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

B-cell lymphoma DNA vaccine VCL-1642.XXX encoding a chimeric immunoglobulin molecule onsisting of tumor-specific variable (Id) regions B16 melanoma DNA vaccine pCMV-hTRP-1 encoding TRP-1 B16 melanoma DNA vaccine pSin-hTRP-1 encoding TRP-1 B16 melanoma DNA vaccine pSin-mTRP-1 B16 Vaccine adjuvanted by Loxoribine Cancer DNA Vaccine encoding Endoglin Boosted with Recombinant Endoglin Protein Cancer DNA Vaccine MIDGE/hNIS encoding hNIS protein Cancer DNA vaccine p.DOM-AH1 encoding fragment C Cancer DNA Vaccine pLXSHDmB7-2 encoding Cd86 Cancer DNA Vaccine pSURV encoding Survivin Cancer EPCAM protein vaccine Cancer Her-2/neu Protein Subunit Vaccine Cancer Retroviral Vector Vaccine encoding Cd40lg Cancer Subunit DC (Tat-CEA) vaccine Cancer Subunit NY-ESO-1 Protein Vaccine Cancer Vaccine using E7 protein of human papillomavirus 16 and Algammulin Cancer Vaccine using E7 protein of human papillomavirus 16 and Quil-A Cancer VEGFA protein vaccine Carcinoma DNA vaccine pVAX/E2A Lung metastasis DNA vaccine pCEP4-MUC1 encoding MUC1 Lymphoma DNA vaccine VαVβVβ-FrC Melanoma DNA vaccine gp75 DNA encoding melanosomal membrane glycoproteins, gp75 Melanoma DNA vaccine hTRP2 encoding TRP-2 Melanoma DNA vaccine hugp100 encoding gp100 Melanoma DNA vaccine pCR3.1-VS-HSP65-TP-GRP6-M2 encoding 6 tandem repeats of GRP Melanoma DNA vaccine pN4a-MAGE-1-GM Melanoma DNA vaccine pNL3-MAGE-1-GM Melanoma DNA vaccine pUB-TRP-2 encoding a fusion protein linking murine ubiquitin (UB) to the N-terminus of the full-length mTRP-2 Melanoma DNA vaccine SINCp c-muMUC18 encoding MCAM/MUC18 Melanoma DNA vaccine VR-S8/VR-IL2/AD-S8 encoding survivin Melanoma DNA vaccine VR1012/mPmel17 Melanoma recombinant vector vaccine rVVmTRP-1 encoding TRP-1 Neuroblastoma DNA vaccine HuDsec Prostate cancer DNA vaccine encoding PSCA Prostate cancer DNA vaccine pcDNA3-STEAP encoding six-transmembrane epithelial antigen Prostate cancer DNA vaccine pmPSCA encoding PSCA Prostate cancer DNA vaccine PSCA-HSP encoding PSCA and HSP Prostate cancer DNA vaccine psig-3P-Fc encoding 3P(hPSM, hPAP, or hPSA)-Fc fusion protein Prostate cancer DNA vaccine pVax-PSA Recombinant Colorectal cancer antigen GA733 Vaccine Synthetic MUC1 Peptide 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 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_0004427
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Human
  • Antigen: Immunoglobulin
  • Immunoglobulin gene engineering:
    • Type: DNA vaccine construction
    • Description: The DNA encoded a chimeric immunoglobulin molecule containing variable heavy and light chain immunoglobulin sequences derived from each patient’s tumor (Timmerman et al., 2002).
    • Detailed Gene Information: Click Here.
  • Vector: VCL-1632 (Timmerman et al., 2002)
  • Preparation: The vaccine is made of DNA encoded a chimeric immunoglobulin molecule containing variable heavy and light chain immunoglobulin sequences derived from each patient’s tumor, linked to the IgG2a and κ mouse immunoglobulin (MsIg) heavy- and light-chain constant regions chains, respectively (Timmerman et al., 2002). DNA used could also encode an ld/GM-CSF (Syrengelas et al., 1996).
  • Immunization Route: Intramuscular injection (i.m.)
  • Description: This vaccine has been used in clinical trials involving B-cell lymphoma. (Syrengelas et al., 1996)
  • Vaccine Ontology ID: VO_0004430
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • TYRP1 gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Tyrp1 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Vector: pSport-hTRP-1, based on pSport-B-gal (Leitner et al., 2003)
  • Preparation: Vaccine is made with naked DNA encoding an alphavirus replicon (self-replicating mRNA) and the self/tumor antigen tyrosinase-related protein-1 with antigen expression controlled by cytomegalovirus (CMV) promoter (Leitner et al., 2003).
  • Immunization Route: Gene gun
  • Description: This vaccine has been used in research involving melanoma. (Leitner et al., 2003)
  • Vaccine Ontology ID: VO_0004429
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Tyrp1 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Vector: pSin-B-gal (Leitner et al., 2003)
  • Preparation: The replicase-based plasmids (pSin-mTRP-1, pSin-hTRP-1) are derived from pSin-β-gal (Leitner et al., 2003).
  • Immunization Route: Gene gun
  • Description: This vaccine has been used in research involving melanoma. (Leitner et al., 2003)
  • Vaccine Ontology ID: VO_0004518
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Tyrp1 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Vector: pSin-B-gal (Leitner et al., 2003)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0001380
  • Type: Live, attenuated vaccine
  • Status: Research
  • Antigen: Irradiated B16 cells (Pope et al., 1994).
  • Adjuvant:
  • Preparation: Loxoribine can be used to promote inhibition of B16 metastasis (Pope et al., 1994).
  • Immunization Route: Intraperitoneal injection (i.p.)
  • Description: This vaccine has been used in clinical trials involving melanoma. (Pope et al., 1994)
  • Vaccine Ontology ID: VO_0011367
  • Type: DNA vaccine
  • Status: Research
  • Eng gene engineering:
    • Type: DNA Vaccine Construction and Recombinant Prot
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3.1(+) (Tan et al., 2007)
  • Immunization Route: i.m. injection for DNA, s.c. for protein
  • Vaccine Ontology ID: VO_0011364
  • Type: DNA vaccine
  • Status: Research
  • SLC5A5 gene engineering:
    • Type: DNA vaccine construction
    • Description: Human sodium iodide symporter (hNIS) is a transmembrane protein that actively transports iodide ions into thyroid cells. hNIS is over-expressed in some cases of the thyroid cancers compared with the surrounding normal tissues and has been considered to be an attractive target for immunotherapy. Minimalistic immunogenically defined gene expression (MIDGE) was used as a vector system (Choi et al., 2007).
    • Detailed Gene Information: Click Here.
  • Vector: Minimalistic immunogenically defined gene expression (MIDGE), derived from hNIS plasmid, pcDNA3.1-hNIS (Choi et al., 2007).
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004425
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Fragment C from tetanus toxin gene engineering:
    • Type: DNA vaccine construction
    • Description: This DNA vaccine expressed the first domain of fragment C (FrC)3 from tetanus toxin (DOM; TT865–1120) with sequence encoding the AH1 CTL epitope fused to the 3 terminus (Buchan et al., 2005).
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3 (Buchan et al., 2005)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0011373
  • Type: DNA vaccine
  • Status: Research
  • Antigen: Cd86
  • Cd86 gene engineering:
    • Type: DNA vaccine construction
    • Description: A DNA fragment encoding the entire open reading frame of murine B7-2 was amplified by reverse transcription-coupled PCR (27) from RNA prepared from LPS-activated murine spleen cells (15). The sense primer (S’-TCGATAGGAATTCGTAGACGTGTTCCAGAACIT3’) consists of an oligonucleotide corresponding to 64 to 83 nucleotides of murine B7-2 cDNA, plus a restriction site for EcoRI. The antisense primer (5’-TACGATACTCGAGTCTCACTGCCTTCACTCTGCAT-3') corresponds to 1018 to 1039 nucleotides of murine B7-2 cDNA, plus a site for Xhol. The PCR product was cloned directly into the vector pLXSHD (provided by Dr. D. Miller, Fred Hutchinson Cancer Research Center, Seattle, WA) (Yang et al., 1995).
    • Detailed Gene Information: Click Here.
  • Vector: pLXSHD
  • Immunization Route: Intraperitoneal injection (i.p.)
  • Vaccine Ontology ID: VO_0011365
  • Type: DNA vaccine
  • Status: Research
  • Survivin gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • CD40 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Vector: pVAX (Lladser et al., 2010)
  • Immunization Route: Intradermal injection (i.d.)
  • Vaccine Ontology ID: VO_0011370
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: EPCAM (GA733)
  • EPCAM gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0011363
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: ERBB2
  • ERBB2 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
    • VO ID: VO_0001147
    • Description: Gerbu Adjuvant and recombinant IL-2 (Wagner et al., 2007).
  • Adjuvant:
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0011372
  • Type: Recombinant vector vaccine
  • Status: Research
  • Antigen: CD40 Ligand
  • Cd40lg gene engineering:
    • Type: Recombinant vector construction
    • Description: Murine CD40 ligand (mCD40L) gene was cloned from a c D N A library generated from activated T cells (Coleclough, 1993) using seminested P C R primers that incorporated a Kpn I site at the 5' end of the gene and a Cta I site at the 3' end of the gene using AmpliTaq (Perkin-Elmer, Branchburg, NJ) according to the manufacturer's protocols. The polymerase chain reaction (PCR) product was inserted into p G E M 7 Z (Pharmacia Biotech Inc., Piscataway, NJ) and the correct D N A sequence of a full-length clone as compared to published data (Armitage et al, 1992) was confirmed. The m u C D 4 0 L gene was then subcloned into the Gla retroviral vector derived from Moloney murine leukemia virus (Genetic Therapy, Inc., Gaithersburg, M D ) using the Xho I and Hind II sites (pGla.mCD40L) and used to manufacture a producer cell line (Grossmann et al., 1997).
    • Detailed Gene Information: Click Here.
  • Vector: Gla retroviral vector
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0011510
  • Type: Subunit vaccine
  • Status: Research
  • CEACAM5 (CEA) gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Vector: Dendritic Cells (Bae et al., 2009).
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0011366
  • Type: Subunit vaccine
  • Status: Research
  • CTAG1B gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004227
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: Peptide GF001, comprising the H-2Db-restricted minimal CTL epitope of HPV16 E7 protein (Fernando et al., 1998).
  • E7 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0004228
  • Type: Subunit vaccine
  • Status: Licensed
  • Antigen: Peptide GF001, comprising the H-2Db-restricted minimal CTL epitope of HPV16 E7 protein (Fernando et al., 1998).
  • E7 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0011371
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: VEGFA
  • VEGFA gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Adjuvant:
    • VO ID: VO_0000127
    • Description: Aluminum hydroxide, VSSP, and CAF01
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0004426
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • E2A gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: pVAX (Lindencrona et al., 2004)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004437
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • MUC1 gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pCEP4 expressed MUC1, a transmembrane molecule whose major extracellular domain is composed of tandem repeat units of 20 amino acids (Kamata et al., 2002).
    • Detailed Gene Information: Click Here.
  • Muc1 gene engineering:
  • Vector: pCEP4 (Kamata et al., 2002)
  • Immunization Route: Intradermal injection (i.d.)
  • Vaccine Ontology ID: VO_0004398
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Fragment C from tetanus toxin gene engineering:
    • Type: DNA vaccine construction
    • Description: This vaccine encoded T-cell antigen receptor Valpha, Vbeta, and Fragment C (Thirdborough et al., 2002).
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3 (Thirdborough et al., 2002)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004440
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • HUMGP 75 gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • TRP-2 gene engineering:
    • Type: Recombinant protein preparation
    • Description: This article informs the gene used in the cancer vaccine engineering. (Weber et al., 1998)
    • Detailed Gene Information: Click Here.
  • Vector: WRG/BEN (Weber et al., 1998)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004442
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Antigen: TRP-2
  • TRP-2 gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pCR3 expressed human-tyrosinase-related protein-2 (hTRP2) (Hawkins et al., 2002).
    • Detailed Gene Information: Click Here.
  • Vector: pCR3
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004438
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • gp100 (PMEL) gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pWRG1644 expressed the human melanoma-associated antigen, gp100 (Rakhmilevich et al., 2001).
    • Detailed Gene Information: Click Here.
  • Vector: pWRG1644 (Rakhmilevich et al., 2001)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004433
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • GRP gene engineering:
    • Type: DNA vaccine construction
    • Description: This vaccine encoded six tandem repeats of a fragment of GRP from amino acids 18 to 27 (GRP6) flanked by helper T-cell epitopes for increased immunogenicity, including HSP65, a tetanus toxoid fragment from amino acids 830 to 844 (T), pan-HLA-DR-binding epitope (PADRE) (P), and two repeats of a mycobacterial HSP70 fragment from amino acids 407 to 426 (M) (Fang et al., 2009).
    • Detailed Gene Information: Click Here.
  • Vector: pCR3.1 (Fang et al., 2009)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004435
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • GM-CSF (Mus musculus) gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pNGVL4a expressed GM-CSF and MAGE-1 (Sun et al., 2002).
    • Detailed Gene Information: Click Here.
  • MAGEA1 (MAGE1) gene engineering:
    • Type: Recombinant protein preparation
    • Description: (Sun et al., 2002)
    • Detailed Gene Information: Click Here.
  • Vector: pNGVL4a (Sun et al., 2002)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004434
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • GM-CSF (Mus musculus) gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pNGVL3 expressed GM-CSF and MAGE-1 (Sun et al., 2002).
    • Detailed Gene Information: Click Here.
  • MAGEA1 (MAGE1) gene engineering:
    • Type: Recombinant protein preparation
    • Description: (Sun et al., 2002)
    • Detailed Gene Information: Click Here.
  • Vector: pNGVL3 (Sun et al., 2002)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004443
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Ubiquitin gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Trp2 gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA 3.1 (-). (Zhang et al., 2005)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004436
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Mcam gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector SINCp expressed murine melanoma cell adhesion molecule (MCAM/MUC18) (Leslie et al., 2007).
    • Detailed Gene Information: Click Here.
  • Vector: SINCp (Leslie et al., 2007)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004432
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Survivin gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector VR expressed novel truncations of survivin, is overexpressed in major types of cancer and is considered an ideal ‘‘universal’’ tumor-associated antigen (Zhang et al., 2012).
    • Detailed Gene Information: Click Here.
  • Vector: VR (Zhang et al., 2012)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004439
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Pmel17 gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • Vector: VR1012 (Wagner et al., 2000)
  • Immunization Route: Intradermal injection (i.d.)
  • Vaccine Ontology ID: VO_0004441
  • Type: Recombinant vector vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • TRP-1 gene engineering:
    • Type: Recombinant vector construction
    • Description: Vector pSC65 and rVV expressed tyrosinase-related protein TRP-1 (Overwijk et al., 1999).
    • Detailed Gene Information: Click Here.
  • Vector: pSC65 and recombinant vaccinia virus (Overwijk et al., 1999)
  • Immunization Route: Intravenous injection (i.v.)
  • Vaccine Ontology ID: VO_0004444
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • HuD gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pcDNA 3.1 expressed Hu proteins which are the human homologues of the Drosophila protein elav, including HuD (Carpentier et al., 1998).
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA 3.1 (Carpentier et al., 1998)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004451
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Prostate stem cell antigen gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pcDNA3 expressed prostate stem cell antigen (PSCA) (Garcia-Hernandez et al., 2008).
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3 prime, Venezuelan equine encephalitis virus replicons boost (Garcia-Hernandez et al., 2008)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004445
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Steap1 gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pcDNA3 expressed six-transmembrane epithelial antigen of the prostate (Garcia-Hernandez et al., 2007).
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3 prime, Venezuelan equine encephalitis virus-like replicon particles boost (Garcia-Hernandez et al., 2007)
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0004452
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Prostate stem cell antigen gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pIRES2 DsRed2 expressed murine prostate stem cell antigen (PSCA) (Ahmad et al., 2009).
    • Detailed Gene Information: Click Here.
  • Vector: pIRES2 DsRed2 (Ahmad et al., 2009)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004450
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • HSP47 gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pcDNA3.1(+) expressed heat shock proteins (Zhang et al., 2007).
    • Detailed Gene Information: Click Here.
  • Prostate stem cell antigen gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pcDNA3.1(+) expressed prostate stem cell antigen (PSCA) (Zhang et al., 2007).
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3.1(+) (Zhang et al., 2007)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004449
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Prostate-specific membrane antigen gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • PAP gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • IgG Fc gene engineering:
    • Type: DNA vaccine construction
    • Detailed Gene Information: Click Here.
  • KLK3 gene engineering:
    • Type: Recombinant protein preparation
    • Detailed Gene Information: Click Here.
  • Vector: pcDNA3 (Qin et al., 2005)
  • Immunization Route: Gene gun
  • Vaccine Ontology ID: VO_0004448
  • Type: DNA vaccine
  • Status: Research
  • Host Species as Laboratory Animal Model: Mouse
  • Prostate specific antigen gene engineering:
    • Type: DNA vaccine construction
    • Description: Vector pVAX1 expressed prostate -specific antigen (Roos et al., 2005).
    • Detailed Gene Information: Click Here.
  • Vector: pVAX1 (Roos et al., 2005)
  • Immunization Route: Intramuscular injection (i.m.)
  • Vaccine Ontology ID: VO_0004245
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: Recombinant GA733-2 Ag (Basak et al., 2000).
  • EPCAM gene engineering:
    • Type: Recombinant protein preparation
    • Description: (Basak et al., 2000)
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: subcutaneous injection
  • Vaccine Ontology ID: VO_0004258
  • Type: Subunit vaccine
  • Status: Research
  • Antigen: The 140-aa synthetic MUC1 peptide (Soares et al., 2001).
  • MUC1 gene engineering:
    • Type: Recombinant protein preparation
    • Description: This article lays out the relationship between MUC1 and tumor growth used in cancer vaccines. (Soares et al., 2001)
    • Detailed Gene Information: Click Here.
  • Adjuvant:
  • Immunization Route: subcutaneous injection
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 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

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: To test the efficacy of replicase-based TRP-1 plasmids, gene-gun−immunized mice were challenged subcutaneously with B16 melanoma. This vaccine provided strong tumor protection, with around 50% of mice still tumor-free 21 days after challenge (Leitner et al., 2003).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: To test the efficacy of replicase-based TRP-1 plasmids, gene-gun−immunized mice were challenged subcutaneously with B16 melanoma. This vaccine provided strong tumor protection, with around 60% of mice still tumor-free 21 days after challenge (Leitner et al., 2003).

Mouse Response

  • Efficacy: To test the efficacy of replicase-based TRP-1 plasmids, gene-gun−immunized mice were challenged subcutaneously with B16 melanoma. This vaccine provided strong tumor protection, with around 70% of mice still tumor-free 21 days after challenge (Leitner et al., 2003).

Mouse Response

  • Host Strain: C57BL/6J
  • Vaccination Protocol: Mice were injected i.p. with 0.5 ml irradiated cells 21 and 14 days before challenge, and were injected with loxoribine prior to challenge (Pope et al., 1994).
  • Challenge Protocol: Mice were challenged i.v. with live B16 cells (Pope et al., 1994).
  • Efficacy: Mice treated with loxoribine had significant inhibition of tumor growth following challenge with live B16 cells (Pope et al., 1994).

Mouse Response

  • Host Strain: BALB/c, C57BL/6
  • Vaccination Protocol: The method of ppEDG DNA immunization; mice were injected i.m. in both quadriceps. A 1-ml insulin syringe was used for all injections and each single dose consisted of 100 μg that was diluted in normal saline of a total volume of 100 μl and split between both legs. The method of pEDG protein immunization; mice were injected s.c. and each single dose consisted of 10 μg that was also diluted in normal saline of a total volume of 100 μl. To investigate the protective anti-tumor effects mice at 6 to 8 weeks of age were randomly divided into the following 4 groups of 10 animals each. Group 1 (DP) mice were vaccinated with pEDG and ppEDG simultaneously once weekly for 4 continuous weeks. Group 2 (DD) mice were vaccinated with pEDG alone at the same time-points as in the group 1. Group 3 (PP) mice were vaccinated with ppEDG alone. Group 4 (NS) mice were injected with 100 μl normal saline. (Tan et al., 2007).
  • Immune Response: CTL response against endoglin-positive HUVECs, but not against endoglin-negative tumor cells was found in the mice combined DNA with protein vaccination. In addition, combination of endoglin DNA and recombinant protein vaccination significantly induced IFN-gamma secreting cells (Tan et al., 2007).
  • Challenge Protocol: One week after the last vaccination or saline injection all the experimental mice were subcutaneously injected with 2×10^6 live tumor cells (Tan et al., 2007).
  • Efficacy: The results showed that combination of endoglin DNA and protein vaccines could enhance both protective and therapeutic anti-tumor efficacy in both colon carcinoma and Lewis lung carcinoma models. Significant inhibition of tumor angiogenesis was found in the tumor tissues (Tan et al., 2007).

Mouse Response

  • Host Strain: Balb/C
  • Vaccination Protocol: mice (4 per group) were vaccinated i.m. with 100 μg per mouse of pcDNA3.1 or pcDNA3.1/hNIS in a 100 μl volume. The groups primed with MIDGE/hNIS or MIDGE/hNIS-NLS received 54.8 μg/100 μl per mouse, which is an equimolar concentration of the plasmids. One week later, the mice were boosted with the same amount of DNA (Choi et al., 2007).
  • Challenge Protocol: One week after the final vaccination, the mice were challenged subcutaneously (s.c.) with CT26/NIS tumor cells at 5 × 10^4 cells/mouse in the hind-right legs (Choi et al., 2007).
  • Efficacy: Immunization with the hNIS encoding vectors induced antigen-mediated antitumor activity against NIS-expressing CT26 tumors in vivo, with the highest tumor free rate (100%) and lowest tumor growth being observed up to 40 days after the CT26/NIS tumor challenge with MIDGE/hNIS than those resulting from other immunization groups (Choi et al., 2007).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Immune Response: Vaccination was rendered effective by electroporation, priming higher levels of AH1-specific CD8(+) T cells able to protect mice from tumor growth (Buchan et al., 2005).
  • Efficacy: Delivery in a suboptimal volume (2 × 10 μl) did not mediate protection. However, protective efficacy was completely restored when suboptimal volume was combined with electroporation (p < 0.003). Therefore, this vaccine was able to protect from CT26 tumor in vivo (Buchan et al., 2005).

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: For immunization experiments, mice were injected into the shaved right back with live tumor cells by using the same procedure as described above, and tumor nodules were removed surgically at day 10 (Yang et al., 1995).
  • Challenge Protocol: Two weeks after tumor removal, the mice were challenged into the left back or the frank with wt tumor cells at 1 X 10^5 /mouse (P815) or 2 X 10^5 /mouse (Yang et al., 1995).
  • Efficacy: A plasmid containing murine B7-2 (Cd86) cDNA was transfected into the immunogenic mouse mastocytoma P815 of DBA/2 origin. In contrast to the lethal growth of the wild-type (wt) P815 tumor, B7-2-positive (B7-2+) P815 cells inoculated into syngeneic mice regressed, and immunization of mice with such tumor cells protected them against the challenge of wt P815 tumor (Yang et al., 1995).

Mouse Response

  • Host Strain: C57BL/6
  • Vaccination Protocol: C57BL/6 mice were anesthetized with isoflurane and injected intradermally with 40 μg (40 μl of PBS) of plasmid DNA at two sites (20 μg each) near the base of the tail using a 29-gauge insulin-grade syringe. Mice were immunized two times either at days −21 and −7 (early setting) or at days +10 and +17 (late setting) with respect to tumor challenge, referred as day 0 (Lladser et al., 2010).
  • Immune Response: Intradermal DNA EP of mice with a human survivin encoding plasmid generated CD8+ cytotoxic T lymphocyte (CTL) responses cross-reactive with the mouse epitope surv(20-28), as determined by intracellular IFN-gamma staining, suggesting that self-tolerance has been broken. Survivin-specific CTLs displayed an activated effector phenotype as determined by CD44 and CD107 up-regulation. Vaccinated mice displayed specific cytotoxic activity against B16 and peptide-pulsed RMA-S cells in vitro as well as against surv(20-28) peptide-pulsed target cells in vivo (Lladser et al., 2010).
  • Challenge Protocol: Mice were challenged with a lethal dose of syngeneic B16 melanoma cells (Lladser et al., 2010).
  • Efficacy: Intradermal EP with a survivin DNA vaccine suppressed angiogenesis in vivo and elicited protection against highly aggressive syngeneic B16 melanoma tumor challenge (Lladser et al., 2010).

Mouse Response

  • Vaccination Protocol: Mice were immunized with 100 lg of Ab2 BR3E4 in CFA/IFA on days 0, 12, and 33, or with 5 lg alum-precipitated GA733-2E on days 0, 14, and 28 (Maruyama et al., 2000).
  • Challenge Protocol: Immunized mice (4±6/group) were challenged s.c. with 4 x 10^7 CT26-ALGA710-3H cells expressing the GA733 antigen or with parental, antigen-negative
    CT26 cells 2 weeks after the last immunization. Tumors were measured with a caliper twice each week for up to 2 months after the challenge (Maruyama et al., 2000).
  • Efficacy: The full-length GA733 (epcam) antigen expressed by recombinant adenovirus inhibited the growth of established tumors in mice (Maruyama et al., 2000).

Mouse Response

  • Host Strain: FVB/N, BALB/c
  • Vaccination Protocol: FVB/N transgenic mice spontaneously developing c-neu overexpressing breast cancers were used. Mice were immunized with the combination of the three peptides P4, P6, and P7 coupled to tetanus toxoid (TT-conjugates; BALB/c and FVB/N n = 5/group, MMTV-c-neu trangenic mice n = 8/group) using 15 μg of each peptide conjugate or with co-applicated IL-12 (BALB/c and FVB/N n = 5/group, MMTV-c-neu trangenic mice n = 7/group). Control BALB/c and FVB/N mice (n = 5/group) received TT and control MMTV-c-neu transgenic mice received TT (n = 8) or IL-12 (n = 5) alone or remained unimmunized (n = 8) (Wagner et al., 2007).
  • Efficacy: At the time all untreated mice had developed tumors about 40% of peptide-immunized mice and nearly 60% of mice immunized with the peptide vaccine co-applied with IL-12 remained tumor free (Wagner et al., 2007).

Mouse Response

  • Host Strain: A/J
  • Vaccination Protocol: A/J (H^'') female mice (Jackson Labs) received a single subcutaneous (s.c.) injection consisting of 2 X 10* cells total unless otherwise noted in the figure legends. The cells were either neuro-2a/neo cells ( 0 % C D 4 0 L positive), neuro-2a/CD40L (70% CD40L positive), or neuro-2a/neo cells mixed with various
    numbers of neuro-2a/CD40L cells to the appropriate percentages of CD40L-positive cells (Grossmann et al., 1997).
  • Challenge Protocol: Antitumor effects were tested with subsequent challenge with parental neuro-2a cells (Grossmann et al., 1997).
  • Efficacy: Transgenic expression of the CD40L (Cd40lg) increased immune responses against a weakly immunogenic murine tumor, neuro-2a. Tumor cells were transduced with a retroviral construct containing the CD40L gene and co-injected with variable numbers of non-CD40L transduced cells into syngeneic mice. Mice injected with cells that expressed CD40L had a significant reduction in average tumor size as compared to controls (p < 0.0001). In addition, survival of the neuro-2a/CD40L mice was 48 days versus 34 days for the neuro-2a/neo controls (p < 0.02). Expression of CD40L by less than 1.5% of neuro-2a cells was sufficient for significant antitumor effects (p < 0.001). These antitumor effects protected mice from subsequent challenge with parental neuro-2a cells (Grossmann et al., 1997).

Mouse Response

  • Host Strain: B6
  • Vaccination Protocol: To establish a CEA-positive tumour-bearing mouse model, 6-week-old B6 mice were injected in the right flank with MC38-CEA2 cells (1 × 106) each. At 7 days after tumour cell injection, mice were immunized at the tail base with DCs pulsed with CEA or Tat-CEA proteins (1 × 106 cells/mouse in 100 µl PBS) at weekly intervals for 4 weeks. Control mice were injected with PBS only (Bae et al., 2009).
  • Challenge Protocol: To establish a CEA-positive tumour-bearing mouse model, 6-week-old B6 mice were injected in the right flank with MC38-CEA2 cells (1 × 106) each (Bae et al., 2009).
  • Efficacy: In vivo, the DC (Tat-CEA) vaccine delayed tumour growth significantly and prolonged survival of tumour-bearing mice (Bae et al., 2009).

Mouse Response

  • Host Strain: BALB/c and C57BL/6
  • Vaccination Protocol: Mice were immunized s.c. with 100 μl into the scruff of the neck with the NY-ESO-1 vaccine (5 μg of both NY-ESO-1 and ISCOPREP saponin), or with NY-ESO-1 protein (5 μg of protein) or with the ISCOMATRIX adjuvant (5 μg of ISCOPREP saponin) (Maraskovsky et al., 2004).
  • Immune Response: The NY-ESO-1 vaccine induced strong NY-ESO-1-specific IFN-gamma and IgG2a responses in C57BL/6 mice. Furthermore, the NY-ESO-1 vaccine induced NY-ESO-1-specific CD8(+) CTLs in HLA-A2 transgenic mice that were capable of lysing human HLA-A2(+) NY-ESO-1(+) tumor cells (Maraskovsky et al., 2004).
  • Challenge Protocol: B16 melanoma cells were transfected using electroporation with the mammalian expression plasmid, pCDNA3, encoding the cDNA for NY-ESO-1 (Invitrogen, Carlsbad, CA). Selection with G418 (800 μg/ml) and limit-dilution cloning yielded a clone expressing NY-ESO-1 (B16-NY-ESO-1) as determined by IHC and quantitative real-time PCR. C57BL/6 mice were vaccinated twice (at 0 and 4 weeks) with the NY-ESO-1 vaccine, or with the ISCOMATRIX adjuvant alone as a control. Four weeks after the second immunization, mice were challenged with B16-NY-ESO-1. The tumor cells (1 × 10^4) were injected s.c. on the back, and tumor volume was measured over time (Maraskovsky et al., 2004).
  • Efficacy: C57BL/6 mice, immunized with the NY-ESO-1 vaccine, were protected against challenge with a B16 melanoma cell line expressing NY-ESO-1 (Maraskovsky et al., 2004).

Mouse Response

  • Host Strain: C57BL/6J
  • Vaccination Protocol: Mice (8 to 10 per group) were immunized s.c. at the base of the tail with 50 µg of E7GST protein, or 50 µg OVA as control, and 10 µg of Quil-A or 50 µg of Algammulin as adjuvant (Fernando et al., 1998).
  • Challenge Protocol: Mice were challenged with 3 x 10^6 cells of EL4.E7 tumor or 2 x 10^6 cells of C3 cells 14 days after the last immunization (Fernando et al., 1998).
  • Efficacy: Immunization with E7 and Algammulin (an alum-based adjuvant) induced a Th2-like response and provided no tumor protection (Fernando et al., 1998).

Mouse Response

  • Host Strain: C57BL/6J
  • Vaccination Protocol: Mice (8 to 10 per group) were immunized s.c. at the base of the tail with 50 µg of E7GST protein, or 50 µg OVA as control, and 10 µg of Quil-A or 50 µg of Algammulin as adjuvant (Fernando et al., 1998).
  • Challenge Protocol: Mice were challenged with 3 x 10^6 cells of EL4.E7 tumor or 2 x 10^6 cells of C3 cells 14 days after the last immunization (Fernando et al., 1998).
  • Efficacy: Growth of EL4.E7 was reduced following immunization with E7 and Quil-A (an adjuvant that induced a Th1-type response to E7) or with GF001 and Quil-A (Fernando et al., 1998).

Mouse Response

  • Host Strain: C57BL/6
  • Vaccination Protocol: Immunization was done subcutaneously (sc) with 100 μg antigen in 200 μl of the antigen-adjuvant preparation. The immunization scheme included weekly immunizations for six consecutive weeks. Control mice only received vehicle or adjuvant (Morera et al., 2008).
  • Challenge Protocol: Animals were challenged with a sc injection of 2 × 104 B16-F10 melanoma cells, 4 days after the third immunization (Morera et al., 2008).
  • Efficacy: A protein vaccine candidate, based on a human modified VEGF antigen that is expressed at high levels in E. coli was developed. With respect to controls, immunization experiments in C57BL/6 mice using weekly doses of this antigen and three adjuvants of different chemical natures show that time for tumor development after subcutaneous injection of Melanoma B16-F10 cells increases, tumors that develop grow slower, and overall animal survival is higher. Immunization also prevents tumor development in some mice, making them resistant to second tumor challenges. Vaccination of mice with the human modified VEGF recombinant antigen produces antibodies against the human antigen and the homologous mouse VEGF molecule. We also show that sera from immunized mice block human VEGF-induced HUVEC proliferation (Morera et al., 2008).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Eighty percent of B cell competent μMT+/− mice immunized with pVAX/E2A and GM-CSF plasmids were protected from HER-2 expressing tumor challenge. All mice challenged with the HER-2 negative untransfected control line developed tumors. Of note, protection was equal or better in B cell deficient animals, as 90% of the μMT−/− mice that were immunized with pVAX/E2A and GM-CSF plasmids rejected the tumor. All mice immunized with pVAX/E2A without the GM-CSF plasmid developed tumors. Thus, this HER-2 specific resistance to tumor challenge is entirely dependent on co-administration of the GM-CSF plasmid. None of the μMT−/− mice immunized with pVAX/E2A or the combination of GM-CSF and pVAX/E2A were protected from challenge with the HER-2 negative D2F2 control line (Lindencrona et al., 2004).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: The number of lung metastatic nodules three weeks after inoculation of F10-MUC1-C8 cells was significantly lower in mice immunized with pCEP4-MUC1 DNA vaccine 3 times at weekly intervals than in mice immunized with the vector DNA alone or with a single immunization of the DNA vaccine (Kamata et al., 2002).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Vaccination with the DNA construct VαVβCβ-FrC induced strong protection against challenge with C6VL, compared with nonvaccinated control mice and mice vaccinated with FrC alone (P < 0.001). Vaccinations with VαVβ-FrC, VβCβ-FrC, or VαVβCβ alone were also completely ineffective. The protective response generated was specific for C6VL, with no protection induced against TCL-1, an unrelated T-cell tumor. The pattern of protection has been confirmed in two subsequent experiments, with survival rates of mice vaccinated with VαVβCβ-FrC ranging from 50 to 80%, whereas the other constructs were ineffective at prolonging survival (Thirdborough et al., 2002).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Mice immunized with hgp75 were significantly protected from lung metastases compared with control mice (mice immunized with a null vector). There was an 86% decrease in lung nodules (Weber et al., 1998).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Immunization of mice with xenogeneic hTRP2 DNA results in tumor immunity against intravenous tumor challenge with a syngeneic mouse B16F10LM3 melanoma known to express TRP2. All mice (100%) immunized with hTRP2 were protected from challenge with B16F10LM3 melanoma while all control PCR3 vector immunized mice (100%) developed tumors (P < 0.0001). However, growth of established tumors was not inhibited (Hawkins et al., 2002).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Particle-mediated delivery of the gp100 plasmid resulted in substantial protection against B16-gp100 tumors, with 40% of the mice remaining tumor free for at least 2 months. Importantly, co-delivery of mGM-CSF DNA with hugp100 DNA resulted in complete tumor protection in all five vaccinated mice (Rakhmilevich et al., 2001).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: B16-F10 cells injected subcutaneously formed large solid tumors in nonimmunized mice (saline) or in mice injected with a non-GRP-containing plasmid (pCR3.1-VS-HSP65-TP-M2). The tumor sizes decreased progressively in mice immunized with the anti-GRP vaccine (pCR3.1-VS-HSP65-TP-GRP6-M2). B16-F10 tumor cells were implanted intradermally at two sites in the abdominal region. It took approximately 7 days for the cells to form ∼4-mm intradermal tumors in the two control groups; however, the growth of intradermal tumors in pCR3.1-VS-HSP65-TP-GRP6-M2-immunized group was slightly delayed and required almost 11 days to form ∼4-mm tumors. The total number of blood vessels around each implant site from pCR3.1-VS-HSP65-TP-GRP6-M2-immunized mice was significantly lower than that from the saline group (22 ± 4 versus 72 ± 14; P < 0.01) or from non-GRP-containing plasmid-immunized mice (22 ± 4 versus 63 ± 19; P < 0.01). To further test the efficacy of the anti-GRP vaccine, the extent of lung metastasis by intravenously administered tumor cells in the tail vein of immunized mice was evaluated. Metastatic tumor nodules were often detected in the lungs 21 days after injection of tumor cells. The average weight of lungs of mice immunized with pCR3.1-VS-HSP65-TP-GRP6-M2 was significantly lower than that of the saline group (0.215 ± 0.020 g versus 0.301 ± 0.068 g; P < 0.05) or the pCR3.1-VS-HSP65-TP-M2 control group (0.215 ± 0.020 g versus 0.289 ± 0.087 g; P < 0.05), which indicates that fewer metastases were formed in the lungs of the anti-GRP DNA vaccine-immunized group. In addition, the average number of metastatic nodules in mice immunized with pCR3.1-VS-HSP65-TP-GRP6-M2 was significantly less than that in mice that received saline (37.2 ± 9.4 versus 88.0 ± 22.6; P < 0.001) or in mice in the pCR3.1-VS-HSP65-TP-M2 control group (37.2 ± 9.4 versus 79.3.0 ± 16.8; P < 0.001) (Fang et al., 2009).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: C57BL/6 mice were immunized on days 0, 7, and 17 with the different MAGE-1 plasmids. After 6 days, the mice were challenged with B16 melanoma cells that express the human MAGE-1 antigen. Mice immunized with plasmids co-expressing MAGE-1 and GM-CSF had significantly fewer tumor colonies on their lungs than mice from any of the other immunization groups. In fact, 60% of the mice immunized with DNA vaccines that co-express MAGE-1 and GM-CSF had fewer than five tumor colonies on their lungs, whereas this was true of a little less than 20% of the mice immunized with MAGE-1 alone vaccines or co-inoculation with MAGE-1 vaccines and GM-CSF expressing plasmid (Sun et al., 2002).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: C57BL/6 mice were immunized on days 0, 7, and 17 with the different MAGE-1 plasmids. After 6 days, the mice were challenged with B16 melanoma cells that express the human MAGE-1 antigen. Mice immunized with plasmids co-expressing MAGE-1 and GM-CSF had significantly fewer tumor colonies on their lungs than mice from any of the other immunization groups. In fact, 60% of the mice immunized with DNA vaccines that co-express MAGE-1 and GM-CSF had fewer than five tumor colonies on their lungs, whereas this was true of a little less than 20% of the mice immunized with MAGE-1 alone vaccines or co-inoculation with MAGE-1 vaccines and GM-CSF expressing plasmid (Sun et al., 2002).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: A week after the last immunization, mice were challenged subcutaneously with 2 105 B16F1 cells. Control mice and mice immunized with pTRP-2 showed rapid tumor growth, whereas 85% of mice immunized with pUB-TRP-2 were free of tumors and the remaining 15% of the mice exhibited almost complete suppression of tumor growth. Strictly, all mice immunized with pUB-TRP-2 survived over 80 days after implantation of tumor cells, although all mice of the other two groups died within 60 days. Furthermore, immunization with pUB-TRP-2 was also effective in suppressing the growth of B16F10 melanoma cells, a more virulent type of melanoma (Zhang et al., 2005).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Mice vaccinated with SINCp c-muMUC18 had significantly (P=0.037) fewer lung colonies than mice vaccinated with SINCp -gal. The number of lung metastases was reduced in mice vaccinated with SINCp c-muMUC18 (range 0–100; median 1.0) compared with SINCp -gal (range 18–200; median 26). Thus, mice immunized with SINCp c-muMUC18 were significantly protected from lung metastasis formation (Leslie et al., 2007).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Tumors were significantly inhibited in mice boosted with AD-S8 compared with those in mice given VR-S8/VR- IL2 (P < 0.01). Moreover, complete tumor rejection occurred in 5 of 15 mice, while no tumor rejection was observed in the control group. A Kaplan–Meier plot showed that 53.3% of the mice in the AD-S8- boosted group were alive at 50 days with survival prolonged by 35.6%, whereas only 6.7% of the mice in the VR-S8/VR-IL2 and VR-S8 groups were alive with survival prolonged by 17 and 10.5%, respectively (Zhang et al., 2012).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: More than half of the animals that had received VR1012/mPmel17 pDNA were protected completely (eight of 15 = 53.3%) against the highly tumorigenic dose of 1X10^5 M3-7 cells. None of these protected animals developed a tumor at a later time point during an entire observation period of 4 months. By contrast, control animals that had been injected with vector VR1012 pDNA alone consistently failed to reject Pmel17high M3-7 melanoma cells (Wagner et al., 2000).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Challenge of vaccinated β2m knockout mice with B16 melanoma revealed robust protection against melanoma growth (Overwijk et al., 1999).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Immunized animals were challenged by s.c. implantation of a neuroblastoma cell line that constitutively expresses HuD. When compared with controls, mice immunized with the secreted HuD showed significant tumor growth inhibition (51% reduction volume; P = 0.0012), and 14% of them had complete tumor rejection (Carpentier et al., 1998).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Eight-week-old TRAMP mice displaying prostate intraepithelial neoplasia were vaccinated with a heterologous prime/boost strategy consisting of gene gun-delivered PSCA-cDNA followed by Venezuelan equine encephalitis virus replicons encoding PSCA. PSCA-vaccinated TRAMP mice had a 90% survival rate at 12 months of age. In contrast, all control mice had succumbed to prostate cancer or had heavy tumor loads (Garcia-Hernandez et al., 2008).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Survival was significantly prolonged in mice vaccinated with mSTEAP using DNA, VRP (Venezuelan equine encephalitis virus-like replicon particles), or a combined strategy after tumor challenge compared with control mice. Although all STEAP-based vaccination strategies significantly slowed tumor growth in tumor-challenged mice, DNA vaccination followed by boosting with VRP was the most effective way of inducing protective immunity (Garcia-Hernandez et al., 2007).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: These experiments demonstrated 37% relative risk reduction of tumor development in the pmPSCA group, but importantly vaccinated tumor bearing mice also had significantly less tumor burden than the control groups. Tumor growth kinetics indicated slower tumor growth in the pmPSCA treated group (versus empty vector P = 0.04, versus untreated P = 0.01). These results demonstrated that the pmPSCA could provide either complete protection or result in containment of the disease (Ahmad et al., 2009).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: The growth of RM-PSCA tumors was significantly slower in the groups vaccinated with PSCA-HSP, HSP-PSCA, and PSCA + HSP plasmids compared with that in the groups treated with PBS or pcDNA-HSP (P < 0.05). Furthermore, the survival time of mice vaccinated with PSCA-HSP (range 40–69 days) was significantly longer as compared with that of groups injected with HSP-PSCA and PSCA + HSP plasmids (P = 0.016 and P = 0.015, respectively) (Zhang et al., 2007).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: Vaccination with psig-3P-Fc by gene gun inoculation induced strong antitumor response in a mouse tumor model, which significantly inhibited tumor growth and prolonged survival time of the tumor-bearing mice (Qin et al., 2005).

Mouse Response

  • Vaccine Immune Response Type: VO_0000286
  • Efficacy: The pVax-PSA vaccine alone protected 40% of the mice from tumor growth. When either of the cytokine adjuvants (IL-2 or GM-CSF) was co-administered 60% of the mice were protected and when both adjuvants were simultaneously co-administered 80% of the mice were protected. All groups of mice that received the vaccine pVax-PSA (with or without cytokine adjuvants) were significantly different from the control group pVax (P < 0.01) (Roos et al., 2005).

Mouse Response

  • Host Strain: BALB/c
  • Vaccination Protocol: BALB/c mice were immunized s.c. with BV GA733-2F (full-length) proteins (25 μg) in theramide adjuvant (25 μg), 3× at 3-week intervals (Basak et al., 2000).
  • Challenge Protocol: Seventeen days after the last immunization mice were challenged with 2.5 × 10^7 CT26-GA733 tumor cells (Basak et al., 2000).
  • Efficacy: 1 out of 7 mice was tumor free after immunization and challenge with tumor cells (Basak et al., 2000).

Mouse Response

  • Host Strain: MUC1-Tg mice (4–6 wk old) on a C57BL/6 background
  • Vaccination Protocol: Three different immunization protocols were tested in vivo. Mice were immunized with: 1) synthetic MUC1 peptide (100 µg/mouse) coadministered with soluble murine GM-CSF (2 µg/mouse; a generous gift from Immunex, Seattle, WA) injected s.c.; 2) synthetic MUC1 peptide (100 µg/mouse) coadministered with SB-AS2 (50 µl/mouse; a generous gift of SmithKline Beecham Biologicals, Rixensart, Belgium) injected i.m.; or 3) murine DC prepulsed with 20 µg/ml of synthetic MUC1 peptide in AIM-V medium (Life Technologies, Grand Island, NY) overnight (2–5 x 104 DC/mouse injected s.c.) (Soares et al., 2001).
  • Challenge Protocol: Ten days after the last boost, the mice were anesthetized with Metofane (Schering-Plough Animal Health, Omaha, NE) and 5 x 10^4 RMA-MUC1 cells injected s.c. in the shaved right hind flank. Tumor growth was monitored every 2–3 days and tumor size determined with calipers (Soares et al., 2001).
  • Efficacy: WT and MUC1-Tg mice that had been immunized with MUC1 peptide admixed with either GM-CSF or SB-AS2 also had to be sacrificed because they failed to reject the tumors (Soares et al., 2001).
References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References References
Syrengelas et al., 1996: Syrengelas AD, Chen TT, Levy R. DNA immunization induces protective immunity against B-cell lymphoma. Nature medicine. 1996; 2(9); 1038-1041. [PubMed: 8782465].
Timmerman et al., 2002: Timmerman JM, Singh G, Hermanson G, Hobart P, Czerwinski DK, Taidi B, Rajapaksa R, Caspar CB, Van Beckhoven A, Levy R. Immunogenicity of a plasmid DNA vaccine encoding chimeric idiotype in patients with B-cell lymphoma. Cancer research. 2002; 62(20); 5845-5852. [PubMed: 12384547].
Leitner et al., 2003: Leitner WW, Hwang LN, deVeer MJ, Zhou A, Silverman RH, Williams BR, Dubensky TW, Ying H, Restifo NP. Alphavirus-based DNA vaccine breaks immunological tolerance by activating innate antiviral pathways. Nature medicine. 2003; 9(1); 33-39. [PubMed: 12496961].
Leitner et al., 2003: Leitner WW, Hwang LN, deVeer MJ, Zhou A, Silverman RH, Williams BR, Dubensky TW, Ying H, Restifo NP. Alphavirus-based DNA vaccine breaks immunological tolerance by activating innate antiviral pathways. Nature medicine. 2003; 9(1); 33-39. [PubMed: 12496961].
Leitner et al., 2003: Leitner WW, Hwang LN, deVeer MJ, Zhou A, Silverman RH, Williams BR, Dubensky TW, Ying H, Restifo NP. Alphavirus-based DNA vaccine breaks immunological tolerance by activating innate antiviral pathways. Nature medicine. 2003; 9(1); 33-39. [PubMed: 12496961].
Pope et al., 1994: Pope BL, Sigindere J, Chourmouzis E, MacIntyre P, Goodman MG. 7-Allyl-8-oxoguanosine (loxoribine) inhibits the metastasis of B16 melanoma cells and has adjuvant activity in mice immunized with a B16 tumor vaccine. Cancer immunology, immunotherapy : CII. 1994; 38(2); 83-91. [PubMed: 8306370].
Tan et al., 2007: Tan GH, Li YN, Huang FY, Wang H, Bai RZ, Jang J. Combination of recombinant xenogeneic endoglin DNA and protein vaccination enhances anti-tumor effects. Immunological investigations. 2007; 36(4); 423-440. [PubMed: 17691024].
Choi et al., 2007: Choi Y, Jeon YH, Kang JH, Chung JK, Schmidt M, Kim AC. MIDGE/hNIS vaccination generates antigen-associated CD8+IFN-gamma+ T cells and enhances protective antitumor immunity. International journal of cancer. Journal international du cancer. 2007; 120(9); 1942-1950. [PubMed: 17266027].
Buchan et al., 2005: Buchan S, Grønevik E, Mathiesen I, King CA, Stevenson FK, Rice J. Electroporation as a "prime/boost" strategy for naked DNA vaccination against a tumor antigen. Journal of immunology (Baltimore, Md. : 1950). 2005; 174(10); 6292-6298. [PubMed: 15879128].
Yang et al., 1995: Yang G, Hellström KE, Hellström I, Chen L. Antitumor immunity elicited by tumor cells transfected with B7-2, a second ligand for CD28/CTLA-4 costimulatory molecules. Journal of immunology (Baltimore, Md. : 1950). 1995; 154(6); 2794-2800. [PubMed: 7533183].
Lladser et al., 2010: Lladser A, Ljungberg K, Tufvesson H, Tazzari M, Roos AK, Quest AF, Kiessling R. Intradermal DNA electroporation induces survivin-specific CTLs, suppresses angiogenesis and confers protection against mouse melanoma. Cancer immunology, immunotherapy : CII. 2010; 59(1); 81-92. [PubMed: 19526360].
Maruyama et al., 2000: Maruyama H, Zaloudik J, Li W, Sperlagh M, Koido T, Somasundaram R, Scheck S, Prewett M, Herlyn D. Cancer vaccines: single-epitope anti-idiotype vaccine versus multiple-epitope antigen vaccine. Cancer immunology, immunotherapy : CII. 2000; 49(3); 123-132. [PubMed: 10881691].
Wagner et al., 2007: Wagner S, Jasinska J, Breiteneder H, Kundi M, Pehamberger H, Scheiner O, Zielinski CC, Wiedermann U. Delayed tumor onset and reduced tumor growth progression after immunization with a Her-2/neu multi-peptide vaccine and IL-12 in c-neu transgenic mice. Breast cancer research and treatment. 2007; 106(1); 29-38. [PubMed: 17203384].
Grossmann et al., 1997: Grossmann ME, Brown MP, Brenner MK. Antitumor responses induced by transgenic expression of CD40 ligand. Human gene therapy. 1997; 8(16); 1935-1943. [PubMed: 9382959].
Bae et al., 2009: Bae MY, Cho NH, Seong SY. Protective anti-tumour immune responses by murine dendritic cells pulsed with recombinant Tat-carcinoembryonic antigen derived from Escherichia coli. Clinical and experimental immunology. 2009; 157(1); 128-138. [PubMed: 19659778].
Maraskovsky et al., 2004: Maraskovsky E, Sjölander S, Drane DP, Schnurr M, Le TT, Mateo L, Luft T, Masterman KA, Tai TY, Chen Q, Green S, Sjölander A, Pearse MJ, Lemonnier FA, Chen W, Cebon J, Suhrbier A. NY-ESO-1 protein formulated in ISCOMATRIX adjuvant is a potent anticancer vaccine inducing both humoral and CD8+ t-cell-mediated immunity and protection against NY-ESO-1+ tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2004; 10(8); 2879-2890. [PubMed: 15102697].
Fernando et al., 1998: Fernando GJ, Stewart TJ, Tindle RW, Frazer IH. Th2-type CD4+ cells neither enhance nor suppress antitumor CTL activity in a mouse tumor model. Journal of immunology (Baltimore, Md. : 1950). 1998; 161(5); 2421-2427. [PubMed: 9725239].
Fernando et al., 1998: Fernando GJ, Stewart TJ, Tindle RW, Frazer IH. Th2-type CD4+ cells neither enhance nor suppress antitumor CTL activity in a mouse tumor model. Journal of immunology (Baltimore, Md. : 1950). 1998; 161(5); 2421-2427. [PubMed: 9725239].
Morera et al., 2008: Morera Y, Bequet-Romero M, Ayala M, Lamdán H, Agger EM, Andersen P, Gavilondo JV. Anti-tumoral effect of active immunotherapy in C57BL/6 mice using a recombinant human VEGF protein as antigen and three chemically unrelated adjuvants. Angiogenesis. 2008; 11(4); 381-393. [PubMed: 19034678].
Lindencrona et al., 2004: Lindencrona JA, Preiss S, Kammertoens T, Schüler T, Piechocki M, Wei WZ, Seliger B, Blankenstein T, Kiessling R. CD4+ T cell-mediated HER-2/neu-specific tumor rejection in the absence of B cells. International journal of cancer. Journal international du cancer. 2004; 109(2); 259-264. [PubMed: 14750178].
Kamata et al., 2002: Kamata M, Denda-Nagai K, Kubota N, Aida S, Takeda K, Irimura T. Vaccination of mice with MUC1 cDNA suppresses the development of lung metastases. Clinical & experimental metastasis. 2002; 19(8); 689-696. [PubMed: 12553374].
Thirdborough et al., 2002: Thirdborough SM, Radcliffe JN, Friedmann PS, Stevenson FK. Vaccination with DNA encoding a single-chain TCR fusion protein induces anticlonotypic immunity and protects against T-cell lymphoma. Cancer research. 2002; 62(6); 1757-1760. [PubMed: 11912151].
Weber et al., 1998: Weber LW, Bowne WB, Wolchok JD, Srinivasan R, Qin J, Moroi Y, Clynes R, Song P, Lewis JJ, Houghton AN. Tumor immunity and autoimmunity induced by immunization with homologous DNA. The Journal of clinical investigation. 1998; 102(6); 1258-1264. [PubMed: 9739060].
Hawkins et al., 2002: Hawkins WG, Gold JS, Blachere NE, Bowne WB, Hoos A, Lewis JJ, Houghton AN. Xenogeneic DNA immunization in melanoma models for minimal residual disease. The Journal of surgical research. 2002; 102(2); 137-143. [PubMed: 11796010].
Rakhmilevich et al., 2001: Rakhmilevich AL, Imboden M, Hao Z, Macklin MD, Roberts T, Wright KM, Albertini MR, Yang NS, Sondel PM. Effective particle-mediated vaccination against mouse melanoma by coadministration of plasmid DNA encoding Gp100 and granulocyte-macrophage colony-stimulating factor. Clinical cancer research : an official journal of the American Association for Cancer Research. 2001; 7(4); 952-961. [PubMed: 11309346].
Fang et al., 2009: Fang J, Lu Y, Ouyang K, Wu G, Zhang H, Liu Y, Chen Y, Lin M, Wang H, Jin L, Cao R, Roque RS, Zong L, Liu J, Li T. Specific antibodies elicited by a novel DNA vaccine targeting gastrin-releasing peptide inhibit murine melanoma growth in vivo. Clinical and vaccine immunology : CVI. 2009; 16(7); 1033-1039. [PubMed: 19458203].
Sun et al., 2002: Sun X, Hodge LM, Jones HP, Tabor L, Simecka JW. Co-expression of granulocyte-macrophage colony-stimulating factor with antigen enhances humoral and tumor immunity after DNA vaccination. Vaccine. 2002; 20(9-10); 1466-1474. [PubMed: 11818167].
Sun et al., 2002: Sun X, Hodge LM, Jones HP, Tabor L, Simecka JW. Co-expression of granulocyte-macrophage colony-stimulating factor with antigen enhances humoral and tumor immunity after DNA vaccination. Vaccine. 2002; 20(9-10); 1466-1474. [PubMed: 11818167].
Zhang et al., 2005: Zhang M, Obata C, Hisaeda H, Ishii K, Murata S, Chiba T, Tanaka K, Li Y, Furue M, Chou B, Imai T, Duan X, Himeno K. A novel DNA vaccine based on ubiquitin-proteasome pathway targeting 'self'-antigens expressed in melanoma/melanocyte. Gene therapy. 2005; 12(13); 1049-1057. [PubMed: 15800663].
Leslie et al., 2007: Leslie MC, Zhao YJ, Lachman LB, Hwu P, Wu GJ, Bar-Eli M. Immunization against MUC18/MCAM, a novel antigen that drives melanoma invasion and metastasis. Gene therapy. 2007; 14(4); 316-323. [PubMed: 17024104].
Zhang et al., 2012: Zhang H, Wang Y, Liu C, Zhang L, Xia Q, Zhang Y, Wu J, Jiang C, Chen Y, Wu Y, Zha X, Yu X, Kong W. DNA and adenovirus tumor vaccine expressing truncated survivin generates specific immune responses and anti-tumor effects in a murine melanoma model. Cancer immunology, immunotherapy : CII. 2012; ; . [PubMed: 22706381].
Wagner et al., 2000: Wagner SN, Wagner C, Lührs P, Weimann TK, Kutil R, Goos M, Stingl G, Schneeberger A. Intracutaneous genetic immunization with autologous melanoma-associated antigen Pmel17/gp100 induces T cell-mediated tumor protection in vivo. The Journal of investigative dermatology. 2000; 115(6); 1082-1087. [PubMed: 11121145].
Overwijk et al., 1999: Overwijk WW, Lee DS, Surman DR, Irvine KR, Touloukian CE, Chan CC, Carroll MW, Moss B, Rosenberg SA, Restifo NP. Vaccination with a recombinant vaccinia virus encoding a "self" antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4(+) T lymphocytes. Proceedings of the National Academy of Sciences of the United States of America. 1999; 96(6); 2982-2987. [PubMed: 10077623].
Carpentier et al., 1998: Carpentier AF, Rosenfeld MR, Delattre JY, Whalen RG, Posner JB, Dalmau J. DNA vaccination with HuD inhibits growth of a neuroblastoma in mice. Clinical cancer research : an official journal of the American Association for Cancer Research. 1998; 4(11); 2819-2824. [PubMed: 9829748].
Garcia-Hernandez et al., 2008: Garcia-Hernandez Mde L, Gray A, Hubby B, Klinger OJ, Kast WM. Prostate stem cell antigen vaccination induces a long-term protective immune response against prostate cancer in the absence of autoimmunity. Cancer research. 2008; 68(3); 861-869. [PubMed: 18245488].
Garcia-Hernandez et al., 2007: Garcia-Hernandez Mde L, Gray A, Hubby B, Kast WM. In vivo effects of vaccination with six-transmembrane epithelial antigen of the prostate: a candidate antigen for treating prostate cancer. Cancer research. 2007; 67(3); 1344-1351. [PubMed: 17283172].
Ahmad et al., 2009: Ahmad S, Casey G, Sweeney P, Tangney M, O'Sullivan GC. Prostate stem cell antigen DNA vaccination breaks tolerance to self-antigen and inhibits prostate cancer growth. Molecular therapy : the journal of the American Society of Gene Therapy. 2009; 17(6); 1101-1108. [PubMed: 19337234].
Zhang et al., 2007: Zhang X, Yu C, Zhao J, Fu L, Yi S, Liu S, Yu T, Chen W. Vaccination with a DNA vaccine based on human PSCA and HSP70 adjuvant enhances the antigen-specific CD8+ T-cell response and inhibits the PSCA+ tumors growth in mice. The journal of gene medicine. 2007; 9(8); 715-726. [PubMed: 17595048].
Qin et al., 2005: Qin H, Zhou C, Wang D, Ma W, Liang X, Lin C, Zhang Y, Zhang S. Specific antitumor immune response induced by a novel DNA vaccine composed of multiple CTL and T helper cell epitopes of prostate cancer associated antigens. Immunology letters. 2005; 99(1); 85-93. [PubMed: 15894116].
Roos et al., 2005: Roos AK, Pavlenko M, Charo J, Egevad L, Pisa P. Induction of PSA-specific CTLs and anti-tumor immunity by a genetic prostate cancer vaccine. The Prostate. 2005; 62(3); 217-223. [PubMed: 15389792].
Basak et al., 2000: Basak S, Eck S, Gutzmer R, Smith AJ, Birebent B, Purev E, Staib L, Somasundaram R, Zaloudik J, Li W, Jacob L, Mitchell E, Speicher D, Herlyn D. Colorectal cancer vaccines: antiidiotypic antibody, recombinant protein, and viral vector. Annals of the New York Academy of Sciences. 2000; 910; 237-252. [PubMed: 10911917].
Soares et al., 2001: Soares MM, Mehta V, Finn OJ. Three different vaccines based on the 140-amino acid MUC1 peptide with seven tandemly repeated tumor-specific epitopes elicit distinct immune effector mechanisms in wild-type versus MUC1-transgenic mice with different potential for tumor rejection. Journal of immunology (Baltimore, Md. : 1950). 2001; 166(11); 6555-6563. [PubMed: 11359807].