Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 64(10): 1215-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26076666

ABSTRACT

The MYCN oncogene is a strong genetic marker associated with poor prognosis in neuroblastoma (NB). Therefore, MYCN gene amplification and subsequent overexpression provide a possible target for new treatment approaches in NB. We first identified an inverse correlation of MYCN expression with CD45 mRNA in 101 NB tumor samples. KEGG mapping further revealed that MYCN expression was associated with immune-suppressive pathways characterized by a down-regulation of T cell activation and up-regulation of T cell inhibitory gene transcripts. We then aimed to investigate whether DNA vaccination against MYCN is effective to induce an antigen-specific and T cell-mediated immune response. For this purpose, we generated a MYCN-expressing syngeneic mouse model by MYCN gene transfer to NXS2 cells. MYCN-DNA vaccines were engineered based on the pCMV-F3Ub plasmid backbone to drive ubiquitinated full-length MYCN-cDNA and minigene expression. Vaccines were delivered orally with attenuated S. typhimurium strain SL7207 as a carrier. Immunization with both MYCN-DNA vaccines significantly reduced primary tumor growth of MYCN-expressing NB cells in contrast to negative controls. The immune response was mediated by tumor-infiltrating T cells in vivo, which revealed MYCN-specific and MHC class I-restricted lysis of inducible MYCN-expressing NB target cells in vitro. Finally, these antigen-specific T cells also killed MYCN-negative mammary carcinoma cells pulsed with MYCN peptides in contrast to controls. In summary, we demonstrate proof of concept that MYCN can be targeted by DNA vaccination, which may provide an approach to overcoming MYCN immune-suppressive activities in patients with MYCN-amplified disease.


Subject(s)
Carcinoma/immunology , Epitopes, B-Lymphocyte/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Animal/immunology , Neuroblastoma/immunology , Proto-Oncogene Proteins/metabolism , Salmonella Vaccines/administration & dosage , Salmonella typhimurium/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, DNA/administration & dosage , Administration, Oral , Animals , Carcinoma/microbiology , Cell Line, Tumor , Cytotoxicity, Immunologic , Epitopes, B-Lymphocyte/genetics , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/microbiology , Mice , Mice, Inbred Strains , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neoplasms, Experimental , Neuroblastoma/genetics , Neuroblastoma/microbiology , Peptide Fragments , Proto-Oncogene Proteins/genetics , Transgenes/genetics , Tumor Burden , Vaccination
2.
Cancer Immunol Immunother ; 64(5): 621-34, 2015 May.
Article in English | MEDLINE | ID: mdl-25711293

ABSTRACT

The disialoganglioside GD2 is a well-established target antigen for passive immunotherapy in neuroblastoma (NB). Despite the recent success of passive immunotherapy with the anti-GD2 antibody ch14.18 and cytokines, treatment of high-risk NB remains challenging. We expanded the approach of GD2-specific, antibody-based immunotherapy to an application of a GD2-specific natural killer (NK) cell line, NK-92-scFv(ch14.18)-zeta. NK-92-scFv(ch14.18)-zeta is genetically engineered to express a GD2-specific chimeric antigen receptor generated from ch14.18. Here, we show that chimeric receptor expression enables NK-92-scFv(ch14.18)-zeta to effectively lyse GD2(+) NB cells also including partially or multidrug-resistant lines. Our data suggest that recognition of GD2 by the chimeric receptor is the primary mechanism involved in NK-92-scFv(ch14.18)-zeta-mediated lysis and is independent of activating NK cell receptor/ligand interactions. Furthermore, we demonstrate that NK-92-scFv(ch14.18)-zeta is able to mediate a significant anti-tumor response in vivo in a drug-resistant GD2(+) NB xenograft mouse model. NK-92-scFv(ch14.18)-zeta is an NB-specific NK cell line that has potential for future clinical development due to its high stability and activity toward GD2(+) NB cell lines.


Subject(s)
Drug Resistance, Neoplasm , Gangliosides/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Neuroblastoma/therapy , Animals , Antibodies, Anti-Idiotypic/immunology , Cell Line , Cytotoxicity, Immunologic/immunology , Female , Gangliosides/genetics , Genetic Engineering , Humans , Mice , Mice, Inbred NOD , Neoplasm Transplantation , Neuroblastoma/immunology , Receptors, Antigen/biosynthesis , Receptors, Antigen/immunology , Single-Chain Antibodies/immunology
3.
Cancer Immunol Immunother ; 62(6): 999-1010, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23591980

ABSTRACT

PURPOSE: Immunotherapy targeting disialoganglioside GD(2) emerges as an important treatment option for neuroblastoma, a pediatric malignancy characterized by poor outcome. Here, we report the induction of a GD(2)-specific immune response with ganglidiomab, a new anti-idiotype antibody to anti-GD(2) antibodies of the 14.18 family. EXPERIMENTAL DESIGN AND RESULTS: Ganglidiomab was generated following immunization of Balb/c mice with 14G2a, and splenocytes were harvested to generate hybridoma cells. Clones were screened by ELISA for mouse antibody binding to hu14.18. One positive clone was selected to purify and characterize the secreted IgG protein (κ, IgG(1)). This antibody bound to anti-GD(2) antibodies 14G2a, ch14.18/CHO, hu14.18, and to immunocytokines ch14.18-IL2 and hu14.18-IL2 as well as to NK-92 cells expressing scFv(ch14.18)-zeta receptor. Binding of these anti-GD(2) antibodies to the nominal antigen GD(2) as well as GD(2)-specific lysis of neuroblastoma cells by NK-92-scFv(ch14.18)-zeta cells was competitively inhibited by ganglidiomab, proving GD(2) surrogate function and anti-idiotype characteristics. The dissociation constants of ganglidiomab from anti-GD(2) antibodies ranged from 10.8 ± 5.01 to 53.5 ± 1.92 nM as determined by Biacore analyses. The sequences of framework and complementarity-determining regions of ganglidiomab were identified. Finally, we demonstrated induction of a GD(2)-specific humoral immune response after vaccination of mice with ganglidiomab effective in mediating GD(2)-specific killing of neuroblastoma cells. CONCLUSION: We generated and characterized a novel anti-idiotype antibody ganglidiomab and demonstrated activity against neuroblastoma.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/immunology , Cancer Vaccines/immunology , Gangliosides/immunology , Neuroblastoma/immunology , Amino Acid Sequence , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Anti-Idiotypic/chemistry , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Base Sequence , Binding, Competitive/immunology , Cell Line, Tumor , Gangliosides/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , Neuroblastoma/therapy , Protein Binding/immunology , Sequence Alignment
4.
Cancer Lett ; 331(2): 167-73, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23337288

ABSTRACT

Attenuated Salmonella is an approved oral life vaccine that is currently entering pre-clinical cancer vaccination studies as a promising DNA carrier. In a syngeneic mouse model for neuroblastoma, oral gavage of Salmonella typhimurium (SL7207) carrying recent generated survivin DNA vaccines induced a stronger cellular anti-NB immune response than gene gun application or injection of lentivirally transduced bone marrow-derived DCs. The level of Salmonella-associated side effects was not significant as indicated by unaffected survivin-mediated hematopoiesis and wound healing. We believe that our findings provide an important baseline to translate Salmonella-based DNA vaccination into a clinical application for neuroblastoma.


Subject(s)
Disease Models, Animal , Neuroblastoma/therapy , Salmonella/immunology , Vaccines, DNA/administration & dosage , Animals , Biolistics , Cell Line, Tumor , Female , Mice , Neuroblastoma/pathology , Plasmids
5.
J Mol Med (Berl) ; 91(4): 459-72, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23052481

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor in children. Combining passive immunotherapy with an antibody to the disialoganglioside GD2 (ch14.18/SP2/0) and cytokines with 13-cis-retinoic acid for post-myeloablative maintenance therapy increased survival in high-risk NB, but the overall prognosis for these children is still in need of improvement. Fenretinide (4-HPR) is a synthetic retinoid that has shown clinical activity in recurrent NB and is cytotoxic to a variety of cancer cells, in part via the accumulation of dihydroceramides, which are precursors of GD2. We investigated the effect of 4-HPR on CHO-derived, ch14.18-mediated anti-NB effector functions, complement-dependent cytotoxicity (CDC), and antibody-dependent and antibody-independent cellular cytotoxicity (ADCC and AICC, respectively). Here, we demonstrate for the first time that pretreatment of fenretinide-resistant NB cells with 4-HPR significantly enhanced ch14.18/CHO-mediated CDC and ADCC and AICC by both human natural killer cells and peripheral blood mononuclear cells. Treatment with 4-HPR increased GD2 and death receptor (DR) expression in resistant NB cells and induced an enhanced granzyme B and perforin production by effector cells. Blocking of ganglioside synthesis with a glucosylceramide synthase inhibitor abrogated the increased ADCC response but had no effect on the AICC, indicating that GD2 induced by 4-HPR mediates the sensitization of NB cells for ADCC. We also showed that 4-HPR induced increased GD2 and DR expression in a resistant NB xenograft model that was associated with an increased ADCC and AICC response using explanted tumor target cells from 4-HPR-treated mice. In summary, these findings provide an important baseline for the combination of 4-HPR and passive immunotherapy with ch14.18/CHO in future clinical trials for high-risk NB patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Fenretinide/pharmacology , Killer Cells, Natural/immunology , Neuroblastoma/immunology , Animals , Cell Line, Tumor , Coculture Techniques , Complement System Proteins/immunology , Female , Gangliosides/metabolism , Humans , Mice , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Receptors, Death Domain/metabolism , Xenograft Model Antitumor Assays
6.
J Cell Mol Med ; 16(3): 569-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21595822

ABSTRACT

Treatment of high-risk neuroblastoma (NB) represents a major challenge in paediatric oncology. Alternative therapeutic strategies include antibodies targeting the disialoganglioside GD(2) , which is expressed at high levels on NB cells, and infusion of donor-derived natural killer (NK) cells. To combine specific antibody-mediated recognition of NB cells with the potent cytotoxic activity of NK cells, here we generated clonal derivatives of the clinically applicable human NK cell line NK-92 that stably express a GD(2) -specific chimeric antigen receptor (CAR) comprising an anti-GD(2) ch14.18 single chain Fv antibody fusion protein with CD3-ζ chain as a signalling moiety. CAR expression by gene-modified NK cells facilitated effective recognition and elimination of established GD(2) expressing NB cells, which were resistant to parental NK-92. In the case of intrinsically NK-sensitive NB cell lines, we observed markedly increased cell killing activity of retargeted NK-92 cells. Enhanced cell killing was strictly dependent on specific recognition of the target antigen and could be blocked by GD(2) -specific antibody or anti-idiotypic antibody occupying the CAR's cell recognition domain. Importantly, strongly enhanced cytotoxicity of the GD(2) -specific NK cells was also found against primary NB cells and GD(2) expressing tumour cells of other origins, demonstrating the potential clinical utility of the retargeted effector cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Jejunal Neoplasms/therapy , Killer Cells, Natural/metabolism , Neuroblastoma/therapy , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line, Tumor , Child , Gangliosides/genetics , Gangliosides/immunology , Gene Expression , Genetic Engineering , Genetic Vectors , Humans , Immunotherapy, Adoptive , Jejunal Neoplasms/immunology , Jejunal Neoplasms/secondary , Jejunum/immunology , Jejunum/pathology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Neuroblastoma/immunology , Neuroblastoma/secondary , Receptors, Antigen/genetics , Receptors, Antigen/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Retroviridae , Transduction, Genetic
7.
Eur J Cancer ; 45(16): 2915-21, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19695868

ABSTRACT

The likelihood of identifying peptides of sufficient quality for the development of effective cancer vaccines by screening of phage display libraries is low. Here, we introduce the sequential application of systematic amino acid substitution by SPOT synthesis. After the substitution of two amino acids within the sequence of a phage display-derived mimotope of disialoganglioside GD2 (mimotope MA), the novel mimotope C3 showed improved GD2 mimicry in vitro. Peptide vaccination with the C3 mimotope induced an 18-fold increased anti-GD2 serum response associated with reduction of primary tumour growth and spontaneous metastasis in contrast to MA mimotope controls in a syngeneic neuroblastoma model. In summary, SPOT provides an ideal optimisation tool for the development of phage display-derived cancer vaccines.


Subject(s)
Amino Acid Substitution/genetics , Cancer Vaccines/genetics , Gangliosides/genetics , Molecular Mimicry/genetics , Neuroblastoma/prevention & control , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Gangliosides/immunology , Humans , Immunity, Active/genetics , Immunity, Active/immunology , Immunoglobulin G/metabolism , Mice , Neoplasm Transplantation , Neuroblastoma/pathology , Protein Array Analysis/methods
8.
Mol Cancer Ther ; 8(8): 2392-401, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19671753

ABSTRACT

Neuroblastoma (NB) is a challenging malignancy of the sympathetic nervous tissue characterized by a very poor prognosis. One important marker for NB is the expression of tyrosine hydroxylase (TH), the first-step enzyme of catecholamine biosynthesis. We could show stable and high TH gene expression in 67 NB samples independent of the clinical stage. Based on this observation, we addressed the question of whether xenogeneic TH DNA vaccination is effective in inducing an anti-NB immune response. For this purpose, we generated three DNA vaccines based on pCMV-F3Ub and pBUD-CE4.1 plasmids encoding for human (h)THcDNA (A), hTH minigene (B), and hTHcDNA in combination with the proinflammatory cytokine interleukin 12 (C), and tested prophylactic and therapeutic efficacy to suppress primary tumor growth and spontaneous metastasis. Here we report that xenogeneic TH DNA vaccination was effective in eradicating established primary tumors and inhibiting metastasis. Interestingly, this effect could not be enhanced by adding the Th1 cytokine interleukin 12. However, increased IFN-gamma production and NB cytotoxicity of effector cells harvested from vaccinated mice suggested the participation of tumor-specific CTLs in the immune response. The depletion of CD8(+)T cells completely abrogated the hTH vaccine-mediated anti-NB immune response. Furthermore, rechallenging of surviving mice resulted in reduced primary tumor growth, indicating the induction of a memory immune response. In conclusion, xenogeneic immunization with TH-derived DNA vaccines is effective against NB, and may open a new venue for a novel and effective immunotherapeutic strategy against this challenging childhood tumor.


Subject(s)
Antigens, Heterophile/administration & dosage , Neuroblastoma/therapy , Tyrosine 3-Monooxygenase/genetics , Vaccines, DNA/administration & dosage , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Immunization , Interleukin-12/metabolism , Mice , Neuroblastoma/immunology , Neuroblastoma/pathology , Tyrosine 3-Monooxygenase/metabolism , Vaccines, DNA/immunology
9.
Bioconjug Chem ; 20(8): 1587-94, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19572629

ABSTRACT

Children with relapsed and refractory acute lymphoblastic leukemia (ALL) still face a critical prognosis. We tested the hypothesis that targeted calicheamicin theta (θ) using an anti-CD19-immunoconjugate may provide an effective treatment strategy for CD19(+) ALL. Calicheamicin θ is a rationally designed prodrug of the natural enediyene calicheamicin γ, obtained by total synthesis. It offers the advantage of increased in vivo stability and 1000-fold higher antitumor potency over calicheamicin γ. First, we demonstrate efficacy of calicheamicin θ against primary pre-B leukemic cells and multidrug-resistant leukemia cell lines (IC(50) = 10(-9) to 10(-12) M). Second, conjugation of calicheamicin θ to an internalizing murine anti-CD19 monoclonal antibody was demonstrated to affect neither calicheamicin θ mediated cytotoxicity nor binding of the antibody to the target molecule. Third, anti-CD19-calicheamicin θ immunoconjugate revealed a maximum tolerated dose of 10 µg/kg and CD19-specific and long-lasting eradication of established leukemia was demonstrated in a xenograft model. Finally, we show that the antileukemic effect of anti-CD19-calicheamicin θ is mediated by induction of apoptosis proceeding through the caspase-mediated mitochondrial pathway. On the basis of these results, we conclude that anti-CD19-calicheamicin θ immunoconjugates may offer a novel and effective approach for the treatment of relapsed CD19(+) ALL.


Subject(s)
Aminoglycosides/therapeutic use , Antigens, CD19/metabolism , Antineoplastic Agents/therapeutic use , Enediynes/therapeutic use , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Aminoglycosides/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antigens, CD19/immunology , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enediynes/chemistry , Female , Humans , Immunoconjugates/immunology , Mice , Mice, SCID , Molecular Structure , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
10.
Int J Cancer ; 125(1): 104-14, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19291796

ABSTRACT

The inhibitor of apoptosis protein survivin is highly expressed in neuroblastoma (NB) and survivin-specific T cells were identified in Stage 4 patients. Therefore, we generated a novel survivin minigene DNA vaccine (pUS-high) encoding exclusively for survivin-derived peptides with superior MHC class I (H2-K(k)) binding affinities and tested its efficacy to suppress tumor growth and metastases in a syngeneic NB mouse model. Vaccination was performed by oral gavage of attenuated Salmonella typhimurium SL7207 carrying pUS-high. Mice receiving the pUS-high in the prophylactic setting presented a 48-52% reduction in s.c. tumor volume, weight and liver metastasis level in contrast to empty vector controls. This response was as effective as a survivin full-length vaccine and was associated with an increased target cell lysis, increased presence of CD8(+) T-cells at the primary tumor site and enhanced production of proinflammatory cytokines by systemic CD8(+) T cells. Furthermore, depletion of CD8(+) but not CD4(+) T-cells completely abrogated the pUS-high mediated primary tumor growth suppression, demonstrating a CD8(+) T-cell mediated effect. Therapeutic vaccination with pUS-high led to complete NB eradication in over 50% of immunized mice and surviving mice showed an over 80% reduction in primary tumor growth upon rechallenge in contrast to controls. In summary, survivin-based DNA vaccination is effective against NB and the rational minigene design provides a promising approach to circumvent potentially hazardous effects of using full length antiapoptotic genes as DNA vaccines.


Subject(s)
Microtubule-Associated Proteins/genetics , Neuroblastoma/prevention & control , Vaccines, DNA/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Drug Design , Female , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Immunoenzyme Techniques , Inhibitor of Apoptosis Proteins , Mice , Mice, Inbred A , Neuroblastoma/immunology , Peptide Fragments/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Survivin , Vaccination
11.
Mol Cancer Ther ; 7(7): 2241-51, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645033

ABSTRACT

Therapeutic vaccination against tumor antigens without induction of autoimmunity remains a major challenge in cancer immunotherapy. Here, we show for the first time effective therapeutic vaccination followed by suppression of established spontaneous neuroblastoma metastases using a tyrosine hydroxylase (TH) DNA minigene vaccine. We identified three novel mouse TH (mTH3) derived peptides with high predicted binding affinity to MHC class I antigen H2-K(k) according to the prediction program SYFPEITHI and computer modeling of epitopes into the MHC class I antigen binding groove. Subsequently, a DNA minigene vaccine was generated based on the expression vector pCMV-F3Ub encoding mutated ubiquitin (Gly(76) to Ala(76)) and mTH3. Prophylactic and therapeutic efficacies of this vaccine were established following oral delivery with attenuated Salmonella typhimurium SL7207. Only mice immunized with mTH3 were free of spontaneous liver metastases. This effect was clearly dependent on ubiquitin and high affinity of the mTH epitopes to MHC class I antigens. Specifically, we showed a crucial role for minigene expression as a stable ubiquitin-Ala(76) fusion peptide for vaccine efficacy. The immune response following the mTH3 DNA minigene vaccination was mediated by CD8(+) T cells as indicated by infiltration of primary tumors and TH-specific cytolytic activity in vitro. Importantly, no cell infiltration was detectable in TH-expressing adrenal medulla, indicating the absence of autoimmunity. In summary, we show effective therapeutic vaccination against neuroblastoma with a novel rationally designed TH minigene vaccine without induction of autoimmunity providing an important baseline for future clinical application of this strategy.


Subject(s)
Drug Design , Immunity/immunology , Neuroblastoma/immunology , Neuroblastoma/prevention & control , Tyrosine 3-Monooxygenase/immunology , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Antibody Specificity/immunology , COS Cells , Chlorocebus aethiops , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , T-Lymphocytes/immunology , Ubiquitin/metabolism , Vaccination
12.
Pediatr Blood Cancer ; 50(2): 284-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17366537

ABSTRACT

BACKGROUND: The replacement of established evidence-based cancer therapy protocols (mainstream therapy) by unevaluated complementary and alternative medicine (CAM) is a challenge in pediatric oncology. We tested the hypothesis that oral application of L-lysine and ascorbic acid (Lysin C Drink) in combination with epigallocatechin-gallate (EGCG) and amino-acids (Epican forte) is effective in a preclinical model of neuroblastoma. METHODS: Primary tumors and spontaneous metastases were induced in A/J mice by injection of NXS2 neuroblastoma cells. Mice were treated by daily oral gavage with L-lysine and ascorbic acid (Lysin C Drink) (equivalent to 150 mg ascorbic acid/day/mouse) (treatment A) or with EGCG plus ascorbic- and amino-acids (Epican forte) (9.2 mg/mouse) (treatment B). Treatment A was started in the prophylactic setting (7 days before tumor cell injection) as well as in the therapeutic setting (1 day after tumor cell inoculation). Finally, treatment B was evaluated alone and in combination with treatment A in the therapeutic setting. The effect on primary tumor growth and the development of spontaneous liver metastases was evaluated. RESULTS: L-lysine and ascorbic acid (Lysin C Drink) and EGCG plus ascorbic- and amino-acids (Epican forte) are ineffective in reduction of primary tumor growth and prevention of spontaneous liver metastases in this model. CONCLUSIONS: Neither a formal clinical development nor the use of these substances can be recommended for neuroblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/drug therapy , Animals , Ascorbic Acid/administration & dosage , Catechin/administration & dosage , Catechin/analogs & derivatives , Dietary Supplements , Disease Models, Animal , Female , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms, Experimental/secondary , Lysine/administration & dosage , Mice , Mice, Inbred A , Neuroblastoma/pathology , Neuroblastoma/secondary , Proline/administration & dosage
13.
Cancer Res ; 67(5): 2331-8, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332365

ABSTRACT

Fractalkine (FKN) is a unique CX3C chemokine (CX3CL1) known to induce both adhesion and migration of leukocytes mediated by a membrane-bound and a soluble form, respectively. Its function is mediated through CX3C receptor (CX3CR), which is expressed by T(H)1 immune cells including T cells and natural killer (NK) cells. FKN was shown to be expressed in >90% of 68 neuroblastoma samples as determined by cDNA microarray analysis. Here, we characterized the effect of FKN in the neuroblastoma microenvironment using a syngeneic model genetically engineered to secrete FKN. We show FKN-mediated migration, adhesion, and IFN-gamma secretion of immune effector cells, but limited antineuroblastoma activity, in vitro and in vivo. Therefore, we tested the hypothesis that a combined increase of FKN and interleukin-2 (IL-2) in the neuroblastoma microenvironment induces an effective antitumor immune response. For this purpose, IL-2 was targeted to ganglioside GD2, which is highly expressed on neuroblastoma tissue, using an anti-GD2 antibody IL-2 immunocytokine (ch14.18-IL-2). Only mice bearing FKN- and IL-2-enriched neuroblastoma tumors exhibited a reduction in primary tumor growth and a complete eradication of experimental liver metastases. The depletion of T cells and NK cells in vivo abrogated the effect, and these effector cells showed the highest cytolytic activity in vitro. Finally, only the FKN- and IL-2-enriched neuroblastoma microenvironment resulted in T-cell activation and the release of proinflammatory cytokines. In summary, we showed for the first time the immunologic mechanisms by which targeted IL-2 treatment of neuroblastoma with an FKN-rich microenvironment induces an effective antitumor response.


Subject(s)
Chemokines, CX3C/metabolism , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , Neuroblastoma/pathology , Neuroblastoma/therapy , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Chemokine CX3CL1 , Chemokines, CX3C/genetics , Chemokines, CX3C/physiology , Female , Gene Targeting , Humans , Immunity, Cellular , Immunotherapy/methods , Interleukin-2/genetics , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred Strains , Neoplasm Metastasis , Neoplasm Transplantation , Neuroblastoma/immunology , Neuroblastoma/metabolism
14.
Cancer Res ; 66(21): 10567-75, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17079481

ABSTRACT

Disialoganglioside GD2 is an established target for immunotherapy in neuroblastoma. We tested the hypothesis that active immunization against the glycolipid GD2 using DNA vaccines encoding for cyclic GD2-mimicking decapeptides (i.e., GD2 mimotopes) is effective against neuroblastoma. For this purpose, two GD2 peptide mimotopes (MA and MD) were selected based on docking experiments to anti-GD2 antibody ch14.18 (binding free energy: -41.23 kJ/mol for MA and -48.06 kJ/mol for MD) and Biacore analysis (K(d) = 12.3 x 10(-5) mol/L for MA and 5.3 x 10(-5) mol/L for MD), showing a higher affinity of MD over MA. These sequences were selected for DNA vaccine design based on pSecTag2-A (pSA) also including a T-cell helper epitope. GD2 mimicry was shown following transfection of CHO-1 cells with pSA-MA and pSA-MD DNA vaccines, with twice-higher signal intensity for cells expressing MD over MA. Finally, these DNA vaccines were tested for induction of tumor protective immunity in a syngeneic neuroblastoma model following oral DNA vaccine delivery with attenuated Salmonella typhimurium (SL 7207). Only mice receiving the DNA vaccines revealed a reduction of spontaneous liver metastases. The highest anti-GD2 humoral immune response and natural killer cell activation was observed in mice immunized with the pSA-MD, a finding consistent with superior calculated binding free energy, dissociation constant, and GD2 mimicry potential for GD2 mimotope MD over MA. In summary, we show that DNA immunization with pSA-MD may provide a useful strategy for active immunization against neuroblastoma.


Subject(s)
Cancer Vaccines/immunology , Gangliosides/immunology , Neuroblastoma/secondary , Vaccines, DNA/immunology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Gangliosides/genetics , Interferon-gamma/biosynthesis , Mice , Neoplasm Metastasis/prevention & control , Neuroblastoma/immunology , Neuroblastoma/prevention & control , Vaccination
15.
Cancer Lett ; 228(1-2): 187-93, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-15953676

ABSTRACT

The induction of tumor protective immunity against neuroblastoma remains a major challenge for active immunotherapy. Fractalkine is a unique Th1 CX3C chemokine known to induce adhesion and migration of leukocytes mediated by both, a membrane-bound and soluble form, respectively. Here, we tested the hypothesis that chemokine gene therapy with fractalkine (FKN) induces an effective anti-neuroblastoma immune response amplified by targeted IL-2 using the anti-GD2 antibody ch14.18 fused with IL-2 (ch14.18-IL-2). For this purpose, NXS2 cells were genetically engineered to stably produce murine FKN (NXS2-FKN). Transcription and expression of the mFKN gene in tumor tissue of mice inoculated with NXS2-FKN cells were demonstrated in vivo. Importantly, mFKN exhibited a reduction in primary tumor growth and spontaneous liver metastases in syngenic A/J mice. This effect was boosted by targeted IL-2 using small non-curative doses of ch14-18-IL-2. The amplification of the FKN induced immune response was specific, since a non-specific antibody-IL-2 fusion protein ch225-IL-2 was ineffective. In summary, we demonstrated for the first time that chemokine gene therapy is amplified by targeted IL-2 suggesting a combination of both strategies as an adjuvant therapy for neuroblastoma.


Subject(s)
Chemokines, CX3C/genetics , Genetic Therapy , Interleukin-2/therapeutic use , Membrane Proteins/genetics , Neuroblastoma/therapy , Animals , Base Sequence , Chemokine CX3CL1 , DNA Primers , Gene Transfer Techniques , Humans , Liver Neoplasms/secondary , Mice , Neuroblastoma/drug therapy
16.
Ann N Y Acad Sci ; 1028: 113-21, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15650237

ABSTRACT

The disruption of self-tolerance against neuroblastoma is the ultimate goal of an effective DNA-vaccine. We demonstrate the induction of protective immunity against syngeneic murine NXS2 neuroblastoma in A/J mice following vaccination with tyrosine hydroxylase (TH)-derived antigens. Oral gene delivery was accomplished using an attenuated strain of Salmonella typhimurium as a carrier harboring vectors encoding for mouse tyrosine hydroxylase (mTH) antigens. Vaccination was effective in protecting animals from a lethal challenge with wild-type NXS2 tumor cells. These findings were extended by comparing efficacy of mTH minigene vaccines with a minigene vaccine comprising three novel epitopes isolated fom NXS2 neuroblastoma cells. For this purpose, MHC class I was immunoprecipitated from NXS2 cell lysates, and peptides were eluted and examined in tandem-mass spectrometry analysis. This led to the identification of three novel natural MHC class I peptide ligands: TEALPVKLI, from ribonucleotide reductase M2; NEYIMSLI, from Ser/Thr protein phosphatase 2A; and FEMVSTLI, of unknown origin. Two minigenes were constructed, one encoding for the three novel epitopes and the second for three known mTH-derived epitopes with high predicted binding affinity to MHC class I, by cloning them into the mammalian expression vector pCMV-3FUB. Immunized mice showed a reduction in primary tumor growth and the absence of spontaneous liver metastasis in the majority of animals. Importantly, there was no significant difference between the two minigenes, suggesting that, compared with tumor peptide isolation, mTH epitope prediction is similarly effective for designing efficient DNA-minigene vaccines. In summary, these findings establish proof of the concept that disruption of self-tolerance against neuroblastoma-associated epitopes may be an effective adjuvant therapeutic strategy.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Gene Transfer Techniques , Immunization, Passive/methods , Immunotherapy, Active/methods , Neuroblastoma/therapy , Vaccines, DNA , Animals , Cell Line, Tumor , Cloning, Molecular , DNA/chemistry , Epitopes/chemistry , Genetic Vectors , Humans , Immunoprecipitation , Immunotherapy/methods , Inflammation , Ligands , Mass Spectrometry , Mice , Models, Biological , Neuroblastoma/chemistry , Neuroblastoma/genetics , Peptides/chemistry , Tyrosine 3-Monooxygenase/genetics
17.
Expert Opin Biol Ther ; 3(1): 187-92, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12718741

ABSTRACT

The AACR Special Conference 2002 for Ubiquitination in Normal and Cancer Cells took place in Vancouver, BC, Canada. It was, indeed, special: a "special" location, a "special" audience and excellent talks that gave a detailed insight into the ubiquitination/proteasome-field. The following meeting highlights try to give a summary of some topics covered at the meeting, from basic research to successful applications of therapeutic agents, starting with cellular regulation, describing recently discovered structural features of enzymes involved in de-/ubiquitination, and, finally, presenting proteasome inhibition as a new approach in cancer chemotherapy.


Subject(s)
Neoplasms/metabolism , Ubiquitin/metabolism , Animals , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/therapeutic use , Humans , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Multienzyme Complexes/therapeutic use , Neoplasms/drug therapy , Proteasome Endopeptidase Complex , Ubiquitin/therapeutic use , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/therapeutic use
18.
Blood ; 102(1): 246-53, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12623853

ABSTRACT

Effective therapy of high-risk leukemia with established cytotoxic drugs may be limited by poor antitumor efficacy, systemic toxicity, and the induction of drug resistance. Here, we provide the first evidence that hydrolytically activated prodrugs may overcome these problems. For this purpose, VP16 was functionally blocked by hydrolytically cleavable carbonate linkers with unique characteristics to generate 2 novel prodrugs of VP16. First, we established a more than 3-log higher efficacy of the 2 prodrugs compared with VP16 on a panel of naturally drug-resistant tumor cell lines. Second, the prodrugs did overcome VP16-induced multidrug resistance-1 gene (MDR-1)-mediated multidrug resistance in vitro in a newly established VP16-resistant T-cell leukemia cell line MOVP-3 by functionally blocking MDR-1-mediated efflux. Third, in vivo studies showed a maximum tolerated dose of ProVP16-II (> 45mg/kg), which was at least 3-fold higher than that of VP16 (15 mg/kg). Finally, tests of ProVP16-II in a multidrug-resistant xenograft model of T-cell leukemia expressing MDR-1 indicated that only the mice treated with this prodrug revealed a complete and long-lasting regression of established, drug-resistant leukemia. In summary, the hydrolytically activated etoposide prodrugs proved effective against multidrug-resistant T-cell leukemia in vitro and in vivo and provide proof of concept for a highly promising new strategy for the treatment of MDR-1 drug-resistant malignancies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Etoposide/pharmacology , Leukemia, T-Cell/pathology , Animals , Cell Survival/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Etoposide/chemistry , Female , Humans , Hydrolysis , Inhibitory Concentration 50 , Leukemia, T-Cell/drug therapy , Mice , Mice, Inbred Strains , Prodrugs/chemistry , Prodrugs/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...