Development of malaria vaccines that block transmission of parasites by mosquito vectors
Hajime Hisaeda and Koji Yasutomo

Department of Immunology and Parasitology, The University of Tokushima School of Medicine, Tokushima , Japan

Abstract: Malaria is still one of the infectious diseases urgently requiring control and causes socioeconomic burdens on people residing in developing countries. Malaria vaccines are expected to control the disease. However, there is no effective vaccine available despite the intense efforts of malaria scientists. One strategy for a malaria vaccine is to prevent parasite spread by means of interfering with parasite development in mosquito vectors, which is the so-called transmission-blocking vaccine (TBV). We will here review the current progress of TBV. J. Med. Invest. 49:118-123, 2002

Keywords:malaria vaccine, transmission-blocking vaccine, P. falciparum, P. vivax

INTRODUCTION
Malaria is a disease caused by infection with protozoan parasite genus Plasmodium, of which four species infect humans. It produces up to 500 million new infections and 2 million deaths every year (1). Chemotherapy and vector control have been insufficient to control the disease because of the emergence and spread of parasites and their vectors resistant to these old chemical based control methods (2, 3). Thus, malaria vaccines are urgently needed to control malaria especially caused by two major species, P. falciparum and P. vivax. The malaria parasite has a complicated life cycle (Fig. 1), which makes difficult to develop a universal, effective and long lasting vaccine. However, several studies suggested that vaccines against each parasite stage are feasible as part of a control strategy.

LIFE CYCLE OF MALARIA PARASITE AND VACCINE TARGETS
Infection is initiated by inoculation of sporozoites, an infectious form of the parasite, through mosquito bites. Once inoculated, sporozoites spend less than 30 minutes in the blood before entering heptocytes or being eradicated. In heptocytes, a single sporozoite develops into 30,000-40,000 merozoites, each of which, when released into the bloodstream, continues its life cycle in red blood cells. Specific antibodies to sporozoites can inhibit entry to hepatocytes (4). During hepatic development, a variety of liver-stage-specific antigens are synthesized by the parasite and are presented in context with MHC class I molecules and recognized by CD8+ T cells (5). Vaccines against parasites before merozoite release called liver-stage vaccine are expected to induce antibodies that inhibit invasion of hepatocytes and cytotoxic T cells that destroy infected liver cells. Successful vaccines can reduce the chance of a person becoming sick because all symptoms appear during the asexual growth stage in the blood cycle.
In the bloodstream each merozoite invades a red blood cell and asexually divides up to 24 merozites in the host cell over a period of 2 to 3 days. Mature parasites rupture the host cell to infect new red blood cells, which causes the periodic elevation of body temperature (fever). Antibodies to merozoite can inhibit invasion of red blood cells (6). Antibodies to parasite-derived molecules on the surface of infected red blood cells can prevent adhesion to capillary endothelial cells, responsible for pathogenicity of major complications such as cerebral malaria. Vaccines against parasites undergoing the blood cycle are called blood-stage vaccines. Effective vaccines will reduce disease severity and the risk of death during infection.
A part of the blood-stage parasite develops to a sexual parasite, male and female gametocytes, instead of repeating the asexual cycle. Gametocytes are ingested by mosquitoes during blood sucking and they undergo the sexual process (sporogony). The TBVs focused on here are designed to prevent parasites spread by inhibiting development of parasites within mosquito vectors.

TRANSMISSION-BLOCKING VACCINE AND ITS EFFECT
Gametocytes ingested by susceptible species of mosquito shed the host erythrocyte and the female develops into a macrogamete and the male becomes eight microgametes. Gametes mate, and the fertilized zygotes transform to motile ookinetes that penetrate the mosquito midgut peritrophic matrix and midgut epithelial cells. Then they develop into oocysts on the serous membrane. Matured oocysts release sporozoites that migrate to the salivary glands to prepare the next infection. TBVs are designed to induce antibodies that disrupt at least one of these steps when ingested with parasite-infected blood. Unlike two other malaria vaccines, TBV does not provide direct protection to individuals infected vaccine. However, this vaccine was suggested to be one of the important strategies for integrated malaria control program. Primarily, TBV will reduce the incidence of malaria patients in the community whose residence is vaccinated. In addition, this would control the escape of parasite mutants from vaccines targeted to other stages of the parasites life cycle (7), which would prolong the effective life of other malaria vaccines.

TRANSMISSION-BLOCKING VACCINE CANDIDATES
Target molecules for TBV must be expressed by the sexual stage parasites, gametocytes to ookinetes (Fig. 2). Several candidate molecules expressed by the sexual stage parasites have been reported. They are mostly from P. falciparum responsible for nine of ten deaths.
1. Pfs230
Pfs230 is cloned as a molecule expressed by gametocytes of P. falciprum and its molecular size is 230 kDa (8). The expression remains after gametes fertilize and transform to zygotes (9). Antibodies against Pfs230 completely block transmission of the parasite to mosquito vectors when ingested with a gametocytemic bloodmeal through a membrane-feeding apparatus (10).
2. Pfs48/45
Pfs48/45 is also a molecule expressed by gametocytes of P. falciparum and shows on electric doublet of 48 and 45 kDa on polyacrylamide gel electrophoresis (11). The expression pattern is similar to that of Pfs230. A rodent parasite disrupted gene encoding P48/45 homologue demonstrates that P48/45 family is essential for male gamete fertility (12). Antibodies against this molecule also completely block parasite transmission to mosquitoes (13).
3. Pfg27
Pfg27 is expressed as early as gametocytogenesis (14). P. falciparum disrupted gene encoding Pfg27 fails to develop the sexual stage parasites, suggesting that this molecule functions at an early stage of gametocytogenesis in vertebrate hosts (15). This also generates antibodies that block parasite transmission (16).
4. Chitinase
Ookinetes secrete chitinase, an enzyme that hydrolyses chitin, a polymerized form of N-acetyl glucosamine (17). Chitin is a major component of mosquito peritrophic matrix and its rigid structure was suggested to function as a physical barrier to parasite invasion. Chitinase is essential to penetrate the peritrophic matrix because a chitinase inhibitor and disruptant of chitinase gene suppresses oocyst formation (18, 19). Thus, this represents a target molecule not only for the transmission-blocking vaccine strategy but also drug development (20).
5. Pfs25/Pvs25, Pfs28/Pvs28
The leading candidates for a transmission-blocking vaccine against P. falciparum have been cloned (21). Pfs25 and Pfs28 are expressed on the surface of ookinetes and are composed of four tandem epidermal growth factor-like domains, putatively anchored to the surface membrane by a glycosylphosphatidylinositol-attached motif. Yeast-produced recombinant proteins elicit transmission-blocking antibodies in animal models(22). Their homologs of P. vivax, Pvs25 and Pvs28, were recently cloned (23).
DEVELOPMENT OF TBV AGAINST VIVAX MALARIA BASED ON Pvs25
Among these vaccine candidates, the most advanced is P25 and P28 family proteins of P. falciparum and P. vivax, Pfs25, 28 and Pvs25, 28. TBV against P. vivax based on Pvs25 and Pvs28 were recently developed. Pvs25 and Pvs28 are successfully expressed as recombinant proteins by yeast Saccharomyces cerevisiae and the products are highly immunogenic and induce antibodies that block oocyst formation in mosquito when mixed with a gametocytemic blood meal (24). Thus, both proteins are potentially vaccine candidates. Gene knock-out experiments demonstrate that rodent malaria parasites deficient for the P25 and P28 proteins fail to generate oocysts in mosquitoes, suggesting a synergistic role in oocyst formation (25). This suggests that maximum inhibition will be obtained when antibodies to Pvs25 and Pvs28 exist. However, detailed experiments revealed that no synergistic effect of anti-Pvs25 and -Pvs28 antiserum on oocyst formation was observed (26). Together with low yield, the high frequency of variants, and the unresponsiveness in mice in MHC-dependent manner of Pvs28, a single use of Pvs25 as a vaccine was determined as the best candidate for TBV against P. vivax. Thereafter, Pvs25 has been tested for immunogenicity in non-human primates (27). A clinical grade Pvs25 is currently being produced and phase I trials will soon be conducted at the Malaria Vaccine Development Unit, NIH (Stowers, personal communication).

ACKNOWLEDGEMENTS
The authors would like to thank Ms. Kiyoko Suzuki and Ms. Yasuko Koto of Environmental and Toxicological Sciences Research Group, National Institute of Radiological Sciences for kind help in retrieval of scientific references for this review.

OBSTACLES TO BE OVERCOME
The most serious obstacles to developing a malaria vaccine (and other vaccines) are antigenic diversity, lack of in vitro assay system, difficulty in conducting human trials, and selection of an adjuvant delivery system. Comparing with two other malaria vaccines, these obstacles are easier to be overcome for the TBV based on Pvs25. Antigenic variation is much less frequent in P25 family proteins (23, 28). The molecules are not exposed to host immune systems because they are expressed only after fertilization in the mosquito. However, this results in another problem. Immune responses to the vaccine would not be boosted during natural infection. Therefore, formulations may need to be developed to extend the effective life of TBV-induced immunity.
The immune mechanism of TBV is antibody-mediated inhibition of parasites' development in mosquitoes. This has enabled an ex vivo assay to be established. Mosquitoes were fed with gametocyte-infected blood through a membrane in the presence of immune serum. Thereafter, the number of oocysts is counted. Using this assay, the transmission-blocking activity of serum from human volunteers will be evaluated without requiring experimental infection of each volunteer with malaria. This is a big advantage to design clinical trials. In terms of the adjuvant and delivery systems, we could be optimistic, because alum adjuvant (only adjuvant applicable for human use) combined with Pvs25 could induce high titers of antibody that has strong a transmission-blocking activity in rodents and non-human primates. Moreover, DNA vaccination also induces a high titer of antibodies (29, Kumar, personal communication).

CONCLUDING REMARKS
Although TBV does not provide direct protection to individuals, this is an important strategy for controlling malaria. Since TBV could prevent the spread of escape of mutants from other vaccines, this must be a member of a multi-component malaria vaccine. The biggest problem common to developing malaria vaccine is the relatively low level of commercial interest. This is because the potential market with endemic malaria is mainly in the developing countries who do not have economic support. However, economic support will be obtained in the future (ex. The Bill Gates Foundation for Malaria Vaccine Development was established).

REFERENCES
1.World Health Organization:World malaria situation in 1994. Wkly Epidemiol Rec 72:285-292, 1997
2.Harinasuta T, Suntharasamai P, Viravan C:Chloroquine-resistant falciparum malaria in Thailand. Lancet 2:657-660, 1965
3.Roberts DR, Ander RG:Insecticide resistance issues in vector-borne disease control. Am J Trop Med Hyg 50:21-34, 1994
4.Nussenzweig RS, Vanderberg J, Most H, Orton C:Protective immunity produced by the infectio of x-irradiated sporozoites of Plasmodium berbhei. Nature 216:160-162, 1967
5.Hoffman SL, Doolan DL:Malaria vaccines-targeting infected hepatocytes. Nat Med 6:1218-1219, 2000
6.Good MF, Kaslow DC, Miller LH:Pathways and strategies for developing malaria blood-stage vaccine. Ann Rev Immunol 16:57-87, 1998
7.Carter R, Mendis KN, Miller HL, Molineaux L, Saul A:Malaria transmission-blocking vaccines - how can their development be supported? Nat Med 6:241-244, 2000
8.Williamson KC, Criscio MD, Kaslow DC:Cloning and expression of the gene for Plasmodium falciparum transission-blocking target antigen, Pfs230. Mol Biochem Parasitol 58:355-358, 1993
9.Read D, Lensen AH, Begarnie S, Haley S, Raza A, Carter R:Transmission-blocking antibodies against multiple, non-variant target epitopes of the Plasmodium falciparum gamete surface antigen pfs230 are all complement-fixing. Parasite Immunol 16:511-519, 1994
10.Roeffen W, Geeraedts F, Eling W, Beckers P, Wizel B, Kumar N, Lensen T, Sauerwein R:Transmission blockade of Plasmodium falciparum malaria by anti-Pfs230-specific antibodies is isotype dependent. Infect Immun 63:467-471, 1995
11.Kocken CH, Jansen J, Kaan Am, Beckers PJ, Ponnudurai T, Kaslow DC, Konings RN, Schoenmakers JG:Cloning and expression of the gene coding for the transmission blocking target antigen Pfs48/45 of Plasmodium falciparum. Mol Biochem Parasitol 61:59-68, 1993
12.van Dijk MR, Janse CJ, Thompson J, Waters AP, Braks JA, Dodemont HJ, Stunnenberg HG, van Gemert GJ, Sauerwein RW, Eling W:A central role for P48/45 in malaria parasite male gamete fertility. Cell 104:153-164, 2001
13.Roeffen W, Lensen T, Mulder B, Teelen K, Sauerwein R, van Druten J, Eling W, Meuwissen JH, Beckers PJ:A comparison of transmission-blocking activity with reactivity in a Plasmodium falciparum 48/45-kD molecule-specific competition enzyme-linked immunosorbent assay. A J Trop Med Hyg 52:60-65, 1995
14.Alano P, Premawansa W, Bruce MC, Carter R:A stage specific gene expressed at the onset of gametocytogenesis in Plasmodium falciparum. Mol Biochem Parasitol 46:81-88, 1991
15.Lobo CA, Fujioka H, Aikawa M, Kumar N:Disruption of the Pfg27 locus by homologous recombination leads to loss of the sexual phenotype in P. falciparum. Mol Cell 3:793-798, 1999
16.Ploton IN, Wizel B, Viscidi R, Kumar N:Mapping of two overlapping linear epitopes in Pfg27 recognized by Plasmodium falciparum transmission-blocking monoclonal antibodies. Vaccine 13:1161-1169, 1995
17.Huber M, Cabib E, Miller LH:Malaria parasite chitinase and penetration of the mosquito peritrophic membrane. Proc Natl Acd Sci USA 88:2807-2810, 1991
18.Shahabudding M, Toyoshima T, Aikawa M, Kaslow DC:Transmission-blocking activity of a chitinase inhibitor and activation of malarial parasite chitinase by mosquito protease. Proc Natl Acd Sci USA 90:4266-4270, 1993
19.Tsai YL, Hayward RE, Langer RC, Fidock DA, Vinetz JM:Disruption of Plasmodium falciparum chitinase markedly impairs parasite invasion of mosquito midgut. Infect Immun 69:4048-4054, 2001
20.Langr RC, Vinetz JM:Plasmodium ookineter-secreted chitinase and parasite penetration of th mosquito peritrophic matrix. Trends Parasitol 17:269-272, 2001
21.Kaslow DC Quankyi IA, Syin C, Raum MB, Keister DB, Coligan JE, McCutchan TF, Miller LH:A vaccine candidate from the sexual stage of human malaria that contains EGF-like domains. Nature 333:74-46, 1988
22.Kaslow DC, Bathurst IC, Lensen T, Ponnuduraoi T, Barr PJ, Keister DB:Saccharomyces cerevisiae recombinant Pfs25 adsorbed to alum elicits antibodies that block transmission of Plasmodium falciparum. Infect Immun 62:5576-5580, 1994
23.Tsuboi T Kaslow DC, Gozar MMG, Tachibana M, Cao Y-M, Torii M:Sequence polymorphism in two novel Plasmodium vivax ookinete surface proteins, Pvs25 and Pvs28, that are malaria transmission-blocking vaccine candidates. Mol Med 4:772-778, 1998
24.Hisaeda H, Stowers AW, Tsuboi T, Collins WE, Sattabongkot JS, Suwanabun N, Torii M, Kaslow DC:Antibodies to malaria vaccine candidates Pvs25 and Pvs28 completely block the ability of Plasmodium vivax to infect mosquitoes. Infect Immun 68:6618-6623, 2000
25.Sinden-Kiamos I, Vlachou D, Margos G, Beetsma A, Waters AP, Sinden R, Louis C:Distinct roles fo Pbs21 and Pbs25 in the in vitro ookinete to oocyst transformation of Plasmodium berghei. J Cell Sci 113:3419-3426, 2000
26.Hisaeda H, Collins WE, Saul A, Stower AW:Antibodies to Plasmodium vivax transmission-blocking vaccine candidate antigens Pvs25 and Pvs28 dono show synergism. Vaccine 20:763-770, 2001
27.Arevalo-Herrera M, Herrera S:Plasmodium vivax malaria vaccine development. Mol Immunol 38:443-455, 2001
28.Kaslow DC, Quankyi IA, Keister DB:Minimal variation in a vaccine candidate from the sexual stage of Plasmodium falciparum. Mol Biochem Parasitol 32:101-103, 1989
29.Lobo CA, Khar R, Kumar N:Immunization of mice with DNA-based Pfs25 elicits potent malaria transmission-blocking antibodies. Infect Immun 67:1688-1693, 1999

Received for publication June 21, 2002;accepted July 4, 2002.

Address correspondence and reprint requests to Koji Yasutomo, M.D., Ph.D., Department of Immunology & Parasitology, The University of Tokushima School of Medicine, Kuramoto-cho, Tokushima 770-8503, Japan and Fax:+81-88-633-7114.