Skip to main content Accessibility help
×
Hostname: page-component-84b7d79bbc-5lx2p Total loading time: 0 Render date: 2024-07-30T18:39:36.076Z Has data issue: false hasContentIssue false

84 - Therapeutic applications of anti-sense mechanisms for the treatment of cancer

from Part 4 - Pharmacologic targeting of oncogenic pathways

Published online by Cambridge University Press:  05 February 2015

A. Robert MacLeod
Affiliation:
Department of Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA, USA
C. Frank Bennett
Affiliation:
Department of Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Our understanding of the molecular pathways controlling tumor growth, survival, and metastasis has increased at a staggering pace over the last decades. Large-scale institutional efforts such as The Cancer Genome Atlas projects have identified previously unknown mutations with high frequencies in various cancers (1). These data, when combined with pathway analysis tools, point us towards genetic events most likely to be important “drivers” of cancer and to those that may be simply “passenger” mutations (2,3). In addition, the widespread availability of functional genomic tools, such as siRNA, shRNA vectors, and genetically engineered mouse models (GEM models), and anti-sense technology, has allowed individual laboratories to carefully evaluate the role of selected genes and pathways, defining their contributions to various aspects of tumorigenesis. Importantly, from these efforts, novel ideas about the fundamentals of the cancer process itself have recently emerged. These include the supporting role of non-tumor cells in the tumor micro-environment, the concept of cancer stem cells, epithelial-to-mesenchymal transitions, the various mechanisms that tumors employ to ensure immune evasion, the principle of oncogene addiction, and the increasingly interesting contribution of non-coding RNAs, to name only a few (4–7). These insights have given us hope that the recent migration of cancer treatment away from untargeted cytotoxic therapy to selective molecular-targeted therapies will ultimately be translated into novel life-saving cancer treatments.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 893 - 902
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008;455:1061–8.CrossRef
Chin, L, Gray, JW. Translating insights from the cancer genome into clinical practice. Nature 2008;452:553–63.CrossRef
Kim, H, Huang, W, Jiang, X, et al. Integrative genome analysis reveals an oncomir/oncogene cluster regulating glioblastoma survivorship. Proceedings of the National Academy of Sciences USA 2010;107:2183–8.CrossRef
Yu, H, Pardoll, D, Jove, R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nature Reviews Cancer 2009;9:798–809.CrossRef
Marshall, L, White, RJ. Non-coding RNA production by RNA polymerase III is implicated in cancer. Nature Reviews Cancer 2008;8:911–14.CrossRef
Ryan, BM, Robles, AI, Harris, CC. Genetic variation in microRNA networks: the implications for cancer research. Nature Reviews Cancer 2010;10:389–402.CrossRef
Weinstein, IB, Joe, A. Oncogene addiction. Cancer Research 2008;68:3077–80.CrossRef
Gerber, DE, Minna, JD. ALK inhibition for non-small cell lung cancer: from discovery to therapy in record time. Cancer Cell 2010;18:548–51.CrossRef
Flaherty, KT, McArthur, G. BRAF, a target in melanoma: implications for solid tumor drug development. Cancer 2010;116:4902–13.CrossRef
Garzon, R, Marcucci, G, Croce, CM. Targeting microRNAs in cancer: rationale, strategies and challenges. Nature Reviews Drug Discovery 2010;9:775–89.CrossRef
Eddy, SR. Non-coding RNA genes and the modern RNA world. Nature Reviews Genetics 2001;2:919–29.CrossRef
Cole-Strauss, A, Yoon, K, Xiang, Y, et al. Correction of the mutation responsible for sickle cell anemia by an RNA-DNA oligonucleotide. Science 1996;273:1386–9.CrossRef
Wang, G, Seidman, MM, Glazer, PM. Mutagenesis in mammalian cells induced by triple helix formation and transcription-coupled repair. Science 1996;271:802–5.CrossRef
Bielinska, A, Shivdasani, RA, Zhang, LQ, Nabel, GJ. Regulation of gene expression with double-stranded phosphorothioate oligonucleotides. Science 1990;250:997–1000.CrossRef
Wang, S, Zhao, X, Suran, R, et al. Knocking down gene function with an RNA aptamer expressed as part of an intron. Nucleic Acids Research 2010;38:e154.
Helene, C, Toulme, JJ. Specific regulation of gene expression by antisense, sense and antigene nucleic acids. Biochimica et Biophysica Acta 1990;1049:99–125.CrossRef
Siwkowski, AM, Malik, L, Esau, CC, et al. Identification and functional validation of PNAs that inhibit murine CD40 expression by redirection of splicing. Nucleic Acids Research 2004;32:2695–706.CrossRef
Vickers, TA, Koo, S, Bennett, CF, et al. Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents: a comparative analysis. Journal of Biological Chemistry 2003;278:7108–18.CrossRefGoogle ScholarPubMed
Baker, BF, Lot, SS, Condon, TP, et al. 2ʹ-O-(2-methoxy)ethyl-modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 mRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. Journal of Biological Chemistry 1997;272:11 994–2000.CrossRefGoogle ScholarPubMed
Bennett, CF, Chiang, MY, Chan, H, Shoemaker, JE, Mirabelli, CK. Cationic lipids enhance cellular uptake and activity of phosphorothioate antisense oligonucleotides. Molecular Pharmacology 1992;41:1023–33.
Miraglia, L, Watt, AT, Graham, MJ, Crooke, ST. Variations in mRNA content have no effect on the potency of antisense oligonucleotides. Antisense Nucleic Acid Drug Devopment 2000;10:453–61.CrossRef
Lima, WF, Rose, JB, Nichols, JG, et al. Human RNase H1 discriminates between subtle variations in the structure of the heteroduplex substrate. Molecular Pharmacology 2007;71:83–91.
Cerritelli, SM, Crouch, RJ. Ribonucelase H: the enzymes in eukaryotes. FEBS Journal 2009;276:1494–505.CrossRefGoogle Scholar
Monia, BP, Lesnik, EA, Gonzalez, C, et al. Evaluation of 2ʹ-modified oligonucleotides containing 2ʹ-deoxy gaps as antisense inhibitors of gene expression. Journal of Biological Chemistry 1993;268:14 514–22.Google ScholarPubMed
McKay, RA, Miraglia, LJ, Cummins, LL, et al. Characterization of a potent and specific class of antisense oligonucleotide inhibitor of human protein kinase C-alpha expression. Journal of Biological Chemistry 1999;274:1715–22.CrossRefGoogle ScholarPubMed
Seth, PPS, Siwkowski, A, Allerson, CR, et al. Short antisense oligonucleotides with novel 2ʹ-4ʹ conformational restricted nucleoside analogues show improved potency without incremental toxicity in animals. Journal of Medicinal Chemistry 2009;52:10–13.CrossRefGoogle Scholar
Matranga, C, Tomari, Y, Shin, C, Bartel, DP, Zamore, PD. Passenger-strand cleavage facilities assembly of siRNA into Ago2-containing RNAi enzyme complexes. Cell 2005;123:607–20.CrossRef
Ghilidiyal, M, Zamore, PD. Small RNAs: an expanding universe. Nature Reviews Genetics 2009;10:94–108.CrossRef
Liu, J, Carmell, MA, Rivas, FV, et al. Argonaute2 is the catalytic engine of mammalian RNAi. Science 2004;305:1437–41.CrossRef
Wang, Y, Sheng, G, Juranek, S, Tuschl, T, Patel, DJ. Structure of the guide-strand-containing argonaute silencing complex. Nature 2008;456:209–13.CrossRef
Elbashir, SM, Harborth, J, Lendeckel, W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001;411:494–8.CrossRef
Lima, WF, Wu, H, Nichols, JG, et al. Binding and cleavage specificities of human Argonaute2. Journal of Biological Chemistry 2009;284:26 017–28.CrossRefGoogle ScholarPubMed
Lima, WF, Rose, JB, Nichols, JG, et al. The positional influence of the helical geometry of the heteroduplex substrate on human RNase H1 catalysis. Molecular Pharmacology 2007;71:73–82.
Lima, WF, Rose, JB, Nichols, JG, et al. Human RNase H1 discriminates between subtle variations in the structure of the heteroduplex substrate. Molecular Pharmacology 2007;71:83–91.
Elbashir, SM, Martinez, J, Patkaniowska, A, Lendeckel, W, Tuschl, T. Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO Journal 2001;20:6877–88.CrossRefGoogle ScholarPubMed
Kole, R, Shukla, RR, Akhtar, S. Pre-mRNA splicing as a target for antisense oligonucleotides. Advanced Drug Delivery Reviews 1991;6:271–86.CrossRef
Kinali, M, Arechavala-Gomeza, V, Feng, L, et al. Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurology 2009;8:918–28.CrossRef
van Deutekom, JC, Janson, AA, Ginjaar, IB, et al. Local dystrophin restoration with antisense oligonucleotide PRO051. New England Journal of Medicine 2007;357:2677–86.CrossRefGoogle ScholarPubMed
Hua, Y, Sahashi, K, Hung, G, et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes and Development 2010;24:1634–44.CrossRef
Taylor, J, Zhang, QQ, Wyatt, JR, Dean, NM. Induction of endogenous Bcl-xS through the control of Bcl-x pre-mRNA splicing by antisense oligonucleotides. Nature Biotechnology 1999;17:1097–100.CrossRef
Clower, CV, Chatterjee, D, Wang, Z, et al. The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proceedings of the National Academy of Sciences USA 2010;107:1894–9.CrossRef
Taylor, JK, Dean, NM. Regulation of pre-mRNA splicing by antisense oligonucleotides. Current Opinion in Drug Discovery and Development 1999;2:147–51.
Alló, M, Buggiano, V, Fededa, JP, et al. Control of alternative splicing through siRNA-mediated transcriptional gene silencing. Nature Structural and Molecular Biology 2009;16:717–24.CrossRef
Dominski, Z, Kole, R. Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides. Proceedings of the National Academy of Sciences USA 1993;90:8673–7.CrossRef
Massirer, KB, Pasquinelli, AE. The evolving role of microRNAs in animal gene expression. Bioessays 2006;28:449–52.CrossRef
Bartel, DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;136:215–33.CrossRef
Mayr, C, Hemann, MT, Bartel, DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 2007;315:1576–9.CrossRef
Cimmino, A, Calin, GA, Fabbri, M, et al. mir-15 and miR-16 induce apoptosis by targeting BCL2. Proceedings of the National Academy of Sciences USA 2005;102:13 944–9.
Volinia, S, Calin, GA, Liu, CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences USA 2006;103:2257–61.CrossRef
He, L, Thomson, JM, Hemann, MT, et al. A microRNA polycistron as a potential human oncogene. Nature 2005;435:828–33.CrossRef
He, L, He, X, Lim, LP, et al. A microRNA component of the p53 tumour suppressor network. Nature 2007;447:1130–4.CrossRef
Medina, PP, Nolde, M, Slack, FL. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 2010;467:86–90.CrossRef
Esau, C, Kang, X, Peralta, E, et al. MicroRNA-143 regulates adipocyte differentiation. Journal of Biological Chemistry 2004;279:52 361–5.CrossRefGoogle ScholarPubMed
Davis, S, Propp, S, Freier, SM, et al. Potent inhibition of microRNA in vivo without degradation. Nucleic Acids Research 2009;37:70–7.CrossRef
Esau, C, Davis, S, Murray, SF, et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metabolism 2006;3:87–98.CrossRef
Krützfeldt, J, Kuwajima, S, Braich, R, et al. Silencing of microRNAs in vivo with “antagomirs”. Nature 2005;438:685–9.CrossRef
Elyakim, E, Sitbon, E, Faerman, A, et al. hsa-miR-191 is a candidate oncogene target for hepatocellular carcinoma therapy. Cancer Research 2010;70:8077–87.CrossRef
Raal, FJ, Santos, RD, Blom, DJ, et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet 2010;375:998–1006.CrossRef
Whitehead, KA, Langer, R, Anderson, DG. Knocking down barriers: advances in siRNA delivery. Nature Reviews Drug Discovery 2009;8:129–38.CrossRef
Hughes, JA, Rao, GA. Targeted polymers for gene delivery. Expert Opinion on Drug Delivery 2005;2:145–57.CrossRef
Davis, ME, Zuckerman, JE, Choi, CH, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010;464:1067–70.CrossRef
Davis, ME. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Molecular Pharmacology 2009;6:659–68.CrossRef
Alnylam Pharmaceuticals. Alnylam demonstrates RNAi in man with systemically delivered RNAi therapeutics. Press release; January 4, 2011. . (Retrieved January 2011).
Santel, A, Aleku, M, Röder, N, et al. Atu027 prevents pulmonary metastasis in experimental and spontaneous mouse metastasis models. Clinical Cancer Research 2010;16:5469–80.CrossRef
Bennett, CF, Swayze, EE. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annual Review of Pharmacology and Toxicology 2010;50:259–93.CrossRef
Croasdell, G. American Diabetes Association: 70th scientific sessions: research on novel therapeutics, part 2. IDrugs 2010;13:598–600.
Excaliard Pharmaceuticals, Inc. Excaliard announces positive results from three phase 2 clinical trials for EXC 001, showing significant improvement in both hypertrophic and fine line scarring. Press release; January 10, 2011. (Retrieved January 2011.)
Gleave, M, Miyake, H, Zangemeister-Wittke, U, Jansen, B. Antisense therapy: current status in prostate cancer and other malignancies. Cancer Metastasis Review 2002;21:79–92.CrossRef
Zellweger, T, Miyake, H, July, LV, et al. Chemonsensitization of human renal cell cancer using antisense oligonucleotides targeting the antiapoptotic gene clusterin. Neoplasia 2001;3:360–7.CrossRef
Zhang, H, Kim, JK, Edwards, CA, et al. Clusterin inhibits apoptosis by interacting with Bax. Nature Cell Biology 2005;7:909–15.CrossRef
Zoubeidi, A, Ettinger, S, Beraldi, E, et al. Clusterin facilitates COMMD1 and I-kappaB degradation to enhance NF-kappaB activity in prostate cancer cells. Molecular Cancer Research 2010;8:119–30.CrossRef
Redondo, M, Villar, E, Torres-Munoz, J, et al. Overexpression of cluster in in human breast carcinoma. American Journal of Pathology 2000;157:393–9.CrossRefGoogle Scholar
Miyake, H, Gleave, M, Kamidono, S, Hara, I. Overexpression of clusterin in transitional cell carcinoma of the bladder is related to disease progression and recurrence. Urology 2002;59:150–4.CrossRef
Miyake, H, Hara, S, Arakawa, S, Kamidono, S, Hara, I. Over expression of clusterin is an independent prognostic factor for nonpapillary renal cell carcinoma. Journal of Urology 2002;167:703–6.CrossRefGoogle ScholarPubMed
Chen, X, Halberg, RB, Ehrhardt, WM, Torrealba, J, Dove, WF. Clusterin as a biomarker in murine and human intestinal neoplasia. Proceedings of the National Academy of Sciences USA 2003;100:9530–5.CrossRef
Steinberg, J, Oyasu, R, Lang, S, et al. Intracellular levels of SGP-2 (Clusterin) correlate with tumor grade in prostate cancer. Clinical Cancer Research 1997;3:1707–11.
July, LV, Akbari, M, Zellweger, T, et al. Clusterin expression is significantly enhanced in prostate cancer cells following androgen withdrawal therapy. Prostate 2002;50:179–88.CrossRef
Chi, KN, Eisenhauer, E, Fazli, L, et al. A Phase I pharmacokinetic and pharmacodynamic study of OGX-011, a 2′-methoxyethyl antisense oligonucleotide to clusterin, in patients with localized prostate cancer. Journal of the National Cancer Institute 2005;97:1287–96.CrossRefGoogle ScholarPubMed
Chi, KN, Hotte, SJ, Yu, EY, et al. Randomized Phase II study of docetaxel and prednisone with or without OGX-011 in patients with metastatic castration-resistant prostate cancer. Journal of Clinical Oncology 2010;28:4247–54.CrossRefGoogle ScholarPubMed
Ambrosini, G, Adida, C, Altieri, DC. A novel anti-apoptosis gene, survivin, expressed in cancer lymphoma. Nature Medicine 1997;3:917–21.CrossRef
Li, F, Ackermann, EJ, Bennett, CF, et al. Pleiotropic cell-division defects and apoptosis induced by interference with survivin function. Nature Cell Biology 1999;1:461–6.CrossRef
Altieri, DC. Validating survivin as a cancer therapeutic target. Nature Reviews Cancer 2003;3:46–54.CrossRef
Talbot, DC, Ranson, M, Davies, J, et al. Tumor survivin is downregulated by the antisense oligonucleotide LY2181308: a proof-of-concept, first-in-human dose study. Clinical Cancer Research 2010;16:6150–8.CrossRef
Deveraux, QL, Takahashi, R, Salvesen, GS, et al. X-linked IAP is a direct inhibitor of cell death proteases. Nature 1997;388:300–4.CrossRef
Sun, C, Cal, M, Gunasekera, A, et al. NMR structure and mutagenesis of the inhibitor-of apoptosis protein XIAP. Nature 1999;401:818–22.CrossRef
Riedl, SJ, Renatus, M, Schwarzenbacher, R, et al. Structural basis for the inhibition of caspase-3 by XIAP. Cell 2001;104:791–800.CrossRef
Shiozaki, EN, Chai, J, Rigotti, DJ, et al. Mechanism of XIAP-mediated inhibition of caspase-9. Molecular Cell 2003;11:519–27.CrossRef
Schimmer, AD, Welsh, K, Pinilla, C, et al. Small molecule antagonists of apoptosis suppressor XIAP exhibit broad antitumor activity. Cancer Cell 2004;5:25–35.CrossRef
Carter, BZ, Gronda, M, Wang, Z, et al. Small molecule XIAP inhibitors derepress downstream effector caspases and induce apoptosis of acute myeloid leukemia cells. Blood 2005;105:4043–50.CrossRef
Carter, BZ, Milella, M, Tsao, T, et al. Regulation and targeting of antiapoptotic XIAP in acute myeloid leukemia. Leukemia 2003;17:2081–9.CrossRef
Schimmer, AD, Estey, EH, Borthkur, G, et al. Phase I/II trial of AEG35156 X-linked inhibitor of apoptosis protein antisense oligonucleotide combined with idarubicin and cytarabine in patients with relapsed or primary refractory acute myeloid leukemia. Journal of Clinical Oncology 2009;27:4741–6.CrossRefGoogle ScholarPubMed
Netti, PA, Hamberg, LM, Babich, JW, et al. Enhancement of fluid filtration across tumor vessels: implication for delivery of macromolecules. Proceedings of the National Academy of Sciences USA 1999;96:3137–42.CrossRef
Fukumura, D, Jain, RK. Tumor microvasculature and microenvironment: targets for anti-angiogenesis and normalization. Microvascular Research 2007;74:72–84.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×