Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

30
CHAPTER  10 Control of Important Helminthic Infections: Vaccine Development as Part of the Solution Robert Bergquist* and  Sara Lustigman Contents  10. 1. Introduction  298 10.2. Type of Vaccines  300 10. 3. Vaccine Design  301 10.3.1. Correl ate studies  302 10.3.2. Adjuvants  303 10.4. Progr ess in Vaccine Devel opment  304 10.4.1. Cestode infections  305 10.4.2. Nematodes  307 10.4.3. Tremat odes  311 10.5. Indus trial Vaccine Production  314 10.6. Concluding Remarks  317 References 319 Abstract  Among the tools available for the contr ol of helminth infecti ons, chemotherapy has come to totally dominate the field. In the veter inary field, development of drug resistance has appeared but this is not (yet) a problem in the control of human di seases. Al though there is no vaccine commerci ally avail able for any human parasitic infection yet, recent progress in vaccine develop- ment is making this a future possibility for several diseases. The goal of chemotherapy is to alleviate infection and morbidity in the definitive host, or reduce transmission, while the effect of available  Advances in Parasitology, Volume 73  # 2010 Elsevier Ltd. ISSN 0065-308X, DOI: 10.1016/S0065-308X(10)73010-4 All rights reserved. *  Ingerod, Brastad, Sweden { Laboratory of Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, USA 297

Transcript of Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

Page 1: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 1/30

Page 2: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 2/30

vaccine candidates would mainly be to influence transmission

through targeting the intermediate or reservoir host, when the

infection is zoonotic. Apart from this general scheme, there are

also vaccine candidates targeting the parasites in the definitive

host, in particular the early developmental stages, which shouldreduce the risk of drug failure. Since the biological targets in most

cases are different, vaccination would be synergistic with drug

therapy. This review covers diseases caused by helminthes in

both humans and animals and includes examples of diseases caused

by cestodes, nematodes and trematodes. The focus is on infections

for which vaccine development has been undertaken for a long

time, resulting in products that could realistically become

integrated into control strategies in the near future.

10.1. INTRODUCTION

Initially, based on the early success of anti-viral and anti-bacterial vac-cines, it was believed that development of vaccines against parasiteswould be likewise. The first vaccine candidates of this kind consisted of attenuated or killed whole organisms, but the results were not entirely

successful and work therefore turned towards native parasite antigensand recombinant subunit vaccines. At this point, it was already clear thatthe way forward would be far from simple. Even after the first stumbling block, large-scale antigen production, had been removed by the advent of recombinant DNA technology in the 1980s, work on anti-parasite vac-cines did not immediately take off. The situation has improved since then but chemotherapy still completely dominates the control measures forparasitic diseases of humans and animals alike. However, long-term drugtreatment represents a continuous expense and drug resistance is a per-manent threat; in the veterinary field often a reality. The goal of vaccinedevelopment in this area is not to produce a vaccine capable of inducingsterilizing immunity but to create an adjunct to chemotherapy that wouldreduce the likelihood of vaccinated individuals developing severe infec-tions and thus reduce the burden of disease throughout the world. Anintegrated approach, that is the follow-up of initial drug treatment withvaccination to achieve long-term protection (Bergquist et al., 2008), hasmuch to offer but a repository of specific vaccines useful against thevariety of infectious agents that make up the neglected tropical diseases(NTD) must first be established.

Commercially provided antiparasitic drugs with broad-spectrumaction have successfully been used to control parasitic diseases in live-stock and other domestic animals. However, frequent emergence of drugresistance in the target parasites has become a challenge. In addition,

298   Robert Bergquist and Sara Lustigman

Page 3: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 3/30

issues regarding drug residues in the environment and food chain havecome to the forefront, boosting interest in alternative control methods andrenewed the appeal of vaccines.

Approaches based on molecular biology technology have resulted inthe elucidation of entire parasite genomes, as well as the identification of individual genes (Abubucker et al., 2008; Berriman et al., 2009; Brindleyet al., 2009; Ghedin et al., 2007; Schistosoma japonicum Genome Sequencingand Functional Analysis Consortium et al., 2009). Without doubt, furtherunderstanding of the role of gene products in parasite biology will lead tothe identification of novel parasite vaccine target antigens. However, forthis aim to be fully realized, strong investment in basic research on thecomplex interplay between parasite and host is necessary. Despite long-

term work on vaccine development, notably in the fields of hookworminfection, leishmaniasis, malaria, onchocerciasis and schistosomiasis, wehave yet to see an effective vaccine being implemented against a humanparasitic disease. On the other hand, for some of these infections, experi-ments using animal models have shown strong promise and there has been clear progress on vaccines against veterinary helminthic infections(Rickard et al., 1995). This may reflect fewer problems to be solved at the biological level with regard to animal vaccine development, but the moreprobable reasons are the much stricter regulatory demands governing

products for human use. The marketability of veterinary products isanother factor, as people who require parasite vaccines are generally notin a position to pay for them.

With regard to veterinary vaccines, the immunological control of Fasciolain sheep and cattle is within reach (Tendler and Simpson, 2008), and highlyeffective recombinant vaccines have already been developed for the preven-tion of   Taenia ovis   in sheep,   T. saginata   in cattle,   T. solium   in pigs andEchinococcus granulosus in livestock (Lightowlers, 2006a,b). While vaccinesagainst   T. ovis   and   T. saginata   are purely intended for assistance forfarming, vaccines against T. solium and E. granulosus vould primarily be beneficial to humans. It should, in this connection, be mentioned that trans-mission-blocking   Schistosoma japonicum   vaccines, which also belong tothis category, are currently in field trials (Da’dara et al., 2008; McManuset al., 2009).

Parasites survive in the host by avoiding or confusing immuneresponses against them, for example through stimulation of factorsdown-regulating the cellular response or through non-specific activationof B-cells. Parasite antigens are complex and difficult to characterise andthe host commonly responds with a range of various defence mechan-isms. Western blotting (Burnette, 1981) is the traditional approach toidentifying potential antigens for vaccine development but few of theantibodies identified by this technique are protective, as they are mostoften raised against non-related intracellular proteins which are released

Vaccine Development and Control of Helminthic Infections   299

Page 4: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 4/30

when the parasite disintegrates. Protein polymorphism representsanother problem (Rosenzvit et al., 2006) that could make a the vaccineineffective against serotypes not included in the original design. Finally,

laboratory strains of parasites may become atypical, lose their naturalrange of polymorphism or even develop new polymorphisms notseen in nature by being maintained through long-term laboratorypropagation.

10.2. TYPE OF VACCINES

Vaccine production involves a spectrum of designs that can be based onanything between attenuated organisms and complex compounds pro-duced by molecular biology. Although the simplest approaches do notgenerally result in useful products, it does not follow that the mostcomplicated approaches are necessarily superior. To facilitate orientation,a brief guide to the different vaccine types is presented:

1.  Vaccines consisting of whole parasites killed in various ways (heating,irradiation, etc.) that conserve their structures.

2.  Attenuated organisms—live parasites treated so they cannot persist or

cause injury when injected.3.   Recombinant vaccines—well-characterised gene products (antigens)

that can be produced in large quantities, consistently and cheaply.4.   Peptide vaccines—short synthetic protein sequences including the

desired immuno-dominant epitopes.5.   Recombinant-vector vaccines—attenuated virus or bacteria whose

genome has been supplied with extraneous DNA encoding parasiteantigenic determinants to be replicated and expressed by the vector.

6.  DNA vaccines—plasmids containing the immune-dominant sequence

(s) capable of producing the desired parasite antigen intracellularly(in host cells) in an approach similar to that of recombinant-vectorvaccines.

Synthetic peptide vaccines are excellent from the point of view thatthey do not carry any danger of contamination with other proteins.In addition, epitopes from different proteins can be incorporated into asingle construct. However, peptides are not immunogenic and are there-fore utterly dependent on adjuvants. Although in theory this very fact

permits the option to select predominantly cell-mediated immunity orantibody production (Al-Sherbiny et al., 2003), we do not yet have accessto the adjuvants needed to achieve this modulation precisely. However,once the field has advanced further and products have been cleared for

300   Robert Bergquist and Sara Lustigman

Page 5: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 5/30

Page 6: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 6/30

assistance in antigen discovery comes from the possibility of probingpreparations by Western blotting (Burnette, 1981) against protective serato identify the most antigenic components. The proteins in question can

 be chromatographically analysed and the preferred band(s) identified,extracted and subjected to liquid chromatography followed by massspectrometry for identification. The subsets of proteins, selected on the basis of their involvement in the host–parasite relationship and parasite biology, can then be produced in large amounts by means of recombinantDNA techniques for the routinely required efficacy-testing in animalmodels.

10.3.1. Correlate studiesOnce a panel of protective antigens has been established, the next step isto find out how animals and humans in endemic areas react immunologi-cally to natural infection. The investigation of individual immuneresponses to specific antigens, focusing interest on the underlyingmechanisms in resistance at the molecular level, is one of the keyapproaches to vaccine design. Well-researched antigen panels have beenset up for various parasites with the aim of identifying antibody andcytokine correlates of apparent resistance and apparent susceptibility.The approach was pioneered for S. mansoni antigens (Acosta et al., 2002;Al-Sherbiny et al., 2003; Ribeiro de Jesus et al., 2000). In a comprehensivestudy of this subject, carried out by the Egyptian Reference DiagnosticCenter (ERDC) in Cairo, immune responses against a panel of 10 priorityS. mansoni   vaccine candidate molecules were determined in cohorts of humans living in areas where they were exposed to infection daily, andthe results compared with the corresponding data emanating from para-sitological diagnosis (Al-Sherbiny et al., 2003). Responses significantlycorrelating either with resistance or with susceptibility to re-infectionwere demonstrated and it was also possible to group the protectiveresponses to some of the antigens as belonging either to the Th1 or tothe Th2 sphere. This type of study is instructive not only for identifyingthe few antigens that directly and exclusively correlate with resistance, but also by indicating which of them can be modulated to produce thesought-after immune response. In a similar approach from the field of filariasis, Vedi et al. (2008) have identified a 2.0 kb cDNA clone coding forBrugia malayi  heavy chain myosin which exhibited strong immunoreac-tivity with bancroftian sera from endemic putatively immune humansubjects. Similarly, sera from individuals immune against   Onchocercavolvolus  have been used successfully to clone vaccine candidates subse-quently shown to be protective against  O. volvulus   infective third-stagelarvae in a mouse model (Lustigman et al., 2003).

302   Robert Bergquist and Sara Lustigman

Page 7: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 7/30

Page 8: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 8/30

worse than those produced by potassium aluminium sulphate (alum), theonly widely used adjuvant in human vaccines today. However, sincealum probably acts more as a depot than anything else (Lindblad, 2004),

the need for specific adjuvants is growing. Already, an adjuvant fromGlaxoSmithKline (London, UK) based on MPL and Quil (ASO4) has beenlicensed for use in human vaccines in more than 100 countries(Pichichero, 2008).

10.4. PROGRESS IN VACCINE DEVELOPMENT

Overall, the development of vaccines against parasitic diseases in humans

has not been taken seriously by donor agencies. The main reason for thisis the easy access to cheap, effective drugs which, apart from malariachemotherapy, have not been prone to produce resistance in their targetparasites. In the veterinary field, there is a stronger need for vaccines asdrug resistance is commonplace. This is probably due to the particularlyhigh drug pressure realized when herds and flocks are exposed to regularrecurrent blanket treatments. Also humans are subjected to repeatedchemotherapy against various helminth infections but the risk for drugresistance is relatively low as the treatment cycles promoted by control

programmes rely on lower doses spaced by relatively long intermissions.Still, development of resistance against drugs used for control of humaninfections can definitely not be ruled out, but it may take longer than the20 years commonly mentioned by veterinarians. Notably, the emergenceof resistant strains of the  O. volvulus  parasite is suggested by reports of onchocerciasis patients failing to respond to ivermectin treatment(Churcher et al., 2009). In addition, an increased frequency of a resistantallele has been identified in Wuchereria bancrofti microfilariae in patients inareas subjected to mass drug administration (MDA) with benzimidazole(Schwab et al., 2005). Both drugs are used for MDA of onchocerciasisand/or lymphatic filariasis (LF).

A typical characteristic of parasitic diseases is that the life cycles of theinfectious agents require that their life forms oscillate between differenthosts which often involve vertebrates both as intermediate and as defini-tive host. Although the intermediate host can also be a gastropod or aninsect (as in schistosomiasis and malaria, respectively), vertebrate hostsother than humans can also play the role of disease reservoir. For exam-ple, in contrast to other schistosome species, S. japonicum infects a broadrange of animals which are sometimes more important for the transmis-sion of the disease than humans (Gray et al., 2007; Wang et al., 2005).Against this background, one might think that vaccination of key verte- brate species involved in the propagation of diseases eventually implicat-ing humans should be part of control activities, yet it is not. In the absence

304   Robert Bergquist and Sara Lustigman

Page 9: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 9/30

of any human vaccine against parasites, veterinary vaccines should be triedfor the control of human endemic diseases. It is a positive sign that this issubject to research (Da’dara et al., 2008; Gray et al., 2007; Guo et al., 2006;

Lightowlers, 2004). The current situation with respect to vaccine develop-ment is reviewed below with respect to major zoonotic helminthiases suchas cysticercosis, echinococcosis and schistosomiasis japonica as well as forhuman helminth hookworm infection, LF and onchocerciasis.

10.4.1. Cestode infections

T. solium (see life cycle in the back of volume 72), the cestode whose larvalencystation gives rise to neurocysticercosis in humans, belongs to a zoonotic

disease complex that is endemic in a large number of countries where pigsare kept under unhygienic conditions and without adequate sanitary dis-posal of human faeces. Since the pig is the obligate intermediate host forT. solium, and effective drugs are available, it should be possible to curb thetransmission of this major parasitic disease that frequently affects humanhealth and economy in the developing countries. Since a veterinary vaccinehas also been developed against this disease (Lightowlers, 2004), it is almostsurprising that neurocysticercosis has not already been eliminated. Theexplanation for this lack of progress is that the pig readily establishes new

tapeworm infections in humans, leading to renewed transmission after eachround of treatment. A detailed review of the disease and its present situa-tion is given by Willingham et al. (2010).

A well-designed vaccine strategy should have a good chance to preventinfection in the pig and thus interrupt the parasite life cycle. Indeed,vaccine research has been moving well ahead during the last decade asthe work to decipher this parasite genetically has provided novel antigens.For example, Almeida et al. (2009) generated more than 1500 high-qualityExpressed Sequence Tags (ESTs) from 20 cDNA  T. solium mini-libraries.Identification of protective antigens and their production by recombinantDNA technology has been researched by Lightowlers (2003 and 2004).Tenyears ago, Johnson et al. (1989) published the cloning of a recombinantT. ovis  antigen which stimulated high levels of protective immunity insheep. Relatively rapidly, this was followed by the development of effec-tive vaccines not only against T. ovis in sheep but also against T. saginata incattle,   T. solium   in pigs and   E. granulosus   in livestock, indicating thatreliable, high-level protection against a complex metazoan parasite can be achieved using defined recombinant antigens (Lightowlers, 2006a,b;Lightowlers et al., 2003).

Several approaches based on recombinant antigens have been madetowards development of vaccines against   T. solium. For example, twohighly immunogenic oncosphere antigens, TSOL18 and TSOL45, have been shown to induce near-complete protection against experimental

Vaccine Development and Control of Helminthic Infections   305

Page 10: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 10/30

challenge infection in four separate vaccine trials in pigs (Kyngdon et al.,2006; Lightowlers, 2004) and another oncospheral stage-specific 45Wprotein has shown similar results (Luo et al., 2009). In addition, a review

on vaccines against cysticercosis by Sciutto et al. (2008) highlights work onS3Pvac, the only synthetic peptide vaccine that has been tested andproved effective in the field against naturally acquired disease. Thus,from the technical point of view, we have access to vaccines with thecapability to break the parasite life cycle in the pig intermediate host.If correctly deployed and monitored, any of these vaccines would withoutdoubt rapidly lead to the eradication of human cysticercosis.

E. granulosus (for life cycle see back of volume 72 and McManus, 2010)is another cestode stimulating strong immune responses which open

possibilities for the development of vaccines directed against this infec-tion, both in the intermediate and in the definitive host. An effectivestrategy would be to stop the development of adult gravid tapeworms indogs with a vaccine, so preventing the oncosphere from producing hyda-tid cysts in animals, for example sheep. This is a straightforward strategywhich relies on the fact that echinococcosis induces cell-mediated cellularresponses as well as significant antibody production in their human andintermediate hosts. However, for the approach to be successful, a thor-ough understanding of the immune mechanisms involved is required.

The many recent reviews of immunity mechanisms at work in echino-coccosis are evidence of the large body of knowledge amassed so far, forexample Amri et al. (2009), Gottstein and Hemphill (2008), Lightowlers(2006a), Lightowlers and Heath (2004), Torgerson (2009) and Zhang et al.(2008). Clearly, a very high coverage of flocks would need to be achievedfor vaccination alone to be effective, but a combination of vaccination of the sheep and drug treatment of surrounding dogs could achieve a goodlevel of transmission control, even with a less-than-perfect vaccine aspointed out by Torgerson (2006).

EG95, the recombinant E. granulosus vaccine based on an oncosphereprotein containing a glycosylphosphatydilinositol (GPI) anchor and afibronectin domain, is strongly immunogenic and induces effective pro-tection against challenge infection (Gauci et al., 2005). However, its immu-nological coverage may not be as broad as desired, since the E. granulosusgenome also contains several other EG95-related genes which may affectthe efficacy of this vaccine (Chow et al., 2008). Further sequencing hasuncovered a large amount of variability in this organism (Haag et al.,2009) confirming this suspicion. Nevertheless, as the sheep-dog strain of the parasite is responsible for most cases of human disease, EG95 remainspromising.

Although it would be of huge benefit in reducing the effective periodrequired to stop transmission of  E. granulosus, no effective vaccine existsagainst canine echinococcosis. Apart from that, however, a long list of 

306   Robert Bergquist and Sara Lustigman

Page 11: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 11/30

deliverables needs to be consulted to efficiently approach this goal, forexample improved diagnostics, characterization of infection dynamicsand determination of the longevity of protection induced, to mention

 just a few. In addition, further down the road, mathematical models willneed to be developed to facilitate impact evaluation. Even incorporationof only a few of these measures should increase the efficiency of controland reduce the time required to achieve prevention of disease transmis-sion. Above all, the scarcity of vaccine candidates for immune protectionagainst adult tapeworms reflects the lack of immune correlates and theambiguity of natural immunity in dogs (Craig et al., 2007; Zhang andMcManus, 2008).

Before leaving the topic of vaccines against echinococcosis,  E. multi-

locularis (see life cycle on page tbc) should briefly be mentioned. UnlikeE. granulosus, this species produces multiple, small cysts that spreadthroughout the body in many mammals, including rodents and humans.Although this infection is mainly distributed in the northern hemisphereand is much less common than that caused by E. granulosus, it is referredto here since there are prospects that a vaccine can be developed against it.A tetraspanin candidate (E24) has been cloned from a full-length cDNAlibrary emanating from the   E. multilocularis   metacestode. Antibodiesagainst this antigen specifically recognize a 25 kDa cyst antigen from

the germinal layer of the  E. multilocularis   metacestode, highlighting itspotential both for diagnostics and vaccine development (Dang et al.,2009a,b)

10.4.2. Nematodes

The Phylum Nematoda (roundworms) includes tens of thousands of oftenvery diverse species, a large number of which are parasitic. Nematodescausing disease in humans include filarids, hookworms, pinworms andwhipworms, as well as individual species such as Ascaris lumbricoides andTrichinella spiralis. While drug treatment is an adequate approach for mostof these, it is realized that the long-term control of some of them, that ishuman hookworm infection (due to   Ancylostoma duodenale   or   Necatoramericanus), LF (due to   W. bancrofti   or   B. malayi,) and river blindness(caused by   O. volvulus) will not be possible with drugs alone. Whileregular, annual or semi-annual chemotherapy is an important part of any public health interventions, high rates of re-infection and the spectreof diminished efficacy of drugs used often and repeatedly, conspire tochip away at the sustainability of this approach. Indeed, macrofilarialdrug cure, such as treatment with the adulticide melarsomine, can evenreduce natural (or induced) protective immunity as shown by a longitu-dinal study in a bovine model (Tchakoute et al., 2006). However, currentlythe treatment is with ivermectin, which has potent efficacy against the

Vaccine Development and Control of Helminthic Infections   307

Page 12: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 12/30

microfilariae only. Duerr et al. (2008), on the other hand, have suggestedthat the resistance against filarial parasites includes a time-dependentcomponent caused by an early immune response with short-term mem-

ory. While vaccine studies are moving forward, clinical studies to investi-gate this conjecture are warranted.Hookworm (see life cycles on back of volume 72) is a leading cause of 

maternal and child morbidity in the developing countries of the tropics andsubtropics. Together, the two species of hookworms that infect humans,N. americanus and A. duodenale, infect more than 500 million people world-wide (Hotez and Kamath, 2009; Hotez et al., 2008). The former species iscommon in the Americas, sub-Saharan Africa and Southeast Asia, with A. duodenale mainly found in the Middle East, North Africa and India.

The excretory/secretory (ES) component, a mixture of proteins, carbo-hydrates and lipids emanating from the parasite, represents the host–parasite interface and is probably involved in modulation of the hostimmune responses to promote the survival of the parasite. The doghookworm A. caninum is the common model for the study of hookworminfection and information from its genome coupled with functional geno-mics and proteomics is accelerating the move towards human hookwormcontrol. This work has resulted in the identification of a suite of ESproteins which are important for the parasitic lifestyle and which provide

insights into the biology of hookworm infection. For example, Abubuckeret al. (2008)   generated 104,000 genome survey sequences (GSSs) andassembled them into 57.6 Mb of unique sequence, while  Mulvenna et al.(2009) identified 105 different proteins and characterised much of the ESproteome.

Vaccine research targets both the larval and adult stages of the worm but vaccine candidates based on the larval forms are in the lead. Thecurrently most promising vaccine candidate is the  N. americanus  ASP-2(Na-ASP-2) antigen, first shown in secretions from  A. duodenale  but lateralso isolated from N. americanus  (Diemert et al., 2008). A well-controlledstudy has shown this candidate to be safe in animals and capable of inducing protective responses, consisting of both specific IgG antibodiesand cellular immune responses. (Bethony et al., 2008). A Phase I safetytrial has been completed in the United States, while corresponding Phase Itrials in endemic areas are underway (Bethony et al., 2008).

With regard to potential adult worm antigens, vaccine-orientedresearch has focused on how the worm feeds, specifically investigatinghow to interfere with the action of the enzymes involved in the breakingdown of haemoglobin. Indeed, several of the proteins involved in theproteolytic cascade utilized by the adult worm to degrade haemoglobulinfrom host erythrocytes, and thus essential for its nutrition and survival,have been shown to induce protective immune responses. Among these,cysteine protease-haemoglobinase (CP-2) (Loukas et al., 2004), aspartic

308   Robert Bergquist and Sara Lustigman

Page 13: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 13/30

protease-haemoglobinase (APR-1) (Loukas et al., 2005) and glutathioneS-transferase (GST) (Zhan et al., 2005) have been selected as possibletargets as they are essential for the digestive pathway. In fact, work

already carried out has shown that vaccination with the two formerantigens has been shown to reduce blood loss and faecal egg counts indogs (Loukas et al., 2004, 2005). Although an attack against the larvalstages by the Na-ASP-2 antigen, combined with interference of the adultworm’s digestive pathway, is theoretically attractive, we are not yet there.Nevertheless, great strides have been made and a partially effectivevaccine should soon be within reach.

LF, also known as elephantiasis, directly affects more than 120 millionpeople with about 10 times more at risk in the 80 countries where the

infection is now endemic (WHO fact sheet No. 102 on lymphatic filariasis,2010). One-third of those infected live in India, one-third in Africa andmost of the remainder in Southeast Asia. The overall prevalence of LF isincreasing due to the rapid, unplanned growth of cities producingexpanded breeding sites for the mosquitoes that transmit the disease.The causative filarial worms  W. bancrofti  and B. malayi  (see life cycle on back of volume 72) lodge in the lymphatic system of humans, the defini-tive host of these parasites. Despite the hostile environment surroundingthem, these nematodes can survive up to 6 years, which they accomplish

 by adopting various immunomodulatory strategies. During its lifespan,each worm produces millions of microfilariae that eventually reach andcirculate in the blood, assuring transmission to the mosquito vector.Application of drugs targeting the first-stage-larvae (L1) can block trans-mission but there are also drawbacks such as inadequate drug coverage,reappearance of infection through migration of infected people into con-trolled areas and partial success leading to reduced compliance. This hasled to a call for complementary approaches that include both improvedchemotherapy and vaccine development. Mouse and gerbil models of Brugia   infection have provided information on the immune responseelicited by the different stages of these nematodes. Studies by Lawrenceand Devaney (2001) reinforce the concept that the different developmen-tal stages each have their own mechanism of modulating responses lead-ing to the down-regulation of potentially host beneficial immuneresponses. In an approach similar to that used by Almeida et al. (2009)for identifying potential vaccine antigens from   T. solium,   Nagaraj et al.(2008)   have carried out a large-scale analysis of excreted or secretedproteins inferred from EST data. Although this inventory of known andnovel excreted or secreted proteins covers an enormous range of nema-todes, it can be used as a source of new vaccine candidates against filarialworms infecting humans.

A large-scale, proteomic analysis to identify the ES products of the L3,L3 to L4 moulting, adult male, adult female and microfilarial stages of the

Vaccine Development and Control of Helminthic Infections   309

Page 14: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 14/30

filarial parasite   B. malayi   has recently been published (Bennuru et al.,2009). This analysis provides extended insight into the host–parasiteinteraction, while the reported abundance of a number of previously

characterised immunomodulatory proteins in the ES of microfilariaeincreases the chances of identifying novel vaccine candidates.Various vaccine candidates have already been put forward. For exam-

ple, Veerapathran et al. (2009), focusing on the key role of GST for thesurvival of the parasite in the host, achieved 61% protection againstB. malayi   challenge infection in subsequent vaccination studies in the jird model. This work built on the effect of human and mice anti-GSTantibodies in an antibody-dependent cellular cytotoxicity (ADCC) assay(Veerapathran et al., 2009). In another development,   Vedi et al. (2008)

showed the potential of  B. malayi  recombinant myosin as a vaccine in arodent model. The authors reported a 76% reduction in microfilarial burden and a 54–58% lower adult worm establishment that was conferredthrough the induction of both humoral and cellular immunities (Vediet al., 2008). There are also other antigens worth mentioning, for exampleALT-2 (Ramachandran et al., 2004), a microfilarial soluble 38 kDa proteaseisolated from   B. malayi   (Krithika et al., 2005) and a zinc-containing175 kDa collagenase (Pokharel et al., 2006) that have shown significantprotection against   B. malayi   in animal models. Any of these antigens

have the potential to be developed into a useful vaccine but there is stilla long way before a vaccine against LF will reach the stage of clinicaltrials.

Contrary to LF, in which the adult worms cause the pathology, themicrofilariae constitute the culprit in onchocerciasis, a disease caused bythe filarial worm O. volvulus. The infection is transmitted by the Simulium blackfly vector and is almost exclusively found in Africa. However,isolated foci also exist in Yemen and six countries in central and SouthAmerica (WHO, 2009a). Onchocerciasis affects an estimated 40 millionpeople, causing visual impairment in half a million; the disease alsocauses depigmentation and a severe, unrelenting itching. For years, con-trol activities were based on insecticides sprayed by aircraft over the blackfly breading sites but with the donation of MectizanÒ (ivermectin), by the U.S. Company Merck & Co. in 1987 (Colatrella, 2008), controloperations changed towards chemotherapy. Regrettably, resistance tothis drug appeared in the veterinary field early on (Coles et al., 2005;Egerton et al., 1988; Xu et al., 1998). The risk for a parallel situation vis-a-vis O. volvulus is therefore a worry and it seems already to be emerging(Boussinesq, 2008; Lustigman and McCarter, 2007; Osei-Atweneboanaet al., 2007). Above all, this development is a prompt for immediate andincreased activities in the vaccine field.

Like so many other parasitic diseases, there is a lack of good animalmodels as O. volvulus can develop fully only in humans. In a recent review

310   Robert Bergquist and Sara Lustigman

Page 15: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 15/30

(Allen et al., 2008), however,   Litomosoides sigmodontis   in mice andO. ochengi   in cattle are placed in the context of how these models can better our ability to control the human disease. The case for vaccine

development with regard to  O. volvulus  has recently been reviewed byLustigman and Abraham (2009)   and several other authors have alsostressed the need for a vaccine (Boussinesq, 2008; Cook et al., 2001).Importantly, protective immunity against   O. volvulus   larvae has now been definitively demonstrated in humans, cattle and mice, thereby prov-ing the conceptual underpinnings that a vaccine can be produced againstthis infection (Lustigman and Abraham, 2009). As noted by   Nutman(2002), an additional modality, complementing chemotherapy and vectorcontrol, is conditional to eliminate onchocerciasis. This makes it all the

more important to develop a vaccine before resistance has spread widelyrendering an integrated approach impossible. Vaccine studies supported by the Edna McConnell Clark Foundation USA resulted in the identifica-tion of 15 protective antigens out of 44 screened that induced significant but partial protection, using the diffusion chamber model in mice(Lustigman et al., 2002). Additional numbers of antigens with protectiveproperties have been reported in the last decade, for example Ov-FBA-1(McCarthy et al., 2002), Ov-ASP-1 (MacDonald et al., 2004), Ov-ALT-1(Wu et al., 2004), Ov-GAPDH (Erttmann et al., 2005), Ov-AST-1 (Borchert

et al., 2007) and paramyosin (Erttmann and Bu¨

ttner, 2009). However,as with vaccines against LF, much more work is needed before a vaccinecan be put to use. Recent reports using the cow model and  O. ochengihave clearly proved the possibility of developing vaccines againstO. volvulus   as well (Achukwi et al., 2007; Makepeace et al., 2009;Tchakoute et al., 2006).

10.4.3. Trematodes

Several species of flatworm threaten human health. Some exist only inSoutheast Asia, that is   Clonorchis sinensis,   Paragonimus   spp. andOpisthorchis   spp. (Keiser and Utzinger, 2005), while others are globallydistributed, for example Fasciola and  Schistosoma. The diseases caused byClonorchis,  Paragonimus,   Opisthorchis   and also   Fasciola   may be groupedtogether as foodborne treamatodiases due to the way they are transmit-ted. Apart from F. hepatica (McManus and Dalton, 2006; Vilar et al., 2003),the few antigens reported for this group mainly regard diagnostic use butreports on protective antigens have started to appear (Zhou et al., 2008).Interestingly, in this connection, the schistosomiasis Sm14-FABP vaccinecandidate (Tendler and Simpson, 2008; Vilar et al., 2003) cross-reacts withFasciola antigens (see below).

With close to 800 million people in 74 countries at risk, and directlyaffecting more than 200 million (Steinmann et al., 2006; WHO, 2009b),

Vaccine Development and Control of Helminthic Infections   311

Page 16: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 16/30

schistosomiasis is the second-most, socio-economically devastating para-sitic disease after malaria. Five species cause human schistosomiasis butonly two exist in Southeast Asia (for life cycle see back of volume 72).

S. japonicum   is the only species in the People’s Republic of China(P.R. China) and the Philippines, while small pockets of S. mekongi infectionconstitute serious, local problems in Cambodia and Lao People’s Demo-cratic Republic (Attwood et al., 2008). In contrast to all other schistosomiasisspecies, S. japonicum is a zoonotic infection affecting a wide range of animals,including wild and domestic ungulates as well as rodents, which all act asreservoirs. The only available drug is praziquantel, which is also one of thefew not fully subsidized drugs. With an estimated 423 million tabletsneeded globally every year (The Carter Center Schistosomiasis Control

Program, Atlanta, USA, 2010), the total expenditure needed for control isstaggering even though the average cost per dose is less than 20 cents.Although modern schistosomiasis control has clearly shown that che-

motherapy alone is capable of reducing morbidity in the human host(WHO, 2002a,b), rapid re-infection is a reminder that the impact of drugtreatment on transmission is marginal. The case for schistosomiasisvaccine development is based on the understanding that vaccination,even if not 100% effective, would contribute to long-term reduction of egg-excretion from the host. An effective vaccine would also contribute to

a positive trade-off regarding the aggressive inflammatory response thathas been observed following interrupted chemotherapy in children livingin high-transmission areas (Olveda et al., 1996; Reimert et al., 2008). Theunderlying reason for this ‘rebound morbidity’ is unclear but is probablydue to interruption of the Th1 response reducing the modulation thatnormally takes place during the course of natural infection.

Theargumentssupporting theutility of a vaccine againstschistosomiasis, based on more than 50 years of laboratory and field research, are strong.For example, it has long been known that humans living in schistosome-endemic areas develop some degree of protection naturally (Butterworthet al., 1985) and the injection of mice with irradiated schistosome cercariaeconsistently induce 60–85% protection (Dean, 1983). Vaccine developmentwas originally focused on   S. mansoni   but a panel of well-characterisedS. japonicum antigens have now also shown protective efficacy in animals justifying support for further consideration (McManus and Loukas, 2008).Due to its wide spectrum of final hosts, a ‘transmission-blocking’ veterinaryvaccine is the priority in areas in which S. japonicum is endemic. The possi- bility that this approach could pay off is supported by studies in P.R. Chinashowing that the animal–snail–human transmission cycle is more prominentthan the human–snail–human cycle in sustaining the infection in the field(Gray et al., 2007). An additional advantage in S. japonicum experimentationis that the access to full-size animal models escapes the limitations of themouse model ( Johansen et al., 2000; Zhu et al., 2006).

312   Robert Bergquist and Sara Lustigman

Page 17: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 17/30

More than a hundred schistosome antigens have been identified andcharacterised but few have shown sufficient promise to be selected forfurther development. Despite the fact that Sh28-GST, a  S. haematobium

GST molecule, is the current lead candidate, the great majority of  Schisto-soma   candidate vaccine antigens, reviewed by   Bergquist and Colley(1998), derive from  S. mansoni. The most well-researched are Sm28-GSTand Sh28-GST (Capron, 1998; Capron et al., 2005), paramyosin (Pearceet al., 1988), triose phosphate isomerase (Sm28-TPI) (Harn et al., 1992),Sm37-GADPH (Goudot-Crozel et al., 1989), Sm14-FABP (Tendler andSimpson, 2008; Vilar et al., 2003) and Sm-p80 calpain (Ahmad et al.,2009; Siddiqui et al., 2005). There are also multiple antigenic peptide(MAP) constructs made from various integrated membrane antigens

such as Sm10, Sm23, Sm28-TPI and Sm28-GST (Argiro et al., 2000; Ferruet al., 1997; Ribeiro de Jesus et al., 2000). The average protection of theseantigens, which have been tested either as native, full-length antigens,recombinant antigens, MAP constructs or as DNA vaccines, is around50% (in some cases higher) in the various animal models used.

The vaccine candidates mentioned above have been developed duringthe last two decades. Meanwhile, technology has become progressivelymore sophisticated resulting in an unprecedented expansion of parasitesequence databases. This accumulation of molecular data allows rational

vaccine discovery such as the two novel  S. mansoni   vaccine candidatesdetected at the parasite surface by proteomics which was recentlyreported by DeMarco and Verjovski-Almeida (2009).

Although fund raising for vaccine development has become anincreasingly uphill exercise, nationally available funds have beeninvested in Brazil and France, supporting the road toward clinical trialsfor Sm14-FASB and Sh28-GST, respectively. Industrial scale-up was firstachieved for Sh28-GST, now in clinical trials under the name of Bilhvax(Capron et al., 2005). This vaccine candidate has successfully passedPhase I/II clinical trials and been shown to be safe, producing Th2cytokines (IL-5 and IL-13) followed by high titres of neutralizing antibo-dies after three injections. Chemotherapy followed by immunization wasfelt to be the most appropriate modality and the best time to give thevaccine seems to be about three months after treatment, when patientshave switched to the Th2 type of response which takes time to occur andis generally not seen until after drug treatment. Therefore, the Phase IItrials of Bilhvax were based on this model, including both primary clinicaland secondary parasitological endpoints in measuring efficacy.

Industrial scale-up has also been achieved for Sm14-FABP (Tendlerand Simpson, 2008). Interestingly, thanks to a shared antigen betweenFasciola and Schistosoma, both natural infection and experimental animalresearch show cross-protection (Vilar et al., 2003). The former parasitecauses great losses in sheep and cattle breeding and can also infect

Vaccine Development and Control of Helminthic Infections   313

Page 18: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 18/30

humans. Commercial interest in a vaccine for veterinary applications hascontributed to move this vaccine candidate into advanced veterinary fieldtrials which made it possible to piggyback the adoption of the same

molecule for developing a human vaccine against schistosomiasis(Tendler and Simpson, 2008). A collaborative initiative in Brazil for thescale-up of Sm14-FABP according to Good Manufacturing Practice (GMP)was established between the government-funded research centreOswaldo Cruz Foundation (FIOCRUZ) and Butantan, a producer of vac-cines for the Brazilian Ministry of Health. The previously used laboratory-scale expression system was substituted for systems based on vectorsappropriate for GMP production. Studies of the gene structure of Sm14were undertaken and provided the basis for functional and structural

analysis to access the preparation of a more stable form of the antigen based on site-directed mutagenesis. Stability and functionality (fatty acid binding) quality control assays were designed and developed. The newconstruct provided a highly purified protein in large yields with pre-served protective activity for both parasites opening the way to Phase Isafety trials (Ramos et al., 2003, 2009).

As   S. japonicum   is a zoonotic infection, the possibility of creating atransmission-blocking vaccine for livestock offers a shortcut in the devel-opment of vaccines for P.R. China and the Philippines. This and the recent

publication of the  S. japonicum   genome (Schistosoma japonicum  GenomeSequencing and Functional Analysis Consortium et al., 2009) have nodoubt contributed to the increase in activities focused on this species inthe last few years (Da’dara et al., 2008; McManus and Dalton, 2006;McManus and Loukas 2008; Zhu et al., 2004, 2006). Based on the notionthat reduced schistosome infection in water buffaloes would also reducedisease transmission to humans, randomised double blind trials in water buffaloes using DNA vaccines encoding well-researched   S. japonicumantigens (Sj28-TPI, Sj23) have taken place in P.R. China (Da’dara et al.,2008). The results were all close to 50% protection which exceeds thehypothetical level predicted by mathematical modelling to be needed toachieve a significant reduction of schistosome transmission.

10.5. INDUSTRIAL VACCINE PRODUCTION

It has been a long road in the development of vaccines against parasiticdiseases, and as scientific hurdles are overcome and we are reaching theindustrial level, the identification of partners and financial support growsin importance. This undertaking is as critical as dealing with the science, but the problems may be unfamiliar as they are played out in the realworld of commerce and politics. The commercial-scale production of GMP-grade material, required at the clinical-trials level, is not only

314   Robert Bergquist and Sara Lustigman

Page 19: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 19/30

more expensive than the previous developmental steps but can also bemore difficult. Indeed, the transfer from the research laboratory to indus-try amounts to a real bottleneck, capable of making or breaking a vaccine

candidate. In fact, two of the most promising schistosomiasis vaccinecandidates had to be shelved for this reason (Bergquist and Colley,1998). In one case, sustained industrial production was not possible evenafter large amounts had been made in the laboratory and commercial-grade production had originally achieved production at the gram-level by the commercial partner. Another predicament, stressed by Lightowlers(2006a), is that the uncertain market potential of the product generallydashes the hope to attract the interest of bio-pharmaceutical companiesin the industrialized countries. However, there are real possibilities in

the developing, endemic countries as they have a vested interest inproducing vaccines for their own needs. For example, the vaccine produc-tion facility at the Research Institute of Tropical Medicine (RITM) in thePhilippines, which was established using a modular, turnkey approach forthe production of certified GMP-grade biological materials according toStandard Operating Procedures (SOP). At present, RITM is producingBCG to cover national needs. However, with this facility in place, addinga few more modules would not be an insurmountable barrier. Thus, multi-purpose industrial plants can be established in endemic countries, not

only for use as vaccine research/development laboratories but also for batch scale-up for clinical trials and, eventually, for full-scale vaccineproduction.

Vaccine development requires long-term commitment as well as sus-tained, high-level funding (Todd and Colley, 2002) and, as shown inFig. 10.1, the process is one of increasing risks. Once promising antigenshave been identified and tested in pilot studies, the researchers must learnto master laboratory large-scale production and focus on implementationof the vaccine in the field. At this point, the workload multiplies asactivities become more multifaceted requiring a different infrastructure,and when this has been put in place, the demanding phase of applied fieldstudies begins. Like the move from the bench to the field, the change fromexperimental approaches to industrial GMP-grade production of antigenmaterial is one of increasing complexity. In fact, there are steps involvedin the process (ringed in the figure) which are critical to the developmen-tal chain: for example without convincing, independent protection stud-ies the project must revert back to the bench and, if large amounts of standard material cannot be produced in a sustainable manner, the devel-opmental chain breaks and no further work is possible even with vaccinecandidates shown to be protective and overall strongly experimentallysupported. Finally, after safety and immunogenicity have been shown(Phase I), there is still no guarantee that the vaccine will prove effective inthe field (Phase II/III). However, when all is said and done, the fact that

Vaccine Development and Control of Helminthic Infections   315

Page 20: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 20/30

we are entering this phase is testimony to a series of successes in thelaboratory and field which augur well for the future.

Clinical-trials are time consuming and constitute the most labour-intensive part of the whole process. A Phase I trial can be carried outwith limited staff and funding but the steps to follow require multifacetedactivities. Already at the Phase II level, the need for staff multiplies andresource consumption increases logarithmically, as this step entails vac-cination and follow-up testing of large numbers of people in an endemicarea. The even longer observation periods needed to follow the immuno-logical responses in the subjects participating in trials to show proof of efficacy (Phase III) may well hamper securing the funds needed. The finallevel (Phase IV) represents the follow-up of an already licensed productfor hidden side effects, an activity that requires even larger populationdata and goes on for a long time indeed. After that, regulatory authorities,involving many administrative levels, will have their say and the hurdlesto be overcome at this stage often require considerable additional input.The consequence is that the time from discovery to ready product takesdecades rather than years.

The situation regarding vaccines for the veterinary market are some-what less constrained than that of vaccines aimed to be used in humans.

Antigens andhost responses

Identification

Phase I

Field studies

Antigenproduction

Industrial-scale

Clinical trials

Vaccine development:

discovery to implementation

Main objective

Study areaSubjectsNumberTime

Endpoints

Safety

Nonendemic

Healthy adults> 20

About 3 months

Adverse effects

Laboratory-scale

Selection

Phase IV

Expenditure

Protection studies

Efficacy General impact

Endemic Endemic

Infected/healthy Infected/healthy>1000 Many thousands

Up to 5 years > 5 years

Immunogenicity

Endemic

Infected/noninf.>100

About 3 years

Immune responses Protection Long-term effects

Phase II Phase III

FIGURE 10.1   Many steps are involved in the process of vaccine development, most of 

them more technically demanding (symbolized by larger script) than the previous one.

The three encircled stages in this process are critical, that is convincing protection in

animal models, ability to scale-up antigen production and showing impact in the field

(evidence of human protection).

316   Robert Bergquist and Sara Lustigman

Page 21: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 21/30

This may explain the rapid progress of vaccines against  E. granulosus,T. solium and F. hepatica. Rather than intended for prevention of infectionsin livestock per se, some vaccines against zoonotic parasites, notably that

against   T. solium, have been developed to assist with the control of transmission of the human disease. Although animals rather thanhumans are the recipients of these vaccines, the main aim is not protectionof livestock but the control of human disease resulting from ingestion of infected meat. However, as these diseases occur primarily in the develop-ing world, vaccines against them are of little commercial interest. Forexample, the T. ovis  vaccine was registered by the New Zealand AnimalRemedies Board in February 1994 (Rickard et al., 1995). In spite of the highefficacy of this vaccine, which has the capability to wipe out neurocysti-

cercosis if properly applied, the product has yet to be generally applied inthe endemic areas on a large scale.Table 10.1 summarizes the current situation in a way that illuminates

differences and similarities between vaccine-related activities in the vari-ous fields covered. An obvious difference is that progress has been par-ticularly strong in the field of schistosomiasis vaccines. This does notreflect that this is an easier parasite to work with but is rather a reflectionof the relatively large amounts of financial support available for thisparasite in the 1980s and 1990s. At this time, the UNICEF/UNDP/

World Bank/WHO Special Programme for Research and Training inTropical Diseases (TDR), the United States National Institutes of Health(NIH) and the Edna McConnell Clark Foundation (EMCF) (USA) ran largewell-funded research programmes; first on schistosomiasis and later ononchocerciasis. In addition, the United States Agency for InternationalDevelopment (USAID) and the Government of Egypt supported a 10-year general Schistosomiasis Research Project (SRP) continued by SVDP,a specific Schistosomiasis Vaccine Development Project (Bergquist andColley, 1998).

10.6. CONCLUDING REMARKS

Vector control and drug administration have reduced infection and dis-ease rates for many parasitic diseases significantly. However, as we arecurrently learning from the field of onchocerciasis, ivermectin’s selectiveactivity on microfilariae, the need for 10–15 years of annual treatments,logistical obstacles and the emergence of drug-resistant strains demandalternative strategies. This is also the case for schistosomiasis, in whichlarge-scale drug distribution is proving a stopgap solution which must befollowed up by the development of an integrated control process.

Helminthic diseases of both humans and animals tend to occur togethergeographically but have historically been targeted by disease-specific

Vaccine Development and Control of Helminthic Infections   317

Page 22: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 22/30

TABLE 10.1   Overview of candidates for some helminth vaccines

Parasite Antigen discovery Industrial scale-up Phase I trial Phase

T. solium*    >10

E. granulosus   >20

E. multilocularis   <10

Hookworm   >10 1** 

W. bancrofti   >10

B. malayi   >10

O. volvulus   >10S. mansoni   >100***  3

S. haematobium   >10*** 

S. japonicum   <100*** 

S. mekongi   ?

Food-borne infection   <5

*  Vaccine for pigs intended to benefit the human host by breaking the transmission cycle.**  Two vaccine candidates upscaled.

***  Corresponding antigens exist in the various species; only a few are species-specific.

Page 23: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 23/30

treatment programs which are now being integrated into a strategy of preventive chemotherapy. Although these programmes have made con-siderable progress reducing prevalence and intensity of disease, this

approach runs the risk not only of having to be continued indefinitely but also of inducing drug resistance without the long-term protection avaccine can offer as a complementary intervention.

A particularly interesting observation is that transmission-blockingvaccines developed for intermediate or reservoir hosts have largely beensuccessful, for example the pioneering T. solium vaccine initiative againsthuman cysticercosis is being emulated by a vaccine against S. japonicumtargeting the water buffalo reservoir. The fact that these veterinary vac-cines also contribute to creation of healthier animal herds is worth

highlighting.The successful vaccines against taeniases show that it is indeed possi- ble to create a sterilizing vaccine against a multicellular parasite. Regret-fully though, it seems that, despite the availability of excellent vaccines,they will not be widely applied without a commercial incentive. This canonly be remedied by convincing international donor agencies and privatefoundations to support large-scale vaccination projects and/or by publichealth provision of vaccines through local production in the endemiccountries themselves.

The current accumulation of molecular data and expansion of parasitesequence databases is providing a fresh start by permitting a more ratio-nal approach to vaccine discovery. A range of new vaccines can beexpected during the next few years provided international donor organi-zations and private foundations can agree on a joint, major NTD vaccineinitiative. The task ahead is to assure product delivery by convincingpotential donors that vaccine production in the developing world is arealistic goal worth supporting.

REFERENCES

Abubucker, S., Martin, J., Yin, Y., Fulton, L., Yang, S.P., Hallsworth-Pepin, K., et al., 2008. Thecanine hookworm genome: analysis and classification of   Ancylostoma caninum  surveysequences. Mol. Biochem. Parasitol. 157, 187–192.

Achukwi, M.D., Harnett, W., Enyong, P., Renz, A., 2007. Successful vaccination againstOnchocerca ochengi   infestation in cattle using live   Onchocerca volvulus   infective larvae.Parasite Immunol. 29, 113–116.

Acosta, L.P., Waine, G., Aligui, G.D., Tiu, W.U., Olveda, R.M., 2002. Immune correlate study

on human  Schistosoma japonicum  in a well-defined population in Leyte Philippines: II.Cellular immune responses to S. japonicum recombinant and native antigens. Acta Trop.84, 137–149.

Ahmad, G., Zhang, W., Torben, W., Haskins, C., Diggs, C., Noor, Z., Le, L., et al., 2009. Prime- boost and recombinant protein vaccination strategies using Sm-p80 protects against

Vaccine Development and Control of Helminthic Infections   319

Page 24: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 24/30

Schistosoma mansoni infection in the mouse model to levels previously attainable only bythe irradiated cercarial vaccine. Parasitol. Res. 105, 1767–1777.

Allen, J.E., Adjei, O., Bain, O., Hoerauf, A., Hoffmann, W.H., Makepeace, B.L., et al., 2008.Of mice, cattle, and humans: the immunology and treatment of river blindness. PLoS

Negl. Trop. Dis. 2, e217.Almeida, C.R., Stoco, P.H., Wagner, G., Sincero, T.C., Rotava, G., Bayer-Santos, E., et al., 2009.

Transcriptome analysis of   Taenia solium   cysticerci using Open Reading Frame ESTs(ORESTES). Parasit. Vectors 2, 35.

Al-Sherbiny, M., Osman, A., Barakat, R., El Morshedy, H., Bergquist, R., Olds, R., 2003.In vitro  cellular and humoral responses to  Schistosoma mansoni   vaccine candidate anti-gens. Acta Trop. 88, 117–130.

Amri, M., Mezioug, D., Touil-Boukoffa, C., 2009. Involvement of IL-10 and IL-4 in evasionstrategies of  Echinococcus granulosus  to host immune response. Eur. Cytokine Netw. 20,63–68.

Argiro, L., Henri, S., Dessein, H., Kouriba, B., Dessein, A.J., Bourgois, A., 2000. Induction of a

protection against   S. mansoni   with a MAP containing epitopes of Sm37-GAPDH andSm10-DLC. Effect of coadsorption with GM-CSF on alum. Vaccine 18, 2033–2038.

Attwood, S.W., Fatih, F.A., Upatham, E.S., 2008. DNA-sequence variation among Schistosomamekongi populations and related taxa; phylogeography and the current distribution of Asian schistosomiasis. PLoS Negl. Trop. Dis. 2, e200.

Bennuru, S., Semnani, R., Meng, Z., Ribeiro, J.M., Veenstra, T.D., Nutman, T.B., 2009.  Brugiamalayi excreted/secreted proteins at the host/parasite interface: stage- and gender-specificproteomic profiling. PLoS Negl. Trop. Dis. 3, e410.

Bergquist, N.R., Colley, D.G., 1998. Schistosomiasis vaccine: research to development. Para-sitol. Today 14, 99–104.

Bergquist, R., Utzinger, J., McManus, D.P., 2008. Trick or treat: the role of vaccines in

integrated schistosomiasis control. PLoS Negl. Trop. Dis. 2, e244.Berriman, M., Haas, B.J., LoVerde, P.T., Wilson, R.A., Dillon, G.P., Cerqueira, G.C., et al.,

2009. The genome of the blood fluke  Schistosoma mansoni. Nature 460, 352–358.Bethony, J.M., Simon, G., Diemert, D.J., Bethony, J.M., Simon, G., Diemer, D.J., et al., 2008.

Randomized, placebo-controlled, double-blind trial of the Na-ASP-2 hookworm vaccinein unexposed adults. Vaccine 26, 2408–2417.

Borchert, N., Becker-Pauly, C., Wagner, A., Fischer, P., Stocker, W., Brattig, N.W., 2007.Identification and characterization of onchoastacin, an astacin-like metalloproteinasefrom the filaria Onchocerca volvulus. Microbes Infect. 9, 498–506.

Boussinesq, M., 2008. Onchocerciasis control: biological research is still needed. Parasite 15,510–514.

Brindley, P.J., Mitreva, M., Ghedin, E., Lustigman, S., 2009. Helminth genomics: the implica-tions for human health. PLoS Negl. Trop. Dis. 3, e538.

Burnette, W.N., 1981. "Western blotting": electrophoretic transfer of proteins from sodiumdodecyl sulfate – polyacrylamide gels to unmodified nitrocellulose and radiographicdetection with antibody and radioiodinated protein A. Anal. Biochem. 112, 195–203.

Butterworth, A.E., Capron, M., Cordingley, J.S., Dalton, P.R., Dunne, D.W., Kariuki, H.C.,et al., 1985. Immunity after treatment of human schistosomiasis mansoni. II. Identificationof resistant individuals, and analysis of their immune responses. Trans. R. Soc. Trop.Med. Hyg. 79, 393–408.

Capron, A., 1998. Schistosomiasis: forty years’ war on the worm. Parasitol. Today 14,379–384.

Capron, A., Riveau, G., Capron, M., Trottein, F., 2005. Schistosomes: the road from host-parasite interactions to vaccines in clinical trials. Trends Parasitol. 21, 143–149.

Chow, C., Gauci, C.G., Vural, G., Jenkins, D.J., Heath, D.D., Rosenzvit, M.C., et al., 2008.Echinococcus granulosus: variability of the host-protective EG95 vaccine antigen in G6 andG7 genotypic variants. Exp. Parasitol. 119, 499–505.

320   Robert Bergquist and Sara Lustigman

Page 25: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 25/30

Churcher, T.S., Pion, S.D., Osei-Atweneboana, M.Y., Prichard, R.K., Awadzi, K.,Boussinesq, M., et al., 2009. Identifying sub-optimal responses to ivermectin in thetreatment of River Blindness. Proc. Natl. Acad. Sci. U.S.A 106, 16716–16721.

Colatrella, B., 2008. The Mectizan Donation Program: 20 years of successful collaboration – a

retrospective. Ann. Trop. Med. Parasitol. 102 (Suppl. 1), 7–11.Coles, G.C., Rhodes, A.C., Wolstenholme, A.J., 2005. Rapid selection for ivermectin resis-

tance in Haemonchus contortus. Vet. Parasitol. 129, 345–347.Cook, J.A., Steel, C., Ottesen, E.A., 2001. Towards a vaccine for onchocerciasis. Trends

Parasitol. 17, 555–558.Coupar, B.E., Oke, P.G., Andrew, M.E., 2000. Insertion sites for recombinant vaccinia virus

construction: effects on expression of a foreign protein. J. Gen. Virol. 81, 431–439.Craig, P.S., McManus, D.P., Lightowlers, M.W., Chabalgoity, J.A., Garcia, H.H., Gavidia, C.M.,

et al., 2007. Prevention and control of cystic echinococcosis. Lancet Infect. Dis. 7, 385–394.Da’dara, A.A., Li, Y.S., Xiong, T., Zhou, J., Williams, G.M., McManus, et al., 2008. DNA-based

vaccines protect against zoonotic schistosomiasis in water buffalo. Vaccine 26, 3617–3625.

Dang, Z., Watanabe, J., Kajino, K., Oku, Y., Matsumoto, J., Yagi, K., et al., 2009a. Molecularcloning and characterization of a T24-like protein in   Echinococcus multilocularis. Mol.Biochem. Parasitol. 168, 117–119.

Dang, Z., Yagi, K., Oku, Y., Kouguchi, H., Kajino, K., Watanabe, J., et al., 2009b. Evaluation of Echinococcus multilocularis  tetraspanins as vaccine candidates against primary alveolarechinococcosis. Vaccine (Sep 25).

Dean, D.A., 1983. Schistosoma and related genera: acquired resistance in mice. Exp. Parasitol.55, 1–104.

DeMarco, R., Verjovski-Almeida, S., 2009. Schistosomes – proteomics studies for potentialnovel vaccines and drug targets. Drug Discov. Today 14, 472–478.

Diemert, D.J., Bethony, J.M., Hotez, P.J., 2008. Hookworm vaccines. Clin. Infect. Dis. 46,

282–288.Duerr, H.P., Hoffmann, W.H., Eichner, M., 2008. Does resistance to filarial reinfections

 become leaky over time? Trends Parasitol. 24, 350–354.Egerton, J.R., Suhayda, D., Eary, C.H., 1988. Laboratory selection of  Haemonchus contortus for

resistance to ivermectin. J. Parasitol. 74, 614–617.Erttmann, K.D., Buttner, D.W., 2009. Immunohistological studies on   Onchocerca volvulus

paramyosin. Parasitol. Res. 105, 1371–1374.Erttmann, K.D., Kleensang, A., Schneider, E., Hammerschmidt, S., Buttner, D.W., Gallin, M.,

2005. Cloning, characterization and DNA immunization of an   Onchocerca volvulusglyceraldehyde-3-phosphate dehydrogenase (Ov-GAPDH). Biochim. Biophys. Acta.1741, 85–94.

Ferru, I., Georges, B., Bossus, M., Estaquier, J., Delacre, M., Harn, D.A., et al., 1997. Analysisof the immune response elicited by a multiple antigen peptide (MAP) composed of twodistinct protective antigens derived from the parasite Schistosoma mansoni. Parasit. Immu-nol. 19, 1–11.

Fitzpatrick, J.M., Peak, E., Perally, S., Chalmers, I.W., Barrett, J., Yoshino, T.P., et al., 2009.Anti-schistosomal intervention targets identified by lifecycle transcriptomic analyses.PLoS Negl. Trop. Dis. 3, e543.

Gauci, C., Heath, D., Chow, C., Lightowlers, M.W., 2005. Hydatid disease: vaccinology anddevelopment of the EG95 recombinant vaccine. Expert. Rev. Vaccines 4, 103–112.

Ghedin, E., Wang, S., Spiro, D., Caler, E., Zhao, Q., Crabtree, J., et al., 2007. Draft genome of the filarial nematode parasite Brugia malayi. Science 317, 1756–1760.

Gottstein, B., Hemphill, A., 2008. Echinococcus multilocularis: the parasite–host interplay. Exp.Parasitol. 119, 447–452.

Goudot-Crozel, V., Caillol, D., Djabali, M., Dessein, A.J., 1989. The major parasite surfaceantigen associated with human resistance to schistosomiasis is a 37-kD glyceraldehyde-3P-dehydrogenase. J. Exp. Med. 170, 2065–2080.

Vaccine Development and Control of Helminthic Infections   321

Page 26: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 26/30

Gray, D.J., Williams, G.M., Li, Y., Chen, H., Li, R.S., Forsyth, S.J., et al., 2007. A cluster-randomized bovine intervention trial against   Schistosoma japonicum   in the People’sRepublic of China: design and baseline results. Am. J. Trop. Med. Hyg. 77, 866–874.

Grossmann, C., Tenbusch, M., Nchinda, G., Temchura, V., Nabi, G., Stone, G.W., et al., 2009.

Enhancement of the priming efficacy of DNA vaccines encoding dendritic cell-targetedantigens by synergistic toll-like receptor ligands. BMC Immunol. 3, 43.

Guo, J., Li, Y., Gray, D., Ning, A., Hu, G., Chen, H., et al., 2006. A drug-based interventionstudy on the importance of buffaloes for human Schistosoma japonicum infection aroundPoyang Lake, People’s Republic of China. Am. J. Trop. Med. Hyg. 74, 335–341.

Haag, K.L., Gottstein, B., Ayala, F.J., 2009. The EG95 antigen of  Echinococcus  spp. containspositively selected amino acids, which may influence host specificity and vaccine effi-cacy. PLoS One 4, e5362.

Harn, D.A., Gu, W., Oligino, L.D., Mitsuyama, M., Gebremichael, A., Richter, D., 1992.A protective monoclonal antibody specifically recognizes and alters the catalytic activityof schistosome triose-phosphate isomerase. J. Immunol. 148, 562–567.

Hauguel, T.M., Hackett, C.J., 2008. Rationally designed vaccine adjuvants: separating effi-cacy from toxicity. Front Biosci. 13, 2806–2813.

Hotez, P.J., Kamath, A., 2009. Neglected tropical diseases in sub-Saharan Africa: review of their prevalence, distribution, and disease burden. PLoS Negl. Trop. Dis. 3, e412.

Hotez, P.J., Bottazzi, M.E., Franco-Paredes, C., Ault, S.K., Periago, M.R., 2008. Theneglected tropical diseases of Latin America and the Caribbean: a review of disease

 burden and distribution and a roadmap for control and elimination. PLoS Negl. Trop.Dis. 2, e300.

Hu, W., Yan, Q., Shen, D.K., Liu, F., Zhu, Z.D., Song, H.D., et al., 2003. Evolutionary and biomedical implications of a Schistosoma japonicum complementary DNA resource. Nat.Genet. 35, 139–147.

 Jiang, Z.H., Koganty, R.R., 2003. Synthetic vaccines: the role of adjuvants in immune target-ing. Curr. Med. Chem. 10, 1423–1439.

 Johansen, M.V., Bogh, H.O., Nansen, P., Christensen, N.O., 2000.   Schistosoma japonicuminfection in the pig as a model for human schistosomiasis japonica. Acta Trop. 76, 85–99.

 Johnson, K.S., Harrison, G.B., Lightowlers, M.W., O’Hoy, K.L., Cougle, W.G., Dempster, R.P.,et al., 1989. Vaccination against ovine cysticercosis using a defined recombinant antigen.Nature 338, 585–587.

Keiser, J., Utzinger, J., 2005. Emerging foodborne trematodiasis. Emerg. Infect. Dis. 11,1507–1514.

Krithika, K.N., Dabir, P., Kulkarni, S., Anandharaman, V., Reddy, M.V., 2005. Identificationof 38 kDa  Brugia malayi   microfilarial protease as a vaccine candidate for lymphatic

filariasis. Indian J. Exp. Biol. 43, 759–768.Kwissa, M., Kasturi, S.P., Pulendran, B., 2007. The science of adjuvants. Expert Rev. Vaccines

6, 673–684.Kyngdon, C.T., Gauci, C.G., Gonzalez, A.E., Flisser, A., Zoli, A., Read, A.J., et al., 2006.

Antibody responses and epitope specificities to the  Taenia solium cysticercosis vaccinesTSOL18 and TSOL45-1A. Parasite Immunol. 28, 191–199.

Lafuente, E.M., Reche, P.A., 2009. Prediction of MHC-peptide binding: a systematic andcomprehensive overview. Curr. Pharm. Des. 15, 3209–3220.

Lawrence, R.A., Devaney, E., 2001. Lymphatic filariasis: parallels between the immunologyof infection in humans and mice. Parasite Immunol. 23, 353–361.

Lightowlers, M.W., 2003. Vaccines for prevention of cysticercosis. Acta Trop. 87, 129–135.Lightowlers, M.W., 2004. Vaccination for the prevention of cysticercosis. Dev. Biol. (Basel)

119, 361–368.Lightowlers, M.W., 2006a. Cestode vaccines: origins, current status and future prospects.

Parasitology 133 (Suppl.), S27–S42.

322   Robert Bergquist and Sara Lustigman

Page 27: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 27/30

Page 28: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 28/30

Nutman, T.B., 2002. Future directions for vaccine-related onchocerciasis research. TrendsParasitol. 18, 237–239.

Olveda, R.M., Daniel, B.L., Ramirez, B.D., Aligui, G.D., Acosta, L.P., Fevidal, P., et al., 1996.Schistosomiasis japonica in the Philippines: the long-term impact of population-based

chemotherapy on infection, transmission, and morbidity. J. Infect. Dis. 174, 163–172.Osei-Atweneboana, M.Y., Eng, J.K., Boakye, D.A., Gyapong, J.O., Prichard, R.K., 2007. Preva-

lence and intensity of  Onchocerca volvulus infection and efficacy of ivermectin in endemiccommunities in Ghana: a two-phase epidemiological study. Lancet 369, 2021–2029.

Pearce, E.J., James, S.L., Hieny, S., Lanar, D.E., Sher, A., 1988. Induction of protectiveimmunity against   Schistosoma mansoni   by vaccination with schistosome paramyosin(Sm97), a nonsurface parasite antigen. Proc. Natl. Acad. Sci. U.S.A 85, 5678–5682.

Persing, D.H., Coler, R.N., Lacy, M.J., Johnson, D.A., Baldridge, J.R., et al., 2002. Taking toll:lipid A mimetics as adjuvant and immunomodulators. Trends Microbiol. 10 (Suppl.),32–37.

Pichichero, M.E., 2008. Improving vaccine delivery using novel adjuvant systems. Hum.

Vaccin. 4, 262–270.Pokharel, D.R., Rai, R., Nandakumar Kodumudi, K., Reddy, M.V., Rathaur, S., 2006. Vacci-

nation with   Setaria cervi  175 kDa collagenase induces high level of protection againstBrugia malayi infection in jirds. Vaccine 24, 6208–6215.

Prieur, E., Gilbert, S.C., Schneider, J., Moore, A.C., Sheu, E.G., Goonetilleke, N., et al., 2004.A Plasmodium falciparum candidate vaccine based on a six-antigen polyprotein encoded

 by recombinant poxviruses. Proc. Natl. Acad. Sci. U.S.A 101, 290–295.Ramachandran, S., Kumar, M.P., Rami, R.M., Chinnaiah, H.B., Nutman, T., Kaliraj, P., et al.,

2004. The larval specific lymphatic filarial ALT-2: induction of protection using protein orDNA vaccination. Microbiol. Immunol. 48, 945–955.

Ramos, C.R., Figueredo, R.C., Pertinhez, T.A., Vilar, M.M., do Nascimento, A.L., Tendler, M.,

et al., 2003. Gene structure and M20T polymorphism of the  Schistosoma mansoni  Sm14fatty acid-binding protein. Molecular, functioanl, and immunoprotection analysis. J. Biol.Chem. 278, 12745–12751.

Ramos, C.R., Spisni, A., Oyama, S., Jr., Sforca, M.L., Ramos, H.R., Vilar, M.M., et al., 2009.Stability improvement of the fatty acid binding protein Sm14 from  S. mansoni  by Cysreplacement: structural and functional characterization of a vaccine candidate. Biochim.Biophys. Acta 1794, 655–662.

Reimert, C.M., Tukahebwa, E.M., Kabatereine, Narcis B., David, W., Dunne, D.W., et al.,2008. Assessment of  Schistosoma mansoni  induced intestinal inflammation by means of eosinophil cationic protein, eosinophil protein X and myeloperoxidase before and aftertreatment with praziquantel. Acta Trop. 105, 253–259.

Ribeiro de Jesus, A., Arau jo, I., Bacellar, O., Magalhaes, A., Pearce, E., Harn, D., et al., 2000.Human immune responses to   Schistosoma mansoni  vaccine candidate antigens. Infect.Immun. 68, 2797–2803.

Rickard, M.D., Harrison, G.B., Heath, D.D., Lightowlers, M.W., 1995. Taenia ovis recombinantvaccine – ’quo vadit’. Parasitology 110 (Suppl.), S5–S9.

Rosenzvit, M.C., Camicia, F., Kamenetzky, L., Muzulin, P.M., Gutierrez, A.M., 2006. Identi-fication and intra-specific variability analysis of secreted and membrane-bound proteinsfrom Echinococcus granulosus. Parasitol. Int. 55 (Suppl.), S63–S67.

Schistosoma japonicum Genome Sequencing and Functional Analysis Consortium, Zhou, Y.,Zheng, F., Liu, F., Hu, W., Wang, Z.Q., Gang, L., et al., 2009. The Schistosoma japonicumgenome reveals features of host-parasite interplay. Nature 460, 345–351.

Schwab, A.E., Boakye, D.A., Kyelem, D., Prichard, R.K., 2005. Detection of benzimidazoleresistance-associated mutations in the filarial nematode Wuchereria bancrofti and evidencefor selection by albendazole and ivermectin combination treatment. Am. J. Trop. Med.Hyg. 73, 234–238.

324   Robert Bergquist and Sara Lustigman

Page 29: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 29/30

Sciutto, E., Fragoso, G., de Aluja, A.S., Hernandez, M., Rosas, G., Larralde, C., 2008. Vaccinesagainst cysticercosis. Curr. Top. Med. Chem. 8, 415–423.

Siddiqui, A.A., Pinkston, J.R., Quinlin, M.L., Kavikondala, V., Rewers-Felkins, K.A.,Phillips, T., et al., 2005. Characterization of protective immunity induced against Schisto-

soma mansoni via DNA priming with the large subunit of calpain (Sm-p80) in the presenceof genetic adjuvants. Parasite 12, 3–8.

Steeghs, L., Tommassen, J., Leusen, J.H., van de Winkel, J.G., van der Ley, P., 2004. Teasingapart structural determinants of ‘toxic’ and ‘adjuvanticy’: implications for meningococcalvaccine development. J. Endotoxin Res. 10, 113–119.

Steinmann, P., Keiser, J., Bos, R., Tanner, M., Utzinger, J., 2006. Schistosomiasis and waterresources development: systematic review, meta-analysis, and estimates of people at risk.Lancet Infect. Dis. 6, 411–425.

Tchakoute, V.L., Graham, S.P., Jensen, S.A., Makepeace, B.L., Nfon, C.K., Njongmeta, L.M.,et al., 2006. In a bovine model of onchocerciasis, protective immunity exists naturally, isabsent in drug-cured hosts, and is induced by vaccination. Proc. Natl. Acad. Sci. U.S.A.

103, 5971–5976.Tendler, M., Simpson, A.J., 2008. The biotechnology-value chain: development of Sm14 as a

schistosomiasis vaccine. Acta Trop. 108, 263–266.The Carter Center Schistosomiasis Control Program, Atlanta, USA, 2010.  http://www.car-

tercenter.org/health/schistosomiasis/index.html.Todd, C.W., Colley, D.G., 2002. Practical and ethical issues in the development of a vaccine

against Schistosomiasis mansoni. Am. J. Trop. Med. Hyg. 66, 348–358.Torgerson, P.R., 2006. Mathematical models for the control of cystic echinococcosis. Parasitol.

Int. 55, S253–258.Torgerson, P.R., 2009. Dogs, vaccines and Echinococcus. Trends Parasitol. 25, 57–58.Vedi, S., Dangi, A., Hajela, K., Misra-Bhattacharya, S., 2008. Vaccination with 73 kDa recom-

 binant heavy chain myosin generates high level of protection against   Brugia malayichallenge in jird and mastomys models. Vaccine 26, 5997–6005.

Veerapathran, A., Dakshinamoorthy, G., Gnanasekar, M.,Reddy, M.V.,Kalyanasundaram, R.E.,2009. Evaluation of  Wuchereria bancrofti GST as a vaccine candidate for lymphatic filariasis.PLoS Negl. Trop. Dis. 3, e457.

Vilar, M.M., Barrientos, F., Almeida, M., Thaumaturgo, N., Simpson, A., Garratt, R., et al.,2003. An experimental bivalent peptide vaccine against schistosomiasis and fascioliasis.Vaccine 22, 137–144.

Wang, T.P., Vang Johansen, M., Zhang, S.Q., Wang, F.F., Wu, W.D., Zhang, G.H., et al., 2005.Transmission of  Schistosoma japonicum by humans and domestic animals in the YangtzeRiver valley, Anhui province, China. Acta Trop. 96, 198–204.

WHO, 2002a. Fact sheet N 102 on lymphatic filariasis. http://www.who.int/mediacentre/factsheets/fs102/en.

WHO, 2002b. Prevention and control of schistosomiasis and soil-transmitted helminthiasis:report of a WHO expert committee. WHO Tech. Rep. Ser. 912, pp. 1–57.

WHO, 2009a. African Programme for Onchocerciasis Control (APOC). http://www.who.int/apoc/onchocerciasis/en.

WHO, 2009b. Preventive chemotherapy databank. http://www.who.int/neglected_diseases/preventive_chemotherapy/databank/en/index.html.

Willingham III, A.L., Wu, H.-W., Conlan, J., Satrija, F., 2010. Combating   Taenia soliumCysticercosis in Southeast Asia: an Opportunity for improving human health andlivestock Production. Adv. Parasitol. 72, 235–259.

Wu, Y., Egerton, G., Pappin, D.J., Harrison, R.A., Wilkinson, M.C., Underwood, A., et al.,2004. The Secreted Larval Acidic Proteins (SLAPs) of  Onchocerca   spp. are encoded byorthologues of the alt gene family of  Brugia malayi  and have host protective potential.Mol. Biochem. Parasitol. 134, 213–224.

Vaccine Development and Control of Helminthic Infections   325

Page 30: Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

8/19/2019 Bergquist 2010 Chapter 10 – Control of Important Helminthic Infections

http://slidepdf.com/reader/full/bergquist-2010-chapter-10-control-of-important-helminthic-infections 30/30

Xu, M., Molento, M., Blackhall, W., Ribeiro, P., Beech, R., Prichard, R., 1998. Ivermectinresistance in nematodes may be caused by alteration of P-glycoprotein homolog. Mol.Biochem. Parasitol. 91, 327–335.

Zhan, B., Liu, S., Perally, S., Xue, J., Fujiwara, R., Brophy, P., et al., 2005. Biochemical

characterization and vaccine potential of a heme-binding glutathione transferase fromthe adult hookworm Ancylostoma caninum. Infect. Immun. 73, 6903–6911.

Zhang, W., McManus, D.P., 2008. Vaccination of dogs against   Echinococcosus granulosus:a means to control hydatid disease? Trends Parasitol. 24, 419–424.

Zhang, W., Ross, A.G., McManus, D.P., 2008. Mechanisms of immunity in hydatid disease:implications for vaccine development. J. Immunol. 181, 6679–6685.

Zhou, Z., Xia, H., Hu, X., Huang, Y., Li, Y., Li, L., et al., 2008. Oral administration of a  Bacillussubtilis spore-based vaccine expressing  Clonorchis sinensis  tegumental protein 22.3 kDaconfers protection against Clonorchis sinensis. Vaccine 26, 1817–1825.

Zhu, Y., Ren, J., Da’dara, A., Harn, D., Xu, M., Si, J., et al., 2004. The protective effect of aSchistosoma japonicum Chinese strain 23 kDa plasmid DNA vaccine in pigs is enhanced

with IL-12. Vaccine 23, 78–83.Zhu, Y., Si, J., Harn, D.A., Xu, M., Ren, J., Yu, C., et al., 2006.  Schistosoma japonicum triose-

phosphate isomerase plasmid DNA vaccine protects pigs against challenge infection.Parasitology 132, 67–71.

326   Robert Bergquist and Sara Lustigman