1
|
Jong KXJ, Mohamed EHM, Syafruddin SE, Faruqu FN, Vellasamy KM, Ibrahim K, Ibrahim ZA. IL-8 and PI3K pathway influence the susceptibility of TRAIL-sensitive colorectal cancer cells to TRAIL-induced cell death. Mol Biol Rep 2024; 51:978. [PMID: 39269555 DOI: 10.1007/s11033-024-09895-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is an apoptosis inducer that exhibits an ideal therapeutic safety profile with less adverse effects than conventional chemotherapy. However, the occurrence of TRAIL resistance has been reported in various cancers including colorectal cancer (CRC). Substantial efforts have been channelled towards managing TRAIL resistance including identifying molecular targets. Interleukins (ILs) have been recently shown to play critical roles in modulating TRAIL sensitivity in cancer cells. METHODS AND RESULTS This study investigated the roles of two ILs, IL-8 and IL⍺, in TRAIL resistance in CRC. TRAIL-resistant HT-29 and TRAIL-sensitive HCT 116 cells, were treated with human recombinant IL-8 and IL-1⍺. The results indicated that treatment with IL-8 (2.5 ng/mL) significantly protected TRAIL-sensitive HCT 116 cells from TRAIL-induced cell death (p < 0.05). However, IL-1⍺ did not play a role in modulating CRC cells' responses to TRAIL. Data from RT-qPCR and Western blotting revealed the molecular regulations of IL-8 on TRAIL decoy receptor genes (OPG) and autophagy-related genes (BECN1 and LC3B) expression. The activation of the phosphoinositide 3-kinase (PI3K) pathway was shown to counteract TRAIL-induced cell death. By inhibiting its activation with wortmannin, the protective role of IL-8 against TRAIL treatment was reversed, suggesting the involvement of the PI3K pathway. CONCLUSION Collectively, findings from this study identified the role of IL-8 and PI3K in modulating CRC cells' sensitivity to TRAIL. Further validation of these two potential molecular targets is warranted to overcome TRAIL resistance in CRC.
Collapse
Affiliation(s)
- Kelly Xue Jing Jong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | | | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Centre, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Farid Nazer Faruqu
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kumutha Malar Vellasamy
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kamariah Ibrahim
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Zaridatul Aini Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
2
|
Habibizadeh M, Lotfollahzadeh S, Mahdavi P, Mohammadi S, Tavallaei O. Nanoparticle-mediated gene delivery of TRAIL to resistant cancer cells: A review. Heliyon 2024; 10:e36057. [PMID: 39247341 PMCID: PMC11379606 DOI: 10.1016/j.heliyon.2024.e36057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as APO2L, has emerged as a highly potential anticancer agent because of its capacity to effectively trigger apoptosis in tumor cells by specifically binding to either of its death receptors (DR4 or DR5) while having no adverse effects on normal cells. Nevertheless, its practical use has been hindered by its inefficient pharmacokinetics characteristics, the challenges involved in its administration and delivery to targeted cells, and the resistance exhibited by most cancer cells towards TRAIL. Gene therapy, as a promising approach would be able to potentially circumvent TRAIL-based cancer therapy challenges mainly through localized TRAIL expression and generating a bystander impact. Among different strategies, using nanoparticles in TRAIL gene delivery allows for precise targeting, and overcoming TRAIL resistance by combination therapy. In this review, we go over potential mechanisms by which cancer cells achieve resistance to TRAIL and provide an overview of different carriers for delivering of the TRAIL gene to resistant cancer cells, focusing on different types of nanoparticles utilized in this context. We will also explore the challenges, and investigate future perspectives of this nanomedicine approach for cancer therapy.
Collapse
Affiliation(s)
- Mina Habibizadeh
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Lotfollahzadeh
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mahdavi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soheila Mohammadi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Omid Tavallaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
3
|
Jong KXJ, Mohamed EHM, Ibrahim ZA. Escaping cell death via TRAIL decoy receptors: a systematic review of their roles and expressions in colorectal cancer. Apoptosis 2022; 27:787-799. [PMID: 36207556 DOI: 10.1007/s10495-022-01774-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2022] [Indexed: 11/02/2022]
Abstract
The development of targeted therapy such as tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-based therapy has gained increasing attention as a promising new approach in cancer therapy. TRAIL specifically targets cancer cells while sparing the normal cells, thus, limiting the known side effects of the majority anti-cancer therapies. As more extensive research and clinical trials are conducted, resistance to TRAIL molecule has become one of the significant issues associated with the failure of TRAIL in treating colorectal cancer (CRC). To date, the exact mechanism by which TRAIL resistance may have occurred remains unknown. Interestingly, recent studies have revealed the critical role of the TRAIL decoy receptor family; consisting of decoy receptor 1 (DcR1; also known as TRAIL-R3), decoy receptor 2 (DcR2; also known as TRAIL-R4), and osteoprotegerin (OPG) in driving TRAIL resistance. This review highlights the expression of the decoy receptors in CRC and its possible association with the reduction in sensitivity towards TRAIL treatment based on the currently available in vitro, in vivo, and human studies. Additionally, discrepancies between the outcomes from different research groups are discussed, and essential areas are highlighted for future investigation of the roles of decoy receptors in modulating TRAIL-induced apoptosis. Overcoming TRAIL resistance through modulating the expression(s) and elucidating the role(s) of TRAIL decoy receptors hold great promise for TRAIL-based therapies to be extensively explored in treating human cancers including CRC.
Collapse
Affiliation(s)
- Kelly Xue Jing Jong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Zaridatul Aini Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
4
|
Grayson KA, Hope JM, Wang W, Reinhart-King CA, King MR. Taxanes Sensitize Prostate Cancer Cells to TRAIL-Induced Apoptotic Synergy via Endoplasmic Reticulum Stress. Mol Cancer Ther 2021; 20:833-845. [PMID: 33632873 PMCID: PMC10789445 DOI: 10.1158/1535-7163.mct-20-0495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 01/05/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022]
Abstract
Docetaxel and cabazitaxel are guideline-chemotherapy treatments for metastatic castration-resistant prostate cancer (mCRPC), which comprises the majority of prostate cancer deaths. TNF-related apoptosis inducing ligand (TRAIL) is an anticancer agent that is selectively cytotoxic to cancer cells; however, many human cancers are resistant to TRAIL. In this study, we sensitized androgen-independent and TRAIL-resistant prostate cancer cells to TRAIL-mediated apoptosis via taxane therapy and examined the mechanism of sensitization. DU145 and PC3 cells displayed no significant reduction in cell viability when treated with soluble TRAIL, docetaxel, or cabazitaxel alone indicating that both cell lines are resistant to TRAIL and taxanes individually. Taxane and TRAIL combination synergistically amplified apoptosis strongly suggesting that taxanes sensitize prostate cancer cells to TRAIL. A Jun N-terminal kinases (JNK) inhibitor inhibited apoptosis in treated cells and significantly reduced death receptor expression indicating JNK activation by ER stress sensitizes PCa cells to TRAIL-induced apoptosis by upregulating DR4/DR5 expression. In addition, suppression of C/EBP homologous protein (CHOP) reduced TRAIL sensitization in both cell lines indicating that ER stress-related apoptosis is mediated, in part, by CHOP. Cytochrome c knockdown showed a significant decrease in sensitivity in PC3 cells, but not in Bax-deficient DU145 cells. A computational model was used to simulate apoptosis for cells treated with taxane and TRAIL therapy as demonstrated in in vitro experiments. Pretreatment with taxanes sensitized cells to apoptosis induced by TRAIL-mediated apoptosis, demonstrating that combining TRAIL with ER stress inducers is a promising therapy to reverse TRAIL resistance to treat mCRPC.
Collapse
Affiliation(s)
- Korie A Grayson
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Jacob M Hope
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
5
|
Enhanced inhibition of tumor growth using TRAIL-overexpressing adipose-derived stem cells in combination with the chemotherapeutic agent CPT-11 in castration-resistant prostate cancer. Prostate Int 2020; 9:31-41. [PMID: 33912512 PMCID: PMC8053698 DOI: 10.1016/j.prnil.2020.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/29/2020] [Accepted: 07/05/2020] [Indexed: 12/20/2022] Open
Abstract
Background This study investigated the inhibition of tumor growth in castrate-resistant prostate cancer (CRPC)-bearing mice by tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)-overexpressing adipose-derived stem cells (ADSCs) (hTERT-ADSC.sTRAIL), which was enhanced by combined treatment with CPT-11. Materials and methods An hTERT-ADSC.sTRAIL cell line was established by transfection with a lentiviral vector (CLV-Ubic) encoding the human sTRAIL gene. Quantitative polymerase chain reaction and Western blots were performed to confirm gene overexpression. An invasion study for the selective migration ability toward PC3 cells was performed. In the in vivo study, the tumor volume in mice treated with ADSC. sTRAIL and CPT-11 was measured. Results Carboxylesterase was generated from hTERT-ADSCs. The gene expression of sTRAIL from hTERT-ADSC.sTRAIL was shown. The directional migration of ADSC.sTRAIL cells toward PC3 cells was significantly stimulated by PC3 cells in vitro (P < 0.05). In the in vitro study, the viability of PC3 cells significantly decreased in the presence of ADSC.sTRAIL (62.7 ± 2.0%) and CPT-11 compared with that of CPT-11 alone (83.0 ± 1.0%) at a cell ratio as low as 0.05 (PC3: ADSC.sTRAIL) (P < 0.05). The proportion of apoptotic PC3 cells significantly increased in the presence of ADSC.sTRAIL (37.2 ± 2.1%) and CPT-11 compared with that of CPT-11 alone (16.5 ± 1.0%) (P < 0.05). In the in vivo study, the inhibition of tumor growth in CRPC-bearing mice by TRAIL-overexpressing adipose stem cells was enhanced by combined treatment with the chemotherapeutic agent CPT-11 compared with that in the treatment with cpt-11 alone. Immunohistochemical staining of the removed tumors showed anti-TRAIL–positive cells and apoptotic bodies after hTERT-ADSC.sTRAIL treatment or combined treatment with hTERT-ADSC.sTRAIL and CPT-11. Conclusions Therapeutic stem cells expressing sTRAIL genes combined with CPT-11 can provide a new strategy for treating CRPC in clinical trials using the patients’ own ADSCs.
Collapse
|
6
|
Goklany S, Lu P, Godeshala S, Hall A, Garrett-Mayer E, Voelkel-Johnson C, Rege K. Delivery of TRAIL-expressing plasmid DNA to cancer cells in vitro and in vivo using aminoglycoside-derived polymers. J Mater Chem B 2019; 7:7014-7025. [PMID: 31633707 DOI: 10.1039/c9tb01286a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a death ligand that can preferentially induce apoptosis in cancer cells over normal cells. The transmembrane form of TRAIL has been shown to elicit much stronger activity than its soluble counterpart but delivery is a potential challenge. Here, we investigated the potential of aminoglycoside-derived polymers to enhance delivery of a plasmid (pEF-TRAIL) that expresses the transmembrane form of TRAIL in order to determine the effect on cell death in vitro and tumor growth in vivo. Transgene delivery efficacy and toxicity of aminoglycoside-derived polymers was first evaluated using a GFP-expressing plasmid (pEF-GFP) at different plasmid amounts and plasmid : polymer ratios in UMUC3 bladder cancer and HeLa cervical cancer cells. Delivery of the TRAIL plasmid using aminoglycoside-derived polymers resulted in up to 60% cell death in UMUC3 and HeLa cells; TRAIL protein expression was confirmed using Western blots. TRAIL plasmid delivery resulted in a decrease in cellular procaspase-8 and an increase in TRAIL receptor DR5 levels, suggesting a role for the death receptor and caspase cascade in TRAIL-mediated apoptosis. The TRAIL plasmid did not cause cell death in normal human or mouse fibroblasts. The in vivo delivery of the TRAIL plasmid using a paromomycin-derived polymer resulted in significant reduction in tumor burden and increased survival in tumor-bearing live mice.
Collapse
Affiliation(s)
- Sheba Goklany
- Chemical Engineering, Arizona State University, 501 E. Tyler Mall, ECG 303, Tempe, AZ 85287-6106, USA.
| | | | | | | | | | | | | |
Collapse
|
7
|
Shishodia G, Koul S, Dong Q, Koul HK. Tetrandrine (TET) Induces Death Receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5) and Sensitizes Prostate Cancer Cells to TRAIL-Induced Apoptosis. Mol Cancer Ther 2018; 17:1217-1228. [PMID: 29549167 PMCID: PMC10186773 DOI: 10.1158/1535-7163.mct-17-1157] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/02/2018] [Accepted: 03/12/2018] [Indexed: 11/16/2022]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells, but not in normal cells; as such, it is a promising therapeutic agent. However, therapeutic resistance limits its clinical use in many malignancies, including prostate cancer. Strategies to sensitize cancer cells to TRAIL are urgently needed. We demonstrate here that small-molecule tetrandrine (TET) potentially sensitizes previously resistant (LNCaP and C4-2B cells) and mildly sensitive (PC3 cells) prostate cancer cells to TRAIL-induced apoptosis, and they do so by upregulating mRNA expression and protein levels of death receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5). Using shRNA knockdown, we show critical requirement of DR4 and DR5 in sensitization of prostate cancer cells to TRAIL. We show that double knockdown of DR4 and DR5 abrogated the apoptotic effects of TET and TRAIL. We also demonstrate that TET-induced DR4 and DR5 expression is independent of p53 status. Given that loss of p53 is associated with progression of prostate cancer to CRPC and NEPC, our results show that TET, by acting as a TRAIL-sensitizing agent in prostate cancer, could serve as a potential therapeutic agent in CRPC and NEPC, for which there is no cure to date. Mol Cancer Ther; 17(6); 1217-28. ©2018 AACR.
Collapse
|
8
|
Chen L, Wolff DW, Xie Y, Lin MF, Tu Y. Cyproterone acetate enhances TRAIL-induced androgen-independent prostate cancer cell apoptosis via up-regulation of death receptor 5. BMC Cancer 2017; 17:179. [PMID: 28270124 PMCID: PMC5341373 DOI: 10.1186/s12885-017-3153-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Virtually all prostate cancer deaths occur due to obtaining the castration-resistant phenotype after prostate cancer cells escaped from apoptosis and/or growth suppression initially induced by androgen receptor blockade. TNF-related apoptosis-inducing ligand (TRAIL) was an attractive cancer therapeutic agent due to its minimal toxicity to normal cells and remarkable apoptotic activity in tumor cells. However, most localized cancers including prostate cancer are resistant to TRAIL-induced apoptosis, thereby creating a therapeutic challenge of inducing TRAIL sensitivity in cancer cells. Herein the effects of cyproterone acetate, an antiandrogen steroid, on the TRAIL-induced apoptosis of androgen receptor-negative prostate cancer cells are reported. METHODS Cell apoptosis was assessed by both annexin V/propidium iodide labeling and poly (ADP-ribose) polymerase cleavage assays. Gene and protein expression changes were determined by quantitative real-time PCR and western blot assays. The effect of cyproterone acetate on gene promoter activity was determined by luciferase reporter assay. RESULTS Cyproterone acetate but not AR antagonist bicalutamide dramatically increased the susceptibility of androgen receptor-negative human prostate cancer PC-3 and DU145 cells to TRAIL-induced apoptosis but no effects on immortalized human prostate stromal PS30 cells and human embryonic kidney HEK293 cells. Further investigation of the TRAIL-induced apoptosis pathway revealed that cyproterone acetate exerted its effect by selectively increasing death receptor 5 (DR5) mRNA and protein expression. Cyproterone acetate treatment also increased DR5 gene promoter activity, which could be abolished by mutation of a consensus binding domain of transcription factor CCAAT-enhancer-binding protein homologous protein (CHOP) in the DR5 gene promoter. Cyproterone acetate increases CHOP expression in a concentration and time-dependent manner and endoplasmic reticulum stress reducer 4-phenylbutyrate could block cyproterone acetate-induced CHOP and DR5 up-regulation. More importantly, siRNA silencing of CHOP significantly reduced cyproterone acetate-induced DR5 up-regulation and TRAIL sensitivity in prostate cancer cells. CONCLUSIONS Our study shows a novel effect of cyproterone acetate on apoptosis pathways in prostate cancer cells and raises the possibility that a combination of TRAIL with cyproterone acetate could be a promising strategy for treating castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Linjie Chen
- Department of Pharmacology, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178 USA
| | - Dennis W. Wolff
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC USA
| | - Yan Xie
- Department of Pharmacology, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178 USA
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yaping Tu
- Department of Pharmacology, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178 USA
| |
Collapse
|
9
|
Tzeng SY, Wilson DR, Hansen SK, Quiñones-Hinojosa A, Green JJ. Polymeric nanoparticle-based delivery of TRAIL DNA for cancer-specific killing. Bioeng Transl Med 2016; 1:149-159. [PMID: 28349127 PMCID: PMC5365091 DOI: 10.1002/btm2.10019] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Lack of specificity in cancer therapeutics severely limits the efficacy of many existing treatment modalities. The use of Tumor Necrosis Factor-related Apoptosis-Inducing Ligand (TRAIL) is of interest to the field due to this protein's ability to cause cell death specifically in cancer cells without harming the surrounding healthy tissue. Here, we report that polymeric nanoparticles, based on synthetic poly(beta-amino ester)s (PBAEs) and containing DNA, are able to selectively transfect cancer cells in vitro over healthy cells of the same tissue type. Moreover, PBAE-based nanoparticles containing TRAIL DNA are able to transfect several human cancer cell cultures in vitro and cause cell death. While certain cell types, including human glioblastoma (GBM), showed resistance to TRAIL, we found that the expression of TRAIL-binding surface proteins was predictive of each cell type's resistance to TRAIL therapy. We demonstrate a non-viral nanomedicine approach to cancer gene therapy that can improve cancer specificity via both biomaterial selection and through the use of cancer-targeting genetic cargo.
Collapse
Affiliation(s)
- Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David R Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sarah K Hansen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Material Science and Engineering, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
10
|
Hartung F, Pardo LA. Guiding TRAIL to cancer cells through Kv10.1 potassium channel overcomes resistance to doxorubicin. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:709-719. [PMID: 27350552 PMCID: PMC5045482 DOI: 10.1007/s00249-016-1149-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/24/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022]
Abstract
Resisting cell death is one of the hallmarks of cancer, and represents a common problem resulting in ineffective cancer therapy. To overcome resistance to apoptosis, we designed an antibody-based therapy strategy using Kv10.1 as a target. Kv10.1 is a voltage-gated potassium channel, which has been identified as a tumor marker several years ago. The agent consists of a Kv10.1-specific single-chain antibody fused to the soluble tumor necrosis factor-related apoptosis-inducing ligand (scFv62-TRAIL). We combined scFv62-TRAIL with different chemotherapeutic drugs, all of which failed to induce apoptosis when used alone. In the combination, we could overcome the resistance and selectively induce apoptosis. Among the drugs, doxorubicin showed the most promising effect. Additionally, we observed improved efficacy by pre-treating the cells with doxorubicin before scFv62-TRAIL application. Expression analysis of the TRAIL death receptors suggests a doxorubicin-induced increase in the abundance of receptors as the mechanism for sensitization. Furthermore, we confirmed the anti-tumor effect and efficacy of our combination strategy in vivo in SCID mice bearing subcutaneous tumors. In conclusion, we propose a novel strategy to overcome resistance to chemotherapy in cancer cells. Doxorubicin and scFv62-TRAIL reciprocally sensitize the cells to each other, specifically in Kv10.1-positive tumor cells.
Collapse
Affiliation(s)
- Franziska Hartung
- Oncophysiology Group, Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075, Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group, Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075, Göttingen, Germany.
| |
Collapse
|
11
|
Zhao Y, Chen R, Wang Y, Qing C, Wang W, Yang Y. In Vitro and In Vivo Efficacy Studies of Lavender angustifolia Essential Oil and Its Active Constituents on the Proliferation of Human Prostate Cancer. Integr Cancer Ther 2016; 16:215-226. [PMID: 27151584 PMCID: PMC5739122 DOI: 10.1177/1534735416645408] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Lavandula angustifolia is the most widely cultivated Lavandula species. The extraction of its flower and leaves has been used as herbal medicine. In this study, the in vitro antitumor activities were tested on human prostate cancer PC-3 and DU145 cell lines. Flow cytometry technology was applied to study apoptosis induction and cell cycle arrest. The PC-3 cell line was used to establish subcutaneous xenograft tumors in nude mice. Paraffin sections from xenograft tumor specimens were used in the TUNEL (terminal deocynucleotide transferase dUTP nick end labeling) assay and an immunohistochemistry assay to detect cell proliferation markers Ki67 and PCNA. Lavender essential oil, linalool, and linalyl acetate showed stronger inhibitory effect on PC-3 cells than on DU145 cells. The apoptotic cell populations observed in PC-3 cells treated with lavender essential oil, linalool, and linalyl acetate were 74.76%, 67.11%, and 56.14%, respectively. The PC-3 cells were mainly arrested in the G2/M phase. In the xenograft model with PC-3 cell transplantation, essential oil and linalool significantly suppressed tumor growth. The immunosignals of Ki67 and PCNA in the essential oil, linalool, and linalyl acetate treatment groups were significantly lower than that of the control group in xenograft tumor sections. The TUNEL assay indicated that each of the 3 phytochemicals significantly induced apoptosis compared to the control group. This study provides novel insight and evidence on the antiproliferative effect of L angustifolia essential oil and its major constituents on human prostate cancer. The antitumor effect was associated with cell proliferation inhibition and apoptosis induction in xenograft tumors.
Collapse
Affiliation(s)
- Yunqi Zhao
- 1 Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Ran Chen
- 1 Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yun Wang
- 1 Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Chen Qing
- 1 Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Wei Wang
- 1 Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yixin Yang
- 2 Emporia State University, Emporia, KS, USA
| |
Collapse
|
12
|
Antitumor effect of TRAIL on oral squamous cell carcinoma using magnetic nanoparticle-mediated gene expression. Cell Biochem Biophys 2015; 69:663-72. [PMID: 24563116 DOI: 10.1007/s12013-014-9849-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
We developed a new magnetic nanovector to improve the efficiency and targeting of transgene therapy for oral squamous cell carcinoma (OSCC). Positively charged polymer PEI-modified Fe(3)O(4) magnetic nanoparticles were tested as gene transfer vectors in the presence of a magnetic field. The Fe(3)O(4) nanoparticles were prepared by a co-precipitation method and had good dispersibility in water. These nanoparticles modified by PEI were combined with negatively charged pACTERT-EGFP via electrostatic interaction. The transfection efficiency of the magnetic nano-gene vector with the magnetic field was determined by a fluorescence-inverted microscope and flow cytometry. The results showed significant improvement compared with the control group (p < 0.05). The magnetic complexes also exhibited up to 6-times higher transfection efficiency compared with commonly used PEI or lipofectin. On the basis of these results, the antitumor effect with suicide gene therapy using pACTERT-TRAIL in vitro and vivo was evaluated. In vitro apoptosis was determined with the Annexin V-FITC Apoptosis Detection Kit. The results suggested that PEI-modified Fe(3)O(4) nanoparticles could mediate the killing of Tca83 cells. Furthermore, treatment with pACTERT-TRAIL delivered by magnetic nanoparticles showed a significant cytostatic effect through the induction of apoptosis in a xenograft model. This indicates that magnetic nano-gene vectors could improve the transgene efficiency for Tca83 cells and could exhibit antitumor functions with the plasmid pACTERT-TRAIL. This may be a new way to treat OSCC.
Collapse
|
13
|
Santha S, Viswakarma N, Das S, Rana A, Rana B. Tumor Necrosis Factor-related Apoptosis-inducing Ligand (TRAIL)-Troglitazone-induced Apoptosis in Prostate Cancer Cells Involve AMP-activated Protein Kinase. J Biol Chem 2015. [PMID: 26198640 DOI: 10.1074/jbc.m115.663526] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer (PCa) is one of the most frequently diagnosed cancers in men with limited treatment options for the hormone-resistant forms. Development of novel therapeutic options is critically needed to target advanced forms. Here we demonstrate that combinatorial treatment with the thiazolidinedione troglitazone (TZD) and TNF-related apoptosis-inducing ligand (TRAIL) can induce significant apoptosis in various PCa cells independent of androgen receptor status. Because TZD is known to activate AMP-activated protein kinase (AMPK), we determined whether AMPK is a molecular target mediating this apoptotic cascade by utilizing PCa cell lines stably overexpressing AMPKα1 dominant negative (C4-2-DN) or empty vector (C4-2-EV). Our results indicated a significantly higher degree of apoptosis with TRAIL-TZD combination in C4-2-EV cells compared with C4-2-DN cells. Similarly, results from a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay showed a larger reduction of viability of C4-2-EV cells compared with C4-2-DN cells when treated with TRAIL-TZD, thus suggesting that C4-2-DN cells were more apoptosis-resistant. Additionally, siRNA-mediated knockdown of endogenous AMPKα1 expression showed a reduction of TRAIL-TZD-induced apoptosis, further confirming the participation of AMPK in mediating this apoptosis. Apoptosis induction by this combinatorial treatment was also associated with a cleavage of β-catenin that was inhibited in both C4-2-DN cells and those cells in which AMPKα1 was knocked down. In addition, time course studies showed an increase in pACC(S79) (AMPK target) levels coinciding with the time of apoptosis. These studies indicate the involvement of AMPK in TRAIL-TZD-mediated apoptosis and β-catenin cleavage and suggest the possibility of utilizing AMPK as a therapeutic target in apoptosis-resistant prostate cancer.
Collapse
Affiliation(s)
- Sreevidya Santha
- From the Department of Medicine, Division of Gastroenterology & Nutrition and
| | - Navin Viswakarma
- the Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Subhasis Das
- the Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Ajay Rana
- the Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, Illinois 60153 and the Hines VA Medical Center, Hines, Illinois 60141
| | - Basabi Rana
- From the Department of Medicine, Division of Gastroenterology & Nutrition and the Hines VA Medical Center, Hines, Illinois 60141
| |
Collapse
|
14
|
Rozanov D, Cheltsov A, Sergienko E, Vasile S, Golubkov V, Aleshin AE, Levin T, Traer E, Hann B, Freimuth J, Alexeev N, Alekseyev MA, Budko SP, Bächinger HP, Spellman P. TRAIL-Based High Throughput Screening Reveals a Link between TRAIL-Mediated Apoptosis and Glutathione Reductase, a Key Component of Oxidative Stress Response. PLoS One 2015; 10:e0129566. [PMID: 26075913 PMCID: PMC4468210 DOI: 10.1371/journal.pone.0129566] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/11/2015] [Indexed: 02/07/2023] Open
Abstract
A high throughput screen for compounds that induce TRAIL-mediated apoptosis identified ML100 as an active chemical probe, which potentiated TRAIL activity in prostate carcinoma PPC-1 and melanoma MDA-MB-435 cells. Follow-up in silico modeling and profiling in cell-based assays allowed us to identify NSC130362, pharmacophore analog of ML100 that induced 65-95% cytotoxicity in cancer cells and did not affect the viability of human primary hepatocytes. In agreement with the activation of the apoptotic pathway, both ML100 and NSC130362 synergistically with TRAIL induced caspase-3/7 activity in MDA-MB-435 cells. Subsequent affinity chromatography and inhibition studies convincingly demonstrated that glutathione reductase (GSR), a key component of the oxidative stress response, is a target of NSC130362. In accordance with the role of GSR in the TRAIL pathway, GSR gene silencing potentiated TRAIL activity in MDA-MB-435 cells but not in human hepatocytes. Inhibition of GSR activity resulted in the induction of oxidative stress, as was evidenced by an increase in intracellular reactive oxygen species (ROS) and peroxidation of mitochondrial membrane after NSC130362 treatment in MDA-MB-435 cells but not in human hepatocytes. The antioxidant reduced glutathione (GSH) fully protected MDA-MB-435 cells from cell lysis induced by NSC130362 and TRAIL, thereby further confirming the interplay between GSR and TRAIL. As a consequence of activation of oxidative stress, combined treatment of different oxidative stress inducers and NSC130362 promoted cell death in a variety of cancer cells but not in hepatocytes in cell-based assays and in in vivo, in a mouse tumor xenograft model.
Collapse
Affiliation(s)
- Dmitri Rozanov
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| | | | | | - Stefan Vasile
- The Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, Orlando, Florida, United States of America
| | - Vladislav Golubkov
- Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Alexander E. Aleshin
- Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Trevor Levin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Elie Traer
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Byron Hann
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, United States of America
| | - Julia Freimuth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, United States of America
| | - Nikita Alexeev
- Computational Biology Institute, George Washington University, Ashburn, Virginia, United States of America
- Department of Mathematics and Mechanics, Saint Petersburg State University, Saint Petersburg, Russia
| | - Max A. Alekseyev
- Computational Biology Institute, George Washington University, Ashburn, Virginia, United States of America
| | - Sergey P Budko
- Research Department, Shriners Hospital for Children, Portland, Oregon, United States of America
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Hans Peter Bächinger
- Research Department, Shriners Hospital for Children, Portland, Oregon, United States of America
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Paul Spellman
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States of America
| |
Collapse
|
15
|
Sundaram K, Sambandam Y, Balasubramanian S, Pillai B, Voelkel-Johnson C, Ries WL, Reddy SV. STAT-6 mediates TRAIL induced RANK ligand expression in stromal/preosteoblast cells. Bone 2015; 71:137-44. [PMID: 25445452 DOI: 10.1016/j.bone.2014.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 10/14/2014] [Accepted: 10/23/2014] [Indexed: 02/02/2023]
Abstract
Receptor activator of nuclear factor kappa-B ligand (RANKL) is a critical osteoclastogenic factor expressed in bone marrow stromal/osteoblast lineage cells. Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) levels are elevated in pathologic conditions such as multiple myeloma and inflammatory arthritis, and have been positively correlated with osteolytic markers. Osteoprotegerin (OPG) which inhibits osteoclastogenesis is a decoy receptor for RANKL and also known to interact with TRAIL. Herein, we show that TRAIL increases DR5 and DcR1 receptors but no change in the levels of DR4 and DcR2 expression in human bone marrow derived stromal/preosteoblast (SAKA-T) cell line. We further demonstrated that TRAIL treatment significantly decreased OPG mRNA expression. Interestingly, TRAIL treatment induced RANKL mRNA expression in these cells. In addition, TRAIL significantly increased NF-kB and c-Jun N-terminal kinase (JNK) activity. Human transcription factor array screening by real-time RT-PCR identified TRAIL up-regulation of the signal transducers and activators of the transcription (STAT)-6 expression in SAKA-T cells. TRAIL stimulation induced p-STAT-6 expression in human bone marrow derived primary stromal/preosteoblast cells. Confocal microscopy analysis further revealed p-STAT-6 nuclear localization in SAKA-T cells. Chromatin immunoprecipitation (ChIP) assay confirmed p-STAT-6 binding to the hRANKL gene distal promoter region. In addition, siRNA suppression of STAT-6 expression inhibits TRAIL increased hRANKL gene promoter activity. Thus, our results suggest that TRAIL induces RANKL expression through a STAT-6 dependent transcriptional regulatory mechanism in bone marrow stromal/preosteoblast cells.
Collapse
Affiliation(s)
- Kumaran Sundaram
- Charles P. Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA
| | - Yuvaraj Sambandam
- Charles P. Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA
| | | | - Balakrishnan Pillai
- Charles P. Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA
| | | | - William L Ries
- College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Sakamuri V Reddy
- Charles P. Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
16
|
Wang H, Xu C, Kong X, Li X, Kong X, Wang Y, Ding X, Yang Q. Trail resistance induces epithelial-mesenchymal transition and enhances invasiveness by suppressing PTEN via miR-221 in breast cancer. PLoS One 2014; 9:e99067. [PMID: 24905916 PMCID: PMC4048247 DOI: 10.1371/journal.pone.0099067] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 05/12/2014] [Indexed: 01/05/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis of cancer cells and is verified effective to various cancers. However, a variety of breast cancer cell lines are resistant to TRAIL and the mechanisms of resistance are largely unknown. In our present experiment, we successfully utilized breast cancer cell line MDA-MB-231 to establish TRAIL-resistant cell line. We found resistance to TRAIL could induce epithelial-mesenchymal transition (EMT) and enhance invasiveness. We further demonstrated PTEN was down-regulated in TRAIL-resistant cells. Silencing miR-221, PTEN expression was up-regulated, the process of EMT could be reversed, and the ability of migration and invasion were correspondingly weakened. We also demonstrated knockdown of miR-221 could reverse resistance to TRAIL partially by targeting PTEN. Our findings suggest that resistance to TRAIL could induce EMT and enhance invasiveness by suppressing PTEN via miR-221. Re-expression of miR-221 or targeting PTEN might serve as potential therapeutic approaches for the treatment of Trail-resistant breast cancer.
Collapse
Affiliation(s)
- Haiji Wang
- Department of Oncology, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Oncology, Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong Province, China
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Chunyuan Xu
- Department of Oncology, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province, China
| | - Xiaoli Kong
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xiaoyan Li
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xiangnan Kong
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xia Ding
- Department of Oncology, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- * E-mail:
| |
Collapse
|
17
|
Gasmi J, Thomas Sanderson J. Jacaric acid and its octadecatrienoic acid geoisomers induce apoptosis selectively in cancerous human prostate cells: a mechanistic and 3-D structure-activity study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:734-742. [PMID: 23453308 DOI: 10.1016/j.phymed.2013.01.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 12/20/2012] [Accepted: 01/27/2013] [Indexed: 06/01/2023]
Abstract
Plant-derived non-essential fatty acids are important dietary nutrients, and some are purported to have chemopreventive properties against various cancers, including that of the prostate. In this study, we determined the ability of seven dietary C-18 fatty acids to cause cytotoxicity and induce apoptosis in various types of human prostate cancer cells. These fatty acids included jacaric and punicic acid found in jacaranda and pomegranate seed oil, respectively, three octadecatrienoic geometric isomers (alpha- and beta-calendic and catalpic acid) and two mono-unsaturated C-18 fatty acids (trans- and cis-vaccenic acid). Jacaric acid and four of its octadecatrienoic geoisomers selectively induced apoptosis in hormone-dependent (LNCaP) and -independent (PC-3) human prostate cancer cells, whilst not affecting the viability of normal human prostate epithelial cells (RWPE-1). Jacaric acid induced concentration- and time-depedent LNCaP cell death through activation of intrinsic and extrinsic apoptotic pathways resulting in cleavage of PARP-1, modulation of pro- and antiapoptotic Bcl-2 family of proteins and increased cleavage of caspase-3, -8 and -9. Moreover, activation of a cell death-inducing signalling cascade involving death receptor 5 was observed. Jacaric acid induced apoptosis in PC-3 cells by activation of the intrinsic pathway only. The spatial conformation cis, trans, cis of jacaric and punicic acid was shown to play a key role in the increased potency and efficacy of these two fatty acids in comparison to the five other C-18 fatty acids tested. Three-dimensional conformational analysis using the PubChem Database (http://pubchem.ncbi.nlm.nih.gov) showed that the cytotoxic potency of the C-18 fatty acids was related to their degree of conformational similarity to our cytotoxic reference compound, punicic acid, based on optimized shape (ST) and feature (CT) similarity scores, with jacaric acid being most 'biosimilar' (ST(ST-opt)=0.81; CT(CT-opt)=0.45). This 3-D analysis of structural similarity enabled us to rank geoisomeric fatty acids according to cytotoxic potency, whereas a 2-D positional assessment of cis/trans structure did not. Our findings provide mechanistic evidence that nutrition-derived non-essential fatty acids have chemopreventive biological activities and Exhibit 3-D structure-activity relationships that could be exploited to develop new strategies for the prevention or treatment of prostate cancer regardless of hormone dependency.
Collapse
Affiliation(s)
- Jihane Gasmi
- Institut National de la Recherche Scientifique (INRS), Institut Armand-Frappier, Université du Québec, Laval, Québec, Canada
| | | |
Collapse
|
18
|
Qiu Y, Zhang Z, Shi J, Liu S, Liu Y, Zheng D. A novel anti-DR5 chimeric antibody and epirubicin synergistically suppress tumor growth. IUBMB Life 2012; 64:757-65. [PMID: 22815059 DOI: 10.1002/iub.1064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/23/2012] [Indexed: 12/20/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in a variety of tumor cells. TRAIL receptor 2 (DR5) expression is high in tumor cells, transformed cells, and clinical tumor specimens and is low in most normal cells and tissues; therefore, DR5 is considered an attractive target for cancer therapy. In this study, HMCAZ5, a novel mouse-human chimeric antibody based on AD5-10, was generated and stably expressed in CHO-dhfr(-) cells. Highly purified HMCAZ5 exhibits a high affinity for the receptor that is equal to the parental mouse antibody, induces apoptosis in various cancer cells but not in normal hepatocytes, and elicits both antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in various human cancer cells. The anthracycline anticancer drug epirubicin (EPB) synergizes the cytotoxicity of HMCAZ5 in cancer cells by upregulating DR5 expression on the cell surfaces, enhancing p53 expression, Bid cleavage, and JNK phosphorylation and downregulating c-FLIP expression and Akt phosphorylation. Moreover, HMCAZ5 alone suppresses tumor growth, and EPB augments the tumoricidal activity in human colorectal and hepatocellular tumor xenografts in athymic nude mice. These data suggest that the anti-DR5 chimeric antibody HMCAZ5 may have a clinical use and represents a useful immunological strategy, in combination with chemotherapy, for the treatment of cancer.
Collapse
Affiliation(s)
- Yuhe Qiu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
19
|
Zheng L, Weilun Z, Minghong J, Yaxi Z, Shilian L, Yanxin L, Dexian Z. Adeno-associated virus-mediated doxycycline-regulatable TRAIL expression suppresses growth of human breast carcinoma in nude mice. BMC Cancer 2012; 12:153. [PMID: 22530952 PMCID: PMC3404920 DOI: 10.1186/1471-2407-12-153] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 04/24/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) functions as a cytokine to selectively kill various cancer cells without toxicity to most normal cells. Numerous studies have demonstrated the potential use of recombinant soluble TRAIL as a cancer therapeutic agent. We have showed previous administration of a recombinant adeno-associated virus (rAAV) vector expressing soluble TRAIL results in an efficient suppression of human tumor growth in nude mice. In the present study, we introduced Tet-On gene expression system into the rAAV vector to control the soluble TRAIL expression and evaluate the efficiency of the system in cancer gene therapy. METHODS Controllability of the Tet-On system was determined by luciferase activity assay, and Western blotting and enzyme-linked immunoabsorbent assay. Cell viability was determined by MTT assay. The breast cancer xenograft animal model was established and recombinant virus was administrated through tail vein injection to evaluate the tumoricidal activity. RESULTS The expression of soluble TRAIL could be strictly controlled by the Tet-On system in both normal and cancer cells. Transduction of human cancer cell lines with rAAV-TRE-TRAIL&rAAV-Tet-On under the presence of inducer doxycycline resulted in a considerable cell death by apoptosis. Intravenous injection of the recombinant virus efficiently suppressed the growth of human breast carcinoma in nude mice when activated by doxycycline. CONCLUSION These data suggest that rAAV-mediated soluble TRAIL expression under the control of the Tet-On system is a promising strategy for breast cancer therapy.
Collapse
Affiliation(s)
- Liu Zheng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Zhang Weilun
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Jiang Minghong
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Zhang Yaxi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Liu Shilian
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Liu Yanxin
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Zheng Dexian
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005, China
| |
Collapse
|
20
|
Histone deacetylase inhibitors restore cell surface expression of the coxsackie adenovirus receptor and enhance CMV promoter activity in castration-resistant prostate cancer cells. Prostate Cancer 2012; 2012:137163. [PMID: 22288017 PMCID: PMC3263646 DOI: 10.1155/2012/137163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/27/2011] [Indexed: 12/20/2022] Open
Abstract
Adenoviral gene therapy using the death receptor ligand TRAIL as the therapeutic transgene can be safely administered via intraprostatic injection but has not been evaluated for efficacy in patients. Here we investigated the efficacy of adenoviral TRAIL gene therapy in a model of castration resistant prostate cancer and found that intratumoral injections can significantly delay tumor growth but cannot eliminate established lesions. We hypothesized that an underlying cause is inefficient adenoviral delivery. Using the LNCaP progression model of prostate cancer we show that surface CAR expression decreases with increasing tumorigenicity and that castration resistant C4-2b cells were more difficult to transduce with adenovirus than castration sensitive LNCaP cells. Many genes, including CAR, are epigenetically silenced during transformation but a new class of chemotherapeutic agents, known as histone deacetylase inhibitors (HDACi), can reverse this process. We demonstrate that HDACi restore CAR expression and infectivity in C4-2b cells and enhance caspase activation in response to infection with a TRAIL adenovirus. We also show that in cells with high surface CAR expression, HDACi further enhance transgene expression from the CMV promoter. Thus HDACi have multiple beneficial effects, which may enhance not only viral but also non-viral gene therapy of castration resistant prostate cancer.
Collapse
|
21
|
Iyer N, Marathe SA, Chaudhuri D, Garai P, Chakravortty D. Immunomodulation using agonists and antagonists: potential clinical applications. Expert Opin Investig Drugs 2012; 21:67-81. [PMID: 22149716 DOI: 10.1517/13543784.2012.642367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Extensive studies have gone into understanding the differential role of the innate and adaptive arms of the immune system in the context of various diseases. Receptor-ligand interactions are responsible for mediating cross-talk between the innate and adaptive arms of the immune system, so as to effectively counter the pathogenic challenge. While TLRs remain the best studied innate immune receptor, many other receptor families are now coming to the fore for their role in various pathologies. Research has focused on the discovery of novel agonists and antagonists for these receptors as potential therapeutics. AREAS COVERED In this review, we present an overview of the recent advances in the discovery of drugs targeting important receptors such as G-protein coupled receptors, TRAIL-R, IL-1β receptor, PPARs, etc. All these receptors play a critical role in the modulation of the immune response. We focus on the recent paradigms applied for the generation of specific and effective therapeutics for these receptors and their status in clinical trials. EXPERT OPINION Non-specific activation by antagonist/agonist is a difficult problem to dodge. This demands innovation in ligand designing with the use of strategies such as allosterism and dual-specific ligands. Rigorous preclinical and clinical studies are required in transforming a compound to a therapeutic.
Collapse
Affiliation(s)
- Namrata Iyer
- Indian Institute of Science, Centre for Infectious Disease Research and Biosafety Laboratories, Department of Microbiology and Cell Biology, Bangalore 560012, India
| | | | | | | | | |
Collapse
|
22
|
|
23
|
Tumor cell-selective apoptosis induction through targeting of K(V)10.1 via bifunctional TRAIL antibody. Mol Cancer 2011; 10:109. [PMID: 21899742 PMCID: PMC3179451 DOI: 10.1186/1476-4598-10-109] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 09/07/2011] [Indexed: 11/26/2022] Open
Abstract
Background The search for strategies to target ion channels for therapeutic applications has become of increasing interest. Especially, the potassium channel KV10.1 (Ether-á-go-go) is attractive as target since this surface protein is virtually not detected in normal tissue outside the central nervous system, but is expressed in approximately 70% of tumors from different origins. Methods We designed a single-chain antibody against an extracellular region of KV10.1 (scFv62) and fused it to the human soluble TRAIL. The KV10.1-specific scFv62 antibody -TRAIL fusion protein was expressed in CHO-K1 cells, purified by chromatography and tested for biological activity. Results Prostate cancer cells, either positive or negative for KV10.1 were treated with the purified construct. After sensitization with cytotoxic drugs, scFv62-TRAIL induced apoptosis only in KV10.1-positive cancer cells, but not in non-tumor cells, nor in tumor cells lacking KV10.1 expression. In co-cultures with KV10.1-positive cancer cells the fusion protein also induced apoptosis in bystander KV10.1-negative cancer cells, while normal prostate epithelial cells were not affected when present as bystander. Conclusions KV10.1 represents a novel therapeutic target for cancer. We could design a strategy that selectively kills tumor cells based on a KV10.1-specific antibody.
Collapse
|
24
|
TRAIL and doxorubicin combination enhances anti-glioblastoma effect based on passive tumor targeting of liposomes. J Control Release 2011; 154:93-102. [DOI: 10.1016/j.jconrel.2011.05.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/23/2011] [Accepted: 05/06/2011] [Indexed: 11/22/2022]
|
25
|
Balyasnikova IV, Ferguson SD, Han Y, Liu F, Lesniak MS. Therapeutic effect of neural stem cells expressing TRAIL and bortezomib in mice with glioma xenografts. Cancer Lett 2011; 310:148-59. [PMID: 21802840 DOI: 10.1016/j.canlet.2011.06.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 06/02/2011] [Accepted: 06/23/2011] [Indexed: 12/21/2022]
Abstract
Treatment of glioblastoma remains a challenge in neuro-oncology. We investigated if treatment with neural stem cells engineered to express membrane-bound TRAIL (NSCs-mTRAIL) alone or in combination with proteasome inhibitors is a feasible therapeutic approach for experimental glioma. Glioma cells showed resistance to soluble TRAIL and proteasome inhibitors alone, but responded well to their combined treatment. In co-culture with NSCs-mTRAIL, glioma cells appeared to be more prone to apoptosis than to treatment with soluble TRAIL, which was enhanced by proteasome inhibitor bortezomib. In vivo, the survival of animals bearing intracranial glial xenografts was significantly improved by NSCs-mTRAIL. The addition of bortezomib further enhanced the efficacy of NSCs-TRAIL treated group in one of examined tumor models. These data demonstrate that therapy with NSCs-mTRAIL is a potent cell based approach for treatment of glioma. Such an approach warrants further search for therapeutics capable of increasing sensitivity of glioma cells to mTRAIL in vivo.
Collapse
|
26
|
Yasukochi A, Kawakubo T, Nakamura S, Yamamoto K. Cathepsin E enhances anticancer activity of doxorubicin on human prostate cancer cells showing resistance to TRAIL-mediated apoptosis. Biol Chem 2011; 391:947-58. [PMID: 20482316 DOI: 10.1515/bc.2010.087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We previously described that cathepsin E specifically induces growth arrest and apoptosis in several human prostate cancer cell lines in vitro by catalyzing the proteolytic release of soluble tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) from the tumor cell surface. It also prevents tumor growth and metastasis in vivo through multiple mechanisms, including induction of apoptosis, angiogenesis inhibition and enhanced immune responses. Using the prostate cancer cell line PPC-1, which is relatively resistant to cell death by doxorubicin (40-50% cytotoxicity), we first report that a combination treatment with cathepsin E can overcome resistance of the cells to this agent. In vitro studies showed that combined treatment of PPC-1 cells with the two agents synergistically induces viability loss, mainly owing to down-regulation of a short form of the FLICE inhibitory protein FLIP. The enhanced antitumor activity was corroborated by in vivo studies with athymic mice bearing PPC-1 xenografts. Intratumoral application of cathepsin E in doxorubicin-treated mice results in tumor cell apoptosis and tumor regression in xenografts by enhanced TRAIL-induced apoptosis through doxorubicin-induced c-FLIP down-regulation and by a decrease in tumor cell proliferation. These results indicate that combination of cathepsin E and doxorubicin is sufficient to overcome resistance to TRAIL-mediated apoptosis in chemoresistant prostate cancer PPC-1 cells, thus indicating therapeutic potential for clinical use.
Collapse
Affiliation(s)
- Atsushi Yasukochi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | |
Collapse
|
27
|
Lee ALZ, Dhillon SHK, Wang Y, Pervaiz S, Fan W, Yang YY. Synergistic anti-cancer effects via co-delivery of TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) and doxorubicin using micellar nanoparticles. MOLECULAR BIOSYSTEMS 2011; 7:1512-22. [DOI: 10.1039/c0mb00266f] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
28
|
Wang Y, Huang F, Cai H, Wu Y, He G, Tan WS. The efficacy of combination therapy using adeno-associated virus-TRAIL targeting to telomerase activity and cisplatin in a mice model of hepatocellular carcinoma. J Cancer Res Clin Oncol 2010; 136:1827-37. [PMID: 20213096 DOI: 10.1007/s00432-010-0841-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Accepted: 02/15/2010] [Indexed: 12/20/2022]
Abstract
PURPOSE TNF-related apoptosis-inducing ligand (TRAIL) functions as a soluble cytokine and has been demonstrated significant antitumor activity against a variety of cancer cell lines without toxicity to most normal cells. Cisplatin is a potent anticancer agent and is widely used in the clinical for treatment of human cancers. Adeno-associated virus (AAV2) is a promising gene delivery vehicle for its advantage of low pathogenicity and long-term gene expression. However, lack of tissue specificity caused low efficiency of AAV transfer to target cells. The promoter of human telomerase reverse transcriptase (hTERT) is a good candidate to enhance targeting efficiency of AAV in cancer cells. Although AAV-mediated TRAIL controlled by hTERT promoter (AAV-hTERT-TRAIL) has obvious antitumor activity, the tumor cannot be completely eradicated. In this study, we first examined the effectiveness of combination therapy of cisplatin and AAV-hTERT-TRAIL on human hepatocellular carcinoma (HCC) in vitro and in vivo. METHODS For in vitro experiments, tumor cell lines were treated with cisplatin, virus, or both. The transgene TRAIL expression controlled by hTERT promoter was evaluated in BEL7404 HCC cell line. Cytotoxicity was performed by MTT analysis. Cell apoptosis was detected by flow cytometry analysis. The in vivo antitumor efficacy of combination treatment with cisplatin and AAV-hTERT-TRAIL was assessed in human hepatocellular carcinoma xenografts mouse model. RESULTS The enhanced TRAIL expression was observed in BEL7404 cells treated with AAV-hTERT-TRAIL plus cisplatin. Treatment with both AAV-hTERT-TRAIL and cisplatin exhibited stronger cytotoxicity and induced more significant apoptosis in cancer cells compared with AAV-hTERT-TRAIL or cisplatin alone, respectively. Moreover, in animal experiments, the combined treatment greatly suppressed tumor growth and resulted in tumor cell death. CONCLUSIONS AAV-mediated therapeutic gene expression in combination with chemotherapy provides a promising therapeutic strategy for human cancers. These data suggest that combined use of AAV-hTERT-TRAIL and cisplatin may have potential clinical application.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Blotting, Western
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Line
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cisplatin/pharmacology
- Combined Modality Therapy
- Dependovirus/genetics
- Female
- Humans
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Promoter Regions, Genetic/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- TNF-Related Apoptosis-Inducing Ligand/genetics
- TNF-Related Apoptosis-Inducing Ligand/metabolism
- Telomerase/genetics
- Telomerase/metabolism
- Transduction, Genetic
- Tumor Burden/drug effects
- Tumor Burden/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yigang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | | | | | | | | | | |
Collapse
|
29
|
Mahmood Z, Shukla Y. Death receptors: Targets for cancer therapy. Exp Cell Res 2010; 316:887-99. [DOI: 10.1016/j.yexcr.2009.12.011] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 12/08/2009] [Accepted: 12/13/2009] [Indexed: 12/24/2022]
|
30
|
Zhang J, Huang S, Zhang H, Wang H, Guo H, Qian G, Fan X, Lu J, Hoffman AR, Hu JF, Ge S. Targeted knockdown of Bcl2 in tumor cells using a synthetic TRAIL 3'-UTR microRNA. Int J Cancer 2010; 126:2229-39. [PMID: 19676053 DOI: 10.1002/ijc.24821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Targeting tumor-related overexpression of anti-apoptotic Bcl2 protein by RNAi has been suggested as a potential treatment for cancer. However, the stability of RNAi and its delivery are still major obstacles to the clinical testing of Bcl2 RNAi. Here, we explore a novel strategy of expressing a synthetic Bcl2 microRNA (smRNA) in the 3' untranslated region (UTR) of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), an apoptosis-inducing protein without apparent toxic effects in normal cells. TRAIL was specifically expressed from the human telomerase reverse transcriptase promoter (pTRT) that is active in many human tumors. Using this approach, we demonstrated that pTRT drove the tumor-specific expression of Bcl2 smRNA, which was processed by the host RNAi machinery and silenced endogenous Bcl2 expression in tumor cells. Bcl2 smRNA induced tumor cell apoptosis by activating caspase-3 and led to significant sensitization of tumor cells to TRAIL-induced apoptosis, while normal cells were spared. We also showed that the combined therapy of TRAIL-induced apoptosis and Bcl2 downregulation was superior to the mono-therapy of TRAIL or Bcl2 smRNA alone. This study proves a general paradigm for cancer therapy by using 3' UTR microRNA technology.
Collapse
Affiliation(s)
- Jianjun Zhang
- Department of Biochemistry and Molecular Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Holoch PA, Griffith TS. TNF-related apoptosis-inducing ligand (TRAIL): a new path to anti-cancer therapies. Eur J Pharmacol 2009; 625:63-72. [PMID: 19836385 PMCID: PMC2783837 DOI: 10.1016/j.ejphar.2009.06.066] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 06/08/2009] [Accepted: 06/22/2009] [Indexed: 12/31/2022]
Abstract
Since its discovery in 1995, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor super family, has been under intense focus because of its remarkable ability to induce apoptosis in malignant human cells while leaving normal cells unscathed. Consequently, activation of the apoptotic signaling pathway from the death-inducing TRAIL receptors provides an attractive, biologically-targeted approach to cancer therapy. A great deal of research has focused on deciphering the TRAIL receptor signaling cascade and intracellular regulation of this pathway, as many human tumor cells possess mechanisms of resistance to TRAIL-induced apoptosis. This review focuses on the current state of knowledge regarding TRAIL signaling and resistance, the preclinical development of therapies targeted at TRAIL receptors and modulators of the pathway, and the results of clinical trials for cancer treatment that have emerged from this base of knowledge. TRAIL-based approaches to cancer therapy vary from systemic administration of recombinant, soluble TRAIL protein with or without the combination of traditional chemotherapy, radiation or novel anti-cancer agents to agonistic monoclonal antibodies directed against functional TRAIL receptors to TRAIL gene transfer therapy. A better understanding of TRAIL resistance mechanisms may allow for the development of more effective therapies that exploit this cell-mediated pathway to apoptosis.
Collapse
Affiliation(s)
- Peter A Holoch
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA 52242, USA
| | | |
Collapse
|
32
|
Kasman LM, Barua S, Lu P, Rege K, Voelkel-Johnson C. Polymer-enhanced adenoviral transduction of CAR-negative bladder cancer cells. Mol Pharm 2009; 6:1612-9. [PMID: 19655763 DOI: 10.1021/mp9000958] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The application of adenoviral gene therapy for cancer is limited by immune clearance of the virus as well as poor transduction efficiency, since the protein used for viral entry (CAR) serves physiological functions in adhesion and is frequently decreased among cancer cells. Cationic polymers have been used to enhance adenoviral gene delivery, but novel polymers with low toxicity are needed to realize this approach. We recently identified polymers that were characterized by high transfection efficiency of plasmid DNA and a low toxicity profile. In this study we evaluated the novel cationic polymer EGDE-3,3' for its potential to increase adenoviral transduction of the CAR-negative bladder cancer cell line TCCSUP. The amount of adenovirus required to transduce 50-60% of the cells was reduced 100-fold when Ad.GFP was preincubated with the EGDE-3,3' polymer. Polyethyleneimine (pEI), a positively charged polymer currently used as a standard for enhancing adenoviral transduction, also increased infectivity, but transgene expression was consistently higher with EGDE-3,3'. In addition, EGDE-3,3'-supplemented transduction of an adenovirus expressing an apoptosis inducing transgene, Ad.GFP-TRAIL, significantly enhanced the amount of cell death. Thus, our results indicate that novel biocompatible polymers may be useful in improving the delivery of adenoviral gene therapy.
Collapse
Affiliation(s)
- Laura M Kasman
- Department of Microbiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | |
Collapse
|
33
|
Yang B, Wu X, Mao Y, Bao W, Gao L, Zhou P, Xie R, Zhou L, Zhu J. Dual-targeted antitumor effects against brainstem glioma by intravenous delivery of tumor necrosis factor-related, apoptosis-inducing, ligand-engineered human mesenchymal stem cells. Neurosurgery 2009; 65:610-24; discussion 624. [PMID: 19687708 DOI: 10.1227/01.neu.0000350227.61132.a7] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE We sought to explore the dual-targeted antitumor effects of membrane-spanned, tumor necrosis factor-related, apoptosis-inducing ligand (TRAIL)-engineered human mesenchymal stem cells (hMSCs) against brainstem gliomas. METHODS The migration capacity of hMSCs toward gliomas was studied by the Transwell system in vitro and by intravenous injection of hMSCs in glioma-bearing mice in vivo. MSCs were engineered with native full-length human TRAIL (hTRAIL) by a recombinant adeno-associated virus (rAAV) vector (rAAV-hTRAIL). The targeted antiglioma effect was analyzed by coculture of hTRAIL-engineered MSCs with glioma in vitro and by systematic delivery of hTRAIL-engineered MSCs to established human brainstem glioma xenografts. RESULTS We demonstrated systematically that transplanted MSCs migrated to a brainstem glioma with a high specificity. MSCs penetrated the vessels surrounding the tumor, then streamed in a chain pattern toward the glioma, eventually surrounding the tumor. Membrane-spanned, TRAIL-engineered MSCs not only expressed full-length TRAIL in MSC surface, but secreted some soluble TRAIL in medium. After being infected with rAAV-hTRAIL, hMSCs showed no increase in apoptosis. After coculture of hTRAIL-engineered MSCs and U87MG cells, the apoptosis of U87MG cells significantly increased more than soluble TRAIL-treated U87MG cells. Systematic delivery of hTRAIL-engineered MSCs to established human brainstem glioma xenografts resulted in the potent induction of apoptosis in gliomas, but not in normal brain and prolonged survival to 38.0 +/- 10.46 days compared with phosphate-buffered saline (16.0 +/- 0.66 days), soluble TRAIL (19.0 +/- 1.65 days), and hMSC-LacZ (14.0 +/- 0.59 days) treated groups. CONCLUSION Systematically transplanted MSCs migrated to gliomas with a high specificity. Systematic delivery of MSC-hTRAIL can prolong the survival of brainstem glioma-bearing mice, presumably through a dual-targeted effect of membrane-spanned, TRAIL-engineered MSCs in the tumor microenvironment. MSCs may be an effective vehicle for the targeted delivery of therapeutic agents to brainstem gliomas.
Collapse
Affiliation(s)
- Bojie Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Jiménez JA, Li X, Zhang YP, Bae KH, Mohammadi Y, Pandya P, Kao C, Gardner TA. Antitumor activity of Ad-IU2, a prostate-specific replication-competent adenovirus encoding the apoptosis inducer, TRAIL. Cancer Gene Ther 2009; 17:180-91. [PMID: 19798123 PMCID: PMC2821463 DOI: 10.1038/cgt.2009.62] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
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.
Collapse
Affiliation(s)
- J A Jiménez
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Rozanov DV, Savinov AY, Golubkov VS, Rozanova OL, Postnova TI, Sergienko EA, Vasile S, Aleshin AE, Rega MF, Pellecchia M, Strongin AY. Engineering a leucine zipper-TRAIL homotrimer with improved cytotoxicity in tumor cells. Mol Cancer Ther 2009; 8:1515-25. [PMID: 19509255 DOI: 10.1158/1535-7163.mct-09-0202] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Successful cancer therapies aim to induce selective apoptosis in neoplastic cells. The current suboptimal efficiency and selectivity drugs have therapeutic limitations and induce concomitant side effects. Recently, novel cancer therapies based on the use of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) have emerged. TRAIL, a key component of the natural antitumor immune response, selectively kills many tumor cell types. Earlier studies with recombinant TRAIL, however, revealed its many shortcomings including a short half-life, off-target toxicity, and existence of TRAIL-resistant tumor cells. We improved the efficacy of recombinant TRAIL by redesigning its structure and the expression and purification procedures. The result is a highly stable leucine zipper (LZ)-TRAIL chimera that is simple to produce and purify. This chimera functions as a trimer in a manner that is similar to natural TRAIL. The formulation of the recombinant LZ-TRAIL we have developed has displayed high specific activity in both cell-based assays in vitro and animal tests in vivo. Our results have shown that the half-life of LZ-TRAIL is improved and now exceeds 1 h in mice compared with a half-life of only minutes reported earlier for recombinant TRAIL. We have concluded that our LZ-TRAIL construct will serve as a foundation for a new generation of fully human LZ-TRAIL proteins suitable for use in preclinical and clinical studies and for effective combination therapies to overcome tumor resistance to TRAIL.
Collapse
Affiliation(s)
- Dmitri V Rozanov
- Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Members of the tumor necrosis factor (TNF) superfamily influence a variety of immunological functions, including cellular activation, proliferation, and death, upon interaction with a corresponding superfamily of receptors. Whereas interest in the apoptosis-inducing molecules TNF and Fas ligand has peaked because of their participation in events such as autoimmune disorders, activation-induced cell death, immune privilege, and tumor evasion from the immune system, another death-inducing family member, TNF-related apoptosis-inducing ligand (TRAIL), or Apo-2 ligand, has generated excitement because of its unique ability to induce apoptosis in a wide range of transformed cell lines but not in normal tissues. TRAIL is well tolerated when given to healthy animals, and no observable histological or functional changes have been observed in any of the tissues or organs examined. Moreover, multiple injections of soluble TRAIL into mice beginning the day after tumor implantation can significantly suppress the growth of the tumors, with many animals becoming tumor-free. One potential drawback to these findings is that large amounts of soluble TRAIL may be required to inhibit tumor formation, possibly because of the pharmacokinetic profile of soluble TRAIL that indicates that, after intravenous injection, the majority of the protein is rapidly cleared. Increasing the in vivo half-life of recombinant soluble TRAIL or developing an alternative means of delivery may increase the relative tumoricidal activity of TRAIL such that larger, more established tumors could be eradicated as efficiently as smaller tumors. The information presented here describes the production of an adenoviral vector engineered to carry the complementary DNA (cDNA) for murine TRAIL (hTRAIL).
Collapse
|
37
|
Wang Y, Huang F, Cai H, Zhong S, Liu X, Tan WS. Potent antitumor effect of TRAIL mediated by a novel adeno-associated viral vector targeting to telomerase activity for human hepatocellular carcinoma. J Gene Med 2008; 10:518-26. [PMID: 18338833 DOI: 10.1002/jgm.1177] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adeno-associated virus (AAV) has rapidly become a promising gene delivery vehicle for its excellent advantages of low pathogenicity and long-term gene expression. However, lack of tissue specificity caused low efficiency of AAV transfer to target cells. The promoter of human telomerase reverse transcriptase (hTERT) has been implicated in mediating gene expression in cancer cells as hTERT is transcriptionally upregulated in most cancer cells. Thereby, the hTERT promoter becomes a good candidate to enhance the targeting efficiency of AAV in cancer cells. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) functions as a soluble cytokine to selectively kill various cancer cells without toxicity to most normal cells. It remains to be determined whether the hTERT promoter can efficiently mediate TRAIL gene therapy in cancer cells using AAV vector. METHODS A novel AAV vector containing the TRAIL gene under the control of the hTERT promoter (AAV-hTERT-TRAIL) was generated. The specific expression of hTERT-controlled genes was evaluated in cell lines. The antitumor efficacy of AAV-hTERT-TRAIL was assessed in tumor cell lines and human hepatocellular carcinoma xenograft mouse model. RESULTS TRAIL expression was observed in tumor cells infected with AAV-hTERT-TRAIL at both the protein and mRNA level. AAV-hTERT-TRAIL displayed cancer-specific cytotoxicity and induced tumor cell apoptosis. Moreover, in animal experiments, intratumoral administration of AAV-hTERT-TRAIL significantly suppressed the growth of xenograft tumors and resulted in tumor cell death. CONCLUSIONS AAVs in combination with hTERT-mediated therapeutic gene expression provide a promising targeting approach for developing effective therapy for human cancers. These data suggest that AAV-hTERT-TRAIL is a potent therapeutic agent for cancer therapy.
Collapse
Affiliation(s)
- Yigang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | | | | | | | | | | |
Collapse
|
38
|
White-Gilbertson S, Rubinchik S, Voelkel-Johnson C. Transformation, translation and TRAIL: an unexpected intersection. Cytokine Growth Factor Rev 2008; 19:167-72. [PMID: 18353705 DOI: 10.1016/j.cytogfr.2008.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with roles in tumor surveillance and tolerance. TRAIL selectively induces apoptosis in many malignant but not normal cells but the underlying cause for spontaneous TRAIL sensitivity remains elusive. We propose a novel hypothesis that links TRAIL sensitivity to translational arrest following stresses that inactivate eukaryotic elongation factor 2 (EF2). Affected cells experience a reduction in apoptotic threshold because, due to their short half-lives, levels of anti-apoptotic proteins quickly drop off once translation elongation is inhibited leaving pro-apoptotic proteins unchallenged. This change in protein profile renders affected cells sensitive to TRAIL-mediated apoptosis and places EF2 into the role of a sensor for cellular damage.
Collapse
Affiliation(s)
- Shai White-Gilbertson
- Medical University of South Carolina, Department of Microbiology and Immunology, PO Box 250504/BSB201, 173 Ashley Avenue, Charleston, SC 29425, USA.
| | | | | |
Collapse
|
39
|
Wang X, Wang C, Qin YW, Yan SK, Gao YR. The association of up-regulation of X-linked inhibitor of apoptosis protein with cell adhesion-mediated drug resistance in U937 cells. Hematol Oncol 2008; 26:21-6. [PMID: 17721914 DOI: 10.1002/hon.828] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
An increasing body of evidence indicates that environmental factors may contribute to the drug resistance of acute myeloid leukaemia (AML). CAM-DR (cell adhesion-mediated drug resistance) is a reversible, de novo drug resistance induced by adhesion of tumour cell lines to fibronectin (FN). Adhesion was demonstrated to directly regulate the apoptotic machinery. And it was observed in previous studies that high levels of X-linked inhibitor of apoptosis protein (XIAP) were related to resistance to chemotherapeutics in many cancer cell lines. However, whether XIAP is relevant to CAM-DR of AML cells is unknown. In this report, we demonstrated that the mRNA and protein levels of XIAP were increased by 96.15% and 120.92%, respectively in U937 cells cocultured with FN as compared with controls. Antisense oligonucleotides targeting XIAP down-regulated the expression of XIAP and sensitized U937 cells to daunorubicin. In addition, we investigated the signalling pathway involved in the upregulation of XIAP. The levels of phosphorylated Akt (Ser473) were elevated in U937/FN cells and the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 suppressed XIAP expression and restored the chemosensitivity to daunorubicin. Our findings suggested that adhesion-dependent activation of the PI3K/Akt/XIAP pathway may be one of the factors involved in the CAM-DR of U937 cells. Targeting this pathway may be a useful approach to improve the therapeutic responsiveness of leukaemia cells.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Hematology, Cancer Hospital of Tianjin, Tianjin Medical University, Tianjin 300060, P.R. China
| | | | | | | | | |
Collapse
|
40
|
Mohr A, Lyons M, Deedigan L, Harte T, Shaw G, Howard L, Barry F, O'Brien T, Zwacka R. Mesenchymal stem cells expressing TRAIL lead to tumour growth inhibition in an experimental lung cancer model. J Cell Mol Med 2008; 12:2628-43. [PMID: 18373740 PMCID: PMC3828879 DOI: 10.1111/j.1582-4934.2008.00317.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lung cancer is a major public health problem in the western world, and gene therapy strategies to tackle this disease systemically are often impaired by inefficient delivery of the vector to the tumour tissue. Some of the main factors inhibiting systemic delivery are found in the blood stream in the form of red and white blood cells (WBCs) and serum components. Mesenchymal stem cells (MSCs) have been shown to home to tumour sites and could potentially act as a shield and vehicle for a tumouricidal gene therapy vector. Here, we describe the ability of an adenoviral vector expressing TRAIL (Ad.TR) to transduce MSCs and show the apoptosis-inducing activity of these TRAIL-carrying MSCs on A549 lung carcinoma cells. Intriguingly, using MSCs transduced with Ad.enhanced-green-fluorescent-protein (EGFP) we could show transfer of viral DNA to cocultured A549 cells resulting in transgenic protein production in these cells, which was not inhibited by exposure of MSCs to human serum containing high levels of adenovirus neutralizing antibodies. Furthermore, Ad.TR-transduced MSCs were shown not to induce T-cell proliferation, which may have resulted in cytotoxic T-cell-mediated apoptosis induction in the Ad.TR-transduced MSCs. Apoptosis was also induced in A549 cells by Ad.TR-transduced MSCs in the presence of physiological concentrations of WBC, erythrocytes and sera from human donors that inhibit or neutralize adenovirus alone. Moreover, we could show tumour growth reduction with TRAIL-loaded MSCs in an A549 xenograft mouse model. This is the first study that demonstrates the potential therapeutic utility of Ad.TR-transduced MSCs in cancer cells and the stability of this vector in the context of the blood environment.
Collapse
Affiliation(s)
- Andrea Mohr
- Molecular Therapeutics Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cisplatin enhances the antitumor effect of tumor necrosis factor-related apoptosis-inducing ligand gene therapy via recruitment of the mitochondria-dependent death signaling pathway. Cancer Gene Ther 2008; 15:356-70. [DOI: 10.1038/sj.cgt.7701120] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
42
|
Nastiuk KL, Yoo K, Lo K, Su K, Yeung P, Kutaka J, Danielpour D, Krolewski JJ. FLICE-Like Inhibitory Protein Blocks Transforming Growth Factor β1–Induced Caspase Activation and Apoptosis in Prostate Epithelial Cells. Mol Cancer Res 2008; 6:231-42. [DOI: 10.1158/1541-7786.mcr-07-0386] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Kikuchi S, Nagai T, Kunitama M, Kirito K, Ozawa K, Komatsu N. Active FKHRL1 overcomes imatinib resistance in chronic myelogenous leukemia-derived cell lines via the production of tumor necrosis factor-related apoptosis-inducing ligand. Cancer Sci 2007; 98:1949-58. [PMID: 17900262 PMCID: PMC11158645 DOI: 10.1111/j.1349-7006.2007.00623.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Revised: 08/12/2007] [Accepted: 08/22/2007] [Indexed: 12/20/2022] Open
Abstract
FKHRL1 (also called FOXO3a) is a member of the Forkhead Box, class O (FOXO) subfamily of forkhead transcription factors and functions downstream of Bcr-Abl tyrosine kinase as a phosphorylated inactive form in chronic myelogenous leukemia (CML). The Bcr-Abl tyrosine kinase inhibitor imatinib induces cell cycle arrest and subsequent apoptosis via the conversion of FKHRL1 from the phosphorylated inactive form to the dephosphorylated active form in CML-derived cell lines. In the present study, we examined whether active FKHRL1 can overcome resistance to imatinib. To this end, we generated a 4-hydroxytamoxifen-inducible active FKHRL1 (FKHRL1-TM; a triple mutant of FKHRL1 in which all three Akt phosphorylation sites have been mutated)-estrogen receptor fusion protein expression system in CML-derived imatinib-resistant cell lines. 4-Hydroxytamoxifen inhibited cell growth and cell cycle progression, and subsequently induced apoptosis, accompanied by upregulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Thus, active FKHRL1 antagonized deregulated proliferation and induced apoptosis in these cell lines. In addition, imatinib-resistant cells underwent apoptosis after transfection with full-length TRAIL cDNA. Collectively, our results suggest that active FKHRL1 can overcome imatinib resistance in CML cells, in part via TRAIL production.
Collapse
Affiliation(s)
- Satoru Kikuchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi-ken 329-0498, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Voortman J, Resende TP, Abou El Hassan MAI, Giaccone G, Kruyt FAE. TRAIL therapy in non-small cell lung cancer cells: sensitization to death receptor-mediated apoptosis by proteasome inhibitor bortezomib. Mol Cancer Ther 2007; 6:2103-12. [PMID: 17620439 DOI: 10.1158/1535-7163.mct-07-0167] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activation of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor pathway is a promising therapeutic strategy to selectively eradicate cancer cells, including non-small cell lung cancer (NSCLC) cells. Recombinant human (rh) TRAIL/Apo-2L, a TRAIL-encoding adenovirus, and monoclonal antibodies directed against TRAIL receptors R1 and R2 were used to study cytotoxicity of TRAIL therapy in NSCLC cells. NSCLC cells showed differential sensitivity to TRAIL therapy, regardless of the agent used. Combination treatment of bortezomib and rhTRAIL led to synergistic apoptosis induction in NSCLC cell lines. Enhancement of rhTRAIL-induced apoptosis by bortezomib was caspase dependent, implicating extrinsic as well as intrinsic apoptosis activation, as shown by increased processing of caspase-8 as well as caspase-9, and could be abrogated completely by overexpression of caspase-8 inhibitor cytokine response modifier A (CrmA), and partially by overexpression of Bcl-2. Enhanced surface expression of TRAIL-R2, but also TRAIL-R1, was associated with bortezomib treatment, which is likely to contribute to the increased processing of caspase-8 in the combination treatment. Furthermore, TRAIL-induced activation of prosurvival transcription factor nuclear factor-kappaB was prevented by cotreatment with bortezomib, which may contribute to the observed synergistic apoptosis induction. Our preclinical data indicate that combination therapy of TRAIL and bortezomib may be an effective strategy for NSCLC.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Boronic Acids/pharmacology
- Bortezomib
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Caspase Inhibitors
- Cell Line, Tumor
- Drug Synergism
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Models, Biological
- NF-kappa B/metabolism
- Proteasome Inhibitors
- Pyrazines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Death Domain/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
Collapse
Affiliation(s)
- Jens Voortman
- Department of Medical Oncology, CCA 2.36, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
45
|
Figueiredo ML, Kao C, Wu L. Advances in preclinical investigation of prostate cancer gene therapy. Mol Ther 2007; 15:1053-64. [PMID: 17457317 PMCID: PMC2826150 DOI: 10.1038/sj.mt.6300181] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
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.
Collapse
Affiliation(s)
- Marxa L Figueiredo
- Department of Urology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Chinghai Kao
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lily Wu
- Department of Urology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
46
|
Kasman L, Lu P, Voelkel-Johnson C. The histone deacetylase inhibitors depsipeptide and MS-275, enhance TRAIL gene therapy of LNCaP prostate cancer cells without adverse effects in normal prostate epithelial cells. Cancer Gene Ther 2006; 14:327-34. [PMID: 17186014 DOI: 10.1038/sj.cgt.7701017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Gene therapy of cancer using adenovirus as a single treatment modality has met limited success and efforts to enhance therapeutic outcomes have included combination of gene therapy with chemotherapy. The goal of this study was to investigate which chemotherapeutic agents may be suitable for combination with gene therapy of prostate cancer. Using an adenovirus expressing green fluorescent protein (GFP), we determined the effect of cisplatin, gemcitabine, doxorubicin, depsipeptide and MS-275 on adenoviral infectivity and transgene expression in LNCaP cells. We found that the two histone deacetylase inhibitors (HDACi), depsipeptide and MS-275, and to a lesser extent doxorubicin, increased infectivity and transgene expression. However, only the HDACi selectively increased infectivity in LNCaP cells while doxorubicin increased infectivity to a greater extent in normal prostate epithelial cells (PrEC). The increase in infectivity but not transgene expression correlated to increased surface expression of coxsackie and adenovirus receptor (CAR). Increased transgene expression following infection with an adenovirus expressing tumor necrosis factor-related apoptosis inducing ligand (TRAIL) was observed only in LNCaP cells treated with depsipeptide or MS-275. Combination of TRAIL gene therapy with HDACi but not doxorubicin resulted in increased induction of apoptosis in LNCaP cells. In contrast, apoptosis was not enhanced by HDACi in normal PrEC. These results suggest that combination of HDACi with adenoviral TRAIL gene therapy may be a new therapeutic approach for the treatment of prostate cancer that warrants further investigation.
Collapse
Affiliation(s)
- L Kasman
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29403, USA
| | | | | |
Collapse
|
47
|
Yang F, Shi P, Xi X, Yi S, Li H, Sun Q, Sun M. Recombinant adenoviruses expressing TRAIL demonstrate antitumor effects on non-small cell lung cancer (NSCLC). Med Oncol 2006; 23:191-204. [PMID: 16720919 DOI: 10.1385/mo:23:2:191] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2005] [Revised: 11/30/1999] [Accepted: 09/13/2005] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in a variety of malignant cells, but not in normal cells. This preferential toxicity to the abnormal cells renders TRAIL potentially a very powerful therapeutic weapon against cancer. However, a requirement for large quantities of TRAIL to suppress tumor growth in vivo is one of the major factors that has hindered it from being widely applied clinically. To overcome this, we constructed a replication-deficient adenovirus that carries a human full-length TRAIL gene (Ad-TRAIL) and tested its efficacy against a lung cancer model system in comparison to that of the recombinant soluble TRAIL protein. METHODS To investigate the antitumor activity and therapeutic value of the Ad-TRAIL on the non-small cell lung cancer (NSCLC), four NSCLC cell lines, namely, YTMLC, GLC, A549, and H460 cells, were used. TRAIL protein expression was determined by Western blotting and flow cytometry. Cell viability was analyzed by proliferation assay, and DNA ladder and cell-cycle analysis were used to identify apoptosis. To further evaluate the effect of Ad-TRAIL in vivo, YTMLC cells were inoculated to the subcutis of nude mice. The Ad-TRAIL was subsequently administered into the established tumors. Tumor growth and the TRAIL toxicity were evaluated after treatment. RESULTS YTMLC cells infected with Ad-TRAIL showed decreased cell viability and a higher percentage of apoptosis. Similar, Ad-TRAIL treatment also significantly suppressed tumor growth in vivo. CONCLUSIONS TRAIL gene therapy provides a promising therapy for the treatment of NSCLC.
Collapse
MESH Headings
- Adenoviridae
- Animals
- Apoptosis/genetics
- Apoptosis Regulatory Proteins/biosynthesis
- Apoptosis Regulatory Proteins/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/ultrastructure
- Cell Line, Tumor
- Cell Proliferation
- Genetic Therapy
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/therapy
- Lung Neoplasms/ultrastructure
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/therapy
- Neoplasms, Experimental/ultrastructure
- TNF-Related Apoptosis-Inducing Ligand
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- F Yang
- Institute of Medical Biology, Chinese Academy of Medical Science, Jiaoling Road 379, Kunming, Yunnan 650118, P.R. China
| | | | | | | | | | | | | |
Collapse
|
48
|
Miller-Jensen K, Janes KA, Wong YL, Griffith LG, Lauffenburger DA. Adenoviral vector saturates Akt pro-survival signaling and blocks insulin-mediated rescue of tumor necrosis-factor-induced apoptosis. J Cell Sci 2006; 119:3788-98. [PMID: 16940353 DOI: 10.1242/jcs.03102] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recombinant adenoviruses are used extensively as delivery vectors in clinical gene therapy and in molecular biology, but little is known about how the viral carrier itself contributes to cellular responses. Here we show that infection with an E1/E3-deleted adenoviral vector (Adv) sensitizes human epithelial cells to tumor necrosis factor (TNF)-induced apoptosis. To explore the mechanism of Adv-mediated sensitization, we measured activity time courses for three protein kinases (MK2, IKK and JNK1) centrally involved in the TNF-receptor signaling network, as well as two kinases (Akt and ERK) activated by growth factors. Both the pro-apoptotic signal MK2 and the anti-apoptotic signal Akt were upregulated when Adv-infected cells were stimulated with TNF, and MK2 and Akt each contributed significantly to TNF-induced cell fate. Surprisingly, further activation of Akt in Adv-infected cells via insulin treatment did not significantly reduce apoptosis or MK2 activity. We show that the ineffectiveness of insulin-mediated anti-apoptotic signaling through Akt is due to saturation of Akt-effector substrate phosphorylation in Adv-infected cells. Normalizing Akt signaling relative to its Adv-induced baseline activity identified a global dose-response curve that relates Akt signaling to cellular survival. Thus, the background Akt activity induced by Adv limits the transmission of anti-apoptotic signals in response to further cytokine or growth-factor stimulation. The phenotypic and intracellular synergy between Adv and TNF may have implications for interpreting cellular responses in gene-therapy and laboratory applications.
Collapse
Affiliation(s)
- Kathryn Miller-Jensen
- Biotechnology Process Engineering Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | |
Collapse
|
49
|
Desai P, Jiménez JA, Kao C, Gardner TA. Future innovations in treating advanced prostate cancer. Urol Clin North Am 2006; 33:247-72, viii. [PMID: 16631463 DOI: 10.1016/j.ucl.2005.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many novel techniques for the treatment of prostate cancer are being aggressively investigated because prostate cancer is prevalent in the population and the current treatments for advanced prostate cancer are woefully inadequate. Although the current treatment options prolong life, most patients will eventually experience local recurrence or develop advanced disease. A greater understanding of the molecular events underlying cancer has enabled investigators to explore gene therapy approaches that are targeted against these molecular events. This article discusses antiangiogenic therapy, immune based therapy, and gene therapy. Any of these experimental modalities could be developed to replace hormone ablation therapy which causes unpleasant side effects, decreases the quality of life of the patient, and only temporarily controls the disease.
Collapse
Affiliation(s)
- Pratik Desai
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
50
|
Norris JS, Bielawska A, Day T, El-Zawahri A, ElOjeimy S, Hannun Y, Holman D, Hyer M, Landon C, Lowe S, Dong JY, McKillop J, Norris K, Obeid L, Rubinchik S, Tavassoli M, Tomlinson S, Voelkel-Johnson C, Liu X. Combined therapeutic use of AdGFPFasL and small molecule inhibitors of ceramide metabolism in prostate and head and neck cancers: a status report. Cancer Gene Ther 2006; 13:1045-51. [PMID: 16763610 DOI: 10.1038/sj.cgt.7700965] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
As of January 2005, there were 1020 gene therapy clinical trials ongoing worldwide with 675 or 66.2% devoted to cancer gene therapy. The majority are occurring in the US and Europe (http://www.wiley.co.uk/genetherapy/clinical/). At the present time, to our knowledge there are no trials that employ gene delivery of Fas Ligand (FasL). As an important note, and in contrast to somatic cell therapy trials, there are no reported deaths due to therapeutic vector administration in any cancer gene therapy trial. That said, from our studies and from the published literature, the issue of gene delivery remains the major obstacle to successfully employing gene therapy for cancer treatment. Numerous laboratories are studying this with many different approaches. My co-workers and I have focused on the delivery issue by using various approaches that address tumor targeting and transgene expression. In addition, we are focusing on enhancing tumor cell killing via the bystander effect and through use of small molecules to enhance bystander activity.
Collapse
Affiliation(s)
- J S Norris
- Department of Microbiology, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|