3,436
Views
44
CrossRef citations to date
0
Altmetric
Review

Chikungunya vaccines in development

, , , &
Pages 716-731 | Received 15 Jul 2015, Accepted 23 Sep 2015, Published online: 05 May 2016

ABSTRACT

Chikungunya virus has become a global health threat, spreading to the industrial world of Europe and the Americas; no treatment or prophylactic vaccine is available. Since the late 1960s much effort has been put into the development of a vaccine, and several heterogeneous strategies have already been explored. Only two candidates have recently qualified to enter clinical phase II trials, a chikungunya virus-like particle-based vaccine and a recombinant live attenuated measles virus-vectored vaccine.

This review focuses on the current status of vaccine development against chikungunya virus in humans and discusses the diversity of immunization strategies, results of recent human trials and promising vaccine candidates.

Introduction

Chikungunya virus (CHIKV) is an arthritogenic arbovirus belonging to the alphavirus genus of Togaviridae, transmitted to humans by infected female Aedes arthropods. Apart from acute infections causing fever and severe joint pain, CHIKV can cause chronic rheumatism with long-lasting debilitating arthralgia.Citation1,2 The term Chikungunya (CHIK) means “to be contorted” in the Kimakonde language, describing the crouched physical appearance of anguished patients. Like Dengue virus (DENV), CHIKV is maintained by a primate-mosquito-primate cycle with 2 species of Culicidae (Aedes aegypti/Stegomyia aegypti and Aedes albopictus/Stegomyia albopicta), among others, serving as main arthropod vectors, both of which have been implicated in large CHIKV outbreaks. Infection by mosquito bites occur throughout the day, with highest occurrence at dusk, both in- and outdoors. The distribution of Aedes aegypti is largely restricted to tropical and sub-tropical urban areas. In contrast, Aedes albopictus, the Asian tiger mosquito, has undergone a dramatic global spread beyond its original boundaries, invading temperate climate zones of the industrial world and spreading CHIKV to new geographic regions.Citation3

Epidemiology

So far, CHIKV has been identified in over 60 countries in Europe, the Americas, Asia and Africa.Citation4 The first isolation of CHIKV was recorded in 1952 in present-day Tanzania.Citation5 While outbreaks in the 20th century in Southern- and Southeast Asia have remained small and localized, an increasing number of endemic outbreaks on all continents have been reported since 2000. The largest outbreaks were recorded in the Democratic Republic of Congo (50,000 reported cases),Citation6 on Lamu Island in Kenya (13,500 reported cases, which represents 75% of the island's population),Citation7 and on La Réunion Island (266,000 reported cases)Citation8; the biggest outbreak of all was recorded in India (1,300,000 reported cases).Citation9

Europe

The establishment of Aedes albopictus in Europe allowed CHIKV to spread beyond (sub)tropical regions.Citation10 The first ever autochthon CHIKV transmission recorded in Europe was in Northeast Italy in August 2007, probably transmitted by a traveler from Southwest India, resulting in 205 confirmed cases of CHIKV infection.Citation11 One hundred twenty-six laboratory confirmed imported CHIK cases were recorded in mainland France in the summer of 2014.Citation12 Chikungunya virus, however, is not endemic in Europe and the risk of infection is mainly associated with traveling.

The Americas

In 2013, a member of the old Asian lineage CHIKV strain vectored by Aedes aegypti was introduced into the French part of the Caribbean Island of St. Martin and spread through Florida and South America. Until January 2015, 1,094,661 suspected CHIKV infections with 26,606 laboratory-confirmed cases were reported in the Americas,Citation13 yet its spread has probably remained geographically limited;Citation14 Aedes albopictus is the primary mosquito spreading CHIKV into climate zones. The etiologic Asian lineage strain of the Caribbean outbreak, however, ineffectively infects this arthropod due to a lack of E1-adaptive protein substitution.Citation15

In this context, as reported by the CDC, 325 CHIK cases have been documented in 37 U.S. states as of September 2015. All cases were associated with travel and occurred in people returning from outbreak zones.Citation16

Genetic diversity

Chikungunya virus is endemic in tropical and subtropical areas of Africa, Southeast Asia and India. CHIKV circulates in an enzootic cycle of non-human primates (NHP) and arboreal mosquitoes. Transmission of CHIKV into an urban human-mosquito cycle causes regular outbreaks in naïve populations.Citation14

CHIKV is an enveloped, single-stranded positive-sense RNA virus. RNA viruses are genetically very diverse,Citation17 with high mutation rates improving viral fitness and pathogenicity to ensure their survival. The genome of CHIKV is about 11.7 kb and encodes 2 open reading frames (ORF), flanked by 5′ and 3′ un-translated regions. The 5′ ORF is translated from genomic RNA by a cap-dependent mechanism resulting in the formation of 5 structural (envelope proteins E1-E3 forming trimeric spikes on the virions' surface, capsid and 6K/TF) and 4 nonstructural proteins (nsP1–4).Citation18 While glycoprotein (GP) E1 is responsible for fusion within endosomes of target cells and nucleocapsid release, GP E2 interacts with cellular receptors for cell entry.Citation19 The small GP E3 mediates pH-protection during virus biogenesis and prevents E1 from premature fusion.Citation20 The capsid protein of alphavirus serves as a serine protease for self-cleavage, is necessary for the interaction with viral spike proteins during virion formation and serves a major function in nucleocapsid formation.Citation21 The 6K/TF protein is essential for formation and budding of new virions.Citation22

Two-thirds of CHIK virus' RNA encodes for non-structural polyprotein precursors nsP1-4 serving RNA helicase, nucleoside triphosphatase and RNA dependent 5′ triphosphatase enzymatic activity.Citation23

Three major CHIKV genotypes have been isolated - an Asian, a West African and an East/Central/Southern African subtype (ECSA). Sequencing ECSA and Asian genotype strains obtained during Malaysian outbreaks in 2006 and 2008 revealed 96.8% amino acid similarity.Citation24 The greatest genetic diversity was found in ns-proteins, the 6K and the E3 epitope. Furthermore, the genome length differs somewhat between genotypes and is longer in West African (11,843 to 11,881 nucleotides) and Asian (11,777 to 11,999 nucleotides) strains than in the ECSA lineage (11,557 to 11,789 nucleotides).Citation25 The Indian Ocean lineage (IOL), responsible for CHIKV epidemics in Europe in 2007 and 2010, evolved from the ECSA enzootic genotype and was primarily isolated in 2004 during an outbreak in coastal Kenya and the islands of the Indian Ocean. Analysis of the IOL genomic sequence revealed a new viral variant characterized by the substitution of Alanine instead of Valine (A226V) within the E1 protein, the major envelope surface protein. These novel mutations in the envelope glycoproteins suggest adaptive evolution of the virus to local vector abundance and allows CHIKV to use Aedes albopictus in addition to Aedes aegypti as a vector thereby increasing its distribution beyond tropical areas to the Western world. Analysis of A226V showed that the new mutation provided better vector specificity and improved fitness of CHIKV.Citation26

Aedes albopictus is considered the most invasive mosquito species, with a high ability to adapt to different environments and strong competitive tendencies. Its global distribution and introduction to moderate climate zones during the past 30 years was facilitated by globalization, increased human travel, climate change, and the mosquitos' high adaptability to cold temperatures.Citation27

Disease pathogenesis

Data on the pathophysiology of CHIKV infection are based on in vitro experiments, animal models and human studies. However the molecular mechanisms of virus host-cell interactions and the pathogenesis of disease chronification are not fully understood.

While infected female Aedes mosquitoes are feeding on blood, CHIKV gets inoculated intradermally, along with salivary molecules, including proteins and ribonucleic acid.Citation28-30 These salivary components alter host hemostasis for blood feeding purposes and enhance pathogen transmission by modifying immune processesCitation31-34 toward a T(H)2 response, while T(H)1 cells and antiviral cytokines are suppressed.Citation32,34

Both hematopoietic and non-hematopoietic cells are involved in the host control of CHIKV infection.Citation35,36 Local epithelial and endothelial cells, primary fibroblasts as well as monocyte-derived macrophages are susceptible to the virus and allow for its replication.Citation37 Studies on immunocompetent mice identified dermal fibroblasts and skin macrophages as primary target cells (dermal injection phase).Citation37,38 An innate immune response mediated through the recognition of pathogen-associated molecular patterns by pattern recognition receptors (toll like receptor and retinoic acid inducible gene I-like receptors)Citation39 leads to a release of inflammatory cytokines, evoking a pyrogenic reaction. Type I IFN response is critical in the early phase of infection for viral clearance. Accordingly, IFN signaling and chemokine levels (IL-1ß, IL-6, MCP-1) correlate with disease severity, viral load (IL-6, MCP-1)Citation40 and persistent arthralgia (IL-6).Citation38,39 Interferons increase the expression of prostaglandins with ensuing nociceptor activation and sensitization, causing CHIK-characteristic joint pain.Citation41 In this context, high levels of IFNα were more likely to be found in patients with persistent polyarthritis than in those without.Citation42

Migration of infected cells into the draining lymph nodes (lymphatic replication phase) and through the lymph circulation into the blood (viremic phase)Citation39 is followed by viral dissemination throughout the body into peripheral tissue, involving liver cells, muscle cells, joint cellsCitation38,43 and - at least in immunodeficient mice - stromal cells of the central nervous system.Citation38 Long-term and recurrent arthralgia in CHIKV infection might be related to the persistence of CHIKV in synovial macrophages,Citation1,44 which provide a protective cellular reservoir. In human synovial fibroblasts, CHIKV induces miRNA 146a expression, inhibiting TRAF6, IRAK1 and IRAK2, which enhances its replication and interferes with pro-inflammatory pathways, i.e. NFkB- signaling.Citation45 In addition, IL-6 is considered a critical driver of long-lasting joint pain and CHIK-related rheumatic complaints by dysregulating the RANKL (Receptor Activator of NF-κB Ligand)/osteoprotegerin ratio, causing enhanced and sustained activity of osteoclasts and osteoclastogenesis.Citation46,47

However, CHIKV infection commonly results in convalescent adaptive immune protection.Citation41 Adaptive immunity is characterized by CD8+ T cell response during the acute phase of infection and a CD4+ T cell dominant immune response in advanced stages.Citation42 Although the mechanisms of the T lymphocyte response to CHIKV infection are not completely understood, it is considered necessary to maintain long-term immunity.Citation48

CHIKV specific antibodies are detectable as early as one week after infection.Citation43 Immunoglobulin synthesis during the acute viremic phase of CHIK disease has been intensively studied, providing the basis for vaccine development and the use of CHIKV antibodies as passive immunization.Citation49-51

Immunopathogenesis of DENV compared to CHIKV infection

Dengue virus has been reported to infect the same cell types as CHIKV;Citation52 a type-I interferon immune response is triggered by toll-like receptors and DExD/H box RNA helicases;Citation53,54 however, inhibitory mechanisms of the DENV against innate immunity responses are also established.Citation55,56

Interferon response factors (IRF) 3 and 7 limit disease severity in CHIKV infection, protecting against haemorrhagic fever and shock.Citation52 In DENV infection, IRF-7 is described to be crucial in early disease response, together with the transcription factors STAT1-3, IRF9, IRF1, CEBPB, and SP1.Citation57 Inadequate IFN α/β–signaling is thought to contribute to complications in both CHIKV and DENV infections.Citation52

In contrast to CHIKV, DENV infection does not become chronic.Citation58 Exosomes have been shown to contribute to disease limitation in Dengue Virus-2 (DENV-2) infections, conferring IFN inducible transmembrane protein 3 (IFITM3) to neighboring cells.Citation59 IFITM 1, 2 and 3 proteins exhibit antiviral properties and IFITM3 is suggested to reduce DENV-2 cell penetration.Citation60,61 This mechanism might be different from CHIKV infection, although this has yet to be ruled out. Furthermore, it is indicated that monocytes and macrophages, which are of importance in the development of CHIKV chronicity, substantially contribute to DENV infection control.Citation62

Clinical presentation

Acute infection

CHIKV infects all age groups and both sexes at an equal rate.Citation63 It was previously believed that asymptomatic seroconversion was a rare event, occurring in less than 15% of cases.Citation1 Recent data from a prospective cohort study in the Philippines, however, suggest (at least for the Asian genotype) that asymptomatic infections account for the majority of cases depending on age, with a subclinical-to-symptomatic infection ratio of 2:1 in 6-month to 5 year olds and 12:1 in those 50 years of age and over.Citation64 The majority of patients present with rapid-onset fever (usually >39°C), indicating acute viremia, headache, myalgia and joint pain after a mean incubation period of 3 days (range 2–12 days).Citation65 Lymphocytopenia is the main viremia-related laboratory finding.Citation1 The duration and intensity of acute infection correlates with the viral load (up to one billion viral genome copies per mL blood) until viremia ends (5–7 days after onset of symptoms) and immunoglobulin M (IgM) is detectable.Citation1,65 Both IgM and Immunoglobulin G (IgG) levels positively correlate with disease severity.Citation66

Severe joint pain and arthritis is the primary symptom of CHIKV infection and helps to differentiate it from DENV infection, which is one of the most common causes of fever in travelers returning from the (sub)tropics.

CHIKV-related arthritis usually coincidences with the onset of myalgia and fever and presents as severe symmetric (60%) polyarthralgia (positive predictive value for CHIK virema >80%)Citation1 manifesting in distal joints involving the knees, ankles, wrists and hands.Citation67 The axial skeleton is affected in up to 50% of cases.Citation68,69 Three-fourths of patients experience a diffuse maculopapular rash lasting about one week, usually on the trunk and resembling rash-pattern of DENV infection.Citation69 Infrequently, less specific symptoms seen in CHIKV infection include throat discomfort, abdominal pain, constipation, conjunctivitis, pruritus and lymphadenopathy.Citation70

CHIKV infection is commonly a self-limiting disease. During recent outbreaks, however, complicated cases occurred, necessitating hospitalization. The enhanced disease severity observed likely has multiple causes including a more comprehensive recording of CHIK cases, larger-scale outbreaks, better viral adaptability to arthropod vectors, and new mutations of circulating CHIKV - increasing viral fitness and pathogenicity. This is supported by recent data from a neonatal mouse model, which indicate that increased CHIKV virulence may be based on a strains' ability to infect the host's myofibers.Citation71 Josseran et al published a case fatality rate of 1/1000 patients during the 2006 outbreak on La Réunion Island.Citation72 Severe complications including encephalitis, myocarditis, hepatitis and multi-organ failure mainly occured in multimorbid patients with chronic heart, kidney or neurological disorders, patients with diabetes, neonates, young children and elderly over 65 years of age. During the La Réunion outbreak, the mortality of hospitalized patients (17 per 105) with a severely complicated form of the disease was approximately 35%, highlighting the potential fatality of CHIKV infection.Citation73

Persistent symptoms

Persistent pain and chronic musculoskeletal complaints are critical sequelae of CHIKV infection. A study of 180 patients with CHIKV infection found chronic symptom persistence, mostly musculoskeletal complaints, in 60% during a follow-up period of 36 months.Citation74

According to Simon and colleagues, however, 2 distinct patterns of disease chronicity must be distinguished.Citation75 While the vast majority of patients with pain persisting beyond 3 months of infection suffer from heterogeneous musculoskeletal complaints (but do not have polyarthritis), about 5% of patients develop chronic inflammatory rheumatism (including spondyloarthritis, rheumatoid arthritis, or polyarthritis). Rapid differentiation between these 2 entities is important from both a diagnostic and a therapeutic point of view. While the former likely responds well to prolonged therapy with non-steroidal anti inflammatory drugs (NSAIDs), the latter may need early treatment with a disease-modifying antirheumatic drug (DMARD) to timely counteract the potentially destructive process of rheumatic inflammation.

In this context, patients with diabetes mellitus and those with pre-existing traumatic or rheumatic joint disorders have an increased risk of persisting joint pain.Citation74 It is not fully understood why previously injured joints are more severely affected and more susceptible to chronic infection. Similarities in immune responses and inflammatory pathways, including activation of synovial macrophages and increased osteoclastogenesis as the crucial event inducing bone loss, however, suggest a pathogenic relationship between classic rheumatoid arthritis and post-CHIK arthritis.Citation76 Alternatively, the reaction to CHIKV may simply aggravate or drive pre-existing inflammation. Although chronic viremia does not seem to occur in CHIKV infection, analysis of muscle biopsies in a patient suffering from long-term myalgia revealed persistence of the virus within the muscle tissue.Citation77 Moreover, CHIKV can reside and replicate in synovial macrophages.Citation78 This is in contrast to most other arboviruses and is presumed to be the underlying cause of chronic joint pain. Analyzing data from the La Réunion outbreak, Schilte and colleagues estimated the costs of long-term arthralgia at 250€ per patient per year, which, although probably overestimated due to selection bias, underlines the economic burden on the health care system.Citation74

Diagnosis

Given the high predictive value of debilitating arthralgia accompanied by high fever in a CHIK endemic region, diagnosis is mainly clinical. However, different laboratory methods may be used to confirm diagnosis.

Viral culturing based on serum inoculation of mosquito cell cultures, mosquitoes, mammalian cell cultures or mice remains the gold standard, allowing further virus characterization,Citation79-81 but is not used routinely. During the first 8 days of the appearance of symptoms, reverse-transcriptase polymerase chain reaction (RT-PCR) detects serum CHIKV RNA with variable sensitivity. While viral culturing is limited by its restricted availability and temporary extra effort, RT-PCR additionally provides the possibility of genotyping to compare different samples and to detect other arboviruses in a multiplex format.Citation82 PCR assays have been performed on synovial tissue samples and fluids to confirm viral persistence within joints, but this is not recommended as a routine diagnostic test. Foissac and colleagues suggest that post-CHIK chronic rheumatism should be considered in patients unresponsive or dependent on steroid therapy beyond 3 months of disease onset.Citation83

CHIKV serum IgM, assessed by enzyme-linked immunosorbent assay (ELISA), normally presents at the end of the first week of symptoms (days 3-8), indicating disappearance of viremia, reaches peak levels 3-5 weeks after the onset of acute illness, and wanes over a one- to 3-month period. IgG levels, primarily IgG3 isotype,Citation84 are usually detectable as of day 4-10 and persist for years.Citation85,86 A 4-fold increase in IgG levels indicates seroconversion. Continuing high IgM antibody titers are thought to result from limited antibody clearance. Unfortunately, no specific assay exists for the assessment of chronicity of CHIK disease.Citation1 Studies investigating persistent arthralgia following CHIKV infection assessed chronic joint symptoms on a clinical basis by medical examination and by interviewing patients using different questionnaires, including questions about the frequency and location of symptoms, intensity and quality of their pain, and its impact on their everyday activites.Citation87,88

Treatment and prevention

No specific therapy or preventive treatment for CHIK disease in humans is currently available. According to the guidelines on the clinical management of CHIK fever by WHO, therapy is entirely supportive and limited to administering fluids, rest, physiotherapy and the administration of NSAIDs, chloroquine (in the case of refractory arthralgia) and short-term steroids for the management of osteoarticular and ocular manifestations.Citation68,89 In particular, there is no consensus on how to treat and when to initiate therapy in post-CHIK rheumatoid arthritis.

However, re-emerging CHIKV outbreaks during past years sparked research on new strategies for the prevention and treatment of CHIKV infection.

Several antiviral agents including ribavirin and IFNα inhibit CHIKV replication in vitro.Citation90 Doxycycline combined with ribavirin significantly reduces the viral load and the extent of inflammation in ICR mice.Citation91 Chloroquine sulfate failed to be effective in a placebo-controlled, double-blind, randomized clinical trial embarked during the 2006 CHIKV outbreak on La Réunion Island.Citation92 Further DMARDs have been tested only in a few patients. Ganu and colleagueCitation93 assessed the efficacy of methotrexate (MTX) in 16 patients with poor to moderate response to hydroxychloroquine and sulfasalazine combination therapy after 3 months. At a 2 year-follow up, out of 14 patients started on MTX all but one experienced a moderate (21%) to good (71%) response. Beneficial use of MTX in the treatment of post-CHIK arthritis is supported by several other reports.Citation94,95 Very recently, Javelle et al published their results of a 6-year case series retrospective study in Réunion Island of patients suffering from CHIK arthritis, in whom treatment with MTX achieved a positive response in 75% (n = 54) of the patients, and provided the first diagnostic and therapeutic algorithm on how to treat rheumatic disorders that persist after an acute CHIKV infection.Citation96 However, DMARDs other than chloroquine have not been evaluated in large-scale clinical trials and have not yet been implemented in WHO's guidelines on the clinical management of CHIK fever.Citation89

Newly invented antivirals include polymerase- and protease-inhibitors. Favipiravir, a small molecule inhibitor with broad-spectrum antiviral activity currently approved in Japan for the treatment of influenza virus infection, which protected mice from lethal CHIKV infection,Citation97 is one promising candidate.

A further, quite novel, approach is the use of small, interfering RNA sequences and short hairpin RNAs to inhibit CHIKV protein synthesis by post-transcriptional gene silencing.Citation98,99 Although the data surrounding these new agents are promising, their efficacy has yet to be proven in human trials and their indications must be clearly defined.

Passive immunization against CHIKV

gives an overview of different monoclonal antibodies tested against CHIKV.

Table 1. Monoclonal antibodies against CHIKV.

Human protection from CHIKV infection is considered primarily mediated by humoral memory host response and the presence of neutralizing antibodies targeting the virions' outer surfaces of envelope glycoproteins.Citation100 Similarily, several studies support the efficacy of monoclonal antibodies (mAbs) as post-exposure therapy against alpha virus infections. Two recombinant IgG1 human mAbs, 5F10 and 8B10, directed against the CHIKV E1/E2 trimer significantly delayed CHIKV-related mortality in mice and conferred a 100% protection rate upon lethal CHIKV challenge.Citation101-103

Several further CHIKV-neutralizing mAbs (targeting either the GP E1,Citation104 E2Citation105,106 or the caspid proteinCitation107) have been satisfyingly tested in vitro and in animal models,Citation50,105,106 but need to be evaluated in human clinical trials.

Likewise, no data exist on the optimal therapeutic window within which mAbs should be administered in humans. However, a timely infusion seems reasonable. Screening a panel of 230 mouse antibodies, Pal and colleagues recently identified 4 neutralizing mAbs (CHK-102, CHK-152, CHK-166, CHK-263), protecting immunocompromised mice from lethal CHIKV infection. However, no survival benefit from mAb injection was observed in the presence of overt disease 72 hours after infection. In contrast, combined mAb-therapy protected mice when given, at the latest, 24 hours before CHIKV induced death.Citation50 The authors suggested that combining mAbs may also confer synergistic efficacy in humans. Not only the time of mAb administration but other factors including viral burden and virulence, the mAb-dosage, the neutralizing potency of the particular mAb, the synergistic effects of combination therapies, and the possible emergence of viral escape mutants are likely to influence the success of this intervention. Future clinical trials in humans have to answer these questions and need to identify the optimal therapeutic window.

Theoretical risks of passive immunization may include the occurrence of adverse immune reaction (IgE-mediated anaphylaxis and anaphylactoid reactions), acquired immunodeficiency, mAb-specific adverse reactions like thrombotic disorders and cardiotoxicity and the selection of resistant virus variants.Citation108

In view of the high costs, the use of immunotherpeutics against CHIKV infection will not be widely affordable in developing countries. Its use will be restricted to high risk populations including pregnant women, neonates and patients with a complicated form of the disease.Citation51

In addition to their prophylactic and therapeutic potentials, however, mAbs provide a tool to understand host-CHIKV interactions, facilitating the development of active immunization strategies.Citation101

Active immunization against CHIKV

summarizes the most researched vaccine types over the past 4 decades.

Table 2. Studies evaluating heterogeneous vaccine types within the past 44 years. IRES, Internal Ribosome Entry Site; NHP, non-human primates; nsP, non-structural protein.

Vector control through the use of larvicides and adulticides, the removal of larval habitats, limitation of human-vector contact and public education is critical to further control CHIKV outbreaks.Citation109

However active immunization is still considered to be the most cost-effective preventive health intervention. Due to relatively low antigenetic diversity, the development of a CHIK vaccine is a viable goal. Funding of studies on orphan vaccines for low-prevalence infections, however, is limited by comprehensible disincentives. Limited global economic demand, low market potential, and high costs of vaccine development, coupled with unsafe investment returns (in light of unpredictable dynamics in vector distribution and the limited purchasing power of low-income countries) are factors that keep pharmaceutical companies from investing in the resource-consuming process of vaccine development, marketing and distribution. The absence of patent protection in some developing countries further impedes the willingness of pharma companies to invest in vaccine development.Citation110

The ongoing geographic vector spread beyond tropical areas, as well as the genetic plasticity of CHIKV, however, raise concerns about the spread of the disease, the possibility of further epidemics, and highlight the need for effective and comprehensive preventive measures against human CHIKV infection.

Viral epidemics may have a severe long-term impact on a country's economy due to increased medical costs and a decline in tourism - a critical source of economic prosperity in many affected countries.Citation111,112 The CHIKV outbreak on La Réunion, which infected about 300,000 people in 2006, diminished tourism by 60% and the associated economic burden was estimated to be as high as 44 million Euro.Citation113 The economic burden related to such an outbreak is estimated to be more than 300 times greater than the costs associated with preventive measures.Citation114

Though various types of vaccinations against CHIKV infection have been introduced and tested during the past decades, only 3 of them have reached clinical trials phase I/II testing.

Attempts to develop vaccinations against CHIKV are manifold and include live-attenuated vaccines, chimeric alpha-virus candidates, adenovirus-, poxvirus- and DNA-based vaccines, subunit formulations based on recombinant envelope proteins of CHIKV and inactivated Virus-like particles (VLPs).Citation22,115-127 Extensive research on immunization against CHIKV was done in animal models mainly in mice and NHP ().

Clinical trials investigating CHIK vaccines

Back in 1967, early attempts at immunization used a formalin-inactivated tissue culture CHIKV, prepared in bank-frozen green monkey kidney cells.Citation128-130 The vaccine was derived from a clinical CHIKV isolate (strain 15561) obtained during an outbreak in Thailand in 1962. Nine years later, in 1971, Harrison et al tested this live vaccine in 2 cohorts (n=8 each) of 16 healthy volunteers aged 21-25 years, assigned to receive two 0.5mL or 1mL doses twice in a 28-day sequence. The vaccine induced robust immunity (100% seroconversion rate 2 weeks after the second vaccination) and had an excellent safety profile without the occurrence of any local or systemic adverse events. Its development was stopped due to high manufacturing costs.

Based on a lot from the 15561 strain-vaccine, the United States Army Medical Research Institute of Infectious Diseases developed another live virus vaccine (TSI-GSD-218) by serial passage in MRC-5 cells.Citation131 After successful evaluation in a small phase I study, the TSI-GSD-218 vaccine, produced at the Salk Institute, Swiftwater entered phase II in 2000. Seventy-three healthy adults were vaccinated in a double-blind, placebo-controlled manner.Citation131,132 The vaccine was highly immunogenic after one-time immunization and showed an overall acceptable safety profile. However 8% of subjects reported arthralgia, generating concerns about incomplete viral inactivation. Lack of funding and higher priority development efforts led to a halt in further development.Citation116

Recent vaccines in development

To date 3 experimental vaccines have advanced to the stage of human testing. Two candidates (the VRC-CHKVCitation133 and the MV-CHIKCitation127 vaccine) finished phase I, in 2014/15. The third candidate (CHIKV/IRESCitation115 vaccine) yielded promising efficacy and safety results in mice and macaques and plans are in place for a phase I trial. summarizes the advantages and disadvantages of different types of vaccines currently under development.

Table 3. Potential assets and drawbacks of CHIK vaccines that have reached human clinical trial testing.

CHIKV/IRESv1+v2 vaccines

IRES-based attenuated live vaccines derived from the CHIKV-La Réunion strain.

Preclinical development efforts

Just recently, Roy and colleagues published their results of an animal trial testing 2 live-attenuated vaccines (CHIKV/IRESv1+v2) in 3 cohorts of cynomolgus macaques (n = 4 per cohort).Citation115 Seven animals served as sham-controls. The candidate-CHIKV was attenuated by inserting a picornavirus (encephalomyocarditis virus) internal ribosome entry site (IRES) element into the genome of the 2006 La Réunion outbreak strain.Citation134 IRES sequence insertion down-regulates the expression of CHIKV structural genes and impedes infection of vector mosquitoes due to inefficient IRES translation in insects. The precursor vaccine (IRESv1) was previously shown to induce robust immunogenicity in miceCitation135,136 and cross-protected from the closely related (85% homology) African O' nyong'nyong alphavirus.Citation137 Single-vaccination of NHP with IRESv2 was highly immunogenic without any signs of disease. Antibody titers were first detected 15 days after immunization. Neutralizing antibodies assessment was done using 50% and 80% plaque reduction neutralization tests (PRNTs).

In vaccinated cynomolgus macaques no viremia occurred within 3 days upon subcutaneous challenge with wild-type CHIKV La Réunion strain 13 weeks after immunization.

The IRES vaccines were developed to overcome considerable safety issues associated with other attenuated live-virus vaccines and have 2 advantages: First of all, IRES sequence insertion is considered more stable compared to traditional attenuating single-point mutations, which may harbor the risk of mutation-reversion and recovery of pathogenicity in vivo; Secondly, the IRES element disables the vaccine strain to infect insect cells,Citation115 making a vaccine derived in this way particularly suitable for safe use in CHIKV-endemic regions with high vector density and frequent mosquito exposure.

The CHIKV/IRES vaccines are encouraging candidates, but have yet to demonstrate their efficacy in human trials.

VRC-CHKVLP059-00-VP

Virus-like particles containing envelope proteins from the West African CHIKV strain – The VRC-311 trial.

Preclinical development efforts

Enveloped VLPs (eVLPs), containing outer-structure proteins, expressed on a host cell-derived membrane without viral nucleic acid, are considered safe and induce robust immunity by eliciting high titer virus neutralizing antibodies. The VRC-CHKV vaccine is composed of CHIK-VLPs manufactured at the VRC, National Institute of Allergy and Infectious Diseases (Vaccine Pilot Plant operated by Leidos Biomedical Research) by plasmid-transfected embryo kidney cells. The VLPs used comprise capsid and envelope glycoproteins E1 and E2 from the West African CHIKV strain 37997. Several previous studies proving efficacy and safety of eVLPs in miceCitation138 and NHPCitation139 advanced the development status of CHIK-VLP vaccines,Citation140 warranting their advancement into human trials.

Clinical development efforts in humans

Chang and colleagues recently tested an adjuvans-free non-replicating CHIK VLP-based vaccine (Vaccine Research Center (VRC) CHIK virus candidate vaccine VRC-CHKVLP059-00-VP) in a 3 dose escalation (10μg, n = 5; 20μg, n = 10; 40μg, n = 10) phase I trial, funded by the National Institute of Health (NIH) intramural research program.Citation133 Vaccinations were administered intramuscularly at weeks 0, 4 and 20 and the CHIKV specific immune response was assessed by neutralizing antibody assays and ELISA.

This single center trial enrolled 25 healthy adults aged 18-50 years of largely (76%) Caucasian origin from December 2011 to March 2012. Overall, the VRC-CHKV vaccine was well tolerated, without the occurrence of any serious adverse events. No patient experienced arthralgia after immunization with VRC-CHKV. It was highly immunogenic which was reflected by a 100% seroconversion rate in all dose cohorts after a booster immunization. Injection site-related tenderness occurred in 36% of participants but was mild, as were systemic reactions (40% of vaccinees) including myalgia, malaise, headache and nausea. Although no neutralization geometric mean titers (GMT)-threshold protecting against CHIKV infection is known, the one month-titer levels achieved after 3-time vaccination with VRC-CHKV were comparable to convalescent titers after natural CHIKV infection and are considered to be protective. This compares to the rather low PRNT50 of <200 detectable 11 months after (albeit single-) immunization with the live attenuated vaccine TSI-GSD-218.Citation132 More importantly, neutralizing antibody titers were still detectable in all dose cohorts 6 months after immunization with VRC-CHKV, suggesting robust immunogenicity. The results of this trial confirm the findings of the preclinical study on VRC-CHKV in NHP.Citation139 The human trial, however, was limited by its small sample size.

MV-CHIK

Live attenuated measles virus (MV)-based vaccine expressing surface proteins from the La Réunion ECSA CHIKV strain – The MV-CHIK trial

Preclinical development efforts

Originally developed as a vector to express heterologous viral antigens at the Institut Pasteur (Paris, France), the live attenuated Schwarz strain has proven to elicit robust humoral and cellular immune responses, yielding protective immunity against various arthropod-borne diseases (including DENV and West Nile virus) in mice and monkeys.Citation141-143 Measles virus vaccines have been mass-produced at low cost for about 50 years, are well established, safe and provide good boosterability with a humoral and cellular mediated long-term memory effect.Citation144

In 2013, Brandler et al introduced the CHIKV La Réunion strain-06-46 (obtained from viremic patients) subgenomic open reading frame encoding for the structural genes C-E3-E2-6K-E1 into the MV vector.Citation48 The La Réunion strain belongs to the ECSA lineage of CHIKV, which was responsible for most Indian Ocean epidemics in the past. Preclinical studies on the efficacy and safety of MV-CHIK were done in CD46 expressing transgenic mice lacking IFNα/β receptors (CD46-IFNAR). The live attenuated MV-vaccine expressing CHIKV envelope and capsid proteins induced a robust neutralizing immune response and completely protected the mice from a lethal CHIKV challenge after one or 2 immunizations. The passive transfer of MV-CHIK pre-immune sera containing neutralizing antibodies conferred full protection to mice.Citation48 The immunogenicity of the recombinant vaccine was also demonstrated in NHP (data not published; Dr. Ramsauer K., personal communication, August 25th 2015).

Based on these results, the MV-CHIK single center trialCitation127 was conducted from November 2013 to June 2014 at the Medical University of Vienna, Austria.

Clinical development efforts in humans

Ramsauer et al recently investigated the immunogenicity, safety and tolerability of a live attenuated recombinant viral-vectored vaccine based on the MV-Schwarz strain, MV-CHIK. This first-in-man trial tested the vaccine's safety and immunogenicity in the presence of pre-existing measles immunity in a randomized, double-blind, controlled dose escalation design (1.5 × 104 - 3 × 105 50% tissue culture infection dose (TCID50) per individual) in 42 healthy adults aged 18-45 years.

In the per protocol analysis (n = 36), MV-CHIK raised neutralizing antibodies, as assessed by PRNT50, in a dose-dependent manner and yielded a 100% seroconversion rate after the second immunization in all 3 dose cohorts. The GMT persisted over a 3-month period in the high dose group. As with the VRC-CHKV vaccine, no MV-CHIK-related serious adverse events were observed. Transient musculoskeletal pain was reported in 12% of subjects at the first follow-up visit and, overall, 6 participants experienced local pain or erythema, headache and pyrexia. Local reactions, however, were considered related to the 1mL-inoculation volume along with the vaccination's salt buffer content rather than the active ingredient.

The MV-CHIK trial enrolled 42 volunteers (compared to 25 subjects in the VRC 311 trial), mostly of Caucasian origin (98%; compared to 76% in VRC 311 trial). MV-CHIK participants were block randomized to receive booster immunization 4 or 13 weeks after the initial injection. This allowed for the assessment of both short-term immunogenicity/booster ability, a vaccine characteristic that could be relevant for travelers and tourists, as well as single-dose-related, 3-month persistence of neutralizing antibody titers. In contrast, antibody titers yielded by the VRC-CHKV vaccine were assessed and detectable up to 180 days after vaccination.Citation133 Due to inter-assay differences, however, neutralizing antibody titers cannot be directly compared.

Discussion

MV-based vaccines may be considered suitable for long-term protective mass immunization, given the long-lasting neutralizing antibody response achieved by the measles vaccine. It has been hypothesized that immunity by previous MV infection or vaccination might interfere with the protective efficacy and immunogenicity of a recombinant MV-based vaccine. This is a particular concern for this type of vaccine. However, large-scale clinical trials in humans have demonstrated a boostered anti-measles antibody production upon revaccination of previously MV-immunized individuals.Citation145,146 A preclinical study tended in the same direction: Immunity to MV did not impair the protective capacity of MV-CHIKV in transgenic CD46/IFNAR mice.Citation48 Likewise, in healthy adults, the immunogenicity of MV-CHIK was independent from the baseline immunity.Citation127

However, given the high measles vaccination-coverage rate (>80% worldwide), pre-existing immunity against MV remains an interesting challenge that must be addressed in future human trials.

An advantage of both the VLP- and the MV-based vaccine could be the lack of a live and replicating CHIKV, rendering both candidates unlikely to induce CHIK-like adverse events including arthralgia and chronic rheumatism. Accordingly, no single subject vaccinated with VRC-CHKV complained of arthralgia, underlining the vaccine's favorable tolerability profile. In addition, VLP based vaccines can be considered safe for immunization of immunosuppressed people. However, the establishment of a chronic infection by replication-efficient CHIK virions may constitute a threat to patients with immunodeficiency. In this context, Seymour and colleagues very recently established a rodent model of chronic CHIKV infection in lymphocyte-deficient RAG1 knock-out mice lacking adaptive immunity for safety evaluation of the live-attenuated CHIK/IRES vaccine candidate under immunocompromised conditions.Citation147 This is a critical safety issue when using live virus vaccines, given the high rates of HIV infection and malnourishment in several developing countries. As already demonstrated for a type-1-IFN deficient state, CHIK/IRES vaccination was safe and did not cause virus persistence despite a lack of adaptive immunity. These findings warrant further evaluation of the CHIK/IRES vaccine in a clinical trial, including patients with compromised host-immune defense. Likewise, preclinical studies on the efficacy of MV-CHIK were conducted in IFN receptor-deficient mice. However, as live vaccines may lead to uncontrolled viral replication under immune-compromised conditions, this safety aspect needs to be addressed in adequately designed trials.

In contrast to a VLP-based vaccine, a live-attenuated vaccine does not require the use of an adjuvant for sufficient long-term protection. VLP-based vaccines are both safe and strongly immunogenic, but, may need several adjuvanted administrations in order to induce complete immunity.Citation139 Although other studies investigating the efficacy of adjuvant-free eVLP-vaccines suggest that single-immunization might be sufficient,Citation133,140 this must be confirmed for recombinant CHIKV VLPs in (further) human trials.

Adjuvants are added to a vaccine to enhance its immunogenicity and to reduce the number of immunizations needed to achieve a sufficient antibody titer. However, adjuvanting may enhance reactogenicity and impair a vaccine's tolerability profile, depending on the adjuvant used.Citation148 A large meta-analysis comparing different influenza vaccines found that local adverse reactions were significantly more common with oil-in-water adjuvants than with adjuvant-free and aluminum-salt based vaccines.Citation149 Furthermore, adjuvants are thought to be implicated in the initiation or exacerbation of autoimmune disorders, a phenomenon known as Autoimmune/inflammatory Syndrome Induced by Adjuvant (ASIA) syndrome.Citation150 Currently, however, there is insufficient evidence supporting a definitive relationship between the use of adjuvants and the occurrence of autoimmune disorders. Because autoimmunity likely depends on several factors including the individual's genetic predisposition, this is an issue that should be considered when developing a human vaccine.

The lack of an adjuvant, however, may not only translate into a better tolerability profile but may also reduce the costs of manufacturing a vaccine, because relatively lower doses might be effective. This would be an important factor in the realization of large-scale use in low-income countries, considering the cost-intensive manufacturing process of VLPs in vitro. In this respect, plant “biofarming” technology is a promising, cost-effective approach to produce VLP-based vaccines,Citation151 characterized by high scalability, low production costs, lack of human and animal pathogens and the potential to produce oral and parenteral vaccine types. Several plant-based vaccines have already been investigated - including those for swine influenza, rabies and hepatitis B,Citation152 - but, have not yet been marketed. Another promising possible tool for the mass-production of eVLP-based vaccines is the baculovirus expression vector/insect cell (BEVS/IC) system.Citation154 A concise review on the current status of the (e)VLP vaccines development was recently published by Pijlman.Citation153 The use of insect cells as an expression system offers several advantages, including: A high rate of cell division; Growth in the absence of serum; And a high degree of complexity of generated VLPs.Citation155 Using baculovirus as an expression vector allows for short turnaround times, which provides product development flexibility especially needed for a quick response to pandemic outbreaks.

Active immunizations against CHIKV need to produce protective immunity against all CHIKV genotypes. Immunity to only one strain could possibly increase disease severity upon reinfection with another viral strain, as seen in secondary wild-type DENV infection. Antibody-dependent disease enhancement (ADE) has not been described in CHIKV infection yet, but it has been suspected to play a role in infections by other arboviruses,Citation156 including the closely-related alpha virus Ross River virus.Citation157 In this context, Hallengärd and colleagues recently observed increased disease severity in vaccinated mice with low anti-CHIKV-IgG titers upon subsequent re-infection with a heterogenic CHIKV strain.Citation121 Although no human data exist supporting the hypothesis of vaccination-related ADE, this issue should be addressed when conducting a clinical trial enrolling subjects at risk of CHIKV infection.

Though MV-CHIK is derived from the La Réunion strain, virus neutralization assays used an attenuated CHIKV strain (clone 25/strain 181) based on an Asian lineage virus isolate. Thus, the MV-CHIK vaccine presumably elicits a cross-neutralizing immune response in vivo.Citation127

This is in agreement with results from preclinical mice-studies, where MV-CHIK conferred protection against the homologous (06-49) and heterologous (India, Congo, Thailand) CHIKV strains.Citation48

In comparison, the amount of cross-reactive neutralizing activity against the ECSA outbreak strain OPY1 induced by the VRC-CHKV vaccine suggests that cross-protection could also be achieved for several strains, including the type circulating in the Americas.Citation133

Efficacy trials in humans

No vaccine against CHIK disease has yet undergone efficacy testing in humans. Vaccine-efficacy against virologically confirmed CHIKV infection can only be determined in large scaled clinical trials including people at risk of CHIKV infection in affected countries.

This, however, may be a difficult task to undertake considering the unpredictability of sporadic CHIKV outbreaks and the high rate of asymptomatic infections (at least with the Asian genotype, as recently reported). Alternatively, levels of neutralizing antibodies could serve as surrogate parameter for vaccine efficacy.Citation158 This is an established concept for several licensed vaccines including those against influenzaCitation159 and measles,Citation160 where antibody titers correlate well with human protection, which would dramatically facilitate the marketing approval for a vaccine against CHIK disease.

Future perspectives

Treatment: Long-term administration of NSAIDs continues to be the mainstay of therapy for patients with post-CHIK chronic pain and musculoskeletal disorders. However, no consensus exists on the proper diagnosis and treatment of CHIK-related chronic inflammatory rheumatism. There is an obvious need for randomized clinical trials testing the efficacy of different DMARDs in the treatment of post-CHIK rheumatoid arthritis, as well as the implementation of valid regulating guidelines.

Prophylaxis: No CHIK vaccine is currently under clinical investigation. Searching European clinical trials databases (clinicaltrials.gov; clinicaltrialsregister.eu) and the NIH Clinical Research Studies registry (clinicalstudies.info.nih.gov; researchmatch.org), we found only one trial that is currently testing the efficacy and safety of an anti-CHIKV hyperimmune intravenous immunoglobulin for passive immunization against CHIK disease in high-risk neonates born to mothers with presumed active CHIKV infection (clinicaltrials.gov Identifier: NCT02230163).

MV-CHIK: The evaluation of MV-CHIK in a randomized controlled multicenter phase II trial addressing safety and the persistence of functional anti-CHIKV antibodies over an extended period of time in a larger and more heterogeneous population will start in late 2015 (Dr. Ramsauer K., personal communication, June 25th 2015).

VRC-CHKVLP059-00-VP: Likewise, the VRC 311 study team plans to initiate a phase II trial in late 2015 (Dr. Ledgerwood J., personal communication, July 7th 2015).

CHIK/IRES: The CHIK/IRES vaccine, which has been successfully tested in (immunosuppressed) mice and NHP, is projected for a phase I clinical trial by Takeda Inc.,Citation161 but there is no date set for the IND submission (Dr. Weaver S.C., personal communication, June 29th, 2015).

Conclusions

Recent dynamics in the geographic distribution of CHIKV with 2.5 million people infected over the past yearsCitation162 emphasize the vital necessity of a sufficient immunization coverage rate and demand further vaccine research to limit spreading of the virus, as well as to reduce the economic burden of increased healthcare costs.

Substantial progress was achieved in the research and development of vaccinations against CHIK within past years. Varied approaches yielded encouraging possiblities, yet, substantial challenges remain. An ideal vaccine should be highly immunogenic, safe, adjuvant-free, have minimal side effects, and should confer long-term immunity after a single vaccination – and at a low cost. Mass immunization necessitates affordable vaccines.

Virus-like particle-based vaccines combine high safety with strong efficacy but may be constrained by their comparably lower long-term protective capacity, the need for multiple-dosing and higher manufacturing costs thereby limiting their use in low-income countries. Optimal strategies for cost-effective VLP production must be implemented in order to provide high scalability, sufficient quality and good immunogenic potency of VLPs. Live-virus vaccines elicit strong immune responses and long-term protection without the need for an adjuvant. However, there are numerous safety concerns that limit their usability in CHIKV-endemic regions - particularly for immunosuppressed patients: Unstable attenuation; Viral recovery to wild type virulence; Chronic infection; and transmission to vector insects.

Further vaccination strategies have been explored with promising preclinical results including a chimeric alphavirusCitation117 and a DNA replicon vaccine encoding the CHIKV envelope (tDREP-Env),Citation121 p62-E1 protein vaccines,Citation121 GP E2 protein subunits,Citation124 adenovirus-basedCitation118 vaccine and the recombinant-modified vaccinia virus Ankara vaccine expressing CHIKV antigens (MVA-CHIKV).Citation121 Future studies will define their role in the ongoing fight against CHIKV.

Abbreviations

CHIK=

Chikungunya

CHIKV=

Chikungunya Virus

DENV=

Dengue Virus

ECSA=

East/Central/Southern African subtype

ELISA=

Enzyme-linked Immunosorbent-assay

GMT=

Geometric Mean Titers

GP=

Glycoprotein

IgG=

Immunoglobulin G

IgM=

Immunoglobulin M

IFN(s)=

Interferon(s)

IOL=

Indian Ocean Lineage

IRES=

Internal Ribosome Entry Site

mAbs=

Monoclonal Antibodies

MTX=

Methotrexat

MV=

Measles Virus

NHP=

Non-Human Primate

nsP=

Non Structural Proteins

PRNT=

Plaque Reduction Neutralization Tests

RT-PCR=

Reverse-Transcriptase Polymerase Chain Reaction

(e)VLP=

(enveloped) Virus-Like Particles

VRC=

Vaccine Research Center

Disclosure of potential conflicts of interest

MS was an investigator of the MV-CHIK trial (PMID: 25739878). BJ served as a regulatory advisor to Themis GmbH. NB, PPW and CS declare no competing interests.

Acknowledgment

We would like to thank Stacy Dahl, M.A., B.A. (professional lector), for linguistic editing of the manuscript.

References

  • Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med 2015; 372:1231-9; PMID:25806915; http://dx.doi.org/10.1056/NEJMra1406035
  • Robinson MC. An epidemic of virus disease in Southern Province, Tanganyika Territory, in 1952-53. I. Clinical features. Trans R Soc Trop Med Hyg 1955; 49:28-32; PMID:14373834; http://dx.doi.org/10.1016/0035-9203(55)90080-8
  • Rezza G. Dengue and chikungunya: long-distance spread and outbreaks in naive areas. Pathog Glob Health 2014; 108:349-55; PMID:25491436; http://dx.doi.org/10.1179/2047773214Y.0000000163
  • WHO. Chikungunya Fact sheet N°327. 2015; http://www.who.int/mediacentre/factsheets/fs327/en/ (accessed June 18th, 2015).
  • Ross RW. The Newala epidemic. III. The virus: isolation, pathogenic properties and relationship to the epidemic. J Hyg 1956; 54:177-91; PMID:13346078; http://dx.doi.org/10.1017/S0022172400044442
  • Moyen N, Thiberville SD, Pastorino B, Nougairede A, Thirion L, Mombouli JV, Dimi Y, Leparc-Goffart I, Capobianchi MR, Lepfoundzou AD, et al. First reported chikungunya fever outbreak in the republic of Congo, 2011. PloS one 2014; 9:e115938; PMID:25541718; http://dx.doi.org/10.1371/journal.pone.0115938
  • Chretien JP, Anyamba A, Bedno SA, Breiman RF, Sang R, Sergon K, Powers AM, Onyango CO, Small J, Tucker CJ, et al. Drought-associated chikungunya emergence along coastal East Africa. Am J Trop Med Hyg 2007; 76:405-7; PMID:17360859
  • Economopoulou A, Dominguez M, Helynck B, Sissoko D, Wichmann O, Quenel P, Germonneau P, Quatresous I. Atypical Chikungunya virus infections: clinical manifestations, mortality and risk factors for severe disease during the 2005-2006 outbreak on Reunion. Epidemiol Infect 2009; 137:534-41; PMID:18694529; http://dx.doi.org/10.1017/S0950268808001167
  • Mudur G. Failure to control mosquitoes has led to two fever epidemics in India. BMJ 2006; 333:773; PMID:17038726; http://dx.doi.org/10.1136/bmj.333.7572.773-c
  • Gould EA, Gallian P, De Lamballerie X, Charrel RN. First cases of autochthonous dengue fever and chikungunya fever in France: from bad dream to reality! Clin Microbiol Infect 2010; 16:1702-4; PMID:21040155; http://dx.doi.org/10.1111/j.1469-0691.2010.03386.x
  • Rezza G, Nicoletti L, Angelini R, Romi R, Finarelli AC, Panning M, Cordioli P, Fortuna C, Boros S, Magurano F, et al. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet 2007; 370:1840-6; PMID:18061059; http://dx.doi.org/10.1016/S0140-6736(07)61779-6
  • Paty MC, Six C, Charlet F, Heuze G, Cochet A, Wiegandt A, Chappert JL, Dejour-Salamanca D, Guinard A, Soler P, et al. Large number of imported chikungunya cases in mainland France, 2014: a challenge for surveillance and response. Euro Surveill 2014; 19:20856; PMID:25060572
  • Halstead SB. Reappearance of chikungunya, formerly called dengue, in the americas. Emerg Infect Dis 2015; 21:557-61; PMID:25816211; http://dx.doi.org/10.3201/eid2104.141723
  • Weaver SC. Arrival of chikungunya virus in the new world: prospects for spread and impact on public health. PLoS Negl Trop Dis 2014; 8:e2921; PMID:24967777; http://dx.doi.org/10.1371/journal.pntd.0002921
  • Tsetsarkin KA, Chen R, Leal G, Forrester N, Higgs S, Huang J, Weaver SC. Chikungunya virus emergence is constrained in Asia by lineage-specific adaptive landscapes. Proc Natl Acad Sci U S A 2011; 108:7872-7; PMID:21518887; http://dx.doi.org/10.1073/pnas.1018344108
  • CDC. 2015 provisional data for the United States. 2015; http://www.cdc.gov/chikungunya/geo/united-states-2015.html (accessed August 26th, 2015).
  • Zheng K, Li J, Zhang Q, Liang M, Li C, Lin M, Huang J, Li H, Xiang D, Wang N, et al. Genetic analysis of chikungunya viruses imported to mainland China in 2008. Virol J 2010; 7:8; PMID:20078896; http://dx.doi.org/10.1186/1743-422X-7-8
  • Fros JJ, Liu WJ, Prow NA, Geertsema C, Ligtenberg M, Vanlandingham DL, Schnettler E, Vlak JM, Suhrbier A, Khromykh AA, et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferon-stimulated JAK-STAT signaling. J Virol 2010; 84:10877-87; PMID:20686047; http://dx.doi.org/10.1128/JVI.00949-10
  • Dudha N, Rana J, Rajasekharan S, Gabrani R, Gupta A, Chaudhary VK, Gupta S. Host-pathogen interactome analysis of Chikungunya virus envelope proteins E1 and E2. Virus Genes 2015; 50:200-9; PMID:25563600; http://dx.doi.org/10.1007/s11262-014-1161-x
  • Uchime O, Fields W, Kielian M. The role of E3 in pH protection during alphavirus assembly and exit. J Virol 2013; 87:10255-62; PMID:23864626; http://dx.doi.org/10.1128/JVI.01507-13
  • Choi HK, Tong L, Minor W, Dumas P, Boege U, Rossmann MG, Wengler G. Structure of Sindbis virus core protein reveals a chymotrypsin-like serine proteinase and the organization of the virion. Nature 1991; 354:37-43; PMID:1944569; http://dx.doi.org/10.1038/354037a0
  • Hallengard D, Kakoulidou M, Lulla A, Kummerer BM, Johansson DX, Mutso M, Lulla V, Fazakerley JK, Roques P, Le Grand R, et al. Novel attenuated Chikungunya vaccine candidates elicit protective immunity in C57BL/6 mice. J Virol 2014; 88:2858-66; PMID:24371047; http://dx.doi.org/10.1128/JVI.03453-13
  • Saisawang C, Sillapee P, Sinsirimongkol K, Ubol S, Smith DR, Ketterman AJ. Full length and protease domain activity of Chikungunya virus nsP2 differ from other alphavirus nsP2 proteases in recognition of small peptide substrates. Biosci Rep 2015; 35
  • Sam IC, Loong SK, Michael JC, Chua CL, Wan Sulaiman WY, Vythilingam I, Chan SY, Chiam CW, Yeong YS, AbuBakar S, et al. Genotypic and phenotypic characterization of Chikungunya virus of different genotypes from Malaysia. PloS one 2012; 7:e50476; PMID:23209750; http://dx.doi.org/10.1371/journal.pone.0050476
  • Volk SM, Chen R, Tsetsarkin KA, Adams AP, Garcia TI, Sall AA, Nasar F, Schuh AJ, Holmes EC, Higgs S, et al. Genome-scale phylogenetic analyses of chikungunya virus reveal independent emergences of recent epidemics and various evolutionary rates. J Virol 2010; 84:6497-504; PMID:20410280; http://dx.doi.org/10.1128/JVI.01603-09
  • Tsetsarkin KA, Vanlandingham DL, McGee CE, Higgs S. A single mutation in chikungunya virus affects vector specificity and epidemic potential. PLoS Pathog 2007; 3:e201; PMID:18069894; http://dx.doi.org/10.1371/journal.ppat.0030201
  • ECDC. Aedes albopictus. 2014; http://ecdc.europa.eu/en/healthtopics/vectors/mosquitoes/Pages/aedes-albopictus.aspx (accessed July 10th, 2015)
  • Ribeiro JM, Rossignol PA, Spielman A. Role of mosquito saliva in blood vessel location. J Exp Biol 1984; 108:1-7; PMID:6707570
  • Ribeiro JM, Arca B, Lombardo F, Calvo E, Phan VM, Chandra PK, Wikel SK. An annotated catalogue of salivary gland transcripts in the adult female mosquito, Aedes aegypti. BMC Genomics 2007; 8:6; PMID:17204158; http://dx.doi.org/10.1186/1471-2164-8-6
  • Maharaj PD, Widen SG, Huang J, Wood TG, Thangamani S. Discovery of mosquito saliva microRNAs during CHIKV infection. PLoS Negl Trop Dis 2015; 9:e0003386; PMID:25612225; http://dx.doi.org/10.1371/journal.pntd.0003386
  • Puiprom O, Morales Vargas RE, Potiwat R, Chaichana P, Ikuta K, Ramasoota P, Okabayashi T. Characterization of chikungunya virus infection of a human keratinocyte cell line: role of mosquito salivary gland protein in suppressing the host immune response. Infect Genet Evol 2013; 17:210-5; PMID:23583544; http://dx.doi.org/10.1016/j.meegid.2013.04.005
  • Schneider BS, Higgs S. The enhancement of arbovirus transmission and disease by mosquito saliva is associated with modulation of the host immune response. Trans R Soc Trop Med Hyg 2008; 102:400-8; PMID:18342898; http://dx.doi.org/10.1016/j.trstmh.2008.01.024
  • Calvo E, Mans BJ, Andersen JF, Ribeiro JM. Function and evolution of a mosquito salivary protein family. J Biol Chem 2006; 281:1935-42; PMID:16301315; http://dx.doi.org/10.1074/jbc.M510359200
  • Thangamani S, Higgs S, Ziegler S, Vanlandingham D, Tesh R, Wikel S. Host immune response to mosquito-transmitted chikungunya virus differs from that elicited by needle inoculated virus. PloS one 2010; 5:e12137; PMID:20711354; http://dx.doi.org/10.1371/journal.pone.0012137
  • Schilte C, Couderc T, Chretien F, Sourisseau M, Gangneux N, Guivel-Benhassine F, Kraxner A, Tschopp J, Higgs S, Michault A, et al. Type I IFN controls chikungunya virus via its action on nonhematopoietic cells. J Exp Med 2010; 207:429-42; PMID:20123960; http://dx.doi.org/10.1084/jem.20090851
  • Her Z, Malleret B, Chan M, Ong EK, Wong SC, Kwek DJ, Tolou H, Lin RT, Tambyah PA, Renia L, et al. Active infection of human blood monocytes by Chikungunya virus triggers an innate immune response. J Immunol 2010; 184:5903-13; http://dx.doi.org/10.4049/jimmunol.0904181
  • Sourisseau M, Schilte C, Casartelli N, Trouillet C, Guivel-Benhassine F, Rudnicka D, Sol-Foulon N, Le Roux K, Prevost MC, Fsihi H, et al. Characterization of reemerging chikungunya virus. PLoS Pathog 2007; 3:e89; PMID:17604450; http://dx.doi.org/10.1371/journal.ppat.0030089
  • Couderc T, Chretien F, Schilte C, Disson O, Brigitte M, Guivel-Benhassine F, Touret Y, Barau G, Cayet N, Schuffenecker I, et al. A mouse model for Chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease. PLoS Pathog 2008; 4:e29; PMID:18282093; http://dx.doi.org/10.1371/journal.ppat.0040029
  • Kam YW, Ong EK, Renia L, Tong JC, Ng LF. Immuno-biology of Chikungunya and implications for disease intervention. Microbes and infection / Institut Pasteur 2009; 11:1186-96; PMID:19737625; http://dx.doi.org/10.1016/j.micinf.2009.09.003
  • Chow A, Her Z, Ong EK, Chen JM, Dimatatac F, Kwek DJ, Barkham T, Yang H, Renia L, Leo YS, et al. Persistent arthralgia induced by Chikungunya virus infection is associated with interleukin-6 and granulocyte macrophage colony-stimulating factor. J Infect Dis 2011; 203:149-57; PMID:21288813; http://dx.doi.org/10.1093/infdis/jiq042
  • Gasque P, Couderc T, Lecuit M, Roques P, Ng LF. Chikungunya virus pathogenesis and immunity. Vector Borne Zoonotic Dis 2015; 15:241-9; PMID:25897810; http://dx.doi.org/10.1089/vbz.2014.1710
  • Wauquier N, Becquart P, Nkoghe D, Padilla C, Ndjoyi-Mbiguino A, Leroy EM. The acute phase of Chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J Infect Dis 2011; 204:115-23; PMID:21628665; http://dx.doi.org/10.1093/infdis/jiq006
  • Schwartz O, Albert ML. Biology and pathogenesis of chikungunya virus. Nat Rev Microbiol 2010; 8:491-500; PMID:20551973; http://dx.doi.org/10.1038/nrmicro2368
  • Labadie K, Larcher T, Joubert C, Mannioui A, Delache B, Brochard P, Guigand L, Dubreil L, Lebon P, Verrier B, et al. Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J Clin Investig 2010; 120:894-906; PMID:20179353; http://dx.doi.org/10.1172/JCI40104
  • Selvamani SP, Mishra R, Singh SK. Chikungunya virus exploits miR-146a to regulate NF-kappaB pathway in human synovial fibroblasts. PloS one 2014; 9:e103624; PMID:25083878; http://dx.doi.org/10.1371/journal.pone.0103624
  • Chen W, Foo SS, Rulli NE, Taylor A, Sheng KC, Herrero LJ, Herring BL, Lidbury BA, Li RW, Walsh NC, et al. Arthritogenic alphaviral infection perturbs osteoblast function and triggers pathologic bone loss. Proc Natl Acad Sci U S A 2014; 111:6040-5; PMID:24733914; http://dx.doi.org/10.1073/pnas.1318859111
  • Noret M, Herrero L, Rulli N, Rolph M, Smith PN, Li RW, Roques P, Gras G, Mahalingam S. Interleukin 6, RANKL, and osteoprotegerin expression by chikungunya virus-infected human osteoblasts. J Infect Dis 2012; 206:455-7: 7-9; http://dx.doi.org/10.1093/infdis/jis368
  • Brandler S, Ruffie C, Combredet C, Brault JB, Najburg V, Prevost MC, Habel A, Tauber E, Despres P, Tangy F. A recombinant measles vaccine expressing chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine 2013; 31:3718-25; PMID:23742993; http://dx.doi.org/10.1016/j.vaccine.2013.05.086
  • Brehin AC, Rubrecht L, Navarro-Sanchez ME, Marechal V, Frenkiel MP, Lapalud P, Laune D, Sall AA, Despres P. Production and characterization of mouse monoclonal antibodies reactive to Chikungunya envelope E2 glycoprotein. Virology 2008; 371:185-95; PMID:17949772; http://dx.doi.org/10.1016/j.virol.2007.09.028
  • Pal P, Dowd KA, Brien JD, Edeling MA, Gorlatov S, Johnson S, Lee I, Akahata W, Nabel GJ, Richter MK, et al. Development of a highly protective combination monoclonal antibody therapy against Chikungunya virus. PLoS Pathog 2013; 9:e1003312; PMID:23637602; http://dx.doi.org/10.1371/journal.ppat.1003312
  • Couderc T, Khandoudi N, Grandadam M, Visse C, Gangneux N, Bagot S, Prost JF, Lecuit M. Prophylaxis and therapy for Chikungunya virus infection. J Infect Dis 2009; 200:516-23; PMID:19572805; http://dx.doi.org/10.1086/600381
  • Rudd PA, Wilson J, Gardner J, Larcher T, Babarit C, Le TT, Anraku I, Kumagai Y, Loo YM, Gale M, Jr., et al. Interferon response factors 3 and 7 protect against Chikungunya virus hemorrhagic fever and shock. J Virol 2012; 86:9888-98; PMID:22761364; http://dx.doi.org/10.1128/JVI.00956-12
  • Munoz-Jordan JL. Subversion of interferon by dengue virus. Curr Top Microbiol Immunol 2010; 338:35-44; PMID:19802576
  • Meylan E, Tschopp J. Toll-like receptors and RNA helicases: two parallel ways to trigger antiviral responses. Mol Cell 2006; 22:561-9; PMID:16762830; http://dx.doi.org/10.1016/j.molcel.2006.05.012
  • Morrison J, Aguirre S, Fernandez-Sesma A. Innate immunity evasion by Dengue virus. Viruses 2012; 4:397-413; PMID:22590678; http://dx.doi.org/10.3390/v4030397
  • Rodriguez-Madoz JR, Belicha-Villanueva A, Bernal-Rubio D, Ashour J, Ayllon J, Fernandez-Sesma A. Inhibition of the type I interferon response in human dendritic cells by dengue virus infection requires a catalytically active NS2B3 complex. J Virol 2010; 84:9760-74; PMID:20660196; http://dx.doi.org/10.1128/JVI.01051-10
  • Hoang LT, Lynn DJ, Henn M, Birren BW, Lennon NJ, Le PT, Duong KT, Nguyen TT, Mai LN, Farrar JJ, et al. The early whole-blood transcriptional signature of dengue virus and features associated with progression to dengue shock syndrome in Vietnamese children and young adults. J Virol 2010; 84:12982-94; PMID:20943967; http://dx.doi.org/10.1128/JVI.01224-10
  • Ramirez-Ronda CH, Garcia CD. Dengue in the Western Hemisphere. Infect Dis Clin North Am 1994; 8:107-28; PMID:7912702
  • Chahar HS, Bao X, Casola A. Exosomes and Their Role in the Life Cycle and Pathogenesis of RNA Viruses. Viruses 2015; 7:3204-25; PMID:26102580; http://dx.doi.org/10.3390/v7062770
  • Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, Feeley EM, Ryan BJ, Weyer JL, van der Weyden L, Fikrig E, et al. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell 2009; 139:1243-54; PMID:20064371; http://dx.doi.org/10.1016/j.cell.2009.12.017
  • Zhu X, He Z, Yuan J, Wen W, Huang X, Hu Y, Lin C, Pan J, Li R, Deng H, et al. IFITM3-containing exosome as a novel mediator for anti-viral response in dengue virus infection. Cell Microbiol 2015; 17:105-18; PMID:25131332; http://dx.doi.org/10.1111/cmi.12339
  • Fink K, Ng C, Nkenfou C, Vasudevan SG, van Rooijen N, Schul W. Depletion of macrophages in mice results in higher dengue virus titers and highlights the role of macrophages for virus control. Eur J Immunol 2009; 39:2809-21; PMID:19637226; http://dx.doi.org/10.1002/eji.200939389
  • Kalantri SP, Joshi R, Riley LW. Chikungunya epidemic: an Indian perspective. Natl Med J India 2006; 19:315-22; PMID:17343016
  • Yoon IK, Alera MT, Lago CB, Tac-An IA, Villa D, Fernandez S, Thaisomboonsuk B, Klungthong C, Levy JW, Velasco JM, et al. High rate of subclinical chikungunya virus infection and association of neutralizing antibody with protection in a prospective cohort in the Philippines. PLoS Negl Trop Dis 2015; 9:e0003764; PMID:25951202; http://dx.doi.org/10.1371/journal.pntd.0003764
  • Staikowsky F, Talarmin F, Grivard P, Souab A, Schuffenecker I, Le Roux K, Lecuit M, Michault A. Prospective study of Chikungunya virus acute infection in the Island of La Reunion during the 2005-2006 outbreak. PloS one 2009; 4:e7603; PMID:19893613; http://dx.doi.org/10.1371/journal.pone.0007603
  • Reddy V, Mani RS, Desai A, Ravi V. Correlation of plasma viral loads and presence of Chikungunya IgM antibodies with cytokine/chemokine levels during acute Chikungunya virus infection. J Med Virol 2014; 86:1393-401; PMID:24523146; http://dx.doi.org/10.1002/jmv.23875
  • Gerardin P, Fianu A, Michault A, Mussard C, Boussaid K, Rollot O, Grivard P, Kassab S, Bouquillard E, Borgherini G, et al. Predictors of Chikungunya rheumatism: a prognostic survey ancillary to the TELECHIK cohort study. Arthritis Res Ther 2013; 15:R9; PMID:23302155; http://dx.doi.org/10.1186/ar4137
  • Simon F, Parola P, Grandadam M, Fourcade S, Oliver M, Brouqui P, Hance P, Kraemer P, Ali Mohamed A, de Lamballerie X, et al. Chikungunya infection: an emerging rheumatism among travelers returned from Indian Ocean islands. Report of 47 cases. Medicine 2007; 86:123-37; PMID:17505252; http://dx.doi.org/10.1097/MD/0b013e31806010a5
  • Taubitz W, Cramer JP, Kapaun A, Pfeffer M, Drosten C, Dobler G, Burchard GD, Loscher T. Chikungunya fever in travelers: clinical presentation and course. Clin Infect Dis 2007; 45:e1-4; PMID:17554689; http://dx.doi.org/10.1086/518701
  • Mohan A. Chikungunya fever: clinical manifestations & management. Indian J Med Res 2006; 124:471-4; PMID:17213512
  • Rohatgi A, Corbo JC, Monte K, Higgs S, Vanlandingham DL, Kardon G, Lenschow DJ. Infection of myofibers contributes to increased pathogenicity during infection with an epidemic strain of chikungunya virus. J Virol 2014; 88:2414-25; PMID:24335291; http://dx.doi.org/10.1128/JVI.02716-13
  • Josseran L, Paquet C, Zehgnoun A, Caillere N, Le Tertre A, Solet JL, Ledrans M. Chikungunya disease outbreak, Reunion Island. Emerg Infect Dis 2006; 12:1994-5; PMID:17354339; http://dx.doi.org/10.3201/eid1212.060710
  • Renault P, Solet JL, Sissoko D, Balleydier E, Larrieu S, Filleul L, Lassalle C, Thiria J, Rachou E, de Valk H, et al. A major epidemic of chikungunya virus infection on Reunion Island, France, 2005-2006. Am J Trop Med Hyg 2007; 77:727-31; PMID:17978079
  • Schilte C, Staikowsky F, Couderc T, Madec Y, Carpentier F, Kassab S, Albert ML, Lecuit M, Michault A. Chikungunya virus-associated long-term arthralgia: a 36-month prospective longitudinal study. PLoS Negl Trop Dis 2013; 7:e2137; PMID:23556021; http://dx.doi.org/10.1371/journal.pntd.0002137
  • Simon F, Javelle E, Gasque P. Chikungunya Virus Infections. N Engl J Med 2015; 373:93-4; PMID:26132957; http://dx.doi.org/10.1056/NEJMc1505501
  • Chen W, Foo SS, Sims NA, Herrero LJ, Walsh NC, Mahalingam S. Arthritogenic alphaviruses: new insights into arthritis and bone pathology. Trends Microbiol 2015; 23:35-43; PMID:25449049; http://dx.doi.org/10.1016/j.tim.2014.09.005
  • Ozden S, Huerre M, Riviere JP, Coffey LL, Afonso PV, Mouly V, de Monredon J, Roger JC, El Amrani M, Yvin JL, et al. Human muscle satellite cells as targets of Chikungunya virus infection. PloS one 2007; 2:e527; PMID:17565380; http://dx.doi.org/10.1371/journal.pone.0000527
  • Hoarau JJ, Jaffar Bandjee MC, Krejbich Trotot P, Das T, Li-Pat-Yuen G, Dassa B, Denizot M, Guichard E, Ribera A, Henni T, et al. Persistent chronic inflammation and infection by Chikungunya arthritogenic alphavirus in spite of a robust host immune response. J Immunol 2010; 184:5914-27; http://dx.doi.org/10.4049/jimmunol.0900255
  • Pialoux G, Gauzere BA, Jaureguiberry S, Strobel M. Chikungunya, an epidemic arbovirosis. Lancet Infect Dis 2007; 7:319-27; PMID:17448935; http://dx.doi.org/10.1016/S1473-3099(07)70107-X
  • Sudeep AB, Parashar D. Chikungunya: an overview. J Biosci 2008; 33:443-9; PMID:19208970; http://dx.doi.org/10.1007/s12038-008-0063-2
  • Chevillon C, Briant L, Renaud F, Devaux C. The Chikungunya threat: an ecological and evolutionary perspective. Trends Microbiol 2008; 16:80-8; PMID:18191569; http://dx.doi.org/10.1016/j.tim.2007.12.003
  • Magurano F, Zammarchi L, Baggieri M, Fortuna C, Farese A, Benedetti E, Fiorentini C, Rezza G, Nicoletti L, Bartoloni A. Chikungunya from the Caribbean: the importance of appropriate laboratory tests to confirm the diagnosis. Vector Borne Zoonotic Dis 2015; 15:258-60; PMID:25897812; http://dx.doi.org/10.1089/vbz.2014.1724
  • Foissac M, Javelle E, Ray S, Guerin B, Simon F. Post-Chikungunya rheumatoid arthritis, Saint Martin. Emerg Infect Dis 2015; 21:530-2; PMID:25695499; http://dx.doi.org/10.3201/eid2103.141397
  • Kam YW, Lee WW, Simarmata D, Harjanto S, Teng TS, Tolou H, Chow A, Lin RT, Leo YS, Renia L, et al. Longitudinal analysis of the human antibody response to Chikungunya virus infection: implications for serodiagnosis and vaccine development. J Virol 2012; 86:13005-15; PMID:23015702; http://dx.doi.org/10.1128/JVI.01780-12
  • Panning M, Grywna K, van Esbroeck M, Emmerich P, Drosten C. Chikungunya fever in travelers returning to Europe from the Indian Ocean region, 2006. Emerg Infect Dis 2008; 14:416-22; PMID:18325256; http://dx.doi.org/10.3201/eid1403.070906
  • Pierro A, Rossini G, Gaibani P, Finarelli AC, Moro ML, Landini MP, Sambri V. Persistence of anti-chikungunya virus-specific antibodies in a cohort of patients followed from the acute phase of infection after the 2007 outbreak in Italy. New Microbes New Infect 2015; 7:23-5; PMID:26106482; http://dx.doi.org/10.1016/j.nmni.2015.04.002
  • Borgherini G, Poubeau P, Jossaume A, Gouix A, Cotte L, Michault A, Arvin-Berod C, Paganin F. Persistent arthralgia associated with chikungunya virus: a study of 88 adult patients on reunion island. Clin Infect Dis 2008; 47:469-75; PMID:18611153; http://dx.doi.org/10.1086/590003
  • de Andrade DC, Jean S, Clavelou P, Dallel R, Bouhassira D. Chronic pain associated with the Chikungunya Fever: long lasting burden of an acute illness. BMC Infect Dis 2010; 10:31; PMID:20170492; http://dx.doi.org/10.1186/1471-2334-10-31
  • WHO. Guidelines on Clinical Management of Chikungunya Fever. 2008; http://www.wpro.who.int/mvp/topics/ntd/Clinical_Mgnt_Chikungunya_WHO_SEARO.pdf (accessed August 25th, 2015)
  • Briolant S, Garin D, Scaramozzino N, Jouan A, Crance JM. In vitro inhibition of Chikungunya and Semliki Forest viruses replication by antiviral compounds: synergistic effect of interferon-alpha and ribavirin combination. Antiviral Res 2004; 61:111-7; PMID:14670584; http://dx.doi.org/10.1016/j.antiviral.2003.09.005
  • Rothan HA, Bahrani H, Mohamed Z, Teoh TC, Shankar EM, Rahman NA, Yusof R. A combination of doxycycline and ribavirin alleviated chikungunya infection. PloS one 2015; 10:e0126360; PMID:25970853; http://dx.doi.org/10.1371/journal.pone.0126360
  • De Lamballerie X, Boisson V, Reynier JC, Enault S, Charrel RN, Flahault A, Roques P, Le Grand R. On chikungunya acute infection and chloroquine treatment. Vector Borne Zoonotic Dis 2008; 8:837-9; PMID:18620511; http://dx.doi.org/10.1089/vbz.2008.0049
  • Ganu MA, Ganu AS. Post-chikungunya chronic arthritis–our experience with DMARDs over two year follow up. J Assoc Physicians India 2011; 59:83-6; PMID:21751641
  • Ribera A, Degasne I, Jaffar Bandjee MC, Gasque P. ; Chronic rheumatic manifestations following chikungunya virus infection: clinical description and therapeutic considerations. Med Trop 2012; 72 Spec No:83-5; PMID:22693935
  • Bouquillard E, Combe B. A report of 21 cases of rheumatoid arthritis following Chikungunya fever. A mean follow-up of two years. Joint Bone Spine 2009; 76:654-7; PMID:19945329; http://dx.doi.org/10.1016/j.jbspin.2009.08.005
  • Javelle E, Ribera A, Degasne I, Gauzere BA, Marimoutou C, Simon F. Specific management of post-chikungunya rheumatic disorders: a retrospective study of 159 cases in Reunion Island from 2006-2012. PLoS Negl Trop Dis 2015; 9:e0003603; PMID:25760632; http://dx.doi.org/10.1371/journal.pntd.0003603
  • Delang L, Segura Guerrero N, Tas A, Querat G, Pastorino B, Froeyen M, Dallmeier K, Jochmans D, Herdewijn P, Bello F, et al. Mutations in the chikungunya virus non-structural proteins cause resistance to favipiravir (T-705), a broad-spectrum antiviral. J Antimicrob Chemother 2014; 69:2770-84; PMID:24951535; http://dx.doi.org/10.1093/jac/dku209
  • Dash PK, Tiwari M, Santhosh SR, Parida M, Lakshmana Rao PV. RNA interference mediated inhibition of Chikungunya virus replication in mammalian cells. Biochem Biophys Res Commun 2008; 376:718-22; PMID:18805396; http://dx.doi.org/10.1016/j.bbrc.2008.09.040
  • Lam S, Chen KC, Ng MM, Chu JJ. Expression of plasmid-based shRNA against the E1 and nsP1 genes effectively silenced Chikungunya virus replication. PloS one 2012; 7:e46396; PMID:23056297; http://dx.doi.org/10.1371/journal.pone.0046396
  • Fong RH, Banik SS, Mattia K, Barnes T, Tucker D, Liss N, Lu K, Selvarajah S, Srinivasan S, Mabila M, et al. Exposure of epitope residues on the outer face of the chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol 2014; 88:14364-79; PMID:25275138; http://dx.doi.org/10.1128/JVI.01943-14
  • Warter L, Lee CY, Thiagarajan R, Grandadam M, Lebecque S, Lin RT, Bertin-Maghit S, Ng LF, Abastado JP, Despres P, et al. Chikungunya virus envelope-specific human monoclonal antibodies with broad neutralization potency. J Immunol 2011; 186:3258-64; http://dx.doi.org/10.4049/jimmunol.1003139
  • Lee CY, Kam YW, Fric J, Malleret B, Koh EG, Prakash C, Huang W, Lee WW, Lin C, Lin RT, et al. Chikungunya virus neutralization antigens and direct cell-to-cell transmission are revealed by human antibody-escape mutants. PLoS Pathog 2011; 7:e1002390; PMID:22144891; http://dx.doi.org/10.1371/journal.ppat.1002390
  • Fric J, Bertin-Maghit S, Wang CI, Nardin A, Warter L. Use of human monoclonal antibodies to treat Chikungunya virus infection. J Infect Dis 2013; 207:319-22; PMID:23125446; http://dx.doi.org/10.1093/infdis/jis674
  • Masrinoul P, Puiprom O, Tanaka A, Kuwahara M, Chaichana P, Ikuta K, Ramasoota P, Okabayashi T. Monoclonal antibody targeting chikungunya virus envelope 1 protein inhibits virus release. Virology 2014; 464-465:111-7; PMID:25063884
  • Goh LY, Hobson-Peters J, Prow NA, Gardner J, Bielefeldt-Ohmann H, Pyke AT, Suhrbier A, Hall RA. Neutralizing monoclonal antibodies to the E2 protein of chikungunya virus protects against disease in a mouse model. Clin Immunol 2013; 149:487-97; PMID:24239837; http://dx.doi.org/10.1016/j.clim.2013.10.004
  • Selvarajah S, Sexton NR, Kahle KM, Fong RH, Mattia KA, Gardner J, Lu K, Liss NM, Salvador B, Tucker DF, et al. A neutralizing monoclonal antibody targeting the acid-sensitive region in chikungunya virus E2 protects from disease. PLoS Negl Trop Dis 2013; 7:e2423; PMID:24069479; http://dx.doi.org/10.1371/journal.pntd.0002423
  • Goh LY, Hobson-Peters J, Prow NA, Gardner J, Bielefeldt-Ohmann H, Suhrbier A, Hall RA. Monoclonal antibodies specific for the capsid protein of chikungunya virus suitable for multiple applications. J Gen Virol 2015; 96:507-12; PMID:25480927; http://dx.doi.org/10.1099/jgv.0.000002
  • Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJ. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov 2010; 9:325-38; PMID:20305665; http://dx.doi.org/10.1038/nrd3003
  • CDC. CHIKUNGUNYA information for vector control programs. 2015; http://www.cdc.gov/chikungunya/pdfs/CHIKV_VectorControl.pdf (accessed July 9th, 2015)
  • Lang J, Wood SC. Development of orphan vaccines: an industry perspective. Emerg Infect Dis 1999; 5:749-56; PMID:10603207; http://dx.doi.org/10.3201/eid0506.990602
  • Kumar CJ, Baboo CA, Krishnan BU, Kumar A, Joy S, Jose T, Philip A, Sambasivaiah K, Hegde BM. The socioeconomic impact of the chikungunya viral epidemic in India. Open Med 2007; 1:e150-2; PMID:21673944
  • Seyler T, Hutin Y, Ramanchandran V, Ramakrishnan R, Manickam P, Murhekar M. Estimating the burden of disease and the economic cost attributable to chikungunya, Andhra Pradesh, India, 2005-2006. Trans R Soc Trop Med Hyg 2010; 104:133-8; PMID:19709705; http://dx.doi.org/10.1016/j.trstmh.2009.07.014
  • Soumahoro MK, Boelle PY, Gauzere BA, Atsou K, Pelat C, Lambert B, La Ruche G, Gastellu-Etchegorry M, Renault P, Sarazin M, et al. The Chikungunya epidemic on La Reunion Island in 2005-2006: a cost-of-illness study. PLoS Negl Trop Dis 2011; 5:e1197; PMID:21695162; http://dx.doi.org/10.1371/journal.pntd.0001197
  • Vazquez-Prokopec GM, Chaves LF, Ritchie SA, Davis J, Kitron U. Unforeseen costs of cutting mosquito surveillance budgets. PLoS Negl Trop Dis 2010; 4:e858; PMID:21049010; http://dx.doi.org/10.1371/journal.pntd.0000858
  • Roy CJ, Adams AP, Wang E, Plante K, Gorchakov R, Seymour RL, Vinet-Oliphant H, Weaver SC. Chikungunya vaccine candidate is highly attenuated and protects nonhuman primates against telemetrically monitored disease following a single dose. J Infect Dis 2014; 209:1891-9; PMID:24403555; http://dx.doi.org/10.1093/infdis/jiu014
  • Hoke CH, Jr., Pace-Templeton J, Pittman P, Malinoski FJ, Gibbs P, Ulderich T, Mathers M, Fogtman B, Glass P, Vaughn DW. US Military contributions to the global response to pandemic chikungunya. Vaccine 2012; 30:6713-20; PMID:22940380; http://dx.doi.org/10.1016/j.vaccine.2012.08.025
  • Wang E, Volkova E, Adams AP, Forrester N, Xiao SY, Frolov I, Weaver SC. Chimeric alphavirus vaccine candidates for chikungunya. Vaccine 2008; 26:5030-9; PMID:18692107; http://dx.doi.org/10.1016/j.vaccine.2008.07.054
  • Wang D, Suhrbier A, Penn-Nicholson A, Woraratanadharm J, Gardner J, Luo M, Le TT, Anraku I, Sakalian M, Einfeld D, et al. A complex adenovirus vaccine against chikungunya virus provides complete protection against viraemia and arthritis. Vaccine 2011; 29:2803-9; PMID:21320541; http://dx.doi.org/10.1016/j.vaccine.2011.01.108
  • Chattopadhyay A, Wang E, Seymour R, Weaver SC, Rose JK. A chimeric vesiculo/alphavirus is an effective alphavirus vaccine. J Virol 2013; 87:395-402; PMID:23077320; http://dx.doi.org/10.1128/JVI.01860-12
  • Garcia-Arriaza J, Cepeda V, Hallengard D, Sorzano CO, Kummerer BM, Liljestrom P, Esteban M. A novel poxvirus-based vaccine, MVA-CHIKV, is highly immunogenic and protects mice against chikungunya infection. J Virol 2014; 88:3527-47; PMID:24403588; http://dx.doi.org/10.1128/JVI.03418-13
  • Hallengard D, Lum FM, Kummerer BM, Lulla A, Lulla V, Garcia-Arriaza J, Fazakerley JK, Roques P, Le Grand R, Merits A, et al. Prime-boost immunization strategies against Chikungunya virus. J Virol 2014; 88:13333-43; PMID:25210177; http://dx.doi.org/10.1128/JVI.01926-14
  • Tretyakova I, Hearn J, Wang E, Weaver S, Pushko P. DNA vaccine initiates replication of live attenuated chikungunya virus in vitro and elicits protective immune response in mice. J Infect Dis 2014; 209:1882-90; PMID:24585894; http://dx.doi.org/10.1093/infdis/jiu114
  • Khan M, Dhanwani R, Rao PV, Parida M. Subunit vaccine formulations based on recombinant envelope proteins of Chikungunya virus elicit balanced Th1/Th2 response and virus-neutralizing antibodies in mice. Virus Res 2012; 167:236-46; PMID:22610133; http://dx.doi.org/10.1016/j.virusres.2012.05.004
  • Kumar M, Sudeep AB, Arankalle VA. Evaluation of recombinant E2 protein-based and whole-virus inactivated candidate vaccines against chikungunya virus. Vaccine 2012; 30:6142-9; PMID:22884660; http://dx.doi.org/10.1016/j.vaccine.2012.07.072
  • Tiwari M, Parida M, Santhosh SR, Khan M, Dash PK, Rao PV. Assessment of immunogenic potential of Vero adapted formalin inactivated vaccine derived from novel ECSA genotype of Chikungunya virus. Vaccine 2009; 27:2513-22; PMID:19368794; http://dx.doi.org/10.1016/j.vaccine.2009.02.062
  • Wagner JM, Pajerowski JD, Daniels CL, McHugh PM, Flynn JA, Balliet JW, Casimiro DR, Subramanian S. Enhanced production of Chikungunya virus-like particles using a high-pH adapted spodoptera frugiperda insect cell line. PloS one 2014; 9:e94401; PMID:24713807; http://dx.doi.org/10.1371/journal.pone.0094401
  • Ramsauer K, Schwameis M, Firbas C, Mullner M, Putnak RJ, Thomas SJ, Despres P, Tauber E, Jilma B, Tangy F. Immunogenicity, safety, and tolerability of a recombinant measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. Lancet Infect Dis 2015; 15:519-27; PMID:25739878; http://dx.doi.org/10.1016/S1473-3099(15)70043-5
  • Harrison VR, Eckels KH, Bartelloni PJ, Hampton C. Production and evaluation of a formalin-killed Chikungunya vaccine. J Immunol 1971; 107:643-7
  • Harrison VR, Marshall JD, Guilloud NB. The presence of antibody to Chikungunya and other serologically related viruses in the sera of sub-human primate imports to the United States. J Immunol 1967; 98:979-81
  • Harrison VR, Binn LN, Randall R. Comparative immunogenicities of chikungunya vaccines prepared in avian and mammalian tissues. Am J Trop Med Hyg 1967; 16:786-91; PMID:4965219
  • McClain DJ, Pittman PR, Ramsburg HH, Nelson GO, Rossi CA, Mangiafico JA, Schmaljohn AL, Malinoski FJ. Immunologic interference from sequential administration of live attenuated alphavirus vaccines. J Infect Dis 1998; 177:634-41; PMID:9498442; http://dx.doi.org/10.1086/514240
  • Edelman R, Tacket CO, Wasserman SS, Bodison SA, Perry JG, Mangiafico JA. Phase II safety and immunogenicity study of live chikungunya virus vaccine TSI-GSD-218. Am J Trop Med Hyg 2000; 62:681-5; PMID:11304054
  • Chang LJ, Dowd KA, Mendoza FH, Saunders JG, Sitar S, Plummer SH, Yamshchikov G, Sarwar UN, Hu Z, Enama ME, et al. Safety and tolerability of chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation trial. Lancet 2014; 384:2046-52; PMID:25132507; http://dx.doi.org/10.1016/S0140-6736(14)61185-5
  • Tsetsarkin K, Higgs S, McGee CE, De Lamballerie X, Charrel RN, Vanlandingham DL. Infectious clones of Chikungunya virus (La Reunion isolate) for vector competence studies. Vector Borne Zoonotic Dis 2006; 6:325-37; PMID:17187566; http://dx.doi.org/10.1089/vbz.2006.6.325
  • Plante K, Wang E, Partidos CD, Weger J, Gorchakov R, Tsetsarkin K, Borland EM, Powers AM, Seymour R, Stinchcomb DT, et al. Novel chikungunya vaccine candidate with an IRES-based attenuation and host range alteration mechanism. PLoS Pathog 2011; 7:e1002142; PMID:21829348; http://dx.doi.org/10.1371/journal.ppat.1002142
  • Chu H, Das SC, Fuchs JF, Suresh M, Weaver SC, Stinchcomb DT, Partidos CD, Osorio JE. Deciphering the protective role of adaptive immunity to CHIKV/IRES a novel candidate vaccine against Chikungunya in the A129 mouse model. Vaccine 2013; 31:3353-60; PMID:23727003; http://dx.doi.org/10.1016/j.vaccine.2013.05.059
  • Partidos CD, Paykel J, Weger J, Borland EM, Powers AM, Seymour R, Weaver SC, Stinchcomb DT, Osorio JE. Cross-protective immunity against o'nyong-nyong virus afforded by a novel recombinant chikungunya vaccine. Vaccine 2012; 30:4638-43; PMID:22583812; http://dx.doi.org/10.1016/j.vaccine.2012.04.099
  • Metz SW, Martina BE, van den Doel P, Geertsema C, Osterhaus AD, Vlak JM, Pijlman GP. Chikungunya virus-like particles are more immunogenic in a lethal AG129 mouse model compared to glycoprotein E1 or E2 subunits. Vaccine 2013; 31:6092-6; PMID:24099875; http://dx.doi.org/10.1016/j.vaccine.2013.09.045
  • Akahata W, Yang ZY, Andersen H, Sun S, Holdaway HA, Kong WP, Lewis MG, Higgs S, Rossmann MG, Rao S, et al. A virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat Med 2010; 16:334-8; PMID:20111039; http://dx.doi.org/10.1038/nm.2105
  • Metz SW, Gardner J, Geertsema C, Le TT, Goh L, Vlak JM, Suhrbier A, Pijlman GP. Effective chikungunya virus-like particle vaccine produced in insect cells. PLoS Negl Trop Dis 2013; 7:e2124; PMID:23516657; http://dx.doi.org/10.1371/journal.pntd.0002124
  • Brandler S, Lucas-Hourani M, Moris A, Frenkiel MP, Combredet C, Fevrier M, Bedouelle H, Schwartz O, Despres P, Tangy F. Pediatric measles vaccine expressing a dengue antigen induces durable serotype-specific neutralizing antibodies to dengue virus. PLoS Negl Trop Dis 2007; 1:e96; PMID:18160988; http://dx.doi.org/10.1371/journal.pntd.0000096
  • Brandler S, Ruffie C, Najburg V, Frenkiel MP, Bedouelle H, Despres P, Tangy F. Pediatric measles vaccine expressing a dengue tetravalent antigen elicits neutralizing antibodies against all four dengue viruses. Vaccine 2010; 28:6730-9; PMID:20688034; http://dx.doi.org/10.1016/j.vaccine.2010.07.073
  • Despres P, Combredet C, Frenkiel MP, Lorin C, Brahic M, Tangy F. Live measles vaccine expressing the secreted form of the West Nile virus envelope glycoprotein protects against West Nile virus encephalitis. J Infect Dis 2005; 191:207-14; PMID:15609230; http://dx.doi.org/10.1086/426824
  • Hilleman MR. Current overview of the pathogenesis and prophylaxis of measles with focus on practical implications. Vaccine 2001; 20:651-65; PMID:11738730; http://dx.doi.org/10.1016/S0264-410X(01)00384-X
  • Wong-Chew RM, Beeler JA, Audet S, Santos JI. Cellular and humoral immune responses to measles in immune adults re-immunized with measles vaccine. J Med Virol 2003; 70:276-80; PMID:12696117; http://dx.doi.org/10.1002/jmv.10390
  • Rager-Zisman B, Bazarsky E, Skibin A, Chamney S, Belmaker I, Shai I, Kordysh E, Griffin DE. The effect of measles-mumps-rubella (MMR) immunization on the immune responses of previously immunized primary school children. Vaccine 2003; 21:2580-8; PMID:12744894; http://dx.doi.org/10.1016/S0264-410X(03)00053-7
  • Seymour RL, Adams AP, Leal G, Alcorn MD, Weaver SC. A Rodent Model of Chikungunya Virus Infection in RAG1 -/- Mice, with Features of Persistence, for Vaccine Safety Evaluation. PLoS Negl Trop Dis 2015; 9:e0003800; PMID:26115459; http://dx.doi.org/10.1371/journal.pntd.0003800
  • Petrovsky N, Aguilar JC. Vaccine adjuvants: current state and future trends. Immunol Cell Biol 2004; 82:488-96; PMID:15479434; http://dx.doi.org/10.1111/j.0818-9641.2004.01272.x
  • Manzoli L, De Vito C, Salanti G, D'Addario M, Villari P, Ioannidis JP. Meta-analysis of the immunogenicity and tolerability of pandemic influenza A 2009 (H1N1) vaccines. PloS one 2011; 6:e24384; PMID:21915319; http://dx.doi.org/10.1371/journal.pone.0024384
  • Pellegrino P, Clementi E, Radice S. On vaccine's adjuvants and autoimmunity: Current evidence and future perspectives. Autoimmun Rev 2015; 14(10):880-8; PMID:26031899
  • Salazar-Gonzalez JA, Angulo C, Rosales-Mendoza S. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine 2015; 33(31):3650-8; PMID:26073010
  • Yusibov V, Streatfield SJ, Kushnir N. Clinical development of plant-produced recombinant pharmaceuticals: vaccines, antibodies and beyond. Hum vaccin 2011; 7:313-21; PMID:21346417; http://dx.doi.org/10.4161/hv.7.3.14207
  • Pijlman GP. Enveloped virus-like particles as vaccines against pathogenic arboviruses. Biotechnol J 2015; 10:659-70; PMID:25692281; http://dx.doi.org/10.1002/biot.201400427
  • Metz SW, Pijlman GP. Arbovirus vaccines; opportunities for the baculovirus-insect cell expression system. J Invertebr pathol 2011; 107 Suppl:S16-30; PMID:21784227; http://dx.doi.org/10.1016/j.jip.2011.05.002
  • Vicente T, Roldao A, Peixoto C, Carrondo MJ, Alves PM. Large-scale production and purification of VLP-based vaccines. J Invertebr pathol 2011; 107 Suppl:S42-8; PMID:21784230; http://dx.doi.org/10.1016/j.jip.2011.05.004
  • Barrett AD, Gould EA. Antibody-mediated early death in vivo after infection with yellow fever virus. J Gen Virol 1986; 67 (Pt 11):2539-42; PMID:3783130; http://dx.doi.org/10.1099/0022-1317-67-11-2539
  • Lidbury BA, Mahalingam S. Specific ablation of antiviral gene expression in macrophages by antibody-dependent enhancement of Ross River virus infection. J Virol 2000; 74:8376-81; PMID:10954537; http://dx.doi.org/10.1128/JVI.74.18.8376-8381.2000
  • Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11:1087-101; PMID:23151166; http://dx.doi.org/10.1586/erv.12.84
  • Dowdle WR, Coleman MT, Mostow SR, Kaye HS, Schoenbaum SC. Inactivated influenza vaccines. 2. Laboratory indices of protection. Postgrad Med J 1973; 49:159-63; PMID:4803439; http://dx.doi.org/10.1136/pgmj.49.569.159
  • Chen RT, Markowitz LE, Albrecht P, Stewart JA, Mofenson LM, Preblud SR, Orenstein WA. Measles antibody: reevaluation of protective titers. J Infect Dis 1990; 162:1036-42; PMID:2230231; http://dx.doi.org/10.1093/infdis/162.5.1036
  • Ahola T, Courderc T, Ng LF, Hallengard D, Powers A, Lecuit M, Esteban M, Merits A, Roques P, Liljestrom P. Therapeutics and vaccines against chikungunya virus. Vector Borne Zoonotic Dis 2015; 15:250-7; PMID:25897811; http://dx.doi.org/10.1089/vbz.2014.1681
  • Fischer M, Staples JE, Arboviral Diseases Branch NCfE, Zoonotic Infectious Diseases CDC. Notes from the field: chikungunya virus spreads in the Americas - Caribbean and South America, 2013-2014. MMWR Morb Mortal Wkly Rep 2014; 63:500-1; PMID:24898168
  • Levitt NH, Ramsburg HH, Hasty SE, Repik PM, Cole FE, Jr., Lupton HW. Development of an attenuated strain of chikungunya virus for use in vaccine production. Vaccine 1986; 4:157-62; PMID:3020820; http://dx.doi.org/10.1016/0264-410X(86)90003-4
  • Piper A, Ribeiro M, Smith KM, Briggs CM, Huitt E, Nanda K, Spears CJ, Quiles M, Cullen J, Thomas ME, et al. Chikungunya virus host range E2 transmembrane deletion mutants induce protective immunity against challenge in C57BL/6J mice. J Virol 2013; 87:6748-57; PMID:23552427; http://dx.doi.org/10.1128/JVI.03357-12
  • Nakao E, Hotta S. Immunogenicity of purified, inactivated chikungunya virus in monkeys. Bull World Health Organ 1973; 48:559-62; PMID:4204490
  • Muthumani K, Lankaraman KM, Laddy DJ, Sundaram SG, Chung CW, Sako E, Wu L, Khan A, Sardesai N, Kim JJ, et al. Immunogenicity of novel consensus-based DNA vaccines against Chikungunya virus. Vaccine 2008; 26:5128-34; PMID:18471943; http://dx.doi.org/10.1016/j.vaccine.2008.03.060
  • Mallilankaraman K, Shedlock DJ, Bao H, Kawalekar OU, Fagone P, Ramanathan AA, Ferraro B, Stabenow J, Vijayachari P, Sundaram SG, et al. A DNA vaccine against chikungunya virus is protective in mice and induces neutralizing antibodies in mice and nonhuman primates. PLoS Negl Trop Dis 2011; 5:e928; PMID:21264351; http://dx.doi.org/10.1371/journal.pntd.0000928
  • Bao H, Ramanathan AA, Kawalakar O, Sundaram SG, Tingey C, Bian CB, Muruganandam N, Vijayachari P, Sardesai NY, Weiner DB, et al. Nonstructural protein 2 (nsP2) of Chikungunya virus (CHIKV) enhances protective immunity mediated by a CHIKV envelope protein expressing DNA Vaccine. Viral immunology 2013; 26:75-83; PMID:23409931; http://dx.doi.org/10.1089/vim.2012.0061
  • Akahata W, Nabel GJ. A specific domain of the Chikungunya virus E2 protein regulates particle formation in human cells: implications for alphavirus vaccine design. J Virol 2012; 86:8879-83; PMID:22647698; http://dx.doi.org/10.1128/JVI.00370-12
  • Billeter MA, Naim HY, Udem SA. Reverse genetics of measles virus and resulting multivalent recombinant vaccines: applications of recombinant measles viruses. Curr Top Microbiol Immunol 2009; 329:129-62; PMID:19198565
  • Brandler S, Tangy F. Recombinant vector derived from live attenuated measles virus: potential for flavivirus vaccines. Comp Immunol Microbiol Infect Dis 2008; 31:271-91; PMID:17869338; http://dx.doi.org/10.1016/j.cimid.2007.07.012
  • Pal P, Fox JM, Hawman DW, Huang YJ, Messaoudi I, Kreklywich C, Denton M, Legasse AW, Smith PP, Johnson S, et al. Chikungunya viruses that escape monoclonal antibody therapy are clinically attenuated, stable, and not purified in mosquitoes. J Virol 2014; 88:8213-26; PMID:4135940; doi: 10.1128/JVI.01032-14
  • Gardner CL, Hritz J, Sun C, Vanlandingham DL, Song TY, Ghedin E, Higgs S, Klimstra WB, Ryman KD. Deliberate attenuation of chikungunya virus by adaptation to heparan sulfate-dependent infectivity: a model for rational arboviral vaccine design. PLoS Negl Trop Dis 2014; 8:e2719; PMID:3930508; http://dx.doi.org/10.1371/journal.pntd.0002719