93 results on '"Chinghai Kao"'
Search Results
2. Supplementary Fig. 1 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Specificity of TFE3 antibody across TFE3 oncoproteins
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table S7. from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
A complete list of targeted genes in the PI3K/AKT signaling pathway with their associated miRNA from cluster 3.
- Published
- 2023
- Full Text
- View/download PDF
4. Table S1 from Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy
- Author
-
Roberto Pili, Scott I. Abrams, Dominic Smiraglia, David E. Nelson, Timothy L. Ratliff, Bennett D. Elzey, Chinghai Kao, Luigi Fontana, Michael Ciesielski, Sreenivasulu Chintala, May Elbanna, Hayley Affronti, Remi Adelaiye-Ogala, Li Shen, Nur P. Damayanti, and Ashley Orillion
- Abstract
List of chemokines/cytokines altered with PR/MR
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure Legend from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Supplementary Figure Legend
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figures S1-S10 from Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy
- Author
-
Roberto Pili, Scott I. Abrams, Dominic Smiraglia, David E. Nelson, Timothy L. Ratliff, Bennett D. Elzey, Chinghai Kao, Luigi Fontana, Michael Ciesielski, Sreenivasulu Chintala, May Elbanna, Hayley Affronti, Remi Adelaiye-Ogala, Li Shen, Nur P. Damayanti, and Ashley Orillion
- Abstract
Additional data on the role of PR/MR on tumor microenvironment
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer. AR expression has also been reported in other solid tumors, including renal cell carcinoma (RCC), but its biological role here remains unclear. Through integrative analysis of a reverse phase protein array, we discovered increased expression of AR in an RCC patient–derived xenograft model of acquired resistance to the receptor tyrosine kinase inhibitor (RTKi) sunitinib. AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro. An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib-induced AR transcriptional activity was associated with increased phosphorylation of serine 81 (pS81) on AR. Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Enzalutamide-induced AR degradation was rescued by either proteasome inhibition or by knockdown of the AR ubiquitin ligase speckle-type POZ protein (SPOP). In vivo treatment with enzalutamide and sunitinib demonstrated that this combination efficiently induced tumor regression in a RCC model following acquired sunitinib resistance. Overall, our results suggest the potential role of AR as a target for therapeutic interventions, in combination with RTKi, to overcome drug resistance in RCC.Significance: These findings highlight the therapeutic potential of targeting the androgen receptor to overcome RCC resistance to receptor tyrosine kinase inhibitors. Cancer Res; 78(11); 2886–96. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Purpose:Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease.Experimental Design:We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models.Results:The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3–tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation.Conclusions:These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3–tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3–tRCC.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Fig. 3 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Pie chart for TFE3 binding sites
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy
- Author
-
Roberto Pili, Scott I. Abrams, Dominic Smiraglia, David E. Nelson, Timothy L. Ratliff, Bennett D. Elzey, Chinghai Kao, Luigi Fontana, Michael Ciesielski, Sreenivasulu Chintala, May Elbanna, Hayley Affronti, Remi Adelaiye-Ogala, Li Shen, Nur P. Damayanti, and Ashley Orillion
- Abstract
Purpose:Diet and healthy weight are established means of reducing cancer incidence and mortality. However, the impact of diet modifications on the tumor microenvironment and antitumor immunity is not well defined. Immunosuppressive tumor-associated macrophages (TAMs) are associated with poor clinical outcomes and are potentially modifiable through dietary interventions. We tested the hypothesis that dietary protein restriction modifies macrophage function toward antitumor phenotypes.Experimental Design:Macrophage functional status under different tissue culture conditions and in vivo was assessed by Western blot, immunofluorescence, qRT-PCR, and cytokine array analyses. Tumor growth in the context of protein or amino acid (AA) restriction and immunotherapy, namely, a survivin peptide–based vaccine or a PD-1 inhibitor, was examined in animal models of prostate (RP-B6Myc) and renal (RENCA) cell carcinoma. All tests were two-sided.Results:Protein or AA-restricted macrophages exhibited enhanced tumoricidal, proinflammatory phenotypes, and in two syngeneic tumor models, protein or AA-restricted diets elicited reduced TAM infiltration, tumor growth, and increased response to immunotherapies. Further, we identified a distinct molecular mechanism by which AA-restriction reprograms macrophage function via a ROS/mTOR-centric cascade.Conclusions:Dietary protein restriction alters TAM activity and enhances the tumoricidal capacity of this critical innate immune cell type, providing the rationale for clinical testing of this supportive tool in patients receiving cancer immunotherapies.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Fig. 6 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Effect of different rapamycin concentrations
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Video S2. from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Enzalutamide prevents Sunitinib induced AR nuclear localization. Real-time fluorescence live cell imaging of 786-0 cells expressing AR-EGFP pretreated with 500 nM Enzalutamide for 30 minutes does not show increase in AR nuclear localization following Sunitinib treatment (5uM Sunitinib t=30s).
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Fig. 5 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Enriched KEGG PI3K/AKT signaling pathway visualization
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Fig. 4 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Pie chart for TFE3 binding sites
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Fig. 2 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
ChiP-seq for TFE3 in UOK-146
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Fig. 8 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Schema for multimodal inhibition in tRCC
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figures from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Enzalutamide modulates the protein expression of sunitinib-induced AR regulated genes.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure Legends from Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy
- Author
-
Roberto Pili, Scott I. Abrams, Dominic Smiraglia, David E. Nelson, Timothy L. Ratliff, Bennett D. Elzey, Chinghai Kao, Luigi Fontana, Michael Ciesielski, Sreenivasulu Chintala, May Elbanna, Hayley Affronti, Remi Adelaiye-Ogala, Li Shen, Nur P. Damayanti, and Ashley Orillion
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figures 1-3 from Androgen-Independent Molecular Imaging Vectors to Detect Castration-Resistant and Metastatic Prostate Cancer
- Author
-
Lily Wu, Chinghai Kao, Liu H. Wei, Makoto Sato, and Ziyue Karen Jiang
- Abstract
PDF file - 729K
- Published
- 2023
- Full Text
- View/download PDF
20. Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Venkata Nithinsai Chintala, George M. Yousef, Sreenivasulu Chintala, Anthony C. Wood, Nur P. Damayanti, Remi Adelaiye-Ogala, Chinghai Kao, Sheng-Yu Ku, Mary W. Ferris, Roberto Pili, Peter C. Hollenhorst, May Elbanna, Justin A. Budka, Eric C. Kauffman, Heba W.Z. Khella, Ashley Orillion, W. Marston Linehan, and Khunsha Ahmed
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Oncogene Proteins, Fusion ,Antineoplastic Agents ,Biology ,Article ,Deep sequencing ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Biomarkers, Tumor ,Animals ,Humans ,Gene silencing ,Gene Silencing ,Carcinoma, Renal Cell ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Regulation of gene expression ,Binding Sites ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,TOR Serine-Threonine Kinases ,Xenograft Model Antitumor Assays ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Insulin Receptor Substrate Proteins ,Cancer research ,TFEB ,Female ,Chromatin immunoprecipitation ,Protein Binding ,Signal Transduction - Abstract
Purpose: Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease. Experimental Design: We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models. Results: The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3–tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation. Conclusions: These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3–tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3–tRCC.
- Published
- 2018
- Full Text
- View/download PDF
21. Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma
- Author
-
Ayumi Hashimoto, Peter Ordentlich, Ashley Orillion, Roberto Pili, Chinghai Kao, Bennett D. Elzey, Sreevani Arisa, Remi Adelaiye-Ogala, Dmitry I. Gabrilovich, Sreenivasulu Chintala, Nur P. Damayanti, and Li Shen
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Pyridines ,medicine.drug_class ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Biology ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Carcinoma, Non-Small-Cell Lung ,Immune Tolerance ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Carcinoma, Renal Cell ,Tumor microenvironment ,Entinostat ,Myeloid-Derived Suppressor Cells ,Histone deacetylase inhibitor ,Immunotherapy ,Histone Deacetylase Inhibitors ,Disease Models, Animal ,030104 developmental biology ,Cytokine ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Benzamides ,Immunology ,Cancer research ,biology.protein ,Myeloid-derived Suppressor Cell - Abstract
Purpose: Recent advances in immunotherapy highlight the antitumor effects of immune checkpoint inhibition despite a relatively limited subset of patients receiving clinical benefit. The selective class I histone deacetylase inhibitor entinostat has been reported to have immunomodulatory activity including targeting of immune suppressor cells in the tumor microenvironment. Thus, we decided to assess whether entinostat could enhance anti–PD-1 treatment and investigate those alterations in the immunosuppressive tumor microenvironment that contribute to the combined antitumor activity. Experimental Design: We utilized syngeneic mouse models of lung (LLC) and renal cell (RENCA) carcinoma and assessed immune correlates, tumor growth, and survival following treatment with entinostat (5 or 10 mg/kg, p.o.) and a PD-1 inhibitor (10 and 20 mg/kg, s.c.). Results: Entinostat enhanced the antitumor effect of PD-1 inhibition in two syngeneic mouse tumor models by reducing tumor growth and increasing survival. Entinostat inhibited the immunosuppressive function of both polymorphonuclear (PMN)- and monocytic-myeloid derived suppressor cell (M-MDSC) populations. Analysis of MDSC response to entinostat revealed significantly reduced arginase-1, iNOS, and COX-2 levels, suggesting potential mechanisms for the altered function. We also observed significant alterations in cytokine/chemokine release in vivo with a shift toward a tumor-suppressive microenvironment. Conclusions: Our results demonstrate that entinostat enhances the antitumor effect of PD-1 targeting through functional inhibition of MDSCs and a transition away from an immune-suppressive tumor microenvironment. These data provide a mechanistic rationale for the clinical testing and potential markers of response of this novel combination in solid tumor patients. Clin Cancer Res; 23(17); 5187–201. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
22. Overcoming cellular senescence in human cancer pathogenesis
- Author
-
YEager, Thomas R., DeVries, Sandy, Jarrard, David F., Chinghai Kao, Nakada, Stepehn Y., Moon, Timothy D., Bruskewitz, Reginald, Stadler, Walter M., Meisner, Lorraine F., Gilchrist, Kennedy W., Newton, Michael A., Waldman, Frederic M., and Reznikoff, Catherine A.
- Subjects
Cells -- Aging ,Cancer -- Genetic aspects ,Biological sciences - Abstract
Overcoming cellular senescence and a minimum of two genetic changes in several combinations seem essential for the human cancer pathogenesis. The genetic changes should include either p16 or pRb loss and involve an alteration such as +20q11-q12 or -8p21-pter. The research details are provided.
- Published
- 1998
23. Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy
- Author
-
Hayley C. Affronti, Ashley Orillion, Sreenivasulu Chintala, Dominic J. Smiraglia, David E. Nelson, Nur P. Damayanti, Michael J. Ciesielski, Chinghai Kao, Luigi Fontana, Remi Adelaiye-Ogala, May Elbanna, Li Shen, Timothy L. Ratliff, Bennett D. Elzey, Roberto Pili, and Scott I. Abrams
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Mice, Transgenic ,Proinflammatory cytokine ,Immunomodulation ,03 medical and health sciences ,Mice ,Western blot ,Cell Line, Tumor ,Neoplasms ,Survivin ,Diet, Protein-Restricted ,Polyamines ,Medicine ,Animals ,Humans ,Amino Acids ,Tumor microenvironment ,Innate immune system ,medicine.diagnostic_test ,business.industry ,Macrophages ,Immunotherapy ,Macrophage Activation ,Gastrointestinal Microbiome ,Disease Models, Animal ,030104 developmental biology ,Cytokine ,Oncology ,Cell culture ,Cancer research ,Cytokines ,Dietary Proteins ,business - Abstract
Purpose: Diet and healthy weight are established means of reducing cancer incidence and mortality. However, the impact of diet modifications on the tumor microenvironment and antitumor immunity is not well defined. Immunosuppressive tumor-associated macrophages (TAMs) are associated with poor clinical outcomes and are potentially modifiable through dietary interventions. We tested the hypothesis that dietary protein restriction modifies macrophage function toward antitumor phenotypes. Experimental Design: Macrophage functional status under different tissue culture conditions and in vivo was assessed by Western blot, immunofluorescence, qRT-PCR, and cytokine array analyses. Tumor growth in the context of protein or amino acid (AA) restriction and immunotherapy, namely, a survivin peptide–based vaccine or a PD-1 inhibitor, was examined in animal models of prostate (RP-B6Myc) and renal (RENCA) cell carcinoma. All tests were two-sided. Results: Protein or AA-restricted macrophages exhibited enhanced tumoricidal, proinflammatory phenotypes, and in two syngeneic tumor models, protein or AA-restricted diets elicited reduced TAM infiltration, tumor growth, and increased response to immunotherapies. Further, we identified a distinct molecular mechanism by which AA-restriction reprograms macrophage function via a ROS/mTOR-centric cascade. Conclusions: Dietary protein restriction alters TAM activity and enhances the tumoricidal capacity of this critical innate immune cell type, providing the rationale for clinical testing of this supportive tool in patients receiving cancer immunotherapies.
- Published
- 2018
24. Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Remi Adelaiye-Ogala, Ashley Orillion, Sreenivasulu Chintala, Sreevani Arisa, Nur P. Damayanti, Chinghai Kao, Mark Titus, and Roberto Pili
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Mice, SCID ,SPOP ,urologic and male genital diseases ,Article ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Nitriles ,Phenylthiohydantoin ,medicine ,Sunitinib ,Enzalutamide ,Animals ,Humans ,Pyrroles ,Phosphorylation ,Carcinoma, Renal Cell ,Protein Kinase Inhibitors ,Gene knockdown ,biology ,Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,female genital diseases and pregnancy complications ,Kidney Neoplasms ,Ubiquitin ligase ,Androgen receptor ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Benzamides ,biology.protein ,Cancer research ,Female ,Tissue Kallikreins ,medicine.drug ,Signal Transduction - Abstract
Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer. AR expression has also been reported in other solid tumors, including renal cell carcinoma (RCC), but its biological role here remains unclear. Through integrative analysis of a reverse phase protein array, we discovered increased expression of AR in an RCC patient–derived xenograft model of acquired resistance to the receptor tyrosine kinase inhibitor (RTKi) sunitinib. AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro. An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib-induced AR transcriptional activity was associated with increased phosphorylation of serine 81 (pS81) on AR. Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Enzalutamide-induced AR degradation was rescued by either proteasome inhibition or by knockdown of the AR ubiquitin ligase speckle-type POZ protein (SPOP). In vivo treatment with enzalutamide and sunitinib demonstrated that this combination efficiently induced tumor regression in a RCC model following acquired sunitinib resistance. Overall, our results suggest the potential role of AR as a target for therapeutic interventions, in combination with RTKi, to overcome drug resistance in RCC. Significance: These findings highlight the therapeutic potential of targeting the androgen receptor to overcome RCC resistance to receptor tyrosine kinase inhibitors. Cancer Res; 78(11); 2886–96. ©2018 AACR.
- Published
- 2017
25. The role of interleukin-12 on modulating myeloid-derived suppressor cells, increasing overall survival and reducing metastasis
- Author
-
Catherine E. Steding, Meei Huey Jeng, Bennett D. Elzey, Chinghai Kao, Yanping Zhang, and Sung tse Wu
- Subjects
biology ,Immunology ,medicine.disease ,Metastasis ,Nitric oxide synthase ,Immune system ,Cell culture ,medicine ,Cancer research ,Interleukin 12 ,biology.protein ,Myeloid-derived Suppressor Cell ,Immunology and Allergy ,Macrophage ,CD8 - Abstract
Myeloid-derived suppressor cells (MDSC) are important to the tumour microenvironment as they actively suppress the immune system and promote tumour progression and metastasis. These cells block T-cell activation in the tumour microenvironment, preventing anti-tumour immune activity. The ability of a treatment to alter the suppressive function of these cells and promote an immune response is essential to enhancing overall therapeutic efficacy. Interleukin-12 (IL-12) has the potential not only to promote anti-tumour immune responses but also to block the activity of cells capable of immune suppression. This paper identifies a novel role for IL-12 as a modulator of MDSC activity, with implications for IL-12 as a therapeutic agent. Treatment with IL-12 was found to alter the suppressive function of MDSC by fundamentally altering the cells. Interleukin-12-treated MDSC exhibited up-regulation of surface markers indicative of mature cells as well as decreases in nitric oxide synthase and interferon-γ mRNA both in vitro and in vivo. Treatment with IL-12 was also found to have significant therapeutic benefit by decreasing the percentage of MDSC in the tumour microenvironment and increasing the percentage of active CD8(+) T cells. Treatment with IL-12 resulted in an increase in overall survival accompanied by a reduction in metastasis. The findings in this paper identify IL-12 as a modulator of immune suppression with significant potential as a therapeutic agent for metastatic breast cancer.
- Published
- 2011
- Full Text
- View/download PDF
26. Abstract 3109: Investigating the role of wild-type TFE3 in renal cell carcinoma cells harboring TFE3 fusions with spliceosome machinery associated genes
- Author
-
Roberto Pili, May Elbanna, Chinghai Kao, Nur P. Damayanti, and Khunsha Ahmed
- Subjects
Cancer Research ,Gene knockdown ,Stromal cell ,Cell ,TFE3 ,Biology ,Cell biology ,Fusion gene ,medicine.anatomical_structure ,Oncology ,RNA interference ,Cancer cell ,medicine ,Ectopic expression - Abstract
Background: Translocation Renal Cell Carcinoma (tRCC) represents an aggressive subtype of kidney cancer associated with various gene fusions involving translocation of one of two members of micropthalmia transcription factor (MiT) family, TFE3 or TFEB. Despite the identification of multiple TFE3 gene fusions in tRCC, heterogeneous phenotype and various dysregulated signaling pathways resulting from variety of gene fusion partners pose a challenge to establish effective treatments for these patients. In this work, we assessed the role of wild type TFE3 (wt-TFE3) in RCC cell bearing TFE3 fusion with genes associated with pre-mRNA splicing factor machinery (NONO, SFPQ and PRCC).Methods: Endogenous SFPQ-TFE3 expressing cells, RP-RO7, were generated from patient derived xenograft established in NSG mice. UOK-109 and UOK-146 (kindly provided by Dr. Marston Lenehan, NCI) were used as endogenous NONO-TFE3 and PRCC-TFE3 expressing cells, respectively. RNA interference (RNAi) mediated knockdown with differential exon targeting strategy was employed to study wt-TFE3 loss of function in RP-R07, UOK-109 and UOK-146, respectively. Real time monitoring of cell viability assay with multiplex readout were developed to investigate the effect of wt-TFE3 loss of function in 2D and 3D culture system. Two different 3D models were used;1) Tumor growth model, in which cancer cell interaction with extracellular matrix and stromal cells were represented in a multiculture-spheroid system; 2) Invasion model, in which cell ability to invade basement membrane barrier was modeled with matrix restricted spheroid, and the invasion trajectories were observed and quantified. Exogenous expression of wt-TFE3-EGFP was utilized to study the protein subcellular localization in RP-R07, UOK-109 and UOK-146 2D cultures and 3D culture system.Results: Consistent with the expression of chimeric TFE3, wt-TFE3-EGFP ectopic expression demonstrated strong nuclear localization in RP-R07, UOK-109 and UOK-146. Multiplex readout of cells viability assay showed that transient loss of wt-TFE3 in RP-R07, UOK-109 and UOK-146 curtails the proliferation rate of these cells in 2D and 3D models in fusion partner dependent manner (P Citation Format: Nur P. Damayanti, Khunsha Ahmed, May Elbanna, Chinghai Kao, Roberto Pili. Investigating the role of wild-type TFE3 in renal cell carcinoma cells harboring TFE3 fusions with spliceosome machinery associated genes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3109.
- Published
- 2018
- Full Text
- View/download PDF
27. Docetaxel increases antitumor efficacy of oncolytic prostate-restricted replicative adenovirus by enhancing cell killing and virus distribution
- Author
-
Meei Huey Jeng, Yong Tang, Xiong Li, Chinghai Kao, Youhong Liu, and Phipps Roger
- Subjects
Male ,Oncolytic adenovirus ,Genetic enhancement ,Genetic Vectors ,Docetaxel ,Biology ,urologic and male genital diseases ,Virus Replication ,medicine.disease_cause ,p38 Mitogen-Activated Protein Kinases ,Article ,Virus ,Adenoviridae ,Genes, Reporter ,Cell Line, Tumor ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Transgenes ,Promoter Regions, Genetic ,Molecular Biology ,Genetics (clinical) ,Cell Death ,Integrin beta1 ,Integrin beta3 ,Prostatic Neoplasms ,Genetic Therapy ,Oncolytic virus ,Enzyme Activation ,Gene Expression Regulation, Neoplastic ,Cell killing ,Apoptosis ,Cancer research ,Molecular Medicine ,Taxoids ,Neoplasm Transplantation ,medicine.drug - Abstract
Background We explored multiple molecular mechanisms of the combination of docetaxel and an oncolytic prostate-restricted replication competent adenovirus (Ad) (PRRA) in advanced prostate cancer (PCa) models. The combinational therapy has potential to overcome the therapeutic limitations of poor virus distribution inside solid tumors. Methods We evaluated the effect of docetaxel on the antitumor efficacy and efficiency of virus transduction, transgene expression and virus distribution of PRRA in a prostate-specific antigen/prostate-specific membrane antigen-positive tumor xenograft model. We also evaluated the effect of docetaxel on apoptosis induction, cell killing and the efficiency of transgene expression and virus replication in vitro. Results Tumor growth inhibition was significantly enhanced when docetaxel was administrated before intratumor injection of PRRA. In vivo dual-photon microscopy and ex vivo fluorescence microscopy and immunohistochemistry showed that docetaxel increased transgene expression and expanded virus distribution. The combination of docetaxel and PRRA also increased cell apoptosis. In vitro, docetaxel significantly increased cell killing in PRRA-treated PCa cells. Docetaxel significantly increased Ad-mediated trangene expression independent of Ad binding receptors and replication capability. Docetaxel increased the activity of cytomegalovirus (CMV) promoter but not of a chimeric prostate-specific enhancer, resulting in higher transgene expression. The enhanced CMV promoter activity resulted from activation of p38 mitogen-activated protein kinase (MAPK) because inhibition of p38 MAPK blocked the docetaxel-induced increase in CMV promoter activity. Conclusions Combining docetaxel with an oncolytic PRRA improved therapeutic potential by expanding virus distribution and enhancing cell apoptosis and killing. These studies suggested a novel mechanism for enhancing the effect of therapeutic genes delivered by a PRRA. Copyright © 2010 John Wiley & Sons, Ltd.
- Published
- 2010
- Full Text
- View/download PDF
28. Suppression of Renal Cell Carcinoma Growth and Metastasis with Sustained Antiangiogenic Gene Therapy
- Author
-
Juan A. Jiménez, Catherine E. Steding, Kyung-Hee Bae, Matthew J. Mellon, Thomas A. Gardner, and Chinghai Kao
- Subjects
Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Recombinant Fusion Proteins ,Mice, Nude ,Malignancy ,Article ,Metastasis ,Metastasis Suppression ,Mice ,Renal cell carcinoma ,Cell Line, Tumor ,Genetics ,Carcinoma ,Animals ,Humans ,Medicine ,Angiostatins ,Carcinoma, Renal Cell ,Molecular Biology ,Kidney ,Neovascularization, Pathologic ,business.industry ,Liver Neoplasms ,Cancer ,Genetic Therapy ,medicine.disease ,Kidney Neoplasms ,Endostatins ,medicine.anatomical_structure ,Molecular Medicine ,business ,Kidney disease - Abstract
Renal cell carcinoma (RCC) is the third most common urologic neoplasm. This aggressive malignancy has proven refractory to conventional treatment options. Antiangiogenic agents have shown early success in treating metastatic disease. The highly vascular nature of RCC appears particularly susceptible to this approach. This study investigates the potential of sustained expression of an endostatin-angiostatin fusion protein in an early-stage model of RCC to inhibit tumor growth and metastasis. Subcutaneous RCC-29 tumors were induced in athymic nude mice. Once tumors reached volumes of 10 and 25 mm(3), subjects received intratumoral injections of a nonreplicating adenoviral vector every 20 days until the conclusion of the trial. The mice were randomly assigned to three treatment groups: saline control, viral Ad-GFP control, and Ad-EndoAngio. Tumor volumes were measured twice weekly for 80 days. During days 40-50 of the trial, subjects underwent dual-photon optical imaging of the tumor vasculature to ascertain angiogenic changes. All animals underwent postmortem histopathological analysis to assess for metastatic disease in the kidney, lung, liver, brain, and spleen. Results indicate that tumors treated with Ad-EndoAngio displayed 97% growth reduction compared with controls (p < 0.001). Further, in vivo tumor vascular imaging illustrated a reduction in blood vessel number and lumen diameter size. Kaplan-Meier analysis suggested dramatic survival advantage with Ad-EndoAngio treatment. Importantly, histopathological examination demonstrated marked lung and liver metastasis suppression in the treatment arms. These results suggest that sustained EndoAngio gene therapy has effective antiangiogenic action against human RCC tumors and possesses potential as a novel treatment for metastatic renal cell carcinoma.
- Published
- 2008
- Full Text
- View/download PDF
29. Long-Term Outcome of Phase I/II Clinical Trial of Ad-OC-TK/VAL Gene Therapy for Hormone-Refractory Metastatic Prostate Cancer
- Author
-
Leland W.K. Chung, Takatsugu Okegawa, Shuji Terao, Kohzo Fuji, Toshiro Shirakawa, Kazuro Sugimura, Katsuyuki Hamada, Kazushi Tanaka, Masafumi Matsuo, Chinghai Kao, Isao Hara, Sadao Kamidono, Atsushi Takenaka, Thomas A. Gardner, Akinobu Gotoh, Masato Fujisawa, Eiji Higashihara, and Nobuyuki Hinata
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Genetic Vectors ,Osteocalcin ,Acyclovir ,Antineoplastic Agents ,Bone Neoplasms ,Docetaxel ,Antiviral Agents ,Thymidine Kinase ,Bone and Bones ,Adenoviridae ,Metastasis ,Prostate cancer ,PSA Failure ,Internal medicine ,Genetics ,Humans ,Medicine ,Promoter Regions, Genetic ,Molecular Biology ,Aged ,business.industry ,Prostatic Neoplasms ,Bone metastasis ,Cancer ,Androgen Antagonists ,Valine ,Combination chemotherapy ,Genetic Therapy ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Surgery ,Radiography ,Valacyclovir ,Molecular Medicine ,Taxoids ,Estramustine ,business ,medicine.drug - Abstract
We evaluated the long-term safety and efficacy of Ad-OC-TK (recombinant adenoviral vector carrying an osteocalcin promoter-driven herpes simplex virus thymidine kinase gene) plus VAL (valacyclovir) gene therapy for hormone-refractory prostate cancer. Ad-OC-TK/VAL therapy is the first in vivo adenovirus-mediated gene therapy to be used to treat metastatic prostate cancer, including bone metastasis. Six patients were enrolled in this trial, and two doses of Ad-OC-TK (2.5 x 10(9) or 2.5 x 10(10) plaque-forming units) were injected into locally recurrent tumor or bone metastasis on day 1 and day 8. Patients were also given VAL (3 g/day) for 21 days. Safety and efficacy were evaluated for at least 8 months in each patient. All patients tolerated this therapy with no serious adverse events. One prostate-specific antigen (PSA) response (from 318.3 to 4.9 ng/ml) was observed with a time to PSA progression (TTP) of 12 months. Docetaxel (30 mg/m2 per week) and estramustine (560 mg/day) combination chemotherapy (DE) was given to three docetaxel-naive patients on PSA failure after gene therapy. All three patients had a PSA response to DE therapy with 21, 7, and 4 months of TTP. These results suggest that additional trials are warranted.
- Published
- 2007
- Full Text
- View/download PDF
30. Fas Ligand Delivery by a Prostate-Restricted Replicative Adenovirus Enhances Safety and Antitumor Efficacy
- Author
-
Meei Huey Jeng, Shaobo Zhang, Yan Ping Zhang, Thomas A. Gardner, Chinghai Kao, Xinzhu Pu, You Hong Liu, and Xiong Li
- Subjects
Glutamate Carboxypeptidase II ,Male ,Cancer Research ,Fas Ligand Protein ,medicine.medical_treatment ,Genetic Vectors ,Apoptosis ,Virus Replication ,Fas ligand ,Adenoviridae ,Mice ,Prostate cancer ,Immune system ,Antigen ,medicine ,Animals ,Humans ,business.industry ,Cytoplasmic Vesicles ,Prostate ,Prostatic Neoplasms ,Genetic Therapy ,Prostate-Specific Antigen ,medicine.disease ,Xenograft Model Antitumor Assays ,Cytokine ,Oncology ,Antigens, Surface ,Immunology ,Androgens ,Cancer research ,Systemic administration ,Tumor necrosis factor alpha ,business - Abstract
Purpose: Recent studies showed that Fas ligand (FasL) induced apoptosis in tumor cells and suppressed the immune response in several types of tumors. However, the toxicity of FasL limited further administration. This study delivered FasL in prostate cancer cells using an improved prostate-restricted replicative adenovirus (PRRA), thereby improving the antitumor effect while decreasing systemic toxicity. Experimental Design: We designed a FasL-armed PRRA, called AdIU3, by placing adenoviral E1a and E4 genes, FasL cDNA, and E1b gene under the control of two individual PSES enhancers. Tissue-specific viral replication and FasL expression were analyzed, and the tumor killing effect of AdIU3 was investigated both in vitro and in vivo using androgen-independent CWR22rv s.c. models via local administration and bone models via systemic administration. The safety of systemic administration of AdIU3 was evaluated. AdCMVFasL, in which FasL was controlled by a universal cytomegalovirus (CMV) promoter, was used as a control. Results: AdIU3 enhanced FasL expression in prostate-specific antigen (PSA)/prostate-specific membrane antigen (PSMA)-positive cells but not in PSA/PMSA-negative cells. It induced apoptosis and killed PSA/PMSA-positive prostate cancer cells but spared normal human fibroblasts, hepatocytes, and negative cells. The increase in killing activity was confirmed to result in part from a bystander killing effect. Furthermore, AdIU3 was more effective than a plain PRRA in inhibiting the growth of androgen-independent prostate cancer xenografts and bone tumor formation. Importantly, systemic administration of AdIU3 resulted in undetectable toxicity, whereas the same doses of AdCMVFasL killed all mice due to multiviscera failure in 16 h. Conclusions: AdIU3 decreased the toxicity of FasL by controlling its expression with PSES, with greatly enhanced prostate cancer antitumor efficacy. The results suggested that toxic antitumor factors can be delivered safely by a PRRA.
- Published
- 2007
- Full Text
- View/download PDF
31. Advances in Preclinical Investigation of Prostate Cancer Gene Therapy
- Author
-
Lily Wu, Chinghai Kao, and Marxa L. Figueiredo
- Subjects
Male ,Genetic enhancement ,Genetic Vectors ,Bioinformatics ,Models, Biological ,Article ,Viral vector ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Combined Modality Therapy ,Molecular Biology ,030304 developmental biology ,Pharmacology ,Regulation of gene expression ,0303 health sciences ,Reporter gene ,business.industry ,Prostatic Neoplasms ,Genetic Therapy ,medicine.disease ,3. Good health ,Oncolytic virus ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030220 oncology & carcinogenesis ,Immunology ,Molecular Medicine ,Recurrent prostate cancer ,business - Abstract
Treating recurrent prostate cancer poses a great challenge to clinicians. Research efforts in the last decade have shown that adenoviral vector-based gene therapy is a promising approach that could expand the arsenal against prostate cancer. This maturing field is at the stage of being able to translate many preclinical discoveries into clinical practices. At this juncture, it is important to highlight the promising strategies including prostate-targeted gene expression, the use of oncolytic vectors, therapy coupled to reporter gene imaging, and combined treatment modalities. In fact, the early stages of clinical investigation employing combined, multimodal gene therapy focused on loco-regional tumor eradication and showed promising results. Clinicians and scientists should seize the momentum of progress to push forward to improve the therapeutic outcome for the patients.
- Published
- 2007
- Full Text
- View/download PDF
32. A conditionally replicative, Wnt/β-catenin pathway-based adenovirus therapy for anaplastic thyroid cancer
- Author
-
Chinghai Kao, Kenneth P. Nephew, Thomas A. Gardner, Xiaochun Li, Lang Li, and Phillip H. Abbosh
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Genetic Vectors ,Mice, Nude ,Biology ,Virus Replication ,Adenoviridae ,Mice ,medicine ,Animals ,Thyroid Neoplasms ,Vector (molecular biology) ,Anaplastic thyroid cancer ,Molecular Biology ,Thyroid cancer ,beta Catenin ,Thyroid ,Wnt signaling pathway ,Genetic Therapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Wnt Proteins ,Cell killing ,medicine.anatomical_structure ,Viral replication ,Catenin ,Cancer research ,Molecular Medicine - Abstract
Thyroid cancer affects between 10,000 and 15,000 people per year in the US. Typically, this disease can be controlled with surgical resection and radioiodide treatment. However, resistance to these conventional therapies is observed in some patients, who develop intractable anaplastic thyroid cancer (ATC), for which no effective therapies exist. Recently, a sizable fraction of undifferentiated or poorly differentiated thyroid cancers were shown to contain mutations in beta-catenin, an oncogenic protein involved in the etiology of cancers of many tissues. We developed a conditionally replicative adenovirus (named 'HILMI') which, by virtue of TCF response elements drives E1A and E1B expression, replicates specifically in cells with an active Wnt/beta-catenin pathway. We show that several thyroid cancer cell lines, derived from undifferentiated or anaplastic tissues and possessing an active Wnt/beta-catenin pathway, are susceptible to cell killing by HILMI. Furthermore, viral replication in ATC cells as xenograft tumors in nude mice was observed, and prolonged survival of mice with ATC tumors was observed following administration of the HILMI therapeutic vector. The results warrant further development of this therapeutic approach for ATC patients.
- Published
- 2007
- Full Text
- View/download PDF
33. Adenoviral Vectors Expressing Human Endostatin–Angiostatin and Soluble Tie2: Enhanced Suppression of Tumor Growth and Antiangiogenic Effects in a Prostate Tumor Model
- Author
-
Bruce A. Molitoris, Chinghai Kao, Constance J. Temm, Nandita S. Raikwar, Thomas A. Gardner, and Sudhanshu P. Raikwar
- Subjects
Male ,Umbilical Veins ,Angiogenesis ,Angiogenesis Inhibitors ,Receptor tyrosine kinase ,Metastasis ,Mice ,Prostate cancer ,Drug Discovery ,Cells, Cultured ,Angiostatin ,Neovascularization, Pathologic ,biology ,Gene Transfer Techniques ,Receptor, TIE-2 ,Endostatins ,Drug Combinations ,Molecular Medicine ,Proteoglycans ,Collagen ,Endostatin ,Blotting, Western ,Genetic Vectors ,Disease-Free Survival ,Article ,Adenoviridae ,Cell Line ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Angiostatins ,Molecular Biology ,Cell Proliferation ,Pharmacology ,Photons ,Matrigel ,business.industry ,Prostatic Neoplasms ,Cancer ,Genetic Therapy ,medicine.disease ,Immunology ,Cancer research ,biology.protein ,Endothelium, Vascular ,Laminin ,business ,Neoplasm Transplantation - Abstract
Angiogenesis is essential for prostate cancer development and metastasis. Antiangiogenic therapy targeting tumor neovasculature, therefore, represents a promising approach for prostate cancer treatment. We hypothesized that adenoviral-mediated delivery of a combination of antiangiogenic factors might have an enhanced antitumor response. We developed the adenoviral vectors Ad-hEndo-angio, expressing a unique, chimeric human endostatin–angiostatin fusion protein, and Ad-sTie2, expressing a soluble form of endothelium-specific receptor tyrosine kinase Tie2. Matrigel angiogenesis assays using Ad-hEndo-angio revealed significant inhibition of tubular network formation and endothelial sprouting compared to Ad-sTie2. In vivo studies in a bilateral PC-3 tumor xenograft model following either intratumoral or systemic administration of Ad-hEndo-angio led to enhanced tumor growth suppression compared to Ad-sTie2. A novel finding is that an intratumoral, combination therapy employing one-half the dose of Ad-hEndo-angio as well as Ad-sTie2 led to a complete regression of the injected, as well as the contralateral uninjected, tumor and prolonged the tumor-free survival in 80% of the animals. In addition, a novel, real-time, intravital imaging modality was used to monitor antiangiogenic responses following adenoviral-mediated gene transfer. These results suggest that a combinatorial antiangiogenic gene therapy approach involving Ad-hEndo-angio and Ad-sTie2 could become a novel form of treatment for localized human prostate cancer.
- Published
- 2005
- Full Text
- View/download PDF
34. Transcriptional targeting modalities in breast cancer gene therapy using adenovirus vectors controlled by α-lactalbumin promoter
- Author
-
Jie Zhang, Thomas A. Gardner, Sudhanshu P. Raikwar, Kyung Hee Bae, Xiong Li, Sang Jin Lee, Huanling Gao, Chinghai Kao, Meei Huey Jeng, Yan Ping Zhang, Edyta Vieth, Dale VanderPutten, and Gary D. Hutchins
- Subjects
Male ,Cancer Research ,animal structures ,Genetic enhancement ,Blotting, Western ,Genetic Vectors ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Biology ,Virus Replication ,Adenoviridae ,Mice ,Breast cancer ,Antigen ,Cell Line, Tumor ,Gene expression ,medicine ,Animals ,Humans ,Adenovirus E1B Proteins ,Mammary Glands, Human ,Promoter Regions, Genetic ,skin and connective tissue diseases ,Gene ,DNA Primers ,Base Sequence ,Virulence ,Reverse Transcriptase Polymerase Chain Reaction ,Prostatic Neoplasms ,Cancer ,Genetic Therapy ,medicine.disease ,Molecular biology ,Oncology ,Viral replication ,Cell culture ,Lactalbumin ,Cancer research ,Female ,Adenovirus E1A Proteins - Abstract
The breast-specific antigen α-lactalbumin is expressed in >60% of breast cancer tissues. To evaluate the effect of gene therapy for breast cancer by controlling adenovirus replication with human α-lactalbumin promoter, we investigated the activity of a 762-bp human α-lactalbumin promoter. α-Lactalbumin promoter showed significantly higher activity in MDA-MB-435S and T47D breast cancer cells than in normal breast cell lines or other tumor cell lines. We then developed two novel breast cancer–restricted replicative adenoviruses, AdALAE1a and AdE1aALAE1b. In AdALAE1a, expression of adenoviral E1a gene is under the control of α-lactalbumin promoter, and in AdE1aALAE1b, expression of both E1a and E1b genes is under the control of a single α-lactalbumin promoter. Both breast cancer–restricted replicative adenoviruses showed viral replication efficiency and tumor cell-killing capability similar to wild-type adenovirus in MDA-MB-435S and T47D cells. The replication efficiency and tumor cell-killing capability of both viruses were attenuated significantly in cells that did not support α-lactalbumin promoter. AdE1aALAE1b showed better breast cancer–restricted replication than AdALAE1a, suggesting that a transcriptional targeting modality with α-lactalbumin promoter controlling both E1a and E1b gene expression is superior to α-lactalbumin promoter controlling only E1a gene expression. Importantly, we found that AdE1aALAE1b could be used to target hormone-independent breast tumors in vivo by inhibiting the growth of MDA-MB-435S s.c. tumors. These data showed that α-lactalbumin promoter could regulate the replication of adenovirus to target hormone-independent breast cancers, suggesting that α-lactalbumin promoter can be used to develop a novel therapeutic modality for hormone-independent breast cancer. [Mol Cancer Ther 2005;4(12):1850–9]
- Published
- 2005
- Full Text
- View/download PDF
35. Targeting Prostate Cancer with Conditionally Replicative Adenovirus Using PSMA Enhancer
- Author
-
Sang Jin Lee, Sang Don Lee, Xiong Li, Chaeyong Jung, Thomas A. Gardner, Hong Sup Kim, Yan-Ping Zhang, Meei Huey Jeng, Chinghai Kao, and Kyung Hee Bae
- Subjects
PCA3 ,Glutamate Carboxypeptidase II ,Male ,Transcription, Genetic ,viruses ,urologic and male genital diseases ,Virus Replication ,Adenoviridae ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,Cell Line, Tumor ,Drug Discovery ,medicine ,Genetics ,Humans ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,business.industry ,Virion ,Cancer ,Prostatic Neoplasms ,Genetic Therapy ,medicine.disease ,Virology ,3. Good health ,Blot ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Viral replication ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Molecular Medicine ,Adenovirus E1A Proteins ,business ,Neoplasm Transplantation - Abstract
Prostate cancer is the second most commonly diagnosed cancer in men and accounts for significant mortality and morbidity in the United States. Initially androgen-dependent, prostate cancer ultimately becomes androgen-independent, which makes the disease extremely difficult to cure. In this study, we examined the use of conditionally replication-competent adenovirus for the treatment of hormone-independent prostate cancer. We utilized PSME, an enhancer element for prostate-specific PSMA expression, to control viral E1A protein expression and achieve exclusive virus replication in prostate. Western blotting confirmed that PSME mediated high E1A protein expression in PSMA-positive, androgen-independent prostate cancer cells (C4-2 and CWR22rv), but was much less active in PSMA-negative cancer cells (PC-3 and A549). Consistent with E1A protein expression, the recombinant adenovirus Ad5-PSME-E1a replicated in C4-2 and CWR22rv almost as efficiently as wild type with low levels of androgen, but its replication was significantly attenuated in PSMA-negative cells. In the in vitro killing assay, Ad5-PSME-E1a lysed all C4-2 and CWR22rv cells 5 days after infection, with minimal effect on PSMA-negative cells. In addition, injections of 1.7 x 10(8) plaque-forming units in a CWR22rv xenograft model in nude mice induced significant tumor growth delay, with a substantial necrotic area. These studies suggest that PSME-driven replication-competent adenovirus may be a new therapeutic modality for prostate cancer patients after hormone ablation therapy.
- Published
- 2004
- Full Text
- View/download PDF
36. High-Level Expression of EphA2 Receptor Tyrosine Kinase in Prostatic Intraepithelial Neoplasia
- Author
-
Lang Li, Michael S. Kinch, Shaobo Zhang, Lee Ann Baldridge, John N. Eble, Thomas A. Gardner, Zhiqiang Hu, Thomas M. Ulbright, Chong-Xian Pan, Guangyuan Zeng, Liang Cheng, Chinghai Kao, Michael O. Koch, and David A. Flockhart
- Subjects
Male ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Adenocarcinoma ,Biology ,Receptor tyrosine kinase ,Pathology and Forensic Medicine ,Mice ,Prostate ,medicine ,Carcinoma ,Animals ,Humans ,Neoplastic transformation ,Prostatic Intraepithelial Neoplasia ,Intraepithelial neoplasia ,Prostatectomy ,Receptor, EphA2 ,Antibodies, Monoclonal ,Prostatic Neoplasms ,medicine.disease ,Immunohistochemistry ,medicine.anatomical_structure ,biology.protein ,Regular Articles - Abstract
EphA2 is a transmembrane receptor tyrosine kinase that is overexpressed in many carcinomas. Specific targeting of EphA2 with monoclonal antibodies is sufficient to inhibit the growth, migration and invasiveness of aggressive cancers in animal models. Using immunohistochemical analyses, we measured the expression of EphA2 in prostatic adenocarcinoma, high-grade prostatic intraepithelial neoplasia, and adjacent benign prostate tissue from ninety-three radical prostatectomy specimens. These results were related to multiple clinical and pathologicalcharacteristics. The fraction of cells staining positively with EphA2 in benign prostatic epithelium (mean, 12%) was significantly lower than that in high-grade prostatic intraepithelial neoplasia (mean, 67%, P < 0.001) and prostatic adenocarcinoma (mean, 85%, P < 0.001). Moreover, the intensity of EphA2 immunoreactivity in prostatic adenocarcinoma was significantly higher than in benign prostatic tissue (P < 0.001) or high-grade prostatic intraepithelial neoplasia (P < 0.001). Benign prostatic epithelium showed weak or no immunoreactivity for EphA2 in all cases examined. Whereas EphA2 immunoreactivity related to neoplastic transformation, it did not correlate with other clinical and pathological parameters examined. Our data suggest that EphA2 levels increase as prostatic epithelial cells progress toward a more aggressive phenotype. Progressively higher levels of EphA2 in high-grade prostatic intraepithelial neoplasia and prostatic carcinoma are consistent with recent evidence that EphA2 functions as a powerful oncogene. Moreover, the presence of high levels of EphA2 in these cells suggests opportunities for prostate cancer prevention and treatment.
- Published
- 2003
- Full Text
- View/download PDF
37. Phase I Dose Escalation Clinical Trial of Adenovirus Vector Carrying Osteocalcin Promoter-Driven Herpes Simplex Virus Thymidine Kinase in Localized and Metastatic Hormone-Refractory Prostate Cancer
- Author
-
Kenneth S. Koeneman, Akinobu Gotoh, Yoshitaka Wada, So Dug Lim, Shigeji Matsubara, Margaret E. Black, Leland W. K. Chung, Hua Yang, Jay Y. Gillenwater, Chinghai Kao, Ling Yang, Hiroyuki Kubo, Thomas A. Gardner, Masayuki Nakagawa, Haiyen E. Zhau, and Mahul B. Amin
- Subjects
Male ,PCA3 ,Stromal cell ,Genetic Vectors ,Osteocalcin ,Thymidine Kinase ,Adenoviridae ,Metastasis ,Prostate cancer ,Genetics ,Humans ,Simplexvirus ,Medicine ,Neoplasm Metastasis ,Promoter Regions, Genetic ,Molecular Biology ,Aged ,Dose-Response Relationship, Drug ,biology ,business.industry ,Prostatic Neoplasms ,Bone metastasis ,Cancer ,Middle Aged ,medicine.disease ,Thymidine kinase ,biology.protein ,Cancer research ,Molecular Medicine ,business - Abstract
Osteocalcin (OC), a major noncollagenous bone matrix protein, is expressed prevalently in prostate cancer epithelial cells, adjacent fibromuscular stromal cells, and osteoblasts in locally recurrent prostate cancer and prostate cancer bone metastasis [Matsubara, S., Wada, Y., Gardner, T.A., Egawa, M., Park, M.S., Hsieh, C.L., Zhau, H.E., Kao, C., Kamidono, S., Gillenwater, J.Y., and Chung, L.W. (2001). Cancer Res. 61, 6012-6019]. We constructed an adenovirus vector carrying osteocalcin promoter-driven herpes simplex virus thymidine kinase (Ad-OC-hsv-TK) to cotarget prostate cancer cells and their surrounding stromal cells. A phase I dose escalation clinical trial of the intralesional administration of Ad-OC-hsv-TK followed by oral valacyclovir was conducted at the University of Virginia (Charlottesville, VA) in 11 men with localized recurrent and metastatic hormone-refractory prostate cancer (2 local recurrent, 5 osseous metastasis, and 4 lymph node metastasis) in order to determine the usefulness of this vector for the palliation of androgen-independent prostate cancer metastasis. This is the first clinical trial in which therapeutic adenoviruses are injected directly into prostate cancer lymph node and bone metastasis. Results show that (1). all patients tolerated this therapy with no serious adverse events; (2). local cell death was observed in treated lesions in seven patients (63.6%) as assessed by TUNEL assay, and histomorphological change (mediation of fibrosis) was detected in all posttreated specimens; (3). one patient showed stabilization of the treated lesion for 317 days with no alternative therapy. Of the two patients who complained of tumor-associated symptoms before the treatment, one patient with bone pain had resolution of pain, although significant remission of treated lesions was not observed by image examination; (4). CD8-positive T cells were predominant compared with CD4-positive T cells, B cells (L26 positive), and natural killer cells (CD56 positive) in posttreated tissue specimens; (5). levels of HSV TK gene transduction correlated well with coxsackie-adenovirus receptor expression but less well with the titers of adenovirus injected; and (6). intrinsic OC expression and the efficiency of HSV TK gene transduction affected the levels of HSV TK protein expression in clinical specimens. Our data suggest that this form of gene therapy requires further development for the treatment of androgen-independent prostate cancer metastasis although histopathological and immunohistochemical evidence of apoptosis was observed in the specimens treated. Further studies including the development of viral delivery will enhance the efficacy of Ad-OC-hsv-TK.
- Published
- 2003
- Full Text
- View/download PDF
38. Novel Prostate-Specific Promoter Derived from PSA and PSMA Enhancers
- Author
-
Rong Yu, Chinghai Kao, Liang Cheng, Thomas A. Gardner, Chaeyong Jung, Meei Huey Jeng, Sang Jin Lee, Hong Sup Kim, Fan Yeung, and KangRyul Lee
- Subjects
Glutamate Carboxypeptidase II ,Male ,PCA3 ,Genetic enhancement ,Genetic Vectors ,Molecular Sequence Data ,Carboxypeptidases ,Biology ,urologic and male genital diseases ,Adenoviridae ,Viral vector ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,Drug Discovery ,Genetics ,medicine ,Glutamate carboxypeptidase II ,Humans ,Luciferase ,Promoter Regions, Genetic ,Molecular Biology ,030304 developmental biology ,Recombination, Genetic ,Pharmacology ,0303 health sciences ,Base Sequence ,Prostate-Specific Antigen ,medicine.disease ,Molecular biology ,3. Good health ,Enhancer Elements, Genetic ,medicine.anatomical_structure ,Organ Specificity ,Cell culture ,030220 oncology & carcinogenesis ,Antigens, Surface ,Cancer research ,Molecular Medicine - Abstract
The expression of prostate-specific membrane antigen (PSMA) and prostate-specific antigen (PSA), two well characterized marker proteins, remains highly active in the hormone refractory stage of prostate cancer. In this study, an artificial chimeric enhancer (PSES) composed of two modified regulatory elements controlling the expression of PSA and PSMA genes was tested for its promoter activity and tissue specificity using the reporter system. As a result, this novel PSES promoter remained silent in PSA- and PSMA-negative prostate and non-prostate cancer cell lines, but mediated high levels of luciferase in PSA- and PSMA-expressing prostate cancer cell lines in the presence and absence of androgen. To determine whether PSES could be used for in vivo gene therapy of prostate cancer, a recombinant adenovirus, Ad-PSES-luc, was constructed. Luciferase activity in prostate cancer cell lines mediated by Ad-PSES-luc was 400- to 1000-fold higher than in several other non-prostate cell lines, suggesting the high tissue-specificity of the PSES promoter in an adenoviral vector. Finally, recombinant virus Ad-PSES-luc was injected into mice to evaluate the tissue-discriminatory promoter activity in an experimental animal. Unlike Ad-CMV-luc, the luciferase activity from systemic injection of Ad-PSES-luc was fairly low in all major organs. However, when injected into prostate, Ad-PSES-luc drove high luciferase activity almost exclusively in prostate and not in other tissues. Our results demonstrated the potential use of PSES for the treatment of androgen-independent prostate cancer patients.
- Published
- 2002
- Full Text
- View/download PDF
39. Abstract 250: Methionine restriction increases macrophage tumoricidal activity and significantly inhibits prostate cancer growth
- Author
-
Roberto Pili, Luigi Fontana, May Elbanna, Sreevani Arisa, Remi Adelaiye-Ogala, Bennett D. Elzey, Li Shen, Ashley Orillion, Chinghai Kao, Sreenivasulu Chintala, and Nur P. Damayanti
- Subjects
Cancer Research ,Methionine ,medicine.medical_treatment ,Cancer ,Immunotherapy ,Biology ,medicine.disease ,Prostate cancer ,chemistry.chemical_compound ,Immune system ,Oncology ,Low-protein diet ,chemistry ,Immunology ,Cancer cell ,medicine ,Cancer research ,PI3K/AKT/mTOR pathway - Abstract
Background: Our previous work showed a significant reduction of tumor growth, macrophage infiltration, circulating IGF-1, and mTOR activation with low protein diet in a patient derived xenograft model of prostate cancer. The evolutionarily conserved, nutrient sensing, mTOR pathway plays a central role in both development of advanced stage prostate cancer and immune responsiveness. This study presents novel data on the impact of dietary protein modification on the function of the host immune system in response to prostate cancer and immunotherapy. Methods: Our In vitro studies utilized bone marrow or tumor derived (RP-B6 Myc) macrophages. In vivo studies utilized the recently characterized RP-B6 Myc model. Mice were fed ad libitum control or methionine restricted diets for four weeks prior to S.C. implantation with ~1mm2 tumor pieces. Treatment of survivin peptide vaccine (1mg/ml S.C. 1 X week) and anti-PD-1 (20mg/kg I.P. 2 X week) began at ~50mm2 tumor size. Tumor volumes were blindly recorded 2 X week. End point analyses include: tumor weight, flow cytometric analysis, proteomic profiler analyses, and microbiome analyses from each diet and treatment group. Results: We show here that while methionine restriction (MR) has little impact on our RP-B6 Myc prostate cancer cell line, it does yield a significant alteration in both the polarization and function of M1 and M2 macrophages. In the in vitro MR conditions, we observed significantly enhanced polarization of M1 macrophages and reduced polarization of M2 macrophages. Functional analysis revealed increased tumoricidal activity of both M1, ‘antitumor’, and M2, ‘pro-tumor,’ macrophages suggesting a flip in M2 function from tumor-promoting to tumoricidal. Further analysis of the released cytokines in MR media conditions yielded significant increase of antitumor cytokines & chemokines, such as IL-12, IL-27, CXCL9, CXCL10, CXCL11, CCL2, CCL4, and TNF-alpha, a double-edged sword which in our system correlates with decreased cancer cell viability upon co-culture with amino acid restricted M1 and M2 macrophages. Our preliminary results show dietary MR yielding a significant inhibition of prostate cancer growth in the RP-B6-Myc model and our ongoing study with survivin peptide vaccine and anti-PD-1 immunotherapies will be available to present in the conference. Conclusions: Our data suggest that restricting methionine is sufficient to alter both the polarization and tumoricidal function of macrophages. Our preliminarily results show that restricted dietary methionine is able to inhibit prostate cancer growth, modify the host immune response and better inhibit prostate cancer growth alone or in combination with immunotherapy. These results provide a strong basis to consider diet restriction as a means to limit cancer growth targeting tumor immune system and potentially to enhance cancer patient responses to immunotherapy. Citation Format: Ashley R. Orillion, Sreenivasulu Chintala, Remi Adelaiye-Ogala, Li Shen, Nur Damayanti, May Elbanna, Sreevani Arisa, Bennett Elzey, Chinghai Kao, Luigi Fontana, Roberto Pili. Methionine restriction increases macrophage tumoricidal activity and significantly inhibits prostate cancer growth [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 250. doi:10.1158/1538-7445.AM2017-250
- Published
- 2017
- Full Text
- View/download PDF
40. Abstract 4170: Targeting androgen receptor overcomes resistance to tyrosine kinase inhibitors in advanced clear cell renal cell carcinoma
- Author
-
Sreenivasulu Chintala, Remi Adelaiye-Ogala, Nur P. Damayanti, Roberto Pili, Ashley Orillion, Kiersten Marie Miles, May Elbanna, Ben Elzey, Piergiorgio Pettazzoni, Giulio Draetta, and Chinghai Kao
- Subjects
Androgen receptor ,Cancer Research ,medicine.medical_specialty ,Clear cell renal cell carcinoma ,Endocrinology ,Oncology ,business.industry ,Internal medicine ,Cancer research ,Medicine ,business ,medicine.disease ,Tyrosine kinase - Abstract
Background: Advanced renal cell carcinoma (RCC) responds initially to anti-angiogenic therapies but eventually develop resistance which may be driven by the expression of key intratumoral pro-survival protein and overall kinome reprogramming. Androgen receptor (AR) expression has been reported in clear cell renal cell carcinoma (ccRCC) but its biological role remains elusive and its association with resistance to tyrosine kinase inhibitors (TKIs) has not been studied. Here we report the role of AR and its association with resistance to TKIs in advanced RCC. Methods: Human RCC cell lines; 786-0, 786-0R (with induced sunitinib resistance), ACHN, URMC2 (with intrinsic sunitinib resistance) were used to assess the combination effect of sunitinib and the AR antagonist enzalutamide in vitro. In vivo studies utilizing 786-0 tumors assessed the combination effect of enzalutamide and sunitinib following acquired resistance to sunitinib. AR expression was detected by qRT-PCR and Western blot analysis, and AR localization by immunofluorescence in treated and non-treated cells. Gene array was used to assess 93 AR associated genes in sensitive and resistant cells. Results: Quantitative RT-PCR data revealed a significant increase in AR expression with sunitinib resistance (>100 folds; p>0.001). In vitro and in vivo studies indicated significant increase in nuclear and cytoplasmic expression of AR with sunitinib treatment in resistant cells which was abrogated with single agent enzalutamide and combination treatment. More interestingly, combination treatment of enzalutamide and sunitinib significantly decreased cell growth and induced tumor regression in vitro and in vivo, respectively. Conclusion: Our data suggest the role of AR both in intrinsic and acquired sunitinib resistance in ccRCC and provide a rationale for combination strategies to restore TKI sensitivity that can be tested in the clinical setting. Citation Format: Remi M. Adelaiye-Ogala, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Ben Elzey, Nur Damayanti, Kiersten M. Miles, Chinghai Kao, Piergiorgio Pettazzoni, Giulio F. Draetta, Roberto Pili. Targeting androgen receptor overcomes resistance to tyrosine kinase inhibitors in advanced clear cell renal cell carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4170. doi:10.1158/1538-7445.AM2017-4170
- Published
- 2017
- Full Text
- View/download PDF
41. Serum-Free Recombinant Production of Adenovirus Using a Hollow Fiber Capillary System
- Author
-
Ling Yang, Thomas A. Gardner, Leland W. K. Chung, J. J S Cadwell, Chinghai Kao, and Song-Chu Ko
- Subjects
Recombination, Genetic ,Chemistry ,Capillary action ,Genetic enhancement ,Genetic Vectors ,Cell Culture Techniques ,Recombinant virus ,medicine.disease_cause ,Virology ,Culture Media, Serum-Free ,General Biochemistry, Genetics and Molecular Biology ,Adenoviridae ,law.invention ,Fiber cell ,law ,Cell culture ,Recombinant DNA ,medicine ,Fiber ,Biotechnology ,Biomedical engineering - Abstract
A novel method for the production of adenoviral vectors on a scale sufficient to support most research applications and early phase clinical trials is presented. This method utilizes serum-free cell culture medium and a hollow fiber cell culture apparatus. Significantly less time and space are required than in conventional methods, and the resulting adenovirus is collected in a much smaller volume, simplifying the purification steps. The protocol described is a reproducible, convenient, biologically safe, and environmentally sound method for the production of adenoviral vectors for laboratory use and has the potential to scale-up the adenovirus production for clinical use.
- Published
- 2001
- Full Text
- View/download PDF
42. Regions of prostate-specific antigen (PSA) promoter confer androgen-independent expression of PSA in prostate cancer cells
- Author
-
Chinghai Kao, Jan Trapman, Xiaoyan Li, Leland W. K. Chung, Fan Yeung, Justin Ellett, and Pathology
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Molecular Sequence Data ,Response Elements ,urologic and male genital diseases ,Biochemistry ,Prostate cancer ,Antigen ,SDG 3 - Good Health and Well-being ,Prostate ,Internal medicine ,LNCaP ,Tumor Cells, Cultured ,medicine ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Base Sequence ,Chemistry ,Prostatic Neoplasms ,Cell Biology ,Prostate-Specific Antigen ,medicine.disease ,Androgen ,Androgen receptor ,Prostate-specific antigen ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,Receptors, Androgen ,Androgens ,Cancer research - Abstract
Prostate-specific antigen (PSA) is expressed primarily by both normal prostate epithelium and the vast majority of prostate cancers. Increases in serum PSA during endocrine therapy are generally considered as evidence for prostate cancer recurrence or progression to androgen independence. The mechanisms by which PSA up-regulation occurs in androgen-refractory prostate cancer cells are unknown. In this study, by using LNCaP and its lineage-derived androgen-independent PSA-producing subline, C4-2, we identified two cis-elements within the 5.8-kilobase pair PSA promoter that are essential for the androgen-independent activity of PSA promoter in prostate cancer cells. First, a previously reported 440-bp androgen-responsive element enhancer core (AREc) was found to be important for the high basal PSA promoter activity in C4-2 cells. Both mutation analysis and supershift experiments demonstrated that androgen receptor (AR) binds to the AREs within the AREc and activate the basal PSA promoter activity in C4-2 cells under androgen-deprived conditions. Second, a 150-bp pN/H region was demonstrated to be a strong AR-independent positive-regulatory element of the PSA promoter in both LNCaP and C4-2 cells. Through DNase I footprinting and linker scan mutagenesis, a 17-bp RI site was identified as the key cis-element within the pN/H region. Data from electrophoretic mobility shift analysis and UV cross-linking experiments further indicated that a 45-kDa (p45) cell-specific transcription factor associates with RI in prostate cancer cells and may be responsible for driving the PSA promoter activity independent of androgen and AR. Furthermore, by juxtaposing AREc and pN/H, we produced a chimeric PSA promoter (supra-PSA) that exhibits 2-3-fold higher activity than the wild type PSA promoter in both LNCaP and C4-2 cells.
- Published
- 2000
43. Establishing human prostate cancer cell xenografts in bone: Induction of osteoblastic reaction by prostate-specific antigen-producing tumors in athymic and SCID/bg mice using LNCaP and lineage-derived metastatic sublines
- Author
-
Haiyen E. Zhau, Quanjun Cui, Tony T. Wu, Cheryl F. Murphy, Gary Balian, Hua Yang, Chinghai Kao, Robert A. Sikes, George N. Thalmann, and Leland W. K. Chung
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Cancer ,medicine.disease ,Metastasis ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Prostate ,LNCaP ,Cancer cell ,medicine ,Bone marrow ,business ,Lymph node - Abstract
LNCaP lineage-derived human prostate cancer cell lines C4-2 and C4-2B4 acquire androgen independence and osseous metastatic potential in vivo. Using C4-2 and C4-2B4 the goals of the current investigation were 1) to establish an ideal bone xenograft model for prostate cancer cells in intact athymic or SCID/bg mice using an intraosseous route of tumor cell administration and 2) to compare prostate cancer metastasis by administering cells either through intravenous (i.v.) or intracardiac administration in athymic or SCID/bg mice. Subsequent to tumor cell administration, prostate cancer growth in the skeleton was assessed by radiographic bone density, serum prostate-specific antigen (PSA) levels, presence of hematogenous prostate cancer cells and histopathologic evaluation of tumor specimens in the lymph node and skeleton. Our results show that whereas LNCaP cells injected intracardially failed to develop metastasis, C4-2 cells injected similarly had the highest metastatic capability in SCID/bg mice. Retroperitoneal and mediastinal lymph node metastases were noted in 3/7 animals, whereas 2/7 animals developed osteoblastic spine metastases. Intracardiac injection of C4-2 in athymic hosts produced spinal metastases in 1/5 animals at 8-12 weeks post-injection; PC-3 injected intracardially also metastasized to the bone but yielded osteolytic responses. Intravenous injection of either LNCaP or C4-2 failed to establish tumor colonies. Intrailiac injection of C4-2 but not LNCaP nor C4-2B4 cells in athymic mice established rapidly growing tumors in 4/8 animals at 2-7 weeks after inoculation. Intrafemoral injection of C4-2 (9/16) and C4-2B4 (5/18) but not LNCaP (0/13) cells resulted in the development of osteoblastic bone lesions in athymic mice (mean: 6 weeks, range: 3-12 weeks). In SCID/bg mice, intrafemoral injection of LNCaP (6/8), C4-2 (8/8) and C4-2B4 (8/8) cells formed PSA-producing, osteoblastic tumors in the bone marrow space within 3-5 weeks after tumor cell inoculation. A stepwise increase of serum PSA was detected in all animals. Reverse transcription-polymerase chain reaction (RT-PCR) to detect hematogenously disseminated prostate cancer cells could not be correlated to either serum PSA level or histological evidence of tumor cells in the marrow space. We have thus established a PSA-producing and osteoblastic human prostate cancer xenograft model in mice.
- Published
- 1998
- Full Text
- View/download PDF
44. Reconstitution of Estrogen-Dependent Transcriptional Activation of an Adenoviral Target Gene in Select Regions of the Rat Mammary Gland1
- Author
-
Chinghai Kao, Lakshmi Sivaraman, Susanne Krnacik, Daniel Medina, Leland W. K. Chung, Meei-Huey Jeng, Orla M. Conneely, and Bert W. O'Malley
- Subjects
Regulation of gene expression ,Reporter gene ,medicine.medical_specialty ,medicine.drug_class ,Gene targeting ,Estrogen receptor ,Biology ,Gene delivery ,Endocrinology ,Estrogen ,Internal medicine ,Gene expression ,medicine ,Transcriptional regulation - Abstract
Estrogen regulates proliferation and morphogenesis of mammary ductal epithelium by interacting with a specific intracellular estrogen receptor (ER) that acts as a hormone-dependent transcriptional regulator of gene expression. The mechanisms by which ER regulates transcription in response to estrogen have been analyzed extensively in tissue culture and in cell-free systems. These studies have demonstrated that the transcriptional activity of ER is strongly influenced by cellular context and highlight the need to address ER transcriptional activity in an appropriate cellular background. Thus, to gain insight into the mechanistic role of ER in mammary epithelial morphogenesis, we have used an adenoviral gene delivery strategy to introduce an estrogen-responsive reporter gene into the mammary epithelium and to monitor the activity of endogenous ERs in their natural environment where cellular context including stromal-epithelial interactions can be taken into account. Using this approach, we first demonstrated highly efficient adenoviral delivery throughout the mammary epithelium using a beta-galactosidase (betagal) reporter gene under the control of the constitutively active cytomegalovirus (CMV) promoter. Next, we constructed an adenoviral vector by substituting the CMV promoter with an estrogen-dependent promoter fragment-linked betagal (Ad-ERE-tk-betagal). This adenoviral reporter system provides evidence that ER positive mammary epithelial cells display a differential sensitivity in a region-specific manner toward estrogen induction. Our data suggest that the availability of factor(s) other than ER is necessary for ER-mediated gene activation and may be important in modulating the differential responses of mammary epithelial cells to estrogen.
- Published
- 1998
- Full Text
- View/download PDF
45. Overcoming cellular senescence in human cancer pathogenesis
- Author
-
Frederic M. Waldman, Stephen Y. Nakada, Sandy DeVries, Michael A. Newton, Thomas R. Yeager, David F. Jarrard, Walter M. Stadler, Timothy D. Moon, Kennedy W. Gilchrist, Chinghai Kao, Reginald C. Bruskewitz, Catherine A. Reznikoff, and Lorraine F. Meisner
- Subjects
Adult ,Male ,Senescence ,Tumor suppressor gene ,Blotting, Western ,Chromosome Disorders ,Biology ,medicine.disease_cause ,Epithelium ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,Neoplasm Invasiveness ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Aged ,Aged, 80 and over ,Chromosome Aberrations ,Carcinoma, Transitional Cell ,Mutation ,Retinoblastoma ,Middle Aged ,medicine.disease ,Blotting, Southern ,Transitional cell carcinoma ,Urinary Bladder Neoplasms ,Tumor progression ,Immunology ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Cell aging ,Research Paper ,Developmental Biology - Abstract
Elevation of p16, the CDKN2/p16 tumor suppressor gene (TSG) product, occurs at senescence in normal human uroepithelial cells (HUC). Immortal HUCs and bladder cancer cell lines show either alteration of p16 or pRb, the product of the retinoblastoma (RB) TSG. In addition, many human cancers show p16 or pRb alteration along with other genetic alterations that we associated with immortalization, including +20q and −3p. These observations led us to hypothesize that p16 elevation plays a critical role in senescence cell cycle arrest and that overcoming this block is an important step in tumorigenesis in vivo, as well as immortalization in vitro. Using a novel approach, we tested these hypotheses in the present study by examining p16 and pRb status in primary culture (P0) and after passage in vitro of transitional cell carcinoma (TCC) biopsies that represented both superficial bladder tumors and invasive bladder cancers. We demonstrated that all superficial TCCs showed elevated p16 after limited passage in vitro and then senesced, like normal HUCs. In contrast, all muscle invasive TCCs contained either a p16 or a pRb alteration at P0 and all spontaneously bypassed senescence (P = 0.001). Comparative genomic hybridization (CGH) was used to identify regions of chromosome loss or gain in all TCC samples. The application of a statistical model to the CGH data showed a high probability of elevated alteration rates of +20q11−q12 (0.99) and +8p22−pter (0.94) in the immortal muscle invasive TCCs, and of −9q (0.99) in the superficial TCCs. Three myoinvasive TCCs lost 3p13−p14. In this study, four of six myoinvasive TCCs also showedTP53 mutation that associated well with genome instability (P = 0.001), as previously hypothesized. Notably,TP53 mutation, which has been used as a marker of tumor progression in many human cancers, was less significant in associating with progression in this study (P = 0.04) than was p16 or pRb alteration (P = 0.001). Thus, these data support a new model in which overcoming senescence plays a critical role in human cancer pathogenesis and requires at least two genetic changes that occur in several combinations that can include either p16 or pRb loss and at least one additional alteration, such as +20q11−q12, −3p13−p14, or −8p21−pter.
- Published
- 1998
- Full Text
- View/download PDF
46. Abstract 897: Methionine restriction alters functional polarization of macrophages in a murine model of prostate cancer
- Author
-
Roberto Pili, Li Shen, May Elbanna, Eric Ciamporcero, Sreevani Arisa, Remi Adelaiye-Ogala, Luigi Fontana, Chinghai Kao, Ashley Orillion, Ben Elzey, and Sreenivasulu Chintala
- Subjects
Cancer Research ,Tumor microenvironment ,Methionine ,Innate immune system ,medicine.medical_treatment ,Growth factor ,Macrophage polarization ,Biology ,Molecular biology ,chemistry.chemical_compound ,Immune system ,Oncology ,chemistry ,Low-protein diet ,medicine ,Cancer research ,PI3K/AKT/mTOR pathway - Abstract
Background: Epidemiological studies link prostate cancer (CaP) to dietary intake. Recent research has shown that individuals consuming a Western diet, high in protein, have higher circulating IGF-1 (insulin-like growth factor 1) which feeds into the nutrient sensing mTOR pathway. This evolutionarily conserved pathway is central to the development of advanced stage and castration resistant CaP. Our previous published studies have shown that dietary protein restriction inhibits tumor growth via mTOR pathway alteration and reduces circulating IGF-1 levels in vivo. The current study hypothesizes that dietary methionine restriction will alter the functionality of immune cells enhancing the ability of the immune system to respond to cancerous insults. Methods: Mice were fed low (7% protein) or control protein (21% protein) diets. In a prevention study mice consumed modified diets for four weeks prior to being inoculated with CaP tumors. In a simultaneous intervention study mice were inoculated with CaP tumors first then four weeks later were placed on the modified diets. Results: In the low protein diet group, we observed that pro-tumor M2 polarized macrophages were significantly reduced in the tumor microenvironment (TME) for both prevention (p = 0.012) and intervention diet (p = 0.0003) studies as compared to the control diet, in addition to observation of mTOR inhibition and IGF-1 reduction. Besides protein content in diet, we also analyzed the amino acid composition. Multiple groups have shown that methionine restriction (MR) inhibits mTOR activation, alters the immune system, and prolongs the survival of rodents. The mechanism by which MR impacts the innate immune system is still poorly understood. In our preliminary in vitro studies, we observed that modification of a single amino acid (AA), such as methionine, is sufficient to inhibit activation of both the mTOR pathway and M2 polarization (Arginase1 qRT-PCR of two samples in quadruplicate (p = 0.0732)), while increasing the presence of M1 macrophage marker iNOS (p = 0.2689) in bone marrow derived macrophages (BMDMs). Thus, we hypothesize that modulating specific amino acids in the diet is sufficient to alter macrophage expression of M1/M2 characteristics, and their functions in the TME. Conclusions: Preliminarily data shows that altering one specific AA intricately linked to the mTOR pathway is sufficient to change macrophage polarization status both at the protein and gene expression levels, suggesting that dietary alteration of a single AA is capable of altering macrophage function. The results of the study provide the basis for translational use of dietary means to alter the immune system and improve the therapeutic effects of immunotherapies. Citation Format: Ashley Orillion, Remi Adelaiye-Ogala, Li Shen, Eric Ciamporcero, May Elbanna, Sreenivasulu Chintala, Sreevani Arisa, Ben Elzey, Chinghai Kao, Luigi Fontana, Roberto Pili. Methionine restriction alters functional polarization of macrophages in a murine model of prostate cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 897.
- Published
- 2016
- Full Text
- View/download PDF
47. An Ultrasonically Powered Implantable Micro-Oxygen Generator (IMOG)
- Author
-
Babak Ziaie, Ning Cao, Seung Hyun Song, Song-Chu Ko, Teimour Maleki, and Chinghai Kao
- Subjects
Materials science ,Oxygen concentrator ,Biomedical Engineering ,Mice, Nude ,chemistry.chemical_element ,Models, Biological ,Oxygen ,Electrolysis ,Mice ,Tumor Microenvironment ,Animals ,Humans ,Ultrasonics ,THERAPY ,STIMULATION ,CORROSION ,TITANIUM ,CARBOGEN ,ALLOYS ,TUMORS ,Hypoxia ,Hypodermic needle ,Luminescent Agents ,business.industry ,Ultrasound ,Reproducibility of Results ,Water ,Equipment Design ,Oxygenation ,Tumor Oxygenation ,Electronics, Medical ,Nanoscience and Nanotechnology ,Pancreatic Neoplasms ,chemistry ,Electrode ,Microtechnology ,Ultrasonic sensor ,business ,Biomedical engineering - Abstract
In this paper, we present an ultrasonically powered implantable micro-oxygen generator (IMOG) that is capable of in situ tumor oxygenation through water electrolysis. Such active mode of oxygen generation is not affected by increased interstitial pressure or abnormal blood vessels that typically limit the systemic delivery of oxygen to hypoxic regions of solid tumors. Wireless ultrasonic powering (2.15 MHz) was employed to increase the penetration depth and eliminate the directional sensitivity associated with magnetic methods. In addition, ultrasonic powering allowed for further reduction in the total size of the implant by eliminating the need for a large area inductor. IMOG has an overall dimension of 1.2 mm x 1.3 mm x 8 mm, small enough to be implanted using a hypodermic needle or a trocar. In vitro and ex vivo experiments showed that IMOG is capable of generating more than 150 mu A which, in turn, can create 0.525 mu L/min of oxygen through electrolytic disassociation. In vivo experiments in a well-known hypoxic pancreatic tumor models (1 cm(3) in size) also verified adequate in situ tumor oxygenation in less than 10 min.
- Published
- 2011
48. Androgen-independent Molecular Imaging Vectors to Detect Castration-Resistant and Metastatic Prostate Cancer
- Author
-
Liu H. Wei, Chinghai Kao, Ziyue Karen Jiang, Lily Wu, and Makoto Sato
- Subjects
PCA3 ,Male ,Transcriptional Activation ,Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.drug_class ,Genetic Vectors ,Biology ,Regulatory Sequences, Nucleic Acid ,urologic and male genital diseases ,Article ,Androgen deprivation therapy ,Prostate cancer ,Mice ,Prostate ,Bone Marrow ,Genes, Reporter ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Tibia ,Cancer ,Prostatic Neoplasms ,Prostate-Specific Antigen ,Androgen ,medicine.disease ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,Prostate-specific antigen ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Oncology ,Positron-Emission Tomography ,Cancer research ,Androgens ,Gene Fusion ,Orchiectomy - Abstract
Prostate-specific promoters are frequently employed in gene-mediated molecular imaging and therapeutic vectors to diagnose and treat castration-resistant prostate cancer (CRPC) that emerges from hormone ablation therapy. Many of the conventional prostate-specific promoters rely on the androgen axis to drive gene expression. However, considering the cancer heterogeneity and varying androgen receptor status, we herein evaluated the utility of prostate-specific enhancing sequence (PSES), an androgen-independent promoter in CRPC. The PSES is a fused enhancer derived from the prostate-specific antigen (PSA) and prostate-specific membrane antigen gene regulatory region. We augmented the activity of PSES by the two-step transcriptional amplification (TSTA) system to drive the expression of imaging reporter genes for either bioluminescent or positron emission tomography (PET) imaging. The engineered PSES–TSTA system exhibits greatly elevated transcriptional activity, androgen independency, and strong prostate specificity, verified in cell culture and preclinical animal experimentations. These advantageous features of PSES–TSTA elicit superior gene expression capability for CRPC in comparison with the androgen-dependent PSA promoter–driven system. In preclinical settings, we showed robust PET imaging capacity of PSES–TSTA in a castrated prostate xenograft model. Moreover, intravenous administrated PSES–TSTA bioluminescent vector correctly identified tibial bone marrow metastases in 9 of 9 animals, whereas NaF- and FDG-PET was unable to detect the lesions. Taken together, this study showed the promising utility of a potent, androgen-independent, and prostate cancer–specific expression system in directing gene-based molecular imaging in CRPC, even in the context of androgen deprivation therapy. Cancer Res; 71(19); 6250–60. ©2011 AACR.
- Published
- 2011
49. Comparative studies of prostate cancers among United States, Chinese, and Japanese patients: characterization of histopathology, tumor angiogenesis, neuroendocrine factors, and p53 protein accumulations
- Author
-
Lian Sheng Zhao, Rodney Davis, Judd W. Moul, Bao Qi Chen, Haiyen E. Zhau, Ninjia Zheng, M. Craig Hall, Patricia Troncoso, Leland W.K. Chung, Chinghai Kao, Ming Fu Ye, W. Fraser Symmans, Lian Sheng Xiao, Munekado Kojima, and J. L. Palmer
- Subjects
PCA3 ,medicine.medical_specialty ,Pathology ,biology ,business.industry ,Angiogenesis ,Urology ,Chromogranin A ,medicine.disease ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Prostate ,biology.protein ,Medicine ,Immunohistochemistry ,Histopathology ,Stage (cooking) ,business - Abstract
Although interracial differences of prostate cancer progression are well recognized, their underlying molecular and cellular mechanisms remain obscure. We compared the histopathologic, immunohistochemical, and molecular characteristics of unselected prostate cancer tissues obtained from U.S., Chinese, and Japanese men. Histopathologic analyses indicated that 74.4% of the prostate cancers in Chinese men were poorly differentiated, compared with 28.6% and 32.8% of the prostate cancers in U.S. and Japanese men, respectively. These differences cannot be attributed to patient age, clinical stage of disease, or methods of tissue sampling. The high proportion of poorly differentiated prostate cancer tissues in the Chinese group was not related to the patients' access to medical service or to geographic background within China. Significantly higher levels of tumor angiogenesis (2- to 4-fold), serotonin (2- to 20-fold), and bombesin (7- to 16-fold), but not chromogranin A, were found in the tissue specimens obtained from Chinese prostate cancer patients compared with those from U.S. and Japanese patients. We also observed marked interracial differences in p53 protein accumulation. The protein was present in 90.2% of Chinese specimens; 17.4% of specimens from U.S. whites; 7.1% of specimens from Japanese men; and 3.7% of specimens from U.S. blacks. Results from multivariate logistic regression analysis demonstrated that p53 protein accumulation, angiogenesis, and serotonin expression in the normal stroma area correlate independently with Chinese versus non-Chinese patient populations.
- Published
- 2011
50. Antitumor activity of Ad-IU2, a prostate-specific replication-competent adenovirus encoding the apoptosis inducer, TRAIL
- Author
-
Chinghai Kao, Thomas A. Gardner, Juan A. Jiménez, Xiong Li, Pankita H. Pandya, Yan-Ping Zhang, Kyung Hee Bae, and Yousef Mohammadi
- Subjects
Glutamate Carboxypeptidase II ,Male ,Cancer Research ,Blotting, Western ,Genetic Vectors ,Mice, Nude ,Apoptosis ,TRAIL ,Biology ,medicine.disease_cause ,urologic and male genital diseases ,PSES ,Article ,Adenoviridae ,Cell Line ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,Antigen ,LNCaP ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Adenovirus ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Prostate Cancer ,Prostatic Neoplasms ,Gene Therapy ,Prostate-Specific Antigen ,medicine.disease ,Molecular biology ,3. Good health ,Oncolytic virus ,030220 oncology & carcinogenesis ,Antigens, Surface ,Cancer research ,Molecular Medicine ,Tumor necrosis factor alpha - Abstract
In this study, we investigated the preclinical utility and antitumor efficacy of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) delivered by Ad-IU2, a prostate-specific replication-competent adenovirus (PSRCA), against androgen-independent prostate cancer. Through transcriptional control of adenoviral early genes E1a, E1b and E4, as well as TRAIL by two bidirectional prostate-specific enhancing sequences (PSES), expression of TRAIL as well adenoviral replication was limited to prostate-specific antigen (PSA) and prostate-specific membrane antigen (PSMA)-positive cells. Ad-IU2 induced 5-fold greater apoptosis selectively in PSA/PSMA-positive CWR22rv and C4-2 cells than an oncolytic adenoviral control. Furthermore, prolonged infection with Ad-IU2 reversed TRAIL resistance in LNCaP cells. Ad-IU2 exhibited superior killing efficiency in PSA/PSMA-positive prostate cancer cells at doses 5- to 8-fold lower than required by a PSRCA to produce a similar effect. This cytotoxic effect was not observed in non-prostatic cells, however. As an enhancement of its therapeutic efficacy, Ad-IU2 exerted a TRAIL-mediated bystander effect through direct cell-to-cell contact and soluble factors such as apoptotic bodies. In vivo, Ad-IU2 markedly suppressed the growth of subcutaneous androgen-independent CWR22rv xenografts compared to a PSRCA at six weeks post-treatment (3.1- vs. 17.1-fold growth of tumor). This study demonstrates the potential clinical utility of a PSRCA armed with an apoptosis-inducing ligand.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.