VIOLIN Logo
VO Banner
Search: for Help
About
Introduction
Statistics
VIOLIN News
Your VIOLIN
Register or Login
Submission
Tutorial
Vaccine & Components
Vaxquery
Vaxgen
VBLAST
Protegen
VirmugenDB
DNAVaxDB
CanVaxKB
Vaxjo
Vaxvec
Vevax
Huvax
Cov19VaxKB
Host Responses
VaximmutorDB
VIGET
Vaxafe
Vaxar
Vaxism
Vaccine Literature
VO-SciMiner
Litesearch
Vaxmesh
Vaxlert
Vaccine Design
Vaxign2
Vaxign
Community Efforts
Vaccine Ontology
ICoVax 2012
ICoVax 2013
Advisory Committee
Vaccine Society
Vaxperts
VaxPub
VaxCom
VaxLaw
VaxMedia
VaxMeet
VaxFund
VaxCareer
Data Exchange
V-Utilities
VIOLINML
Help & Documents
Publications
Documents
FAQs
Links
Acknowledgements
Disclaimer
Contact Us
UM Logo

Vaccine Comparison

P. berghei CS Protein Subunit Vaccine P. berghei DNA vaccine CSP-3p28 P. berghei DNA vaccine encoding PbCSP P. berghei MSP1 Protein Vaccine P. berghei p36p mutant vaccine P. chabaudi AMA1 Protein Vaccine P. falciparum CS expressed in irradiated P. berghei as Vaccine P. falciparum MSP4 with AFCo1 Adjuvant P. falciparum pfCelTos protein vaccine P. falciparum Pfen Protein Subunit Vaccine P. yoelii DNA vaccine encoding MSP1 P. yoelii DNA vaccine encoding PyHEP17 Protein P. yoelii DNA vaccine encoding PySSP2 P. yoelii DNA vaccine pDIP/PyCSP. 1 P. yoelii DNA vaccine pPyHsp60-VR1012 P. yoelii MSP1 and MSP4/5 Proteins Subunit Vaccine P. yoelii p36/p52 mutant vaccine P. yoelii TyCS-VLP Vaccine P. yoelii UIS3 mutant vaccine Plasmodium FabB/FabF mutant vaccine rBCGMSP1-15 Recombinant ABRA protein vaccine
Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information Vaccine Information
  • Vaccine Ontology ID: VO_0011549
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: A tandem repeat of the B cell immunodominant repeat epitope (DPPPPNPN)2D of the malaria parasite Plasmodium berghei circumsporozoite protein (P4c-Mal) (Kaba et al., 2009).
  • CS from P. berghei str. ANKA gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Immunization Route: Intraperitoneal injection (i.p.)
  • Vaccine Ontology ID: VO_0004594
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Antigen: CSP and 3 copies of the p28 fragment of C3d (Bergmann-Leitner et al., 2007)
  • CS from P. berghei str. ANKA gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: pBLUESCRIPT (Bergmann-Leitner et al., 2007)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004591
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • CS from P. berghei str. ANKA gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3.1 (Yoshida et al., 2000)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004065
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: Recombinant MSP1 (rPbMSP1)
  • MSP1 from P. berghei gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: Intraperitoneal injection (i.p.)
  • Vaccine Ontology ID: VO_0003008
  • Type: Live, attenuated vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • P36p gene engineering:
    • Type: Gene mutation
    • Description: This p36p mutant is from Plasmodium berghei (Douradinha et al., 2007).
    • Detailed Gene Information: Click Here.
  • Immunization Route: Intravenous injection (i.v.)
  • Vaccine Ontology ID: VO_0004194
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: Recombinant ectodomain of P. chabaudi adami (DS stain) AMA1 (denoted rAMA1B)
  • AMA-1 from P. chabaudi gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: Intraperitoneal injection (i.p.)
  • Vaccine Ontology ID: VO_0004794
  • Type: Recombinant vector vaccine
  • Status: Research
  • Antigen: CSP from P. falciparum
  • CS from P. falciparum gene engineering:
    • Type: Recombinant vector construction
    • Description: P. berghei ANKA cloned lines were transfected with the P. falciparum CSP gene (Grüner et al., 2007).
    • Detailed Gene Information: Click Here.
  • Immunization Route: non-specified injection
  • Vaccine Ontology ID: VO_0004243
  • Type: Subunit vaccine
  • Status: Research
  • MSP4 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: Not specified
  • Vaccine Ontology ID: VO_0004204
  • Type: Subunit vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: mouse
  • pfCelTOS gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
    • VO ID: VO_0001268
    • Description: Montanide ISA 720
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0011418
  • Type: Subunit vaccine
  • Status: Research
  • Pfen gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: Intraperitoneal injection (i.p.)
  • Vaccine Ontology ID: VO_0004593
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • MSP1 from P. yoelii str. 17XNL gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: pJW4304 (Sakai et al., 2003)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004162
  • Type: DNA vaccine
  • Status: Research
  • HEP17 gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: nkCMVintpolyli (Doolan et al., 1996)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004590
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • SSP2 gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: nkCMVint or VR1012 (Hoffman et al., 1997)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004589
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • CS from P. yoelii gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: pBC12/CMV/IL-2 (Hoffman et al., 1994)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004592
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • HSP60 from P. yoelii gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: VR1012 (Sanchez et al., 2001)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0011439
  • Type: Subunit vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: mouse
  • MSP4/5 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Immunization Route: Orally
  • Vaccine Ontology ID: VO_0003009
  • Type: Live, attenuated vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Py36 gene engineering:
    • Type: Gene mutation
    • Description: This p36/p52 mutant is from Plasmodium yoelii (Labaied et al., 2007).
    • Detailed Gene Information: Click Here.
  • Py52 gene engineering:
    • Type: Gene mutation
    • Description: This p36/p52 mutant is from Plasmodium yoelii (Labaied et al., 2007).
    • Detailed Gene Information: Click Here.
  • Immunization Route: Intravenous injection (i.v.)
  • Vaccine Ontology ID: VO_0004271
  • Type: Subunit vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: mouse
  • Antigen: CD8+ T cell epitope (SYVPSAEQI) of the circumsporozoite (CS) protein of Plasmodium yoelii (Oliveira-Ferreira et al., 2000).
  • Adjuvant:
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0003011
  • Type: Live, attenuated vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Antigen: UIS3(Tarun et al., 2007)
  • UIS3 gene engineering:
    • Type: Gene mutation
    • Description: This UIS3 mutant is from Plasmodium yoelii (Tarun et al., 2007).
    • Detailed Gene Information: Click Here.
  • Immunization Route: Intravenous injection (i.v.)
  • Vaccine Ontology ID: VO_0003007
  • Type: Live, attenuated vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • FabB/FabF gene engineering:
    • Type: Gene mutation
    • Description: This FabB/FabF mutant is from Plasmodium (Butler et al., 2011).
    • Detailed Gene Information: Click Here.
  • Immunization Route: Intravenous injection (i.v.)
  • Vaccine Ontology ID: VO_0004791
  • Type: Recombinant vector vaccine
  • Status: Research
  • Host Species for Licensed Use: Mouse
  • Antigen: MSP1-15 as a fusion protein with the α antigen of Mycobacterium kansasii (α-k), which is secreted from the rBCG vaccine vector (Matsumoto et al., 1998).
  • MSP1 from P. yoelii str. 17XNL gene engineering:
    • Type: Recombinant vector construction
    • Description: A 2.4-kbp fragment containing an α-k–MSP1-15 hybrid gene was subcloned into pSO246. The final construct (designated pSOMSP1-15) was transformed into BCG Tokyo by electroporation (Matsumoto et al., 1998).
    • Detailed Gene Information: Click Here.
  • Immunization Route: Intravenous injection (i.v.)
  • Vaccine Ontology ID: VO_0000778
  • Type: Subunit vaccine
  • Host Species as Laboratory Animal Model: mouse
  • Antigen: The acidic basic repeat antigen (ABRA) of Plasmodium falciparum is a vaccine candidate against erythrocytic stages of malaria (Kushwaha et al., 2001).
  • ABRA gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
    • VO ID: VO_0000139
    • Description: Complete Freund's adjuvant (CFA) was used as the first adjuvant. Boosters of the same amount of protein were given in incomplete Freund's adjuvant (IFA) (Kushwaha et al., 2001).
  • Preparation: The four fragments of ABRA, including N-terminal [ABRA (N); aa 24–369], middle [ABRA (M); aa 370–507], N-terminal + middle [ABRA (P); aa 24–507] and the C-terminal [ABRA (C); aa 508–743], were expressed as fusions with either maltose binding protein (MBP) or 6X histidine tag at their amino terminii using pMALc-2 vector from NEB (New England Biolabs, Beverly, MA, USA) or pQE30 vector (Qiagen GmbH, Germany), respectively. These recombinant proteins were purified to near homogeneity by affinity chromatography of the soluble fraction, concentrated, and the purity of the protein judged by SDS-PAGE.
Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response Host Response

Mouse Response

  • Host Strain: BALB/c, C57BL/6
  • Vaccination Protocol: Mice were randomly divided into groups of 5 or 10 and immunized i.p. three times at 14-day intervals. Where indicated, a positive control group was immunized with irradiated P. berghei sporozoites (Kaba et al., 2009).
  • Challenge Protocol: P. berghei sporozoites (ANKA strain), maintained by cyclical transmission in mice and Anopheles stephensi, were dissected from mosquitoes 21–23 days after their infectious blood meal and used within 6 h. Fourteen days after the final immunization or at other specific times on long-term memory experiments, mice were challenged with a lethal dose of live P. berghei sporozoites by i.v. inoculation. C57BL/6, MHC KO, and nude mice were injected with 1000 sporozoites and BALB/c mice were injected with 4000 sporozoites per mouse (Kaba et al., 2009).
  • Efficacy: More than 95% of mice immunized with P4c-Mal, both with and without Montanide ISA-720, or R-PbCSP in Montanide ISA-720 did not develop any parasitemia and thus showed complete protection against challenge with viable sporozoites (Fig. 2B). This ability to prevent parasitemia and thus prevent malaria following sporozoite challenge is equivalent to what is only achieved with the whole, irradiated sporozoite immunization regime. In contrast, as few as 5% of animals administered saline, saline and Montanide ISA-720, or R-PbCSP in saline did not develop parasites and survived until 11 days post challenge. No animal was observed with blood stage parasites that did not die naturally or was killed according to protocol. These results show that immunization with P4c-Mal had a significant ability to induce a protective immune response in the presence as well as in the absence of adjuvant (Kaba et al., 2009).

Mouse Response

  • Vaccine Immune Response Type: VO_0003057
  • Efficacy: Vaccination with CSP-3p28 resulted in better (100%) protection than CSP alone (60%) against P. berghei sporozoites at the 6-week challenge (p = 0.043) suggesting that the addition of 3 copies of the p28 peptide to CSP results in the generation of a better vaccine (Bergmann-Leitner et al., 2007).

Mouse Response

  • Vaccine Immune Response Type: VO_0003057
  • Efficacy: Protection obtained by gene gun delivery into the liver once (73%) was significantly higher than that by the material into the skin twice (31%) (Yoshida et al., 2000).

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: Lyophilized rPbMSP1 was mixed with alum on the day of injection. Each vaccine formulation, containing 10 ug was administered through IP route to mice .
  • Challenge Protocol: For challenge study, mice were intraperitoneally inoculated with parasitized erythrocytes at a density of either 10^6 or 10^5 parasitized
    erythrocytes per mouse (Wan et al., 2007).
  • Efficacy: Eight out of ten mice vaccinated with rMSP1 in alum survived challenge with P. berghei (Wan et al., 2007).

Mouse Response

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: Groups of three to five mice were immunized i.p. with 15 µg of rAMA1B emulsified in Montanide ISA720. Four weeks later, a booster immunization was given using the same amount of rAMA1B emulsified with Montanide ISA720. Controls were immunized with PBS emulsified in Montanide ISA720 (Xu et al., 2000).
  • Challenge Protocol: Ten days after being given a booster immunization the mice were challenged i.v. with 1 x 10^5 P. chabaudi adami parasitized erythrocytes (Xu et al., 2000).
  • Efficacy: Immunized mice demonstrated significantly lower peak parasitemias compared with PBS-immunized mice, showing that rAMA1B immunization confers protection against challenge with P. chabaudi (Xu et al., 2000).

Mouse Response

  • Host Strain: BALB/cJ
  • Vaccination Protocol: In order to induce sterile immunity in all the animals, BALB/cJ mice were immunized with 12,000 rad-irradiated P. berghei sporozoites as follows: one dose of 75,000 sporozoites followed by two booster doses of 25,00 of P. berghei sporozoites on days 15 and 21. In [BALB/c×C57BL/6] F1 mice immunisation was made with 3 injections of 10,000 P. berghei irradiated sporozoites at days 0, 15 and 21 (Grüner et al., 2007).
  • Challenge Protocol: Control mice and mice immunized with irradiated sporozoites (transfected with P. falciparum CS) were challenged intravenously with 5,000 P. berghei or P. berghei [PfCS] sporozoites (Grüner et al., 2007).
  • Efficacy: Mice immunized with irradiated sporozoites were protected from challenge (Grüner et al., 2007).

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: MIce were immunized at 14-day intervals with three doses of 10 μg MSPs (Bracho et al., 2009).
  • Immune Response: AFCo1 significantly enhanced the IgG and T-cell response against MSP4, with a potency equivalent to CFA, with the response being characterized by both IgG1 and IgG2a isotypes, increased interferon gamma production and a strong DTH response, consistent with the ability of AFCo1 to induce Th1-like immune responses (Bracho et al., 2009).

Mouse Response

  • Host Strain: Balb/c-J
  • Vaccination Protocol: Mice were immunized subcutaneously in the scruff of the neck three times with 25 or 10 or 1 µg/dose of recombinant PfCelTOS or saline emulsified in Montanide ISA 720 (Bergmann-Leitner et al., 2010).
  • Challenge Protocol: Fourteen days after the final immunization, mice were challenged by subcutaneous inoculation (into the inguinal region) with 4,000 P. berghei sporozoites for Balb/c and 15,000 P. berghei sporozoites for CD-1 mice, dissected from infected mosquito salivary glands. The challenge dose was determined by titration studies in each mouse strain and compared to the different challenge routes. Infection was determined by the presence of blood stage parasites in Giemsa stained thin blood smears on day 6 and day 8 after challenge. Animals that were not infected at that time were tested again on day 14. Mice that remained un-infected by day 14 were classified as sterilely protected. We used this evaluation schedule because animals that are infected with P. berghei ANKA strain malaria parasites do not self-cure.
  • Efficacy Detail:
    No. Efficacy method Result Description Group Efficacy Detail
    1 CFU Immunization with PfCelTOS resulted in potent humoral and cellular immune responses and most importantly induced sterile protection against a heterologous challenge with P. berghei sporozoites in a proportion of both inbred and outbred mice (Bergmann-Leitner et al., 2010).

Mouse Response

  • Host Strain: Swiss
  • Vaccination Protocol: Mice were injected intraperitoneally with r-Pfen emulsified in Freund's adjuvant at 21-day intervals (the first injection was 100 μg of r-Pfen in complete Freund's adjuvant, followed by 50 μg for the two boosters in incomplete Freund's adjuvant). In one control group, mice were injected in parallel with a recombinant Drosophila odorant binding protein OSF (as an irrelevant His-tagged protein control) emulsified in complete Freund's adjuvant. The other control group received no injections. After three immunizations, the antibody titers against r-Pfen were monitored (Pal-Bhowmick et al., 2007).
  • Challenge Protocol: Mice having anti-r-Pfen antibody titers greater than 1:300,000 were then challenged with the lethal strain of P. yoelii (strain 17XL; 10^6 parasites per mouse), and parasitemia was monitored daily (Pal-Bhowmick et al., 2007).
  • Efficacy: All the control mice and the mice immunized with the irrelevant His-tagged protein developed a high degree of parasitemia (>17% on average) by day 4 postchallenge, whereas r-Pfen-immunized mice showed <1% parasitemia at that time point. The highest average parasitemia values were 70% and 40% for nonimmunized mice and mice injected with irrelevant His-tagged protein, respectively. However, among the mice immunized with r-Pfen, there was significant delay in the increase in parasitemia, and the highest average parasitemia was about 20% on day 8 postchallenge. The averages of these groups were compared using one-way analysis of variance, which showed that the mice immunized with enolase were significantly protected (Pal-Bhowmick et al., 2007).

Mouse Response

  • Vaccine Immune Response Type: VO_0003057
  • Challenge Protocol: Two weeks after the final immunization, the mice were challenged i.p. with 10^5 P. yoelii pRBC. The course of infection was monitored by microscopic examination of tail-blood smears stained with Gimsa. (Sakai et al., 2003)
  • Efficacy: MSP1 vaccine alone conferred partial protection. Vaccination with MSP1 + IL-12 conferred the strongest protective immunity against the infection. Only two of the six mice immunized with MSP1 alone survived, while five of the six mice immunized with MSP1 + IL-12 survived (Sakai et al., 2003).

Mouse Response

  • Host Strain: BALB/cByJ, A/J, B10.BR, B10.Q and C57BL/6
  • Vaccination Protocol: Female 6- to 8-wk-old mice were immunized three times at 3-wk intervals intramuscularly in each tibialis anterior muscle with 50 μg of PyHEP17 DNA in 50 μl of saline or unmodified nkCMVintpolyli plasmid. 2 wk after the third immunization, mice were challenged by tail-vein injection with 100 infectious sporozoites or 200 infected erythrocytes (Doolan et al., 1996).
  • Challenge Detail:
    No. Pathogen Name Dose Route Age Interval
    1 Plasmodium yoelii 100 CFU in volume ml Tail vein injection day 14 day
  • Efficacy Detail:
    No. Efficacy method Result Description Group Efficacy Detail
    1 In vivo protection study CFU Immunization with PyHEP17 DNA partially protected three of the five strains >20% against challenge with 100 infectious sporozoites. A 2 to 6 day delay in the onset of parasitemia in some nonprotected mice was consistent with partial immunity, which eliminated up to 90% of infected hepatocytes (Doolan et al., 1996).

Mouse Response

  • Vaccine Immune Response Type: VO_0003057
  • Efficacy: The full-length gene of PySSP2 in the nkCMVint vector induced specific antibodies and protected 50% of immunized mice. Subsequently, outbred CD-l mice were immunized with nkCMVint and VR1012 vector based PySSP2 DNA vaccines and as many as 33% were protected (Hoffman et al., 1997).

Mouse Response

  • Vaccine Immune Response Type: VO_0003057
  • Efficacy: Mice immunized with three doses of pDIP/PyCSP.1 and challenged with 5 × 105 P. yoelii sporozoites had a significant reduction in liver stage infection compared with mice immunized with the empty plasmid. Most importantly, 9 of 16 mice were protected against challenge (Hoffman et al., 1994).

Mouse Response

  • Vaccine Immune Response Type: VO_0003057
  • Efficacy: In experiment 1, 40% of mice immunized with the combination of pPyHsp60-VR1012 and pmurGM-CSF did not develop parasitemia during the 14 days postchallenge. Only this group had statistically significant protection on day 14 as compared with the pooled controls (two-tailed Fisher's exact test: P = 0.031 , group 1.B versus group 1.H + group 1.I). However, in experiment 2 (identical immunization schedule), immunized mice only experienced delayed parasitemia, rather than protection from parasitemia (Sanchez et al., 2001).

Mouse Response

  • Vaccination Protocol: One group of mice was treated by gavage with 25 μg of EcMSP4/5, and the other group was treated by gavage with 25 μg of EcMSP4/5 plus an amount of GST-PyMSP119 equivalent to 25 μg of PyMSP119 (Wang et al., 2004).
  • Challenge Protocol: In order to examine the protective efficacy of the induced antibodies, the immunized mice were challenged at 2 weeks after the sixth immunization with a lethal dose of 10^5 P. yoelii YM parasites (Wang et al., 2004).
  • Efficacy: Oral immunization of mice with Escherichia coli-expressed Plasmodium yoelii merozoite surface protein 4/5 or the C-terminal 19-kDa fragment of merozoite surface protein 1 induced systemic antibody responses and protected mice against lethal malaria infection. All of the eight immunized mice survived the challenge, with peak parasitemia levels between 0.2 and 55.2% (Wang et al., 2004).

Mouse Response

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: All the immunizations with TyCS-VLP carrying the CTL epitope of the P. yoelii circumsporozoite protein (SYVPSAEQI), except the dose response and the route of immunization experiments, consisted of 50 mg per mouse injected intramuscularly (i.m.) in the leg quadriceps (Oliveira-Ferreira et al., 2000).
  • Challenge Protocol: Mice were challenged with 75 sporozoites per mice administered i.v. (Oliveira-Ferreira et al., 2000).
  • Efficacy: 2/8 mice immunized with the TyCs-VLP vaccine were protected from challenge with P. yoelii sporozoites (Oliveira-Ferreira et al., 2000).

Mouse Response

  • Persistence: A UIS3 mutant is attenuated in mice (Tarun et al., 2007).
  • Efficacy: A UIS3 mutant provided complete protection in mice after two doses from challenge with wild type Plasmodium yoelii (Tarun et al., 2007).

Mouse Response

  • Persistence: A FabB/FabF genetically attenuated parasite is attenuated in mice (Butler et al., 2011).
  • Efficacy: A FabB/FabF genetically attenuated parasite induces complete protection in mice from challenge with wild type Plasmodium (Butler et al., 2011).

Mouse Response

  • Host Strain: C57BL/6, C3H/He, A/J
  • Vaccination Protocol: C3H/He mice were immunized intravenously with 10^6 CFU of rBCGMSP1-15 in 200 μl of PBS containing 0.1% PBS-T80. A control group of mice was injected with 106 CFU of BCG in 200 μl of PBS-T80 or PBS-T80 only. 30 days later, the same amount of each sample was injected intraperitoneally to boost the immune response (Matsumoto et al., 1998).
  • Vaccine Immune Response Type: VO_0003057
  • Challenge Protocol: Mice were challenged with 10^4 P. yoelii 17XL-parasitized erythrocytes intravenously or intraperitoneally 1 month after the final immunization (Matsumoto et al., 1998).
  • Efficacy: 3 out of 7 mice immunized with GST-MSP1-15 in RAS and 2 out of 8 mice immunized with GST-MSP1-15 in IFA survived the infection. 6 out of 7 mice immunized with rBCGMSP1-15 survived the infection. Data showed the three adjuvants examined are effective for vaccination with MSP1-15, while their efficacy levels differ. The rBCG system was the most effective for vaccination (Matsumoto et al., 1998).

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: Groups of BALB/c mice were immunized by i.p. injection with ABRA protein/MBP emulsified in complete Freund's adjuvant (CFA). Control mice received only PBS in the adjuvant. Boosters of the same amount of protein were given in incomplete Freund's adjuvant (IFA). Sera were collected from each group and treated as described earlier (Kushwaha et al., 2001).
  • Persistence: Humoral and cell-mediated response was still robust up to 70 days post-immunization (Kushwaha et al., 2001).
  • Immune Response: Results showed that ABRA (M), ABRA (C) and ABRA (P) were highly immunogenic in these animals; end point titres greater than 10^5 were observed in these constructs. Mice were immunized using standard methods. Relative concentrations of the antibodies elicited by them in mice at different intervals of immunization were measured at a dilution of 1 : 3000. ABRA (N) and ABRA (C) did not show a boostable antibody response; two secondary immunizations did not result in any increase in the antibody titre. Immunogenicity studies with these constructs in rabbits and mice indicated that the N-terminal region is the least immunogenic part of ABRA. T-cell proliferation experiments in mice immunized with these constructs revealed that the T-cell epitopes were localized in the middle portion of the protein (Kushwaha et al., 2001).
  • Efficacy: The purified immunoglobulin G specific to middle and C-terminal fragments prevented parasite growth at levels approaching 80-90% (Kushwaha et al., 2001).
  • Description: This antibody response was the focus of intense interest when it was found that mice could be rendered resistant to sporozoite challenge by passive immunisation with monoclonal antibodies against circumsporozoite protein (Kwiatkowski et al., 1997).
References References References References References References References References References References References References References References References References References References References References References References
Kaba et al., 2009: Kaba SA, Brando C, Guo Q, Mittelholzer C, Raman S, Tropel D, Aebi U, Burkhard P, Lanar DE. A nonadjuvanted polypeptide nanoparticle vaccine confers long-lasting protection against rodent malaria. Journal of immunology (Baltimore, Md. : 1950). 2009; 183(11); 7268-7277. [PubMed: 19915055].
Bergmann-Leitner et al., 2007: Bergmann-Leitner ES, Duncan EH, Leitner WW, Neutzner A, Savranskaya T, Angov E, Tsokos GC. C3d-defined complement receptor-binding peptide p28 conjugated to circumsporozoite protein provides protection against Plasmodium berghei. Vaccine. 2007; 25(45); 7732-7736. [PubMed: 17931754].
Yoshida et al., 2000: Yoshida S, Kashiwamura SI, Hosoya Y, Luo E, Matsuoka H, Ishii A, Fujimura A, Kobayashi E. Direct immunization of malaria DNA vaccine into the liver by gene gun protects against lethal challenge of Plasmodium berghei sporozoite. Biochemical and biophysical research communications. 2000; 271(1); 107-115. [PubMed: 10777689].
Wan et al., 2007: Wan Omar A, Roslaini AM, Ngah ZU, Azahari AA, Zahedi M, Baharudin O. A recombinant 19 kDa Plasmodium berghei merozoite surface protein 1 formulated with alum induces protective immune response in mice. Tropical biomedicine. 2007; 24(1); 119-126. [PubMed: 17568385].
Douradinha et al., 2007: Douradinha B, van Dijk MR, Ataide R, van Gemert GJ, Thompson J, Franetich JF, Mazier D, Luty AJ, Sauerwein R, Janse CJ, Waters AP, Mota MM. Genetically attenuated P36p-deficient Plasmodium berghei sporozoites confer long-lasting and partial cross-species protection. International journal for parasitology. 2007; 37(13); 1511-1519. [PubMed: 17604034].
Xu et al., 2000: Xu H, Hodder AN, Yan H, Crewther PE, Anders RF, Good MF. CD4+ T cells acting independently of antibody contribute to protective immunity to Plasmodium chabaudi infection after apical membrane antigen 1 immunization. Journal of immunology (Baltimore, Md. : 1950). 2000; 165(1); 389-396. [PubMed: 10861076].
Grüner et al., 2007: Grüner AC, Mauduit M, Tewari R, Romero JF, Depinay N, Kayibanda M, Lallemand E, Chavatte JM, Crisanti A, Sinnis P, Mazier D, Corradin G, Snounou G, Rénia L. Sterile protection against malaria is independent of immune responses to the circumsporozoite protein. PloS one. 2007; 2(12); e1371. [PubMed: 18159254].
Bracho et al., 2009: Bracho G, Zayas C, Wang L, Coppel R, Pérez O, Petrovsky N. AFCo1, a meningococcal B-derived cochleate adjuvant, strongly enhances antibody and T-cell immunity against Plasmodium falciparum merozoite surface protein 4 and 5. Malaria journal. 2009; 8; 35. [PubMed: 19250541].
Bergmann-Leitner et al., 2010: Bergmann-Leitner ES, Mease RM, De La Vega P, Savranskaya T, Polhemus M, Ockenhouse C, Angov E. Immunization with pre-erythrocytic antigen CelTOS from Plasmodium falciparum elicits cross-species protection against heterologous challenge with Plasmodium berghei. PloS one. 2010; 5(8); e12294. [PubMed: 20808868].
Pal-Bhowmick et al., 2007: Pal-Bhowmick I, Mehta M, Coppens I, Sharma S, Jarori GK. Protective properties and surface localization of Plasmodium falciparum enolase. Infection and immunity. 2007; 75(11); 5500-5508. [PubMed: 17785475].
Sakai et al., 2003: Sakai T, Hisaeda H, Nakano Y, Zhang M, Takashima M, Ishii K, Maekawa Y, Matsumoto S, Nitta Y, Miyazaki Ji, Yamamoto S, Himeno K. Gene gun-based co-immunization of merozoite surface protein-1 cDNA with IL-12 expression plasmid confers protection against lethal Plasmodium yoelii in A/J mice. Vaccine. 2003; 21(13-14); 1432-1444. [PubMed: 12615440].
Doolan et al., 1996: Doolan DL, Sedegah M, Hedstrom RC, Hobart P, Charoenvit Y, Hoffman SL. Circumventing genetic restriction of protection against malaria with multigene DNA immunization: CD8+ cell-, interferon gamma-, and nitric oxide-dependent immunity. The Journal of experimental medicine. 1996; 183(4); 1739-1746. [PubMed: 8666931].
Hoffman et al., 1997: Hoffman SL, Doolan DL, Sedegah M, Aguiar JC, Wang R, Malik A, Gramzinski RA, Weiss WR, Hobart P, Norman JA, Margalith M, Hedstrom RC. Strategy for development of a pre-erythrocytic Plasmodium falciparum DNA vaccine for human use. Vaccine. 1997; 15(8); 842-845. [PubMed: 9234529].
Hoffman et al., 1994: Hoffman SL, Sedegah M, Hedstrom RC. Protection against malaria by immunization with a Plasmodium yoelii circumsporozoite protein nucleic acid vaccine. Vaccine. 1994; 12(16); 1529-1533. [PubMed: 7879419].
Sanchez et al., 2001: Sanchez GI, Sedegah M, Rogers WO, Jones TR, Sacci J, Witney A, Carucci DJ, Kumar N, Hoffman SL. Immunogenicity and protective efficacy of a Plasmodium yoelii Hsp60 DNA vaccine in BALB/c mice. Infection and immunity. 2001; 69(6); 3897-3905. [PubMed: 11349057].
Wang et al., 2004: Wang L, Goschnick MW, Coppel RL. Oral immunization with a combination of Plasmodium yoelii merozoite surface proteins 1 and 4/5 enhances protection against lethal malaria challenge. Infection and immunity. 2004; 72(10); 6172-6175. [PubMed: 15385527].
Labaied et al., 2007: Labaied M, Harupa A, Dumpit RF, Coppens I, Mikolajczak SA, Kappe SH. Plasmodium yoelii sporozoites with simultaneous deletion of P52 and P36 are completely attenuated and confer sterile immunity against infection. Infection and immunity. 2007; 75(8); 3758-3768. [PubMed: 17517871].
Oliveira-Ferreira et al., 2000: Oliveira-Ferreira J, Miyahira Y, Layton GT, Savage N, Esteban M, Rodriguez D, Rodriguez JR, Nussenzweig RS, Zavala F. Immunogenicity of Ty-VLP bearing a CD8(+) T cell epitope of the CS protein of P. yoelii: enhanced memory response by boosting with recombinant vaccinia virus. Vaccine. 2000; 18(17); 1863-1869. [PubMed: 10699335].
Tarun et al., 2007: Tarun AS, Dumpit RF, Camargo N, Labaied M, Liu P, Takagi A, Wang R, Kappe SH. Protracted sterile protection with Plasmodium yoelii pre-erythrocytic genetically attenuated parasite malaria vaccines is independent of significant liver-stage persistence and is mediated by CD8+ T cells. The Journal of infectious diseases. 2007; 196(4); 608-616. [PubMed: 17624848].
Butler et al., 2011: Butler NS, Schmidt NW, Vaughan AM, Aly AS, Kappe SH, Harty JT. Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell host & microbe. 2011; 9(6); 451-462. [PubMed: 21669394].
Matsumoto et al., 1998: Matsumoto S, Yukitake H, Kanbara H, Yamada T. Recombinant Mycobacterium bovis bacillus Calmette-Guérin secreting merozoite surface protein 1 (MSP1) induces protection against rodent malaria parasite infection depending on MSP1-stimulated interferon gamma and parasite-specific antibodies. The Journal of experimental medicine. 1998; 188(5); 845-854. [PubMed: 9730886].
Kushwaha et al., 2001: Kushwaha A, Rao PP, Suresh RP, Chauhan VS. Immunogenicity of recombinant fragments of Plasmodium falciparum acidic basic repeat antigen produced in Escherichia coli. Parasite immunology. 2001 Aug; 23(8); 435-44. [PubMed: 11489167].
Kwiatkowski et al., 1997: Kwiatkowski D, Marsh K. Development of a malaria vaccine. Lancet. 1997 Dec 6; 350(9092); 1696-701. [PubMed: 9400530].