1
|
Zeh N, Schlossbauer P, Raab N, Klingler F, Handrick R, Otte K. Cell line development for continuous high cell density biomanufacturing: Exploiting hypoxia for improved productivity. Metab Eng Commun 2021; 13:e00181. [PMID: 34401326 PMCID: PMC8348152 DOI: 10.1016/j.mec.2021.e00181] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 01/18/2023] Open
Abstract
Oxygen deficiency (hypoxia) induces adverse effects during biotherapeutic protein production leading to reduced productivity and cell growth. Hypoxic conditions occur during classical batch fermentations using high cell densities or perfusion processes. Here we present an effort to create novel engineered Chinese hamster ovary (CHO) cell lines by exploiting encountered hypoxic bioprocess conditions to reinforce cellular production capacities. After verifying the conservation of the hypoxia-responsive pathway in CHO cell lines by analyzing oxygen sensing proteins HIF1a, HIF1β and VDL, hypoxia-response-elements (HREs) were functionally analyzed and used to create hypoxia-responsive expression vectors. Subsequently engineered hypoxia sensitive CHO cell lines significantly induced protein expression (SEAP) during adverse oxygen limitation encountered during batch fermentations as well as high cell density perfusion processes (2.7 fold). We also exploited this novel cell system to establish a highly effective oxygen shift as innovative bioprocessing strategy using hypoxia induction to improve production titers. Thus, substantial improvements can be made to optimize CHO cell productivity for novel bioprocessing challenges as oxygen limitation, providing an avenue to establish better cell systems by exploiting adverse process conditions for optimized biotherapeutic production.
Collapse
Affiliation(s)
- Nikolas Zeh
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Patrick Schlossbauer
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Nadja Raab
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Florian Klingler
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - René Handrick
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| |
Collapse
|
2
|
Zhang W, Zhang C, Tian W, Qin J, Chen J, Zhang Q, Fang L, Zheng J. Efficacy of an Oncolytic Adenovirus Driven by a Chimeric Promoter and Armed with Decorin Against Renal Cell Carcinoma. Hum Gene Ther 2020; 31:651-663. [PMID: 32216478 DOI: 10.1089/hum.2019.352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Virus-targeted therapy for tumors can effectively prolong the survival rate of patients and has become a new trend for cancer biotherapy. Oncolytic adenovirus (OAd) can specifically replicate in tumor cells, allowing the therapeutic genes carried to be rapidly copied. As known, solid tumors are always hypoxic, and researchers often overlook a key point, the replication of OAd depends not only on its own activity but also on the cellular hypoxic environment in which the virus replicates. In this study, we constructed an OAd carrying Decorin, HRE-Ki67-Decorin, combining the Ki67 promoter upstreamed with hypoxia-response element (HRE) sequences to drive adenoviral E1A. The OAd HRE-Ki67-Decorin had better replication ability under hypoxic conditions, downregulated cellular immunosuppressed growth factor TGF-β. In addition, HRE-Ki67-Decorin was potent in suppressing tumor growth and participated in the assembly of tumor extracellular matrix by expressing Decorin in subcutaneous renal cancer cell tumor models. Tumor sections from HRE-Ki67-Decorin-treated tissues had less collagen fibers and more spread of virus among tumor tissues. These results indicated that chimeric HRE-Ki67 promoter-regulated OAd carrying Decorin might be an effective anticancer treatment strategy.
Collapse
Affiliation(s)
- Wen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and
| | - Chen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and
| | - Weiping Tian
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and
| | - Jing Qin
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and
| | - Jing Chen
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qi Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; and.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Yu K, Xiang L, Li S, Wang S, Chen C, Mu H. HIF1α promotes prostate cancer progression by increasing ATG5 expression. Anim Cells Syst (Seoul) 2019; 23:326-334. [PMID: 31700698 PMCID: PMC6830197 DOI: 10.1080/19768354.2019.1658637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/09/2019] [Accepted: 07/26/2019] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer among men. However, the major modifiable risk factors for PCa are poorly known and its specific mechanism of progression remains unclear. Here we reported that, in prostate cancer cells, the autophagy level was elevated under hypoxic condition, as well as the mRNA and protein level of ATG5, which is an important gene related to autophagy. Furthermore, we found HIF1α could directly bind to the promoter of ATG5 and promote the expression of ATG5 on transcriptional level by luciferase assay and ChIP assay. Intriguingly, overexpression of HIF1α by HIF1α-M could increase tumor size and the effect could be abolished by knockdown ATG5 by si-ATG5 in BALB/cA-nu/nu nude mice. Importantly, HIF1α could also promote the metastasis of PC-3 cells by upregulating the ATG5 and autophagy level and knockdown ATG5 and inhibition autophagy both could abolish the effect of overexpression of HIF1α on the migration of PC-3 cells. Taken together, our results, for the first time, proved that HIF1α could promote the proliferation and migration of PC-3 cells by direct upregulating ATG5 and autophagy level in PC-3 prostate cancer cells. Our findings not only provide new perspective for the relationship between hypoxia and autophagy, but also add new potential therapeutic regimens for the treatment of prostate cancers.
Collapse
Affiliation(s)
- Kaiyuan Yu
- The Second Affiliated Hospital & YuYing Children's Hospital of Wenzhou Medical University, Wenzhou City, People's Republic of China
| | - Luxia Xiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shaoxun Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shuaibin Wang
- The Second Affiliated Hospital & YuYing Children's Hospital of Wenzhou Medical University, Wenzhou City, People's Republic of China
| | - Chaohao Chen
- The Second Affiliated Hospital & YuYing Children's Hospital of Wenzhou Medical University, Wenzhou City, People's Republic of China
| | - Haiqi Mu
- The Second Affiliated Hospital & YuYing Children's Hospital of Wenzhou Medical University, Wenzhou City, People's Republic of China
| |
Collapse
|
4
|
Javan B, Shahbazi M. Constructing a Novel Hypoxia-Inducible Bidirectional shRNA Expression Vector for Simultaneous Gene Silencing in Colorectal Cancer Gene Therapy. Cancer Biother Radiopharm 2018; 33:118-123. [PMID: 29641253 DOI: 10.1089/cbr.2017.2401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Nonspecific siRNA expression limits its application in cancer gene therapy. Therefore, a tightly regulated and reversibly inducible RNAi system is required to conditionally control the gene expression. This investigation aims at constructing a hypoxia/colorectal tumor dual-specific bidirectional short hairpin RNA (shRNA) expression vector. MATERIALS AND METHODS First, carcinoma embryonic antigen (CEA) promoter designed in two directions. Then, pRNA-bipHRE-CEA vector was constructed by insertion of the vascular endothelial growth factor enhancer between two promoters for hypoxic cancer-specific gene expression. To confirm the therapeutic effect of the dual-specific vector, two shRNA oligonucleotides were inserted in the downstream of each promoter. QRT-polymerase chain reaction and western blot assays were performed to estimate the mRNA and protein expression levels. RESULTS Both mRNA and protein levels were significantly reduced (50%-60%) in the hypoxic colorectal cancer-treated cells when compared with the controls. CONCLUSION The novel bidirectional hypoxia-inducible shRNA expression vector may be efficient in colorectal cancer-specific gene therapy.
Collapse
Affiliation(s)
- Bita Javan
- 1 Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences , Gorgan, Iran .,2 Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences , Gorgan, Iran
| | - Majid Shahbazi
- 1 Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences , Gorgan, Iran .,2 Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences , Gorgan, Iran .,3 Arya Tina Gene (ATG), Biopharmaceutical Company , Gorgan, Iran
| |
Collapse
|
5
|
Javan B, Atyabi F, Shahbazi M. Hypoxia-inducible bidirectional shRNA expression vector delivery using PEI/chitosan-TBA copolymers for colorectal Cancer gene therapy. Life Sci 2018; 202:140-151. [PMID: 29656061 DOI: 10.1016/j.lfs.2018.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/07/2018] [Accepted: 04/11/2018] [Indexed: 10/17/2022]
Abstract
AIMS This investigation was conducted to construct a hypoxia/colorectal dual-specific bidirectional short hairpin RNA (shRNA) expression vector and to transfect it into the colon cancer cell line HT-29 with PEI/chitosan-TBA nanoparticles for the simultaneous knock down of β-catenin and Bcl-2 under hypoxia. MAIN METHODS To construct a pRNA-bipHRE-CEA vector, the carcinoma embryonic antigen (CEA) promoter designed in two directions and the vascular endothelial growth factor (VEGF) enhancer were inserted between two promoters for hypoxic cancer specific gene expression. To confirm the therapeutic effect of the dual-specific vector, β-catenin and Bcl-2 shRNAs were inserted downstream of each promoter. The physicochemical properties, the cytotoxicity, and the transfection efficiency of these PEI/chitosan-TBA nanoparticles were investigated. In addition, the antitumor effects of the designed vector on the expression of β-catenin and Bcl-2, cell cycle distribution, and apoptosis were investigated in vitro. KEY FINDINGS The silencing effect of the hypoxia-response shRNA expression vector was relatively low (18%-25%) under normoxia, whereas it was significantly increased to approximately 50%-60% in the HT-29 cell line. Moreover, the cancer cells showed significant G0/G1 arrest and increased apoptosis due to gene silencing under hypoxia. Furthermore, MTS assay, fluorescence microscopy images, and flow cytometry analyses confirmed that the PEI/chitosan-TBA blend system provided effective transfection with low cytotoxicity. SIGNIFICANCE This novel hypoxia-responsive shRNA expression vector may be useful for RNA interference (RNAi)-based cancer gene therapy in hypoxic colorectal tumors. Moreover, the PEI/chitosan-TBA copolymer might be a promising gene carrier for use in gene transfer in vivo.
Collapse
Affiliation(s)
- Bita Javan
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran; Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Nanotechnology Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| | - Majid Shahbazi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran; Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Arya Tina Gene (ATG), Biopharmaceutical Company, Gorgan, Iran.
| |
Collapse
|
6
|
A hypoxia- and telomerase-responsive oncolytic adenovirus expressing secretable trimeric TRAIL triggers tumour-specific apoptosis and promotes viral dispersion in TRAIL-resistant glioblastoma. Sci Rep 2018; 8:1420. [PMID: 29362367 PMCID: PMC5780382 DOI: 10.1038/s41598-018-19300-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 12/27/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma is a highly aggressive and malignant type of cancer that is apoptosis resistant and difficult to cure by conventional cancer therapies. In this regard, an oncolytic adenovirus that selectively targets the tumour tissue and induces tumour cell lysis is a promising treatment option. We designed and constructed a hypoxia-responsive and cancer-specific modified human telomerase reverse transcriptase (H5CmTERT) promoter to drive replication of an oncolytic adenovirus (H5CmTERT-Ad). To enhance the anti-tumour efficacy of H5CmTERT-Ad against malignant glioblastoma, we also generated an H5CmTERT-Ad expressing secretable trimeric tumour necrosis factor-related apoptosis-inducing ligand (H5CmTERT-Ad/TRAIL). H5CmTERT promoter-regulated oncolytic adenoviruses showed cancer-specific and superior cell-killing effect in contrast to a cognate control oncolytic adenovirus replicating under the control of the endogenous adenovirus promoter. The cancer cell-killing effects of H5CmTERT-Ad and H5CmTERT-Ad/TRAIL were markedly higher during hypoxia than normoxia owing to hypoxia responsiveness of the promoter. H5CmTERT-Ad/TRAIL showed more potent anti-tumour efficacy than H5CmTERT-Ad did in a xenograft model of TRAIL-resistant subcutaneous and orthotopic glioblastoma through superior induction of apoptosis and more extensive virus distribution in the tumour tissue. Altogether, our findings show that H5CmTERT-Ad/TRAIL can promote dispersion of an oncolytic adenovirus through robust induction of apoptosis in a highly TRAIL-resistant glioblastoma.
Collapse
|
7
|
Javan B, Shahbazi M. Hypoxia-inducible tumour-specific promoters as a dual-targeting transcriptional regulation system for cancer gene therapy. Ecancermedicalscience 2017; 11:751. [PMID: 28798809 PMCID: PMC5533602 DOI: 10.3332/ecancer.2017.751] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Indexed: 12/25/2022] Open
Abstract
Transcriptional targeting is the best approach for specific gene therapy. Hypoxia is a common feature of the tumour microenvironment. Therefore, targeting gene expression in hypoxic cells by placing transgene under the control of a hypoxia-responsive promoter can be a good strategy for cancer-specific gene therapy. The hypoxia-inducible gene expression system has been investigated more in suicide gene therapy and it can also be of great help in knocking down cancer gene therapy with siRNAs. However, this system needs to be optimised to have maximum efficacy with minimum side effects in normal tissues. The combination of tissue-/tumour-specific promoters with HRE core sequences has been found to enhance the specificity and efficacy of this system. In this review, hypoxia-inducible gene expression system as well as gene therapy strategies targeting tumour hypoxia will be discussed. This review will also focus on hypoxia-inducible tumour-specific promoters as a dual-targeting transcriptional regulation systems developed for cancer-specific gene therapy.
Collapse
Affiliation(s)
- Bita Javan
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan 4934174515, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan 4934174515, Iran
| |
Collapse
|
8
|
Quantification of hypoxia-related gene expression as a potential approach for clinical outcome prediction in breast cancer. PLoS One 2017; 12:e0175960. [PMID: 28430808 PMCID: PMC5400273 DOI: 10.1371/journal.pone.0175960] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/03/2017] [Indexed: 01/08/2023] Open
Abstract
Breast cancers are solid tumors frequently characterized by regions with low oxygen concentrations. Cellular adaptations to hypoxia are mainly determined by “hypoxia inducible factors” that mediate transcriptional modifications involved in drug resistance and tumor progression leading to metastasis and relapse occurrence. In this study, we investigated the prognostic value of hypoxia-related gene expression in breast cancer. A systematic review was conducted to select a set of 45 genes involved in hypoxia signaling pathways and breast tumor progression. Gene expression was quantified by RT-qPCR in a retrospective series of 32 patients with invasive ductal carcinoma. Data were analyzed in relation to classical clinicopathological criteria and relapse occurrence. Coordinated overexpression of selected genes was observed in high-grade and HER2+ tumors. Hierarchical cluster analysis of gene expression significantly segregated relapsed patients (p = 0.008, Chi2 test). All genes (except one) were up-regulated and six markers were significantly expressed in tumors from recurrent patients. The expression of this 6-gene set was used to develop a basic algorithm for identifying recurrent patients according to a risk score of relapse. Analysis of Kaplan-Meier relapse-free survival curves allowed the definition of a threshold score of 2 (p = 0.021, Mantel-Haenszel test). The risk of recurrence was increased by 40% in patients with a high score. In addition to classical prognostic factors, we showed that hypoxic markers have potential prognostic value for outcome and late recurrence prediction, leading to improved treatment decision-making for patients with early-stage invasive breast cancer. It will be necessary to validate the clinical relevance of this prognostic approach through independent studies including larger prospective patient cohorts.
Collapse
|
9
|
Zhang Y, Liu NM, Wang Y, Youn JY, Cai H. Endothelial cell calpain as a critical modulator of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1326-1335. [PMID: 28366876 DOI: 10.1016/j.bbadis.2017.03.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/04/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Calpains are a family of calcium-dependent non-lysosomal cysteine proteases. In particular, calpains residing in the endothelial cells play important roles in angiogenesis. It has been shown that calpain activity can be increased in endothelial cells by growth factors, primarily vascular endothelial growth factor (VEGF). VEGF/VEGFR2 induces calpain 2 dependent activation of PI3K/AMPK/Akt/eNOS pathway, and consequent nitric oxide production and physiological angiogenesis. Under pathological conditions such as tumor angiogenesis, endothelial calpains can be activated by hypoxia. This review focuses on the molecular regulatory mechanisms of calpain activation, and the newly identified mechanistic roles and downstream signaling events of calpains in physiological angiogenesis, and in the conditions of pathological tumor angiogenesis and diabetic wound healing, as well as retinopathy and atherosclerosis that are also associated with an increase in calpain activity. Further discussed include the differential strategies of modulating angiogenesis through manipulating calpain expression/activity in different pathological settings. Targeted limitation of angiogenesis in cancer and targeted promotion of angiogenesis in diabetic wound healing via modulations of calpains and calpain-dependent signaling mechanisms are of significant translational potential. Emerging strategies of tissue-specific targeting, environment-dependent targeting, and genome-targeted editing may turn out to be effective regimens for targeted manipulation of angiogenesis through calpain pathways, for differential treatments including both attenuation of tumor angiogenesis and potentiation of diabetic angiogenesis.
Collapse
Affiliation(s)
- Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Norika Mengchia Liu
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Yongchen Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA.
| |
Collapse
|
10
|
Ivey JW, Bonakdar M, Kanitkar A, Davalos RV, Verbridge SS. Improving cancer therapies by targeting the physical and chemical hallmarks of the tumor microenvironment. Cancer Lett 2016; 380:330-9. [PMID: 26724680 PMCID: PMC4919249 DOI: 10.1016/j.canlet.2015.12.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 12/18/2022]
Abstract
Tumors are highly heterogeneous at the patient, tissue, cellular, and molecular levels. This multi-scale heterogeneity poses significant challenges for effective therapies, which ideally must not only distinguish between tumorous and healthy tissue, but also fully address the wide variety of tumorous sub-clones. Commonly used therapies either leverage a biological phenotype of cancer cells (e.g. high rate of proliferation) or indiscriminately kill all the cells present in a targeted volume. Tumor microenvironment (TME) targeting represents a promising therapeutic direction, because a number of TME hallmarks are conserved across different tumor types, despite the underlying genetic heterogeneity. Historically, TME targeting has largely focused on the cells that support tumor growth (e.g. vascular endothelial cells). However, by viewing the intrinsic physical and chemical alterations in the TME as additional therapeutic opportunities rather than barriers, a new class of TME-inspired treatments has great promise to complement or replace existing therapeutic strategies. In this review we summarize the physical and chemical hallmarks of the TME, and discuss how these tumor characteristics either currently are, or may ultimately be targeted to improve cancer therapies.
Collapse
Affiliation(s)
- Jill W Ivey
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA
| | - Mohammad Bonakdar
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Akanksha Kanitkar
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA; Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Scott S Verbridge
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA.
| |
Collapse
|
11
|
Patel A, Sant S. Hypoxic tumor microenvironment: Opportunities to develop targeted therapies. Biotechnol Adv 2016; 34:803-812. [PMID: 27143654 PMCID: PMC4947437 DOI: 10.1016/j.biotechadv.2016.04.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/13/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023]
Abstract
In recent years, there has been great progress in the understanding of tumor biology and its surrounding microenvironment. Solid tumors create regions with low oxygen levels, generally termed as hypoxic regions. These hypoxic areas offer a tremendous opportunity to develop targeted therapies. Hypoxia is not a random by-product of the cellular milieu due to uncontrolled tumor growth; rather it is a constantly evolving participant in overall tumor growth and fate. This article reviews current trends and recent advances in drug therapies and delivery systems targeting hypoxia in the tumor microenvironment. In the first part, we give an account of important physicochemical changes and signaling pathways activated in the hypoxic microenvironment. This is then followed by various treatment strategies including hypoxia-sensitive signaling pathways and approaches to develop hypoxia-targeted drug delivery systems.
Collapse
Affiliation(s)
- Akhil Patel
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
12
|
Hypoxia-Sensitive Materials for Biomedical Applications. Ann Biomed Eng 2016; 44:1931-45. [DOI: 10.1007/s10439-016-1578-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 02/23/2016] [Indexed: 10/22/2022]
|
13
|
Harris BHL, Barberis A, West CML, Buffa FM. Gene Expression Signatures as Biomarkers of Tumour Hypoxia. Clin Oncol (R Coll Radiol) 2015; 27:547-60. [PMID: 26282471 DOI: 10.1016/j.clon.2015.07.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/16/2015] [Indexed: 02/08/2023]
Abstract
Hypoxia is a feature of most solid tumours and is associated with a poor prognosis. The hypoxic environment can reduce the efficacy of radiotherapy and some chemotherapeutics, and has been investigated extensively as a therapeutic target. The clinical use of hypoxia-targeting treatment will benefit from the development of a biomarker to assess tumour hypoxia. There are several possible techniques that measure either the level of oxygen or the tumour molecular response to hypoxia. The latter includes gene expression profiling, which measures the transcriptional response of a tumour to its hypoxic microenvironment. A systematic review identified 32 published hypoxia gene expression signatures. The methods used for their derivation varied, but are broadly classified as: (i) identifying genes with significantly higher or lower expression in cancer cells cultured under hypoxic versus normoxic conditions; (ii) using either previously characterised hypoxia-regulated genes/biomarkers to define hypoxic tumours and then identifying other genes that are over- or under-expressed in the hypoxic tumours. Both generated gene signatures useful in furthering our understanding of hypoxia biology. However, signatures derived using the second method seem to be superior in terms of providing prognostic information. Here we summarise all 32 published hypoxia signatures, discuss their commonalities and differences, and highlight their strengths and limitations. This review also highlights the importance of reproducibility and gene annotation, which must be accounted for to transfer signatures robustly for clinical application as biomarkers.
Collapse
Affiliation(s)
- B H L Harris
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - A Barberis
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - C M L West
- Translational Radiobiology Group, Institute of Cancer Sciences, University of Manchester, Christie Hospital, Manchester M13 9PT, UK
| | - F M Buffa
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
14
|
Biswal MR, Prentice HM, Dorey CK, Blanks JC. A hypoxia-responsive glial cell-specific gene therapy vector for targeting retinal neovascularization. Invest Ophthalmol Vis Sci 2014; 55:8044-53. [PMID: 25377223 DOI: 10.1167/iovs.14-13932] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Müller cells, the major glial cell in the retina, play a significant role in retinal neovascularization in response to tissue hypoxia. We previously designed and tested a vector using a hypoxia-responsive domain and a glial fibrillary acidic protein (GFAP) promoter to drive green fluorescent protein (GFP) expression in Müller cells in the murine model of oxygen-induced retinopathy (OIR). This study compares the efficacy of regulated and unregulated Müller cell delivery of endostatin in preventing neovascularization in the OIR model. METHODS Endostatin cDNA was cloned into plasmids with hypoxia-regulated GFAP or unregulated GFAP promoters, and packaged into self-complementary adeno-associated virus serotype 2 vectors (scAAV2). Before placement in hyperoxia on postnatal day (P)7, mice were given intravitreal injections of regulated or unregulated scAAV2, capsid, or PBS. Five days after return to room air, on P17, neovascular and avascular areas, as well as expression of the transgene and vascular endothelial growth factor (VEGF), were compared in OIR animals treated with a vector, capsid, or PBS. RESULTS The hypoxia-regulated, glial-specific, vector-expressing endostatin reduced neovascularization by 93% and reduced the central vaso-obliteration area by 90%, matching the results with the unregulated GFAP-Endo vector. Retinas treated with the regulated endostatin vector expressed substantial amounts of endostatin protein, and significantly reduced VEGF protein. Endostatin production from the regulated vector was undetectable in retinas with undamaged vasculature. CONCLUSIONS These findings suggest that the hypoxia-regulated, glial cell-specific vector expressing endostatin may be useful for treatment of neovascularization in proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- Manas R Biswal
- Integrative Biology PhD Program, Florida Atlantic University, Boca Raton, Florida, United States
| | - Howard M Prentice
- Center for Complex Systems and Brain Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
| | - C Kathleen Dorey
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| | - Janet C Blanks
- Center for Complex Systems and Brain Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States
| |
Collapse
|
15
|
Hsiao HT, Xing L, Deng X, Sun X, Ling CC, Li GC. Hypoxia-targeted triple suicide gene therapy radiosensitizes human colorectal cancer cells. Oncol Rep 2014; 32:723-9. [PMID: 24912473 PMCID: PMC4091884 DOI: 10.3892/or.2014.3238] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/25/2014] [Indexed: 12/21/2022] Open
Abstract
The hypoxic microenvironment, an important feature of human solid tumors but absent in normal tissue, may provide an opportunity for cancer-specific gene therapy. The purpose of the present study was to investigate whether hypoxia-driven triple suicide gene TK/CD/UPRT expression enhances cytotoxicity to ganciclovir (GCV) and 5-fluorocytosine (5-FC), and sensitizes human colorectal cancer to radiation in vitro and in vivo. Stable transfectant of human colorectal HCT8 cells was established which expressed hypoxia-inducible vectors (HRE-TK/eGFP and HRE-CD/UPRT/mDsRed). Hypoxia-induced expression/function of TK, CD and UPRT was verified by western blot analysis, flow cytometry, fluorescent microscopy and cytotoxicity assay of GCV and 5-FC. Significant radiosensitization effects were detected after 5-FC and GCV treatments under hypoxic conditions. In the tumor xenografts, the distribution of TK/eGFP and CD/UPRT/mDsRed expression visualized with fluorescence microscopy was co-localized with the hypoxia marker pimonidazole positive staining cells. Furthermore, administration of 5-FC and GCV in mice in combination with local irradiation resulted in tumor regression, as compared with prodrug or radiation treatments alone. Our data suggest that the hypoxia-inducible TK/GCV+CDUPRT/5-FC triple suicide gene therapy may have the ability to specifically target hypoxic cancer cells and significantly improve the tumor control in combination with radiotherapy.
Collapse
Affiliation(s)
- Hung Tsung Hsiao
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Ligang Xing
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Xuelong Deng
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Xiaorong Sun
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - C Clifton Ling
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Gloria C Li
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
16
|
Hypoxia as a target for tissue specific gene therapy. J Control Release 2013; 172:484-94. [DOI: 10.1016/j.jconrel.2013.05.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 05/13/2013] [Accepted: 05/24/2013] [Indexed: 12/28/2022]
|
17
|
Zhu W, Zhang H, Shi Y, Song M, Zhu B, Wei L. Oncolytic adenovirus encoding tumor necrosis factor-related apoptosis inducing ligand (TRAIL) inhibits the growth and metastasis of triple-negative breast cancer. Cancer Biol Ther 2013; 14:1016-23. [PMID: 24025362 DOI: 10.4161/cbt.26043] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) is a promising cancer therapeutic target due to its selective apoptosis-inducing effect in cancer cells. To efficiently deliver TRAIL to the tumor cells, an oncolytic adenovirus (p55-hTERT-HRE-TRAIL) carrying the TRAIL coding sequence was constructed. In the present study, we aimed to investigate the effect of p55-hTERT-HRE-TRAIL on the growth and metastasis of triple-negative breast cancer (TNBC). We observed that infection of the recombinant adenovirus resulted in expression of TRAIL and massive cell death in a TNBC cell line MDA-MB-231. This effect is much weaker in MCF-10A, which is a normal breast cell line. Administration of P55-HTERT-HRE-TRAIL significantly reduced orthotopic breast tumor growth and extended survival in a metastatic model. Our results suggest the oncolytic adenovirus armed with P55-HTERT-HRE-TRAIL, which exhibited enhanced anti-tumor activity and improved survival, is a promising candidate for virotherapy of TNBC.
Collapse
Affiliation(s)
- Wei Zhu
- Department of General Surgery; Zhongshan Hospital; Fudan University, Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Shan YF, Fang YF, Wang XQ, Jin R, Zhang QY, Andersson R. Experimental studies on treatment of pancreatic cancer with double-regulated duplicative adenovirus AdTPHre-hEndo carrying human endostatin gene. Pancreatology 2013; 13:393-400. [PMID: 23890138 DOI: 10.1016/j.pan.2013.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/27/2013] [Accepted: 05/31/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gene-virus targeted therapy is a promising new method of treating pancreatic cancer. To increase the efficacy and decrease the side-effect, we constructed a conditionally replicative adenovirus (CRAd) expressing human endostatin, with a human Telomoerase Reverse Transcriptase (hTERT) promoter for the regulation of the early stage of adenovirus expression of gene E1a and a Hypoxia Response Element (HRE) promoter to regulate the gene E1b. METHODS A gene recombination technique was adopted to construct and generate the adenovirus AdTPHre-hEndo. Pancreatic cancer cells were studied both in vitro and in vivo. Western blotting was adopted to observe the expressions of protein E1A and E1B; duplication assay was applied to observe the selective duplication capability of recombinant cells. MTT assay was applied to measure the lethal effects of virus on pancreatic cancer cells, and ELISA was adopted to detect the human endostatin gene expression. A pancreatic cancer transplantation tumor model of nude mice was constructed to observe the antitumor effects of the virus. RESULTS Double-regulated duplicative adenovirus AdTPHre-hEndo genes were successfully constructed. Duplication and lethal assays proved that AdTPHre-hEndo could replicate specifically in pancreatic cancer cells and kill them. The endostatin expression in a cultured supernatant from tumor cells was significantly higher than that obtained from non-duplicative adenovirus vectors carrying that gene. The animal experiment demonstrated that AdTPHre-hEndo has a high capability to limit pancreatic cancer growth. CONCLUSIONS AdTPHre-hEndo has a special ability to duplicate and kill pancreatic cancer cells in in vitro and in vivo experiments, thus providing a new gene-virus-based treatment system for pancreatic cancer.
Collapse
Affiliation(s)
- Yun-feng Shan
- Department of Surgery, First Affiliated Hospital, Wenzhou Medical College, Wenzhou 325000, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|
20
|
Sun X, Xing L, Deng X, Hsiao HT, Manami A, Koutcher JA, Clifton Ling C, Li GC. Hypoxia targeted bifunctional suicide gene expression enhances radiotherapy in vitro and in vivo. Radiother Oncol 2012; 105:57-63. [PMID: 22938726 DOI: 10.1016/j.radonc.2012.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 06/22/2012] [Accepted: 07/17/2012] [Indexed: 12/26/2022]
Abstract
PURPOSE To investigate whether hypoxia targeted bifunctional suicide gene expression-cytosine deaminase (CD) and uracil phosphoribosyltransferase (UPRT) with 5-FC treatments can enhance radiotherapy. MATERIALS AND METHODS Stable transfectants of R3327-AT cells were established which express a triple-fusion-gene: CD, UPRT and monomoric DsRed (mDsRed) controlled by a hypoxia inducible promoter. Hypoxia-induced expression/function of CDUPRTmDsRed was verified by western blot, flow cytometry, fluorescent microscopy, and cytotoxicity assay of 5-FU and 5-FC. Tumor-bearing mice were treated with 5-FC and local radiation. Tumor volume was monitored and compared with those treated with 5-FC or radiation alone. In addition, the CDUPRTmDsRed distribution in hypoxic regions of tumor sections was visualized with fluorescent microscopy. RESULTS Hypoxic induction of CDUPRTmDsRed protein correlated with increased sensitivity to 5-FC and 5-FU. Significant radiosensitization effects were detected after 5-FC treatments under hypoxic conditions. In the tumor xenografts, the distribution of CDUPRTmDsRed expression visualized with fluorescence microscopy was co-localized with the hypoxia marker pimonidazole positive staining cells. Furthermore, administration of 5-FC to mice in combination with local irradiation resulted in significant tumor regression, as in comparison with 5-FC or radiation treatments alone. CONCLUSIONS Our data suggest that the hypoxia-inducible CDUPRT/5-FC gene therapy strategy has the ability to specifically target hypoxic cancer cells and significantly improve the tumor control in combination with radiotherapy.
Collapse
Affiliation(s)
- Xiaorong Sun
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Li J, Zhang G, Liu T, Gu H, Yan L, Chen B. Construction of a novel vector expressing the fusion suicide gene yCDglyTK and hTERT-shRNA and its antitumor effects. Exp Ther Med 2012. [PMID: 23181115 PMCID: PMC3503886 DOI: 10.3892/etm.2012.613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This study aimed to construct a novel recombinant expression vector, pcDNA3.1(-)hTERT-shRNA/yCDglyTK. Its bioactivity and antitumor effects were investigated in the SGC7901 human gastric cancer cell line. Interfering RNA (RNAi) targeting human telomerase reverse transcriptase (hTERT) was applied to construct the pYr1.1-hTERT-shRNA vector. The shRNA expression cassette (including U6 promoter) was subcloned into the pcDNA3.1(-) CV-yCDglyTK vector to build a new vector, pcDNA3.1(-) hTERT-shRNA/yCDglyTK, which was identified by restriction enzyme digestion and gene sequencing. All the plasmids were delivered into SGC7901 cells using calcium phosphate nanoparticles (CPNPs). Expression of yCDglyTK and hTERT was detected by immunofluorescence, real-time PCR and western blot analysis. MTT assays were applied to measure the cytotoxic effect of the plasmids with 5-fluorocytosine (5-FC). Cell apoptosis was detected by flow cytometry. Restriction enzyme digestion and gene sequencing confirmed that the recombinant vector pcDNA3.1(-)hTERT-shRNA/yCDglyTK had been successfully constructed. Immunofluorescence, real-time PCR and western blot analysis showed that yCDglyTK was expressed, and that hTERT expression was inhibited in cells transfected with the recombinant vector. The cells transfected with the recombinant vector were the most sensitive to 5-FC and the apoptosis rates of the cells were also increased. The pcDNA3.1(-)hTERTshRNA/yCDglyTK vector was constructed successfully; it was confirmed that targeting hTERT through RNAi could synergize with suicide gene therapy.
Collapse
Affiliation(s)
- Jia Li
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | | | | | | | | | | |
Collapse
|
22
|
Zhu W, Wei L, Zhang H, Chen J, Qin X. Oncolytic adenovirus armed with IL-24 inhibits the growth of breast cancer in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:51. [PMID: 22640485 PMCID: PMC3511263 DOI: 10.1186/1756-9966-31-51] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/06/2012] [Indexed: 11/18/2022]
Abstract
Background Interleukin-24 (IL-24) is a cytokine that belongs to the IL-10 family. It can selectively induce cancer cell apoptosis which has been utilized as a cancer gene therapy strategy. Methods A recombinant type five adenovirus containing IL-24 gene (designated CNHK600-IL24) was constructed, whose replication is activated only in tumor cells. The replication of CNHK600-IL24 in breast tumor cells and fibroblasts were assessed by TCID50 and MTT assay; the secretion of IL-24 was measured by ELISA and western blotting. The in vivo anti-tumor effect of CNHK600-IL24 was investigated in nude mice carrying orthotopic or metastatic breast tumor. Results We observed that CNHK600-IL24 could replicate efficiently and resulted in high level IL-24 expression and massive cell death in human breast cancer cell MDA-MB-231 but not in normal fibroblast cell MRC-5. In addition, orthotopic breast tumor growth in the nude mice model was significantly suppressed when CNHK600-IL24 was administered. In the metastatic model generated by tail vein injection, CNHK600-IL24 virotherapy significantly improved survival compared with the same virus expressing EGFP (median survival CNHK600-IL24, 55 days vs. CNHK600-EGFP, 41 day, p < 0.05 Mantal-Cox test). A similar phenomenon was observed in the metastatic model achieved by left ventricular injection as suggested by in vivo luminescence imaging of tumor growth. Conclusion The oncolytic adenovirus armed with IL-24, which exhibited enhanced anti-tumor activity and improved survival, is a promising candidate for virotherapy of breast cancer.
Collapse
Affiliation(s)
- Wei Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | | | | | | | | |
Collapse
|
23
|
Immunohistochemical expression of VEGF, HIF1-a, and PlGF in malignant melanomas and dysplastic nevi. Melanoma Res 2012; 21:389-94. [PMID: 21876459 DOI: 10.1097/cmr.0b013e328347ee33] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We evaluated the role of vascular endothelial growth factor (VEGF), placental growth factor (PlGF), and hypoxia-inducible factor 1-a (HIF1-a) in melanoma angiogenesis and investigated their expression in dysplastic nevi, as potential melanoma precursors. In addition, we examined a possible correlation of VEGF expression with PlGF and HIF1-a. These factors were detected immunohistochemically in 95 melanomas of all types and stages and in 28 dysplastic nevi. We used 10 intradermal melanocytic nevi as controls. HIF1-a was expressed in 93 out of 95 (97.89%) melanomas and in none of the dysplastic or control nevi. HIF1-a expression was more intense in melanocytes around necrotic areas but did not correlate with melanoma type, the Clark staging or the Breslow thickness. A strong positive association was detected between HIF1-a and VEGF expression in all cases. VEGF was detected in 82 out of 95 (86.31%) melanomas and in 21 out of 28 (75%) dysplastic nevi, whereas it was expressed weakly in neoplastic cells of the controls. Its expression was more intense in melanomas, especially in nodular melanomas of elevated stage and thickness. PIGF was detected in 46 out of 95 (48.42%) melanomas and in none of the nevi. Expression did not correlate with melanoma staging nor thickness; however, it was more intense in superficial spreading melanomas, where a weak positive association between VEGF and PlGF was also detected. There was no association between HIF1-a and PlGF in any melanoma type. Hypoxia, through the expression of HIF1-a, plays a key role in melanoma progression; it activates VEGF secretion, which induces angiogenesis and metastasis. The role of PlGF seems to be limited.
Collapse
|
24
|
Effects of YC-1 targeting hypoxia-inducible factor 1 alpha in oesophageal squamous carcinoma cell line Eca109 cells. Cell Biol Int 2011; 35:491-7. [DOI: 10.1042/cbi20090419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
25
|
Kwon OJ, Kim PH, Huyn S, Wu L, Kim M, Yun CO. A hypoxia- and {alpha}-fetoprotein-dependent oncolytic adenovirus exhibits specific killing of hepatocellular carcinomas. Clin Cancer Res 2010; 16:6071-82. [PMID: 21169258 DOI: 10.1158/1078-0432.ccr-10-0664] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Oncolytic adenoviruses (Ad) constitute a new promising modality of cancer gene therapy that displays improved efficacy over nonreplicating Ads. We have previously shown that an E1B 19-kDa-deleted oncolytic Ad exhibits a strong cell-killing effect but lacks tumor selectivity. To achieve hepatoma-restricted cytotoxicity and enhance replication of Ad within the context of tumor microenvironment, we used a modified human α-fetoprotein (hAFP) promoter to control the replication of Ad with a hypoxia response element (HRE). EXPERIMENTAL DESIGN We constructed Ad-HRE(6)/hAFPΔ19 and Ad-HRE(12)/hAFPΔ19 that incorporated either 6 or 12 copies of HRE upstream of promoter. The promoter activity and specificity to hepatoma were examined by luciferase assay and fluorescence-activated cell sorting analysis. In addition, the AFP expression- and hypoxia-dependent in vitro cytotoxicity of Ad-HRE(6)/hAFPΔ19 and Ad-HRE(12)/hAFPΔ19 was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and cytopathic effect assay. In vivo tumoricidal activity on subcutaneous and liver orthotopic model was monitored by noninvasive molecular imaging. RESULTS Ad-HRE(12)/hAFPΔ19 exhibited enhanced tumor selectivity and cell-killing activity when compared with Ad-hAFPΔ19. The tumoricidal activity of Ad-HRE(12)/hAFPΔ19 resulted in significant inhibition of tumor growth in both subcutaneous and orthotopic models. Histologic examination of the primary tumor after treatment confirmed accumulation of viral particles near hypoxic areas. Furthermore, Ad-HRE(12)/hAFPΔ19 did not cause severe inflammatory immune response and toxicity after systemic injection. CONCLUSIONS The results presented here show the advantages of incorporating HREs into a hAFP promoter-driven oncolytic virus. This system is unique in that it acts in both a tissue-specific and tumor environment-selective manner. The greatly enhanced selectivity and tumoricidal activity of Ad-HRE(12)/hAFPΔ19 make it a promising therapeutic agent in the treatment of liver cancers.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Brain Korea 21 Project for Medical Sciences, Institute for Cancer Research, Yonsei Cancer Center, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
26
|
Dudek AZ. Endothelial lineage cell as a vehicle for systemic delivery of cancer gene therapy. Transl Res 2010; 156:136-46. [PMID: 20801410 DOI: 10.1016/j.trsl.2010.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 07/12/2010] [Accepted: 07/13/2010] [Indexed: 01/14/2023]
Abstract
A major limitation of cancer gene therapy is the difficulty of delivering a therapeutic gene to distant sites of metastatic disease. A promising strategy to address this difficulty is to use expanded ex vivo cells to produce a therapeutic protein. As with other approaches to gene therapy, this strategy is attractive when the therapeutic protein is unstable ex vivo or has a short circulating half life in vivo. The initial step to develop a cancer gene therapy using autologous cell delivery is the identification of a cell type that migrates to the tumor site, is readily available for harvesting, and is manipulated easily ex vivo. Recent evidence suggests that endothelial progenitor, precursor, and blood outgrowth endothelial cells are attracted to the tumor vasculature by its angiogenic drive. Here, we review recent advances in the study of circulating endothelial cell-mediated tumor vasculogenesis and discuss the advantages and challenges of bringing endothelial lineage-based cancer gene therapy closer to clinical application.
Collapse
Affiliation(s)
- Arkadiusz Z Dudek
- Division of Hematology, Oncologyand Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
27
|
Abstract
Using biochemical, imaging and histological methods, we employed transcriptional targeting to increase the specificity of tumor gene expression in vivo for intravenously administered recombinant adenovirus vectors. Surprisingly, the relative specificity of tumor expression in comparison to other tissues was increased for a constitutively expressing recombinant adenovirus, AdCMVLuc, by simply reducing the viral dose. Even at lower doses, however, the high frequency of viral infection and transgene expression in the liver using constitutive promoters still represents a substantial problem. To further augment tumor specificity, we constructed a series of adenoviruses expressing luciferase from several other promoters and tested their ability to selectively transcribe genes in tumor cells both in vitro and in vivo. Constitutively active viral promoters (RSV, SRα) varied widely in their tumor selectivity, but hypoxia-responsive promoters (carbonic anhydrase 9, PAI-1, SOD2, and several chimeric constructs) demonstrated the most tumor-selective expression. Our results show that tumor targeting to HT1080 fibrosarcomas was readily achieved using transcriptional targeting mechanisms. We attribute the relatively high level of gene transfer and expression in HT1080 tumors in vivo to increased viral access to the tumor, presumably due to discontinuities in tumor vasculature and augmented expression from stress-responsive promoters in the hypoxic and inflammatory tumor microenvironment.
Collapse
Affiliation(s)
- R T Hogg
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | | | | |
Collapse
|
28
|
Abstract
The abnormal decrease or the lack of oxygen supply to cells and tissues is called hypoxia. This condition is commonly seen in various diseases such as rheumatoid arthritis and atherosclerosis, also in solid cancers. Pre-clinical and clinical studies have shown that hypoxic cancers are extremely aggressive, resistant to standard therapies (chemotherapy and radiotherapy), and thus very difficult to eradicate. Hypoxia affects both the tumor and the immune cells via various pathways. This review summarizes the most common effects of hypoxia on immune cells that play a key role in the anti-tumor response, the limitation of current therapies, and the potential solutions that were developed for hypoxic malignancies.
Collapse
Affiliation(s)
- Patricia Yotnda
- Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, 77030, Houston, TX, USA.
| | | | | |
Collapse
|
29
|
Hiley CT, Yuan M, Lemoine NR, Wang Y. Lister strain vaccinia virus, a potential therapeutic vector targeting hypoxic tumours. Gene Ther 2009; 17:281-7. [PMID: 19890355 PMCID: PMC2875103 DOI: 10.1038/gt.2009.132] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hypoxia contributes to the aggressive and treatment-resistant phenotype of pancreatic ductal adenocarcinoma (PDAC). Oncolytic vaccinia virus has potential as an anti-tumour agent but the ability to lyse hypoxic tumour cells is vital for clinical efficacy. We hypothesised that unique aspects of the poxvirus lifecycle would protect it from attenuation in hypoxic conditions. We characterised and compared the viral protein production, viral replication, cytotoxicity and transgene expression of Lister strain vaccinia virus in a panel of pancreatic cancer cell lines after exposure to normoxic or hypoxic conditions. Viral protein production was not affected by hypoxia, and high viral titres were produced in both normoxic and hypoxic conditions. Interestingly there was a five-fold (P<0.001) and 10-fold (P<0.0001) increase in viral cytotoxicity for CFPac1 and MiaPaca2 cell lines respectively in hypoxic conditions. Cytotoxicity was equivalent in the remaining cell lines. Levels of transgene expression (luciferase reporter gene) from the vaccinia viral vector were comparable regardless of the ambient oxygen concentration. The present study suggests that vaccinia virus is a promising vector for targeting pancreatic cancer and potentially other hypoxic tumour types.
Collapse
Affiliation(s)
- C T Hiley
- Centre for Molecular Oncology & Imaging, Institute of Cancer, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | |
Collapse
|
30
|
Lee M. Hypoxia targeting gene expression for breast cancer gene therapy. Adv Drug Deliv Rev 2009; 61:842-9. [PMID: 19426773 DOI: 10.1016/j.addr.2009.04.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2008] [Accepted: 04/28/2009] [Indexed: 01/03/2023]
Abstract
Gene therapy is a promising strategy to treat various inherited and acquired diseases. However, targeting gene expression to specific tissue is required to minimize side effects of gene therapy. Hypoxia is present in the microenvironment of solid tumors such as breast tumors. A hypoxic tumor targeting gene expression system has been developed for cancer gene therapy. In hypoxic tissues, hypoxia inducible factor (HIF)-1alpha is accumulated and stimulates transcription of the genes that have hypoxia response elements (HREs) in their promoters. Therefore, transcriptional regulation with a hypoxia inducible promoter is the most widely used strategy for hypoxic tumors targeting gene therapy. In breast cancer gene therapy, breast tumor specific promoters in combination with HREs have been used to induce gene expression in hypoxic breast tumors. Post-transcriptional regulation using an untranslated region (UTR) is also a useful strategy to increase gene expression in hypoxic tumor tissue. In addition, post-translational regulation with the oxygen-dependent degradation (ODD) domain is effective to eliminate therapeutic gene products and reduce side effects in normal tissue. In combination with the breast tumor specific promoters, hypoxic tumor targeting strategies will be useful for the development of a safe breast cancer gene therapy.
Collapse
Affiliation(s)
- Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea.
| |
Collapse
|
31
|
Kizaka-Kondoh S, Tanaka S, Hiraoka M. Imaging and Targeting of the Hypoxia-inducible Factor 1-active Microenvironment. J Toxicol Pathol 2009; 22:93-100. [PMID: 22271982 PMCID: PMC3246054 DOI: 10.1293/tox.22.93] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 11/28/2008] [Indexed: 12/04/2022] Open
Abstract
Human solid tumors contain hypoxic regions that have considerably lower oxygen tension than normal tissues. They are refractory to radiotherapy and anticancer chemotherapy. Although more than half a century has passed since it was suggested that tumour hypoxia correlates with poor treatment outcomes and contributes to recurrence of cancer, no fundamental solution to this problem has been found. Hypoxia-inducible factor-1(HIF-1) is the main transcription factor that regulates the cellular response to hypoxia. It induces various genes, whose function is strongly associated with malignant alteration of the entire tumour. The cellular changes induced by HIF-1 are extremely important therapeutic targets of cancer therapy, particularly in therapy against refractory cancers. Therefore, targeting strategies to overcome the HIF-1-active microenvironment are important for cancer therapy. To Target HIF-1-active/ hypoxic tumor cells, we developed a fusion protein drug, PTD-ODD-Procaspase-3 that selectively induces cell death in HIF-1-active/hypoxic cells. The drug consists of the following three functional domains: the protein transduction domain (PTD), which efficiently delivers the fusion protein to hypoxic tumor cells, the ODD domain, which has a VHL-mediated protein destruction motif of human HIF-1α protein and confers hypoxia-dependent stabilization to the fusion proteins, and the human procaspase-3 proenzyme responsible for the cytocidal activity of the protein drug. In vivo imaging systems capable of monitoring HIF-1 activity in transplanted human cancer cells in mice are useful in evaluating the efficiency of these drugs and in study of HIF-1-active tumor cells.
Collapse
Affiliation(s)
- Shinae Kizaka-Kondoh
- Innovative Techno-Hub for Integrated Medical Bio-imaging, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shotaro Tanaka
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masahiro Hiraoka
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
32
|
Kizaka-Kondoh S, Tanaka S, Harada H, Hiraoka M. The HIF-1-active microenvironment: an environmental target for cancer therapy. Adv Drug Deliv Rev 2009; 61:623-32. [PMID: 19409433 DOI: 10.1016/j.addr.2009.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 01/28/2009] [Indexed: 12/20/2022]
Abstract
Solid tumors possess unique microenvironments that are exposed to chronic hypoxic conditions, so-called tumor hypoxia. Although more than half a century has passed since it was suggested that tumor hypoxia correlated with bad treatment outcomes and contributed to the recurrence of cancer, no fundamental solution to this problem has yet been found. Hypoxia-inducible factor HIF-1 is the main transcription factor that regulates the cellular response to hypoxia. It induces various genes, whose function is strongly associated with the malignant alteration of the entire tumor. The cellular changes induced by HIF-1 are extremely important therapeutic targets of cancer therapy, particularly in the therapy against refractory cancers. Therefore targeting strategies to overcome the HIF-1-active microenvironment are important for cancer therapy.
Collapse
Affiliation(s)
- Shinae Kizaka-Kondoh
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | |
Collapse
|
33
|
Gomes EM, Rodrigues MS, Phadke AP, Butcher LD, Starling C, Chen S, Chang D, Hernandez-Alcoceba R, Newman JT, Stone MJ, Tong AW. Antitumor activity of an oncolytic adenoviral-CD40 ligand (CD154) transgene construct in human breast cancer cells. Clin Cancer Res 2009; 15:1317-25. [PMID: 19228733 DOI: 10.1158/1078-0432.ccr-08-1360] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE CD40 ligand (CD40L, CD154) plays a central role in immunoregulation and also directly modulates epithelial cell growth and differentiation. We previously showed that the CD40 receptor is commonly expressed in primary breast cancer tissues. In this proof-of-principle study, we examined the breast cancer growth-regulatory activities of an oncolytic adenoviral construct carrying the CD40L transgene (AdEHCD40L). EXPERIMENTAL DESIGN In vitro and in vivo evaluations were carried out on AdEHCD40L to validate selective viral replication and CD40L transgene activity in hypoxia inducing factor-1alpha and estrogen receptor-expressing human breast cancer cells. RESULTS AdEHCD40L inhibited the in vitro growth of CD40+ human breast cancer lines (T-47D, MDA-MB-231, and BT-20) by up to 80% at a low multiplicity of infection of 1. Incorporation of the CD40L transgene reduced the effective dose needed to achieve 50% growth inhibition (ED50) by approximately 10-fold. In contrast, viral and transgene expression of AdEHCD40L, as well its cytotoxicity, was markedly attenuated in nonmalignant cells. Intratumoral injections with AdEHCD40L reduced preexisting MDA-MB-231 xenograft growth in severe combined immunodeficient mice by >99% and was significantly more effective (P<0.003) than parental virus AdEH (69%) or the recombinant CD40L protein (49%). This enhanced antitumor activity correlated with cell cycle blockade and increased apoptosis in AdEHCD40L-infected tumor cells. CONCLUSIONS These novel findings, together with the previously known immune-activating features of CD40L, support the potential applicability of AdEHCD40L for experimental treatment of human breast cancer.
Collapse
Affiliation(s)
- Erica M Gomes
- Cancer Immunology Research Laboratory and Department of Pathology, Baylor Sammons Cancer Center, Dallas, TX 75246, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
The targeting expression of the vascular endothelial growth factor gene in endothelial cells regulated by HRE.ppET-1. ACTA ACUST UNITED AC 2008; 51:959-65. [PMID: 18989637 DOI: 10.1007/s11427-008-0116-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Accepted: 08/05/2008] [Indexed: 10/21/2022]
Abstract
The success of gene therapy depends largely on the efficacy of gene delivery vector systems that can deliver genes to target organs or cells selectively and efficiently with minimal toxicity. Here, we show that by using the HRE.ppET-1 regulatory element, we were able to restrict expression of the transgene of vascular endothelial growth factor (VEGF) to endothelial cells exclusively in hypoxic conditions. Eukaryotic expression vectors such as pEGFP-HRE.ppET-1, pcDNA3.1-VEGF+Pa, pcDNA3.1-ppET-1+ EGF+Pa, and pcDNA3.1-HRE.ppET-1+VEGF+Pa were constructed by using a series of nuclear molecule handling methods like PCR, enzyme digestion. The recombinant vectors were transfected into HUVEC cells and HL7702 cells by the lipofectin method. GFP expression was observed with a fluorescence microscope to validate the specificity of expression in endothelial cells under the regulation of HRE.ppET-1 element. Cobalt chloride (final concentration 100 mumol/L) was added to the medium to mimic hypoxia in vitro. After transfection of vectors, the expression of VEGF mRNA was detected by RT-PCR, and the expression of VEGF was detected by Western blotting and ELISA methods under normoxia and hypoxia, respectively. The cell proliferation rate was detected by the MTT test. The expression of GFP revealed that the exterior gene was transcripted effectively in endothelial cells regulated by the HRE.ppET-1 element, while the expression of GFP was very weak in nonendothelial cells. The results of RT-PCR, Western blotting and ELISA showed that VEGF gene expression in the pcDNA3.1-HRE.ppET-1+VEGF+Pa group and in the pcDNA3.1-ppET-1+VEGF+Pa group was higher in hypoxia than it was in normoxia (P<0.05). The MTT test showed that the proliferation rate of HUVEC transfected with HPVA under hypoxia exceeded that of the control group. We conclude that the HRE.ppET-1 element was expressed specifically in endothelial cells, and can increase the expression of VEGF in hypoxia and stimulate proliferation of endothelial cells. Taking advantage of these facts could greatly improve the efficiency of gene therapy. The vector would be valuable for various gene transfer studies targeting endothelial cells.
Collapse
|
35
|
Systemic therapy for cervical cancer with potentially regulatable oncolytic adenoviruses. PLoS One 2008; 3:e2917. [PMID: 18698374 PMCID: PMC2500220 DOI: 10.1371/journal.pone.0002917] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Accepted: 07/07/2008] [Indexed: 12/20/2022] Open
Abstract
Clinical trials have confirmed the safety of selectively oncolytic adenoviruses for treatment of advanced cancers. However, increasingly effective viruses could result in more toxicity and therefore it would be useful if replication could be abrogated if necessary. We analyzed viruses containing the cyclooxygenase-2 (Cox-2) or vascular endothelial growth factor (VEGF) promoter for controlling replication. Anti-inflammatory agents can lower Cox-2 protein levels and therefore we hypothesized that also the promoter might be affected. As Cox-2 modulates expression of VEGF, also the VEGF promoter might be controllable. First, we evaluated the effect of anti-inflammatory agents on promoter activity or adenovirus infectivity in vitro. Further, we analyzed the oncolytic potency of the viruses in vitro and in vivo with and without the reagents. Moreover, the effect of on virus replication was analyzed. We found that RGD-4C or Ad5/3 modified fibers improved the oncolytic potency of the viruses in vitro and in vivo. We found that both promoters could be downregulated with dexamethasone, sodium salicylate, or salicylic acid. Oncolytic efficacy correlated with the promoter activity and in vitro virus production could be abrogated with the substances. In vivo, we saw good therapeutic efficacy of the viruses in a model of intravenous therapy of metastatic cervical cancer, but the inhibitory effect of dexamethasone was not strong enough to provide significant differences in a complex in vivo environment. Our results suggest that anti-inflammatory drugs may affect the replication of adenovirus, which might be relevant in case of replication associated side effects.
Collapse
|
36
|
Han HX, Tang H. Application of adenoviral vector in biotherapy for hepatitis B virus infection. Shijie Huaren Xiaohua Zazhi 2008; 16:1649-1654. [DOI: 10.11569/wcjd.v16.i15.1649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection is a worldwide public health problem. Especially in China about 120 million are estimated to be HBV chronic carriers. For those infected with HBV, there has been no curable treatment. However, biotherapy provides a new clue for future treatment. An appropriate vector is the essential factor in determining efficiency of biotherapy. Owe to its own properties, Adenoviral vector has gained increasing interest in the biotherapy of HBV infection resently. This review focused on the progress in the biotherapy of HBV infection using adenoviral vector.
Collapse
|
37
|
Adenoviral-vector mediated transfer of HBV-targeted ribonuclease can inhibit HBV replication in vivo. Biochem Biophys Res Commun 2008; 371:541-5. [PMID: 18455508 DOI: 10.1016/j.bbrc.2008.04.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 04/18/2008] [Indexed: 12/26/2022]
Abstract
Hepatitis B virus (HBV)-targeted ribonuclease (HBV-TR) is a fused protein of HBV core protein and a ribonuclease, human eosinophil-derived neurotoxin (hEDN). Our previous results showed that HBV-TR could effectively inhibit HBV replication in vitro. To test whether HBV-TR can inhibit HBV replication in vivo, we constructed a recombinant adenoviral vector expressing HBV-TR (Ad-TR) and used it to treat HBV-transgenic mice. Immunohistochemical staining showed that TR was expressed at varied levels in different tissues of Ad-TR-treated mice. Serum HBsAg concentration was decreased by 64.8% for the Ad-TR-treated mice compared with empty adenoviral vector-treated control mice. The amount of HBV-DNA in the livers of the Ad-TR-treated mice was 0.74 x 10(7) copies/mug of genomic DNA while the amount of HBV-DNA in the livers of the empty adenoviral vector-treated control mice was 2.86 x 10(7) copies/mug of genomic DNA. Serum HBV-DNA of Ad-TR-treated mice was also decreased by 71.4% compared with empty adenoviral vector-treated control mice. In addition, for some Ad-TR-treated mice, the expression of HBsAg in the liver cells turned negative. No discernible adverse effects were observed for Ad-TR-treated mice. Taken together, our results indicated that adenovirus mediated transfer of HBV-TR can inhibit HBV replication in vivo.
Collapse
|
38
|
Su C, Na M, Chen J, Wang X, Liu Y, Wang W, Zhang Q, Li L, Long J, Liu X, Wu M, Fan X, Qian Q. Gene-viral cancer therapy using dual-regulated oncolytic adenovirus with antiangiogenesis gene for increased efficacy. Mol Cancer Res 2008; 6:568-75. [PMID: 18344493 DOI: 10.1158/1541-7786.mcr-07-0073] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Conditionally replicative adenovirus (CRAD) represents a promising approach for cancer therapy. Several CRADs controlled by the human telomerase reverse transcriptase promoter have been developed. However, because of their replicative capacity, the importance of cancer specificity for CRADs needs to be further emphasized. In this study, we have developed a novel dual-regulated CRAD, CNHK500-mE, which has its E1a and E1b gene controlled by the human telomerase reverse transcriptase promoter and the hypoxia response element, respectively. It also carries a mouse endostatin expression cassette controlled by the cytomegalovirus promoter. These properties allow for increased cancer cell targeting specificity and decreased adverse side effects. We showed that CNHK500-mE preferentially replicated in cancer cells. Compared with a replication-defective vector carrying the same endostatin expression cassette, CNHK500-mE-mediated transgene expression level was markedly increased via viral replication within cancer cells. In the nasopharyngeal tumor xenograft model, CNHK500-mE injection resulted in antitumor efficacy at day 7 after therapy. Three weeks later, it led to significant inhibition of xenograft tumor growth due to the combined effects of viral oncolytic therapy and antiangiogenesis gene therapy. Pathologic examination showed that most cancer cells were positive for adenoviral capsid protein and for apoptotic terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling in the CNHK500-mE-treated tumor tissues, and the microvessels in these tumor tissues were diminished in quantity and abnormal in morphology. These results suggest that, as a potential cancer therapeutic agent, the CNHK500-mE is endowed with higher specificity to cancer cells and low cytotoxicity to normal cells.
Collapse
Affiliation(s)
- Changqing Su
- Eastern Hepatobiliary Surgical Hospital, Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Polcicova K, Hrabovska Z, Mistrikova J, Tomaskova J, Pastorek J, Pastorekova S, Kopacek J. Up-regulation of Murid herpesvirus 4 ORF50 by hypoxia: possible implication for virus reactivation from latency. Virus Res 2008; 132:257-62. [PMID: 18221814 DOI: 10.1016/j.virusres.2007.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 12/06/2007] [Accepted: 12/06/2007] [Indexed: 10/22/2022]
Abstract
Murid herpesvirus 4 (MuHV-4) is a member of the Gammaherpesvirus subfamily capable to establish a long-lasting latency and induce occasional malignancies. Because MuHV-4 is associated with cancer in a subset of virus-infected mice and because tumor development is often linked with hypoxia, we studied the influence of hypoxia on the biology of this virus. Using immunofluorescence and FACS analysis we detected increased proportion of MuHV-4 positive cells in the latently infected NB-78 cell line exposed to low oxygen conditions compared to normoxic controls. Moreover, the expression of ORF50, a crucial gene responsible for switch from latency to lytic virus replication, was induced upon the exposure of NB-78 cells to hypoxia. Luciferase reporter assays with ORF50 promoter confirmed the hypoxia-dependent induction. Transient co-transfections with hypoxia inducible factors showed that HIF-2alpha is a more potent activator of ORF50 expression than HIF-1alpha. Our results confirm that the MuHV-4 life cycle is influenced by low oxygen concentration.
Collapse
Affiliation(s)
- Katarina Polcicova
- Institute of Virology, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovak Republic
| | | | | | | | | | | | | |
Collapse
|
40
|
Liu J, Harada H, Ogura M, Shibata T, Hiraoka M. Adenovirus-mediated hypoxia-targeting cytosine deaminase gene therapy enhances radiotherapy in tumour xenografts. Br J Cancer 2007; 96:1871-8. [PMID: 17519901 PMCID: PMC2359966 DOI: 10.1038/sj.bjc.6603812] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hypoxia is closely associated with the radioresistance of tumours; therefore, targeting hypoxic areas is very important for cancer therapy. The aim of this study is to establish such a targeting strategy by applying a bacterial cytosine deaminase (BCD)/5-fluorocytosine (5-FC) gene therapy system and to examine whether the strategy enhances the efficacy of radiotherapy in a tumour xenograft. The hypoxia-responsive promoter 5HREp, in which five copies of the hypoxia-response element (HRE) enhance transcription from a cytomegalovirus minimal promoter, was employed to induce the expression of BCD under hypoxic conditions. The adenoviral vector Ad/5HREp-BCD, encoding the gene 5HREp-BCD, robustly induced BCD expression under hypoxic conditions and this led to significant cytotoxicity in combination with 5-FC in vitro. Intratumoral Ad/5HREp-BCD administration resulted in the expression of BCD at the border between normoxic and necrotic regions. The BCD/5-FC gene therapy enhanced the therapeutic effects of both single (12.5 Gy) and fractionated (3 Gy × 5 days) radiotherapy with few side effects and significantly increased tumour growth doubling time by up to 2.4-fold (P<0.01) and 2.5-fold (P<0.05), respectively. All of these results suggest that the present BCD/5-FC gene therapy has the ability to specifically target hypoxic tumour cells and significantly improves the control of tumour growth after radiotherapy.
Collapse
Affiliation(s)
- J Liu
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - H Harada
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Nano-Medicine Merger Education Unit, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- E-mail:
| | - M Ogura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - T Shibata
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - M Hiraoka
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
41
|
Zhao HC, Zhang Q, Yang Y, Lu MQ, Li H, Xu C, Chen GH. p53-expressing conditionally replicative adenovirus CNHK500-p53 against hepatocellular carcinoma in vitro. World J Gastroenterol 2007; 13:683-91. [PMID: 17278190 PMCID: PMC4066000 DOI: 10.3748/wjg.v13.i5.683] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop a conditionally replicative gene-viral vector system called CNHK500-p53, which contains dual promoters within the E1 region, and combines the advantages of oncolytic virus and gene therapies for hepatocellular carcinoma (HCC).
METHODS: CNHK500-p53 was constructed by using human telomerase reverse transcriptase (hTERT) promoter to drive adenovirus E1a gene and hypoxia response element (HRE) promoter to drive adenovirus E1b gene. p53 gene expressing cassette was inserted into the genome of replicative virus. Viral replication experiments, cytopathic effect (CPE) and methyl thiazolyl tetrazolium (MTT) assay were performed to test the selective replication and oncolytic efficacy of CNHK500-p53.
RESULTS: Immunohistochemistry verified that infection with CNHK500-p53 was associated with selective replication of adenovirus and production of p53 protein in telomerase-positive and hypoxia-inducible factor-dependent HCC cells. p53 protein secreted from HepG2, infected with CNHK500-p53 was significantly higher than that infected with nonreplicative adenovirus Ad-p53 in vitro (388 ± 34.6 μg/L vs 76.3 ± 13.17 μg/L). Viral replication experiments showed that replication of CNHK500-p53 and CNHK500 or WtAd5, was much stronger than that of Ad-p53 in tested HCC cell lines. CPE and MTT assay indicated that CNHK500-p53 selectively replicated in and killed HCC cells while leaving normal cells unaffected.
CONCLUSION: A more efficient gene-viral system is developed by combining selective oncolysis with exogenous expression of p53 against HCC cells.
Collapse
Affiliation(s)
- Hong-Chuan Zhao
- Liver Transplantation Centre, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, China
| | | | | | | | | | | | | |
Collapse
|
42
|
Zhang Q, Chen G, Peng L, Wang X, Yang Y, Liu C, Shi W, Su C, Wu H, Liu X, Wu M, Qian Q. Increased safety with preserved antitumoral efficacy on hepatocellular carcinoma with dual-regulated oncolytic adenovirus. Clin Cancer Res 2007; 12:6523-31. [PMID: 17085667 DOI: 10.1158/1078-0432.ccr-06-1491] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A dual-regulated adenovirus variant CNHK500, in which human telomerase reverse transcriptase promoter drove the adenovirus 5 (Ad5) E1a gene and hypoxia-response promoter controlled the E1b gene, was engineered. This virus has broad anticancer spectrum and higher specificity compared with mono-regulated adenovirus CNHK300. The objective of the current study is to show its antitumor selectivity and therapeutic potential. EXPERIMENTAL DESIGN The antitumor specificity of human telomerase reverse transcriptase and hypoxia response promoters was evaluated in a panel of tumor and normal cells. Under the control of these promoters, the tumor-selective expression of E1a and E1b genes was evaluated. Further in vitro antitumor specificity and potency of this virus were characterized by viral replication and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Subsequently, hepatocellular carcinoma xenografts were established to evaluate CNHK500 antitumor efficacy in vivo by different routes of virus administration and different dosages. RESULTS Human telomerase reverse transcriptase and hypoxia response promoters were activated in a tumor-selective manner or under hypoxia treatment in a broad panel of cells. Selective adenoviral early gene expression, efficient viral replication, and oncolysis were observed in all tested cancer cells with more attenuated replication capacity in normal cells. Significant regression of hepatocellular carcinoma xenografts and prolonged survival were observed by either i.t. or i.v. administration. CONCLUSIONS CNHK500 greatly reduced side effects in normal cells via dual control of adenoviral essential genes while still preserving potent antitumor efficacy on broad-spectrum cancer cells in vitro and in vivo. It can be used as a powerful therapeutic agent not only for liver cancers but also for other solid tumors.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Hypoxia is an integral characteristic of the tumor microenvironment, primarily due to the microvascular defects that accompany the accelerated neoplastic growth. The presence of tumor hypoxic areas correlates with negative outcome after radiotherapy, chemotherapy, and surgery, as hypoxia not only provides an environment directly facilitating chemo- and radio-resistance, but also encourages the evolution of phenotypic changes inducing permanent resistance to treatment and metastatic spread. Therefore, successful treatment of hypoxic cells has the potential to not only improve local control but also impact overall patient survival. Specific and selective targeting of hypoxic tumor areas can be achieved at all three steps of a gene therapy treatment: delivery of the therapeutic gene to the tumor, regulation of gene expression, and therapeutic efficacy. In this review the latest developments and innovations in hypoxia-targeted gene therapy are discussed. In particular, approaches such as hypoxia-conditionally replicating viruses, cellular vehicles, and gene therapy means to disrupt the hypoxia-inducible factor (HIF) signaling are outlined.
Collapse
Affiliation(s)
- Olga Greco
- Tumour Microcirculation Group, University of Sheffield, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | |
Collapse
|
44
|
Matzow T, Cowen RL, Williams KJ, Telfer BA, Flint PJ, Southgate TD, Saunders MP. Hypoxia-targeted over-expression of carboxylesterase as a means of increasing tumour sensitivity to irinotecan (CPT-11). J Gene Med 2007; 9:244-52. [PMID: 17397102 DOI: 10.1002/jgm.1016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The induced expression of carboxylesterase (CE) enzymes, which convert the prodrug irinotecan (CPT-11) into its active cytotoxic metabolite SN-38, constitutes a promising strategy for cancer gene therapy. By incorporating hypoxia-responsive elements (HREs) in conjunction with the transgene, expression can be targeted specifically to hypoxic tissues (such as solid tumours), expressing the hypoxia-inducible factor 1 (HIF-1). We have constructed a recombinant adenoviral vector, AdHRE-rCE, encoding the cDNA for the highly efficient rabbit liver CE (rCE), under the control of a HRE derived from the human phosphoglycerate kinase 1 (PGK-1) gene in conjunction with a minimal SV40 promoter. In vitro, HT1080 fibrosarcoma and SW480 colon carcinoma cells demonstrated an approximately 10-fold hypoxia-dependent induction in CE expression following pre-infection with AdHRE-rCE, which led to a15-30-fold increased sensitivity to CPT-11. Furthermore, in vivo, SW480 tumour xenografts infected with AdHRE-rCE demonstrated a 2-fold decrease in tumour doubling time, when combined with 7 days of CPT-11 treatment, in comparison to mock-infected controls, with rCE expression shown to be limited to hypoxic regions only. As the cytotoxicity of CPT-11 is reduced under hypoxic conditions, over-expression of a highly efficient CE such as rCE under hypoxia control within these hypoxic cells could reverse this effect and, therefore, form the basis for future clinical treatment strategies.
Collapse
Affiliation(s)
- Torkjel Matzow
- Paterson Institute for Cancer Research, Christie Hospital NHS Trust, and School of Pharmacy and Pharmaceutical Sciences, University of Manchester, UK
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Human solid tumors are invariably less well-oxygenated than the normal tissues from which they arose. This so-called tumor hypoxia leads to resistance to radiotherapy and anticancer chemotherapy as well as predisposing for increased tumor metastases. In this chapter, we examine the resistance of tumors to radiotherapy produced by hypoxia and, in particular, address the question of whether this resistance is the result of the physicochemical free radical mechanism that produces resistance to radiation killing of cells in vitro. We conclude that a major part of the resistance, though perhaps not all, is the result of the physicochemical free radical mechanism of the oxygen effect in sensitizing cells to ionizing radiation. However, in modeling studies used to evaluate the effect of fractionated irradiation on tumor response, it is essential to consider the fact that the tumor cells are at a wide range of oxygen concentrations, not just at the extremes of oxygenated and hypoxic. Prolonged hypoxia of the tumor tissue also leads to necrosis, and necrotic regions are also characteristic of solid tumors. These two characteristics--hypoxia and necrosis--represent clear differences between tumors and normal tissues and are potentially exploitable in cancer treatment. We discuss strategies for exploiting these differences. One such strategy is to use drugs that are toxic only under hypoxic conditions. The second strategy is to take advantage of the selective induction under hypoxia of the hypoxia-inducible factor (HIF)-1. Gene therapy strategies based on this strategy are in development. Finally, tumor hypoxia can be exploited using live obligate anaerobes that have been genetically engineered to express enzymes that can activate nontoxic prodrugs into toxic chemotherapeutic agents.
Collapse
Affiliation(s)
- J Martin Brown
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
46
|
Abstract
Tumor-associated vasculature is a relatively accessible component of solid cancers that is essential for tumor survival and growth, providing a vulnerable target for cancer gene therapy administered by intravenous injection. Several features of tumor-associated vasculature are different from normal vasculature, including overexpression of receptors for angiogenic growth factors, markers of vasculogenesis, upregulation of coagulation cascades, aberrant expression of adhesion molecules and molecular consequences of hypoxia. Many of these differences provide candidate targets for tumor-selective 'transductional targeting' of genetically- or chemically modified vectors and upregulated gene expression can also enable 'transcriptional targeting', regulating tumor endothelia-selective expression of transgenes following nonspecific gene delivery. Tumor vasculature also represents an important site of therapeutic action by the secreted products of antiangiogenic gene therapies that are expressed in non-endothelial cells. In this review we assess the challenges faced and the vectors that may be suitable for gene delivery to exploit these targets. We also overview some of the strategies that have been developed to date and highlight the most promising areas of research.
Collapse
Affiliation(s)
- M Bazan-Peregrino
- Department of Clinical Pharmacology, University of Oxford, Radcliffe Infirmary, Woodstock Road, Oxford, UK
| | | | | |
Collapse
|
47
|
Lee M, Choi D, Choi MJ, Jeong JH, Kim WJ, Oh S, Kim YH, Bull DA, Kim SW. Hypoxia-inducible gene expression system using the erythropoietin enhancer and 3′-untranslated region for the VEGF gene therapy. J Control Release 2006; 115:113-9. [PMID: 16962197 DOI: 10.1016/j.jconrel.2006.07.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 07/06/2006] [Accepted: 07/13/2006] [Indexed: 01/10/2023]
Abstract
Gene therapy with the vascular endothelial growth factor (VEGF) gene is a potential treatment for many disorders or injuries with ischemia. However, unregulated expression of VEGF may induce pathological angiogenesis, promoting tumor growth, diabetic proliferative retinopathy and rupture of atherosclerotic plaque. Therefore, the effective regulation of the gene expression is one of the requirements for the VEGF gene therapy. In this research, we evaluated the hypoxia-inducible gene expression system with the erythropoietin (Epo) enhancer and the Epo 3'-untranslated region (UTR). The luciferase plasmids were constructed with the Epo enhancer (pEpo-SV-Luc), the Epo 3'-UTR (pSV-Luc-EpoUTR) or both (pEpo-SV-Luc-EpoUTR). The polyethylenimine/plasmid complexes were transfected to 293 or A7R5 cells and the cells were incubated under normoxia or hypoxia. The results showed that the Epo enhancer or Epo 3'-UTR increased the target gene expression under hypoxia. pEpo-SV-Luc-EpoUTR showed the highest luciferase expression. The VEGF expression plasmid with the Epo enhancer and 3'-UTR was also constructed. The VEGF expression by pEpo-SV-VEGF-EpoUTR showed the highest specificity of the gene expression in the hypoxic cells. The results suggest that the VEGF plasmid with the Epo enhancer and the Epo 3'-UTR may be useful for gene therapy for ischemic diseases.
Collapse
Affiliation(s)
- Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Riddick DS, Lee C, Ramji S, Chinje EC, Cowen RL, Williams KJ, Patterson AV, Stratford IJ, Morrow CS, Townsend AJ, Jounaidi Y, Chen CS, Su T, Lu H, Schwartz PS, Waxman DJ. Cancer chemotherapy and drug metabolism. Drug Metab Dispos 2006; 33:1083-96. [PMID: 16049130 DOI: 10.1124/dmd.105.004374] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Drug-metabolizing enzymes and drug transporters are key determinants of the pharmacokinetics and pharmacodynamics of many antineoplastic agents. Metabolism and transport influence the cytotoxic effects of antineoplastic agents in target tumor cells and normal host tissues. This article summarizes several state-of-the-art approaches to enhancing the effectiveness and safety of cancer therapy based on recent developments in our understanding of antineoplastic drug metabolism and transport. Advances in four interrelated research areas presented at a recent symposium sponsored by the Division for Drug Metabolism of the American Society for Pharmacology and Experimental Therapeutics (Experimental Biology 2004; Washington D.C., April 17-21, 2004) are discussed: 1) interactions of anthracyclines with drug-metabolizing enzymes; 2) use of hypoxia-selective gene-directed enzyme prodrug therapy (GDEPT) in combination with bioreductive prodrugs; 3) synergy between glutathione conjugation and conjugate efflux in conferring resistance to electrophilic toxins; and 4) use of cytochromes P450 as prodrug-activating enzymes in GDEPT strategies. A clear theme emerged from this symposium: drug metabolism and transport processes can be modulated and exploited in ways that may offer distinct therapeutic advantages in the management of patients with cancer.
Collapse
Affiliation(s)
- David S Riddick
- Department of Pharmacology, Medical Sciences Building, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chen HJ, Huang ZH, Su GQ, Wu AG, Yu JL. Targeted killing effect of lentivirus-mediated CD/TK suicide genes on colorectal carcinoma cells. Shijie Huaren Xiaohua Zazhi 2006; 14:1681-1687. [DOI: 10.11569/wcjd.v14.i17.1681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the killing effect of lentivirus-mediated double suicide genes under the regulation of kinase domain-containing receptor (KDR) promoter targeted on colorectal carcinoma cells.
METHODS: 293FT packaging cells were transfected by the constructed plasmids pLenti6/V5-D-KDR-CDglyTK and pLenti6/V5-D-GFP. After blasticidin selection and cell cloning, the infectious viruses were generated. Then SW620 (with KDR expression) and LS174T cells (without KDR expression) were transfected with the obtained viruses by lipofectamin 2000. The transfection efficacy was evaluated by the fluorecence microscopy. The expression of CDglyTK was detected by reverse transcription-polymerase chain reaction (RT-PCR). After treatment with 5-FC and GCV, the killing effects and bystander effect of CD/TK suicide genes on the two kinds of cell lines were assessed.
RESULTS: The transfection efficacy was not significantly different between SW620 and LS174T cells, and elevated with the increase of virus titer. RT-PCR demonstrated that CDglyTK was expressed only in SW620 cells infected by pLenti6/V5-D-KDR-CDglyTK but not in LS147T cells. For the transfected SW620 cells, the survival rate was 32.34% ± 2.42% or 30.56% ± 2.14% when GCV (100 mg/L) or 5-FC (2.0 g/L) was used alone, respectively, and it was 5.36% ± 1.55% when GCV and 5-FC were used in combination. For the transfected LS174T cells, the survival rate was 95.48% ± 1.70% when GCV and 5-FC were used in combination. SW620 cells had a higher sensitivity to the prodrugs than LS174T cells did (P < 0.001), and the effects of double suicide genes were markedly stranger than that of either single gene (P < 0.001). Considerable bystander effect was also observed. When the infected cells covered a percentage of 40%, the survival rate of SW620 cells was 11.42% ± 2.66%, while that of LS174T cells was 99.54% ± 2.61% after treatment with GCV and 5-FC. There was significant difference between the two kinds of cells (P < 0.001).
CONCLUSION: Lentivirus-mediated CD/TK suicide genes driven by KDR promoter have specific killing effect on colorectal carcinoma cells with KDR expression.
Collapse
|
50
|
Lee JY, Lee YS, Kim JM, Kim KL, Lee JS, Jang HS, Shin IS, Suh W, Jeon ES, Byun J, Kim DK. A novel chimeric promoter that is highly responsive to hypoxia and metals. Gene Ther 2006; 13:857-68. [PMID: 16467859 DOI: 10.1038/sj.gt.3302728] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Revised: 12/15/2005] [Accepted: 01/01/2006] [Indexed: 01/16/2023]
Abstract
To develop a potent hypoxia-inducible promoter, we evaluated the usefulness of chimeric combinations of the (Egr-1)-binding site (EBS) from the Egr-1 gene, the metal-response element (MRE) from the metallothionein gene, and the hypoxia-response element (HRE) from the phosphoglycerate kinase 1 gene. In transient transfection assays, combining three copies of HRE (3 x HRE) with either EBS or MRE significantly increased hypoxia responsiveness. When a three-enhancer combination was tested, the EBS-MRE-3 x HRE (E-M-H) gave a hypoxia induction ratio of 69. The expression induced from E-M-H-pGL3 was 2.4-fold higher than that induced from H-pGL3 and even surpassed the expression from a human cytomegalovirus promoter-driven vector. The high inducibility of E-M-H was confirmed by validation studies in different cells and by expressing other cDNAs. Gel shift assays together with functional overexpression studies suggested that increased levels of hypoxia-inducible factor 1alpha, metal transcription factor-1 and Egr-1 may be associated with the high inducibility of the E-M-H chimeric promoter. E-M-H was also induced by hypoxia mimetics such as Co2+ and deferoxamine (DFX) and by hydrogen peroxide. Gene expression from the E-M-H was reversible as shown by the reduced expression of the transgene upon removal of inducers such as hypoxia and DFX. In vivo evaluation of the E-M-H in ischemic muscle revealed that erythropoietin secretion and luciferase and LacZ expression were significantly higher in the E-M-H group than in a control or H group. With its high induction capacity and versatile means of modulation, this novel chimeric promoter should find wide application in the treatment of ischemic diseases and cancer.
Collapse
Affiliation(s)
- J-Y Lee
- Department of Medicine, Cardiac and Vascular Center, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Kangnam-ku, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|