorylated eIF2α orchestrates a coordinated stress response, promoting parasite survival under adverse conditions. The molecular mechanisms underlying eIF2α phosphorylation in L. donovani explores the role of stress-responsive kinases, including eIF2α kinases (e.g., PKR-like endoplasmic reticulum kinase-PERK), in phosphorylating eIF2α in response to specific stress stimuli encountered by the parasite. Phosphorylation of LdeIF2α enhances L. donovani tolerance to stress by reprogramming gene expression and promoting the synthesis of stress-responsive proteins. It was discussed how eIF2α phosphorylation-mediated translational control enables the parasite to adapt to adverse conditions and survive within the host environment. Overall, the findings highlight the importance of eIF2α phosphorylation in regulating L. donovani survival under stress conditions encountered during infection. By modulating protein synthesis and cellular responses, phosphorylated eIF2α enables the parasite to adapt to adverse environments and persist within the host.242 Further, in the context of host-pathogen interactions, it was explored that L. donovani infection delays the host’s apoptotic process by inducing unfolded protein response in macrophages to ensure its survival and infection establishment.243

Protective role of Mevalonate Kinase in Leishmania donovani against oxidative stress: Mevalonate kinase (MVK) enzyme of the mevalonate pathway helps the Leishmania parasite to survive under oxidative stress. The mevalonate pathway is an important metabolic pathway in trypanosomatids, which leads to the synthesis of various isoprenoid precursors and are crucial for trypanosomatid viability.244 Inhibition of MVK leads to decreased viability and altered morphology. MVK expression was found to be up-regulated under oxidative stress conditions, establishing the role of leishmania donovani mevalonate kinase (LdMVK) in stress tolerance. Ergosterol is a crucial isoprenoid obtained through the mevalonate pathway, which are essentially required for the Leishmania parasites as they cannot survive without it.245 Apart from its role in maintaining cellular integrity, it has also been proven to be essential to overcome the variety of stress such as heat, osmotic, and oxidative stress.246,247 Recent studies showed that L. donovani increases MVK-dependent ergosterol biosynthesis and decreases the peroxidation of cellular lipids when exposed to oxidative stress. LdMVK was found to be associated with maintaining plasma membrane integrity and also in preventing the peroxidation of cellular lipids when exposed to oxidative stress.248 This finding highlights the protective role of MVK in L. donovani against oxidative stress through the modulation of ergosterol biosynthesis. By enhancing the parasite’s antioxidant capacity, ergosterol contributes to its resilience to oxidative damage and promotes survival within the host environment.

Adenylate Cyclase mediated Antioxidant Defense Mechanisms in L. donovani: L. donovani, causing the VL disease in humans, applies its sensory system comprising adenylate cyclase (ACs) to modulate its own as well as the host’s machinery. These ACs localize diversely, such as membrane-bound to a cytosolic location in the cell, and catalyze adenosine triphosphate (ATP) into cyclic adenosine monophosphate (cAMP). L.donovani possibly manages the oxidative environments and its cytotoxic effects exerted either exogenously or endogenously by utilizing both the membrane and cytosol localized adenylate cyclases. Localization of ACs decides the stress responses and cellular activity during the parasite’s developmental stages and drug-exposed stages. Apart from the membrane-bound ACs, the cytosolic AC (LdHemAC) is soluble, and hence, not only preferably controls the PKA-cAMP activity but also induces the alignment of anti-oxidant machineries against the intracellular stress. Unlike membrane bound ACs, which are activated by extracellular signals, its activation occurs by internal factors such as oxidative stress. The recent study using the overexpression construct of LdHemAC signifies its role in infectivity and amastigote survival in murine macrophage. At protein level (LdHemAC), it plays a role in aligning the cAMP-PKA signaling axis to control substrate proteins phosphorylated by PKA under oxidative exposures, and modulates host Heme-oxygeenase-1 (HO-1) to counter oxidative stress.249 Studies focusing in this context are appreciated for new findings and their application for therapeutics or diagnosis value.

Hybrid forms: Hybrid forms in Leishmaniasis are the result of genetic exchange between different strains or species of Leishmania parasites. This phenomenon can occur through genetic recombination, which plays a crucial role in the evolution of Leishmania parasites, contributing to their diversity and potential adaptability to different environments or hosts. It can contribute to increased drug resistance and increased virulence.250

Immunology and host immune response in visceral leishmaniasis

Understanding the host immune response to Leishmania infection is crucial for developing effective therapeutic interventions and vaccines against this disease. The overview of the host immune response to Leishmania infection is summarized below:

Innate immune response: Upon infection, Leishmania promastigotes are phagocytosed by macrophages, DCs, and neutrophils. Recognition of leishmania by pattern recognition receptors (PRRs) such as toll-like receptors (TLRs) and C-type lectin receptors (CLRs) triggers the activation of innate immune responses.251,252 Recent studies have highlighted the role of various signaling pathways, including nuclear factor-κ-B (NF-κB) and MAPK pathways, in modulating the innate immune response to Leishmania infection.253

Adaptive immune response: The adaptive immune response plays a critical role in controlling Leishmania infection. CD4+ T cells, particularly Th1 cells, are pivotal in activating macrophages to kill intracellular Leishmania parasites through the production of IFN-γ and TNF-α.244 Conversely, a Th2-polarized response is associated with disease progression, characterized by IL-4, IL-10, and TGF-β production, which inhibit macrophage activation and promote parasite survival.254 Recent studies have elucidated the role of CD8+ T cells, γδ T cells, and regulatory T cells (Tregs) in modulating the adaptive immune response to Leishmania infection.255 CD8+ T cells contribute to parasite clearance through cytotoxic activity, while γδ T cells exhibit both protective and regulatory functions.256 Tregs, on the other hand, suppress effector T cell responses, thereby promoting parasite persistence and disease progression.

Cytokine and chemokine signaling: The balance between pro-inflammatory and anti-inflammatory cytokines and chemokines determines the outcome of Leishmania infection.257 Recent studies have highlighted the role of chemokines such as C-X-C motif chemokine ligand (CXCL10) and chemokine (C-C motif) ligand (CCL2) in recruiting immune cells to the site of infection and shaping the immune response.258,259 Deregulation of cytokine and chemokine signaling contributes to immune evasion mechanisms employed by Leishmania parasites, facilitating their survival and dissemination within the host.260

Immunomodulatory mechanisms: Leishmania parasites employ various strategies to subvert host immune responses and establish chronic infections. These include inhibition of macrophage activation, interference with antigen presentation, induction of host cell apoptosis, and modulation of host signaling pathways.261 Recent advances in understanding the molecular mechanisms underlying parasite-host interactions have identified potential targets for therapeutic intervention.

Genetic susceptibility to visceral leishmaniasis

Genetic factors play a significant role in determining individual susceptibility to VL. Genome-wide association studies have identified polymorphisms in genes involved in immune regulation, such as cytokines, chemokines, and their receptors, that influence susceptibility to Leishmania infection and disease severity.262 Understanding the genetic basis of susceptibility to VL may aid in the development of personalized therapeutic strategies and vaccination approaches. In conclusion, the host immune response to Leishmania infection is complex and multifaceted, involving innate and adaptive immune mechanisms. Recent studies have provided insights into the cellular and molecular processes underlying immune regulation during VL. Further research aimed at deciphering host-parasite interactions, identifying biomarkers of disease progression, and developing novel therapeutic interventions and vaccines is warranted to combat this neglected tropical disease effectively.

Therapeutic interventions and vaccine development

Current treatment options for VL primarily rely on antimonial drugs, amphotericin B and miltefosine, which have limitations such as toxicity, drug resistance, and high cost. Therefore, there is an urgent need for the development of safer, more effective therapeutic interventions and vaccines against VL. Recent advances in vaccine development include recombinant protein vaccines, DNA vaccines, and live-attenuated vaccines targeting Leishmania antigens.263

First generation vaccine

The first generation of leishmaniasis vaccines involved inoculating live Leishmania promastigotes in attempts to create effective immunization against the disease.114 However, due to the complex and unpredictable nature of the parasite’s antigens, modern vaccine development faces significant challenges. In the 1940s, Brazil initiated the development of these first-generation vaccines, which comprised the entire crude antigen of Leishmania. Despite these efforts, their efficacy was limited.264 Some formulations included autoclaved whole cell lysate, either alone or combined with Bacillus Calmette-Guérin (BCG) as an adjuvant, as the antigen source.265

Second-generation vaccine

Live vaccines

These vaccines use genetically altered Leishmania species that are deficient in critical genes, such as cysteine proteases, biopterin transporters, or centrin. Suicidal cassettes are inserted into the genome alternatively.264,266

Recombinant viruses and bacteria employed as the delivery system for vaccinations Leishmania antigen-expressing recombinant live bacteria and viruses are used as vaccines. L.major GP63 mutants of Salmonella thypymurium, L. amazonensis GP46 mutants of Vaccinia virus, and L.infantum LACK mutants of Vaccinia virus all offered protection.267-269

Purified Leishmania antigen as vaccine

Various Leishmania lipophosphoglycans and sub-fraction proteins have been tested as vaccines, but none have progressed to Phase IIa or Phase III trials. Fucose Mannose Ligand (FML)-saponin270 and LiESAp, which contains a 54-kDa excretory secretory protein with MDP,271 are the canine vaccines that have reached Phase III trial.

Recombinant antigens to be used as vaccine

Candidates for different Leishmania recombinant vaccines have been tested singly, in combination, or as polyproteins and chimaeras. Mice have been observed to benefit from the polyprotein formulations of TSA (thiol-specific antioxidant), Lm STI1 (L. major stress-inducible protein 1), and nucleoside hydrolase. Mice were shielded from both CL and VL by the multicomponent Leish-111f fusion protein, which also contained TSA, LmSTI1, and LeIF (Leishmania elongation initiation factor).272,273 While LCR1 protein and A2 amastigote specific protein afforded protection in VL, a plasmid harbouring L.major LACK, PSA-2 recombinant antigen provided protection against CL.274

Third generation vaccine

DNA vaccines offer increased stability compared to recombinant protein vaccines and stimulate immune responses by engaging innate immune mechanisms. Promising candidates include LACK, LeIF, TSA, LmSTI1, H1, CpA + CpB, KMP11, and NH36. Studies have shown that LACK DNA vaccination provides defense against L. major infection in mice,275 while TSA or LmSTI1 DNA vaccines offer protection against CL. In Balb/c mice, the combination of CPa and CPb with histone plasmids (H2A, H2B, H3, and H4) has also demonstrated protection against L. major. NH36 DNA vaccine shields female hamsters from infection by various Leishmania species, while KMP11 vaccination protects hamsters against visceral leishmaniasis.276,277 Co-administration of three antigenic Leishmania infantum poly-A binding proteins through DNA vaccination induces protection against Leishmania major infection in Balb/c mice. The LEISH-F1+MPL-SE vaccine, a second-generation vaccine, is currently undergoing clinical development for human VL, with recent trials in India evaluating its safety and immunogenicity. Other initiatives, such as the RAPSODI synthetic vaccine, DNA-based LEISHDNAVAX, adenovirus-vectored therapeutic vaccine, and novel European-based VL vaccine projects, are also in progress.278 Furthermore, new adjuvants are being developed, and several clinical vaccine trials are underway or planned. Given the rapid advancements in parasite immunology and genetics, the development of an effective and cost-efficient anti-Leishmania vaccine seems feasible. Increased funding in research and development is essential to advance promising vaccine candidates toward clinical application.

Treatment of VL

Monotherapy

The treatment of leishmaniasis presents various challenges and limitations. Several drugs are utilized for treatment, including sodium antimony gluconate (SAG), Miltefosine (Hexadecylphosphocholine), Paromomycin, Amphotericin B, and liposomal formulations such as AmBisome. However, none of these drugs were found perfect for cure. Each drug has its advantages and disadvantages, particularly concerning adverse effects.

Sodium stibogluconate (SSG)

Historically, antimonials such as SSG and MA have been the mainstays of VL treatment in the Indian subcontinent. These drugs are administered intravenously and have been associated with significant toxicity and the emergence of resistance over time, hence are not in use in the Indian subcontinent. The recommended regimen for SSG is 20 mg/kg/day for 28-30 days.279

Miltefosine

Miltefosine is the only oral drug approved for the treatment of VL. It has been widely used in the Indian subcontinent due to its efficacy and ease of administration. Miltefosine is recommended as the first-line treatment for VL in many endemic regions, including India and Nepal. The standard regimen involves oral administration of miltefosine at a dose of 50 mg/day for adults <25 kg and 100 mg/day >50 kg for 28 days.280 The drug is currently in use for PKDL treatment in Kala-azar elimination.

Amphotericin B and liposomal amphotericin B (LAmB)

For VL treatment, Amphotericin B is recommended at a dose of 0.75-1.0 mg/kg for 15-20 infusions intravenously.281 In place of Amp B, LAmB has emerged as the preferred choice for VL treatment in the Indian subcontinent due to its high efficacy and low toxicity profile. It is recommended as the first-line treatment for VL in India and Nepal, particularly in patients with severe disease, HIV co-infection, or treatment failure. The standard regimen involves intravenous administration of a total cumulative dose of 10 mg/kg body weight.282

Paromomycin

Paromomycin is an aminoglycoside antibiotic with anti-leishmanial activity. It is recommended as a second-line treatment option for VL in the Indian subcontinent, particularly in combination with miltefosine. In combination, the paromomycin is administered intramuscularly at a 15 mg/kg/day dose for 21 days.279

Combination therapy

Combination therapy has been explored as a strategy to enhance treatment efficacy and prevent the emergence of drug resistance in VL. Common combinations include miltefosine plus paromomycin and LAmB plus miltefosine. These combinations have shown promising results in clinical trials and are recommended as alternative treatment options for VL in the Indian subcontinent.279,283

Newer therapeutic agents

Several newer therapeutic agents are under investigation for the treatment of VL, including sitamaquine, aminopyrazoles, and nitroimidazoles. These drugs hold promise as alternative or adjunctive treatments for VL, particularly in cases of drug resistance or treatment failure. However, further clinical trials are needed to establish their safety and efficacy in the Indian subcontinent.284

Treatment of VL-HIV Coinfection: An immunosuppressed host condition

Patients with VL who exhibit higher treatment failure or relapse often present co-infections with other pathogens, such as HIV. Treatment with antimonials in immunosuppressed patients, other than L-AmB, can lead to higher mortality and poor recovery responses. The current recommendation for VL-HIV coinfection involves higher doses of L-AmB administered over multiple days or a single higher dose followed by alternative doses, as per WHO guidelines. Miltefosine monotherapy was preferred over antimonials but showed lower efficacy in Ethiopian studies. Combination therapy with L-AmB and miltefosine demonstrated over 80% cure rates in Ethiopia for both primary and relapsed VL patients.

An alternative approach in VL-HIV coinfection involves administering antiretroviral drugs alongside anti-leishmanial drugs, resulting in decreased coinfection incidence in southern Europe, although relapse rates remain high. Secondary prophylaxis, including L-AmB, antimonials, and pentamidine, is recommended for patients with low CD4 counts after parasitological cure, discontinuing maintenance therapy when CD4 levels rise.285,286

Monthly pentamidine infusions in Ethiopia showed high rates of relapse-free survival among VL-treated patients for one year. Concerns over drug resistance and anthroponotic transmission modes highlight the need for first-line options in secondary prophylaxis. Coinfection of Lymphatic filariasis (LF)/VL has been reported in various regions, necessitating effective therapeutic strategies and further research.

Use of Nanotechnology: A new horizon for treatment of leishmaniasis Chemotherapeutic drugs used in the treatment of VL often come with toxic side effects and increased relapse rates, presenting a critical situation for leishmaniasis elimination. There is an urgent need for safer, more efficient, innovative, and cost-effective therapies for VL treatment. Studies suggest that nanotechnology-based techniques and products could be effective agents for targeted drug delivery to parasites. These techniques include lipophilic vesicles, nano-capsules, and solid-liquid nanoparticles (SLNs) composed of polymers, metals, metallic oxides, and carbon nanotubes. These drug-delivery materials are chemically and biologically suitable for cellular environments, offering higher bioavailability and biodegradability. They exhibit lesser toxicity when used for targeted drug delivery and have emerged as promising anti-leishmanial drugs.287,288

Furthermore, a new nano-vaccine concept has been investigated as a carrier for targeted delivery, adding to the potential of nanotechnology in combating leishmaniasis.

The text has been modified as it is not a conclusion of the article. The treatment of VL in the Indian subcontinent has evolved significantly in recent years, with the introduction of drugs like miltefosine and liposomal amphotericin B. Combination therapy and newer therapeutic agents offer additional options for the management of VL, particularly in cases of drug resistance or treatment failure. However, challenges such as drug resistance, toxicity, and access to treatment remain significant barriers to effective VL control in this region. Continued research and development efforts are needed to optimize treatment regimens and improve outcomes for VL patients in the Indian subcontinent.

Challenges

Treatment of VL-HIV, drug resistance with first-line treatment, and concerns about reduced efficacy of other treatment options, such as miltefosine and amphotericin B, clearly highlight the urgent need for alternative therapies and surveillance of drug resistance patterns.

Mechanism of drug resistance in visceral leishmaniasis

Leishmaniasis, a neglected tropical disease caused by protozoan parasites of the genus Leishmania, remains a significant public health concern, particularly in developing countries with poor socioeconomic conditions and limited access to healthcare services.289 Efforts to control the disease are hindered by the emergence of drug resistance in Leishmania species, which complicates treatment efforts and threatens control programs.290 Drug resistance in Leishmania parasites is a complex phenomenon driven by multiple molecular mechanisms. These mechanisms allow the parasites to adapt and evade the effects of antileishmanial drugs, posing significant challenges to effective treatment. At the molecular level, resistance often arises from alterations in crucial cellular pathways.291 For instance, mutations or dysregulation of genes encoding membrane transporters can impede drug uptake, reducing the intracellular concentration of the drug and compromising its efficacy. Additionally, changes in drug targets, such as mutations that prevent drug binding or modify enzymatic activity, can contribute to resistance. Furthermore, alterations in metabolic pathways and the upregulation of detoxification enzymes may decrease the susceptibility of parasites to antileishmanial agents.292 Understanding these diverse molecular mechanisms is essential for developing targeted therapeutic interventions and combating drug-resistant leishmaniasis effectively. We are discussing here a study that provides a comprehensive examination of the multifaceted resistance mechanisms exhibited by Leishmania parasites against SSG, Amphotericin-B (AmB), AmBisome, Miltefosine (MIL), and Paromomycin, encompassing molecular, genetic, immunological, and epidemiological aspects. At the molecular level, resistance to antileishmanial drugs often originates from intricate changes within the parasite cellular machinery. For example, mutations or disruptions in genes encoding essential membrane transporters such as Aquaglyceroporins (AQPs) can hinder the uptake of SSG, leading to a decrease in its concentration and effectiveness within the parasite.293 Similarly, resistance to AmB and its liposomal formulation, AmBisome, may occur due to mutations in genes responsible for Ergosterol biosynthesis. This reduces AmB’s ability to bind to the parasite’s membrane and disrupt its structure.294 Additionally, an increase in the expression of ATP-binding cassette (ABC) transporters can expel AmB from the parasite, further diminishing its concentration.295 MIL resistance may involve mutations in genes encoding phospholipid translocases, which hinder MIL uptake, as well as heightened expression of efflux pumps that remove MIL from the intracellular space. Changes in lipid metabolism and apoptotic pathways also contribute to MIL resistance, allowing parasites to survive despite MIL exposure.296 Parmomycine resistance likely stems from mutations affecting drug targets or metabolic pathways, resulting in reduced drug efficacy.297 Immunologically, resistance alters host-parasite interactions and immune responses, leading to persistent infections that trigger chronic inflammation and tissue damage. Drug-resistant parasites may modify surface molecules or immune responses to evade clearance, complicating treatment efforts.298 From an epidemiological perspective, drug resistance presents challenges for disease management and control by prolonging infections and increasing the risk of transmission within communities. Effective surveillance is crucial for identifying emerging resistance strains, monitoring their spread, and informing treatment protocols to improve patient outcomes and prevent further transmission. In conclusion, a thorough understanding of resistance mechanisms is essential for developing targeted therapeutic approaches and implementing evidence-based strategies to combat drug-resistant leishmaniasis, effectively.

Resistance to sodium stibogluconate (SSG)

SSG stands as a cornerstone in the treatment arsenal against leishmaniasis, exerting its therapeutic effects through a multi-faceted mechanism of action. Upon administration, SSG infiltrates the Leishmania parasite via specialized transport proteins known as AQPs embedded in the parasite’s cell membrane.299 Once inside the parasite, SSG undergoes metabolic conversion into toxic trivalent antimony (Sb3+) ions, which initiate a cascade of biochemical disruptions crucial for the parasite’s survival.292 This process entails inhibition of glycolysis, fatty acid oxidation, and impairment of antioxidant defense mechanisms, ultimately compromising the parasite’s energy production and leading to cellular damage and death.300 One significant mechanism contributing to SSG resistance revolves around AQPs, specialized transport proteins responsible for facilitating SSG uptake into the parasite. Mutations or downregulation of AQPs can disrupt the influx of SSG, reducing its intracellular concentration and impairing its therapeutic effects.301 Additionally, alterations in the parasite’s redox balance and antioxidant defense systems play a crucial role in conferring resistance. Leishmania parasites possess robust antioxidant mechanisms that neutralize the toxic effects of trivalent antimony (Sb3+) ions, the active form of SSG, thereby diminishing its efficacy.302

Further changes in the parasite’s redox balance and antioxidant defense mechanism contribute to SSG resistance. Leishmania parasites employ antioxidant enzymes to counteract the oxidative stress induced by SSG treatment. Recent studies have highlighted the upregulation of antioxidant enzymes, such as tryparedoxin peroxidases and superoxide dismutases in SSG-resistant parasites, enhancing their ability to neutralize ROSs and evade drug-induced cytotoxicity.303 Moreover, alterations in parasite metabolism, particularly in the PPP, have been implicated in SSG resistance. Recent research suggests that resistant Leishmania strains exhibit increased activity of PPP enzymes, such as glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase, allowing them to cope with the oxidative stress imposed by SSG and sustain parasite growth.304 Moreover, changes in drug efflux pumps and metabolic pathways contribute to SSG resistance. Drug efflux pumps actively expel SSG from the intracellular compartment of the parasite, reducing its accumulation and effectiveness.305 Alterations in metabolic pathways further contribute to resistance by providing alternative routes for the parasite to evade the drug’s toxic effects. These multifaceted alterations collectively confer resistance to SSG and pose significant challenges to effective treatment outcomes.

Mechanism of amphotericin-B and amBisome resistance

AmB and its liposomal formulation, AmBisome, are vital antiparasitic drugs commonly employed in the treatment of life-threatening VL and fungal infections. AmB and AmBisome exert their antifungal and anti-Leishmanial effects through a multifaceted mechanism of action that primarily targets the integrity of the cell membrane. It is a polyene class of agents characterized by their ability to bind selectively to Ergosterol, a crucial component of parasitic cell membranes. Upon binding to ergosterol, AmB forms pores or channels in the lipid bilayer, leading to alterations in membrane permeability and disruption of ion homeostasis. These disruptions ultimately culminate in osmotic instability, leakage of cellular contents, and, ultimately, cell death.306 Furthermore, these drugs may also induce oxidative stress and trigger apoptotic pathways in Leishmania parasites, contributing to their antiparasitic effects. Despite their efficacy, the clinical utility of AmB and AmBisome in the treatment of leishmaniasis is hindered by the emergence of drug resistance. One of the primary mechanisms of resistance to AmB and AmBisome in Leishmania parasites involves alterations in the composition of the parasite’s cell membrane. Mutations or downregulation of genes involved in ergosterol biosynthesis, such as those encoding enzymes like lanosterol 14α-demethylase (CYP51) or sterol 24-methyltransferase, can reduce the availability of ergosterol-like molecules in the membrane. This reduction in ergosterol-like molecules decreases the binding affinity of AmB and AmBisome, impairing their ability to induce membrane permeabilization and compromising their efficacy.307 Additionally, alterations in drug uptake and efflux mechanisms contribute to resistance to AmB and AmBisome in Leishmania parasites. Upregulation of ABC transporters, which actively pump drugs out of the cell, has been implicated in resistance to AmB and AmBisome in Leishmania parasites.308 Similarly, alterations in the expression or function of membrane transporters in Leishmania parasites may contribute to resistance by reducing drug uptake or promoting drug efflux. Moreover, alterations in cellular stress response pathways and the activation of detoxification mechanisms can confer resistance to AmB and AmBisome in Leishmania parasites. Parasites may upregulate genes involved in antioxidant defense systems or activate stress response pathways to counteract the cytotoxic effects of these drugs. Additionally, alterations in apoptotic pathways or DNA repair mechanisms may allow parasites to evade the lethal effects of AmB and AmBisome and survive drug exposure.309 Resistance to AmB not only compromises the efficacy of this vital drug but also limits treatment options for patients, particularly in regions where alternative therapies may be limited or unavailable.310 Therefore, there is an urgent need for continued research to elucidate the molecular basis of AmB resistance and identify novel drug targets for therapeutic intervention.311

Mechanism of miltefosine resistance

MIL, the first oral drug approved for the treatment of leishmaniasis, exerts its antiparasitic effects through a multifaceted mechanism of action. As an alkylphosphocholine compound, MIL acts on various cellular components of the Leishmania parasite, targeting lipid metabolism, membrane integrity, and intracellular signaling pathways.312 MIL disrupts the synthesis of phospholipids, essential components of the parasite’s cell membrane, leading to structural alterations and membrane destabilization.313 Additionally, MIL induces apoptosis-like cell death in Leishmania parasites by triggering mitochondrial dysfunction and the release of apoptogenic factors.314 Furthermore, MIL modulates intracellular signaling pathways involved in cell proliferation and survival, leading to the inhibition of parasite growth and replication.315

One of the primary mechanisms of miltefosine resistance involves alterations in the parasite’s lipid metabolism. MIL interferes with lipid biosynthesis and metabolism in Leishmania parasites by inhibiting enzymes involved in phospholipid synthesis.316 Resistance may arise through mutations or changes in the expression of these enzymes, leading to reduced susceptibility to MIL-induced membrane destabilization. Additionally, alterations in lipid composition, such as changes in the ratio of different lipid species, can affect the efficacy of miltefosine. Another mechanism of MIL resistance involves changes in drug uptake and efflux mechanisms. MIL enters Leishmania parasites primarily through a P-type phospholipid translocase (P-type ATPase) localized in the plasma membrane.317 Mutations or alterations in the expression of this transporter can reduce the intracellular accumulation of miltefosine, thereby decreasing its efficacy. Additionally, the upregulation of efflux pumps, such as ABC transporters, can actively expel MIL from the parasite, further reducing its intracellular concentration and contributing to resistance.318 Furthermore, alterations in intracellular signaling pathways may confer resistance to miltefosine. MIL modulates various signaling pathways involved in cell proliferation, survival, and apoptosis in Leishmania parasites. Resistance may arise through mutations or dysregulation of these pathways, allowing parasites to evade the cytotoxic effects of miltefosine and promote their survival. Moreover, alterations in membrane properties and surface molecules may contribute to miltefosine resistance. Changes in the composition or structure of the parasite’s cell membrane can affect the binding affinity of MIL and its ability to induce membrane disruption. Additionally, alterations in surface molecules involved in drug uptake or binding may impact the efficacy of MIL and contribute to resistance.

Lymphatic filariasis

Background

Lymphatic filariasis is a neglected tropical disease that poses a significant public health burden worldwide. It is primarily transmitted through the bites of infected mosquitoes carrying parasites of the Wuchereria bancrofi, Brugia malayi, and B. timori species. Some species of Anopheles, Culex, Aedes, and Mansonia genera are implicated in the transmission of human filarial parasites. Among these, W. bancrofi accounts for more than 90% of lymphatic filariasis cases globally. Many species of several mosquito genera transmit these parasites, viz. Anopheles, Aedes, Culex, and Mansonia. Adult parasites are primarily found in the lymphatic system of humans and produce several thousands of microfilariae (MF) released in the bloodstream.319 The mosquito vector picks up the MF when feeding on an infected human host. They undergo further development in the mosquito host molting to become infective larvae (L3 stage). When this infected mosquito takes the next blood meal, the infective larvae find their way into another human and develop to become adults. In this way, the life cycle of the parasite continues. The impact of lymphatic filariasis on affected individuals and communities is severe. The disease causes chronic and debilitating conditions such as lymphedema, elephantiasis, and hydrocele. These conditions not only lead to physical disability but also result in social stigma, economic hardship, and reduced productivity. The burden of the disease falls disproportionately on marginalized populations in low-resource settings, further exacerbating health inequities.320

To address this global health challenge, the WHO launched the Global Program to Eliminate Lymphatic Filariasis (GPELF) in 2000. The program aimed to eliminate lymphatic filariasis as a public health problem by 2024 and this timeline for elimination has been extended to 2030 now.321 GPELF adopted a comprehensive approach that combined mass drug administration (MDA) with other preventive measures such as vector control, morbidity management, and disability prevention. MDA involves the administration of antiflarial drugs, including Albendazole, Ivermectin, and Diethylcarbamazine (DEC), to entire at-risk eligible populations in endemic areas. The MDA-ineligible section of the population includes children below a certain age (<2 years in case of DA-MDA and <5 years in case of IDA-MDA), pregnant women, and severely ill.322 These drugs target the MFs, the larval stage of the parasites circulating in the bloodstream, and aim to reduce their numbers, prevent transmission, and alleviate symptoms. The success of MDA campaigns in reducing disease burden has been significant, and several countries have made remarkable progress toward elimination goals. However, challenges remain on the path toward the complete elimination of lymphatic filariasis.323 One of the primary challenges is the persistence of adult worms in infected individuals for years, as the currently available drugs primarily target MF. These adult worms can continue to produce MF, perpetuating the transmission cycle of the disease. At present, there is no potent macrofilaricide drug that can penetrate lymphatics and kill adult worms.324,325

Additionally there are concerns about the emergence of drug resistance in the parasite populations. There have been some reports of poor responses to DEC treatment as well as to Ivermectin. Moreover, a mutation associated with resistance to Albendazole has been detected in W. bancrofti populations. Such challenges could impact the effectiveness of MDA interventions. Innovative strategies are needed to overcome these challenges and achieve sustained elimination.323,326-328

The burden and global programme to eliminate lymphatic filariasis (GPELF)

LF is endemic in 73 countries and territories of the tropical and subtropical regions of the globe, with 1.39 billion people at risk of developing this debilitating disease. In India, LF continues to be an important public health problem contributing about 44.3% to the global burden. To eliminate LF, combined drug regimens of antifilarial drugs are being mass administered to populations living in at least 48 endemic countries. At least 36 million people were suffering from chronic disease manifestations, and among them, 25 million men were affected with hydrocele and 15 million with lymphedema. In India, over 670 million people are at risk of infection by W. bancrofti and B. malayi parasites in 339 districts. Infection due to W. bancrofti is widespread in 21 states and UTs, contributing to about 99.4 percent of the total cases. The remaining cases are due to B. malayi, being limited to small pockets in six endemic States. About 0.48 million cases of lymphoedema and 0.18 million cases of hydrocele are line-listed in the endemic districts of the country.324

In 1997, the WHO, via the World Health Assembly Resolution, WHA 50.29, pledged to eliminate LF as a public health problem by 2020. The GPELF, launched in the year 2000, has two principal aims: (i) to interrupt LF transmission and (ii) to manage morbidity and prevent disability. The overwhelming success of the GPELF, so far, has led to a significant decrease in MF and antigenemia prevalence in several countries that have accomplished several rounds of MDA.324,325

The current LF elimination strategy adopted in India

The Government of India prioritized the elimination of LF through the annual MDA program in 2004 and continued with a single dose of diethylcarbamazine citrate (DEC), 6 mg/kg of body weight, plus albendazole annually over 5-6 years. The GOI had set the target to achieve LF elimination by 2015 and now by 2030. In India, as many as 138 districts out of 339 endemic districts have stopped MDA in Gujarat, Goa, Puducherry, Dadra Nagar Haveli, Daman & Diu, Tamil Nadu, Lakshadweep, Andaman & Nicobar Islands. An additional 30 districts are under TAS evaluation. In 171 districts where MDA is implemented, 108 are under DA-MDA, while 63 are under IDA-MDA. In 2023, to accelerate the elimination of LF, the Govt. divided the country such that roughly half of the endemic districts will receive MDA on February 10th, coinciding with National Deworming Day and the remaining districts on August 10th. This division is expected to manage the supply chain and implementation of MDA more effectively.329,330

Despite several rounds of MDA, the progress of LF elimination in the country so far has been suboptimal. Much remains to be achieved as about 50.4% of the total 339 endemic districts are still under MDA. The major challenge in implementing MDA is poor compliance and drug consumption. It is necessary to have a critical look at the causes of slow progress and provide alternatives to address many challenges that are impeding LF elimination in India.331

The challenges and impediments to LF elimination

The Govt. of India is making concerted efforts with the help of various stakeholders, drug donors, and funding partners to eliminate LF by the targeted year 2030. Some of the challenges have been mentioned below and steps have been suggested for consideration by the Govt. to further accelerate the elimination efforts.

Delineation of endemic blocks/districts of the country

From time to time, the addition of the new LF endemic districts to the existing pool is worrisome. That suggests a need to scientifically delineate all the endemic districts in which the status of LF is uncertain in the country. To identify them, especially those bordering the existing MDA districts, systematic surveys to line list the hydrocele and lymphedema cases in the villages and the cities are necessary through Wellness Centres. Additionally, Night blood surveys carried out will inform the prevalence of infection (MF rate) in different blocks of the districts. When above the 1% threshold, the MF rate will also be an indicator of ongoing transmission of the disease and the inclusion of these blocks for the MDA. As currently, block is the intervention unit it is emphasized that the focus of MF surveys should be the blocks, especially those in which a sizable number of chronic cases are present.329,330,332

The challenges with diagnosis and diagnostics

This is a very important issue. Diagnostics conforming to different use cases with high sensitivity and specificity are the cornerstones for determining LF status during different phases of elimination and during sustenance monitoring in the post-elimination phase. The following are the diagnostics and their use cases.

  • Microscopy – detection of MF in the night blood sample – a very useful measure of the prevalence of the source of infection for monitoring the impact of interventions and for active case detection and treatment.

  • Diagnostic test to detect circulating filarial antigens – a sensitive and operationally feasible tool useful for mapping, monitoring, evaluating the impact, stopping decisions, and post-elimination surveillance.

  • An antibody assay that can detect filarial antibody as a marker of exposure to infection – useful to demonstrate active transmission.

Microscopy for examining stained night blood smears is considered a gold standard method to detect MF. However, there are challenges with night blood surveys, which usually start after 9 PM and normally end by midnight when the MF appears in the peripheral blood and is captured in the blood smears. The timing for making a blood smear is critical, which is close to midnight to determine the true MF prevalence. Equipping the wellness center with a microscope, staining facility, and trained staff in all the endemic blocks is a basic requirement in every endemic block in the country.333-335

As for an antigen test, currently, an Alere Filariasis Test Strip (FTSTM) is imported and costs about Rs. 400 per test. The total cost is very high considering the large number of tests needed in our country for post-MDA monitoring and Evaluation and for making stop-MDA decisions. Hence, concerted efforts are needed either to import this technology and manufacture antigen detection tests under a license in India or to take steps to develop indigenous tests.329

A typical filarial antibody detection test is an IgG/IgM Rapid Test Cassette (Whole Blood/Serum/Plasma) that is a rapid chromatographic immunoassay for the qualitative detection of IgG and IgM antibodies to Filariasis parasites (W. Bancrofti and B. Malayi) in whole blood, serum, or plasma to aid in the diagnosis of Filariasis infection. These tests should have a sensitivity and specificity of >95%. Antibody tests are useful as exposure markers in the detection of new disease foci and useful during the post-elimination validation surveys. Indigenous development of these tests should be encouraged.335

Mass Drug Administration (MDA): Challenges to meet the elimination thresholds and stopping MDA strategy

Currently, MDA is the main LF elimination strategy globally and in the country. Two types of MDAs have been rolled out. In 108 endemic districts, a combination of DEC and Albendazole (DA) combination is being used during MDA campaigns, while in 63 districts Ivermectin+Diethylcarbamazine+Albendazole (IDA) combination is being administered. For DA and IDA to be effective, a minimum of 65% and 85% consumption of drugs must be achieved during the MDA campaigns every time. As per the modeling studies, 5-6 rounds of DA and 2-3 rounds of IDA are enough to achieve the LF elimination targets, i.e., <1% Mf rate and <2% filarial antigenemia provided the above coverage is achieved. This is followed by Transmission Assessment Surveys (TAS) in DA-MDA districts wherein antigenemia in children of 6-7 years of age who have lived in areas with 5-6 MDA rounds. The TAS is conducted in the schools following a statistical sampling tool. To stop MDA, the district or block must pass three consecutive transmission assessment surveys as per the guidelines of WHO and NCVBDC.329,336-338 By 2020, as many as 98, 87, and 42 endemic districts had cleared 1st, 2nd, and 3rd transmission assessments, respectively, and stopped MDA. However, the districts that failed TAS 2 and 3 after having passed TAS 1 & 2, respectively, were reverted to MDA. It is noteworthy that DA-MDA was started in 2004. Hence, some districts have undergone 18-19 rounds of MDA and yet not passed TAS suggesting that the desired level of drug consumption targets during these MDA campaigns were not achieved. For IDA-MDA an effective monitoring and evaluation strategy is needed as per WHO.329 WHO recommends the FTS for all areas endemic to W. bancrofti and Brugia Rapid Test for all areas endemic to Brugia spp. The FTS, which measures circulating filarial antigen (CFA), is used in all steps of the GPELF strategy. However, CFA can take 12 months or more to appear after infection and persists several years after adult worms can no longer reproduce or have died. New tools are needed to detect, ideally, the presence of viable worms or MF following the introduction of IDA. WHO suggests community surveys and hence all levels of the population for post-MDA assessment in case of IDA-MDA.338,339

Drug consumption

One challenge to achieving LF elimination is the persistent coverage-compliance gap during annual MDA and the risk of ongoing transmission among never-treated individuals. Ideally, 5-6 rounds of DA-MDA should suffice to meet the elimination threshold provided a minimum level of drug consumption in case of DA (65%) and IDA (85%) are achieved. The biggest challenge faced currently is how to improve drug consumption rates in the MDA blocks. Innovative strategies are needed to make the community MDA-compliant so that the actual consumption is above the threshold. Convincing a non-diseased person to consume a handful of drugs becomes challenging for the health workers leading to drug non-consumption and wastage. One of the ways to improve drug compliance could be fixed-dose combinations of the three drugs into a single or a few tablets. Efforts to reassure community members about adverse events, specific instructions on how to take LF drugs, and improving awareness that MDA participation is part of one’s contribution to promoting community health are essential drivers for the uptake of LF drugs during MDA.331,339,340

Systematic non-compliant

There are about 10% population that is out of the ambit of MDA, like children below 2 years (IDA) and 5 years (DA), pregnant women, and severely ill individuals who are not administered anti-filarial drugs. Hence, meeting the drug consumption targets becomes stiffer.

Migrants

A sizable population migrates out of LF endemic districts to other parts of the country for livelihood and is not available during MDA to receive anti-filarial drugs. These include mostly males between the age groups of 20-50 years. Since they are absent for long periods, they miss drug administration during the mop-up round also. Additionally, many persons are engaged in agriculture, business, industry, etc. who are absent from home during the MDA campaign. These people must be covered during the mop-up rounds to improve drug consumption rates.329

Never treated

It is now recognized that a section of persons does not consume anti-filarial drugs even after several rounds of MDA. The term “never treatment” is used to describe individuals who report never swallowing LF drugs during any MDA rounds. There is poor evidence at present on the proportion of “never treated” out of those who do not consume anti-filarial drugs willfully.

Space for molecular xenomonitoring (MX)

The success of the GPELF through MDA hinges on the availability of sensitive tools to monitor the outcome of the elimination efforts. The current challenge is to have appropriate diagnostic tools and time for cessation of MDA. The decision to stop MDA is complex, and multiple decision support tools have been reported for arriving at the decision. The interruption of transmission of infection is presently being monitored using human infection measures such as MF loads, antigenemia, anti-filarial antibodies in humans, and infection in vectors. Transmission, a key parameter in this regard is a function of both the prevalence of mosquitoes with infective-stage larvae and the man-biting rate and can be monitored by measuring changes in infection status of either vectors or humans. It has two components: transmission from man to mosquito vector (infection) and transmission from the mosquito vector to the human host (infectivity). Direct detection of MF/larval stages of the parasite in the vector is indicative of both the presence of patent (circulating microfilariae) infections in humans and transmission of the infection from humans to the vector.341 Detection of larvae of filarial parasites in vectors by dissection and microscopic examination has been the gold standard method to estimate the transmission levels in endemic settings. However, this method is cumbersome, subjective, has low throughput and is not applicable in areas with ultra-low parasite prevalence in vectors. These attributes make this method unsuitable for use in assessing large-scale programs, such as GPELF. MX and PCR detection of filarial DNA in mosquito vectors are highly sensitive and less invasive tools that help indirectly detect filarial infection in communities. Hence, efforts have been made to develop PCR-based methods for the detection of infection and infective (L3) stage larvae of W. bancrofti and B. malayi. A simple and inexpensive technique for the isolation of DNA from infected mosquitoes has been developed in the ICMR-Vector Control Centre (VCRC) and found to be useful in xenomonitoring of LF. Further, the MX for W. bancrofti developed at VCRC has been evaluated in several geographic locations in India along with other parameters (microfilaria, circulating filarial antigen, and antibody) and found to be 2-3 times more sensitive than the mf survey. The technique utilizes a simple and inexpensive buffer which is routinely used in all molecular biology laboratories and bead beating. The cost of isolation of DNA from vectors by employing this method is a small fraction (US $ 0.06) of that by the published method that utilizes commercial kits (US $ 5.0). This cost-saving will bring down the cost of xenomonitoring immensely which is vital for source-constrained endemic countries currently implementing global programmes like GPELF.335,339 It may be noted that MX for LF has remained largely within the realm of research laboratories for almost 2 decades. The LF elimination programme, launched 2 decades ago, has now reached advanced stages in many countries with several rounds of MDA administered. Now it is the time to deploy the MX in the LF elimination programs globally, through establishing the assay for monitoring the transmission for stopping MDA in an endemic setting and finally proceed to certification of LF elimination in the country.340

DEC fortified salt as an alternate strategy

At this juncture, a well-researched approach, i.e. the use of DEC-fortified salt, also advocated by the WO, as a unique form of MDA, is proposed. As per this strategy, a low dose of DEC (0.2% w/w) is added to the cooking salt at the manufacturing facility of iodized salt and consumed by the LF-endemic communities for about two years. Many examples of successful use of this strategy for LF elimination in small- and large-scale trials have been documented in India and several other endemic countries in the world. Implementing DEC–iodine-fortified salt is a safe, less expensive, more efficient, and prompt approach for achieving the elimination of LF in India. Adverse effects are either absent or minor and self-limiting. The DEC-fortified salt strategy can easily piggyback on the existing countrywide deployment of iodized salt under the National Iodine Deficiency Disorders Control Programme (NIDDCP), which has achieved great success in reducing iodine-deficiency disorders such as hypothyroidism. This existing robust program can be leveraged to launch DEC-fortified salt for the community. If implemented appropriately, this strategy may ensure the complete cessation of LF transmission within two years from its introduction. A detailed and in-depth blueprint may be needed to implement this strategy considering the technical, ethnic, political, cultural, social, and ethical issues related to DEC-fortified salt introduction for LF elimination.342-345

Prospects of a LF vaccine

Extensive research efforts have been dedicated to identifying suitable vaccine candidates using various approaches, including subtractive genomics and reverse vaccinology. Subtractive genomics involves comparing the genomes of the parasite with the host organisms to identify unique proteins or metabolic pathways that could be potential targets for interventions. This approach helps in prioritizing proteins essential for the survival or reproduction of the parasite, that could be explored to develop potential vaccines or new drug therapies. Reverse vaccinology complements subtractive genomics to predict immunogenic epitopes and design and evaluate multi-epitope vaccines using various immune-informatics tools. This computational approach aids in the selection of vaccine candidates with the highest potential for success, minimizing the need for extensive experimental testing. So far development of a vaccine against LF has been a neglected area, with practically little or no research being undertaken to develop an effective vaccine that will have far-reaching implications on disease prevention for billions at risk of LF.343-348

Space for vector control

Vector Control in LF is another neglected area due to the lack of sufficient vector control capacity in the country. Most of the W. bancrofti infections are transmitted by the Culex quinquefasciatus which is a predominant mosquito species in both rural and urban India. This mosquito prefers polluted man made water bodies such as drains, septic tanks, and ditches. These sites can be managed by proper sanitation, routine cleaning, and drainage facilities. It is noted that in the villages, box-type concrete drains are being constructed, which lack gradients, as a result, the wastewater stagnating in them creates ideal conditions for Culex quinquefasciatus breeding. Hence, MDA campaigns are carried out to control the parasite, while little to no effort is directed at the control of vector mosquitoes, which unabatedly transmit the disease. Hence, there is a need for introducing integrated vector management (IVM) which is recommended by the WHO and advocated by the NVBDCP. This will need a multipronged approach, including identification of stubborn LF pockets needing IVM, making species sanitation as part of the ‘Swachh Bharat Mission’ of Govt. of India and improvements in rural and urban drainage systems and advocating LLINs in the endemic blocks to prevent/minimize vector–human contact.332,334,339,349-352

VECTOR BORNE BACTERIAL DISEASES

Background: Vector-borne bacterial diseases pose a significant public health challenge, transmitted primarily through the bites of arthropods such as mosquitoes, fleas, mites, and ticks. The transmission of these diseases is closely linked to environmental factors like climate change, deforestation, urbanization, and poor sanitation, which create favorable conditions for vector proliferation. Outbreaks are more frequent during the monsoon season when stagnant water and increased humidity support vector breeding. Despite advancements in healthcare, these diseases remain endemic in many regions, highlighting the need for strengthened vector control measures, early diagnosis, and targeted public health interventions.353

Lyme disease

Lyme disease is a tick-borne bacterial infection. It is caused by Borrelia burgdorferi and transmitted primarily by Ixodes ticks, commonly known as deer ticks or black-legged ticks. It is characterized by a wide range of symptoms, from localized skin lesions to systemic complications. It is more prevalent in North America and Europe, and cases have been increasingly reported in Asia, including India, especially in regions with high tick activity.354

Epidemiology

Lyme disease is not endemic in India; sporadic cases have been identified, especially in the northern regions, such as Himachal Pradesh, where tick populations are abundant due to forested areas and livestock farming. The disease is more common in cooler climates where ticks thrive. Individuals working in forestry, agriculture, or animal husbandry are at higher risk of exposure to infected ticks.355

Clinical symptoms

Lyme disease progresses through three stages: early localized, early disseminated, and late disseminated.356 Early Localized Stage: The hallmark symptom is erythema migrans, a circular, “bullseye” rash that appears at the site of the tick bite. This occurs within 3 to 30 days after infection. Other early symptoms include fatigue, fever, headache, and muscle or joint pain. Early Disseminated Stage: If untreated, the infection spreads to other parts of the body. Symptoms may include multiple rashes, facial palsy (loss of muscle tone on one or both sides of the face), meningitis, and shooting pains. Late Disseminated Stage: Months to years after infection, untreated patients may develop severe arthritis, especially in the knees, and neurological complications such as neuropathy or cognitive dysfunction may appear.356,357

Diagnosis and management

Lyme disease can be diagnosed through clinical observation, particularly the presence of erythema migrans, and confirmed using serological tests such as enzyme immunoassay (EIA) and Western blot to detect antibodies against Borrelia burgdorferi. Due to a lack of awareness, cases are often misdiagnosed as other tick-borne illnesses.358

Treatment in the early stages involves antibiotics like doxycycline or amoxicillin. In the later stages, intravenous antibiotics may be required to treat severe neurological or cardiac complications.358

Challenges

The lack of awareness among healthcare professionals and the public may lead to misdiagnosis, as the symptoms can resemble other febrile illnesses. The presence of Ixodes ticks in certain northern regions poses a potential public health threat, particularly as climate changes alter tick distribution patterns. Lack of prevention efforts, such as tick control and public education, can make it difficult to reduce exposure.359

Scrub typhus

Scrub typhus is a mite-borne bacterial disease caused by Orientia tsutsugamushi. It is transmitted through the bite of infected chigger mites, commonly found in areas with dense vegetation, such as forests, farmlands, and grasslands.360 The disease is characterized by fever, rash, and multi-organ involvement, and it is a significant cause of acute febrile illness in rural India. Scrub typhus has a long history in India, especially in the Himalayan region, northeastern states, and southern parts of the country.361

Epidemiology

Scrub typhus is endemic in the “tsutsugamushi triangle,” which includes South and Southeast Asia, parts of China, and northern Australia. In India, the disease is widespread in rural and forested areas, particularly in Himachal Pradesh, Uttarakhand, Jammu & Kashmir, and Assam, as well as parts of southern India like Tamil Nadu and Kerala. The disease re-emerged as a major public health concern in India in the early 21st century, with outbreaks frequently reported during the monsoon and post-monsoon periods when chiggers are most active.362,363

Clinical Symptoms

Symptoms typically begin 6 to 21 days after being bitten by an infected mite. The disease presents with high-grade fever, eschar, rash, and systemic involvement in severe cases. Scrub typhus can lead to complications such as pneumonia, myocarditis, meningoencephalitis, acute renal failure, and multi-organ dysfunction. Without treatment, the disease can progress to severe complications and can be fatal, with a mortality rate ranging from 7% to 30%.363,364

Diagnosis and Management

Scrub typhus is diagnosed based on clinical symptoms, particularly in endemic areas, and confirmed by serological tests such as the Weil-Felix test, indirect immunofluorescence assay (IFA), or ELISA.365

Doxycycline is the treatment of choice and is highly effective if started early. In some cases, azithromycin is used as an alternative, especially in children and pregnant women. Prompt treatment with antibiotics significantly reduces the risk of complications and death.366

Challenges

Lack of awareness contributes to delayed diagnosis and treatment. Seasonal outbreaks are common, particularly during the monsoon when the vector population increases. Public health initiatives focused on vector control and educating rural populations on protective measures, such as avoiding areas with dense vegetation and using insect repellents, are essential to controlling the spread of scrub typhus.

Recommondations and way forwards

To address the critical gaps identified in the context of VBDs in India, it is essential to foster innovative research initiatives. The following recommendations are:

  • Establish interdisciplinary research networks that bring together experts from public health, epidemiology, entomology, environmental, and social sciences to develop holistic approaches to VBDs.

  • Collaborate with local communities and stakeholders to incorporate indigenous knowledge and practices into research design and implementation.

  • Conduct comprehensive studies on the ecology and behavior of vectors, including resistance patterns to insecticides and the impact of climate change on vector populations.

  • Deploy smart traps equipped with sensors to monitor vector populations and provide data on their behavior, aiding targeted control measures.

  • Utilize advanced genomic and proteomic technologies to identify novel targets for vector control and to better understand the molecular mechanisms of vector-pathogen interactions.

  • Utilize next generation sequencing to analyze the genetic makeup of VBD pathogens, aiding in the development of targeted vaccines and treatments.

  • Invest in the development of innovative, point-of-care diagnostic tests that are rapid, affordable, and suitable for use in low-resource settings.

  • Research into multiplex diagnostic platforms that can detect multiple VBDs simultaneously, improving efficiency in disease management.

  • Explore the use of novel vaccine platforms, such as mRNA and viral vector technologies, for the rapid development of vaccines against emerging VBDs.

  • Facilitate the establishment of clinical trial networks to expedite the testing of new vaccines and therapeutics in diverse populations.

  • Explore novel delivery mechanisms (e.g., microneedles, nasal sprays) to enhance vaccine uptake and efficacy in diverse populations.

  • Research the behavioral factors influencing the transmission of VBDs and assess community engagement in prevention efforts.

  • Investigate effective health communication strategies to promote awareness and adherence to prevention measures among at-risk populations.

  • Focus on implementation science to evaluate the effectiveness of current vector control measures and public health interventions in real-world settings.

  • Research how to scale up successful interventions and integrate them into national health programs, ensuring sustainability and broad reach.

  • Leverage big data analytics and artificial intelligence to analyze epidemiological trends and predict outbreak patterns, aiding in proactive public health responses.

  • Develop and test mobile health applications for real-time reporting and data collection on VBDs from community health workers and the public.

  • Employ social media analysis tools to monitor public discussions and concerns regarding VBDs, enabling timely responses to misinformation and community engagement.

  • Advocate for increased public and private funding dedicated to innovative research on VBDs, ensuring a sustained commitment to tackling these diseases.

  • Invest in training programs and infrastructure to enhance research capacity within local institutions, fostering a sustainable research ecosystem.

  • Establish networks for sharing knowledge and best practices in the application of novel tools for vector-borne disease research and control.

  • Create feedback mechanisms for communities to report vector-related issues and disease outbreaks, ensuring a responsive public health approach.

  • Create an implementation framework to monitor and evaluate adherence to the guidelines, ensuring continuous improvement based on feedback and emerging evidence.

  • Establish a schedule for regularly reviewing and updating the guidelines to reflect new research findings, epidemiological trends, and advancements in technology.

Implementing these recommendations, India can significantly enhance its capacity to combat VBDs, ultimately leading to improved public health outcomes and improving the quality of life for Indian citizens.

Acknowledgment

We would like to express our sincere gratitude to various research institutions, universities, and public health organizations that provided essential data and research findings, enabling us to present a comprehensive overview of the current situation regarding VBDs in India. Thank you to everyone who has played a role in this endeavor. Together, we can make significant strides in combating VBDs and promoting public health in India.

References

  1. , . Climate change: an enduring challenge for vector-borne disease prevention and control. Nat Immunol. 2020;21:479-83.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  2. , . Climate change and infectious disease in europe: Impact, projection and adaptation. Lancet Reg Health Eur. 2021;9:100230.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  3. , , . Mosquito-borne diseases in India over the past 50 years and their global public health implications: A systematic review. J Am Mosq Control Assoc. 2023;39:258-77.
    [CrossRef] [PubMed] [Google Scholar]
  4. National Centre for Vector Borne Diseases Control, Ministry of Health & Family Welfare, Government of India. Dengue situation in India. Available from: https://ncvbdc.mohfw.gov.in/index1.php?lang=1&level=1&sublinkid=5776&lid=3690 [Last accessed 2024 August 16].
  5. National Centre for Vector Borne Diseases Control (NCVBDC), Ministry of Health & Family Welfare, Government of India. (2023). Chikungunia situation in India. Available from: https://ncvbdc.mohfw.gov.in/index1.php?lang=1&level=2&sublinkid=5967&lid=3765 [Last accessed 2024 August 25].
  6. , , , , , , et al. Zika a vector borne disease detected in newer states of India amidst the COVID-19 pandemic. Frontiers in Microbiology.. 2022;13:888195.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  7. , , , , , , et al. Time to implement tailored interventions in chhattisgarh, east-central India to reach malaria elimination. J Vector Borne Dis. 2024;61:151-7.
    [CrossRef] [PubMed] [Google Scholar]
  8. World Health Organization, Leishmaniasis, 2020 https://www.who.int/health-topics/leishmaniasis [Last accessed 2024 Apr 20].
  9. National Centre for Vector Borne Diseases Control, Ministry of Health & Family Welfare, Government of India. .Magnitude of Diseases, Filaria. Available from: https://ncvbdc.mohfw.gov.in/index4.php?lang=1&level=0&linkid=455&lid=3732 [Last accessed 2024 August 16].
  10. , , , . Larvicidal property and active compound profiling of annona squamosa leaf extracts against two species of diptera, aedes aegypti and anopheles stephensi. J Vector Borne Dis. 2023;60:401-13.
    [CrossRef] [PubMed] [Google Scholar]
  11. , , , , . Vector-borne diseases in central India, with reference to malaria, filaria, dengue and chikungunya. WHO South-East Asia journal of public health. 2014;3:28-35.
    [CrossRef] [PubMed] [Google Scholar]
  12. , , , . Trends in emerging vector-borne viral infections and their outcome in children over two decades. Pediatric Research. 2024;95:464-479.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  13. National Centre for Vector Borne Diseases Control, Ministry of Health & Family Welfare, Government of India. Diseases. Available from: https://ncvbdc.mohfw.gov.in/index4.php?lang=1&level=0&linkid=407&lid=3683 [Last accessed 2024 August 25].
  14. . Dengue: guidelines for diagnosis, treatment, prevention and control. World Health Organization; . Available from: https://www.who.int/publications/i/item/9789241547871 [Last accessed 2024 Apr 20]
  15. World Health Organization “Disease Outbreak News; Dengue – Global situation; 2024. Available from: https://www.who.int/emergencies/disease-outbreak-news/item/2024-DON518 [Last accessed 2024 Apr 20].
  16. National Centre for Vector Borne Diseases Control (NCVBDC), Ministry of Health & Family Welfare, Government of India. Dengue. Available from: https://ncvbdc.mohfw.gov.in/index1.php?lang=1&level=1&sublinkid=5776&lid=3690 [Last accessed 2024 August 20].
  17. , , , , , , et al. Serotype and genotype diversity of dengue viruses circulating in India: A multi-centre retrospective study involving the virus research diagnostic laboratory network in 2018. Int J Infect Dis. 2021;111:242-5.
    [CrossRef] [PubMed] [Google Scholar]
  18. , , , , . Genetic characterization of dengue virus types 1 and 2 in India, with emphasis on the viruses circulating in karnataka. Indian J Med Res. 2017;146:662-5.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  19. , . Current perspectives on the spread of dengue in India. Infect Drug Resist. 2014;7:337-42.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  20. , , , , , . Continuous circulation of DENV-3 genotype 3 in Delhi: A single hospital based surveillance study. medRxiv 2020 2020.04. 25.20078899
    [Google Scholar]
  21. NCVBDC, Dengue situation in India https://ncvbdc.mohfw.gov.in/index4.php?lang=1&level=0&linkid=431&lid=3715 [Last accessed 2024 Apr 22].
  22. , , , , , . Burden of dengue, leishmaniasis and lymphatic filariasis in India and its states from 1990–2019: Analysis from the global burden of disease study (GBD 2019) PLoS ONE. 2023;18:e0292723.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  23. , . A retrospective study of environmental predictors of dengue in delhi from 2015 to 2018 using the generalized linear model. Sci Rep. 2022;12:8109.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  24. World Health Organization Diseases outbreak News-Zika Virus Disease – India. 2021. https://www.who.int/emergencies/disease-outbreak-news/item/zika-virus-disease-india [Last accessed 2024 Apr 25].
  25. . The resurgence of dengue epidemic and climate change in India. Lancet. 2023;401:727-8.
    [CrossRef] [PubMed] [Google Scholar]
  26. , . Dengue detection: Advances and challenges in diagnostic technology. Biosensors and Bioelectronics: X. 2022;10:100100.
    [CrossRef] [Google Scholar]
  27. , , , , , , et al. Evaluation of diagnostic tests: Dengue. Nat Rev Microbiol. 2010;8:S30-8.
    [CrossRef] [PubMed] [Google Scholar]
  28. , , , . Use of the tourniquet test in diagnosing dengue-like syndrome. Bulletin de la Société de pathologie exotique.. 2012;105:361-3.
    [PubMed] [Google Scholar]
  29. , , , , , , et al. Evaluation of dengue NS1 antigen rapid tests and ELISA kits using clinical samples. PLoS One. 2014;9:e113411.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  30. , , , , , . Performance evaluation of a rapid dengue NS1 antigen lateral flow immunoassay test with reference to dengue NS1 antigen-capture ELISA. Journal of Clinical Virology Plus. 2023;3:100144.
    [CrossRef] [Google Scholar]
  31. , , , , , , et al. Diagnostic accuracy of commercially available immunochromatographic rapid tests for diagnosis of dengue in India. J Vector Borne Dis. 2021;58:159-64.
    [CrossRef] [PubMed] [Google Scholar]
  32. , , , , , , et al. Sensitivity and specificity of DPP® fever panel II asia in the diagnosis of malaria, dengue and melioidosis. J Med Microbiol. 2022;71:001584.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  33. , , , . Can rapid dengue diagnostic kits be trusted? A comparative study of commercially available rapid kits for serodiagnosis of dengue fever. J Lab Physicians. 2019;11:63-7.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  34. , , . Management of dengue: an updated review. Indian Journal of Pediatrics.. 2023;90:168-77.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  35. NCVBDC. National Guidelines for Clinical Management of Dengue Fever. https://ncvbdc.mohfw.gov.in/Doc/National%20Guidelines%20for%20Clinical%20Management%20of%20Dengue%20Fever%202023.pdf [Last accessed 2024 Apr 26].
  36. , , . Molecular mechanisms of antiviral agents against dengue virus. Viruses. 2023;15:705.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  37. , , , , . Traditional knowledge to contemporary medication in the treatment of infectious disease dengue: a review. Frontiers in Pharmacology.. 2022;13:750494.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  38. , , , . Ayurvedic and other herbal remedies for dengue: an update. Clinical Complementary Medicine and Pharmacology.. 2022;2:100024.
    [CrossRef] [Google Scholar]
  39. , . Challenges on the development of a dengue vaccine: a comprehensive review of the state of the art. Journal of General Virology.. 2023;104:001831.
    [CrossRef] [Google Scholar]
  40. . Dengue is spreading Can new vaccines and antivirals halt its rise? Nature. 2023;623:470.
    [CrossRef] [PubMed] [Google Scholar]
  41. Takeda Contact ASoDViHC, Teenagers and Adults in India. ClinicalTrials.gov Identifier: NCT06060067.” Dec. 27, 2023.
  42. , . Dengue vaccine development: Global and indian scenarios. Int J Infect Dis. 2019;84S:S80-6.
    [CrossRef] [PubMed] [Google Scholar]
  43. , , , , . Cellular t-cell immune response profiling by tetravalent dengue subunit vaccine (DSV4) candidate in mice. Front Immunol. 2023;14:1128784.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  44. , , . Biological control of mosquito vectors: Past, present, and future. Insects. 2016;7:52.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  45. , , , , , , et al. Prevention and control strategies to counter zika virus, a special focus on intervention approaches against vector mosquitoes—Current updates. Front Microbiol. 2018;9
    [Google Scholar]
  46. , , . Wolbachia-a foe for mosquitoes. Asian Pacific Journal of Tropical Disease. 2014;4:78-81.
    [CrossRef] [Google Scholar]
  47. , , , , , , et al. Sensitivity of wMel and wAlbB wolbachia infections in aedes aegypti puducherry (Indian) strains to heat stress during larval development. Parasit Vectors. 2022;15:221.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  48. , , , , . Evolutionary dynamics of dengue virus in India. PLoS Pathog. 2023;19:e1010862.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  49. , , , , , . Circulation of DENV-3 genotype 3 during 2017 to 2018 in delhi: A single-center hospital-based study. J Lab Physicians. 2021;14:21-6.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  50. , , , , . Circulation of Dengue virus-1 (DENV-1) serotype in Delhi, during 2010–11 after Dengue virus-3 (DENV-3) predominance: a single centre hospital-based study. Journal of vector borne diseases.. 2012;49:82-5.
    [PubMed] [Google Scholar]
  51. , , , , , , et al. Insight into the seroepidemiology and dynamics of circulating serotypes of dengue virus over a 4 year period in western uttar pradesh, India. Access Microbiol. 2023;5:acmi000567.v4.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  52. . When it comes to a dengue vaccine, we might have to accept imperfection. Nat India 2023 https://www.nature.com/articles/d44151-023-00085-1 [Last accessed 2024 Apr 25]
    [CrossRef] [Google Scholar]
  53. Mallapaty S. DENGUE VACCINE POISED FOR ROLL-OUT—BUT CONCERNS LINGER. Nature. 2022;611. https://www.nature.com/articles/d41586-022-03546-2 [Last accessed 2024 Apr 25]
  54. , , . Dengue outbreak update in India: 2022. Indian J Public Health. 2023;67:181-3.
    [CrossRef] [PubMed] [Google Scholar]
  55. , . Challenges & options in dengue prevention & control: A perspective from the 2015 outbreak. Indian Journal of Medical Research.. 2017;145:718-21.
    [Google Scholar]
  56. , , , , , , et al. Strategic considerations on developing a CHIKV vaccine and ensuring equitable access for countries in need. NPJ Vaccines. 2023;8:123.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  57. World Health Oganization. Guidelines for prevention and control of chikungunya fever. 2009. https://www.who.int/publications/i/item/guidelines-for-prevention-and-control-of-chikungunya-fever [Last accessed 2024 Apr 26].
  58. , . Transcontinental movement of asian genotype chikungunya virus. Emerg Infect Dis. 2014;20:1400-2.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  59. , , , , , , et al. Fast expansion of the asian-pacific genotype of the chikungunya virus in Indonesia. Front Cell Infect Microbiol. 2021;11:631508.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  60. , , , , , , et al. Genotypic and phenotypic characterization of chikungunya virus of different genotypes from Malaysia. PLoS One. 2012;7:e50476.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  61. ECDPC Chikungunya virus disease worldwide overview https://www.ecdc.europa.eu/en/chikungunya-monthly [Last accessed 2024 Apr 30].
  62. Indian Council of Medical R, and ICMR. “ICMR Bulletin,” ICMR, vol. 41, no. November-December 2011, pp. 11–12, 2011.
  63. , , , , , , et al. Genome microevolution of chikungunya viruses causing the indian ocean outbreak. PLoS Med. 2006;3:e263.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  64. NCVBDC. Chikungunya Situation in India. 2023. https://ncvbdc.mohfw.gov.in/index4.php?lang=1&level=0&linkid=486&lid=3765 [Last accessed 2024 Apr 30].
  65. , , , , , , et al. Retrospective study of chikungunya outbreak in urban areas of India. Indian J Med Res. 2012;135:351-8.
    [PubMed] [PubMed Central] [Google Scholar]
  66. , , . Development and application of treatment for chikungunya fever. Res Rep Trop Med. 2022;13:55-66.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  67. , , , , , , et al. Effect of a chikungunya virus–Like particle vaccine on safety and tolerability outcomes. JAMA. 2020;323:1369.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  68. , , , , , , et al. Chikungunya virus vaccine candidate incorporating synergistic mutations is attenuated and protects against virulent virus challenge. J Infect Dis. 2023;227:457-65.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  69. Chikungunya vaccine approved. Nature Biotechnology. 2023;41(12):1667.
    [CrossRef] [PubMed] [Google Scholar]
  70. . Vaccines and antivirals: Grand challenges and great opportunities. FrontVirol. 2021;1
    [CrossRef] [Google Scholar]
  71. , , , , , , et al. Emergence of the east-central-south-african genotype of chikungunya virus in Brazil and the city of rio de janeiro may have occurred years before surveillance detection. Sci Rep. 2019;9:2760.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  72. , . Chikungunya vaccine candidates: Current landscape and future prospects. Drug Des Devel Ther. 2022;16:3663-7.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  73. , , , . An overview of indian biomedical research on the chikungunya virus with particular reference to its vaccine, an unmet medical need. Vaccines (Basel). 2023;11:1102.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  74. . Chikungunya virus vaccines: Platforms, progress, and challenges. Curr Top Microbiol Immunol. 2022;435:81-106.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  75. World Health Organization . Japanese encephalitis. Vaccine-Preventable Diseases Surveillance Standards. 2018. https://www.who.int/publications/m/item/vaccine-preventable-diseases-surveillance-standards-je [Last accessed 2024 Apr 22].
  76. NCVBDC. National Programme for Prevention and Control of Japanese Encephalitis/Acute Encephalitis Syndrome-Operational Guidelines. 2014. https://ncvbdc.mohfw.gov.in/Doc/JE-AES-Prevention-Control(NPPCJA).pdf [Last accessed 2024 Apr 25].
  77. , . Overview: Japanese encephalitis. Prog Neurobiol. 2010;91:108-20.
    [CrossRef] [PubMed] [Google Scholar]
  78. NCVBDC FAQ - Japanese Encephalitis. https://ncvbdc.mohfw.gov.in/index1.php?lang=1&level=2&sublinkid=5930&lid=4005 [Last accessed 2024 Apr 22].
  79. , , , , , , et al. Estimated global incidence of Japanese encephalitis:. Bull World Health Org. 2011;89:766-74.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  80. Simon LV, Sandhu DS, Goyal A, Kruse B. Japanese encephalitis. https://www.ncbi.nlm.nih.gov/books/NBK470423/ [Last accessed 2024 Apr 22].
  81. World Health Organization. Global Health Observatory data repository- Japanese encephalitis Reported cases by WHO region-Reported cases by country. 2023. https://www.who.int/data/gho [Last accessed 2024 May 10].
  82. , . Japanese encephalitis vaccines: WHO position paper—February 2015. Weekly Epidemiological Record= Relevé épidémiologique hebdomadaire. 2015;90:69-88.
    [PubMed] [Google Scholar]
  83. , . Epidemiology of Japanese encephalitis: Past, present, and future prospects. Ther Clin Risk Manag. 2015;11:435-48.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  84. Reshma Kulkarni RK, Sapkal G, Himanshu Kaushal HK, Mourya D. Japanese encephalitis: a brief review on Indian perspectives. 2018. https://pmc.ncbi.nlm.nih.gov/articles/PMC6142657/ [Last accessed 2024 May 05].
  85. NCVBDC. National Guidline for clinical Management of Chikunguya.” 2016. https://ncvbdc.mohfw.gov.in/WriteReadData/l892s/77728737401531912419.pdf [Last accessed 2024 May 10].
  86. NCVBDC. Japanese Encephalities- AES/JE Cases and Deaths in the Country Since 2018-2023. https://ncvbdc.mohfw.gov.in/index1.php?lang=1&level=2&sublinkid=5917&lid=4001 [Last accessed 2024 Apr 14].
  87. , , , , . Management of Japanese encephalitis: a current update. Cureus.. 2021;13(4)
    [CrossRef] [Google Scholar]
  88. , , , . Japanese Encephalitis Vaccine: Recommendations of the Advisory Committee on Immunization Practices. MMWR Recomm Rep.. 2019;68:1-33.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  89. , , , . Recent outbreak of zika virus in India amid ongoing COVID-19 and monkeypox outbreak: A call for action. Int J Surg. 2023;109:601-3.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  90. Welfare MoHF. Universal Immunization Program, Immunization Division at MoHFW. https://www.mohfw.gov.in/sites/default/files/Universal.pdf,0- [Last accessed 2024 May 22].
  91. , . Vaccination policy for Japanese encephalitis in India: Tread with caution! Indian Pediatr. 2015;52:837-9.
    [CrossRef] [PubMed] [Google Scholar]
  92. Ministry of Health and Family Welfare GoI, New Delhi. Operational Guide Japanese Encephalitis Vaccination in India. 2012. https://dhsfw.assam.gov.in/sites/default/files/swf_utility_folder/departments/dirfw_lipl_in_oid_5/menu/document/JE%20Ops%20Guide%2029%20Nov%202012_0_0.pdf [Last accessed 2024 May 22].
  93. , , , , , . Introduction of Japanese encephalitis virus genotype i, India. Emerg Infect Dis. 2011;17:319-21.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  94. , , , , , , et al. Shift in dominant genotypes of Japanese encephalitis virus and its impact on current vaccination strategies. Front Microbiol. 2023;14:1302101.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  95. , , , , , . History of ZIKV infections in India and management of disease outbreaks. Front Microbiol. 2018;9:2126.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  96. Schaefer. RWWTJ. Zika Virus 2023. Available from: http://www.ncbi.nlm.nih.gov/books/NBK430981/ [Last accessed 2024 May 24].
  97. , , , , . Differential virulence between asian and african lineages of zika virus. PLoS Negl Trop Dis. 2017;11:e0005821.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  98. , , , , , , et al. Asian and african lineage zika viruses show differential replication and innate immune responses in human dendritic cells and macrophages. Sci Rep. 2019;9:15710.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  99. , , , , , . African and asian strains of zika virus differ in their ability to infect and lyse primitive human placental trophoblast. PLoS One. 2018;13:e0200086.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  100. , , . Zika virus: Current concerns in India. Indian J Med Res. 2017;146:572-5.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  101. Organization WH. WHO guidelines for the prevention of sexual transmission of Zika virus: World Health Organization; 2018. https://www.who.int/publications-detail-redirect/prevention-of-sexual-transmission-of-zika-virus [Last accessed 2024 May 25].
  102. Ministry of Health and Family Welfare GoI, New Delhi. Guidelines on Zika Virus Disease following Epidemic in Brazil and other countries of America. https://idsp.mohfw.gov.in/WriteReadData/l892s/Guidelines_ZikaVIrus.pdf [Last accessed 2024 May 25].
  103. Organization WH. Zika virus disease-symptoms and Treatment. 2016. https://www.who.int/health-topics/zika-virus-disease [Last accessed 2024 May 25].
  104. National Institute of Virology P. Algorithm for Zika virus diagnosis. https://idsp.mohfw.gov.in/WriteReadData/l892s/217966471499669891.pdf [Last accessed 2024 May 25].
  105. , , , . Current advances in zika vaccine development. Vaccines (Basel). 2022;10:1816.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  106. , , , , . Clinical & epidemiological significance of Kyasanur forest disease. Indian Journal of Medical Research.. 2018;148(2):145-50.
    [Google Scholar]
  107. Natarajan Balakrishnan NB. Current scenario of tick-borne diseases in India-a review. 2017.
  108. , . Predicting kyasanur forest disease in resource-limited settings using event-based surveillance and transfer learning. Sci Rep. 2023;13:11067.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  109. . Kyasanur forest disease in India: Innovative options for intervention. Hum Vaccin Immunother. 2019;15:2243-8.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  110. Prevention CfDCa. Kyasanur Forest Disease (KFD). 2024.
  111. , , , , , , et al. Coverage and effectiveness of kyasanur forest disease (KFD) vaccine in karnataka, south India, 2005-10. PLoS Negl Trop Dis. 2013;7:e2025.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  112. , , , , . Experiences of Indian council of medical research with tick-borne zoonotic infections. Indian J Med Res. 2021;153:339-47.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  113. , , , , . Does the rise in cases of kyasanur forest disease call for the implementation of one health in India? IJID Reg. 2023;7:18-21.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  114. , , , , , , et al. Phylogeography of kyasanur forest disease virus in India (1957–2017) reveals evolution and spread in the Western Ghats region. Sci Rep. 2020;10
    [Google Scholar]
  115. , , , , . Historical expansion of Kyasanur forest disease in India from 1957 to 2017: A retrospective analysis. GeoHealth.. 2019;3:44-55.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  116. , , , , , , et al. Epidemiology, pathogenesis, and control of a tick-borne disease-Kyasanur forest disease: Current status and future directions. Front Cell Infect Microbiol. 2018;8:149.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  117. , , , , . Chandipura virus: an emerging tropical pathogen. Acta tropica.. 2012;124(1):1-14.
    [CrossRef] [PubMed] [Google Scholar]
  118. , , , , , , et al. An outbreak of Chandipura virus encephalitis in the Eastern districts of gujarat state, India. Am J Trop Med Hyg. 2005;73:566-70.
    [CrossRef] [PubMed] [Google Scholar]
  119. . India facing largest Chandipura virus outbreak in 20 years. Lancet. 2024;404:919.
    [CrossRef] [PubMed] [Google Scholar]
  120. Chinikar S, Mirahmadi R, Moradi M, Ghiasi SM, Khakifirouz S. Crimean-Congo hemorrhagic fever (CCHF): IntechOpen; 2012. https://www.intechopen.com/chapters/34771 [Last accessed 2024 June 10].
  121. , , , , , , et al. Quantifying the influence of climate, host and change in land-use patterns on occurrence of crimean congo hemorrhagic fever (CCHF) and development of spatial risk map for India. One Health. 2023;17:100609.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  122. , . Diagnosis of crimean-congo hemorrhagic fever. Expert Rev Anti Infect Ther. 2015;13:555-66.
    [CrossRef] [PubMed] [Google Scholar]
  123. , , , , . Emergence of viral hemorrhagic fevers: Is recent outbreak of crimean congo hemorrhagic fever in India an indication? J Postgrad Med. 2012;58:39-46.
    [CrossRef] [PubMed] [Google Scholar]
  124. , , . Malaria elimination in India: Bridging the gap between control and elimination. Indian Pediatr. 2020;57:613-7.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  125. Organization WH. World malaria report 2023. World Health Organization; 2023. https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2023 [Last accessed 2024 June 08].
  126. , , , , . Global burden, distribution, and interventions for infectious diseases of poverty. Infect Dis Poverty. 2014;3:21.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  127. , , . Climate change & infectious diseases in India: Implications for health care providers. Indian J Med Res. 2013;138:847-52.
    [PubMed] [PubMed Central] [Google Scholar]
  128. . World malaria report 2022. World Health Organization; . https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2022 [Last accessed 2024 June 08]
  129. . World malaria report 2021. Geneva: World Health Organization; . Licence: CC BY-NC-SA 3.0 IGO. https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2021 [Last accessed 2024 June 08]
  130. , , , , , . Tracking district-level performance in the context of achieving zero indigenous case status by 2027. PLOS Glob Public Health. 2023;3:e0001292.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  131. Organization WH. Global technical strategy for malaria 2016-2030: World Health Organization; 2015. https://www.who.int/publications/i/item/9789241564991 [Last accessed 2024 June 09].
  132. , , . Strengthening diagnosis is key to eliminating malaria in India. Lancet Infect Dis. 2019;19:1277-8.
    [CrossRef] [PubMed] [Google Scholar]
  133. , , , , , . Age-specific malaria vulnerability and transmission reservoir among children. Glob Pediatr. 2023;6:None. https://www.sciencedirect.com/science/article/pii/S2667009723000519?via%3Dihub [Last accessed 2024 June 10]
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  134. , , . Time to stimulate Plasmodium vivax research in India: A way forward. Asian Pac J Trop Med. 2023;16:1-2.
    [CrossRef] [Google Scholar]
  135. , , . Use of primaquine and glucose-6-phosphate dehydrogenase deficiency testing: Divergent policies and practices in malaria endemic countries. PLoS Negl Trop Dis. 2018;12:e0006230.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  136. , , , . Strengthening malaria microscopy using artificial intelligence-based approaches in India. Lancet Reg Health Southeast Asia. 2022;5:100054.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  137. , , , . Malaria slide bank plays a crucial role in achieving and sustaining malaria elimination in India. Asian Pacific Journal of Tropical Medicine. 2023;16:431-3.
    [CrossRef] [Google Scholar]
  138. , , , , , , et al. Unreported mixed plasmodium species infection may increase vivax malaria in India: a challenge for malaria elimination. Trans R Soc Trop Med Hyg. 2022;116:600-3.
    [CrossRef] [PubMed] [Google Scholar]
  139. , , . School-age children as asymptomatic malaria reservoir in tribal villages of bastar region, chhattisgarh. Indian Pediatr. 2019;56:873-5.
    [CrossRef] [PubMed] [Google Scholar]
  140. , , , , , , et al. Malaria slide bank to strengthen and improve the quality of malaria diagnosis: A national slide repository in India. Am J Trop Med Hyg. 2024;110:921-4.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  141. , , , . The need for practical insecticide-resistance guidelines to effectively inform mosquito-borne disease control programs. Elife. 2021;10:e65655.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  142. , , , , , . Economic burden of ventilator-associated pneumonia in a developing country. J Hosp Infect. 2012;81:128-30.
    [CrossRef] [PubMed] [Google Scholar]
  143. Bloland PB, Williams HA, Population NRCCo. program on forced migration and health at the mailman School of Public Health, Columbia University. Malaria and Mobility-A Brief History and Overview. 2002. https://www.ncbi.nlm.nih.gov/books/NBK221171/ [Last accessed 2024 June 06].
  144. , , , , . Knowledge attitude and practices of mitanin’s (Community health workers) in chhattisgarh: Malaria elimination perspective. Front Public Health. 2022;9
    [Google Scholar]
  145. Breman JG, Egan A, Keusch GT. The intolerable burden of malaria: a new look at the numbers. The Intolerable Burden of Malaria: A New Look at the Numbers: Supplement to Volume 64 (1) of the American Journal of Tropical Medicine and Hygiene: American Society of Tropical Medicine and Hygiene; 2001. https://www.ncbi.nlm.nih.gov/books/NBK2622/ [Last accessed 2024 June 06].
  146. . malERA: An updated research agenda for diagnostics, drugs, vaccines, and vector control in malaria elimination and eradication. PLoS Med. 2017;14:e1002455.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  147. , , , , , , et al. Antimalarial drug policy in India: Past, present & future. Indian J Med Res. 2014;139:205-15.
    [PubMed] [PubMed Central] [Google Scholar]
  148. , , , , , , et al. Low prevalence of antimalarial resistance mutations in India during 2014–2015: Impact of combining first-line therapy with primaquine. The Journal of Infectious Diseases.. 2024;229:1574-83.
    [CrossRef] [PubMed] [Google Scholar]
  149. , , , , , , et al. Assessment of plasmodium falciparum drug resistance associated molecular markers in mandla, madhya pradesh, India. Malar J. 2023;22:375.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  150. , , . Antimalarial drug resistance: Linking plasmodium falciparum parasite biology to the clinic. Nat Med. 2017;23:917-28.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  151. , , , , , . Prevalence of pfhrp2 and/or pfhrp3 gene deletion in plasmodium falciparum population in eight highly endemic states in India. PLoS One. 2016;11:e0157949.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  152. , . Malaria and economic development. Rev Dev Econ. 2013;17:1-15.
    [CrossRef] [Google Scholar]
  153. . Continuing challenge of malaria in India. Curr Sci. 2012;102:678-82.
    [Google Scholar]
  154. , , , , , , et al. Zoonotic malaria transmission and land use change in southeast asia: What is known about the vectors. Malar J. 2022;21:109.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  155. , , , , , , et al. Scientific achievements and reflections after 20 years of vector biology and control research at the pu teuy mosquito field research station, Thailand. Malar J. 2022;21:44.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  156. , , , , , . Identification of potential antimalarial drug candidates targeting falcipain-2 protein of malaria parasite—A computational strategy. BioTech. 2022;11:54.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  157. , , , , , . A computational strategy for systematic virtual screening of plasmodium falciparum heme detoxification protein inhibitors from the drugbank database. J Biomol Struct Dyn 2024:1-16.
    [Google Scholar]
  158. , , . Advances and opportunities in malaria population genomics. Nat Rev Genet. 2021;22:502-17.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  159. , , , , , . Rethinking malaria vaccines: Perspectives on currently approved malaria vaccines in India’s path to elimination. BMJ Glob Health. 2024;9:e016019.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  160. . Malaria transmission and prospects for malaria eradication: The role of the environment. Cold Spring Harb Perspect Med. 2017;7:a025601.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  161. Oaks Jr SC, Mitchell VS, Pearson GW, Carpenter CC. Social and behavioral aspects of malaria. Malaria: Obstacles and opportunities: National Academies Press (US); 1991. https://www.ncbi.nlm.nih.gov/books/NBK234325/ [Last accessed 2024 June 12].
  162. , . Can water, sanitation and hygiene help eliminate stunting? Current evidence and policy implications. Matern Child Nutr. 2016;12:91-105.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  163. , , , , , , et al. Rethinking human resources and capacity building needs for malaria control and elimination in africa. PLOS Glob Public Health. 2022;2:e0000210.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  164. , , , , , . Leishmaniasis in ancient Egypt and Upper Nubia. Emerg Infect Dis. 2006;12:1616-7.
    [CrossRef] [PubMed] [Google Scholar]
  165. , . Leishmaniasis in ancient Egypt and Upper Nubia - molecular detection of Leishmania in human remains from ancient times. BioRxiv. 2018;278994
    [Google Scholar]
  166. . On the possibility of the occurrence of trypanosomiasis in India. Br Med J. 1903;1:1252.
    [CrossRef] [Google Scholar]
  167. . The relationship of morphology to classification of the Leishmaniasis. J Parasitol. 1949;35:149-50.
    [Google Scholar]
  168. . Taxonomy of the genus leishmania: Present and future trends and their implications. Mem Inst Oswaldo Cruz. 1994;89:471-8.
    [CrossRef] [PubMed] [Google Scholar]
  169. , , , , , . Eliminating visceral leishmaniasis in south asia: The road ahead. BMJ. 2019;364:k5224.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  170. , , , , , , et al. Visceral leishmaniasis and HIV co-infection in Bihar, India: Long-term effectiveness and treatment outcomes with liposomal amphotericin B (AmBisome) PLoS Negl Trop Dis. 2014;8:e3053.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  171. , , , , , , et al. AmBisome monotherapy and combination AmBisome–miltefosine therapy for the treatment of visceral leishmaniasis in patients coinfected with human immunodeficiency virus in India: a randomized open-label, parallel-arm, phase 3 trial. Clin Infect Dis. 2022;75:1423-32.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  172. , , , , , . Report of the fifth post-kala-azar dermal leishmaniasis consortium meeting, Colombo, Sri lanka, 14–16 May 2018. Parasites Vectors. 2020;13
    [Google Scholar]
  173. , , , , . Post-kala-azar dermal leishmaniasis in the Indian subcontinent: A threat to the South-East Asia region kala-azar elimination programme. PLoS Negl Trop Dis. 2017;11:e0005877.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  174. , , , , , , et al. A review of leishmaniasis: Current knowledge and future directions. Curr Trop Med Rep. 2021;8:121-32.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  175. , , , , , , et al. Atypical leishmaniasis: A global perspective with emphasis on the indian subcontinent. PLoS Negl Trop Dis. 2018;12:e0006659.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  176. , , , . Reinfection in american cutaneous leishmaniasis: Evaluation of clinical outcomes in the hamster model. Mem Inst Oswaldo Cruz. 1998;93:353-6.
    [CrossRef] [PubMed] [Google Scholar]
  177. , . Cutaneous leishmaniasis: A 2022 updated narrative review into diagnosis and management developments. Am J Clin Dermatol. 2022;23:823-40.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  178. . Kala-Azar elimination programme: Report of a WHO consultation of partners. Geneva, Switzerland: World Health Organization; . 10-11 February 2015. https://iris.who.int/handle/10665/185042 [Last accessed 2024 June 15]
  179. , , , , , , et al. Visceral leishmaniasis: What are the needs for diagnosis, treatment and control? Nat Rev Microbiol. 2007;5:873-82.
    [CrossRef] [PubMed] [Google Scholar]
  180. , . A review on sustainable approaches for management of sand fly vectors of leishmaniasis. Indian J Med Res. 2004;20:507-16.
    [Google Scholar]
  181. , , , , , , et al. The relationship between leishmaniasis and AIDS: The second 10 years. Clin Microbiol Rev. 2008;21:334-59.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  182. , , , , , , et al. Visceral leishmaniasis and HIV coinfection in latin america. PLoS Negl Trop Dis. 2014;8:e3136.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  183. , , , , , , et al. Liposomal amphotericin B for visceral leishmaniasis in human immunodeficiency virus-coinfected patients: 2-year treatment outcomes in bihar, India. Clin Infect Dis. 2011;53:e91-8.
    [CrossRef] [PubMed] [Google Scholar]
  184. , , . Leishmaniasis and poverty. Trends Parasitol. 2006;22:552-7.
    [CrossRef] [PubMed] [Google Scholar]
  185. , , , , , . Visceral leishmaniasis and HIV coinfection in East Africa. PLoS Negl Trop Dis. 2014;8:e2869.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  186. , , . Trends in the epidemiological characteristics of visceral leishmaniasis in children under 5 years old in Brazil from 2001 to 2012. J Trop Med. 2014;14
    [Google Scholar]
  187. , , , , . Polar and subpolar diffuse cutaneous leishmaniasis in Brazil: Clinical and immunopathologic aspects. Int J Dermatol. 1995;34:474-9.
    [CrossRef] [PubMed] [Google Scholar]
  188. . A new form of cutaneous leishmaniasis—dermal leishmanoid. Ind Med Gaz. 1922;57:125.
    [PubMed] [PubMed Central] [Google Scholar]
  189. , , , , . Post-kala-azar dermal leishmaniasis - A study of 11 cases. Indian J Dermatol Venereol Leprol. 2010;76:55-8.
    [Google Scholar]
  190. , , , . Post kala-azar dermal leishmaniasis: An unresolved mystery. Trends Parasitol. 2014;30:65-74.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  191. , , , , . Post-kala-azar dermal leishmaniasis. Lancet Infect Dis. 2003;3:87-98.
    [CrossRef] [PubMed] [Google Scholar]
  192. , . The immune response to leishmania: Mechanisms of parasite control and evasion. Int J Parasitol. 1998;28:121-34.
    [CrossRef] [PubMed] [Google Scholar]
  193. . Human leishmaniasis: Clinical, diagnostic, and chemotherapeutic developments in the last 10 years. Clin Infect Dis. 1997;24:684-703.
    [CrossRef] [PubMed] [Google Scholar]
  194. . Parasitic adaptive mechanisms in infection by leishmania. Exp Mol Pathol. 2002;72:132-41.
    [CrossRef] [PubMed] [Google Scholar]
  195. , . Leishmania donovani promastigotes evade the activation of mitogen‐ activated protein kinases p38, c‐Jun N‐terminal kinase, and extracellular signal‐regulated kinase‐1/2 during infection of naive macrophages. Eur J Immunol. 2000;30:2235-44.
    [CrossRef] [PubMed] [Google Scholar]
  196. , . Leishmania–Host-cell interaction: Complexities and alternative views. Parasitol Today. 2000;16:292-7.
    [CrossRef] [PubMed] [Google Scholar]
  197. , . The mouse macrophage receptor for C3bi (CR3) is a major mechanism in the phagocytosis of leishmania promastigotes. J Immunol. 1985;135:2785-9.
    [CrossRef] [PubMed] [Google Scholar]
  198. , , , , . Immunity to coccidiosis: Genetic influences on lymphocyte and cytokine responses to infection with eimeria vermiformis in inbred mice. Parasite Immunol. 1993;15:11-9.
    [CrossRef] [PubMed] [Google Scholar]
  199. , , . Natural killer cells participate in the early defense against leishmania major infection in mice. Eur J Immunol. 1993;23:2237-41.
    [CrossRef] [PubMed] [Google Scholar]
  200. , , . Prevalência de oclusão normal e má oclusão em escolares da cidade de Bauru (São Paulo). Parte I: relação sagital. Rev odontol Univ Säo Paulo. 1990:130-7.
    [PubMed] [Google Scholar]
  201. , , , , . Leishmania mexicana mutants lacking glycosylphosphatidylinositol (GPI): protein transamidase provide insights into the biosynthesis and functions of GPI-anchored proteins. Mol Biol Cell. 2000;11:1183-95.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  202. , , , , , . Macrophage complement and lectin-like receptors bind leishmania in the absence of serum. J Exp Med. 1985;162:324-31.
    [CrossRef] [PubMed] [Google Scholar]
  203. , . The host response to leishmania infection. Adv Immunol. 2000;74:275-317.
    [CrossRef] [PubMed] [Google Scholar]
  204. , . Interaction of leishmania with the host macrophage. Trends Parasitol. 2002;18:332-4.
    [CrossRef] [PubMed] [Google Scholar]
  205. . Macrophage‐parasite interactions in leishmania infections. J Leukoc Biol. 1990;47:187-93.
    [CrossRef] [PubMed] [Google Scholar]
  206. , . Proline transport in leishmania donovani amastigotes: Dependence on pH gradients and membrane potential. Mol Biochem Parasitol. 1992;51:1-8.
    [CrossRef] [PubMed] [Google Scholar]
  207. . Energy metabolism in Leishmania. Hum. Paras. Dis.. 1985;1:49-64.
    [Google Scholar]
  208. , . The structure, biosynthesis and function of glycosylated phosphatidylinositols in the parasitic protozoa and higher eukaryotes. Biochem J. 1993;294:305-24.
    [CrossRef] [PubMed] [Google Scholar]
  209. , . Disruption of mannose activation in Leishmania mexicana: GDP‐mannose pyrophosphorylase is required for virulence, but not for viability. EMBO J 2001 https://www.embopress.org/doi/full/10.1093/emboj/20.14.3657 [Last accessed 2024 June 10]
    [Google Scholar]
  210. , , , . Evidence that free GPI glycolipids are essential for growth of leishmania mexicana. EMBO J. 1999;18:2746-55.
    [CrossRef] [PubMed] [Google Scholar]
  211. , , , , , , et al. Filamentous proteophosphoglycan secreted by leishmania promastigotes forms gel-like three-dimensional networks that obstruct the digestive tract of infected sandfly vectors. Eur J Cell Biol. 1999;78:675-89.
    [CrossRef] [PubMed] [Google Scholar]
  212. , , . Leishmania:Amastigotes synthesize conserved secretory acid phosphatases during human infection. Exp Parasitol. 1998;89:161-8.
    [CrossRef] [PubMed] [Google Scholar]
  213. , . Identification of a surface membrane proton-translocating ATPase in promastigotes of the parasitic protozoan leishmania donovani. Biochem J. 1988;256:13-21.
    [CrossRef] [PubMed] [Google Scholar]
  214. , , . Transbilayer inhibition of protein kinase C by the lipophosphoglycan from leishmania donovani. Proc Natl Acad Sci U S A. 1996;93:11634-9.
    [CrossRef] [PubMed] [Google Scholar]
  215. , , . Cell biology of leishmania spp.: Invading and evading. Curr Pharm Des. 2002;8:297-318.
    [CrossRef] [PubMed] [Google Scholar]
  216. , . The involvement of the major surface glycoprotein (gp63) of leishmania promastigotes in attachment to macrophages. J Immunol. 1986;136:2613-20.
    [PubMed] [Google Scholar]
  217. , , , , , , et al. The ser-arg-tyr-asp region of the major surface glycoprotein of leishmania mimics the arg-gly-asp-ser cell attachment region of fibronectin. J Biol Chem. 1992;267:13980-5.
    [CrossRef] [PubMed] [Google Scholar]
  218. , , , , , , et al. Stage-specific glycosphingolipids from amastigote forms of leishmania (L.) amazonensis Immunogenicity and role in parasite binding and invasion of macrophages. J Biol Chem. 1993;268:13723-30.
    [PubMed] [Google Scholar]
  219. , , . Macrophage type 3 complement receptors mediate serum-independent binding of leishmania donovani Detection of macrophage-derived complement on the parasite surface by immunoelectron microscopy. J Exp Med. 1986;164:1332-7.
    [CrossRef] [PubMed] [Google Scholar]
  220. , , , . Activated macrophages destroy intracellular leishmania major amastigotes by an l-arginine-dependent killing mechanism. J Immunol. 1990;144:278-83.
    [CrossRef] [PubMed] [Google Scholar]
  221. , , , , . Leishmania major amastigotes initiate the l-arginine-dependent killing mechanism in IFN-gamma-stimulated macrophages by induction of tumor necrosis factor-alpha. J Immunol. 1990;145:4290-7.
    [PubMed] [Google Scholar]
  222. , . Impairment of the oxidative metabolism of mouse peritoneal macrophages by intracellular leishmania spp. Infect Immun. 1987;55:587-93.
    [CrossRef] [PubMed] [Google Scholar]
  223. , , . Activation of phosphotyrosine phosphatase activity attenuates mitogen-activated protein kinase signaling and inhibits c-FOS and nitric oxide synthase expression in macrophages infected with leishmania donovani. Infect Immun. 1999;67:4055-63.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  224. , , , , , , et al. Modulation of interferon-γ-induced macrophage activation by phosphotyrosine phosphatases inhibition. J Biol Chem. 1998;273:13944-9.
    [CrossRef] [PubMed] [Google Scholar]
  225. , , . Glycoinositolphospholipids of leishmania major inhibit nitric oxide synthesis and reduce leishmanicidal activity in murine macrophages. Eur J Immunol. 1995;25:745-50.
    [CrossRef] [PubMed] [Google Scholar]
  226. , , , , , , et al. Disruption of the trypanothione reductase gene of leishmania decreases its ability to survive oxidative stress in macrophages. EMBO J. 1997;16:2590-8.
    [CrossRef] [PubMed] [Google Scholar]
  227. , , , , , , et al. Nitric oxide production by Leishmania-infected macrophages and modulation by cytokines and prostaglandins. Parassitologia.. 2001;43:1-6.
    [PubMed] [Google Scholar]
  228. , . c-fos and tumor necrosis factor gene expression in leishmania donovani-infected macrophages. Mol Cell Biol. 1989;9:5223-7.
    [CrossRef] [PubMed] [Google Scholar]
  229. . Parasite accessory cell interactions in murine leishmaniasis I Evasion and stimulus-dependent suppression of the macrophage interleukin 1 response by leishmania donovani. J Immunol. 1987;138:1919-25.
    [PubMed] [Google Scholar]
  230. , , , , , , et al. Up-regulation of Th1-type responses in mucosal leishmaniasis patients. Infect Immun. 2002;70:6734-40.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  231. , . Leishmania parasites and their ploys to disrupt macrophage activation. Curr Opin Hematol. 2000;7:26-31.
    [CrossRef] [PubMed] [Google Scholar]
  232. , , , . Cloning of syrian hamster (Mesocricetus auratus) cytokine cDNAs and analysis of cytokine mRNA expression in experimental visceral leishmaniasis. Infect Immun. 1998;66:2135-42.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  233. , . Transforming growth factor β and immunosuppression in experimental visceral leishmaniasis. Infect Immun. 1998;66:1233-6.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  234. , , , , . The importance of TGF-β in murine visceral leishmaniasis. J Immunol. 1998;161:6148-55.
    [PubMed] [Google Scholar]
  235. . The amastigote forms of leishmania are experts at exploiting host cell processes to establish infection and persist. Int J Parasitol. 2007;37:1087-96.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  236. , , , , . H-2M molecules, like MHC class II molecules, are targeted to parasitophorous vacuoles of leishmania-infected macrophages and internalized by amastigotes of l. amazonensis and l. mexicana. J Cell Sci. 1999;112:2559-70.
    [CrossRef] [PubMed] [Google Scholar]
  237. , , , , , , et al. Proteome changes associated with leishmania donovani promastigote adaptation to oxidative and nitrosative stresses. J Proteomics. 2013;81:185-99.
    [CrossRef] [PubMed] [Google Scholar]
  238. , , , , , , et al. Metabolic reconfiguration of the central glucose metabolism: A crucial strategy of Leishmania donovani for its survival during oxidative stress. FASEB J. 2015;29:2081-98.
    [CrossRef] [PubMed] [Google Scholar]
  239. , , , , , , et al. Glucose-6-phosphate dehydrogenase and trypanothione reductase interaction protects leishmania donovani from metalloid mediated oxidative stress. Free Radic Biol Med. 2017;106:10-23.
    [CrossRef] [PubMed] [Google Scholar]
  240. , , , , , , et al. Phosphorylation of translation initiation factor 2-alpha in leishmania donovani under stress is necessary for parasite survival. Mol Cell Biol. 2016;37:e00344-16.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  241. , , , , , , et al. Leishmania donovani induced unfolded protein response delays host cell apoptosis in PERK dependent manner. PLoS Negl Trop Dis. 2018;12:e0006646.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  242. . Ergosterol biosynthesis and drug development for chagas disease. Mem Inst Oswaldo Cruz. 2009;104:311-8.
    [CrossRef] [PubMed] [Google Scholar]
  243. , , , , , . Fatty acid and sterol metabolism: Potential antimicrobial targets in apicomplexan and trypanosomatid parasitic protozoa. Mol Biochem Parasitol. 2003;126:129-42.
    [CrossRef] [PubMed] [Google Scholar]
  244. , . Changes in the sterol composition of the plasma membrane affect membrane potential, salt tolerance and the activity of multidrug resistance pumps in saccharomyces cerevisiae. PLoS One. 2015;10:e0139306.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  245. , , , . Elimination of the last reactions in ergosterol biosynthesis alters the resistance of saccharomyces cerevisiae to multiple stresses. FEMS Yeast Res. 2017;17:fox063.
    [Google Scholar]
  246. , , , , , , et al. Mevalonate kinase of leishmania donovani protects parasite against oxidative stress by modulating ergosterol biosynthesis. Microbiol Res. 2021;251:126837.
    [CrossRef] [PubMed] [Google Scholar]
  247. , , , , , , et al. Oxidant activated soluble adenylate cyclase of Leishmania donovani regulates the cAMP–PKA signaling axis for its intra‐ macrophage survival during infection. J Cell Biochem. 2021;122:1413-27.
    [CrossRef] [PubMed] [Google Scholar]
  248. , , , , . Multiple hybrid genotypes of leishmania (viannia) in a focus of mucocutaneous leishmaniasis. Am J Trop Med Hyg. 2007;76:573-8.
    [CrossRef] [PubMed] [Google Scholar]
  249. , . Innate immunity against leishmania infections. Cell Microbiol. 2015;17:1286-94.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  250. , , , , , , et al. Immune responses in leishmaniasis: an overview. Trop Med Infect Dis. 2022;7:54.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  251. , , . Cytokines and signaling networks regulating disease outcomes in leishmaniasis. Infect Immun. 2022;90:e0024822.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  252. , , , , , , et al. Leishmaniasis: Immune cells crosstalk in macrophage polarization. Trop Med Infect Dis. 2023;8:276.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  253. , , , , , . Role of treg, breg and other cytokine sets in host protection and immunopathology during human leishmaniasis: Are they potential valuable markers in clinical settings and vaccine evaluation? Acta Trop. 2023;240:106849.
    [CrossRef] [PubMed] [Google Scholar]
  254. , , , . Leishmania infection induces macrophage vascular endothelial growth Factor A production in an ARNT/HIF-dependent manner. Infect Immun. 2019;87:e00088-19.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  255. , , , , , , et al. Chemokines in leishmaniasis: Map of cell movements highlights the landscape of infection and pathogenesis. Cytokine. 2021;147:155339.
    [CrossRef] [PubMed] [Google Scholar]
  256. , , . Role of chemokine systems in cancer and inflammatory diseases. MedComm (2020). 2022;3:e147.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  257. , . The role of chemokines in orchestrating the immune response to pancreatic ductal adenocarcinoma. Cancers (Basel). 2024;16:559.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  258. , , , , , , et al. Integrative approaches to understand the mastery in manipulation of host cytokine networks by protozoan parasites with emphasis on plasmodium and leishmania species. Front Immunol. 2018;9:296.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  259. , , . Host immune response against leishmaniasis and parasite persistence strategies: A review and assessment of recent research. Biomed Pharmacother. 2021;139:111671.
    [CrossRef] [PubMed] [Google Scholar]
  260. , , . Genetics, transcriptomics and meta-taxonomics in visceral leishmaniasis. Front Cell Infect Microbiol. 2020;10:590888.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  261. . Leishmaniasis: Current status of vaccine development. Clin Microbiol Rev. 2001;14:229-43.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  262. . Vaccines for leishmaniasis in the fore coming 25 years. Vaccine. 2008;26:1709-24.
    [CrossRef] [PubMed] [Google Scholar]
  263. , , , , , , et al. Controlled field trials of a vaccine against new world cutaneous leishmaniasis. Int J Epidemiol. 1986;15:572-80.
    [CrossRef] [PubMed] [Google Scholar]
  264. , , , , , , et al. Vaccination using live attenuated leishmania donovani centrin deleted parasites induces protection in dogs against leishmania infantum. Vaccine. 2015;33:280-8.
    [CrossRef] [PubMed] [Google Scholar]
  265. , , , , , , et al. A heterologous prime–boost regime using DNA and recombinant vaccinia virus expressing the leishmania infantum P36\/LACK antigen protects BALB\/c mice from cutaneous leishmaniasis. Vaccine. 2002;20:1226-31.
    [CrossRef] [PubMed] [Google Scholar]
  266. , , , , , . Oral salmonella typhimurium (AroA-) vaccine expressing a major leishmanial surface protein (gp63) preferentially induces T helper 1 cells and protective immunity against leishmaniasis. J Immunol. 1990;145:2281-5.
    [CrossRef] [PubMed] [Google Scholar]
  267. , , , , , , et al. Recombinant vaccinia viruses expressing GP46/M-2 protect against leishmania infection. Infect Immun. 1993;61:3351-9.
    [CrossRef] [PubMed] [Google Scholar]
  268. , , , . Inhibition of leishmania donovani promastigote internalization into murine macrophages by chemically defined parasite glycoconjugate ligands. Infect Immun. 1989;57:754-63.
    [CrossRef] [PubMed] [Google Scholar]
  269. , , , , , . Protection against experimental visceral leishmaniasis infection in dogs immunized with purified excreted secreted antigens of leishmania infantum promastigotes. Vaccine. 2005;23:2825-40.
    [CrossRef] [PubMed] [Google Scholar]
  270. , , , , , , et al. Immunization with a polyprotein vaccine consisting of the t-cell antigens thiol-specific antioxidant, leishmania major stress-inducible protein 1, and leishmania elongation initiation factor protects against leishmaniasis. Infect Immun. 2002;70:4215-25.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  271. , , , , . Leish-111f, a recombinant polyprotein vaccine that protects against visceral leishmaniasis by elicitation of CD4+ T cells. Infect Immun. 2007;75:4648-54.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  272. , , , , , , et al. Leishmania donovani suppresses activated protein 1 and NF-κB activation in host macrophages via ceramide generation: Involvement of extracellular signal-regulated kinase. Infect Immun. 2002;70:6828-38.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  273. , , , , , , et al. Vaccination with DNA encoding the immunodominant LACK parasite antigen confers protective immunity to mice infected with Leishmania major. J Exp Med. 1997;186:1137-47.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  274. , , , , , . Kinetoplastid membrane protein-11 DNA vaccination induces complete protection against both pentavalent antimonial-sensitive and -resistant strains of leishmania donovani that correlates with inducible nitric oxide synthase activity and IL-4 generation: Evidence for mixed Th1- and Th2-like responses in visceral leishmaniasis. J Immunol. 2005;174:7160-71.
    [CrossRef] [PubMed] [Google Scholar]
  275. , , , , . A combination DNA vaccine encoding nucleoside hydrolase 36 and glycoproteine 63 protects female but not male hamsters against leishmania mexicana. Parasite. 2009;16:227-30.
    [CrossRef] [PubMed] [Google Scholar]
  276. , , , , , , et al. Modular Multiantigen T cell epitope–Enriched DNA vaccine against human leishmaniasis. Sci Transl Med. 2014;6
    [Google Scholar]
  277. , . Visceral leishmaniasis: Recent advances in diagnostics and treatment regimens. Infect Dis Clin North Am. 2019;33:79-9.
    [CrossRef] [PubMed] [Google Scholar]
  278. , , , , , , et al. Oral miltefosine for indian visceral leishmaniasis. N Engl J Med. 2002;347:1739-46.
    [CrossRef] [PubMed] [Google Scholar]
  279. , . Recent developments and future prospects in the treatment of visceral leishmaniasis. Ther Adv Infect Dis. 2016;3:98-109.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  280. , , , , . Single-dose liposomal amphotericin B for visceral leishmaniasis in India. N Engl J Med. 2010;362:504-12.
    [CrossRef] [PubMed] [Google Scholar]
  281. , , , , . Combination therapy against indian visceral leishmaniasis with liposomal Amphotericin B (FungisomeTM) and short-course miltefosine in comparison to miltefosine monotherapy. Am J Trop Med Hyg. 2020;103:308-14.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  282. , , , , , , et al. Antileishmanial aminopyrazoles: Studies into mechanisms and stability of experimental drug resistance. Antimicrob Agents Chemother. 2020;64:e00152-20.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  283. , , . Leishmaniasis diagnosis: An update on the use of parasitological, immunological and molecular methods. J Parasit Dis. 2020;44:253-72.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  284. , , , . Visceral leishmaniasis and HIV coinfection: Current perspectives. HIV AIDS (Auckl). 2018;10:193-201.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  285. , , , , . Applications of nanomaterials in leishmaniasis: a focus on recent advances and challenges. Nanomaterials. 2019;9:1749.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  286. , , , , , , et al. Cationic solid–lipid nanoparticles are as efficient as electroporation in DNA vaccination against visceral leishmaniasis in mice. Parasite Immunol. 2013;35:397-408.
    [CrossRef] [PubMed] [Google Scholar]
  287. , . Is leishmaniasis the new emerging zoonosis in the world? Vet Res Commun. 2023;47:1777-99.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  288. , , , , , , et al. Field-deployable treatments for leishmaniasis: Intrinsic challenges, recent developments and next steps. Res Rep Trop Med. 2023;14:61-85.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  289. , . Drug resistance in leishmaniasis. J Glob Infect Dis. 2010;2:167-76.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  290. , , , , , . Multiple mutations in heterogeneous miltefosine-resistant leishmania major population as determined by whole genome sequencing. PLoS Negl Trop Dis. 2012;6:e1512.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  291. , , , . Antimony resistance and gene expression in leishmania: Spotlight on molecular and proteomic aspects. Parasitology. 2024;151:1-14.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  292. , , , , , , et al. Amphotericin B resistance in leishmania mexicana: Alterations to sterol metabolism and oxidative stress response. PLoS Negl Trop Dis. 2022;16:e0010779.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  293. , , , , , , et al. The role of ATP-binding cassette transporter genes expression in treatment failure cutaneous leishmaniasis. AMB Express. 2022;12:78.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  294. , . Miltefosine resistance in leishmania donovani involves suppression of oxidative stress-induced programmed cell death. Exp Parasitol. 2013;135:397-406.
    [CrossRef] [PubMed] [Google Scholar]
  295. , , , . Elucidation of cellular mechanisms involved in experimental paromomycin resistance in leishmania donovani. Antimicrob Agents Chemother. 2014;58:2580-5.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  296. , , , , . Host–Pathogen interaction in leishmaniasis: Immune response and vaccination strategies. Immuno. 2022;2:218-54.
    [Google Scholar]
  297. , , , , , , et al. Antileishmanial high-throughput drug screening reveals drug candidates with new scaffolds. PLoS Negl Trop Dis. 2010;4:e675.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  298. , , , , , , . Leishmania donovani 60S ribosomal protein L23a modulates the signaling events leading to the activation of hypoxic response. PLoS Negl Trop Dis. 2016;10:e0005004.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  299. , , , , , , et al. High-throughput functional screening identifies low-affinity targets of concanamycin A in Leishmania parasites. PLoS Negl Trop Dis. 2014;8:e2671.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  300. , , , , , , et al. Genome-wide stochastic adaptive DNA amplification at direct and inverted DNA repeats in the parasite leishmania. PLoS Biol. 2014;12:e1001868.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  301. , , , . Ongoing revolution in leishmania drug discovery: From the classical to the non-classical therapeutic approach. Front Cell Infect Microbiol. 2016;6:1-11.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  302. , , , , , , et al. Telomeric gene deletion and intrachromosomal amplification in antimony‐resistant L eishmania. Mol Microbiol. 2013;88:189-202.
    [CrossRef] [PubMed] [Google Scholar]
  303. , . Drug resistance in Leishmania species: consequences, molecular mechanisms, and possible treatments. Mol Biochem Parasitol. 2010;170:87-94.
    [Google Scholar]
  304. , , , , , , et al. Amphotericin primarily kills yeast by simply binding ergosterol. Proc Natl Acad Sci U S A. 2012;109:2234-9.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  305. , . Amphotericin b: Side effects and toxicity. Rev Iberoam Micol. 2009;26:223-7.
    [CrossRef] [PubMed] [Google Scholar]
  306. , , . Drug resistance in leishmaniasis. Clin Microbiol Rev. 2006;19:111-26.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  307. , . Drug transporters in antiparasitic chemotherapy. Mol Aspects Med. 2007;28:168-208.
    [Google Scholar]
  308. , , , , , , et al. Tackling drug resistance and other causes of treatment failure in leishmaniasis. Front Trop Dis. 2022;3
    [CrossRef] [Google Scholar]
  309. , , , , . Experimental strategies to explore drug action and resistance in kinetoplastid parasites. Microorganisms. 2020;8:950.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  310. , , , . Miltefosine: A review of its pharmacology and therapeutic efficacy in the treatment of leishmaniasis. J Antimicrob Chemother. 2012;67:2576-97.
    [CrossRef] [PubMed] [Google Scholar]
  311. , , . Miltefosine inhibits the membrane remodeling caused by phospholipase action by changing membrane physical properties. Biochim Biophys Acta Biomembr. 2020;1862:183407.
    [CrossRef] [PubMed] [Google Scholar]
  312. , , , . Promising aspects of miltefosine and its potential for future leishmaniasis therapy. Curr Sci. 2007;93:1709-1716.
    [Google Scholar]
  313. . Drug resistance in indian visceral leishmaniasis. Tropical Med Int Health. 2001;6:849-54.
    [CrossRef] [Google Scholar]
  314. , , , , , . Characterisation of leishmania donovani promastigotes resistant to hexadecylphosphocholine (miltefosine) Int J Antimicrob Agents. 2003;22:380-7.
    [CrossRef] [PubMed] [Google Scholar]
  315. , , . Current diagnosis and treatment of visceral leishmaniasis. Expert Rev Anti Infect Ther. 2010;8:919-44.
    [CrossRef] [PubMed] [Google Scholar]
  316. , , , , , . Elevated levels of tryparedoxin peroxidase in antimony unresponsive leishmania donovani field isolates. Mol Biochem Parasitol. 2010;173:162-4.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  317. , , , , , , et al. Potential factors influencing lymphatic filariasis transmission in “hotspot” and “control” areas in Ghana: The importance of vectors. Infect Dis Poverty. 2019;8
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  318. . Clinical and pathological aspects of filarial lymphedema and its management. Korean J Parasitol. 2008;46:119-25.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  319. . Ending the neglect to attain the Sustainable Development Goals: A road map for neglected tropical diseases 2021–2030. World Health Organization; . https://www.eliminateschisto.org/sites/gsa/files/content/attachments/2020-06-10/WHO_Revised-Draft-NTD-Roadmap2021-2030-23Apr2020.pdf [Last accessed 2024 June 08]
  320. , , , , , . Phenotypic evidence of emerging ivermectin resistance in onchocerca volvulus. PLoS Negl Trop Dis. 2011;5:e998.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  321. , , , , , , et al. Mass drug administration of ivermectin, diethylcarbamazine, plus albendazole compared with diethylcarbamazine plus albendazole for reduction of lymphatic filariasis endemicity in papua new Guinea: A cluster-randomised trial. The Lancet Infectious Diseases. 2022;22:1200-9.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  322. . Global programme to eliminate lymphatic filariasis: Progress report, 2011. Wkly Epidemiol Rec. 2012;87:346-56.
    [PubMed] [Google Scholar]
  323. , . Global programme to eliminate lymphatic filariasis: progress report, 2019–Programme mondial pour l’élimination de la filariose lymphatique: rapport de situation, 2019. Weekly Epidemiological Record= Relevé épidémiologique hebdomadaire.. 2020;95:509-23.
    [PubMed] [Google Scholar]
  324. , , , . Detection of benzimidazole resistance-associated mutations in the filarial nematode wuchereria bancrofti and evidence for selection by albendazole and ivermectin combination treatment. Am J Trop Med Hyg. 2005;73:234-8.
    [PubMed] [Google Scholar]
  325. , , . Persistence of microfilaremia in bancroftian filariasis after diethylcarbamazine citrate therapy. Trop Med Parasitol. 1988;39:128-30.
    [PubMed] [Google Scholar]
  326. , , , , , , et al. Vaccination greatly reduces disease, disability, death and inequity worldwide. Bull World Health Organ. 2008;86:140-6.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  327. Control NCfVBD. Filaria. 2022. https://ncvbdc.mohfw.gov.in/index1.php?lang=1&level=1&sublinkid=5777&lid=3691 [Last accessed 2024 June 16].
  328. Ministry of Health and Family Welfare GoI, New Delhi. Annual Report. 2020-21. https://www.mohfw.gov.in/?q=en/publications-3 [Last accessed 2024 June 16].
  329. , . Coverage of, and compliance with, mass drug administration under the programme to eliminate lymphatic filariasis in India: A systematic review. Transactions of the Royal Society of Tropical Medicine and Hygiene.. 2014;108:538-49.
    [CrossRef] [PubMed] [Google Scholar]
  330. . Lymphatic filariasis: a handbook of practical entomology for national lymphatic filariasis elimination programmes 2013 Report No.: 9241505648. https://iris.who.int/handle/10665/87989 [Last accessed 2024 June 18]
  331. , , , , , , et al. Effectiveness of a triple-drug regimen for global elimination of lymphatic filariasis: A modelling study. The Lancet Infectious Diseases. 2017;17:451-8.
    [CrossRef] [PubMed] [Google Scholar]
  332. , , , . Strategies and tools for the control/elimination of lymphatic filariasis. Bull World Health Organ. 1997;75:491-503.
    [PubMed] [PubMed Central] [Google Scholar]
  333. Organization WH. Lymphatic filariasis: Fact sheet. 2022. https://www.who.int/news-room/fact-sheets/detail/lymphatic-filariasis, [Last accessed 2024 June 18].
  334. , . Progress in the elimination of lymphatic filariasis in the western pacific region: Successes and challenges. Int Health. 2020;13:S10-6.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  335. , , , , . Elimination of lymphatic filariasis: Current perspectives on mass drug administration. Res Rep Trop Med. 2018;9:25-33.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  336. Foundation TTAGotGAtELFwsftBaMG. DEC-fortified salt for the elimination of lymphatic filariasis: A manual for program managers 2019. https://www.gaelf.org/dec-fortified-salt-for-the-elimination-of-lymphatic-filariasis-a-manual-for-project-managers [Last accessed 2024 June 19].
  337. , . Lymphatic filariasis: The disease and its control Fifth report of the WHO expert committee on filariasis. World Health Organ Tech Rep Ser. 1992;821:1-71.
    [PubMed] [Google Scholar]
  338. , , , , , , et al. Global programme to eliminate lymphatic filariasis: The processes underlying programme success. PLoS Negl Trop Dis. 2014;8:e3328.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  339. , , , , , , et al. Population genetics of the filarial worm wuchereria bancrofti in a post-treatment region of papua new Guinea: Insights into diversity and life history. PLoS Negl Trop Dis. 2013;7:e2308.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  340. , , . Unfulfilled potential: Using diethylcarbamazine-fortified salt to eliminate lymphatic filariasis. Bull World Health Organ. 2007;85:545-9.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  341. , , , , , , et al. Efficacy, safety, and pharmacokinetics of coadministered diethylcarbamazine, albendazole, and ivermectin for treatment of bancroftian filariasis. Clin Infect Dis. 2016;62:334-41.
    [CrossRef] [PubMed] [Google Scholar]
  342. Ouattara AF, Kouadio O, Bjerum C, Koudou BG, Meite A, Kazura JW, et al., editors. High efficacy of single dose of co-administered ivermectin, diethylcarbamazine and albendazole in treatment of lymphatic filariasis in cote d’ivoire. american journal of tropical medicine and hygiene; 2017, Amer Soc Trop Med & Hygiene 8000 Westpark DR, STE 130, Mclean, VA 22101 USA. https://www.researchgate.net/publication/322539488_High_efficacy_of_single_dose_of_co-administered_Ivermectin_Diethylcarbamazine_and_Albendazole_in_treatment_of_Lymphatic_Filariasis_in_Cote_d%27Ivoire [Last accessed 2024 June 22].
  343. . Diethylcarbamazine salt in the control of lymphatic filariasis. Am J Trop Med Hyg. 1994;50:655-62.
    [CrossRef] [PubMed] [Google Scholar]
  344. , , . Advances in vaccine development for human lymphatic filariasis. Trends Parasitol. 2020;36:195-20.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  345. , , , , , . Improving the efficacy of a prophylactic vaccine formulation against lymphatic filariasis. Parasitol Res. 2017;116:2821-30.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  346. , , , , , , et al. The safety of double- and triple-drug community mass drug administration for lymphatic filariasis: A multicenter, open-label, cluster-randomized study. PLoS Med. 2019;16:e1002839.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  347. , , , . Role of vector control in the global program to eliminate lymphatic filariasis. Annu Rev Entomol. 2009;54:469-87.
    [CrossRef] [PubMed] [Google Scholar]
  348. , , . Filariasis vector control down-played due to the belief the drugs will be enough-not true! Entomologia Generalis. 2020. ;40
    [PubMed] [Google Scholar]
  349. , , , , , , et al. Detection of wuchereria bancrofti DNA in wild caught vector and non-vector mosquitoes: Implications for elimination of lymphatic filariasis. Mol Biol Rep. 2024;51:291.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  350. , , . A review of the scope of vector control research for the elimination of lymphatic filariasis in India. Gates Open Res.. 2024;8:7.
    [Google Scholar]
  351. , , , . Vector-borne diseases. In: , , , , , eds. Biodiversity and Health in the Face of Climate Change. Cham: Springer; . p. :67-90.
    [Google Scholar]
  352. , , . The emergence of lyme disease. J Clin Invest. 2004;113:1093-101.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  353. . Epidemiology of lyme disease. Infect Dis Clin North Am. 2015;29:187-210.
    [CrossRef] [PubMed] [Google Scholar]
  354. . Chronic Lyme disease: A review. Infect Dis Clin North Am. 2008;22:341-60.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  355. , , , , . Subjective symptoms after treatment of early lyme disease. Am J Med. 2010;123:79-86.
    [CrossRef] [PubMed] [Google Scholar]
  356. , , , . Lyme disease: Diagnosis and management. Pract Neurol. 2018;18:455-64.
    [CrossRef] [PubMed] [Google Scholar]
  357. . Lyme disease: An overview. Indian Dermatol Online J. 2023;14:594-60.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  358. , . Scrub typhus: An emerging threat. Indian J Dermatol. 2017;62:478-85.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  359. , , , , , . The burden of scrub typhus in India: A systematic review. PLoS Negl Trop Dis. 2021;15:e0009619.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  360. , , , , , , et al. Scrub typhus in northeast India: Epidemiology, clinical presentations, and diagnostic approaches. Trans R Soc Trop Med Hyg. 2024;118:206-22.
    [CrossRef] [PubMed] [Google Scholar]
  361. , , , , , , et al. Molecular epidemiology and genetic diversity of orientia tsutsugamushi from patients with scrub typhus in 3 regions of India. Emerg Infect Dis. 2015;21:64-9.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  362. , , , , . A review of the global epidemiology of scrub typhus. PLoS Negl Trop Dis. 2017;11:e0006062.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  363. . Scrub typhus: Risks, diagnostic issues, and management challenges. Res Rep Trop Med. 2017;8:73-8.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  364. , , , , , , et al. The epidemiology, diagnosis and management of scrub typhus disease in China. Hum Vaccin Immunother. 2021;17:3795-80.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
Show Sections