future prospects and challenges of vaccines against filariasis

11
Review Article Future prospects and challenges of vaccines against filariasis S. A. BABAYAN, 1 J. E. ALLEN 1 & D. W. TAYLOR 1,2 1 Institute of Immunology and Infection Research, and Centre for Immunity, Infection & Evolution, Universityof Edinburgh, Edinburgh, UK, 2 Division of Pathway Medicine, University of Edinburgh, Edinburgh, UK SUMMARY Filarial infections remain a major public health and socio-eco- nomic problem across the tropics, despite considerable effort to reduce disease burden or regionally eliminate the infection with mass drug administration programmes. The sustainabil- ity of these programmes is now open to question owing to a range of issues, not least of which is emerging evidence for drug resistance. Vaccination, if developed appropriately, remains the most cost-effective means of long-term disease control. The rationale for the feasibility of vaccination against filarial parasites including onchocerciasis (river blindness, Onchocerca volvulus) and lymphatic filariasis (Wuchereria bancrofti or Brugia malayi) is founded on evidence from both humans and animal models for the development of protective immunity. Nonetheless, enormous challenges need to be faced in terms of overcoming parasite-induced suppression without inducing pathology as well as the need to both recognize and tackle evolutionary and ecological obstacles to successful vac- cine development. Nonetheless, new technological advances in addition to systems biology approaches offer hope that optimal immune responses can be induced that will prevent infection, disease and or transmission. Keywords experimental models, Filariasis, vaccine INTRODUCTION Filariasis presents major public health problems in more than 90 developing countries. Current control of these infections relies on mass treatment with ivermectin (oncho- cerciasis) or diethylcarbamazine (DEC) (lymphatic filaria- sis), either alone or in combination with albendazole. Major international initiatives such as the African Programme for Onchocerciasis Control (APOC), the Onchocerciasis Elimi- nation Program for the Americas (OEPA) and the Global Programme to Eliminate Lymphatic Filariasis (GPELF) have been established to provide sustained delivery of treat- ment with the aim of eliminating these infections as public health problems (1,2). However, despite the demonstrable achievements of these programmes (1,3), an estimated 160 million people remain infected and a further billion are at risk of infection (4). This situation argues strongly for an alternate control strategy. Vaccination, where it exists, is the most cost-effective method of controlling an infection. Although eight filarial species can parasitize humans, onchocerciasis (Onchocerca volvulus) and lymphatic filariasis (Wuchereria bancrofti and Brugia malayi) are the main targets of vaccine research efforts as they are the major causes of morbidity. Proof-of- Principle of vaccination against filariae has been demon- strated in animal models with live attenuated (5,6) and with recombinant and DNA vaccines (7), and there is con- siderable knowledge about the mechanisms that underlie vaccine-induced protective immunity as discussed below. This knowledge, combined with the application of post- genomic technologies to antigen identification, analysis and delivery, means there has never been greater optimism nor a better opportunity to develop vaccines against filari- al infections. THE NEED FOR A VACCINE Existing drugs and control programmes have important limitations (4,8,9). The single annual dose of ivermectin that is used in mass onchocerciasis treatment programmes is very effective at killing microfilariae and hence reducing morbidity associated with skin and eye disease. However, ivermectin does not kill adult worms, and while prolonged use can reduce female fecundity, sterilization may not be permanent, and once treatment stops, microfilariae can repopulate the skin and return to levels measure before Correspondence: Simon Babayan, Institute of Immunology and Infection Research, and Centre for Immunity, Infection & Evolution, University of Edinburgh, Edinburgh EH9 3JT, UK (e-mail: [email protected]). Disclosures: None. Received: 3 October 2011 Accepted for publication: 1 December 2011 Parasite Immunology, 2012, 34, 243–253 DOI: 10.1111/j.1365-3024.2011.01350.x Ó 2011 Blackwell Publishing Ltd 243

Upload: s-a-babayan

Post on 02-Oct-2016

220 views

Category:

Documents


5 download

TRANSCRIPT

Page 1: Future prospects and challenges of vaccines against filariasis

Review Article

Future prospects and challenges of vaccines against filariasis

S. A. BABAYAN,1 J. E. ALLEN1 & D. W. TAYLOR1,2

1Institute of Immunology and Infection Research, and Centre for Immunity, Infection & Evolution, University of Edinburgh, Edinburgh,UK, 2Division of Pathway Medicine, University of Edinburgh, Edinburgh, UK

SUMMARY

Filarial infections remain a major public health and socio-eco-nomic problem across the tropics, despite considerable effortto reduce disease burden or regionally eliminate the infectionwith mass drug administration programmes. The sustainabil-ity of these programmes is now open to question owing to arange of issues, not least of which is emerging evidence fordrug resistance. Vaccination, if developed appropriately,remains the most cost-effective means of long-term diseasecontrol. The rationale for the feasibility of vaccination againstfilarial parasites including onchocerciasis (river blindness,Onchocerca volvulus) and lymphatic filariasis (Wuchereriabancrofti or Brugia malayi) is founded on evidence from bothhumans and animal models for the development of protectiveimmunity. Nonetheless, enormous challenges need to be facedin terms of overcoming parasite-induced suppression withoutinducing pathology as well as the need to both recognize andtackle evolutionary and ecological obstacles to successful vac-cine development. Nonetheless, new technological advancesin addition to systems biology approaches offer hope thatoptimal immune responses can be induced that will preventinfection, disease and ⁄ or transmission.

Keywords experimental models, Filariasis, vaccine

INTRODUCTION

Filariasis presents major public health problems in morethan 90 developing countries. Current control of theseinfections relies on mass treatment with ivermectin (oncho-cerciasis) or diethylcarbamazine (DEC) (lymphatic filaria-

sis), either alone or in combination with albendazole. Majorinternational initiatives such as the African Programme forOnchocerciasis Control (APOC), the Onchocerciasis Elimi-nation Program for the Americas (OEPA) and the GlobalProgramme to Eliminate Lymphatic Filariasis (GPELF)have been established to provide sustained delivery of treat-ment with the aim of eliminating these infections as publichealth problems (1,2). However, despite the demonstrableachievements of these programmes (1,3), an estimated 160million people remain infected and a further billion are atrisk of infection (4). This situation argues strongly for analternate control strategy.

Vaccination, where it exists, is the most cost-effectivemethod of controlling an infection. Although eight filarialspecies can parasitize humans, onchocerciasis (Onchocercavolvulus) and lymphatic filariasis (Wuchereria bancrofti andBrugia malayi) are the main targets of vaccine researchefforts as they are the major causes of morbidity. Proof-of-Principle of vaccination against filariae has been demon-strated in animal models with live attenuated (5,6) andwith recombinant and DNA vaccines (7), and there is con-siderable knowledge about the mechanisms that underlievaccine-induced protective immunity as discussed below.This knowledge, combined with the application of post-genomic technologies to antigen identification, analysisand delivery, means there has never been greater optimismnor a better opportunity to develop vaccines against filari-al infections.

THE NEED FOR A VACCINE

Existing drugs and control programmes have importantlimitations (4,8,9). The single annual dose of ivermectinthat is used in mass onchocerciasis treatment programmesis very effective at killing microfilariae and hence reducingmorbidity associated with skin and eye disease. However,ivermectin does not kill adult worms, and while prolongeduse can reduce female fecundity, sterilization may not bepermanent, and once treatment stops, microfilariae canrepopulate the skin and return to levels measure before

Correspondence: Simon Babayan, Institute of Immunology andInfection Research, and Centre for Immunity, Infection &Evolution, University of Edinburgh, Edinburgh EH9 3JT, UK(e-mail: [email protected]).Disclosures: None.Received: 3 October 2011Accepted for publication: 1 December 2011

Parasite Immunology, 2012, 34, 243–253 DOI: 10.1111/j.1365-3024.2011.01350.x

� 2011 Blackwell Publishing Ltd 243

Page 2: Future prospects and challenges of vaccines against filariasis

treatment within 1 year. This increases the risk of new dis-ease episodes for the individual as well as resumption oftransmission in the community. Another limitation of iver-mectin is that while generally well tolerated and safe foruse, in a large area of equatorial west and central Africa(10) where O. volvulus is co-endemic with Loa loa, there isa risk of serious adverse reactions to treatment, includingcoma and death associated with massive death of L. loamicrofilariae (11).

Another major concern is emergence of resistance toivermectin. It is now over 25 years since ivermectin hasbeen in regular and widespread use against onchocerciasis,and there is a small but growing literature describinggenetic selection of O. volvulus by this drug (12). The per-sistence of microfilariae in the skin after repeated treat-ment with ivermectin (13) raises questions about theemergence of resistance, and although some debate sur-rounds the explanations for the so-called suboptimalresponses to ivermectin, experience in the veterinary fieldpoints towards the inevitable development of drug resis-tance among filariae.

Fortunately, there has been a long-standing search fornew drugs and, in particular, effective macrofilaricides. Inthis context, a major advance was made with the demon-stration, in cattle, that tetracycline treatment aimed at thefilarial endosymbiont Wolbachia pipensis resulted in deathof adult O. ochengi (14). Wolbachia is found in the major-ity of filariae species (15), but crucially it is not present inL. loa (16). In humans, a 6-week course of doxycyclineresulted in death of 60% adult O. volvulus (17). Doxycy-cline is safe, well tolerated, and its absence from L. loaprovides an alternative for control of onchocerciasis inareas co-endemic for loiasis (18).

For the treatment of lymphatic filariasis, DEC or iver-mectin is used, either alone or in combination with alben-dazole (19). A single dose of DEC significantly reducesblood microfilaraemia over a year, but not all adult wormsare killed (20). Adverse reactions may occur, and theseverity of local inflammatory responses associated withO. volvulus microfilariae in the eye precludes the use ofDEC in regions of Africa where lymphatic filariasis andonchocerciasis are co-endemic. To avoid such risks, iver-mectin is given alone or in combination with albendazole.However, W. bancroti and L. loa are co-endemic in 10African countries, and as mentioned in the case of oncho-cerciasis, the risk of severe adverse reactions associatedwith death of L. loa microfilariae following ivermectin usehas prevented the initiation of lymphatic filariasis masstreatment programmes (21).

Doxycycline again provides an alternate therapy againstW. bancrofti and B. malayi (9,22) with the additional bene-fit of its ability to ameliorate pathology (23,24). However,

doxycycline cannot be given to children under the age of 9(17) who, together with pregnant women and lactatingmothers, are excluded from mass treatment programmes.Children below 5 years are also excluded from ivermectintreatment. This leaves a significant proportion of individu-als exposed to infection. For example, in Gabon where theentire country is co-endemic for O. volvulus and L. loa(10), 20% of the population is under 10 years (UnitedNations, http://esa.un.org/unpd/wpp/index.htm), and simi-lar age profiles are found throughout filarial endemicregions of Africa. Therefore, untreated children representa large reservoir of microfilariae that can contribute totransmission. Furthermore, for the individual, the conse-quences of not receiving treatment would be the prospectof developing progressive filarial disease and more generallong-term health problems as well as associated socio-eco-nomic disadvantage.

The proposition that mass drug treatment can eliminatefilarial infections from Africa is contentious (25,26). Formass treatment to be effective at reducing transmission oflymphatic filariasis, it must be maintained for at least5 years, while it may take at least 15–17 years to reducetransmission of onchocerciasis (WHO estimates). However,reduced transmission does not mean control or elimina-tion of these diseases as public health problems. Introduc-tion of doxycycline as a prescribed treatment for bothdiseases would make a difference for the individual andcommunity, but elimination of these infections from Africausing these tools only seems most unlikely. Vaccinationwould make a major contribution to control of filarialinfections, and its inclusion in national immunizationprogrammes would provide preschool children withprotection.

EVIDENCE FOR PROTECTIVE IMMUNITY

In humans

Filarial infections are characterized by a spectrum of clini-cal and parasitological presentations (4). At one end ofthis spectrum are the severe skin lesions of onchocerciasisand the elephantiasis of lymphatic filariasis. Patients pre-senting with these pathologies have strong Th2 typeresponses including elevated levels of IgE with absent orlow microfilaraemia. Inflammatory responses underlie thepathology and physiological changes seen in hyper-reactive(localized) onchocerciasis (sowda) (27) and lymphoedemaand elephantiasis of lymphatic filariasis (28). Fortunately,such severe disease does not afflict a majority of infectedindividuals and many present with milder symptoms,despite the presence of sometimes massive numbers ofmicrofilariae in the skin (onchocerciasis) or blood (lymphatic

S. A. Babayan et al. Parasite Immunology

244 � 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253

Page 3: Future prospects and challenges of vaccines against filariasis

filariasis). Such individuals frequently exhibit a markedfilarial-specific T-cell unresponsiveness with impairment ofboth Th1 and Th2 pathways (29). Initially, this regulationwas ascribed to predominant IL10 responses, but it isbecoming clear that a range of regulatory events areinvolved including TGF-b, CTLA-4, natural regulatoryT cells and alternatively activated myeloid cells, withenhanced production of IgG4 characteristic of the down-modulated immune response (29,30).

Somewhere [immunologically] between the hyper-reac-tive diseased state and immunosuppressed parasitized indi-viduals lie a third but rare group of individuals whodespite having lived their entire lives (or prolonged peri-ods) in areas endemic for onchocerciasis or lymphatic fila-riasis, and continually exposed to infected vectors, showno parasitological or clinical signs of infection. These indi-viduals are described as putative immune or endemic nor-mals (31) and are frequently cited as providing evidencefor the existence of protective immunity against filarialinfections. If true, these individuals should provide infor-mation about effector mechanisms capable of destroyinginvading L3 that could be vital for successful developmentof vaccines.

In one study, endemic normals were found to producestronger Th2 responses (e.g. more IL5) against L3 antigensthan individuals with patent O. volvulus infections (32).However, half of the endemic normals tested also haddemonstrable Th1 responses as judged by production ofIFNc. Such mixed Th1 ⁄ Th2 responses had been previouslyobserved in endemic normals in other cross-sectional stud-ies of onchocerciasis and lymphatic filariasis (e.g. (33–35)and can be seen in murine models of protection. Whereinvestigated, endemic normals lack the elevated levels offilarial-specific IgG4 associated with a regulated Th2environment seen in individuals with patent infections andpresenting with no or mild disease. In general, endemicnormals have low IgG4 ⁄ IgE ratios, and in one study(36,37), this profile was associated with elevated c-inter-feron levels and again indicative of a mixed Th1 ⁄ Th2response. The concept, or reality, of endemic normalsremains controversial because there is no absolute suretythat an individual is not infected, nor is it practical andethical to carry out the longitudinal studies that mayresolve unanswered questions. It is this situation that bene-fits from and necessitates animal experimentation.

Cattle model

Putative immune humans and endemic normals havedirect counterparts in cattle exposed to O. ochengi (38,39).O. ochengi is the closest known relative to O. volvulus (40)and transmitted by the same zoophilic blackfly vector

(Simulium damnosum sl.). The parasite has a wide distribu-tion in sub-Saharan Africa, and an epidemiological studyin Cameroon (41) has even suggested that the reducedprevalence of onchocerciasis was a consequence of expo-sure to O. ochengi L3: an example of zooprophylaxis (42)?

Adult O. ochengi are found in intradermal nodules thatcan be easily quantified as a measure of drug or vaccineefficacy (43). The immunological cross-protection suggestedby Wahl et al. (41) was subsequently demonstrated follow-ing vaccination of cattle with live O. volvulus L3 (44).Furthermore, vaccination with irradiated O. ochengi L3induced good protection against natural challenge (64%reduction in mean nodule load compared with controls)and 100% protection against experimental challenge (45).

The cellular basis of this protection has yet to be estab-lished. Early parasite-specific response of cattle infectedwith O. ochengi exhibit a mixed Th1 ⁄ Th2 profile, but theonset of patency is coincident with marked inhibition ofIFNc and IL4 production, which has similarities to thehypo-responsiveness seen in patent human filarial infec-tions (46). Eosinophils have been implicated in the killingof adult O. ochengi following temporary depletion ofWolbachia by tetracycline (47), but whether such a Th2-driven response follows vaccination of cattle with irradi-ated L3 remains to be demonstrated.

Rodent models

Vaccination of cattle with irradiation-attenuated L3 followsan established route to immunization against parasites thatwas first exploited in the late 1950s when irradiated L3 lar-vae of Dictyocaulus viviparus were introduced as a commer-cial vaccine against cattle lung worm (48). Subsequently,immunization with irradiation-attenuated infective larvaehas been shown to stimulate significant levels of protectionagainst both platyhelminth [e.g. schistosomes (49)] andnematode species, including filariae (50), and despite theincomplete nature of this protection, irradiated larvaeremain the gold standard by which other vaccination proto-cols are measured. Just how irradiated L3 larvae drive pro-tective responses remains unclear. Immunization withsoluble filarial antigens induces good antibody responses,but these are not protective and infection with nonirradiat-ed L3 larvae rarely evoke a demonstrable protectiveresponse. Early studies performed with irradiated L3 ofO. volvulus (51) and B. malayi (52) in mice suggested thatprotection is associated with a Th2 response that mayinvolve IgE and eosinophils (53,54) – a conclusion alsodrawn from studies on Acanthocheilonema viteae in Meri-ones unguiculatus (55) and B. pahangi in cats (56).

The absence of reagents has limited more detailedimmunological investigation of filarial infections in cats

Volume 34, Number 5, May 2012 Vaccination against filariasis

� 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253 245

Page 4: Future prospects and challenges of vaccines against filariasis

and jirds, and consequently, most attention has focused onmurine models. However, the L3 of human filariae cannotmature and produce patent infections in immunocompe-tent mice, and this has hampered immunological investiga-tions. This constraint was removed when Bain et al.adapted Litomosoides sigmodontis, a natural filariae of thecotton rat Sigmodon hispidus, to the laboratory mouse (57)and opened filarial research to the full extent of immuno-logical tools that are available for laboratory mice, includ-ing genetic manipulation of the host. Patent infections candevelop in BALB ⁄ c mice, while C57BL ⁄ 6 mice are resis-tant to patent infection (58). Amicrofilaraemic infectionalso occurs in BALB ⁄ c mice, and thus, the parasitologicalpresentations seen in human filarial infections are repli-cated by L. sigmodontis in mice.

The L. sigmodontis model has shown that resistance canbe abrogated by knocking out the IL4 gene (59). Indeed, aconsistent finding from both L. sigmodontis studies andearlier work using B. malayi in mice is that IL4 and ⁄ orIL13 are required for containment of the larval stages,both incoming L3s and microfilariae (60–63). Such resultsare in keeping with the general finding that Th2 responsesare needed for immunity to most helminth species (64).However, significant complexity is added by the fact thatBALB ⁄ c mice, which are genetically susceptible, produceIL4 in abundance (59). Progress towards resolution of thisconundrum was made by the demonstration that suscepti-bility relies on the establishment of an earlyCD4+ FoxP3+ T regulatory response (65) and that long-term adult parasite survival requires both T regulatorycells and maintenance of a T-cell hypo-responsive state(66). These observations are arguably some of the mostimportant in filarial immunology because recognition ofinvolvement of Tregs and associated regulatory pathwaysprovide an explanation for the spectrum of parasitologicaland clinical presentations seen in human filarial infections.Furthermore, abrogation of all or part of these regulatoryprocesses may provide the key to successful vaccination.

Mice vaccinated with irradiated L. sigmodontis L3 rou-tinely lead to 70% reduction in worm burden when com-pared with controls when the challenge infection isperformed 2 weeks after the immunization (5,67),although when the challenge is given 5 months after vacci-nation, protection falls to 50–55% (6). This protectioninvolves an antibody-dependent eosinophil-mediated pro-cess in the skin (68–71) and occurs immediately after thedelivery of the infective L3 (5). Involvement of antibodyand eosinophils in protective immunity is consistent withprevious studies using O. volvulus larvae in mice (53,54)and A. viteae in jirds (72).

To date, most studies have focussed on the incomingL3s, but there are good arguments for targeting microfila-

riae, both to block transmission and to reduce risk ofpathology (e.g. onchocerciasis skin disease or tropical pul-monary eosinophilia associated with lymphatic filariasis).Studies in which microfilaria are directly injected in theblood stream of resistant mice have shown that early andrapid (6 day) clearance of microfilariae is an innateresponse that does not depend on production of nitricoxide (63,73,74). A fascinating twist to the story is thatthe presence of adult females enhances microfilarial sur-vival in the blood, which is associated with elevated levelsof IL10 (63). The timing of these events is indicative ofT-independent responses, but there is also good evidencethat IL4- or IL13-driven pathways are necessary for thecontrol of microfilaraemia following natural challengewith L. sigmodontis L3 in both susceptible and resistantstrains (59,61,75,76). Some argue that the IL4 responsesnegatively impact on fecundity rather than being responsi-ble for direct attack on microfilariae (59,61,77). However,IL4 may also act through promoting antibody-dependentcellular cytotoxicity responses that can mediate clearanceof microfilariae (78).

Critically, different effector mechanism may be operat-ing in different models, and at the level of the individual,there may be temporal changes in the relative dominanceof one effector response over another during the course ofan infection. Nonetheless, there is clear evidence that theparasite-driven immunoregulation plays a central role inthe parasitological outcome of filarial infections and thatboth innate and acquired responses are under the influ-ence of the regulatory networks that determine clinicaloutcomes (64). For the most part, infected hosts manageto maintain a balance of regulatory and effector responsesthat, while not completely controlling infection, can pre-vent severe disease. Sometimes, for a fortunate few, thisbalance favours complete elimination of infection.

THE CHALLENGES

Vaccine Targets

Given the complexity of immune responses that areneeded to resist chronic infection and target different par-asite life stages without the induction of immunopathol-ogy, it is no surprise that there is still no vaccine againstfilariasis. Efforts to develop vaccines have focussed on arange of proteins that are associated with immune protec-tion in animal models or are abundant at the L3 stage.These experimental vaccines produced either as recombi-nant proteins or DNA plasmids give highly variableprotection but can sometimes give impressive resultsespecially when combined into multivalent vaccines (seeTable 1) (7,79,80). Defining protective antigens is a

S. A. Babayan et al. Parasite Immunology

246 � 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253

Page 5: Future prospects and challenges of vaccines against filariasis

daunting task when one considers that nematodes havegenome sizes nearly as large as their hosts and indeedmany more candidates will come from the systems biologyapproach discussed below. A particular problem for filarialparasites is which stage to target. Most efforts havefocussed on the incoming L3s, but as discussed above,there are good arguments for targeting MF, both to blocktransmission and to reduce pathology, particularly in thecase of onchocerciasis. Immunity induced by irradiatedL3, which is the current gold standard of antifilarial vacci-nation, is often partial and acts primarily during the earlystages of infection (5). In genetically susceptible hosts, lar-vae that survive vaccination migrate away from the skinand thereby escape further vaccine-induced destruction.This is significant in light of our recent finding that thenematodes are able to respond to the presence ofeosinophils by accelerating their development and increas-ing their fecundity (6,81). As a consequence, the parasite’sresponse to host immunity may reduce vaccine efficacyunless vaccines can also target later life stages moreefficiently.

As described above, the biggest challenge to effectivevaccination may be parasite-driven immunoregulation.Collective work to date has led to the proposal that specif-ically targeting parasite-derived immune modulators(82,83) could allow the host to mount a protective Th2response. Although such a vaccine may allow the host togenerate protective responses more easily against multipleparasite life stages, experimental manipulation shows thatthe balance between regulatory and effector cells is notonly driven by the parasite but, in large part, is deter-mined by the host (66). Thus, simply reducing parasiteimmune modulation may not be sufficient for a vaccine toinduce full protection. Further, overcoming parasite-induced regulation may be an important prerequisite foreffective vaccination, although some level of regulationmust be maintained to avoid inducing pathology. Indeed,mammals may have evolved to respond to nematodes withregulatory rather than effector mechanisms because resis-tance and the associated immunopathology are too costly.So it becomes essential to understand the mechanisms ofparasite tolerance (84), such as T regulatory cells, anddetermine whether vaccines can work with or despitethem.

Pathology

The severe pathology seen in onchocerciasis and lymphaticfilariasis is associated with elevated IgE responses, and yetmany nematode antigens and potential vaccine targets arethemselves allergens with the capacity to induce IgE. Forexample, the venom allergen homologues (VALs), also

called Ancylostoma-secreted proteins (ASPs), are strongcontenders for an anti-filarial vaccine. Considerable hopewas placed on the idea that the filarial field could piggyback on the human trials underway using the Necatoramericanus VAL for immunization against hookworminfection (85). However, these trials faced a set back asindividuals with pre-exisiting IgE responses to the vaccinecandidate exhibited allergic responses that led to the dis-continuation of those trials (86). The hookworm vaccineresearch provides both lessons and guidance for the future.Trials with two recombinant antigens that are protective inanimal models but do not induce IgE in humans are mov-ing forward (86). This suggests that an approach in whichprotection is generated by non-IgE responses is feasibleand can avoid the risks associated with using allergens asvaccine candidates. Nonetheless, considerably more immu-nological research is needed to generate a fuller under-standing of how to generate a protective response andavoid allergic hypersensitivity.

Adjuvants

A majority of protection studies support the concept thatinduction of the Th2 arm of the immune system is anappropriate strategy for vaccination against helminthsincluding filariasis. The basic parameters that lead to Th2cell activation are well known (87), but there are still majorgaps in our understanding of what specific components areneeded to generate and fine-tune the response. For exam-ple, despite indications to the contrary, dendritic cells (DC)are needed to initiate a Th2 response (88), but the specificsignals provided to the DC, and by the DC to the naiveT cell, are still unclear (89). Further, we now understandthat Th2 immunity does not reflect a single T-cell pheno-type but that Th2 cells can be fine-tuned to produce a verydifferent array of cytokines and newly defined T-cell sub-sets such as Th9 may be involved in antihelminth immu-nity. Determining which type of immune response is themost effective against different filarial stages of parasites isno small challenge.

Further, unlike the more advanced fields of viral andbacterial vaccinology in which the triggers of innateimmunity such as TLRs are known, we do not know theinnate signals that promote Th2 responses against filariae.Nonetheless, we do know that the cytokine milieu is a crit-ical factor in inducing a particular T-cell subset (87). Withgreater knowledge of how both effector and regulatorypathways influence expression of protective immunityagainst filariae, the potential exists to orchestrate an effec-tive response by delivery of the appropriate cytokines andsecondary signals to drive naive T cells towards the Th2lineage.

Volume 34, Number 5, May 2012 Vaccination against filariasis

� 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253 247

Page 6: Future prospects and challenges of vaccines against filariasis

From the viewpoint of helminth vaccination, alum, oneof the very few adjuvants licensed for use in humans, is veryeffective at driving a Th2 response although how this isachieved is unknown (90). Fortunately, this is an activeresearch area, and the findings may contribute to the devel-opment of new more effective Th2-inducing adjuvants.

MEETING THE CHALLENGES

Molecular engineering

Our growing understanding of the mechanisms that enableparasites to evade the lethal effects of their host’s immunesystems and particularly their ability to modulate effectorresponses provide an opportunity to target specific para-site and host pathways and interactions. DNA technologyallows the use of two complementary strategies: immuniz-ing against immunomodulators and refining adjuvants.Parasite excretory-secretory products have been identifiedas a source of immunomodulators, but these moleculescan be poorly immunogenic or even block their own pro-cessing by antigen-presenting cells (APC) as may be thecase for cystatins (91,92). Such observations indicated thatcareful structural analyses may be required to identifyregions (e.g. enzymatic or inhibitory sites, post-transla-tional modifications) responsible for immunomodulatoryactivity, so that these may be removed and the immunoge-nicity of protein enhanced. For example, B. malayi cysta-tins have two protease-inhibitory sites, which can beinactivated by point mutations (92). In the L. sigmodontismodel, the inactivated form of the recombinant protein isproving to be far more immunogenic, and thus more pro-tective, than the native protein (unpublished data).

To further overcome the poor immunogenicity of para-site products, vaccines may be formulated or constructedto specifically target antigen-presenting cells such as DC(93) and thereby induce the appropriate level of responsethrough manipulation of costimulatory pathways. Suchapproaches are, in principle, applicable to a wide range ofdiseases, and a wide range on immune pathways, whetherthey utilize recombinant protein or DNA vaccines.

Systems biology

The widespread availability of gene sequencing combinedwith the functional analysis of specific parasite proteinssuch as those the parasite secretes into its environment isallowing the identification of many new vaccine targets.This approach is being expanded exponentially by the evergreater affordability of high-throughput sequencing plat-forms, which is currently being applied to filarial andhost transcriptomes (http://www.filaria.eu/projects/projects/

epiaf.html). We are reaching a point where the greatest diffi-culty is choosing which candidate genes to prioritize. Fur-ther, the exponential increase in our understanding ofimmune gene function and regulation, of cellular activationcascades and of determinants of resistance and pathologyoffers a myriad of possible vaccine approaches. A numberof parasite immune modulatory molecules are currentlybeing tested. However, only a fraction of the total molecularrepertoire can be expected to be tested experimentally, andthe complexity of host effector and regulatory systems simi-larly limits direct manipulation. The solution to this prob-lem may lie with application of systems and pathwayanalyses (94). This approach, which is already providingexceptional benefit to investigators of viral [e.g. (95)] andbacterial diseases [e. g. (96,97)], can now be exploited tohelp disentangle the interactions occurring between theimmune system and eukaryotic parasites.

Immunologists, and in particular those who wish tomanipulate immune responses to combat disease, areincreasingly requiring powerful analytical methods todecide which elements of the immune system to target,whether individual genes or entire pathways. Filarial nema-todes trigger type–2 immune responses, which are necessaryfor controlling the infection. However, there is a trade-offbetween parasite killing and the risk of type–2 immunopa-thology, and most pathology caused by filarial parasitesstems from poorly regulated immune responses. Designingvaccines against these pathogens therefore would greatlybenefit from a systems biology approach to the determinantsof disease and ⁄ or resistance. Such an approach could there-fore inform of the risks of reducing host-generated immuneregulation (e.g. by targeting the TGF-b pathway) or of theimmune pathways that may still mediate protection butwithout triggering parasite developmental cues (81). To gen-erate and test hypotheses about what constitutes a safe andprotective immune response will require strong interdisci-plinary links. Indeed, collaboration between classical parasi-tologists, medics, systems biologists and molecularbiologists is no longer a luxury but a necessity.

Biological Constraints – the long view

The potential for a revolution in vaccine design is huge,and the use of recombinant protein or DNA vaccinesalong with our technological advances in basic vaccinebiology will solve many of the logistical problems that pre-vented the use of attenuated vaccines against filariasis.

Evolution

However, it should be remembered that parasites haveevolved sophisticated mechanisms to evade their host’s

S. A. Babayan et al. Parasite Immunology

248 � 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253

Page 7: Future prospects and challenges of vaccines against filariasis

immune responses. There is a risk that imperfect vaccina-tion may drive the evolution of filarial adaptations thatallow their continued transmission. For instance, a vaccine

that targets one immunosuppressive protein may selectparasites that favour a different immunosuppressive mole-cule. Further, by reducing parasite longevity, we could

Table 1 Examples of protective immunity in evoked by selected filarial recombinant proteins and ⁄ or plasmids carrying filarial antigengenes alone or as multivalent formulations

Antigen Delivery Host Protection AssessmentPredominantresponse(s) References

Brugia malayi BmVAL-1vespid venom allergenhomologue-like protein(also described as VAH)

DNA Mice 39% L3 in IP chambers IgG1, IgG2aIFN-�, IL5

(79)

BmVAL-1 Protein Mice 48% L3 in IP chambers (79)BmVAL-1 DNA + protein

boostMice 54% L3 in IP chambers (79)

BmVAL-1 DNA Jirds 50% Adult worms (79)BmVAL-1 Protein Jirds 40% Adult worms (79)BmVAL-1 DNA + protein

boostJirds 52% Adult worms (79)

BmALT-2 (abundant larvaltranscript)

DNA Mice 51% L3 in IP chambers IgG1, IgG3 (79)

BmALT-2 Protein Mice 65% L3 in IP chambers (79)BmALT-2 + Bm VAL1 DNA + Protein

boostMice 74% L3 in IP chambers (79)

BmALT-2 DNA Jirds 58% Adult worms (79)BmALT-2 Protein Jirds 72% Adult worms (79)BMALT-2 DNA + Protein

boostJirds 78% Adult worms (79)

BmALT-2 + Bm VAL1(prime and boost)

DNA + proteinboost

Mice 82% L3 in IP chambers IgG1, IgG2a, IgG3 (79)

BmALT-2 + Bm VAL1(prime and boost)

DNA + proteinboost

Jirds 85% Adult worms (79)

BmALT-2 + BmVAH DNA Jirds 57% Adult worms IgG2a, IgG2b (7)BmALT-2 + BmVAH Protein Jirds 80% Adult worms IgG1, IgG2a, IgG2b,

IgG3(7)

BmALT-2 Protein Jirds 70% L3 in IP chambers IgG1, IgG3 (7)BmVAH Protein Jirds 60% L3 in IP chambers IgG2a (7)BmALT-2 DNA Mice 34% L3 in IP chambers IgG2a, IgG2b, IgA (107)BmALT-2 Protein Jirds 75% Adult worms IgG1 IL4, IL5 (107)BmALT-2 DNA Jirds 57% Adult worms IgG2a, IFN-g (107)BmALT-2 DNA + protein

boostJirds 64% Adult worms Mixed Th1 ⁄ Th2 (107)

Bm TPX Thioredoxinperoxidase

DNA Mice 37% L3 in IP chambers IgG2a, IgG2b, IgA (108)

BmALT-2 and thioredoxinperoxidase (TPX)

DNA Mice 78% L3 in IP chambers IgG2a, IgG2b, IgA (108)

Onchocerca volvulusGlutathione-S transferase(GST)

Protein Jirds 82.75% Adult worms Mixed Th1 ⁄ Th2 asassessed in miceimmunized withrecombinant protein

(109)

OV-FBA-1 fructose-1,6-bisphosphate aldolase

Protein Mice 50% L3 in IP chambers nd (110)

Recombinant DNA vaccines have been administered by subcutaneous (sc) injection. Inoculation with recombinant proteins has also beenby the sc route. DNA refers to recombinant DNA plasmids containing the filarial antigen gene sequence. Protein refers to recombinantfilarial antigen. Assessment of protective immunity in the mice has been by counting the number of surviving L3 larvae contained in micro-pore chambers that may measure up to 14 mm diameter and 2 mm depth (variation will occur) and which have been surgically implantedinto subcutaneous or tissue or into the peritoneal cavity. In the jird (Meriones unguiculatus) model of B. malayi, adult worm burden is usedto assess efficacy of vaccination. Levels of protection are expressed as a percentage recovery or reduction in parasite burden of vaccinatedanimals versus unvaccinated control animals. For additional information on Onchocerca vaccine trails see (80).

Volume 34, Number 5, May 2012 Vaccination against filariasis

� 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253 249

Page 8: Future prospects and challenges of vaccines against filariasis

select parasites with shorter natural life spans and higherfecundity (98), thereby potentially increasing transmissionand ⁄ or pathology. Our work on L. sigmodontis immune-dependant phenotypic plasticity has shown that such lifehistory shifts can occur within the parasite’s life span (81).Although combined drug and vaccine treatment mayreduce vaccine-escape evolution, such optimism may beunwarranted as persistent drug pressure can lead to theevolution of resistance at little or no fitness cost to para-sites (99). Fortunately, evolutionary theory can informboth vaccine design and intervention strategies, forinstance by providing predictive power to a given vaccina-tion approach, and help weigh trade-offs between mitiga-tion of pathology and elimination of parasite transmission(84,100,101).

Ecology

Finally, there is the further challenge of leaving the labora-tory for the real world. Vaccines are required to protect asufficient number of individuals whose genetic susceptibil-ity to infection varies widely (102,103). In addition to vari-ation in host immune genotype, protective immunity canvary as a result of its prior interactions with other patho-gens as well as nutritional status, age and gender (104).Such sources of variation are known to affect hostimmune responses and susceptibility to infection (105,106)and thus may degrade the efficacy of antifilarial vaccinesbelow their performance in the laboratory. This requiresgreater investment in human and nonlaboratory-based ani-mal immunological studies and particularly longitudinalinvestigations at the community level, as well as acquisi-

tion of knowledge of the genetic make-up of parasite pop-ulations in relation to parasite survival and transmission.

CONCLUSION

Despite these obstacles, there is a room for optimism. Theexistence of natural immunity in people gives hope that vac-cines can be developed as does the success of vaccines inanimal models. Knowledge of antifilarial immunity hasmade enormous advances in recent years, and developmentof the rodent filarial nematode L. sigmodontis has allowedthe full power of mouse genetics and immunology to beapplied to antifilarial vaccine research. At the same time,the introduction of high-throughput technologies enablesexamination of the entire molecular repertoires of both par-asite and hosts. Combined with the application of systemanalyses, these data are being used to identify the pathwaysthat induce and regulate protective immunity. Furthermore,this combination also identifies traits that can lead topathology, and the evolutionary and ecological forces driv-ing potential vaccine failures. Our best hopes may lie withdesigning vaccines that do not exist naturally, i.e. geneticallymodified antigens, and adjuvants that decouple immunopa-thology from protective immune responses.

ACKNOWLEDGEMENTS

The authors work on filariasis has been or is supported byEU contracts FP5 VARBO ICT4-1999-10002, FP6SCOOTT INCO-CT 2006-03232 and FP7 E PIAF 242131,the Welcome Trust salary for S.A.B, and the MRC UK(G0600818 to J.E.A.)

REFERENCES

1 Ottesen EA, Hooper PJ, Bradley M &Biswas G. The global programme to elimi-nate lymphatic filariasis: health impactafter 8 years. PLoS Negl Trop Dis 2008; 2:e317.

2 Cupp EW, Sauerbrey M & Richards F. Elim-ination of human onchocerciasis: history ofprogress and current feasibility using iver-mectin (Mectizan((R))) monotherapy. ActaTrop 2011; 120(Suppl 1): S100–S108.

3 S�k�t�li A, Adeoye G, Eyamba A, et al. Theachievements and challenges of the AfricanProgramme for Onchocerciasis Control(APOC). Ann Trop Med Parasitol 2002; 96:15–28.

4 Taylor MJ, Hoerauf A & Bockarie M. Lym-phatic filariasis and onchocerciasis. Lancet2010; 376: 1175–1185.

5 Le GL, Marechal P, Petit G, Taylor DW,Hoffmann W & Bain O. Early reduction ofthe challenge recovery rate following immu-nization with irradiated infective larvae in a

filaria mouse system. Trop Med Int Health1997; 2: 1170–1174.

6 Babayan SA, Attout T, Harris A, et al. Vac-cination against filarial nematodes with irra-diated larvae provides long-term protectionagainst the third larval stage but not againstsubsequent life cycle stages. Int J Parasitol2006; 36: 903–914.

7 Anand SB, Kodumudi KN, Reddy MV &Kaliraj P. A combination of two Brugia ma-layi filarial vaccine candidate antigens(BmALT-2 and BmVAH) enhances immuneresponses and protection in jirds. J Helmin-thol 2011; 85: 442–452.

8 Bockarie MJ & Deb RM. Elimination oflymphatic filariasis: do we have the drugs tocomplete the job? Curr Opin Infect Dis 2010;23: 617–620.

9 Hoerauf A, Pfarr K, Mand S, Debrah AY &Specht S. Filariasis in Africa – treatmentchallenges and prospects. Clin MicrobiolInfect 2011; 17: 977–985.

10 Zoure HG, Wanji S, Noma M, et al. Thegeographic distribution of Loa loa in Africa:Results of large-scale implementation of therapid assessment procedure for loiasis(RAPLOA). PLoS Negl Trop Dis 2011; 5:e1210.

11 Boussinesq M, Gardon J, Gardon-Wendel N& Chippaux JP. Clinical picture, epidemiol-ogy and outcome of Loa-associated seriousadverse events related to mass ivermectintreatment of onchocerciasis in Cameroon.Filaria J 2003; 2(Suppl 1): S4.

12 Ardelli BF & Prichard RK. Reduced geneticvariation of an Onchocerca volvulus ABCtransporter gene following treatment withivermectin. Trans R Soc Trop Med Hyg2007; 101: 1223–1232.

13 Osei-Atweneboana MY, Awadzi K, AttahSK, Boakye DA, Gyapong JO & PrichardRK. Phenotypic evidence of emerging iver-mectin resistance in Onchocerca volvulus.PLoS Negl Trop Dis 2011; 5: e998.

S. A. Babayan et al. Parasite Immunology

250 � 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253

Page 9: Future prospects and challenges of vaccines against filariasis

14 Langworthy NG, Renz A, Mackenstedt U,et al. Macrofilaricidal activity of tetracyclineagainst the filarial nematode Onchocercaochengi: elimination of Wolbachia precedesworm death and suggests a dependent rela-tionship. Proc Biol Sci 2000; 267: 1063–1069.

15 Ferri E, Bain O, Barbuto M, et al. Newinsights into the evolution of Wolbachiainfections in filarial nematodes inferred froma large range of screened species. PLoS ONE2011; 6: e20843.

16 McGarry HF, Pfarr K, Egerton G, et al.Evidence against Wolbachia symbiosis in Loaloa. Filaria J 2003; 2: 9.

17 Hoerauf A, Specht S, Buttner M, et al. Wol-bachia endobacteria depletion by doxycyclineas antifilarial therapy has macrofilaricidalactivity in onchocerciasis: a randomized pla-cebo-controlled study. Med Microbiol Immu-nol 2008; 197: 295–311.

18 Turner JD, Tendongfor N, Esum M, et al.Macrofilaricidal activity after doxycyclineonly treatment of Onchocerca volvulus in anarea of Loa loa co-endemicity: a randomizedcontrolled trial. PLoS Negl Trop Dis 2010; 4:e660.

19 Bockarie MJ, Taylor MJ & Gyapong JO.Current practices in the management of lym-phatic filariasis. Expert Rev Anti Infect Ther2009; 7: 595–605.

20 Gyapong JO, Kumaraswami V, Biswas G &Ottesen EA. Treatment strategies underpin-ning the global programme to eliminate lym-phatic filariasis. Expert Opin Pharmacother2005; 6: 179–200.

21 WHO. Working to Overcome the GlobalImpact of Neglected Tropical Diseases: FirstWHO Report on Neglected Tropical Dis-eases 2010. Geneva: World Health Organiza-tion, 2011:150.

22 Supali T, Djuardi Y, Pfarr KM, et al. Doxy-cycline treatment of Brugia malayi-infectedpersons reduces microfilaremia and adversereactions after diethylcarbamazine andalbendazole treatment. Clin Infect Dis 2008;46: 1385–1393.

23 Debrah AY, Mand S, Specht S, et al. Doxy-cycline reduces plasma VEGF-C ⁄ sVEGFR-3and improves pathology in lymphatic filaria-sis. PLoS Pathog 2006; 2: e92.

24 Mand S, Pfarr K, Sahoo PK, et al. Macrofil-aricidal activity and amelioration of lympha-tic pathology in bancroftian filariasis after3 weeks of doxycycline followed by single-dose diethylcarbamazine. Am J Trop MedHyg 2009; 81: 702–711.

25 Dadzie Y, Neira M & Hopkins D. Finalreport of the Conference on the eradicabilityof Onchocerciasis. Filaria J 2003; 2: 2.

26 Diawara L, Traore MO, Badji A, et al. Fea-sibility of onchocerciasis elimination withivermectin treatment in endemic foci inAfrica: first evidence from studies in Maliand Senegal. PLoS Negl Trop Dis 2009; 3:e497.

27 Brattig NW. Pathogenesis and host responsesin human onchocerciasis: impact of Oncho-cerca filariae and Wolbachia endobacteria.Microbes Infect 2004; 6: 113–128.

28 Pfarr KM, Debrah AY, Specht S & HoeraufA. Filariasis and lymphoedema. ParasiteImmunol 2009; 31: 664–672.

29 Maizels RM & Yazdanbakhsh M. Immuneregulation by helminth parasites: cellular andmolecular mechanisms. Nat Rev Immunol2003; 3: 733–744.

30 Adjobimey T & Hoerauf A. Induction ofimmunoglobulin G4 in human filariasis: anindicator of immunoregulation. Ann TropMed Parasitol 2010; 104: 455–464.

31 Hoerauf A & Brattig N. Resistance and sus-ceptibility in human onchocerciasis ? beyondTh1 vs Th2. Trends Parasitol 2002; 18: 25–31.

32 Turaga PS, Tierney TJ, Bennett KE, et al.Immunity to onchocerciasis: cells from puta-tively immune individuals produce enhancedlevels of interleukin-5, gamma interferon,and granulocyte-macrophage colony-stimu-lating factor in response to Onchocerca volvu-lus larval and male worm antigens. InfectImmun 2000; 68: 1905–1911.

33 Doetze A. Production of both IFN-gammaand IL-5 by Onchocerca volvulus S1 antigen-specific CD4+ T cells from putatively immuneindividuals. Int Immunol 1997; 9: 721–729.

34 Elson LH, Calvopina HM, Paredes YW,et al. Immunity to onchocerciasis: putativeimmune persons produce a Thl-like responseto Onchocerca volvulus. J Infect Dis 1995;171: 652–658.

35 Soboslay PT, Geiger SM, Weiss N, et al. Thediverse expression of immunity in humans atdistinct states of Onchocerca volvulus infec-tion. Immunology 1997; 90: 592–599.

36 Nielsen NO, Bloch P & Simonsen PE. Lym-phatic filariasis-specific immune responses inrelation to lymphoedema grade and infectionstatus. II. Humoral responses. Trans R SocTrop Med Hyg 2002; 96: 453–458.

37 Nielsen NO, Bloch P & Simonsen PE. Lym-phatic filariasis-specific immune responses inrelation to lymphoedema grade and infectionstatus. I. Cellular responses. Trans R SocTrop Med Hyg 2002; 96: 446–452.

38 Trees AJ, Wahl G, Kl�ger S & Renz A. Age-related differences in parasitosis may indicateacquired immunity against microfilariae incattle naturally infected with Onchocerca och-engi. Parasitology 1992; 104: 247.

39 Wahl G, Achu-Kwi MD, Mbah D, Dawa O& Renz A. Bovine onchocercosis in northCameroon. Vet Parasitol 1994; 52: 297–311.

40 Xie H, Bain O & Williams SA. Molecularphylogenetic studies on filarial parasitesbased on 5S ribosomal spacer sequences.Parasite 1994; 1: 141–151.

41 Wahl G, Enyong P, Ngosso A, et al. Oncho-cerca ochengi: epidemiological evidence ofcross-protection against Onchocerca volvulusin man. Parasitology 1998; 116: 349–362.

42 Seidenfaden R, Fischer A, Bonow I, EkaleD, Tanya V & Renz A. Combined benefitsof annual mass treatment with ivermectinand cattle zooprophylaxis on the severity ofhuman onchocerciasis in northern Camer-oon. Trop Med Int Health 2001; 6: 715–725.

43 Trees AJ. The geographic distribution of Loaloa in Africa: results of large-scale implemen-

tation of the rapid assessment procedure forloiasis (raploa). Parasitology Today 1992; 8:337–339.

44 Achukwi MD, Harnett W, Enyong P & RenzA. Successful vaccination against Onchocercaochengi infestation in cattle using live Oncho-cerca volvulus infective larvae. Parasite Immu-nol 2007; 29: 113–116.

45 Tchakoute VL, Graham SP, Jensen SA, et al.In a bovine model of onchocerciasis, protec-tive immunity exists naturally, is absent indrug-cured hosts, and is induced by vaccina-tion. Proc Natl Acad Sci USA 2006; 103:5971–5976.

46 Graham SP, Trees AJ, Collins RA, et al.Down-regulated lymphoproliferation coin-cides with parasite maturation and with thecollapse of both gamma interferon and inter-leukin-4 responses in a bovine model ofonchocerciasis. Infect Immun 2001; 69: 4313–4319.

47 Nfon CK, Makepeace BL, Njongmeta LM,Tanya VN, Bain O & Trees AJ. Eosinophilscontribute to killing of adult Onchocerca och-engi within onchocercomata following elimi-nation of Wolbachia. Microbes Infect 2006; 8:2698–2705.

48 Jarrett WF, Jennings FW, McIntyre WI,Mulligan W & Urquhart GM. Irradiatedhelminth larvae in vaccination. Proc R SocMed 1958; 51: 743–744.

49 Bickle QD. Radiation-attenuated schistosomevaccination – a brief historical perspective.Parasitology 2009; 136: 1621–1632.

50 Schrempf-Eppstein B, Kern A, Textor G &Lucius R. Acanthocheilonema viteae: vaccina-tion with irradiated L3 induces resistance inthree species of rodents (Meriones unguicula-tus, Mastomys coucha, Mesocricetus auratus).Trop Med Int Health 1997; 2: 104–110.

51 Lange AM, Yutanawiboonchai W, Lok JB,Trpis M & Abraham D. Induction of protec-tive immunity against larval Onchocerca vol-vulus in a mouse model. Am J Trop MedHyg 1993; 49: 783–788.

52 Osborne J. The L3 of Brugia induces a Th2-polarized response following activation of anIL-4-producing CD4-CD8- alphabeta T cellpopulation. Int Immunol 1998; 10: 1583–1590.

53 Carlow CK & Bianco AE. Transfer of immu-nity to the microfilariae of Onchocerca lienal-is in mice. Trop Med Parasitol 1987; 38: 283–286.

54 Abraham D, Leon O, Schnyder-Candrian S,et al. Immunoglobulin E and eosinophil-dependent protective immunity to larvalOnchocerca volvulus in mice immunized withirradiated larvae. Infect Immun 2004; 72:810–817.

55 Hartmann S, Sollwedel A, Hoffmann A,Sonnenburg B & Lucius R. Characterizationof IgE responses in a rodent model of filaria-sis and the allergenic potential of filarialantigens using an in vitro assay. ParasiteImmunol 2003; 25: 9–16.

56 Baldwin CI, de Medeiros F & Denham DA.IgE responses in cats infected with Brugia pah-angi. Parasite Immunol 1993; 15: 291–296.

Volume 34, Number 5, May 2012 Vaccination against filariasis

� 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253 251

Page 10: Future prospects and challenges of vaccines against filariasis

57 Petit G, Diagne M, Marechal P, Owen D,Taylor D & Bain O. Maturation of the filariaLitomosoides sigmodontis in BALB ⁄ c mice;comparative susceptibility of nine otherinbred strains. Ann Parasitol Hum Comp1992; 67: 144–150.

58 Graham AL, Taylor MD, Le Goff L, LambTJ, Magennis M & Allen JE. Quantitativeappraisal of murine filariasis confirms hoststrain differences but reveals that BALB ⁄ cfemales are more susceptible than males toLitomosoides sigmodontis. Microbes Infect2005; 7: 612–618.

59 Le Goff L, Lamb TJ, Graham AL, HarcusY & Allen JE. IL-4 is required to preventfilarial nematode development in resistantbut not susceptible strains of mice. Int JParasitol 2002; 32: 1277–1284.

60 Al-Qaoud K. The Xid defect imparts suscep-tibility to experimental murine filariosis –association with a lack of antibody andIL-10 production by B cells in response tophosphorylcholine. Int Immunol 1998; 10:17–25.

61 Volkmann L, Saeftel M, Bain O, Fischer K,Fleischer B & Hoerauf A. Interleukin-4 isessential for the control of microfilariae inmurine infection with the filaria Litomoso-ides sigmodontis. Infect Immun 2001; 69:2950–2956.

62 Lawrence RA. Immunity to filarial nema-todes. Vet Parasitol 2001; 100: 33–44.

63 Hoffmann WH, Pfaff AW, Schulz-Key H &Soboslav PT. Determinants for resistanceand susceptibility to microfilaraemia inLitomosoides sigmodontis filariasis. Parasitol-ogy 2001; 122: 641–649.

64 Allen JE & Maizels RM. Diversity and dia-logue in immunity to helminths. Nat RevImmunol 2011; 11: 375–388.

65 Taylor MD, van der Werf N, Harris A, et al.Early recruitment of natural CD4+ Foxp3+Treg cells by infective larvae determines theoutcome of filarial infection. Eur J Immunol2009; 39: 192–206.

66 Taylor MD, LeGoff L, Harris A, Malone E,Allen JE & Maizels RM. Removal ofregulatory T cell activity reverses hypore-sponsiveness and leads to filarial parasiteclearance in vivo. J Immunol 2005; 174:4924–4933.

67 Le Goff L, Martin C, Oswald IP, et al. Para-sitology and immunology of mice vaccinatedwith irradiated Litomosoides sigmodontis lar-vae. Parasitology 2000; 120: 271–280.

68 Wagner U, Hirzmann J, Hintz M, et al.Characterization of the DMAE-modifiedjuvenile excretory-secretory protein Juv-p120of Litomosoides sigmodontis. Mol BiochemParasitol 2011; 176: 80–89.

69 Martin C, Saeftel M, Vuong PN, et al. B-celldeficiency suppresses vaccine-induced protec-tion against murine filariasis but does notincrease the recovery rate for primary infec-tion. Infect Immun 2001; 69: 7067–7073.

70 Martin C, Al-Qaoud KM, Ungeheuer MN,et al. IL-5 is essential for vaccine-inducedprotection and for resolution of primary

infection in murine filariasis. Med MicrobiolImmunol 2000; 189: 67–74.

71 Martin C, Le Goff L, Ungeheuer MN,Vuong PN & Bain O. Drastic reduction of afilarial infection in eosinophilic interleukin-5transgenic mice. Infect Immun 2000; 68:3651–3656.

72 Bleiss W, Oberl�nder U, Hartmann S, et al.Onchocerca ochengi: Mimic, model or modu-lator of O. volvulus? J Parasitol 2002; 88:264–270.

73 Pfaff AW, Schulz-Key H, Soboslay PT, Gei-ger SM & Hoffmann WH. The role of nitricoxide in the innate resistance to microfilariaeof Litomosoides sigmodontis in mice. ParasiteImmunol 2000; 22: 397–405.

74 Gray CA & Lawrence RA. Interferon-gamma and nitric oxide production are notrequired for the immune-mediated clearanceof Brugia malayi microfilariae in mice. Para-site Immunol 2002; 24: 329–336.

75 Volkmann L, Bain O, Saeftel M, et al. Mur-ine filariasis: interleukin 4 and interleukin 5lead to containment of different worm devel-opmental stages. Med Microbiol Immunol2003; 192: 23–31.

76 Specht S, Volkmann L, Wynn T & HoeraufA. Interleukin-10 (IL-10) counterregulatesIL-4-dependent effector mechanisms in mur-ine filariasis. Infect Immun 2004; 72: 6287–6293.

77 Devaney E, Gillan V, Wheatley I, Jenson J,O’Connor R & Balmer P. Interleukin-4 influ-ences the production of microfilariae in amouse model of Brugia infection. ParasiteImmunol 2002; 24: 29–37.

78 Gray CA & Lawrence RA. A role for anti-body and Fc receptor in the clearance ofBrugia malayi microfilariae. Eur J Immunol2002; 32: 1114–1120.

79 Kalyanasundaram R & Balumuri P. Multiva-lent vaccine formulation with BmVAL-1 andBmALT-2 confer significant protectionagainst challenge infections with Brugiamalayi in mice and jirds. Res Rep Trop Med2011; 2011: 45–56.

80 Makepeace BL, Jensen SA, Laney SJ, et al.Immunisation with a multivalent, subunitvaccine reduces patent infection in a naturalbovine model of onchocerciasis duringintense field exposure. PLoS Negl Trop Dis2009; 3: e544.

81 Babayan SA, Read AF, Lawrence RA,Bain O & Allen JE. Filarial parasitesdevelop faster and reproduce earlier inresponse to host immune effectors thatdetermine filarial life expectancy. PLoSBiol 2010; 8: e1000525.

82 Dittrich AM, Erbacher A, Specht S, et al.Helminth infection with Litomosoidessigmodontis induces regulatory T cells andinhibits allergic sensitization, airway inflam-mation, and hyperreactivity in a murineasthma model. J Immunol 2008; 180: 1792–1799.

83 Hoerauf A, Satoguina J, Saeftel M & SpechtS. Immunomodulation by filarial nematodes.Parasite Immunol 2005; 27: 417–429.

84 Schneider DS & Ayres JS. Two ways to sur-vive infection: what resistance and tolerancecan teach us about treating infectious dis-eases. Nat Rev Immunol 2008; 8: 889–895.

85 Bethony JM, Simon G, Diemert DJ, et al.Randomized, placebo-controlled, double-blind trial of the Na-ASP-2 hookworm vac-cine in unexposed adults. Vaccine 2008; 26:2408–2417.

86 Hotez PJ, Bethony JM, Diemert DJ, PearsonM & Loukas A. Developing vaccines tocombat hookworm infection and intestinalschistosomiasis. Nat Rev Microbiol 2010; 8:814–826.

87 Paul WE & Zhu J. How are T(H)2-typeimmune responses initiated and amplified?Nat Rev Immunol 2010; 10: 225–235.

88 Phythian-Adams AT, Cook PC, Lundie RJ,et al. CD11c depletion severely disrupts Th2induction and development in vivo. J ExpMed 2010; 207: 2089–2096.

89 MacDonald AS & Maizels RM. Alarmingdendritic cells for Th2 induction. J Exp Med2008; 205: 13–17.

90 Pulendran B & Ahmed R. Immunologicalmechanisms of vaccination. Nat Immunol2011; 12: 509–517.

91 Schierack P, Lucius R, Sonnenburg B, Schil-ling K & Hartmann S. Parasite-specificimmunomodulatory functions of filarial cyst-atin. Infect Immun 2003; 71: 2422–2429.

92 Manoury B, Gregory WF, Maizels RM &Watts C. Bm-CPI-2, a cystatin homologsecreted by the filarial parasite Brugiamalayi, inhibits class II MHC-restricted anti-gen processing. Curr Biol 2001; 11: 447–451.

93 Nchinda G, Kuroiwa J, Oks M, et al. Theefficacy of DNA vaccination is enhanced inmice by targeting the encoded protein todendritic cells. J Clin Invest 2008; 118: 1427–1436.

94 Oberg AL, Kennedy RB, Li P, OvsyannikovaIG & Poland GA. Systems biologyapproaches to new vaccine development.Curr Opin Immunol 2011; 23: 436–443.

95 Friedel CC & Haas J. Virus-host interacto-mes and global models of virus-infected cells.Trends Microbiol 2011; 19: 501–508.

96 Rodland KD, Adkins JN, Ansong C, et al.Use of high-throughput mass spectrometryto elucidate host-pathogen interactions inSalmonella. Future Microbiol 2008; 3: 625–634.

97 McDermott JE, Yoon H, Nakayasu ES,et al. Technologies and approaches to eluci-date and model the virulence program of sal-monella. Front Microbiol 2011; 2: 121.

98 Gandon S, Jansen VA & van Baalen M.Host life history and the evolution of para-site virulence. Evolution 2001; 55: 1056–1062.

99 Gagneux S, Long CD, Small PM, Van T,Schoolnik GK & Bohannan BJ. The compet-itive cost of antibiotic resistance in Mycobac-terium tuberculosis. Science 2006; 312: 1944–1946.

100 Day T, Galvani A, Struchiner C & GumelA. The evolutionary consequences of vacci-nation. Vaccine 2008; 26: C1–C3.

S. A. Babayan et al. Parasite Immunology

252 � 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253

Page 11: Future prospects and challenges of vaccines against filariasis

101 Lynch PA, Grimm U & Read AF. How willpublic and animal health interventions drivelife-history evolution in parasitic nematodes?Parasitology 2008; 135: 1599–1611.

102 Lazarus R, Vercelli D, Palmer LJ, et al. Sin-gle nucleotide polymorphisms in innateimmunity genes: abundant variation andpotential role in complex human disease.Immunol Rev 2002; 190: 9–25.

103 Carvalho A, Cunha C, Pasqualotto AC, Pit-zurra L, Denning DW & Romani L. Geneticvariability of innate immunity impactshuman susceptibility to fungal diseases. IntJ Infect Dis 2010; 14: e460–e468.

104 Pedersen AB & Babayan SA. Wild immunol-ogy. Mol Ecol 2011; 20: 872–880.

105 Babu S, Bhat SQ, Kumar NP, et al. Attenua-tion of toll-like receptor expression and func-

tion in latent tuberculosis by coexistentfilarial infection with restoration followingantifilarial chemotherapy. PLoS Negl TropDis 2009; 3: e489.

106 Nookala S, Srinivasan S, Kaliraj P, Naraya-nan RB & Nutman TB. Impairment of teta-nus-specific cellular and humoral responsesfollowing tetanus vaccination in human lym-phatic filariasis. Infect Immun 2004; 72:2598–2604.

107 Thirugnanam S, Pandiaraja P, RamaswamyK, et al. Brugia malayi: comparison of pro-tective immune responses induced by Bm-alt-2 DNA, recombinant Bm-ALT-2 protein andprime-boost vaccine regimens in a jirdmodel. Exp Parasitol 2007; 116: 483–491.

108 Anand SB, Murugan V, Prabhu PR,Anandharaman V, Reddy MV & Kaliraj P.

Comparison of immunogenicity, protectiveefficacy of single and cocktail DNA vac-cine of Brugia malayi abundant larvaltranscript (ALT-2) and thioredoxin peroxi-dase (TPX) in mice. Acta Trop 2008; 107:106–112.

109 Rathaur S, Yadav M, Gupta S, Anandhar-aman V & Reddy MV. Filarial glutathione-S-transferase: a potential vaccine candidateagainst lymphatic filariasis. Vaccine 2008; 26:4094–4100.

110 McCarthy JS, Wieseman M, Tropea J, et al.Onchocerca volvulus glycolytic enzyme fruc-tose-1,6-bisphosphate aldolase as a target fora protective immune response in humans.Infect Immun 2002; 70: 851–858.

Volume 34, Number 5, May 2012 Vaccination against filariasis

� 2011 Blackwell Publishing Ltd, Parasite Immunology, 34, 243–253 253