82
qwertyuiopasdfghjklzxcv bnmqwertyuiopasdfghjkl zxcvbnmqwertyuiopasdfg hjklzxcvbnmqwertyuiopa sdfghjklzxcvbnmqwertyui opasdfghjklzxcvbnmqwer tyuiopasdfghjklzxcvbnmq wertyuiopasdfghjklzxcvb nmqwertyuiopasdfghjklzx cvbnmqwertyuiopasdfghj klzxcvbnmqwertyuiopasd fghjklzxcvbnmqwertyuiop asdfghjklzxcvbnmqwerty Human Parasitic Vaccines Dr.M.Muruganandam Human Parasitic Vaccines

HUMAN PARASITIC VACCINE

Embed Size (px)

Citation preview

Page 1: HUMAN PARASITIC VACCINE

qwertyuiopasdfghjklzxcv

bnmqwertyuiopasdfghjkl

zxcvbnmqwertyuiopasdfg

hjklzxcvbnmqwertyuiopa

sdfghjklzxcvbnmqwertyui

opasdfghjklzxcvbnmqwer

tyuiopasdfghjklzxcvbnmq

wertyuiopasdfghjklzxcvb

nmqwertyuiopasdfghjklzx

cvbnmqwertyuiopasdfghj

klzxcvbnmqwertyuiopasd

fghjklzxcvbnmqwertyuiop

asdfghjklzxcvbnmqwerty

Human

Parasitic

Vaccines

Dr.M.Muruganandam

Human Parasitic Vaccines

Page 2: HUMAN PARASITIC VACCINE

Dr.M.Muruganandam

Page 3: HUMAN PARASITIC VACCINE

First Edition -2019

ISBN-978-9982-22-635—6

Publisher

Einsteein Bio-Engineering

Research Foundation.

Author

Dr.M.Muruganandam,

Email- [email protected]

www.vaccinebiotech.blogspot.com

Page 4: HUMAN PARASITIC VACCINE

Preface

Human parasite vaccine

development research is going on around the

world. Still scientists are not reach

appropriate level. Because parasites have

Complex life cycle and they need to complete

their life cycle with the help of different host.

In this book, informations are collected from

various researchers regarding current status

of Parasite vaccines. I thank all of them. I

hope this manuscript will be useful to vaccine

development researchers. Once again thanks

to all.

M.Muruganandam

Page 5: HUMAN PARASITIC VACCINE

Content

1. Parasitic disease Burden

2. Obstacles in vaccine Development

3. Malaria Vaccine

4. Leishmaniasis Vaccine

5. Amoebiasis Vaccine

6. Cestode Vaccine

7. Schistosomiasis Vaccine

8. Hook worms vaccine

9. Filariasis Vaccine

10. Prevention and control methods.

Page 6: HUMAN PARASITIC VACCINE

1. Parasite Disease Burden

Parasite infections are the leading cause of

morbidity as well as mortality in developing and

under developed countries in the world. In the

most places, the infected peoples are children,

aged people and pregnant women who are due to

weakness and poor immunity. Another important

reason is lack of awareness about infections such

as malaria, Leishmaniasis, etc and People are not

followed by proper prevention methods. 1

The intestinal and protozoan infections are the

most parasitic diseases with according to WHO,

3.5 billion people affected and 450 million,

mainly children ill as a result, Co-infection with

different parasites is common and illness is often

made worse by Co-existing malnutrition.2

The parasites have different types of antigens.

Page 7: HUMAN PARASITIC VACCINE

The antigenic variations posses a big problem

during vaccine development. The most of the

parasitic infection leads to immuno suppression

in host. The important antibodies such as IgA,

IgE and IgG play a key role in parasitic immune

responses. The polymorphic nuetrophill and

Eosinophils also plays an important role in cell

mediated immunity.

The parasitic vaccine development is

complicated process, because parasites have

various stages in life cycle in host and Vector.

So identification of proper Immunogens are very

difficult. However many attempts in vaccine

development is going on in all over the world.

The current status of vaccine development is

discussed here.

Reference

1. Muruganandam .M and N. Thiru Kumari (2016) malaria - A

Page 8: HUMAN PARASITIC VACCINE

climate sensitive disease - Proceedings of the Seminar on Global

warming and climatic changes held on 19th Aug.2016. pp. 87-91.

2. Knox. D.P.(2010). Parasite Vaccines: Recent progress in the

problems Associated with their development. The open. Infect.

Disease. jour. 2010,4; 63-73.

Page 9: HUMAN PARASITIC VACCINE

2. Obstacles in vaccine Development.

The existing anti - parasitic drugs have

been in use for decades and drug resistance

among the parasites in gradually expanding

the glaring example of which can be seen in

Plasmodium falciparum. The important

obstacles for vaccine development are as

follows.

Even the simplest parasites have complex

structure and life cycles.

There is a general lack of precise

understanding of the host/parasitic

interaction.1

Due to complex nature of parasites the

Page 10: HUMAN PARASITIC VACCINE

immune system is confronted with a highly

diverse and continuously parasite changing

the spectrum of antigen.

A number of biological characteristics of the

parasite help in making the situation more

difficult, many parasites go through a phase

of sexual reproduction, with the associated

exchange of genetic material. This results in

new parasites with a different genetic and

phenotypic makeup.

There is a different expression of genes

during the successive life cycle stages. This

will create difficult situation to host immune

system for understanding the nature of

Immunogens.

A number of species can express distinct

various stages - Specific molecules which are

antigenically different. This ability allows

Page 11: HUMAN PARASITIC VACCINE

them to avoid the host defence mechanisms.

Parasites avoid, deflect and confuse host

immune system; the right parasite antigen has

not been identified yet because of

complicated life cycle.

The protective host responses are not

understood in most target species. These

parasites candidate antigen also may show

their efficacy.

Absence of genome databases or

bioinformatics algorithms for selecting

candidate antigens of promise.

Another thing is the commercial viability of a

vaccine depends on such factors as

development and production casts, and

specific characteristics, such as storage,

transport conditions and shelf life.

Page 12: HUMAN PARASITIC VACCINE

Current drugs have efficacies approaching

100%: - It will not easy to persuade users that

a vaccine which less than 100% effective can

usefully control the disease2.

Reference

1. Vercruysse.J., Schetters, Tpm,Knox.D.P,Will Adsen,P,Clae

rebout.E. Control of parasite disease using vaccines: an

answer to drug

resistance?Rev.Sci.tech.off.int.Epiz.2007;26:105-115

2.Abhijit Chaudhury(2014).Human parasite vaccine-an

overview .J.clin.Bio.Sci. 2014;4(1):216-21.

Page 13: HUMAN PARASITIC VACCINE

3. Malaria Vaccine

3.1. Malaria

Malaria is the world's most important

tropical parasitic disease. The world wide

prevalence of the disease is in the order of 350 -

500 million clinical cases each year, with an

estimated annual death toll of over |:| million

deaths reported by WHO. Plasmodium is an

unicellular organism. It causes malaria. It is an

endoparasite living in the blood of man. It is

cosmopolitan in distribution. Plasmodium

completes its life cycle in two hosts, namely

man and the female Anopheles mosquito.

The life cycle of plasmodium in man is called

the cycle of Golgi. It occurs in three stages.

They are 1. Pre-erythrocytic cycle 2. Exo-

erythrocytic. Cycle and 3. Endoerythrocytic

cycle. Another one Cycle Present inside is the

mosquito called cycle of Ross.

Page 14: HUMAN PARASITIC VACCINE

3.2 Vaccine

The development of Vaccine for malaria has

turned out to be a highly complex exercise

owing to a multitude of difficulties. (1). A natural

malaria infection does not repeated and

prolonged exposure to malaria parasite over

several years, only partially effective immunity

is acquired, which is short lived and is highly

stage and strain specific. (2,1). This incomplete

immune response is due to the complex biology

of the plasmodium parasite. Its extensive

antigenic diversity and its immune evasion

strategies and all these factors make vaccine

development against malaria challenging (3).

Several vaccine candidates have been

tasted over the years, but without much success.

Now few malaria vaccine candidates entered in

clinical trials. The difficulty of developing a

Page 15: HUMAN PARASITIC VACCINE

highly effective malaria vaccine has led to the

design and assessment of a very wide range of

new approaches (1).

3.3. Irradiated Sporozoites

The observation that inactivated plasmodium

sporozoites could protect against malaria is

about a hundred years old. However, Systematic

demonstration of protection using irradiates

sporozoites occurred in the nineteen - sixties,

providing the impetus for the development of

malaria vaccine. Modern malaria vaccine

development works starts from immunization

studies of mice with irradiated sporozoites,

conducted in the 1960s (4). A high level of

protection could be induced in volunteers, but

required large number of bites by irradiated

infectious mosquitoes (2). Immunization of the

human subjects with irradiated sporozoites

confers high levels of protection against

Page 16: HUMAN PARASITIC VACCINE

experimental infection, suggesting that

vaccination is feasible.

Individuals in malaria endemic countries do

acquire, natural immunity that builds up slowly

requires continual antigenic stimulation and

affects the severity of disease (5). Protection has

been elicited by passive transfer of hyper

immune immuno globulins from malaria

immune adults into malaria native human

volunteers.(6).

3.4. Peptide Vaccine

In 1983, the circum sporozoites protein (CSP) , a

major sporozoites surface antigen became the

First plasmodium gene to be cloned and a CSP -

based vaccine appeared. The emergence of a

peptide based candidate vaccine from Colombia

called SPf66, with apparent efficacy in new

world monkeys and humans (2). Different

recombinant proteins are used as vaccine

Page 17: HUMAN PARASITIC VACCINE

candidate. They receive only limited success.

3.5. Apical membrane Antigen - 1

Apical membrane antigen - 1 (AMA-1) is

another leading candidate. It is present in a type.

I transmembrane Protein located in the

micronemes of the merozoite that is functional

in the rapid invasion of erythrocytes (8). Pre-

Clinical studies showed that Vaccination with

AMA-1 induces antibodies and protection

against homologous parasite challenge in both

rodent and monkey models of malaria infection

(9, 10).

Vaccination of monkeys with recombinant

plasmodium falciparum apical membrane

antigen -1 confers protection against blood stage

malaria. The outcome of a phase 1/2 a study,

which evaluated the safety, immuno genicity and

efficacy of a vaccine compressed of a

recombinant plasmodium falciparum AMA -1

Page 18: HUMAN PARASITIC VACCINE

representing the 3D7 allele formulated with

either the AS01B or ASo2A adjuvant system just

been reported (II).All vaccine formulations

stimulated similar functional antibody responses,

as judged using a growth inhibition essay against

homologous parasites and demonstrate

interferon - gamma (IFN - gamma) responses.

However, volunteer’s challenges with

P.falciparam infected mosquitoes all become

parasitimic although a small but significant

reduction of parasite in the AMA-1 ASo2 A

group was noticed.

3.6. Transmission Blocking Vaccine.

It induces antibodies against the sexual stage

antigens which prevent the development of

infectious sporozoites, the salivary glands of

Anopheles mosquitoes. The leading candidate

vaccine contain the P. falciparum surface

protein antigens Pf S25 and P fs28 or offer

Page 19: HUMAN PARASITIC VACCINE

P.vivax homologous PVS25 and PVS -28(7).

These vaccines are currently being developed at

the NIH as recombinant yeast secreted proteins.

(S. Cerevisiae). Initial human phase I trials have

been conducted for Pf s25 and should follow

soon for PVS25. Other sexual stage - specific

antigens that are being developed as

transmission - blocking vaccines are PfS48/45

and Pfs230.

The concept of attenuation and parasite

challenge to elicit immunity is also being

explored. As noted earlier, an attempt to develop

a commercial attenuated sporozoites vaccine has

been under taken by Sararia Inc. with the

support from the Bill and Melinda Gates

foundation and the NIH and the outcome of

clinical trials is a waited.

Page 20: HUMAN PARASITIC VACCINE

3.7. Erythrocytic Vaccine.

A sexual blood stage strategies aim to elicit

antibodies that will inactivate merozoite and or

target malarial antigens expensed on the RBC

surface, thus inducing antibody dependent

cellular cytotoxicity and complement lysis; they

also are meant to elicit T Cell responses that will

inhibit the development of the parasite in RBCs.

This type of vaccine would mostly serve as a

disease reduction vaccine in endemic countries

by decreasing the exponential multiplication of

merozoites.

The development of MSP-1 as a vaccine

candidate is the subject of a recent detailed

review (12). Briefly, MSP, conferred production

against challenge infection against plasmodium

yoelii in the laboratory mice 13 and passive

immunization with monoclonal antibodies

(MAbs) also provided in the same model,

Page 21: HUMAN PARASITIC VACCINE

highlighting in the importance of antibody for

production. (14,15).

Gene sequence analysis (16) allowed detailed

structural analysis which identified a disual

phide - rich region of approximately 100 amino

acids at the C - terminus which encoded 2

epidermal growth factor (EGF) domains (12).

Subsequent work showed that immunization

with recombinant forms of the EGF domains

protected against challenge infection with blood

stage rodent parasites (17). Evidence that MsP-1 is

important in natural acquired immunity and can

consistently confer protective immunity is

equivocal.12.

3.8. Pre - erythrocytic Vaccine.

Pre - crythrocytic Vaccine strategies aim to

generate an antibody response that will

neutralize sporozoites and present them from

invading the hepatocyte and /or to elicit a cell

Page 22: HUMAN PARASITIC VACCINE

mediated immune response that will inhibit intra

hepatic parasites. This type of Vaccine would

prevent the advert of clinical disease. The most

advanced Pre-erythrocytic (liver stage) vaccine

candidate is derived from the circum sporozoites

Protein. (CSP). The major component of the

surface of the sporozoites Prototype Vaccine

were designed to induce antibody responses

against the repeat epitope of the circum

sporozoites protein and provided the first

evidence that humans could be Protected from

malaria infection with a sub unit Vaccine.(18,19).

The candidate Vaccine were poorly

immunogenic and a vaccine was developed

comprising a typhoid of the circum sporozoites

protein fused to hepatitis B surface antigen

expressed together with unused HBAg (20). From

these prototypes, RTS, S/A SO2 arose, which is

Page 23: HUMAN PARASITIC VACCINE

composed of both the CSP repeat antibody

targets as well as C-terminal non- repeat regions

that pre-targets for cell mediated immunity (21).

These were fused to the hepatitis B surface

antigen and expressed in yeast(21). Initial Phase -

I clinical trial of RTS, S formulated with Glaxo

Smith - Kline ASO2 adjuvant, an adjuvant

vehicle which consists of a lipid emulsion,

showed protection against malaria challenge in

six out of seven volunteers.(5).

More recently, the Vaccine has been formulated

with ASO1, A liposomal vehicle and this

vaccine induced higher levels of antibody and

Th1 response to the GS Protein (22). Vaccination

with these formulation Protects against infection

and reduces blood Parasites Extensive clinical

studies are under way, including one aimed at

combining RTS, with a blood stage antigen

Page 24: HUMAN PARASITIC VACCINE

MSP-1 (21). The MSP -1 is expressed on the

merozoite surface and is also a target for

protective immunity (12).

Reference

1. Hill. AVS. Vaccines against malaria. Phil Trans. R. Soc. 2011;

366 : 2806-14.

2. Patarroyo, ME. Et al., 1988. A synthetic Vaccine protects

human against challenge with a sexual blood stages of

plasmodium falciparum. malaria. Nature. 1988; 332:158-161.

3. Stoute.J.A., A Preliminary evaluation of a recombinant circum

sporozoite Protein Vaccine against Plasmodium falciparum

malaria. RTS. S. Malaria Vaccine Evaluation. Group. N. Engl.

J. med. 1997; 336 : 86 -91.

4. Nussen Zweig. R.S. Vanderberg . J. Most. H, orton. C,

Protective immunity Produced by the injection of X-irradiated

sporozoites of Plasmodium berghel, Nature 1967; 216; 160-

162.

5. WHO, 2008. Immunization, Vaccines and Biologicals.

6. M C Gregor. IA. The Passive transfer of human malarial

immunity. Am.J. Trop. med. Hyg 1964. 13. Suppl.237-9.

Page 25: HUMAN PARASITIC VACCINE

7. Wu. Y., Ellis, R.D, Shaffer. Detal Phase - I trial of malaria

transmission blocking Vaccine candidates Pfs25and PVS25

formulated with motanide I SA51, PLO some 2008;37:2636

8. Tree CK.M., Zacheri.S, Herrmann.S., et al., Functional analysis

of the leading malaria vaccine canditate AMA - 1 - reveals an

essential role for the cytophesmic domain in the invasion

process pLos pathog 2009:5(3): e 1000:322.

9. Collins.W.E., pye.D., crewther.P.E., et al Protective immunity

induced in squirel monkeys with recombinent apical membrane

antigen - 1 of plasmodium fragile Am.J.Trop.med.Hyg.1994.51:

711 - 9,

10. Stowers.Aw, kennedy, M.C., Keegan.BP., soul.A., Long.CA,

miller.L.H. vaccination of monkeys with recombinant

plasmodium falciparum apical membrane antigen - 1 confers

protection against blood - stage malaria Infect Immun.2002: 70:

6961-7.

11. Spring, MD., Cummings, J.F., ockenhouse CF. et al., phase 1/2

A Study of the malaria vaccine candidate apical membrane

antigen - 1 (Ama -1) administrated in adjuvant system A sol B

or Aso2A PLoS one 2009; 4(4): e 5254.

Page 26: HUMAN PARASITIC VACCINE

12. Holder AA. The carboxy - terminus of merozoite surface

protein.1. Structure, Specific antibodies and immunity to

malaria, porasitology .2009; 136: 1445-46.

13. Free man.RR. Holders.AA., charcteristics of the protective

response of BALB/C Mice immunized with a purified

Plasmodium yoelii. Schizont antigen clin.Exp. Immunol . 1983;

54; 609 - 16.

14. Maharajan. W.R., Daly. Tm, We idenzwp, Long. CA., Passive

immunization against murine malaria with an IgG3 monoclonal

antibody I. Immunel. 1984; 132; 3131 - 7.

15. Spencer valero.L.M., ogun SA, Fleck.S.L, et al., Passive

immunization with antibodies against three distinct epitopes on

plasmodium yoeliii; merozoite surface protein I. suppresses

parasitemia Infect immun. 1998; 66: 3925 - 30.

16. Holder. AA., Blackman.MJ, Burghaus.P.A chappel JA, Ling .

IT, Mccallum Deighton.N sha.SA., Malaria merozoite surface

protein (MSPI) - structure, processing and function. Inst.

oswaldo cruz. 1992; 87 (suppl.)3: 37 - 42.

17. Daly. T.M.., Long. CA, Humoral response to a carboxyl-

terminal region of the merozoite surface protein – I, plays a

predominant role in controlling blood stage infection in rodent

Page 27: HUMAN PARASITIC VACCINE

malaria. J. Immiunol. 1995; 155 – 236-43.

18. Hoffmann. SL, wistar.R. Jr. Ballon. WR. Et al Immunity to

malaria and naturally a cqwired antibodies to the circum

sporozoites protein of plasmodium falciparum. N. Engl. J.med.

1086: 315: 601-6.

19. Ballon. WR., Hoffman, Sl, Sherwood. JA et al., Safty and

efficacy of a recommend DNA plasmodium falciparum

sporozoites vaccine Lancet. 1987; 1 (8545) ; 1277-81.

20. Stoute. H.A., Sla ow,M., Heppener DG. et al., preliminary

evaluation of a recombinant circum sporozoites protein vaccine

against plarmodium falciparum malaria. RTS. S. Malaria

vaccine Evalzation Group. N. Engl. J.med. 1997; 336; 86-91

21. Ballon, WR. The development of the RTS. S, Malaria vaccine

candidate challenges and lesion. Parasite. Immunol.2009; 31;

492-500.

22. Garzon.N, Heppener DG, Coten. J., Development of RTS,

S/ASo2, a purified sub-unit based malaria vaccine candidate

formulated with a novel adjuvant Export Rev. Vaccines. 2003;

2231-8.

Page 28: HUMAN PARASITIC VACCINE

4. Leishmania vaccine

4.1Leishmaniasis

It is caused by several species of flagellated

protozoan parasites of the Leishmania genus, is

prevalent in Africa, Latin America, south and

central Asia, the Mediterranean basin and the

middle east,. There are an estimated 12 million

cases annually and disease can cause serious

disfigurement as well as death. Parasite are

usually transmitted from a wild animal

reservoirs (Small rodents, dogs) by biting sand

flies although transmission can occur from

infected humans the disease has Cutaneous,

mucocutaneous and visceral forms. About 1.5 –

2 million occurs annually and epidermis of

visceral leishmaniasis canaries with mortality

rates. The treatment relies on chemotherapy is

expensive and becoming compromised by

emerging during resistance. Recovery from

Page 29: HUMAN PARASITIC VACCINE

infection renders an individual resistant to

subsequent infection indicating that a successful

vaccine is feasible (14). As noted earlier

vaccination is possible with by controlled

infection using infectious materials from lesions.

(1).

4.2 Vaccine

The evidence is that most individuals who

were once infected with Leishmania are resistant

to clinical infections. When later exposed to it

provided the just infection for vaccine

development (2). The leishmaniasis are unique

among parasitic disease because a single vaccine

could successfully present and treat diseases and

the potential to protect against more than one

leishmania parasite species (3). There are

different types of vaccine development process

is going on. Here they discussed in details.

Page 30: HUMAN PARASITIC VACCINE

4.3 Killed vaccine

First generation vaccines consisting of

whole killed leishmania or fractions of the

parasite. Whole killed vaccines have been

experiment with both old and new world

leishmania. Mayrink and his colleague was

developed a killed vaccine composed of five

isolates of leishmania containing four different

species in 1970 (4). Convict and his group in

Venezuela introduced their autoclaved

Leishmania mexicana (L.mexicana + BCG for

immunotherapy (1).

Several prophylactic studies were done with in

conclusive results or low immunity protection

induced by the vaccine. For old leishmaniasis,

autoclaved L.major BCG (ALM + BCG) has

been extensively studied. Two doses of the

vaccine reduced the incidence by 43% in

Page 31: HUMAN PARASITIC VACCINE

Leishmania skin test converted volunteers in

sudan against visceral leishmaniasis involving

2306 volunteers (6). To enhance the.

Immunogenicity of the ALM + BCG vaccine

.ALM was absorbed to alum and the resulting

alum, ALM was mixed with BCG just prior to

injection. It appears to constitute a safe

vaccine and an appropriate candidate for

further development.

4.4. Live vaccine

Live attenuated leishmania vaccine is used

Uzbekistan Iran and Israel. Live virulent L.

major promastigotes are harvested from cultures

and used as vaccine. This is a mixture of live

virulent L.major mixed with killed parasite

registered in Uzbekistan. Adverse side effects

includes development of large persistent lesions,

psoriasis and immunosuperssion (7) Recently use

of L.donovani certain null mutants in mice

Page 32: HUMAN PARASITIC VACCINE

showed clearance of virulent challenge parasites

in 10 weeks after challenge, with significantly

reduced parasite burden in the spleen and no

parasites in the liver (8). The use of live virulent

organisms for vaccination was largely

discontinued and in the 1990 s the focus shifted

to attenuated organisms.

Attenuated parasite vaccine are lively to

mimic natural infection and hence induce an

appropriate immune response possible advantage

is that such immunization will also deliver many

more parasite antigens than the limited number

of possible with subunit or recombinant

antigens. (5)

4.5. Sub unit vaccine

A subunit vaccine utilizing the fucose

mannose ligand (FML) antigen has been shown

to be a potent immunogen in mice and rabbit and

Page 33: HUMAN PARASITIC VACCINE

a sensitive predictive and specific antigen in

serodiagnoses of human and canine Kala- azar (9)

,the main drawback of subunit vaccine is during

the preparation of vaccine more and more

natural pathogenic antigen subunit is necessary.

So the pathogen must be cultured otherwise,

Isolation and collection of parasitic subunit

antigens are very difficult.

4.6. Recombinant DNA vaccine

Second generation vaccine include all

defined vaccines ie) recombinant proteins, DNA

vaccines and combinations. A variety of

leishmania vaccine candidates consists of

recombinant proteins. More recent efforts have

aimed at increasing the immunogenicity of DNA

cloned vaccine including the use of genetic

adjuvant and plasmid based expression of viral

replicons. Some of the important recombinant

protein candidate vaccine include surface

Page 34: HUMAN PARASITIC VACCINE

expressed glycoprotein leishmanolysin (gp63) ,

Leishmania activated C. kinose (LACK)

parasitic surface antigen (PSA) Leishmania

derived recombinant polyprotein (Leish lllf) and

serine proteases (10)

Leish lllf is a single poly protein

composed of three molecules fused in tandem

the L. Major homologue of eukaryotic thiol-

specific antioxidant, TSA; the L. Major stress –

inducible protein – 1 LMSTII; and L.

brazilliensis and initiation factor, Leif. The

Leish- lllf product is the first defined vaccine for

leishmaniasis to go in to human clinical trials

and has completed phase -1 and 2 safety and

immunogenicity testing in normal healthy

human subjects (ll) various subunit recombinant

candidate vaccines have been tested in murine

model of Cutaneous Leishmaniasis (14) A lead

candidate is a surface – expressed glycoprotein

Page 35: HUMAN PARASITIC VACCINE

gp 63 or leishmanolysin. This is a zinc

metalloprotease (13) which is expressed on the

promastigotes surface and mediates

internalisation of promastigotes. Efficacy with

recombinant versions has been patchy. (12)

Gp 63 expressed in E.coli induced partial

protection in monkeys (15) However the native

protein purified from L. Major did protect mica

against challenge with either L.mexicana or L.

major and protection was evident when gp63

was expressed in BCG (16) and attenuated

Salmonella. (17) Studies on mice indicated that

host genetics influence the initiation of

protective immune responses to the vaccine (14)

DNA vaccination is considered on attractive

option for vaccine development. Since this

approach generally leads to the induction of the

Th 1responses. (18) A variety of DNA vaccination

is constructs have been test with variable out

Page 36: HUMAN PARASITIC VACCINE

comes. In addition, some protective immunity

can be stimulated by vaccination with sand fly

salivary proteins, sand flies being the insect

vector for the parasite with a 15 KD a protein

showing promise (19)

4.7. Ideal vaccine

Given the rapid progress in the fields of

parasite immunology and genetic engineering a

successful anti- Leishmania vaccine should be

achievable in the near future. Kedzierski et al (12)

regarding the definition of an ideal anti

leishmania vaccine. One of the requirements of

an ideal anti leishmania vaccine is to be effective

against more than one leishmania species in

order to protect individuals in areas. Where

Cutaneous and visceral leishmaniasis for

example, co-exist, recent evidence (21) suggests

that cross – Protection is possible.

Page 37: HUMAN PARASITIC VACCINE

It is of interest to note that summarizing a

large amount of experimental evidence, Rivier et

al (21) concluded that injection of attenuated

organisms achieved better protection than any

method involving recombinant gp63 as test

antigen delivered with a variety of adjuvant and

delivery systems. The prospect of using

attenuated leishmania vaccine is preference to

submit or recombinant approaches is likely to

come therefore although there would be

problems of are large scale production and

distribution in the field. Based on the past and

present experience on leishmania vaccine

studies, it appears that future experiments should

include appropriate adjuvant as components in

order to achieve effective vaccines against

human leishmaniasis (5)

Page 38: HUMAN PARASITIC VACCINE

Reference

1.Handman E. Leishmaniasis, current status of vaccine

development. Clin microbio. Rev 2001;4;229-43.

2.Reithinger. R, Dujardin. Jc. Louzir.H, pirme z.c., Alexander B,

Brooker S., Cutaneous leishmaniosis. Lancet. Infect.

Dis2007:7:581-96

3.Coler.R.N., Reed.s.,Second generation vaccines against

leishmeniasis Trend parasitol 2005,21:244-49

4.Genaro. O.de Toledo.v.p., Da costa.CA, Hermet mv, Afonsolc,

mayriak.w.,Vaccines for prophylaxis and immunotherapy. Brazis.

Clin. Dermatol. 1996,14:503-12.

5.Mutiso. J.M., Macharia. J.c, k;mn I

chagichis.J.m.,Riko;H.Gicheru.mm; Development of leishmania

vaccines; predicting the future from past and present experience J

Biomed. Res. 2013,27;85-102

6.Khalil.E.A , E.I. Hassan, A.M, Zuhystra.E.E mukhtar.

Mm.,Ghalib. HW,musa. B.et al. Autoclaved leishmania major

vaccine for prevention of visceral leishmaniasis;a randomisede

double blind, BIG-controlled trial in sudan. Lancet. 2000:356:1565-

9.

Page 39: HUMAN PARASITIC VACCINE

7.Khamesipour . A, Rafati.s, Davoudi.N, maboudi. F., Modabber. F.

Leishmaniasis vaccine canditate for development: A global

review.Indian. J.med. Res.2006:123:423-38.

8. Selvapandian. A, Day R., Gannavaram. S.,Lakkalvaouar. I.,

Duncan.R., Salotra, p. et al. Immunology to visceral leishmaniasis

using genetically defined live- attenuated parasite j. Trop med.

:2012:631 460

9..Palacnik-de sousa c.B., Dutra Hs, borofevic.R. Leishmania

donovani surface glyco-conjugate Gp 63 is the major immunogen

component of the fructose mannose ligand (f(ml (Act(9a. Trop.

1993:53:59-72.

10..kedzierski. I., leishmaniosis vaccine where are we today. J.Glob.

Infec. Dis 2010,2;177-85

11..Coler.Rn, Goto.y. Lisa. B, Ramen.v. Reed S.G., leish 111f, a

recombinant polyprokin vaccine that protects against visceral

leishmaniasis by elicitation of CD4+T-Cerlls.

Infect immunol. 2007:75:4648-54

12..Holder. AA.,Blackman M.J., Blackman. M.j., Burghaous.

P.Achappel. J.A., ling. IT., mcclum Deighton.N,shai. S.A. Malaria

merozoite surrface protein (MSPI). Structure, processing and

function mrm. Inst. Oswaldo ruz. 1992;87(suppl) 3;37-42

Page 40: HUMAN PARASITIC VACCINE

13.Chauthuri.G.,Handharm.Pan.A, changike Surface acid protionase

(gp63) of leishmania Mexicana.a metalloenzyme capable of

proteching liposome encaphlated preteins from phagolysohomal

degradation by macrophages. J. Bio. Chem.. 1989;264;7483-9

(14)Kedzicrrki. L. zhs.y. andman.E.,leishmania vaccines: progress

and problems parasitology. 2006;133. Suppl.s87-112.

(15) olobo. Jo, Anjili., co., Gichens MN et al vaccination of vervet

monkeys Cutaneous leishmaniasis using recombinant leishmania

major surface glycoprotein (gp63), vet. Paresite. 1995;60;199-212.

(16)Conell. N.D., Medina A costa E,Mcmaster wr., Bloom BR,

Rusell. DG, Effective immunization against cutaneous

leishmaniosis with recombinant bacile calmette Guerin expressing

bse leishmania surface protirase gp63 proc. Nat. acad Sci.Usa.

1993;90;11:473-7

(17)Yang.Dm, fair wcather.N. Button. Llama master wakah. I,

lpliewfy Oral salmonella typhimurium(Aroa) vaccine expressing a

major leishtimanial surface protein (gp63) pre-ferentially inducer

helper T cells and protective immunity against

leishmaniasis.J.Immanol.1990;145-2281-5

(18)Gurunathan.S.,klinman.Dm,Seder. RA

Page 41: HUMAN PARASITIC VACCINE

DNA vaccines immunology, application and

optimizationAnnRv. Immunol. 2000:18:927-4

19.valenzvela. J.H.,Belkaid.y, Gerfield. Mk,et al., Toward a

defined anti-leishmania vaccine targeting vector antigens.

Characterization of a protective salivary protein.J. EXP, med

2001;194;331-42.

20.Selvapandiyan.A, Dey R. Nylen. S.Duncan., sacks

D,Nakhasi.I, Intrcellular replication deficient. Leishmania

donovani induces long lasting protective immunity against

visceral leishmaniasis. J.Imman2009:183:1813-20

21.Rivier.D,Bovay.p.shah.R,Didisteim.s., masel.J .Vaccination

against leishmania major in a BA mouse model of infection. Role of

adjuvant and mechanism of protection parasite

immunol.1999:21:461-73

Page 42: HUMAN PARASITIC VACCINE

5. Amoebiasis vaccine

Entamoeba histolytica lives inside the larger

intestine of man. So it is an endoparasite. It

causes a disease called amoebiasis or amoebic

dysentery and also causes ulceration of colon.

Entamoeba is cosmopolitan in distribution.

There are many species of endamoeba. The

amoebic cyst is transmitted from one person to

another through the contaminated food and

water.

Amoebiasis vaccine development work is under

experimental level. Researchers identified many

antigens as serine rich binding protein N-

Acetylgalactosamine, 29 KD a cysteine –rich

protein (peroxiredoxin) lipo phaspho glycon and

oral \intranasal administration of lectins (2).

Page 43: HUMAN PARASITIC VACCINE

Most of the antigen efficacy studied in animal

model. Still they are not yet developed best

vaccine.(1)

Reference

1.Abhijit cha udhury (2014)Human parasite vaccines an overview –

Review. J.clin. Bio. Med. Sci. 4(1) ; 216-21.

2.Parija. Sc. Progress in the research on diagnosis and vaccines

in amoebiasis Trop. Parasitol 2011 : 1;48

Page 44: HUMAN PARASITIC VACCINE

6. Cestode Vaccine

6.1. Infection

Man is affected by a number of different

Cestode parasites including the Taenia solium

and T.saginata. The important worldwide

spreading Cestode are Echinococcus granulosus

and E. multiclocularis, T.Solium and T.saginata.

But incidence is considerably higher in

developing countries. {9)

6.2 Disease

The tape worm larval cyst, containing the

infective on chosphere stage is ingested with

poorly cooked infected meat. The adult may live

as long as 25 years and pass gravid proglottids,

containing eggs with the faeces. Adults establish

the intestines. The egg can persist on gestation

for several days and are consumed by cattle or

pigs in which they hatch and form cysticerci.

Page 45: HUMAN PARASITIC VACCINE

The eggs can also infect humans and

cause cysticercosis. Where cysts can establish in

the lungs, liver, eyes and brain resulting in

blindness and neurological disorders. Gastro

intestinal symptoms arise from the presence of

adult tape worm while cysticercosis symptoms

are due to the host inflammatory immune

responses to the parasite and abnormal

mechanical presence of the cyst (9)

6.3. Vaccine

Recombinant antigen vaccines targeting

on chosphere proteins (1) It has been developed

against infection with a number of Taenia

species including Taenia ovis in sheep (2,3).

Taenia sagninata in cattle (4) and Taenia solium

in pigs.(5, 6). These vaccines are based on groups

on chosphere proteins designated 16 k, 18 k and

45 w groups, after their original description in T.

ovis (2) Protection induced by vaccination

Page 46: HUMAN PARASITIC VACCINE

approaches 100%.

The 45w vaccine against Taenia ovis

infection in sheep (2) was the first highly

effective recombinant vaccine against a parasitic

infection. A similar approach was using

immuno blotting and invitro. On chosphere

killing identified a putative protective molecule

(EG 95) in Echinococcus granuloses (7). Despite

these impressive successes, these vaccines have

yet to be applied due to commercial, economic

and socio- economic reasons (1). However it

cannot be over emphasized that this work and

the work immediately below on ticks, is ground

breaking providing proof of concept for the

development of subunit vaccines against

complex metazoan parasites.

Independent vaccine trials for Taenia

solium carried out in pigs with the TSOL 18

antigen in (Transmission Blocking veterinary

Page 47: HUMAN PARASITIC VACCINE

vaccine) Mexico, Peru Honduras and Cameroon

have all achieved 99-100% protection. Results

were published of the first field trial of the

TSOL 18 vaccine, which was carried out in

north Cameroon. The vaccine completely

eliminated the transmission of T.solium by pigs

involved in the trial (8).

Reference

1. Light taulers. MW., Cestode vaccine, origins current status and

future prospects parasitology 2006; 133 : S 27-42.

2. Johnson K.S, Harrison G.B, Light oulers MV. Et al:

Vaccination against ovine cystice-cosis using a defined

recombinant antigen Nature, 1989 : 338: 585-7

3. Harrison. G.B. ; Health DD., Dempster.R. p et al., Identification

and CDNA cloning of two novel low molecular weight host

protective antigens from Taenia olis on cospheres. In J.Parasitol

1996 : 26: 195-204.

4. Light owlers. MW. Ralfe. R. Gauti G.G. Taenia sagninata.

Vaccination against cyliticercoses in cattle with recombinant on

Page 48: HUMAN PARASITIC VACCINE

cosphere antigens.Exp- parasital. 1996: 84 : 330-8.

5. Flisher A., Gakuti., C.G. Zali. A. et al., Induction of protection

against procine cysticercosis by vaccination with recombinant

on cosphere antigens Infect Immun. 2004 : 72: 5292 – 7.

6. Gonzelez. A.E., Gauc. C.G., Barbar.D. et al., Vaccination of

pigs to control human neurocysticercosis AM.J. trop.med. Hyg.

2005 : 72 : 837- 9.

7. Heath. DD., Lawrence. S.B. (1996) Antigenic polypeptides of

Echinococcus granulosus on chosphere and definition of

protective molecules, para. Immunol; 1996 ; 18 : 347-57.

8. AAskna. E, Kyngson.CT., GDauti. GG, Geerts.S Dorny.P.,

Daken.R.D.., et al., Elimination of Taenia solium transmission

to pigs in a field trial of the TSOL 18 vaccine in Cameroon. Int.

J. Parasitel.2010 ; 40 ; 515-519.

9. .Knox.D. (2010) Parasite vaccines : Recent progress in the

problems associatd with their development. The open Infec.

Disea. Jawn. 2010, 4 : 63- 73.

Page 49: HUMAN PARASITIC VACCINE

7. Schistosomiasis vaccine

7.1. Disease

Schistosomiasis affects an estimated 300

million people worldwide. The causative agents

are snail- transmitted, water born parasitic

helminthus. The schistosomiasis have complex

life cycle using two separate hosts to complete

their development. Severe consequence of

infection include bladder cancer or renal failure (

schistosoma haematobium) and liver fibrosis and

portal hypertension (S. mensoni). Again control

is largely dependent on mass chemotherapy but

resistance is a concern. Disease is endemic in

over 70 countries causing tens of thousands of

deaths.

S. haematobium is found in the Middle East

Africa and Southern Europe and affect the

bladder causing urinary schistosomiasis. S.

mansoni is present in most African countries

Page 50: HUMAN PARASITIC VACCINE

north of the equator as well as parts of central

and South America. It causes intestinal

schistosomiasis S.jalonicum is endemic in china

where bovines are the main reservoir as well as

in Indonesia and the Philippines (with dogs and

pigs reservoir) S. bovis is an important parasite

affecting cattle as well as sheep and goats a is

prevalent in south East Asia (1).

7.2 Vaccine target

The most important vaccine target of the

schistosome is the tegument. The tegument is

through to be involved in several key

physiologic processes; parasitic nutrition,

osmoregulation and the evasion of host

immunity. Tetra spanins found in outer tegument

play an important role in maintaining the

integrity of the tegument. Schistosoma mansoni;

TSP-2 has been selected by the H H V I for

development as a human vaccine antigen, (1, 2)

Page 51: HUMAN PARASITIC VACCINE

many proteins of the parasite may act as good

Immunogens. If identify the appropriate

Immunogens, the vaccine development process

is very easy.

7.3 Irradiated vaccine

Irradiated larval vaccine were a focus of

study in the 1970s with several early studies

being conducted in sheep challenged with S.matt

heel and S. bovis. Vaccination of mice with

radiation alternated cercariae reduced worm

burdens by 90% compared to controls and this

vaccine model has been exploited to identify the

antigens responsible for protection. The

approaches being used for vaccine development

have been extensively reviewed. (3)

7.4 Protein vaccine

Several proteins expressed on the surface of

the adult stage with particular current interest in

transmembrane proteins. Tetra spanins are

Page 52: HUMAN PARASITIC VACCINE

proteins which is found n the surface of karyotic

cells, including B and T cells and at least three

of these show promise as vaccine (11). SM 23 is

one of the independently tested WHO\TDR

vaccine candidates and is a testasparin expressed

in the tegument of S. Mansoni SM23 is not

effective when given as a recombinant protein

with alum but is effect when derived as a DNA

vaccine (4) There has been much focus on 26

and28 KD glutathoine –S-transferases (GSTS) .

The GSTS being though to have a detoxification

role for the parasite.

SM 28 –GST is expressed in the sub-

tegumental tissues of the parasite and

vaccination studies in rats and hamsters with the

S. hoematobium equivalent (5) indicated its

significant protective potential. Trials in

primates demonstrated and antifecundity effect

(6). Clinical testing of sh – 28 GST in people

Page 53: HUMAN PARASITIC VACCINE

should that the vaccine was immunogenic and

antibody inhibited the enzymatic activity of the

recombinant protein (7). A concerted effort,

based in Egypt and supported by USA, ID the

schistosomiasis vaccine development program

(SUDP), is examining two S.mansoni antigens,

parasmyosin and a synthetic peptide construct

containing multiple antigen epitopes (MAP)

from the triose phosphate isomerase.(1).

Another vaccine candidate is a 14 KD

fatty acid binding S.mansoni protein (SM 14)

which provided 67% protection against

challenge with S.Mansoni cercarioe (9) SM 14 is

a cytosolic protein expressed in a basal lamella

of the tegument and the get epithelium (8). It is an

attractive vaccine candidate because fatly acids

binding proteins play a key role in Schistosoma

nutrient acquction. Mc manus and laukas (3)

commented that an apparent efficacy ceiling of

Page 54: HUMAN PARASITIC VACCINE

40 to 50% was road block to success. This is

compounded by difficulties in obtaining good

expression levels and in scaling up production

according to good laboratory practice. (1)

7.5 Recombinant vaccine

The Institute pasteur has taken a

recombinant 28 KDa Glutathioe S – transferase

(GST) cloned from S.haematobium through both

phase – 1 and 2.clinical testing in Europe and

West Africa(Senegal and Niger).Sh 28 GST

(Bilhvax) is a recombinant protein formulated

with an aluminium hydroxide adjuvant. Bilhvax

appears to be immunogenic and well to be rated

healthy adults from non- endemic (France) and

S. heamatabium endemic area in Africa. The Sm

– Tsp -2 recombinant Schistosomiasis vaccine

would be intended primary for school aged

children living in the S.mansoni endemic regions

of sub – Saharan Africa and Brazil. The vaccine

Page 55: HUMAN PARASITIC VACCINE

ideally would prevent the real question of

schistosomiasis in the blood streams following

initial treatment with praziquantel (vaccine

linked chemotherapy) – (10).

Recently two new S. Mansoni

tetrasparin (Sm TSP -1 and Sm TSP – 2) were

identified. Recombinant TSP – 2.But not Tsp-

1,is strongly recognized by IgG and Ig G3(but

not IgE) from naturally resistant individually. It

is not recognized by IgG from chronically

infected or unexposed individuals. Both proteins

included good levels of protection in immunized

mice given an S.manloni challenge.

The authors noted that Tsp-2 in particular

provided protection in excels of etc 40% bench

mark set by the words health organization for

progression of Schistosoma vaccine antigens

into clinical trials. other antigens include a 97kd

Page 56: HUMAN PARASITIC VACCINE

a parasmyosin (12) and a 28kd a triose phosphate

isomerace (Tpl:eg) (9).The latter being ubiquitous

in each stage give that the S.mansoni and

S.japnicom sequences are very similar trails of

vaccine efficacy have focused on the latter with

the aim of blocking parasite transmission.(9).

7.6 Nucleic Acid Vaccine

Pigs vaccinated with a TpI DNA vaccine alone

showed reductions in adult worm burdens of

48% with female parasite being more

susceptible(13).Vaccination reduced liver egg

numbers and reduced granulomasize. Moreover,

protective immunity was stimulated in water

buffalo by vaccination with a DNA plasmid

comprising a fusion of S.jalonicum TpI,a heat

shock protein and IL-12(14).

Mc manus and Loukes (3) noted that the data

were encouraging but that extensive trails were

skill need to determine. If a vaccine of this

Page 57: HUMAN PARASITIC VACCINE

nature would reduce transmission as evidenced

by reduced adult worm burdens, egg outputs and

reductions in hepatic eggs. More Effective

antigens may be selected by mining the publicly

available S.mansoni and S.jalonicum

transcryptomes in combination with DNA

micro-array profiling, proteomics, glycomics

and immunomics.

The development of RNA interference

to disrupt gene function is a big step forwarded

and will enhance ability to determine the

functions of schistosome gens/proteins and it is

hoped the techniques will help define those

essential for survival and reproduction. Silencing

the expression of numerous S.mansoni genes has

resulted in phenotypic changes, highlighting

their importance as targets for vaccines and new

drugs.(15).

Page 58: HUMAN PARASITIC VACCINE

Reference

1.knox.Dp.parasite vaccines: :Recent progress in and problems

associated with their development. The open infectious diseases

journal.2010:4:63-73.

2.Fonesca.CT.,carvaloh GBE,Avles cc,demelo.TT.Schisistosoma

tagment proteins in vaccine and diagnosis development: An

update.J.parasitol.Res.2012;2012:541268.

3.McManus.Dp,Loukas.A.current status of vaccine for

schisitomiasis.clin.microbial.Rev.2008;21:225-42.

4.Da’dara.AA,Skelly.PJ.,Wang.M.M,Harn.DA.Immunization with

plasmid DNA encoding the integral remembrance

protein,Sm23,elicite a protective immune response against

schistasome infection in mice vaccine 2001 12.20:359-69.

5.Balloul.Jm,Grzych.Jm,piercc RJ,Capron.,A.A. ,purified 28,000

Dalton protein from Schistosoma mansoni adult worms protects

rats and mice against experimental

schistimiasis.J.Immunal.1987:138:3448-53

6 Boulange r.D.,Reid.G.D,Sturrock .Rf.et al.,Immunization of mice

and balloons with the recombinant Sm 28 GST affects both worm

viability and fecundity after experimental infection with

Page 59: HUMAN PARASITIC VACCINE

Schistosoma mansoni parasite Immunol.1991:13:473-90.

7 Capron.A,Riveau.G,Capron.m.,TrotteinF.Schistosomes.The road

from host parasite interactions to vaccines in clinical trails.Trends

parasite .2005:21;143-09

8 Brito.CF.oleria.Gc.,olivera sc.et.al.,sm14.gene expression in

different stages of the Schistosma mansoni life cycle and

immunolocvalization of the sm14 protein within the adult

worm.Braz.J.med.Biol,Res.2002:35:377-87.

9Harn.DA,Gu.w.olignol DMilsyana.M .Gebre

micheal,A.,Richter.D.A.protective monoclonal antibody

specifically recognized and alters the catalytic activity of

Schistosome triose phosphate isomerase.J.Immunal.1992;148:562-

7.

10.Rofatto.H.K,araujo-montoya Bo,miyasato.P.A,leavanno-

garcia.J.Rodrigue Z.D,Nakano .E etal.,Immunization with

tegurement nucleotides associated with a subcurative praziquantel

treatment reduces worm burden following schistosoma mansoni

challenge,Peer.J.2013.1:e58.

11.Tranm.H,pearson.m.S,Bethony.Jm.et al.,Tetraspanins on the

surface of schistosoma mansoni are protective antigens against

schistosomiasis.Nat.med.2006:12:835.40.

Page 60: HUMAN PARASITIC VACCINE

12.Pearice E J,James .Sl.Hieny.S.Lenar.De,Sher.A.Induction of

protective immunity against schistosoma mansoni by vaccination

with Schistosoma parasmyosin(sm97),a non surface parasite antigen

.proc.Nat ..Acad.Sci.USA.1988;85:5678-82.

13.Zhu.y.Si.J,Harn.DA .et al. ,Schistosoma japonicum trails

phosphate isomerase phasmid DNA vaccine protects pigs against

challenge infection.parasitology.2006;132:67-71.

14.Yu XLHe.YK,Xiong.T.et al.,Protective effects of co-

immunization with sjcTpl-Hsp70 and interleukin-12 DNA vaccines

against Schistosoma japonicum challenge infection inwater

buffalo.Z.hongguo.Ji Sheng Chong Xue Yu Ji Sheng Chong Bing

Zhi 2006,24:433-6.

15.Brindey.PJ,Pearce.EJ,Genetic manipulation of schistosomes

Int.J.Parasitol..2007,37:465-73.

Page 61: HUMAN PARASITIC VACCINE

8. Hook worm vaccine

8.1 Infection

Human hookworm infection is caused by the

nematode parasites Necator Americanus and

Ancylostoma duedenale infections is leading

cause of amoeba and protein malnutrition,

afflicting an estimated 740 million people in the

developing nations of the tropics. The largest

numbers of cases occur in improved rural areas

of Sub-Saharan Africa, Latin America, South

East Asia and China. N.americanus is the most

common book worm worldwide, while

A.duodenste is more geographically restricted.

Hook worm infection arises by skin contact with

infective third stage larvae (L3).

Adult hook worms are blood feeders and

attach to the mucosa of the small intestine and

cause intestinal blood loss. Typically symptoms

Page 62: HUMAN PARASITIC VACCINE

can arise from infections with as little as 50 to

150 of adult parasites.(1).

Infection induces as iron – deficiency

an anaemia which is indirect correlation with

the number of parasites (as measured by

quantitative egg counts).In children, chronic

hook worm infection impairs physical and

intellectual development ,reduces school

performance and attendance and adversely

affects future productivity and wage-earning

potential(1).

8.2 Disease Burden

The major human soil transmitted helmithes

(STH),Ascaris lumbricoides, Trichuris trichiura

and the hook worms occur in 1221 million,795

million and 740 million people, respectively(2)

and are among the most common pathogen of

human in developing countries. There is

considerable epidemiological overlap among

Page 63: HUMAN PARASITIC VACCINE

the STH infections and disability adjusted life

year (DALY) estimate indicate that the disease

burden from human STH infection is nearly

equivalent to better known Conditions such as

malaria and tuberculosis (3).They have broad

ranging effects on pregnancy, childhood

growth, nutrition and cognitive and intellectual

development.(3).

8.3. Control Measures

Control is achieved by population wide

treatment, with benzimidozole based anti

helmetics albendazole and the 2001 world

health assembly advocated the antihelmenthic

treatment of 75% of all at risk school aged

children.However,drug treatment not present

reinfection and the frequent use of drugs

required to maintain control would be likely to

lead to drug resistance. Therefore, there is a

drive to develop a safe and cost effective

Page 64: HUMAN PARASITIC VACCINE

vaccine.

8.4 Vaccine

The selection of the leading hook worm vaccine

candidate ,the Ancylostoma secreted protein

,ASP-2 was based on three major lines of

evidence (4).First human anti ASP-2 antibody

responses are associated with considerably

reduced risk of acquiring heavy hook worm

infection.(5).Second, when recombinant ASP-

2(expressed either in yeast or baculo virus) was

used as vaccine in animal models. it resulted in

reduction in host hookworm burden, hookworm

and fecundity(5-7).Third, anti ASP-2 antibody

inhabits larval penetration through host tissue

invitro.(5). suggesting that the vaccine elicits an

antibody response that interferes with passage

of larval through host tissue of the L3 ES

products tested,ASP-2 provided the greatest

levels of protection in canines(5) and hamster

Page 65: HUMAN PARASITIC VACCINE

models of hookworm infection and was

therefore given the highest priority. The most

consistent and emphatic group of protective

antigens to date identified form blood feeding

parasites are those expressed on the surface of

the gut and presumed to function in blood meal

digestion. Gut membrane proteins have been

effectively employed as vaccine antigens

against cattle ticks(8) and H.contortus (9,10) and

are offering promise as vaccine components

against hookworms .Haemoglobin digestion is

through to be facilitated by a multi-enzyme

cascade in blood feeding-parasites(11,12) and

some of the enzyme involved in this process

have been indentified in hookworms and

Haemonchus with many commonalities evident

between the two and also when compared to the

fluke and malaria parasites.

Page 66: HUMAN PARASITIC VACCINE

Vaccination of dogs with recombinant

AC-APR-1 significantly reduced hook worm

burdens and faecal egg counts and these dogs

were protected against blood loss and did not

develop anaemia (3) Like the AC-CP2 vaccine,

IgG from animals vaccinated with APR-1-

inhibited enzyme activity invitro and antibody

bound insitu to the intestines of worms

recovered from vaccinated dogs, implying that

the vaccine interfere with the parasite’s ability

to digest blood. This was the first report of

recombinant vaccine from a haematophagous

parasite that significantly reduced both parasite

load and blood loss, supporting the

development of APR-1 as a second arm of the

HHV.

A catalytically inactive form of homologue of

APR-1 found in Necator americanus also

Page 67: HUMAN PARASITIC VACCINE

induced protection against. A.caninum in dogs

.Resident in high –transmissions areas for

N.americanus had high circulating IgG to the

inactive Na-APR mutant, indicating that

natural boosting may occur in exposed

humans (13).

8.5 Hurdles In vaccine Development

A number of hurdles complicate the

development of an effective vaccine for hook

worm and for that matter other helmthics.

Some of these include

*The difficulty of maintaining human hook

worms in animal models and the cost of

maintaining hook worm in the laboratory-

canine model.

*The absence of a laboratory animal that is

permissive to human hook worms and can

Page 68: HUMAN PARASITIC VACCINE

accurately reproduce human

disease(anaemia).

*Paucity of invitro functional tests to

determine the efficacy of the immune

response induced by an experimental hook

worm vaccine.

*The lack of protective immune response in

humans and the consequent absence of

correlates of P.rotection that can guide the

discovery of vaccine antigens and be used to

assess their effectiveness in pre clinical and

clinical trials.

*No model of an effective immune response

in humans to determine the biological

consequences of the vaccine in humans (14).

Page 69: HUMAN PARASITIC VACCINE

8.6 History of vaccine Development

In 1964, Miller (15) showed that ancyloshoma

caninum larvae could be alternated

using40,000 rontgens of X.Ray. Industrial

manufacture and US licensing of the

1st,hookworm vaccine commenced in 1970s

,which consisted of gamma-irradiated

infective A.Caninum 1.3 vaccine for canine

(16).This vaccine was discontinued in 1975due

to commercial failure. Although this vaccine

failed commercially it provided compelling

evidence that human hook worm vaccine is a

possibility. The human hookworm vaccine

initiative (HHV1) is the only group currently

working in vaccines targeting this parasite.

Ancylostoma secreted protein-2 of

N.americanus (Na-ASP-2) is a 21KD a

protein, there is secreted by infective

hookworm larvae up to entry, into the host

Page 70: HUMAN PARASITIC VACCINE

and Na-ASp-2 was chosen as a lead

hookworm vaccine candidate (17).In phase –I

study in hookworm native adult living in the

USA,Na-ASP-2 adjuvant with alhydrogel

was well tolerated and immunogenic(18).

However, a phase I safety and

immunogenicity trial of this vaccine is

healthy adult form a hookworm endemic area

in rural Brazil. It had to be halted when 3

participants developed immediate,

generalized urticarial reactions. The urticarial

reactions were associated when elevated

levels of IgE antibodies specific for Na-ASP-

2,which were present before immunization

most likely due to previous hook worm

infection.(19).In November ,2012 Sabin

vaccine Institute an start of the part-II of its

phase –I clinical trial of the Necater

americanus –glutathione S-

Page 71: HUMAN PARASITIC VACCINE

transferase_1(NaGST-1) vaccine candidate

Part-II of the trial commenced in American

has brazil following successful vaccination in

part-I of the study ,which began in

belohorizonte, Brazil in late

2011.Ulternately,Na-GST-1 and Necater

americanus aspartic protease-1(Na,APR-1)

would be used together a bivalent vaccine

and the aim of the vaccine will be reduce

moderate to heavy infections in the host.(20).

Reference

1 Evans .A.C.,Stephenson L.S.,Not by drugs alone the fight against

parastic helmethics world health forum.1995;16(3):258-61.

2.Desilva.NR.,brooker.s.,Hote

Z.PJ,Montresor.A,Engels.D,Savioli.L.Soil-transmitted helmintin

infections updating the global picture.Trends parasital.2003;19:547-

51.

3.Loukas.A.,Bathony.Jm,Mendez.S.et al. ,Vaccination with

recombinant aspartic hemoglobinase reduces parasites load and

Page 72: HUMAN PARASITIC VACCINE

blood loss after hook worm infection in dogs PLos med 2005:2e295.

4.Bethony.Ju.,Loukas.A,hotez.PJ,knox.D.P.Vaccines against blood

feeding Nematodes of human and lives stock parasitology

2006:133(suppl):S.63-79.

5.bethony.j.,Loukas,A,Smout.M.et al.,Antibodies against a secreted

protein from hookworm larvea reduce the intensity of hookworm

larvea reduce the intensity of hookworm infections in humans and

vaccinated laborartory animals.FASEB.J.2005;19:1743-5.

6.Sen.L.Ghosh.K,Binz et al.Hookworm burden reductions in

BALB/emice vaccinated with recombinant Acylostoma secreted

protein(ASPs)from Ancylostoma duodenate

,americanus.vaccine.2000;18:1096-102.

7.Goud.G.N.,Zhan.B,Gosh.K.et al.,Cloning,yeart expression

isolation and vaccine testing of recombinanr Ancylostoma secreted

protein ASp-1 and ASP-2 from Ancylostoma

ceylanicum.J.Infect.Dis.2004;189;919-29.

8.Willadsen.P.,vaccination against ecto parasites

parasitology.2006;133:59-525.

9.Knox.DP.,Smith.WD.,vaccination against gastro intestinal

nematode parastites of ruminants using gut-expressed antigens

Page 73: HUMAN PARASITIC VACCINE

vet.parasital.2001;21-32.

10.Loukas.A.,Bethony.J.M.,mendez.S.et al.vaccination with

recombination aspartic heamoglobinase reduces parasite load and

blood loss after hookworm infection in dogs PLos.med.2005;2:e295.

11.Goldbery.D.E.,Haemoglobin degradation

curr.Top.micro.bio.Immunal.2005;295:275-291

12.Williamson.AL,Brundley.PJ.Knox.DP.Hotez.PJ.Loukas.a.Digest

ive proteases of blood feeding hematodes.Trends parasital

2003;19:417-23.

13 Pearson.m.s.,Bethony .J.m,pickering D.A.etal.,An enzymatically

inactivated hemoglobinate from Necator americanus induces

neutralizing antibodies against multiple hook worm species and

protects dogs against heterologous hook worm infection

FASEB.J.2009:23:3007-19.

14 Abhijit chaudhury (2014).Review Human parastic vaccines an

overview J.clin.Biomed.sci.2014;4(1):216-21.

15 Miller.J.A.,effect of X-irradiation upon the infective larvae of

Ancylostoma caninum and the immunogenic effect in dogs of a

single infection with 40Kr-Irradiated larvea.J.Parasital 1964;50:735-

42.

Page 74: HUMAN PARASITIC VACCINE

16 Miller.T.A,Industrial development and field use of the

canine hook worm vaccine.Adv.parasital.1978:16333-42.

17Loukas.A.,bethony.J.Brooker.S.,Hotez.P.hookworm

vaccines:past,present and future.Lancet,Infect.Dis-2006;6:733-41.

18 Bethony.J.,Simon.G.,Diemert.DJ.,Paranti.D.,Des

rosiers.D,schuck.s.,et al.,Randomized,place controlled double-blind

trial of the Na-ASP-2 hook worm vaccine in exposed adults.Vaccine

.2008;26:2608-17.

19. Diemert.DJ.,pinto.AG.,friere.J.Jariwala.A.,santiago.H,h

amliton.RG.et al.,Generalized urticaria induced by the Na-ASP-2

hook worm vaccine: implications for the development of vaccines

against helmiths.J.Allergy.clin.Immunal.2012:130:169-76.

20. Hotez.PJ.,Diemert

D,Bacon.K.M.,beaumier.C.,Bethony.Jm,Bottazzi.ME et al.,the

human hook worm vaccine vaccine 2013;31.suppl.2:227-32.

Page 75: HUMAN PARASITIC VACCINE

9. Filarial Vaccine

9.1 Infection

Filarial worm (wuchereria benorefti) is a

digenic parasite. Man is the primary host and the

cluex mosquito is the secondary host. It lives in

the lymph nodes and lymph vessels of man. The

sexes are separate. The male is smaller than the

female. The male has a curved posterior end and

a pair of spicules. The male and female are

found coiled together it is viviparous, giving

birth of larvae. The larvae are called

microfilaria. It has a stylet. The microfilaria

passes from the lymph vessels into the blood

vessels.

The larva comes to the peripheral blood vessels

in the night. When the culex mosquito bites a

man containing the larva, which enters the gut of

Page 76: HUMAN PARASITIC VACCINE

mosquito. In the gut ,larva moults twice and

becomes another larva called filariform larva.

The filariform larva penetrates the gut and

migrates to the muscles of the mosquito. Then it

reaches the mouth parts. When the mosquito

bites another man, the larva enters the blood of

the man. It causes the obstruction of the free

flow of lymph. As a result, the lymph glands and

lymph vessels of the affected parts are enlarged.

9.2 Vaccine

To get a vaccine to eradicate the filarial disease

is still for from reach chemotherapy is the only

option left for patients (2). Still there is no vaccine

for human use. Numerous vaccine antigens are

being defined for a wide range of helminthus

parasite species, at greater understanding is

needed to develop vaccine. The vaccine

development process move to several scientific

Page 77: HUMAN PARASITIC VACCINE

generations such as live attenuated

organism.(Using irradiation, biochemical

fractions, recombinant protein, etc.) The

radiation –attenuated larval vaccines

successfully induced protective immunity in the

dogs and cattle. Although they were not widely

adopted(4). The use of extretory-secretory(ES-

products)were able to generate significant

immunity in animal model of Trichinella spirals

infection(5). Ansarietal.,(1) reported that highly

expressed stage specific product ALT

protein(abundant larvae transcripts) was

producing good results. It is not found in mature

adult stage. The animals immunized with ALT-1

proteins shows up to 76% reduction in parasite

survival stage specific proteins are the therefore

strong candidates for future vaccine against

human Filariasis.(3).

Page 78: HUMAN PARASITIC VACCINE

Refernce

1 Ansari.va,Sp.Singh,mbmjulbid,Kuldeep singh and muhamed Arif

(2016). Development of filarial vaccine by targeting stage specific

proteins.J.che.phor.res.2016;8(2);269-274.

2 Sharma.B.(2014).lymphatic filariasis and chemo therepetic

targets. Bietech Anal .Bioche.3:1-147.

3 Knox.DP.,Redmand.DL,Newlands.GF,et al ,The nature and

prospects for gut membrane proteins as vaccine candidates for

Haemonchus contortus and other ruminant Trichus trongyloids. .

Int.J.Parasitol.2003:33:1129-37.

4 Miller .T.A.,Vaccination against the canine hook worm

diseases.Adv.Parasital 1971.153-83.

5. Camphell C..H.The antigenic role of the excretions and secretions

of Trichinella spirallis in the production of immunity in

mice,J.parasital.1955-41:483-91.

Page 79: HUMAN PARASITIC VACCINE

10. Prevention and Control Methods

The following strategies are helpful to minimize

or completely eradicate parasitic infections.

*The complex life cycle of parasite should be

breakdown.

*Vector population should be controlled or

destroyed by suitable vector control programme.

*Stage specific vaccine will be developed and

mixed at appropriate level then used mixer

vaccine.

*First of all, mode of transmission should be

identified clearly then blocks will be developed

and stop the transmission process.

Page 80: HUMAN PARASITIC VACCINE

*Drinking water should be well boiled.

*Food stuffs and water should be protected from

house files and other insects.

*Destroy the mosquito and its larvae by using

biological control methods.

*Avoid active movements; during mosquito’s

peak time .Mosquito proof house should be

constructed. Use always mosquito nets.

*Applying the repellent on the surface of the

body. Spraying bio-pesticides in and around the

houses periodically.

*Integrated management of control is highly

useful to reduce the parasitic infection.

Page 81: HUMAN PARASITIC VACCINE

*Before eating, the hands should be washed

clearly. Finger nails should be clean and closely

cut.

*Fruits and vegetables should be thoroughly

washed.

*Should wearing sleeves and trousers.

*Use good food items which provide more

strength to our immune system. These food

items must be given particularly in endemic

areas and it should be given to children.

*Give immunotherapy to people who live in

dangerous area. It will reduce the disease burden

in future.

*Conduct awareness programme in schools and

Page 82: HUMAN PARASITIC VACCINE

other public area, it will be useful to people.

About the Author

Dr.M.Muruganandam is an

Editor of African journal of Biotechnology

and International journal of Medicine and

Biomedical Research. He is also Reviewer and

Editorial board member in Various National

and International journals. He published more

than hundred publication including ten books.