Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Gene Ther ; 25(1): 54-65, 2018 01.
Article in English | MEDLINE | ID: mdl-28925992

ABSTRACT

Current treatments of hepatocellular carcinoma (HCC) are ineffective and unsatisfactory in many aspects. Cancer-targeting gene virotherapy using oncolytic adenoviruses (OAds) armed with anticancer genes has shown efficacy and safety in clinical trials. Nowadays, both inhibitor of growth 4 (ING4), as a multimodal tumor suppressor gene, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), as a potent apoptosis-inducing gene, are experiencing a renaissance in cancer gene therapy. Herein we investigated the antitumor activity and safety of mono- and combined therapy with OAds armed with ING4 (Ad-ΔB/ING4) and TRAIL (Ad-ΔB/TRAIL) gene, respectively, on preclinical models of human HCC. OAd-mediated expression of ING4 or TRAIL transgene was confirmed. Ad-ΔB/TRAIL and/or Ad-ΔB/ING4 exhibited potent killing effect on human HCC cells (HuH7 and Hep3B) but not on normal liver cells. Most importantly, systemic therapy with Ad-ΔB/ING4 plus Ad-ΔB/TRAIL elicited more eradicative effect on an orthotopic mouse model of human HCC than their monotherapy, without causing obvious overlapping toxicity. Mechanistically, Ad-ΔB/ING4 and Ad-ΔB/TRAIL were remarkably cooperated to induce antitumor apoptosis and immune response, and to repress tumor angiogenesis. This is the first study showing that concomitant therapy with Ad-ΔB/ING4 and Ad-ΔB/TRAIL may provide a potential strategy for HCC therapy and merits further investigations to realize its possible clinical translation.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cell Cycle Proteins/genetics , Genetic Therapy , Homeodomain Proteins/genetics , Liver Neoplasms/therapy , Oncolytic Virotherapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Cytopathogenic Effect, Viral , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Transfection , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Gene Ther ; 21(5): 476-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24598892

ABSTRACT

Oncolytic adenoviruses (Ad) have been developed for the eradication of tumors. Although they hold much promise as a cancer therapy, they have a short blood circulation time and high liver toxicity. An effective strategy to overcome these problems has been complexing Ad with shielding materials. However, the therapeutic efficacy of the Ad complexes has also been an issue because passive accumulation does not allow for sufficient delivery of Ad to the cancer cells. To enhance the therapeutic efficacy of the polymer-coated Ads, the attachment of a targeting moiety to polymer-coated Ad vectors is inescapable. Our lab has previously reported the potential use of Arg-Gly-Asp (RGD)-targeted bioreducible polymers with a polyethylene glycol (PEG) linker for delivering oncolytic Ads. We have shown the enhanced in vitro transduction efficiency and increased cancer-killing effect with producing progeny oncolytic Ad particles. In addition, we have shown significant tumor-growth inhibition of the polymer-shielded Ad in an in vivo lung orthotopic tumor model. The shielding effect of the Ad surface with the polymers allowed evasion of host immune responses and reduction of liver toxicity. This data demonstrates that the RGD-conjugated bioreducible polymer for delivering the oncolytic Ad vectors could be utilized for cancer therapy via systemic administration.


Subject(s)
Adenocarcinoma/therapy , Fibrosarcoma/therapy , Lung Neoplasms/therapy , Oligopeptides/pharmacology , Oncolytic Virotherapy/methods , Adenocarcinoma of Lung , Adenoviridae/chemistry , Cell Line, Tumor , HEK293 Cells , Humans , Liver/injuries , Liver/virology , Oligopeptides/chemistry , Oncolytic Viruses/chemistry , Polyethylene Glycols , Polymers/chemistry , Polymers/pharmacology , RNA Interference , RNA, Small Interfering
3.
Gene Ther ; 21(1): 106-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24225639

ABSTRACT

For successful clinical tumor immunotherapy outcomes, strong immune responses against tumor antigens must be generated. Cell-based vaccines compromise one strategy with which to induce appropriate strong immune responses. Previously, we established a natural killer T-cell (NKT) ligand-loaded, adenoviral vector-transduced B-cell-based anticancer cellular vaccine. To enhance tumor antigen delivery to B cells, we established a modified adenoviral vector (Ad-k35) that encoded a truncated form of the breast cancer antigen Her2/neu (Ad-k35HM) in which fiber structure was substituted with adenovirus serotype 35. We observed increased tumor antigen expression with Ad-k35HM in both human and murine B cells. In addition, an Ad-k35HM-transduced B-cell vaccine elicited strong antigen-specific cellular and humoral immune responses that were further enhanced with the additional loading of soluble NKT ligand KBC009. An Ad-k35HM-transduced, KBC009-loaded B-cell vaccine efficiently suppressed the in vivo growth of established tumors in a mouse model. Moreover, the vaccine elicited human leukocyte antigen (HLA)-A2 epitope-specific cytotoxic T-cell responses in B6.Cg (CB)-Tg (HLA-A/H2-D) 2Enge/Jat mice. These findings indicated that the Ad-k35 could be appropriate for the preclinical and clinical development of B-cell-based anticancer immunotherapies.


Subject(s)
B-Lymphocytes/immunology , Cancer Vaccines , Dependovirus/genetics , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/genetics , Animals , B-Lymphocytes/virology , Cancer Vaccines/immunology , Cells, Cultured , Dependovirus/metabolism , Female , Genetic Vectors , HLA-A2 Antigen/immunology , Humans , Immunotherapy , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Natural Killer T-Cells/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/immunology , Xenograft Model Antitumor Assays
4.
Gene Ther ; 20(9): 880-92, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23514707

ABSTRACT

Adenoviruses (Ad) have been investigated for their efficacy in reducing primary tumors after local intratumoral administration. Despite high Ad concentrations and repetitive administration, the therapeutic efficacy of Ad has been limited because of rapid dissemination of the Ad into the surrounding normal tissues and short maintenance of Ad biological activity in vivo. To maximize the therapeutic potential of Ad-mediated gene therapeutics, we investigated the efficacy of local, sustained Ad delivery, using an injectable alginate gel matrix system. The biological activity of Ad loaded in alginate gel was prolonged compared with naked Ad, as evidenced by the high green fluorescent protein gene transduction efficiency over an extended time period. Moreover, oncolytic Ad encapsulated in alginate gel elicited 1.9- to 2.4-fold greater antitumor activity than naked Ad in both C33A and U343 human tumor xenograft models. Histological and quantitative PCR analysis confirmed that the oncolytic Ad/alginate gel matrix system significantly increased preferential replication and dissemination of oncolytic Ad in a larger area of tumor tissue in vivo. Taken together, these results show that local sustained delivery of oncolytic Ad in alginate gel augments therapeutic effect through selective infection of tumor cells, sustained release and prolonged maintenance of Ad activity.


Subject(s)
Adenoviridae/genetics , Adenoviridae/physiology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Alginates , Animals , Cell Line, Tumor , Genetic Therapy , Glucuronic Acid , Hexuronic Acids , Humans , Mice , Mice, Nude , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Xenograft Model Antitumor Assays
5.
Gene Ther ; 19(7): 711-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21993173

ABSTRACT

Interleukin (IL)-12 and granulocyte-monocyte colony-stimulating factor (GM-CSF) have recently been used as immunotherapeutic agents in cancer gene therapy. IL-12 and GM-CSF have differential roles in the antitumor immune response, as IL-12 targets T, NK and natural killer T (NKT) cells and GM-CSF principally targets antigen-presenting cells (APCs). To strengthen the therapeutic efficacy of these two cytokines, we generated an oncolytic adenovirus (Ad), Ad-ΔB7/IL12/GMCSF, coexpressing IL-12 and GM-CSF. Using a murine B16-F10 syngeneic tumor model, we show that Ad-ΔB7/IL12/GMCSF promoted antitumor responses and increased survival compared with an oncolytic Ad expressing IL-12 or GM-CSF alone (Ad-ΔB7/IL12 or Ad-ΔB7/GMCSF, respectively). By measuring cytotoxic T lymphocyte activity and interferon-γ production, we show that the enhanced therapeutic effect was mediated by the induction of immune cell cytotoxicity. In situ delivery of Ad-ΔB7/IL12/GMCSF resulted in massive infiltration of CD4(+) T cells, CD8(+) T cells, NK cells and CD86(+) APCs into the tissue surrounding the necrotic area of the tumor. Moreover, GM-CSF effectively promoted antitumor immune memory, which was significantly augmented by IL-12. Lastly, IL12-expressing oncolytic Ads prevented tumor-induced thymic atrophy and was associated with reduced apoptosis and increased proliferation in the thymus. Taken together, these data demonstrate that an oncolytic Ad coexpressing IL-12 and GM-CSF is a potential therapeutic tool for the treatment of cancer.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunologic Memory , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Thymus Gland/immunology , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Interleukin-12/genetics , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Oncolytic Viruses/genetics , T-Lymphocytes, Cytotoxic/immunology , Thymus Gland/pathology
6.
Gene Ther ; 19(10): 967-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22033466

ABSTRACT

S100A2, a member of the S100 family of calcium-binding proteins, has been implicated in carcinogenesis as both a tumor suppressor and stimulator. Here, we characterized promoter activity of S100A2, generated an S100A2 promoter-driven conditionally replicative adenovirus (Ad/SA), and evaluated its anti-tumor activity in vitro and in vivo. Promoter activity of S100A2 was greatly restricted to tumor cells, and the S100A2 promoter bound with typical nuclear targets of epidermal growth factor receptor (EGFR) signaling. EGF-stimulated EGFR phosphorylation induced S100A2 expression and further activated E1A expression of Ad/SA, which was restored by EGFR signal inhibition in a concentration-dependent manner in non-small-cell lung carcinoma (NSCLC). In two EGFR-activated tumor xenograft animal models, Ad/SA exhibited potent anti-tumor activity, whereas cetuximab, an EGFR-targeting anticancer drug, was active transiently or ineffective. Combined treatment with cetuximab or cisplatin plus Ad/SA resulted in enhanced anti-tumor activity. Immunohistochemical analysis of tumor sections showed moderate-to-high grade signals for EGFR and adenovirus, and a reduction in viable cells in Ad/SA-treated tumors. Collectively, these results demonstrate that the S100A2 promoter-driven adenovirus is a potent inhibitor of cancers, and further suggest that S100A2 is a target gene of EGFR signaling pathway in NSCLC.


Subject(s)
Adenoviridae/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Chemotactic Factors/genetics , Lung Neoplasms/genetics , Oncolytic Viruses/genetics , Promoter Regions, Genetic , S100 Proteins/genetics , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cetuximab , Cisplatin/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression , Genetic Vectors/genetics , Humans , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy , Virus Replication , Xenograft Model Antitumor Assays
7.
Gene Ther ; 18(9): 898-909, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21451575

ABSTRACT

The oncolytic adenovirus (Ad) is currently being advanced as a promising antitumor remedy as it selectively replicates in tumor cells and can transfer and amplify therapeutic genes. Interleukin (IL)-12 induces a potent antitumor effect by promoting natural killer (NK) cell and cytotoxic T cell activities. IL-18 also augments cytotoxicity of NK cells and proliferation of T cells. This effect further enhances the function of IL-12 in a synergistic manner. Therefore, we investigated for the first time an effective cancer immunogene therapy of syngeneic tumors via intratumoral administration of oncolytic Ad co-expressing IL-12 and IL-18, RdB/IL-12/IL-18. Intratumoral administration of RdB/IL-12/IL-18 improved antitumor effects, as well as increased survival, in B16-F10 murine melanoma model. The ratio of T-helper type 1/2 cytokine as well as the levels of IL-12, IL-18, interferon-γ and granulocyte-macrophage colony-stimulating factor was markedly elevated in RdB/IL-12/IL-18-treated tumors. Mice injected with RdB/IL-12/IL-18 also showed enhanced cytotoxicity of tumor-specific immune cells. Consistent with these results, immense necrosis and infiltration of NK cells, as well as CD4+ and CD8+ T cells, were observed in RdB/IL-12/IL-18-treated tumor tissues. Importantly, tumors treated with RdB/IL-12/IL-18 showed an elevated number of T cells expressing IL-12Rß2 or IL-18Rα. These results provide a new insight into therapeutic mechanisms of IL-12 plus IL-18 and provide a potential clinical cancer immunotherapeutic agent for improved antitumor immunity.


Subject(s)
Genetic Therapy/methods , Interleukin-12/genetics , Interleukin-18/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Oncolytic Virotherapy/methods , Receptors, Interleukin-12/metabolism , Receptors, Interleukin-18/metabolism , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Cell Differentiation , Cell Line, Tumor , Interleukin-12/metabolism , Interleukin-18/metabolism , Male , Mice , Mice, Inbred C57BL , Oncolytic Viruses
8.
Br J Dermatol ; 165(3): 673-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21623756

ABSTRACT

BACKGROUND: Keloids or hypertrophic scars are pathological proliferations of the dermal skin layer resulting from excessive collagen deposition. Because the hormone relaxin (RLX) inhibits collagen synthesis and expression in stimulated fibroblasts, an adenovirus expressing RLX (dE1-RGD/lacZ/RLX) was generated. OBJECTIVES: To investigate the effect of RLX-expressing adenovirus on expression of various extracellular matrix (ECM) components in primary keloid spheroids. METHODS: The expression levels of type I and III collagen, fibronectin and elastin were investigated by immunohistochemistry in primary keloid spheroids transduced with the RLX-expressing adenovirus. RESULTS: Immunohistochemical analysis showed that expression of major ECM components (e.g. type I and III collagen, elastin and fibronectin) was markedly reduced in primary keloid spheroids transduced with dE1-RGD/lacZ/RLX. CONCLUSIONS: These results suggest that the antifibrotic effect of RLX-expressing adenovirus may have therapeutic effects on keloids by reversing pathological fibrosis and preventing keloid recurrence after surgical excision.


Subject(s)
Adenoviridae/genetics , Collagen Type III/metabolism , Collagen Type I/metabolism , Genetic Therapy/methods , Keloid/therapy , Relaxin/genetics , Adult , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Fibrosis/prevention & control , Humans , Immunohistochemistry , Keloid/metabolism , Middle Aged , Transduction, Genetic
9.
Nat Med ; 5(10): 1203-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10502827

ABSTRACT

Angiogenesis inhibitors produced by a primary tumor can create a systemic anti-angiogenic environment and maintain metastatic tumor cells in a state of dormancy. We show here that the gallbladder microenvironment modulates the production of transforming growth factor (TGF)-beta1, a multifunctional cytokine that functions as an endogenous anti-angiogenic and anti-tumor factor in a cranial window preparation. We found that a wide variety of human gallbladder tumors express TGF-beta1 irrespective of histologic type. We implanted a gel impregnated with basic fibroblast growth factor or Mz-ChA-2 tumor in the cranial windows of mice without tumors or mice with subcutaneous or gallbladder tumors to study angiogenesis and tumor growth at a secondary site. Angiogenesis, leukocyte-endothelial interaction in vessels and tumor growth in the cranial window were substantially inhibited in mice with gallbladder tumors. The concentration of TGF-beta1 in the plasma of mice with gallbladder tumors was 300% higher than that in the plasma of mice without tumors or with subcutaneous tumors. In contrast, there was no difference in the plasma levels of other anti- and pro-angiogenic factors. Treatment with neutralizing antibody against TGF-beta1 reversed both angiogenesis suppression and inhibition of leukocyte rolling induced by gallbladder tumors. TGF-beta1 also inhibited Mz-ChA-2 tumor cell proliferation. Our results indicate that the production of anti-angiogenesis/proliferation factors is regulated by tumor-host interactions.


Subject(s)
Brain Neoplasms/blood supply , Carcinoma/metabolism , Gallbladder Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Transforming Growth Factor beta/metabolism , Angiostatins , Animals , Carcinoma/surgery , Gallbladder Neoplasms/surgery , Mice , Mice, SCID , Peptide Fragments/isolation & purification , Plasminogen/isolation & purification , Skull/surgery , Thrombospondin 1/isolation & purification
10.
Gene Ther ; 17(2): 190-201, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19907500

ABSTRACT

The pressing challenge for contemporary gene therapy is to deliver enough therapeutic genes to enough cancer cells in vivo. With the aim of improving viral distribution and tumor penetration, we explored the use of decorin to enhance viral spreading and tumor tissue penetration. We generated decorin-expressing replication-incompetent (dl-LacZ-DCNG, dl-LacZ-DCNQ and dl-LacZ-DCNK) and replication-competent (Ad-DeltaE1B-DCNG, Ad-DeltaE1B-DCNQ and Ad-DeltaE1B-DCNK) adenoviruses (Ads). Point mutants of decorin gene (DCNG), DCNK and DCNQ, have a negative and moderate binding affinity to type-I collagen fibril, respectively. In both tumor spheroids and established solid tumors in vivo, tissue penetration potency of dl-LacZ-DCNG was greatly enhanced than those of dl-LacZ, dl-LacZ-DCNQ and dl-LacZ-DCNK, and this enhanced tissue penetration effect derived from decorin-expressing Ad was dependent on the binding affinity of decorin to collagen fibril. Expression of DCNG enhanced viral spread of replicating Ad, leading to improved tumor reduction and survival benefit. Moreover, the tumoricidal effects of Ad-DeltaE1B-DCNQ and Ad-DeltaE1B-DCNK were lessened, as the binding affinity to collagen was decreased, showing that the increased cancer cell cytotoxicity was driven by the action of decorin on extracellular matrix (ECM). Furthermore, Ad-DeltaE1B-DCNG substantially decreased ECM components within the tumor tissue. Finally, intratumoral injection of Ad-DeltaE1B-DCNG in primary tumor site greatly reduced the formation of B16BL6 melanoma cell pulmonary metastases in mice. Taken together, these data show the utility of decorin as a dispersion agent and highlight its utility and potential in improving the efficacy of replicating Ad-mediated cancer gene therapy.


Subject(s)
Adenoviridae/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Oncolytic Virotherapy/methods , Proteoglycans/genetics , Animals , Cell Line, Tumor , Decorin , Extracellular Matrix Proteins/metabolism , Gene Transfer Techniques , Genetic Therapy , Mice , Mice, Nude , Proteoglycans/metabolism , Spheroids, Cellular/metabolism , Transduction, Genetic , Xenograft Model Antitumor Assays
11.
Gene Ther ; 16(9): 1111-21, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19494843

ABSTRACT

Radiation therapy, a mainstay for anti-tumor therapeutic regimens for a variety of tumor types, triggers tumor cell apoptotic pathways by either directly eliciting DNA damage or indirectly inducing the formation of oxygen radicals. In an effort to augment radiation therapy, we generated a double E1B 19 kDa- and E1B 55 kDa-deleted oncolytic adenovirus (Ad-DeltaE1B19/55). In combination with radiotherapy, greater cytotoxicity was observed for Ad-DeltaE1B19/55 than for the single E1B 55 kDa-deleted oncolytic Ad (Ad-DeltaE1B55). Consistent with this observation, higher levels of p53, phospho-p53, phospho-Chk1, phospho-Chk2, PI3K (phosphatidylinositol-3-kinase), phospho-AKT, cytochrome c, and cleavage of PARP (poly (ADP-ribose) polymerase) and caspase-3 were observed in cells treated with Ad-DeltaE1B19/55 compared with those treated with Ad-DeltaE1B55, indicating that the E1B 19 kDa present in Ad-DeltaE1B55 may partially block radiation-induced apoptosis. A significant therapeutic benefit was also observed in vivo when oncolytic Ads and radiation were combined. Tumors treated with Ad-DeltaE1B19/55 and radiation showed large areas of necrosis and apoptosis with the corresponding induction of p53. Finally, consistent with in vitro observations, the combination of Ad-DeltaE1B19/55 and radiation was more efficacious than the combination of Ad-DeltaE1B55 and radiation. Taken together, these results present a strong therapeutic rationale for combining radiation therapy with E1B 19 kDa-deleted oncolytic Ad.


Subject(s)
Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Uterine Cervical Neoplasms/therapy , Animals , Apoptosis/genetics , Combined Modality Therapy , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
12.
Gene Ther ; 15(9): 635-51, 2008 May.
Article in English | MEDLINE | ID: mdl-18273054

ABSTRACT

RNA interference, due to its target specificity, may be highly effective as a novel therapeutic modality, but direct delivery of synthetic small interfering RNA still remains a major obstacle for this approach. To induce long-term expression and specific gene silencing, novel delivery vector system is also required. In this study, we have generated an efficient oncolytic adenovirus (Ad)-based short hairpin (shRNA) expression system (Ad-DeltaB7-U6shIL8) against IL-8, a potent proangiogenic factor. To demonstrate IL-8-specificity of this newly engineered Ad-based shRNA, we also manufactured replication-incompetent Ads (Ad-DeltaE1-CMVshIL8 and Ad-DeltaE1-U6shIL8) under the control of the cytomegalovirus (CMV) and U6 promoters, respectively. Ad-DeltaE1-U6shIL8 was highly effective in reducing IL-8 expression, and was much more effective in driving IL-8-specific shRNA than the CMV promoter-driven vector. The reduced IL-8 expression then translated into decreased angiogenesis in vitro as measured by migration, tube formation and rat aortic ring sprouting assays. In addition to its effect on endothelial cells, Ad-DeltaE1-U6shIL8 also effectively suppressed the migration and invasion of cancer cells. In vivo, intratumoral injection of Ad-DeltaB7-U6shIL8 significantly inhibited the growth of Hep3B and A549 human tumor xenografts. Histopathological analysis of Ad-DeltaB7-U6shIL8-treated tumors revealed an increase in apoptotic cells and a reduction in vessel density. Finally, Ad-DeltaB7-U6shIL8 was also shown to inhibit the growth of disseminated MDA-MB-231 breast cancer metastases. Taken together, these findings demonstrate the utility and antitumor effectiveness of oncolytic Ad expressing shRNA against IL-8.


Subject(s)
Adenoviridae/genetics , Breast Neoplasms/therapy , Genetic Therapy/methods , Interleukin-8/genetics , Oncolytic Virotherapy/methods , RNA, Small Interfering/administration & dosage , Animals , Cell Line, Tumor , Endothelial Cells/metabolism , Female , Gene Silencing , Genetic Engineering , Humans , Interleukin-8/analysis , Interleukin-8/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Promoter Regions, Genetic , RNA Interference , Transduction, Genetic/methods , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
13.
Cancer Res ; 61(6): 2404-8, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289105

ABSTRACT

Unlike vascular endothelial growth factor (VEGF)-A, the effect of VEGF-C on tumor angiogenesis, vascular permeability, and leukocyte recruitment is not known. To this end, we quantified in vivo growth and vascular function in tumors derived from two VEGF-C-overexpressing (VC+) and mock-transfected cell lines (T241 fibrosarcoma and VEGF-A-/- embryonic stem cells) grown in murine dorsal skinfold chambers. VC+ tumors grew more rapidly than mock-transfected tumors and exhibited parallel increases in tumor angiogenesis. Furthermore, VEGF-C overexpression elevated vascular permeability in T241 tumors, but not in VEGF-A-/- tumors. Surprisingly, unlike VEGF-A, VEGF-C did not increase leukocyte rolling or adhesion in tumor vessels. Administration of VEGF receptor (VEGFR)-2 neutralizing antibody DC101 reduced vascular density and permeability of both VC+ and mock-transduced T241 tumors. These data suggest that VEGFR-2 signaling is critical for tumor angiogenesis and vascular permeability and that VEGFR-3 signaling does not compensate for VEGFR-2 blockade. An alternate VEGFR, VEGFR-1 or neuropilin-1, may modulate adhesion of leukocytes to tumor vessels.


Subject(s)
Endothelial Growth Factors/physiology , Leukocytes/pathology , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/physiopathology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Animals , Capillary Permeability/physiology , Cell Communication/physiology , Cell Division/physiology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/pathology , Mice , Mice, SCID , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA/biosynthesis , RNA/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C
14.
Oncogene ; 35(28): 3718-28, 2016 07 14.
Article in English | MEDLINE | ID: mdl-26568304

ABSTRACT

The multifunctional enzyme transglutaminase 2 (TG2) primarily catalyzes cross-linking reactions of proteins via (γ-glutamyl) lysine bonds. Several recent findings indicate that altered regulation of intracellular TG2 levels affects renal cancer. Elevated TG2 expression is observed in renal cancer. However, the molecular mechanism underlying TG2 degradation is not completely understood. Carboxyl-terminus of Hsp70-interacting protein (CHIP) functions as an ubiquitin E3 ligase. Previous studies reveal that CHIP deficiency mice displayed a reduced life span with accelerated aging in kidney tissues. Here we show that CHIP promotes polyubiquitination of TG2 and its subsequent proteasomal degradation. In addition, TG2 upregulation contributes to enhanced kidney tumorigenesis. Furthermore, CHIP-mediated TG2 downregulation is critical for the suppression of kidney tumor growth and angiogenesis. Notably, our findings are further supported by decreased CHIP expression in human renal cancer tissues and renal cancer cells. The present work reveals that CHIP-mediated TG2 ubiquitination and proteasomal degradation represent a novel regulatory mechanism that controls intracellular TG2 levels. Alterations in this pathway result in TG2 hyperexpression and consequently contribute to renal cancer.


Subject(s)
Carcinoma, Renal Cell/metabolism , GTP-Binding Proteins/metabolism , Kidney Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Transglutaminases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Male , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Glutamine gamma Glutamyltransferase 2 , Proteolysis , Transglutaminases/genetics , Transplantation, Heterologous , Ubiquitin-Protein Ligases/genetics , Ubiquitination
15.
Clin Cancer Res ; 5(12): 3928-41, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10632322

ABSTRACT

Tumor-associated antigens are typically nonimmunogenic in cancer patients, "immune surveillance" having manifestly failed. The fact that most tumor antigens are normal human proteins presents significant obstacles to current cancer immunization approaches that researchers are presently striving to overcome. An alternative strategy bypasses immunization altogether by direct genetic alteration of autologous patient T cells, to create "designer T cells" specific to a particular antigen. Chimeric immunoglobulin-T cell receptors (IgTCR) with a specificity for carcinoembryonic antigen (CEA) were created to evaluate the optimal IgTCR structure for cancer therapy. Antigen-binding domains of a humanized antibody were combined with TCR signaling chains to yield four different chimeric IgTCR: single chain Fv fragment (sFv)-zeta, fragment antigen-binding (Fab)-zeta, sFv-epsilon, and Fab-epsilon. All of the IgTCR were well expressed on T cells, and all showed specific binding and activation, as demonstrated by IL-2 production on contact with immobilized or cellular CEA, excepting sFv-epsilon alone which was inert solely against cellular targets for steric reasons unique to this construct. In contrast to prior studies of isolated TCR chains that related increased tyrosine-based activation motifs in zeta as a reason for superior signaling potency, these tests are the first to show that epsilon and zeta are indistinguishable for T cell signaling when assayed in the context of the intact TCR complex. Further, Fab was equivalent to sFv as an IgTCR component for expression and antigen binding, establishing an important alternative for IgTCR antigen recognition because sFvs may often lose antigen affinity. When IgTCR was expressed on normal human T cells, cytotoxic potency was demonstrated at low E:T ratios, with T cell recycling and progressive tumor cell destruction. Contrary to recent speculations, these observations prove that high affinity TCR interactions are not an impediment to serial target engagement and disengagement by cytotoxic T cells. The multivalent intercellular interactions of target cell binding, activation, and cytotoxicity were resistant to inhibition by soluble CEA. These studies establish a potentially important new immunotherapeutic modality for the treatment of CEA-expressing tumors.


Subject(s)
Carcinoembryonic Antigen/immunology , Epitopes, T-Lymphocyte/immunology , Immunotherapy, Adoptive , T-Lymphocytes/immunology , CD3 Complex/immunology , Carcinoembryonic Antigen/biosynthesis , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/therapy , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Immunoglobulins/metabolism , Interleukin-2/metabolism , Jurkat Cells , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Solubility , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Tumor Cells, Cultured
16.
Neoplasia ; 2(5): 449-59, 2000.
Article in English | MEDLINE | ID: mdl-11191112

ABSTRACT

Immunoglobulin T-cell receptors (IgTCRs) combine the specificity of antibodies with the potency of cellular killing by grafting antibody recognition domains onto TCR signaling chains. IgTCR-modified T cells are thus redirected to kill tumor cells based on their expression of intact antigen on cell surfaces, bypassing the normal mechanism of activation through TCR-peptide-major histocompatibility complex (MHC) recognition. Melanoma is one of the most immunoresponsive of human cancers and has served as a prototype for the development of a number of immunotherapies. The target antigen for this study is the ganglioside GD3, which is highly expressed on metastatic melanoma with only minor immunologic cross-reaction with normal tissues. To determine an optimal configuration for therapy, four combinations of IgTCRs were prepared and studied: sFv-epsilon, sFv-zeta, Fab-epsilon, Fab-zeta. These were expressed on the surface of human T cells by retroviral transduction. IgTCR successfully redirected T-cell effectors in an MHC-unrestricted manner, in this case against a non-T-dependent antigen, with specific binding, activation, and cytotoxicity against GD3+ melanoma cells. Soluble GD3 in concentrations up to 100 microg/ml did not interfere with recognition and binding of membrane-bound antigen. Based on the outcomes of these structural and functional tests, the sFv-zeta construct was selected for clinical development. These results demonstrate key features that emphasize the potential of anti-GD3 IgTCR-modified autologous T cells for melanoma therapies.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Chemotaxis, Leukocyte/physiology , Gangliosides/immunology , Melanoma/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/cytology , Animals , Antibodies, Monoclonal/genetics , Binding Sites , Cell Line , Cytotoxicity, Immunologic , Genes, Immunoglobulin , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunotherapy, Adoptive , Interleukin-2/biosynthesis , Jurkat Cells , Kidney , Membrane Proteins/genetics , Mice , Rats , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Transfection
17.
Cell Death Dis ; 5: e1112, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24625971

ABSTRACT

Although curcumin suppresses the growth of a variety of cancer cells, its poor absorption and low systemic bioavailability have limited its translation into clinics as an anticancer agent. In this study, we show that dimethoxycurcumin (DMC), a methylated, more stable analog of curcumin, is significantly more potent than curcumin in inducing cell death and reducing the clonogenicity of malignant breast cancer cells. Furthermore, DMC reduces the tumor growth of xenografted MDA-MB 435S cells more strongly than curcumin. We found that DMC induces paraptosis accompanied by excessive dilation of mitochondria and the endoplasmic reticulum (ER); this is similar to curcumin, but a much lower concentration of DMC is required to induce this process. DMC inhibits the proteasomal activity more strongly than curcumin, possibly causing severe ER stress and contributing to the observed dilation. DMC treatment upregulates the protein levels of CCAAT-enhancer-binding protein homologous protein (CHOP) and Noxa, and the small interfering RNA-mediated suppression of CHOP, but not Noxa, markedly attenuates DMC-induced ER dilation and cell death. Interestingly, DMC does not affect the viability, proteasomal activity or CHOP protein levels of human mammary epithelial cells, suggesting that DMC effectively induces paraptosis selectively in breast cancer cells, while sparing normal cells. Taken together, these results suggest that DMC triggers a stronger proteasome inhibition and higher induction of CHOP compared with curcumin, giving it more potent anticancer effects on malignant breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Curcumin/analogs & derivatives , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Transcription Factor CHOP/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/pharmacology , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Time Factors , Transcription Factor CHOP/genetics , Transfection , Tumor Burden/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
18.
Cancer Gene Ther ; 20(2): 70-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23306610

ABSTRACT

Recurrent or metastatic cancer in most cases remains an incurable disease, and thus alternative treatment strategies, such as oncolytic virotherapy, are of great interest for clinical application. Oncolytic adenoviruses (Ads) have many advantages as virotherapeutic agents and have been safely employed in the clinics. However, the efficacy of oncolytic Ads is insufficient to eradicate tumors and current clinical applications are restricted to local administration against primary tumors because of immunological obstacles and poor tumor-cell targeting. Thus, alternative viable approaches are needed to establish therapies based on oncolytic Ad that will eliminate both primary and metastatic cancers. To this end, rational design of oncolytic Ads that express immunostimulatory genes has been employed. Even when restricted to local viral delivery, these oncolytic Ad-based immunotherapeutics have been shown to exert systemic antitumor immunity and result in eradication of both primary and metastatic cancers. Moreover, oncolytic Ad-based immunotherapeutics in combination with either dendritic cell-based vaccine or radiotherapy further strengthen the systemic tumor-specific immunity, resulting in complete suppression of both local and distant tumor metastatic growth. This review will focus on the most recent updates in strategies to develop potent oncolytic Ad-based immunotherapeutics for use in cancer gene therapy.


Subject(s)
Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Oncolytic Virotherapy , Oncolytic Viruses , Adenoviridae/genetics , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Humans , Immunotherapy , Neoplasm Metastasis/immunology , Neoplasms/immunology , Neoplasms/pathology , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Xenograft Model Antitumor Assays
19.
Cancer Gene Ther ; 19(11): 741-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23018622

ABSTRACT

The development of effective treatments that enable many patients suffering from cancer to be successfully cured is highly demanded. Angiogenesis, which is a process for the formation of new capillary blood vessels, has a crucial role in solid tumor progression and the development of metastasis. Antiangiogenic therapy designed to prevent tumor angiogenesis, thereby arresting the growth or spread of tumors, has emerged as a non-invasive and safe option for cancer treatment. Due to the fact that integrin receptors are overexpressed on the surface of angiogenic endothelial cells, various strategies have been made to develop targeted delivery systems for cancer gene therapy utilizing integrin-targeting peptides with an exposed arginine-glycine-aspartate (RGD) sequence. The aim of this review is to summarize the progress and prospect of RGD-functionalized nonviral vectors toward targeted delivery of genetic materials in order to achieve an efficient therapeutic outcome for cancer gene therapy, including antiangiogenic therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Genetic Vectors/metabolism , Neoplasms/therapy , Neovascularization, Pathologic/therapy , Oligopeptides/administration & dosage , Amino Acid Sequence , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Animals , Endothelium/metabolism , Endothelium/pathology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Integrins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Oligopeptides/therapeutic use , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/therapeutic use , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/therapeutic use
20.
Cell Death Differ ; 18(4): 589-601, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21052095

ABSTRACT

Collaborator of ARF (CARF) was cloned as an ARF-interacting protein and shown to regulate the p53-p21(WAF1)-HDM2 pathway, which is central to tumor suppression via senescence and apoptosis. We had previously reported that CARF inhibition in cancer cells led to polyploidy and caspase-dependent apoptosis, however, the mechanisms governing this phenomenon remained unknown. Thus, we examined various cell death and survival pathways including the mitochondrial stress, ataxia telangiectasia mutated (ATM)-ATR, Ras-MAP kinase and retinoblastoma cascades. We found that CARF is a pleiotropic regulator with widespread effects; its suppression affected all investigated pathways. Most remarkably, it protected the cells against genotoxicity; CARF knockdown elicited DNA damage response as evidenced by increased levels of phosphorylated ATM and γH2AX, leading to induction of mitotic arrest and eventual apoptosis. We also show that the CARF-silencing-induced apoptosis in vitro translates to in vivo. In a human tumor xenograft mouse model, treatment of developing tumors with short hairpin RNA (shRNA) against CARF via an adenovirus carrier induced complete suppression of tumor growth, suggesting that CARF shRNA is a strong candidate for an anticancer reagent. We demonstrate that CARF has a vital role in genome preservation and tumor suppression and CARF siRNA is an effective novel cancer therapeutic agent.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Neoplasms/therapy , RNA-Binding Proteins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Repair , DNA-Binding Proteins/metabolism , Histones/metabolism , Humans , Mice , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Signal Transduction , Transplantation, Heterologous , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL