Target Validation to Biomarker Development
Tóm tắt
With the growing number of putative molecular targets and increased economic pressure on companies developing novel drugs, particularly in the cancer area, the need to work on highly validated targets is essential. The use of biomarkers for proof of mechanism of action is becoming an important tool in validation efforts in the preclinical phase of drug development, helping to reduce the attrition rate of candidate drugs once they have entered the clinic. In this review, we highlight how RNA interference (RNAi) has become the method of choice to perform both target validation and identification in academia and industry. RNAi takes advantage of a naturally occurring mechanism whereby cells regulate the expression of genes at the post-transcriptional level, and it introduces a new era in loss-of-function experiments, allowing for the rapid measurement of the phenotype observed upon target expression abrogation. Design of both small-interfering RNA and short-hairpin RNA constructs and their delivery into cells have emerged as the most important aspects of this technology, and reduction or measurement of potential unwanted off-target effects must also be taken into consideration. A number of successes have already been described, and several oncology targets and biomarkers have been identified and validated with this technique.
Tài liệu tham khảo
Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 2004; 3(8): 711–5
Same old story? Nat Rev Drug Discov 2007; 6 (2): 97
Collins I, Workman P. New approaches to molecular cancer therapeutics. Nat Chem Biol 2006; 2(12): 689–700
Benson JD, Chen YN, Cornell-Kennon SA, et al. Validating cancer drug targets. Nature 2006; 441(7092): 451–6
Sawyers CL. Opportunities and challenges in the development of kinase inhibitor therapy for cancer. Genes Dev 2003; 17(24): 2998–3010
Sanger Institute. Cancer gene census [online]. Available from URL: http://www.sanger.ac.uk/genetics/CGP/Census/ [Accessed 2008 Feb 25]
Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature 2002; 417(6892): 949–54
Garraway LA, Widlund HR, Rubin MA, et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 2005; 436(7047): 117–22
Kim H, Muller WJ. The role of the epidermal growth factor receptor family in mammary tumorigenesis and metastasis. Exp Cell Res 1999; 253(1): 78–87
Kitayama H, Tsujimura T, Matsumura I, et al. Neoplastic transformation of normal hematopoietic cells by constitutively activating mutations of c-kit receptor tyrosine kinase. Blood 1996; 88(3): 995–1004
Li QX, Tan P, Ke N, et al. Ribozyme technology for cancer gene target identification and validation. Adv Cancer Res 2007; 96: 103–43
Kurreck J. Antisense technologies: improvement through novel chemical modifications. Eur J Biochem 2003; 270(8): 1628–44
Pendergrast PS, Marsh HN, Grate D, et al. Nucleic acid aptamers for target validation and therapeutic applications. J Biomol Tech 2005; 16(3): 224–34
Bennett CF. Efficiency of antisense oligonucleotide drug discovery. Antisense Nucleic Acid Drug Dev 2002; 12(3): 215–24
Scherer LJ, Rossi JJ. Approaches for the sequence-specific knockdown of mRNA. Nat Biotechnol 2003; 21(12): 1457–65
Napoli C, Lemieux C, Jorgensen R. Introduction of a Chimeric chalcone synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell 1990; 2(4): 279–89
Cerutti H, Casas-Mollano JA. On the origin and functions of RNA-mediated silencing: from protists to man. Curr Genet 2006; 50(2): 81–99
Fire A, Xu S, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998; 391(6669): 806–11
Kavi HH, Fernandez HR, Xie W, et al. RNA silencing in Drosophila. FEBS Lett 2005; 579: 5940–9
Qi Y, Hannon GJ. Uncovering RNAi mechanisms in plants: biochemistry enters the foray. FEBS Lett 2005; 579: 5899–903
Elbashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001; 411(6836): 494–8
Kim DH, Rossi JJ. Strategies for silencing human disease using RNA interference. Nat Rev Genet 2007; 8(3): 173–84
Bernstein E, Caudy AA, Hammond SM, et al. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001; 409(6818): 363–6
Amarzguioui M, Rossi JJ, Kim D. Approaches for chemically synthesized siRNA and vector-mediated RNAi. FEBS Lett 2005; 579(26): 5974–81
Berezikov E, Guryev V, van de BJ, et al. Phylogenetic shadowing and computational identification of human microRNA genes. Cell 2005; 120(1): 21–4
Bartel DP, Chen CZ. Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nat Rev Genet 2004; 5(5): 396–400
Esquela-Kerscher A, Slack FJ. Oncomirs: microRNAs with a role in cancer. Nat Rev Cancer 2006; 6(4): 259–69
Wienholds E, Plasterk RH. MicroRNA function in animal development. FEBS Lett 2005; 579(26): 5911–22
Ui-Tei K, Naito Y, Takahashi F, et al. Guidelines for the selection of highly effective siRNA sequences for mammalian and chick RNA interference. Nucleic Acids Res 2004; 32(3): 936–48
Kim DH, Behlke MA, Rose SD, et al. Synthetic dsRNA dicer substrates enhance RNAi potency and efficacy. Nat Biotechnol 2005; 23(2): 222–6
Sledz CA, Holko M, de Veer MJ, et al. Activation of the interferon system by short-interfering RNAs. Nat Cell Biol 2003; 5(9): 834–9
de Fougerolles A, Vornlocher HP, Maraganore J, et al. Interfering with disease: a progress report on siRNA-based therapeutics. Nat Rev Drug Discov 2007; 6(6): 443–53
McCaffrey AP, Meuse L, Pham TT, et al. RNA interference in adult mice. Nature 2002; 418(6893): 38–9
Soutschek J, Akinc A, Bramlage B, et al. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 2004; 432: 173–8
Grimm D, Kay MA. Therapeutic application of RNAi: is mRNA targeting finally ready for prime time?J Clin Invest 2007; 117: 3633–41
Zimmermann TS, Lee AC, Akinc A, et al. RNAi-mediated gene silencing in non-human primates. Nature 2006; 441(7089): 111–4
Doench JG, Petersen CP, Sharp PA. siRNAs can function as miRNAs. Genes Dev 2003; 17(4): 438–42
Jackson AL, Burchard J, Schelter J, et al. Widespread siRNA “off-target” transcript silencing mediated by seed region sequence complementarity. RNA 2006; 12(7): 1179–87
Jackson AL, Burchard J, Leake D, et al. Position-specific chemical modification of siRNAs reduces “off-target” transcript silencing. RNA 2006; 12(7): 1197–205
Mook OR, Baas F, de Wissel MB, et al. Evaluation of locked nucleic acid-modified small interfering RNA in vitro and in vivo. Mol Cancer Ther 2007; 6(3): 833–43
Iorns E, Lord CJ, Turner N, et al. Utilizing RNA interference to enhance cancer drug discovery. Nat Rev Drug Discov 2007; 6(7): 556–68
Berns K, Hijmans EM, Mullenders J, et al. A large-scale RNAi screen in human cells identifies new components of the p53 pathway. Nature 2004; 428(6981): 431–7
Kittler R, Pelletier L, Heninger AK, et al. Genome-scale RNAi profiling of cell division in human tissue culture cells. Nat Cell Biol 2007; 9(12): 1401–12
Moffat J, Grueneberg DA, Yang X, et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell 2006; 124(6): 1283–98
MacKeigan JP, Murphy LO, Blenis J. Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nat Cell Biol 2005; 7(6): 591–600
Krausz E. High-content siRNA screening. Mol Biosyst 2007; 3(4): 232–40
Neumann B, Held M, Liebel U, et al. High-throughput RNAi screening by time-lapse imaging of live human cells. Nat Methods 2006; 3(5): 385–90
Zheng L, Liu J, Batalov S, et al. An approach to genomewide screens of expressed small interfering RNAs in mammalian cells. Proc Natl Acad Sci USA 2004; 101(1): 135–40
Brummelkamp TR, Fabius AW, Mullenders J, et al. An shRNA barcode screen provides insight into cancer cell vulnerability to MDM2 inhibitors. Nat Chem Biol 2006; 2(4): 202–6
Draviam VM, Stegmeier F, Nalepa G, et al. A functional genomic screen identifies a role for TAO1 kinase in spindle-checkpoint signalling. Nat Cell Biol 2007; 9(5): 556–64
Ngo VN, Davis RE, Lamy L, et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature 2006; 441(7089): 106–10
Sarthy AV, Morgan-Lappe SE, Zakula D, et al. Survivin depletion preferentially reduces the survival of activated K-Ras-transformed cells. Mol Cancer Ther 2007; 6(1): 269–76
Rottmann S, Wang Y, Nasoff M, et al. A TRAIL receptor-dependent synthetic lethal relationship between MYC activation and GSK3beta/FBW7 loss of function. Proc Natl Acad Sci U S A 2005; 102(42): 15195–200
Collins CS, Hong J, Sapinoso L, et al. A small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase. Proc Natl Acad Sci U S A 2006; 103(10): 3775–80
Farmer H, McCabe N, Lord CJ, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; 434(7035): 917–21
Gartel AL, Kandel ES. RNA interference in cancer. Biomol Eng 2006; 23(1): 17–34
Wacheck V, Losert D, Gunsberg P, et al. Small interfering RNA targeting bcl-2 sensitizes malignant melanoma. Oligonucleotides 2003; 13(5): 393–400
Spankuch-Schmitt B, Bereiter-Hahn J, Kaufmann M, et al. Effect of RNA silencing of polo-like kinase-1 (PLK1) on apoptosis and spindle formation in human cancer cells. J Natl Cancer Inst 2002; 94(24): 1863–77
Choudhury A, Charo J, Parapuram SK, et al. Small interfering RNA (siRNA) inhibits the expression of the Her2/neu gene, upregulates HLA class I and induces apoptosis of Her2/neu positive tumor cell lines. Int J Cancer 2004; 108(1): 71–7
Brummelkamp TR, Bernards R, Agami R. Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell 2002; 2(3): 243–7
Lage H. Potential applications of RNA interference technology in the treatment of cancer. Future Oncol 2005; 1(1): 103–13
Williams NS, Gaynor RB, Scoggin S, et al. Identification and validation of genes involved in the pathogenesis of colorectal cancer using cDNA microarrays and RNA interference. Clin Cancer Res 2003; 9(3): 931–46
Hoeflich KP, Gray DC, Eby MT, et al. Oncogenic BRAF is required for tumor growth and maintenance in melanoma models. Cancer Res 2006; 66(2): 999–1006
Pan Q, Bao LW, Kleer CG, et al. Protein kinase C epsilon is a predictive biomarker of aggressive breast cancer and a validated target for RNA interference anti-cancer therapy. Cancer Res 2005; 65(18): 8366–71
Bae KM, Wang H, Jiang G, et al. Protein kinase C epsilon is overexpressed in primary human non-small cell lung cancers and functionally required for proliferation of non-small cell lung cancer cells in a p21/Cip1-dependent manner. Cancer Res 2007; 67(13): 6053–63
Pan Q, Bao LW, Teknos TN, et al. Targeted disruption of protein kinase C epsilon reduces cell invasion and motility through inactivation of RhoA and RhoC GTPases in head and neck squamous cell carcinoma. Cancer Res 2006; 66(19): 9379–84
Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001; 69: 89–95
Frank R, Hargreaves R. Clinical biomarkers in drug discovery and development. Nat Rev Drug Discov 2003; 2(7): 566–80
Ackermann BL, Hale JE, Duffin KL. The role of mass spectrometry in biomarker discovery and measurement. Curr Drug Metab 2006; 7(5): 525–39
Ayers M, Fargnoli J, Lewin A, et al. Discovery and validation of biomarkers that respond to treatment with brivanib alaninate, a small-molecule VEGFR-2/ FGFR-1 antagonist. Cancer Res 2007; 67(14): 6899–906
Kovar H. Context matters: the hen or egg problem in Ewing’s sarcoma. Semin Cancer Biol 2005; 15(3): 189–96
Stegmaier K, Wong JS, Ross KN, et al. Signature-based small molecule screening identifies cytosine arabinoside as an EWS/FLI modulator in Ewing sarcoma. PLoS Med 2007; 4(4): e122
Nobelprize.org. The Nobel Prize in physiology or medicine [online]. Available from URL: http://nobelprize.org/nobel_prizes/medicine/laureates/2006/ [Accessed 2008 Feb 25]