We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Published Online:https://doi.org/10.2217/rme.10.36

Pluripotent embryonic stem cells (ESCs), when compared with transformed, primary or engineered cells, have unique characteristics and advantages that have resulted in the development of important cell-based tools in modern drug discovery. However, a key limitation has been the availability of human ESCs from patients with specific medical needs and the broad range of genetic variation represented worldwide. Induced pluripotent stem (iPS) cells are derived from somatic cells that are reprogrammed to a pluripotent stem cell state and have functional characteristics similar to ESCs. The demonstration that human iPS cells can be derived, with relative ease, through the introduction of transcription factor combinations has allowed the generation of disease-specific iPS cell lines. Therefore, iPS cell technology may deliver robust, human pluripotent cell lines from a wide range of clinical phenotypes and genotypes. Although human iPS cell technology is still a new tool in drug discovery, the promise that this technology will impact the discovery of new therapies can be projected based on the uptake of stem cell applications in biopharmaceutical sciences. Here, the near-term opportunities that iPS cells may deliver to drug discoverers to generate and test hypotheses will be discussed, with a focus on the specific strengths and weaknesses of iPS cell technology. Finally, the future perspective will address novel opportunities iPS cells could uniquely deliver to the preclinical development of new drug therapies.

Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

Bibliography

  • Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell126(4),663–676 (2006).▪▪ First paper to describe the reprogramming of a somatic cell into a pluripotent stem cell through the overexpression of four transcription factors.
  • Baker M: iPS cells: potent stuff. Nat. Methods7(1),17–19 (2009).
  • Woltjen K, Michael IP, Mohseni P et al.: Piggybac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature458(7239),766–770 (2009).
  • Soldner F, Hockemeyer D, Beard C et al.: Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell136(5),964–977 (2009).
  • Sommer CA, Sommer AG, Longmire TA et al.: Excision of reprogramming transgenes improves the differentiation potential of iPS cells generated with a single excisable vector. Stem Cells28(1),64–74 (2009).
  • Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S: Generation of mouse induced pluripotent stem cells without viral vectors. Science322(5903),949–953 (2008).
  • Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K: Induced pluripotent stem cells generated without viral integration. Science322(5903),945–949 (2008).
  • Yu J, Hu K, Smuga-Otto K et al.: Human induced pluripotent stem cells free of vector and transgene sequences. Science324(5928),797–801 (2009).
  • Zhou H, Wu S, Joo JY et al.: Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell4(5),381–384 (2009).
  • 10  Kim D, Kim CH, Moon JI et al.: Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell4(6),472–476 (2009).
  • 11  Ichida JK, Blanchard J, Lam K et al.: A small-molecule inhibitor of TGF-β signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell5(5),491–503 (2009).
  • 12  Shi Y, Do JT, Desponts C, Hahm HS, Scholer HR, Ding S: A combined chemical and genetic approach for the generation of induced pluripotent stem cells. Cell Stem Cell2(6),525–528 (2008).
  • 13  Lin T, Ambasudhan R, Yuan X et al.: A chemical platform for improved induction of human iPSCs. Nat. Methods6(11),805–808 (2009).
  • 14  Lyssiotis CA, Foreman RK, Staerk J et al.: Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of KLF4. Proc. Natl Acad. Sci. USA106(22),8912–8917 (2009).
  • 15  Maherali N, Hochedlinger K: TGF-β signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and c-myc. Curr. Biol.19(20),1718–1723 (2009).
  • 16  Huangfu D, Maehr R, Guo W et al.: Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol.26(7),795–797 (2008).
  • 17  Rodriguez-Piza I, Richaud-Patin Y, Vassena R et al.: Reprogramming of human fibroblasts to induced pluripotent stem cells under xeno-free conditions. Stem Cells28(1),36–44 (2009).
  • 18  Takahashi K, Narita M, Yokura M, Ichisaka T, Yamanaka S: Human induced pluripotent stem cells on autologous feeders. PLoS One4(12),e8067 (2009).
  • 19  Stadtfeld M, Maherali N, Borkent M, Hochedlinger K: A reprogrammable mouse strain from gene-targeted embryonic stem cells. Nat. Methods7(1),53–55 (2010).
  • 20  Carey BW, Markoulaki S, Beard C, Hanna J, Jaenisch R: Single-gene transgenic mouse strains for reprogramming adult somatic cells. Nat. Methods7(1),56–59 (2010).
  • 21  Lee H, Park J, Forget BG, Gaines P: Induced pluripotent stem cells in regenerative medicine: an argument for continued research on human embryonic stem cells. Regen. Med.4(5),759–769 (2009).▪▪ Addresses the issues related to human induced pluripotent stem (iPS) and embryonic stem cells, which includes support for continued research and development of new pluripotent stem cell lines using both technologies.
  • 22  McNeish JD: Stem cells as screening tools in drug discovery. Curr. Opin. Pharmacol.7(5),515–520 (2007).
  • 23  Takahashi K, Tanabe K, Ohnuki M et al.: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell131(5),861–872 (2007).
  • 24  Yu J, Vodyanik MA, Smuga-Otto K et al.: Induced pluripotent stem cell lines derived from human somatic cells. Science318(5858),1917–1920 (2007).
  • 25  Chin MH, Mason MJ, Xie W et al.: Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell5(1),111–123 (2009).
  • 26  Wilson KD, Venkatasubrahmanyam S, Jia F, Sun N, Butte AJ, Wu JC: MicroRNA profiling of human-induced pluripotent stem cells. Stem Cells Dev.18(5),749–758 (2009).
  • 27  Kay GF, Penny GD, Patel D, Ashworth A, Brockdorff N, Rastan S: Expression of xist during mouse development suggests a role in the initiation of X chromosome inactivation. Cell72(2),171–182 (1993).
  • 28  Maherali N, Sridharan R, Xie W et al.: Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell1(1),55–70 (2007).
  • 29  Novak K: Stem cell therapies: California dreamin’? Cell140(1),10–12 (2010).
  • 30  Sacca R, Engle SJ, Qin W, Stock JL, McNeish JD: Genetically engineered mouse models in drug discovery research. Methods Mol. Biol.602,37–54 (2010).
  • 31  Doetschman TC, Eistetter H, Katz M, Schmidt W, Kemler R: The in vitro development of blastocyst-derived embryonic stem cell lines: formation of visceral yolk sac, blood islands and myocardium. J. Embryol. Exp. Morphol.87,27–45 (1985).
  • 32  Gorba T, Allsopp TE: Pharmacological potential of embryonic stem cells. Pharmacol. Res.47(4),269–278 (2003).
  • 33  McNeish J: Embryonic stem cells in drug discovery. Nat. Rev. Drug Discov.3(1),70–80 (2004).
  • 34  Pouton CW, Haynes JM: Embryonic stem cells as a source of models for drug discovery. Nat. Rev. Drug Discov.6(8),605–616 (2007).
  • 35  Allen M, Svensson L, Roach M, Hambor J, McNeish J, Gabel CA: Deficiency of the stress kinase p38a results in embryonic lethality: characterization of the kinase dependence of stress responses of enzyme-deficient embryonic stem cells. J. Exp. Med.191(5),859–870 (2000).
  • 36  McNeish J, Roach M, Hambor J et al.: High-throughput chemical library screening in embryonic stem cell-derived neurons identifies potentiators of AMPA-type glutamate receptors. J. Biol. Chem.285(22),17209–17217 (2010).
  • 37  Paquette JA, Kumpf SW, Streck RD, Thomson JJ, Chapin RE, Stedman DB: Assessment of the embryonic stem cell test and application and use in the pharmaceutical industry. Birth Defects Res. B Dev. Reprod. Toxicol.83(2),104–111 (2008).
  • 38  Niclis JC, Trounson, AO, Dottori M et al.: Human embryonic stem cell models of Huntington disease. Reprod. Biomed. Online19(1),106–113 (2009).
  • 39  Li W, Wei W, Zhu S et al.: Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors. Cell Stem Cell4(1),16–19 (2009).
  • 40  Wu Z, Chen J, Ren J et al.: Generation of pig induced pluripotent stem cells with a drug-inducible system. J. Mol. Cell Biol.1(1),46–54 (2009).
  • 41  Ezashi T, Telugu BP, Alexenko AP, Sachdev S, Sinha S, Roberts RM: Derivation of induced pluripotent stem cells from pig somatic cells. Proc. Natl Acad. Sci. USA106(27),10993–10998 (2009).
  • 42  Esteban MA, Xu J, Yang J et al.: Generation of induced pluripotent stem cell lines from Tibetan miniature pig. J. Biol. Chem.284(26),17634–17640 (2009).
  • 43  Liu H, Zhu F, Yong J et al.: Generation of induced pluripotent stem cells from adult rhesus monkey fibroblasts. Cell Stem Cell3(6),587–590 (2008).
  • 44  Shimada H, Nakada A, Hashimoto Y, Shigeno K, Shionoya Y, Nakamura T: Generation of canine induced pluripotent stem cells by retroviral transduction and chemical inhibitors. Mol. Reprod. Dev.77(1),2 (2010).
  • 45  Aasen T, Raya A, Barrero MJ et al.: Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat. Biotechnol.26(11),1276–1284 (2008).
  • 46  Giorgetti A, Montserrat N, Aasen T et al.: Generation of induced pluripotent stem cells from human cord blood using Oct4 and Sox2. Cell Stem Cell5(4),353–357 (2009).
  • 47  Haase A, Olmer R, Schwanke K et al.: Generation of induced pluripotent stem cells from human cord blood. Cell Stem Cell5(4),434–441 (2009).
  • 48  Sun N, Panetta NJ, Gupta DM et al.: Feeder-free derivation of induced pluripotent stem cells from adult human adipose stem cells. Proc. Natl Acad. Sci. USA106(37),15720–15725 (2009).
  • 49  Osafune K, Caron L, Borowiak M et al.: Marked differences in differentiation propensity among human embryonic stem cell lines. Nat. Biotechnol.26(3),313–315 (2008).▪ An important paper demonstrating that all pluripotent stem cells will have unique functional characteristics, which need to be considered in advance of experimental design.
  • 50  Choi KD, Yu J, Smuga-Otto K et al.: Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells27(3),559–567 (2009).
  • 51  Colman A, Dreesen O: Induced pluripotent stem cells and the stability of the differentiated state. EMBO Rep.10(7),714–721 (2009).
  • 52  D’Amour KA, Bang AG, Eliazer S et al.: Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat. Biotechnol.24(11),1392–1401 (2006).
  • 53  Song Z, Cai J, Liu Y et al.: Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Res.19(11),1233–1242 (2009).
  • 54  Kehat I, Kenyagin-Karsenti D, Snir M et al.: Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J. Clin. Invest.108(3),407–414 (2001).
  • 55  Maehr R, Chen S, Snitow M et al.: Generation of pluripotent stem cells from patients with Type 1 diabetes. Proc. Natl Acad. Sci. USA106(37),15768–15773 (2009).
  • 56  Chan EM, Ratanasirintrawoot S, Park IH et al.: Live cell imaging distinguishes bona fide human iPS cells from partially reprogrammed cells. Nat. Biotechnol.27(11),1033–1037 (2009).
  • 57  Laurent LC, Nievergelt CM, Lynch C et al.: Restricted ethnic diversity in human embryonic stem cell lines. Nat. Methods7(1),6–7 (2010).
  • 58  Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L: Highly efficient neural conversion of human ES and iPS cells by dual inhibition of smad signaling. Nat. Biotechnol.27(3),275–280 (2009).
  • 59  Ebert AD, Yu J, Rose FF Jr et al.: Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature457(7227),277–280 (2009).▪▪ One of the first to demonstrate functional phenotypes in patient-derived iPS cells. The iPS cells were derived from patients with spinal muscle atrophy and the differentiated iPS cell lines demonstrated relative motor neuron loss over time and expected responsiveness to small therapeutics.
  • 60  Lamba DA, Mcusic A, Hirata RK, Wang PR, Russell D, Reh TA: Generation, purification and transplantation of photoreceptors derived from human induced pluripotent stem cells. PLoS One5(1),e8763 (2010).
  • 61  Buchholz DE, Hikita ST, Rowland TJ et al.: Derivation of functional retinal pigmented epithelium from induced pluripotent stem cells. Stem Cells27(10),2427–2434 (2009).
  • 62  Taura D, Noguchi M, Sone M et al.: Adipogenic differentiation of human induced pluripotent stem cells: comparison with that of human embryonic stem cells. FEBS Lett.583(6),1029–1033 (2009).
  • 63  Sullivan GJ, Hay DC, Park IH et al.: Generation of functional human hepatic endoderm from human induced pluripotent stem cells. Hepatology51(1),329–335 (2010).
  • 64  Si-Tayeb K, Noto FK, Nagaoka M et al.: Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology51(1),297–305 (2010).
  • 65  Yokoo N, Baba S, Kaichi S et al.: The effects of cardioactive drugs on cardiomyocytes derived from human induced pluripotent stem cells. Biochem. Biophys. Res. Commun.387(3),482–488 (2009).▪ Demonstrates that functional cardiomyocytes can be derived from iPS cells and these cells respond in a predicted manner pharmacologically.
  • 66  Zwaka TP, Thomson JA: Homologous recombination in human embryonic stem cells. Nat. Biotechnol.21(3),319–321 (2003).
  • 67  Hockemeyer D, Soldner F, Beard C et al.: Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat. Biotechnol.27(9),851–857 (2009).
  • 68  Zou J, Maeder ML, Mali P et al.: Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell Stem Cell5(1),97–110 (2009).
  • 69  Root DE, Hacohen N, Hahn WC, Lander ES, Sabatini DM: Genome-scale loss-of-function screening with a lentiviral RNAi library. Nat. Methods3(9),715–719 (2006).
  • 70  Saha K, Jaenisch R: Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell5(6),584–595 (2009).▪▪ A comprehensive review of the strengths and weaknesses of iPS cells for use as preclinical disease models. This article also notes advances in technologies that may prove effective in overcoming limitations in current iPS cell-based applications.
  • 71  Guengerich FP: Cytochrome p450 and chemical toxicology. Chem. Res. Toxicol.21(1),70–83 (2008).
  • 72  Zhou SF: Polymorphism of human cytochrome p450 2D6 and its clinical significance: part I. Clin. Pharmacokinet.48(11),689–723 (2009).
  • 73  Gwathmey JK, Tsaioun K, Hajjar RJ: Cardionomics: a new integrative approach for screening cardiotoxicity of drug candidates. Expert Opin. Drug Metab. Toxicol.5(6),647–660 (2009).
  • 74  Huikuri HV, Castellanos A, Myerburg RJ: Sudden death due to cardiac arrhythmias. N. Engl. J. Med.345(20),1473–1482 (2001).
  • 75  Splawski I, Shen J, Timothy KW et al.: Spectrum of mutations in long-QT syndrome genes. KVLQT1, HERG, SCN5A, KCNE1, and KCNE2.Circulation102(10),1178–1185 (2000).
  • 76  Lin G, Xie Y, Ouyang Q et al.: HLA-matching potential of an established human embryonic stem cell bank in China. Cell Stem Cell5(5),461–465 (2009).
  • 77  Condic ML, Rao M: Regulatory issues for personalized pluripotent cells. Stem Cells26(11),2753–2758 (2008).
  • 78  Li Y, Zeng H, Xu RH, Liu B, Li Z: Vaccination with human pluripotent stem cells generates a broad spectrum of immunological and clinical responses against colon cancer. Stem Cells27(12),3103–3111 (2009).
  • 101  Cellular Dynamics www.cellulardynamics.com
  • 102  Diagnostics 2009: Moving towards personalized medicine www.pwc.com