1
|
Affiliation(s)
- Frank D. Cirisano
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, California
| | - Beth Y. Karlan
- Cedars-Sinai Medical Center, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, 8700 Beverly Boulevard, #1740, Los Angeles, CA 90048; Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, California
| |
Collapse
|
2
|
Lu X, Jia F, Tan X, Wang D, Cao X, Zheng J, Zhang K. Effective Antisense Gene Regulation via Noncationic, Polyethylene Glycol Brushes. J Am Chem Soc 2016; 138:9097-100. [PMID: 27420413 DOI: 10.1021/jacs.6b05787] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Negatively charged nucleic acids are often complexed with polycationic transfection agents before delivery. Herein, we demonstrate that a noncationic, biocompatible polymer, polyethylene glycol, can be used as a transfection vector by forming a brush polymer-DNA conjugate. The brush architecture provides embedded DNA strands with enhanced nuclease stability and improved cell uptake. Because of the biologically benign nature of the polymer component, no cytotoxicity was observed. This approach has the potential to address several long-lasting challenges in oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Xueguang Lu
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| | - Fei Jia
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| | - Xuyu Tan
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| | - Dali Wang
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| | - Xueyan Cao
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| | - Jiamin Zheng
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| | - Ke Zhang
- Department of Chemistry and Chemical Biology, Northeastern University , Boston, Massachusetts 02115, United States
| |
Collapse
|
3
|
pcDNA3.1(-)-mediated ribozyme targeting of HER-2 suppresses breast cancer tumor growth. Mol Biol Rep 2009; 37:1597-604. [PMID: 19444644 DOI: 10.1007/s11033-009-9569-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Accepted: 05/01/2009] [Indexed: 01/11/2023]
Abstract
The HER-2 proto-oncogene (also called c-erbB-2/neu) encodes the protein, p185, which is closely related to the growth and metastasis of adenocarcinoma, and is overexpressed in 25-30% of human breast cancers. In this study, we attempt to reverse the malignant phenotype of the breast cancer cell line, MCF-7, using a HER-2-specific hammerhead ribozyme. Two anti-HER-2 hammerhead ribozymes, RZ1 and RZ2, were synthesized, inserted separately into the nonviral eukaryotic expression vector, pcDNA3.1(-), and transfected into MCF-7 cells. Analyses showed that the HER-2 mRNA and p185, as well as oncogene k-ras were down-regulated remarkably in the ribozyme-transfected cells, while the onco-suppressor gene, p53, was up-regulated. Furthermore, the tumorigenicity of the RZ1-stably transfected MCF-7 cells was decreased dramatically in nude mice. These results demonstrate that the use of anti-HER-2 ribozymes may be a beneficial strategy for gene therapy of breast cancer.
Collapse
|
4
|
Abstract
This review briefly outlines the importance of molecular imaging, particularly imaging of endogenous gene expression for noninvasive genetic analysis of radiographic masses. The concept of antisense imaging agents and the advantages and challenges in the development of hybridization probes for in vivo imaging are described. An overview of the investigations on oncogene expression imaging is given. Finally, the need for further improvement in antisense-based imaging agents and directions to improve oncogene mRNA targeting is stated.
Collapse
Affiliation(s)
- Archana Mukherjee
- Postdoctoral Research Fellow, Department of Radiology, Thomas Jefferson University, 361 JAH, 1020, Locust Street, Philadelphia, PA 19107, USA, , Tel: 215-503-7879, Fax: 215-923-9245
| | - Eric Wickstrom
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233S, 10th street, Suite 219 Philadelphia, PA 19107, USA, , Tel: 215-955-4578, Fax: 215-955-4580
| | - Mathew L. Thakur
- Director, Laboratories of Radiopharmaceutical Research and Molecular Imaging, Department of Radiology, Thomas Jefferson University, Kimmel Cancer Centre. Suite 359 JAH, 1020, Locust Street, Philadelphia, PA 19107, USA, , Tel: 215-503-7874, Fax: 215-923-9245
| |
Collapse
|
5
|
Park JW, Neve RM, Szollosi J, Benz CC. Unraveling the biologic and clinical complexities of HER2. Clin Breast Cancer 2008; 8:392-401. [PMID: 18952552 DOI: 10.3816/cbc.2008.n.047] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It has been over 20 years since the discovery of the human epidermal growth factor receptor 2 (HER2), a tyrosine kinase receptor that is a potent oncoprotein in breast and other cancers and has become an opportune target for therapy. HER2 plays a critical role in normal development, forming homodimers or heterodimers with other HER family members and triggering downstream signaling cascades controlling proliferation, cell survival, and apoptosis. However, amplification of the HER2 gene in cancer cells results in overexpression of HER2 receptors on the cell surface, leading to excessive and dysregulated signaling. HER2-driven signaling also upregulates transcription factors that act on the HER2 promoter, increasing its expression. In breast cancer, HER2 is gene amplified in 20%-25% of primary tumors and is associated with a more aggressive phenotype and poorer prognosis. The key role HER2 plays in tumorigenesis makes it an ideal target for therapy. Trastuzumab, a monoclonal antibody against HER2, inhibits downstream signaling and has proven to be effective against HER2-overexpressing metastatic breast cancer both as a single agent and in combination with chemotherapy. Seminal clinical trial data also show that the use of adjuvant trastuzumab in combination with chemotherapy or as a single agent after chemotherapy significantly increases disease-free and overall survival. Lapatinib, a dual tyrosine kinase inhibitor against HER1 and HER2, has been approved in combination with capecitabine for HER2-overexpressing advanced or metastatic breast cancer, which has progressed following previous anthracycline, taxane, and trastuzumab therapy. Other HER2-targeting strategies are also under active investigation.
Collapse
Affiliation(s)
- John W Park
- Comprehensive Cancer Center, University of California, San Francisco, CA 94115-1710, USA.
| | | | | | | |
Collapse
|
6
|
Tian X, Chakrabarti A, Amirkhanov NV, Aruva MR, Zhang K, Mathew B, Cardi C, Qin W, Sauter ER, Thakur ML, Wickstrom E. External imaging of CCND1, MYC, and KRAS oncogene mRNAs with tumor-targeted radionuclide-PNA-peptide chimeras. Ann N Y Acad Sci 2006; 1059:106-44. [PMID: 16382049 DOI: 10.1196/annals.1339.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In 2005, breast cancer will kill approximately 40,410 women in the U.S., and pancreatic cancer will kill approximately 31,800 men and women in the U.S. Clinical examination and mammography, the currently accepted breast cancer screening methods, miss almost half of breast cancers in women younger than 40 years, approximately one-quarter of cancers in women aged 40-49 years, and one-fifth of cancers in women over 50 years old. Pancreatic cancer progresses rapidly, with only 1% of patients surviving more than 5 years after diagnosis. However, if the disease is diagnosed when it is localized, the 5-year survival is approximately 20%. It would be beneficial to detect breast cancer and pancreatic cancer at the earliest possible stage, when multimodal therapy with surgery, radiotherapy, and chemotherapy have the greatest chance of prolonging survival. Human estrogen receptor-positive breast cancer cells typically display elevated levels of Myc protein due to overexpression of MYC mRNA, elevated cyclin D1 protein due to overexpression of CCND1 mRNA, and elevated insulin-like growth factor 1 receptor (IGF1R) due to overexpression of IGF1R mRNA. We hypothesized that scintigraphic detection of MYC or CCND1 peptide nucleic acid (PNA) probes with an IGF1 peptide loop on the C-terminus, and a Tc-99m-chelator peptide on the N-terminus, could measure levels of MYC or CCND1 mRNA noninvasively in human IGF1R-overexpressing MCF7 breast cancer xenografts in immunocompromised mice. Similarly, human pancreatic cancer cells typically display elevated levels of KRAS mRNA and elevated IGF1R. Hence, we also hypothesized that a KRAS Tc-99m-chelator PNA-peptide probe could detect overexpression of KRAS mRNA in pancreatic cancer xenografts by scintigraphic imaging, or by positron emission tomography (PET) with a KRAS Cu-64-chelator PNA-peptide. Human MCF7 breast cancer xenografts in immunocompromised mice were imaged scintigraphically 4-24 h after tail-vein administration of MYC or CCND1 Tc-99m-chelator PNA-peptides, but not after administration of mismatch controls. Similarly, human Panc-1 pancreatic cancer cells xenografts were imaged scintigraphically 4 and 24 h after tail-vein administration of a KRAS Tc-99m-chelator PNA-peptide, and AsPC1 xenografts were imaged by PET 4 and 24 h after tail-vein adminstration of a KRAS Cu-64-chelator PNA-peptide. The radioprobes distributed normally to the kidneys, livers, tumors, and other tissues. External molecular imaging of oncogene mRNAs in solid tumors with radiolabel-PNA-peptide chimeras might in the future provide additional genetic characterization of pre-invasive and invasive breast cancers.
Collapse
Affiliation(s)
- Xiaobing Tian
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 S. 10th Street, Suite 219, Philadelphia, PA 19107-5541, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Oh BK, Nam JM, Lee SW, Mirkin CA. A fluorophore-based bio-barcode amplification assay for proteins. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2006; 2:103-8. [PMID: 17193564 DOI: 10.1002/smll.200500260] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Byung-Keun Oh
- Department of Chemistry and Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | | | | | | |
Collapse
|
8
|
Wickstrom E, Thakur ML, Sauter ER. Receptor-specific targeting with complementary peptide nucleic acids conjugated to peptide analogs and radionuclides. Int J Pept Res Ther 2005. [DOI: 10.1007/s10989-005-4917-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
9
|
Rohmann A, Lochmann D, Weyermann J, Bertling WM, Dingermann T, Zimmer A. Rapid screening method for antisense oligonucleotides against human growth factor receptor p185(erbB-2). Oligonucleotides 2004; 14:1-9. [PMID: 15104891 DOI: 10.1089/154545704322988003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The aim of this study was the development of an indirect cell proliferation assay as screening tool for antisense oligonucleotides. Unmodified and phosphorothioate-modified oligonucleotides with different amounts of sulfur in the DNA backbone were examined for biologic activity. The human growth factor receptor p185(erbB-2) was chosen as cellular target. High-level expression of this protein can be related to an early event in tumor development and cell proliferation. We correlated the expression of p185(erbB-2) with the cell proliferation of BT-474. Additionally a control cell line (MCF-7) with very low p185(erbB-2) expression was cultivated. Antisense oligonucleotides were transfected as a liposome formulation (Lipofectin), GIBCO-BRL, Eggenstein, Germany). Cell count was correlated with a total protein quantification assay (BCA method). Stability against nuclease digestion was determined with a DNase I assay. Sequence-specific antisense effects on the p185(erbB-2) protein level were determined by Western blot. An antisense phosphorothioate oligonucleotide was identified to inhibit the cell proliferation in comparison to a random control and a negative control oligonucleotide sequence. The comparison of fully thioated, partly thioated, and unmodified oligonucleotides verified the correlation between the enzymatic stability and the biologic activity of the different modifications. Using the unstable oligonucleotides, more treatments were necessary to achieve an antiproliferative effect. In our study, the indirect proliferation assay was found to be a reliable and potent tool for an antisense oligonucleotide screening by targeting the p185(erbB-2) protein.
Collapse
Affiliation(s)
- Anke Rohmann
- Institute for Pharmaceutical Biology and Institute for Pharmaceutical Technology, Johann Wolfgang Goethe-University, 60439 Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
10
|
Tian X, Aruva MR, Rao PS, Qin W, Read P, Sauter ER, Thakur ML, Wickstrom E. Imaging oncogene expression. Ann N Y Acad Sci 2004; 1002:165-88. [PMID: 14751834 DOI: 10.1196/annals.1281.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In 2003, approximately 39,800 women in the US will die from breast cancer. Mammography and physical examination miss up to 40% of early breast cancers. Moreover, if an abnormality is found, an invasive diagnostic procedure must still be performed to determine if the breast contains atypia or cancer, even though approximately 85% of abnormalities are benign. Scintigraphic imaging of gene expression in vivo by noninvasive means could direct physicians to appropriate targets for intervention at the onset of disease and thereby significantly impact patient management. Until now, no method has been available to image specific overexpressed oncogene mRNAs in vivo by scintigraphic imaging. We hypothesize that gamma-emitting Tc-99m-PNA-peptides can be taken up by human ER+ and ER- breast cancer xenografts, hybridize to complementary mRNA targets in those cells, and concentrate sufficiently in tumor tissue to allow noninvasive imaging of oncogene overexpression. To prepare the probes, peptide analogs of insulin-like growth factor 1 (IGF1) were extended from a solid support by Fmoc coupling. Peptide nucleic acid (PNA) dodecamers antisense to CCND1 and MYC mRNAs were then extended from the N-terminus of IGF1, followed by a chelator peptide, using Fmoc coupling for all residues. The cysteine thiols were cyclized on the solid support, either before or after PNA extension. This simplified synthetic approach allows preparation of a variety of multipeptide disulfide-bridged PNA chimeras. A chelating peptide-PNA chimera antisense to MYC mRNA was then labeled efficiently with Tc-99m, yielding a single product. Tissue distribution studies of antisense and mismatch chimeras at 4 h and 24 h after administration displayed modest accumulation in the liver and kidneys, with appreciable levels in tumors. This result enables testing of Tc-99m-peptide-PNA probes to image gene expression in tumors.
Collapse
Affiliation(s)
- Xiaobing Tian
- Department of Biochemistry Molecular Pharmacology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Wickstrom E, Thakur ML, Sauter ER. Receptor-specific targeting with complementary peptide nucleic acids conjugated to peptide analogs and radionuclides. Int J Pept Res Ther 2003. [DOI: 10.1007/bf02484559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
12
|
Shadidi M, Sioud M. Identification of novel carrier peptides for the specific delivery of therapeutics into cancer cells. FASEB J 2003; 17:256-8. [PMID: 12490548 DOI: 10.1096/fj.02-0280fje] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cancer therapy is currently limited by the difficulty of achieving efficient delivery into target cells. To investigate whether therapeutics can be delivered specifically to cancer cells, we have explored the possibility of selecting small peptides that bind specifically, or preferentially, to breast cancer cell lines. By using random peptide phage libraries and an experimental approach that allows the selection of internalized peptides, cell-specific binding peptides have been identified. The peptides define a major core motif (LTVXPWY) that was not found in negative phages. Phage displaying LTVSPWY peptide sequence exhibited a specific binding to breast cancer cells. None of the selected peptides bound to human primary cells from different tissue origin (e.g., epithelial, endothelial, hematopoetic). The potential of the selected peptides to mediate cellular internalization in the context of phages and recombinant GFP-peptide fusions was demonstrated. By linking the LTVSPWY peptide to an antisense phosphorothioate oligonucleotide against the ErbB2 receptor, specific delivery to cancer cells was achieved. In contrast to free antisense, the peptide-antisense conjugates inhibited ErbB2 gene expression. Thus, efficient delivery of antisense oligonucleotides can be achieved by coupling them to cancer cell-specific peptides, identified by a method that did not require any knowledge about their corresponding receptors.
Collapse
Affiliation(s)
- Mohsen Shadidi
- Institute for Cancer Research, Department of Immunology, Molecular Medicine Group, The Norwegian Radium Hospital, Montebello, N-0310 Oslo, Norway
| | | |
Collapse
|
13
|
|
14
|
Affiliation(s)
- Funda Meric
- Department of Surgical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, 77030, USA
| | | | | | | |
Collapse
|
15
|
Rait AS, Pirollo KF, Rait V, Krygier JE, Xiang L, Chang EH. Inhibitory effects of the combination of HER-2 antisense oligonucleotide and chemotherapeutic agents used for the treatment of human breast cancer. Cancer Gene Ther 2001; 8:728-39. [PMID: 11687896 DOI: 10.1038/sj.cgt.7700359] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2001] [Indexed: 11/09/2022]
Abstract
Poor response to chemotherapy in patients with breast cancer is often associated with overexpression of HER-2/neu. Interference with HER-2 mRNA translation by means of antisense oligonucleotides might improve the efficacy of chemotherapy. To test this hypothesis, eight breast cancer cell lines and a normal human fibroblast cell line were examined for their level of HER-2 expression, their sensitivity to phosphorothioate antisense oligonucleotides (AS HER-2 ODN), and to various chemotherapeutic agents, and the combination of the two. No correlation was found between the intrinsic HER-2 level and either the sensitivity to a particular chemotherapeutic agent alone, or the amount of growth inhibition observed with a specific AS HER-2 ODN concentration. Although sequence specificity and extent of AS HER-2 ODN inhibition of HER-2 synthesis were somewhat higher in the HER-2 overexpressing MDA-MB-453 and SK-BR-3 cells, we found that antisense treatment significantly sensitized all of the breast cancer cells, even MDA-MB-231 and MDA-MB-435 cells, with approximately basal levels of HER-2, to various chemotherapeutic agents. In addition, the combination of AS HER-2 ODN and taxol was shown to synergistically induce apoptosis in MDA-MB-435. These results demonstrate that overexpression of HER-2 would not be a prerequisite for the effective use of AS HER-2 ODN as a combination treatment modality for breast cancer and suggest that the use of AS HER-2 ODN, as part of a combination treatment modality, need not be limited to breast tumors that display elevated levels of HER-2.
Collapse
Affiliation(s)
- A S Rait
- Departments of Oncology and Otolaryngology, Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Pancreatic cancer is the fifth leading cause of cancer deaths in the United States with little or no impact from conventional treatment options. Significant advances in understanding basic immunology have renewed interest in using immunotherapy to treat pancreatic cancer. Cancer immunotherapy, including humanized MAbs, cytokines, and potent vaccine strategies, has been successful in animal models and is being evaluated in clinical trials. Gene therapy is also being explored using methods to inactivate oncogenes, replace defective tumor suppressor genes, confer enhanced chemosensitivity to tumor cells, and increase immunogenicity of tumor cells. Angiogenesis, an essential step in the growth and metastasis of pancreatic cancer, has been targeted by many antiangiogenic agents. Several clinical trials have been initiated to evaluate the role of these innovative strategies in patients with pancreatic cancer with increasingly sophisticated correlative studies to learn more about the mechanisms of tumor rejection with these agents. The rapid translation of basic science discoveries to clinical trials should result in the development of new effective treatments for patients with pancreatic cancer.
Collapse
Affiliation(s)
- S M Lieberman
- Department of Surgery, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | |
Collapse
|
17
|
Abstract
Oligonucleotides have shown an ability to target specific oncogene transcripts and inhibit their expression in cells, but the degree to which sustained treatment can suppress the levels of an oncogenic protein enough to benefit a patient remains to be determined. This question has been studied in several ways. First, the relationship of antisense DNA inhibition to the predicted secondary structure of human H-RAS oncogene mRNA was examined in transformed mouse cells that form solid tumors. Inhibition of H-Ras expression was sequence-specific, dose-dependent, and correlated with inhibition of focus formation. The efficacy of the first intron antisense sequence in reducing H-Ras expression was greater than that of the initiation codon target. Second, H-RAS transformed solid tumor cells were pretreated in vitro with normal oligonucleotides, after which tumor growth from the treated cells was tested in nude mice. The three days of treatment with the first intron antisense DNA reduced H-Ras cellular levels by more than 90% whereas a nonspecific control DNA reduced H-Ras levels by approx 20%. Tumor growth of cells treated with H-RAS antisense oligonucleotide was significantly reduced for up to 14 d following the end of treatment and implantation into the mice, whereas the nonspecific control DNA had no significant effect. Third, H-RAS transformed bladder cancer cells were implanted into nude mice, after which the mice were treated for 31 d with oligonucleotide phosphorothioates. Tumor growth in mice treated with H-RAS 12th codon antisense oligonucleotide was reduced by about 80% throughout the treatment period, reiterating the sustained effect seen in pretreated tumor cells. However, the scrambled phosphorothioate control inhibited tumor growth by about 60%, illustrating some nonspecific inhibition. Fourth, K-RAS transformed pancreatic cancer cells were treated in culture and in nude mice. Inhibition of K-Ras expression with a phosphorothioate oligonucleotide directed against a 5'-UTR sequence was sequence-specific and dose-dependent. K-RAS transformed pancreatic cancer cells were implanted into nude mice, after which the mice were treated for 14 d with oligonucleotide phosphorothioates. Tumor growth in mice treated with K-RAS 5'-UTR antisense oligonucleotide was reduced by about 50% throughout the treatment period, reiterating the sustained effect seen with H-RAS transformed cells. In this case, the sense phosphorothioate control did not inhibit tumor growth, demonstrating that nonspecific inhibition is not a characteristic of all phosphorothioate sequences. The next logical steps include testing oligonucleotide efficacy against other tumor types, toxicological testing in higher species, and clinical trials in human subjects.
Collapse
MESH Headings
- 3T3 Cells
- 5' Untranslated Regions
- Animals
- Cell Division
- Cell Line, Transformed
- Codon
- Dose-Response Relationship, Drug
- Female
- Genes, ras/genetics
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mutation
- Neoplasm Transplantation
- Neoplasms, Experimental/therapy
- Nucleic Acid Conformation
- Oligonucleotides, Antisense/pharmacokinetics
- Oligonucleotides, Antisense/therapeutic use
- RNA, Messenger/metabolism
- Thermodynamics
- Time Factors
- Tumor Cells, Cultured
- ras Proteins/biosynthesis
Collapse
Affiliation(s)
- E Wickstrom
- Laboratory of Nucleic Acid Therapeutics, Department of Microbiology and Immunology, Kimmel Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Germann MW, Aramini JM, Kalisch BW, van de Sande JH. Structural, dynamic, and enzymatic properties of mixed alpha/beta-oligonucleotides containing polarity reversals. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2001; 20:493-9. [PMID: 11563065 DOI: 10.1081/ncn-100002324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We employ NMR structure determination, thermodynamics, and enzymatics to uncover the structural, thermodynamic and enzymatic properties of alpha/beta-ODNs containing 3'-3' and 5'-5' linkages. RNase H studies show that alpha/beta-gapmers that are designed to target erbB-2 efficiently elicit RNase H activity. NMR structures of DNA.DNA and DNA.RNA duplexes reveal that single alpha-anomeric residues fit well into either duplex, but alter the dynamic properties of the backbone and deoxyriboses as well as the topology of the minor groove in the DNA.RNA hybrid.
Collapse
Affiliation(s)
- M W Germann
- Department of Microbiology and Immunology, Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia 19107, USA
| | | | | | | |
Collapse
|
19
|
|
20
|
Lui VW, He Y, Huang L. Specific down-regulation of HER-2/neu mediated by a chimeric U6 hammerhead ribozyme results in growth inhibition of human ovarian carcinoma. Mol Ther 2001; 3:169-77. [PMID: 11237673 DOI: 10.1006/mthe.2000.0241] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The U6 expression system was explored for efficient expression of a ribozyme against the human proto-oncogene c-neu. A hammerhead ribozyme (neuRz) and the control mutant ribozyme (MRz) were targeted to cleave c-neu mRNA at the tyrosine kinase domain. In vitro cleavage showed that neuRz was very active while MRz was not. Near-maximal target cleavage observed at a low ribozyme:target ratio (0.1) suggests that neuRz has good activity and turnover capability under physiological conditions, i.e., <5 mM MgCl(2) and 37 degrees C. Chimeric U6 ribozyme was expressed at about 5 x 10(6) copies/cell at 48 h in the ovarian carcinoma cell line SKOV-3.ip1. Partial down-regulation of c-neu mRNA and protein was observed in a dose-dependent manner in cells transiently transfected with U6neuRz- but not with MRz-containing plasmid. Sorted transient transfectants demonstrated dramatic growth inhibition with the neuRz-expressing cells. Our results demonstrate that the U6 expression system is very efficient and suitable for the expression of a hammerhead ribozyme. Moreover, nonviral delivery of the neuRz-expressing plasmid resulted in specific down-regulation of c-neu and, subsequently, growth inhibition of ovarian cancer cells overexpressing c-neu.
Collapse
Affiliation(s)
- V W Lui
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
21
|
Abstract
When epidermal growth factor and its relatives bind the ErbB family of receptors, they trigger a rich network of signalling pathways, culminating in responses ranging from cell division to death, motility to adhesion. The network is often dysregulated in cancer and lends credence to the mantra that molecular understanding yields clinical benefit: over 25,000 women with breast cancer have now been treated with trastuzumab (Herceptin), a recombinant antibody designed to block the receptor ErbB2. Likewise, small-molecule enzyme inhibitors and monoclonal antibodies to ErbB1 are in advanced phases of clinical testing. What can this pathway teach us about translating basic science into clinical use?
Collapse
Affiliation(s)
- Y Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| | | |
Collapse
|
22
|
Scott GK, Chang CH, Erny KM, Xu F, Fredericks WJ, Rauscher FJ, Thor AD, Benz CC. Ets regulation of the erbB2 promoter. Oncogene 2000; 19:6490-502. [PMID: 11175365 DOI: 10.1038/sj.onc.1204041] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Evaluating the chromatinized erbB2 gene in nuclei from breast cancer cells expressing varying levels of ErbB2 transcripts, we identified a nuclease-sensitive site within a 0.22 kb region of maximum enhancer activity centered over a conserved 28 bp polypurine(GGA)-polypyrimidine(TCC) mirror-repeat and an adjacent essential Ets binding site (EBS). Promoter footprinting with nuclear extracts reveals an intense Ets hypersensitivity site at the EBS whose degree of intensity correlates with the level of cellular ErbB2 expression. In vitro mapping assays show that the supercoiled erbB2 promoter forms an internal triplex structure (Hr-DNA) at the mirror-repeat element. Mutations preventing Hr-DNA formation can enhance erbB2 promoter activity in human breast cancer cells, a result consistent with previous demonstration that Ets-erbB2 promoter complexes cannot form when the mirror-repeat is engaged in triplex binding, and new results suggesting that Ets binding induces severe promoter bending that may restrict local triplex formation. In addition to previously described erbB2-regulating breast cancer Ets factors (PEA3, ESX/Elf-3), Elf-1 is now shown to be another endogenously expressed Ets candidate capable of binding to and upregulating the erbB2 promoter. Given current strategies to transcriptionally inhibit ErbB2 overexpression, including development of novel erbB2 promoter-targeted therapeutics, an EBS-targeted approach is presented using chimeric Ets proteins that strongly repress erbB2 promoter activity.
Collapse
Affiliation(s)
- G K Scott
- Division of Oncology-Hematology, Department of Medicine, University of California at San Francisco, San Francisco, California, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Dittmar T, Schäfer F, Brandt BH, Zänker KS. Accelerated assessing of antisense RNA efficacy using a chimeric enhanced green fluorescent protein-antisense RNA-producing vector. ANTISENSE & NUCLEIC ACID DRUG DEVELOPMENT 2000; 10:401-8. [PMID: 11079579 DOI: 10.1089/oli.1.2000.10.401] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The selection of suitable parts of a gene as antisense RNA sequences is largely a matter of trial and error and, as a consequence, a rather time-consuming process. In this study, we present a rapid and reproducible method to bypass this protracted procedure by using a chimeric enhanced green fluorescent protein (EGFP)-antisense RNA-producing vector. The combination of a reporter gene and antisense RNA allows easy measurement by flow cytometry of antisense RNA efficacy in successfully transfected cells shortly after transfection. Four chimeric EGFP-p185c-erbB-2-antisense RNA vectors were constructed and transfected into the p185-c-erbB-2-overexpressing cell line SKBR3. Within 1 week, we were able to estimate the inhibitory capacities of the different antisense RNA sequences used in this study. Our results strongly suggest that a chimeric EGFP-antisense RNA vector is an appropriate tool to expedite the laboratory work and time in screening the efficacy of antisense RNA strategies.
Collapse
Affiliation(s)
- T Dittmar
- Institute of Immunology, University of Witten/Herdecke, Germany
| | | | | | | |
Collapse
|
24
|
Hsieh SS, Malerczyk C, Aigner A, Czubayko F. ERbB-2 expression is rate-limiting for epidermal growth factor-mediated stimulation of ovarian cancer cell proliferation. Int J Cancer 2000; 86:644-51. [PMID: 10797285 DOI: 10.1002/(sici)1097-0215(20000601)86:5<644::aid-ijc7>3.0.co;2-t] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Over-expression of the ErbB-2 proto-oncogene frequently coincides with an aggressive clinical course of certain human adenocarcinomas. The ErbB-2 receptor is a member of the ErbB family of growth factor receptors, and within this complex signaling network, ErbB-2-containing heterodimers are preferentially formed. To assess whether ErbB-2 is a critical component in epidermal growth factor (EGF)-mediated stimulation of tumor cell proliferation, we used as a model SK-OV-3 ovarian cancer cells, which over-express EGF receptor (EGFR) and ErbB-2 receptors. In these cells, we reduced ErbB-2 mRNA and protein expression by transfection with ErbB-2-targeted hammerhead ribozymes and generated cell lines expressing different levels of ErbB-2. In SK-OV-3 cells, ErbB-2 expression conferred a growth advantage and soft agar experiments revealed that ErbB-2 was rate-limiting for anchorage-independent growth. The induction of colony formation by EGF was completely abrogated in ErbB-2-depleted cells, despite unchanged expression levels and tyrosine phosphorylation of the EGFR. The duration of EGF-mediated c-Fos mRNA up-regulation was decreased in parallel with loss of ErbB-2 expression. Furthermore, the rate of spontaneous apoptosis was increased in ErbB-2-depleted cells. Our results demonstrate that in human ovarian cancer cells the EGFR-ErbB-2 heterodimer, and not the EGFR homodimer, can be rate-limiting for EGF-mediated proliferation, thus suggesting that the oncogenic activity of ErbB-2 in human tumors is due in part to its ability to increase the growth response to stroma-derived EGF-like growth factors.
Collapse
Affiliation(s)
- S S Hsieh
- Department of Pharmacology and Toxicology, Philipps-University Marburg, Germany
| | | | | | | |
Collapse
|
25
|
Nagy P, Jenei A, Damjanovich S, Jovin TM, Szölôsi J. Complexity of signal transduction mediated by ErbB2: clues to the potential of receptor-targeted cancer therapy. Pathol Oncol Res 2000; 5:255-71. [PMID: 10607920 DOI: 10.1053/paor.1999.0255] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The erbB2 oncogene belongs to the type I trans-membrane tyrosine kinase family of receptors. Its medical importance stems from its widespread over-expression in breast cancer. This review will focus on the signal transduction through this protein, and explains how the overexpression of erbB2 may result in poor prognosis of breast cancer, and finally it will summerize our current understanding about the therapeutic potential of receptor-targeted therapy in breast cancer. ErbB2 does not have any known ligand which is able to bind to it with high affinity. However the kinase activity of erbB2 can be activated without any ligand, if it is overexpressed, and by heteroassociation with other members of the erbB family (erbB1 or epidermal growth factor receptor, erbB3 and erbB4). This interaction substantially increases the efficiency and diversity of signal transduction through these receptor complexes. In addition, erbB2 forms large scale receptor clusters containing hundreds of proteins. These receptor islands may take part in recruiting cytosolic factors which relay the signal towards the nucleus or the cytoplasm. Overexpression of erbB2 was linked to higher transforming activity, increased metastatic potential, angiogenesis and drug resistence of breast tumor in laboratory experiments. As a corollary of these properties, erbB2 amplification is generally thought to be associated with a poor prognosis in breast cancer patients. These early findings lead to the development of antibodies that down-regulate erbB2. Such a therapeutic approach has already been found effective in experimental tumor models and in clinical trials as well. Further understanding of the importance of erbB2 and growth factor receptors in the transformation of normal cells to malignant ones may once give us a chance to cure erbB2 over-expressing breast cancer.
Collapse
Affiliation(s)
- P Nagy
- Hungarian Academy of Sciences, Biophysical Workgroup, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
26
|
Suzuki T, Anderegg B, Ohkawa T, Irie A, Engebraaten O, Halks-Miller M, Holm PS, Curiel DT, Kashani-Sabet M, Scanlon KJ. Adenovirus-mediated ribozyme targeting of HER-2/neu inhibits in vivo growth of breast cancer cells. Gene Ther 2000; 7:241-8. [PMID: 10694801 DOI: 10.1038/sj.gt.3301065] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
HER-2/neu is overexpressed in 25-30% of human breast cancers. We prepared an anti-HER-2/neu hammerhead ribozyme expressed by a recombinant adenovirus (rAdHER-Rz). Human breast cancer cell lines were transduced with high efficiency, resulting in decreased HER-2/neu expression. In vivo injections of rAdHER-Rz into BT-474 tumors established in nude mice inhibited tumor growth to 20% of mock-treated controls. Similar in vivo effects were shown in MCF-7 cells, which do not overexpress HER-2/neu. The growth inhibitory effects of rAdHER-Rz were greater than those of an antisense-expressing vector. These results suggest the utility of anti-HER-2/neu ribozymes as a rational strategy for gene therapy of breast cancer. Gene Therapy (2000) 7, 241-248.
Collapse
Affiliation(s)
- T Suzuki
- Cancer Research Department, Berlex Biosciences, Richmond, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Affiliation(s)
- B Kuss
- Department of Hematology, St. Bartholomew's London, UK
| | | |
Collapse
|
28
|
Abstract
Molecular medicine and genomics technologies are inseparable for defining new molecular targets. cDNA databases and elementary informatic tools provide instantaneous glimpses of gene families or tissue-restricted expression patterns as a means of new target identification. In addition, cDNA microarrays and two-dimensional gel electrophoresis unmask the expression of genes with unassigned or unexpected functions. Depletion of mRNA with ribozymes or neutralization of proteins with intracellular antibodies enable investigators to reject or embrace new molecular hypotheses about the determinants of disease, pharmacology or toxicology.
Collapse
Affiliation(s)
- D A Jones
- Division of Molecular Pharmacology, Department of Medicinal Chemistry, The Huntsman Cancer Institute, 546 Chipeta Way, Suite 1100, Salt Lake City, Utah 84108, USA.
| | | |
Collapse
|
29
|
Müller M, Dietel M, Turzynski A, Wiechen K. Antisense phosphorothioate oligodeoxynucleotide down-regulation of the insulin-like growth factor I receptor in ovarian cancer cells. Int J Cancer 1998; 77:567-71. [PMID: 9679760 DOI: 10.1002/(sici)1097-0215(19980812)77:4<567::aid-ijc16>3.0.co;2-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The insulin-like growth factors (IGF-I and IGF-II) play a key role in cellular proliferation and are involved in cellular transformation. The expression of the IGF-I receptor has been demonstrated in a variety of human tumor cell lines including ovarian cancer cells. Phosphorothioate antisense oligodeoxynucleotides (S-ODNs) were analyzed for their potential to suppress the IGF-I receptor in the NIH: OVCAR-3 ovarian cancer cell line. The application of the antisense S-ODN reduced potently the cell growth of unstimulated NIH:OVCAR-3 cells, whereas sense and mismatch S-ODNs were without any effect. This effect resembled that of the monoclonal antibody (MAb) alphaIR-3. In contrast to the antisense compound, this MAb only partially inhibited the IGF-I-induced proliferation of ovarian cancer cells. The concentration of the antisense S-ODN to exhibit an identical inhibition of cell proliferation was reduced to 50 nM when the oligonucleotides were delivered by the cationic lipid formulation lipofectin. The specificity of the antisense S-ODN action was confirmed by reduction of the receptor protein and of the receptor mRNA, as assayed by flow cytometry and by Northern blot hybridizations. Our data demonstrate the potency of antisense S-ODNs to target the IGF-I receptor message and show that antisense strategies against the IGF-I receptor may provide new strategies for the therapy of ovarian cancer.
Collapse
Affiliation(s)
- M Müller
- Institute of Pathology, Universitätsklinikum Charité, Medizinische Fakultät der Humboldt-Universität Berlin, Germany
| | | | | | | |
Collapse
|
30
|
Roh H, Pippin J, Boswell C, Drebin JA. Antisense oligonucleotides specific for the HER2/neu oncogene inhibit the growth of human breast carcinoma cells that overexpress HER2/neu. J Surg Res 1998; 77:85-90. [PMID: 9698539 DOI: 10.1006/jsre.1998.5353] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The HER2/neu oncogene encodes a cell surface protein which plays a role in growth factor-stimulated mitogenic signaling. HER2/neu is overexpressed in 30-40% of human breast carcinomas. This study tested the hypothesis that inhibiting HER2/neu expression using a phosphorothioate antisense (AS) oligonucleotide would inhibit the growth of breast cancer cells that overexpress this gene. A human breast carcinoma cell line, BT474, which overexpresses the HER2/neu oncogene was exposed to AS, sense (S), or scrambled antisense (SC) phosphorothioate oligonucleotides in tissue culture. Treatment with AS oligonucleotides specifically downregulated HER2/neu mRNA expression and resulted in lower levels of the HER2/neu protein product, p185; control oligonucleotides had no such effect. AS oligonucleotide treatment significantly inhibited the in vitro growth of BT474 cells, whereas S and SC controls had little effect on BT474 growth. HER2/neu AS oligonucleotide treatment had no effect on the growth of a distinct breast cancer line, MCF7, which expresses low levels of the HER2/neu oncogene. Breast carcinoma cells which overexpress the HER2/neu gene appear to be dependent on continued expression of this oncogene for cell growth. AS oligonucleotide pharmaceuticals which interfere with the expression of the HER2/neu oncogene may be of use in the therapy of some patients with breast carcinoma.
Collapse
Affiliation(s)
- H Roh
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | | | | | | |
Collapse
|
31
|
Allal C, Sixou S, Kravtzoff R, Soulet N, Soula G, Favre G. SupraMolecular BioVectors (SMBV) improve antisense inhibition of erbB-2 expression. Br J Cancer 1998; 77:1448-53. [PMID: 9652760 PMCID: PMC2150199 DOI: 10.1038/bjc.1998.238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
New therapeutic strategies are now being developed against adenocarcinoma associated with erbB-2 amplification, particularly by inhibiting p185erbB-2 expression. Antisense oligodeoxynucleotides seem promising for this purpose as long as they are efficiently protected against degradation and targeted into the cells. We present antisense oligonucleotide carriers, the supramolecular biovectors (SMBVs), for which we have already demonstrated the ability to improve both cellular uptake and protection of oligodeoxynucleotide. The present work demonstrates that SMBVs elicit a specific and non-toxic action of antisense compounds in a cell model, irrespective of their sensitivity to nucleases. This is a major point, considering the specificity problems associated with the use of nuclease-resistant phosphorothioate oligodeoxynucleotide. SMBVs improve antisense efficiency of oligodeoxynucleotide designed against p185erbB-2, with a complete growth arrest of SK-Br-3, human adenocarcinoma mammary cells that overexpress p185erbB-2 and no effect on MCF-7 cells that normally express p185erbB-2. The comparison of SMBVs with DOTAP reveals the statistically higher efficiency of SMBVs, which allows the antisense inhibition of p185erbB-2 expression in 65-75% of SK-Br-3 cells (P < 0.05). The efficiency and controlled synthesis of SMBVs underline their potentialities as oligodeoxynucleotide carriers for in vivo experiments.
Collapse
Affiliation(s)
- C Allal
- Laboratoire d'Oncologie Cellulaire et Moléculaire, EA 2048 MRES, Faculté des Sciences Pharmaceutiques and Centre de Lutte Contre le Cancer Claudius Regaud, Toulouse, France
| | | | | | | | | | | |
Collapse
|
32
|
Yoo JY, Hamburger AW. Changes in heregulin beta1 (HRGbeta1) signaling after inhibition of ErbB-2 expression in a human breast cancer cell line. Mol Cell Endocrinol 1998; 138:163-71. [PMID: 9685225 DOI: 10.1016/s0303-7207(98)00004-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Specific biological responses to the erbB3-erbB4 ligand heregulin (HRG) have been postulated to be due to the formation of heterodimers of those receptors with erbB2. To test the role that erbB2 plays in the response to HRG in a human breast carcinoma cell line, antisense oriented erbB2 was stably transfected into AU565 cells. In the absence of HRG, inhibition of erbB2 expression slowed cell growth, leading to accumulation of cells in the G2/M phase, and suppressed colony growth in soft agar. Low concentrations of HRG induced cell proliferation in both the erbB2-nonexpressing cells and the parental AU565 cells. In contrast, high concentrations of HRG failed to induce differentiation of the erbB2-nonexpressing cells as compared with the parental cells. ErbB3 expression was significantly decreased in the erbB2 nonexpressing cells. ErbB3 was constitutively tyrosine phosphorylated in both the parental AU565 cells and in the erbB2 nonexpressing cells. HRG further increased tyrosine phosphorylation of erbB3 with a maximum response at 1 ng/ml of HRG in erbB2 nonexpressing cells, as compared with 10 ng/ml of HRG in AU565 cells. This finding suggested that the biochemical responsiveness of erbB3 to HRG was changed, but not abrogated, by inhibition of erbB2 expression. These results suggest that inhibition of erbB2 expression modulates, but does not abolish, HRG mediated signal transduction pathways in a human breast cancer cell line.
Collapse
Affiliation(s)
- J Y Yoo
- Molecular and Cellular Biology Program, University of Maryland, School of Medicine, Baltimore 21201, USA
| | | |
Collapse
|
33
|
Wickstrom E, Tyson FL. Differential oligonucleotide activity in cell culture versus mouse models. CIBA FOUNDATION SYMPOSIUM 1998; 209:124-37; discussion 137-41. [PMID: 9383573 DOI: 10.1002/9780470515396.ch10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The usual course of drug discovery begins with the demonstration of compound activity in cells and, usually, a lower level of activity in animals. Successive rounds of drug design may result in a compound with sufficient activity in animals to justify clinical trials. The basic endpoints of therapeutic oligonucleotide experiments include target antigen reduction, target messenger reduction and inhibition of transformed cell proliferation or viral replication. However, one should expect oligonucleotides to exhibit pleiotropic behaviour, as do all other drugs. In an animal oligonucleotides will necessarily bind to and dissociate from all macromolecules encountered in the blood, in tissues, on cell surfaces and within cellular compartments. Contrary to expectations, oligonucleotides designed to be complementary to certain transcripts have sometimes been found moderately effective in cell-free extracts, more effective in cell culture and most effective in animal models. If greater potency against standard endpoints is reported in mouse models than was observed in cell culture, critical examination must consider alternate modes of action in animals that may not apply in cell culture. This counterintuitive paradox will be examined, based on studies of Ha-ras expression in bladder cancer, Ki-ras expression in pancreatic cancer, erbB2 expression in ovarian cancer and c-myc expression in B cell lymphoma.
Collapse
Affiliation(s)
- E Wickstrom
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | |
Collapse
|
34
|
Sacco MG, Gribaldo L, Barbieri O, Turchi G, Zucchi I, Collotta A, Bagnasco L, Barone D, Montagna C, Villa A, Marafante E, Vezzoni P. Establishment and characterization of a new mammary adenocarcinoma cell line derived from MMTV neu transgenic mice. Breast Cancer Res Treat 1998; 47:171-80. [PMID: 9497105 DOI: 10.1023/a:1005988715285] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A new murine cell line, named MG1361, was established from mammary adenocarcinomas arising in a MMTV-neu transgenic mouse lineage where breast tumors develop in 100% of females, due to the overexpression of the activated rat neu oncogene in the mammary gland. The MG1361 cell line shows an epithelial-like morphology, has a poor plating efficiency, low clonogenic capacity, and a doubling time of 23.8 hours. Karyotype and flow cytometry analysis revealed a hypotetraploid number of chromosomes, whereas cell cycle analysis showed 31.2% of cells to be in the G1 phase, 21.4% in S and 47.4% in G2 + M. This cell line maintains a high level of neu expression in vitro. The MG1361 cell line was tumorigenic when inoculated in immunodeficient (nude) mice and the derived tumors showed the same histological features as the primary tumors from which they were isolated. MG1361 cells were positive for specific ER and PgR binding which was competed by tamoxifen, making this cell line useful for the evaluation of endocrine therapy. Moreover, they were sensitive to etoposide treatment, suggesting that they could be a model for the study of chemotherapy-induced apoptosis. As the tumors arising in MMTV-neu transgenic mice have many features in common with human mammary adenocarcinomas (Sacco et al., Gene Therapy 1995; 2: 493-497), this cell line can be utilized to perform basic studies on the role of the neu oncogene in the maintenance of the transformed phenotype, and to test novel protocols of therapeutic strategies.
Collapse
Affiliation(s)
- M G Sacco
- Istituto di Tecnologie Biomediche Avanzate, CNR, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Juhl H, Downing SG, Wellstein A, Czubayko F. HER-2/neu is rate-limiting for ovarian cancer growth. Conditional depletion of HER-2/neu by ribozyme targeting. J Biol Chem 1997; 272:29482-6. [PMID: 9368008 DOI: 10.1074/jbc.272.47.29482] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Amplification and overexpression of the HER-2/neu proto-oncogene frequently coincide with an aggressive clinical course of certain human adenocarcinomas. To assess whether HER-2/neu plays a rate-limiting role in ovarian cancer, we used human SK-OV-3 ovarian cancer cells as a model. We applied a conditional mRNA depletion strategy of HER-2/neu with anti-HER-2/neu-targeted hammerhead ribozymes expressed under the control of a tetracycline-regulated promoter system. In these ovarian cancer cells, we reduced HER-2/neu mRNA, protein expression, and tumor growth in nude mice by transfection with HER-2/neu-targeted ribozymes and generated cell lines expressing different levels of HER-2/neu. Expression of the most effective ribozyme (Rz3) quenched HER-2/neu mRNA levels by >90%. Concomitantly, fluorescence-activated cell sorting analysis revealed that expression of the HER-2/neu-encoded surface glycoprotein was almost completely abrogated. In nude mice, tumor growth was dramatically inhibited in the HER-2/neu-depleted Rz3-expressing SK-OV-3 cells. Furthermore, already established tumors started to regress when Rz3 expression was activated midstream by withdrawal of the tetracycline treatment. This study supports the thesis that HER-2/neu can be rate-limiting for the malignant phenotype of ovarian cancer in a gene dose-dependent manner.
Collapse
Affiliation(s)
- H Juhl
- Lombardi Cancer Center and Department of Pharmacology, Georgetown University, Washington, D. C. 20007, USA
| | | | | | | |
Collapse
|
36
|
Lavrovsky Y, Chen S, Roy AK. Therapeutic potential and mechanism of action of oligonucleotides and ribozymes. BIOCHEMICAL AND MOLECULAR MEDICINE 1997; 62:11-22. [PMID: 9367793 DOI: 10.1006/bmme.1997.2631] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Specific inactivation of gene expression is an attractive approach for rational drug design to combat degenerative diseases and infectious agents. Oligonucleotide-directed triple-helix formation at cis-acting elements of gene promoters, short oligonucleotides containing base sequences that are complementary to the messenger RNA (antisense oligos), and RNA enzymes (ribozymes) that specifically cleave messenger RNA molecules are currently being used both as experimental tools and as therapeutic agents. Mechanisms of action of various oligonucleotide-based drugs, recent developments in the drug-delivery approaches, and future potentials are discussed in this review.
Collapse
Affiliation(s)
- Y Lavrovsky
- Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78284-7762, USA
| | | | | |
Collapse
|
37
|
Casalini P, Ménard S, Malandrin SM, Rigo CM, Colnaghi MI, Cultraro CM, Segal S. Inhibition of tumorigenicity in lung adenocarcinoma cells by c-erbB-2 antisense expression. Int J Cancer 1997; 72:631-6. [PMID: 9259403 DOI: 10.1002/(sici)1097-0215(19970807)72:4<631::aid-ijc14>3.0.co;2-e] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The lung carcinoma cell line Calu3, which overexpresses the c-erbB-2 oncogene, was stably transfected with antisense (AS) cDNA constructs encompassing different regions of the c-erbB-2 gene. Transfected cells were analyzed for their tumorigenic properties in vitro and in nude mice. Two independent clones, AS F1 (low erbB-2 expressor) and AS B12 (high erbB-2 expressor), as well as the polyclonal Calu3/AS 5', were selected for these analyses. In Calu3/AS 5' transfected cells and in the AS F1 clone, c-erbB-2 RNA and protein levels were lower than those detected in the parental cell line and the AS B12 clone. Anchorage-independent growth and tumor take were also significantly reduced. Furthermore, cells derived from primary tumors of Calu3/AS 5', AS F1 and AS B12 lost the AS c-erbB-2 DNA insert but retained the gene for G418 resistance. Our results suggest that a correlation between c-erbB-2 overexpression and tumorigenicity may exist in the Calu3 lung carcinoma cell line.
Collapse
Affiliation(s)
- P Casalini
- Division of Experimental Oncology E, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Berton M, Sixou S, Kravtzoff R, Dartigues C, Imbertie L, Allal C, Favre G. Improved oligonucleotide uptake and stability by a new drug carrier, the SupraMolecular Bio Vector (SMBV). BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1355:7-19. [PMID: 9030197 DOI: 10.1016/s0167-4889(96)00111-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Antisense oligodeoxynucleotides are potential therapeutic agents, but their development is still limited by both a poor cellular uptake and a high degradation rate in biological media. The strategy that we propose to face these problems is to use small synthetic carriers, around 30 nm diameter, the SupraMolecular Bio Vectors (SMBV). We used positively charged SMBV and settled the ionic incorporation of negatively charged oligonucleotides into these carriers. A minimal leakage of 10% of total incorporated oligonucleotides was then measured during two months. Both protection and uptake of oligonucleotides were then analyzed. On the one hand, we showed that the incorporation of oligonucleotides into the selected SMBV allows to significantly increase, 8 times, their half-life, in cell growth medium. On the other hand, the internalization of the SMBV, into cells, by an endosomal pathway has been characterized. The essential point is that the SMBV uptake elicits the simultaneous oligonucleotide uptake. The oligonucleotide amount that goes through cells within 5 h can be up to 30 times higher than for free oligonucleotides and the fraction of oligonucleotides that is present in the cytosol is increased up to 10 fold after incorporation into the SMBV. This study demonstrates the ability of SMBV to improve oligonucleotide cellular behaviour.
Collapse
Affiliation(s)
- M Berton
- EA/UPRES 2048, Laboratoire d'Oncologie Cellulaire et Moléculaire, Faculté des Sciences Pharmaceutiques and Centre Claudius Regaud, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
39
|
Vaughn JP, Stekler J, Demirdji S, Mills JK, Caruthers MH, Iglehart JD, Marks JR. Inhibition of the erbB-2 tyrosine kinase receptor in breast cancer cells by phosphoromonothioate and phosphorodithioate antisense oligonucleotides. Nucleic Acids Res 1996; 24:4558-64. [PMID: 8948649 PMCID: PMC146276 DOI: 10.1093/nar/24.22.4558] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Antisense activity against erbB-2 of a variety of sulfur-modified oligonucleotides was examined in a breast cancer cell line which overexpresses this oncogene. Using a 15 base anti-erbB-2 sequence previously shown to be effective, various backbone configurations containing phosphoromonothioate or phosphorodithioate linkages were evaluated for antisense activity by a two-color flow cytometric assay. This sequence was effective in inhibiting the production of erbB-2 protein when it was configured as a monothioate at each linkage and as an alternating dithioate/phosphodiester. Both of these compounds were also able to specifically inhibit erbB-2 mRNA expression, indicative of RNase H-mediated activity. The same sequence protected by either three dithioate or three monothioate linkages at each end was ineffective as an antisense reagent, suggesting that endonuclease activity is a significant determinant of the stability of oligonucleotides. Finally, the erbB-2 sequence target was shifted in an effort to improve antisense activity. A new lead sequence was identified that was significantly more effective in inhibiting erbB-2 protein levels and retained activity at lower concentrations.
Collapse
Affiliation(s)
- J P Vaughn
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|