کاربرد کشت سه ‌بعدی در بهینه‌سازی تحقیقات دارویی سرطان: از روش‌های سنتی تا تکنولوژی نوین میکروسیالی

نوع مقاله : مقاله مروری

نویسندگان

1 گروه زیست‌فن‌آوری پزشکی، دانشکده‌ی پیراپزشکی، دانشگاه علوم پزشکی ایران، تهران، ایران

2 استادیار، گروه مهندسی زیستی و زیست نانو فن‌آوری، دانشکده‌ی علوم و فن‌آوری‌های زیستی، دانشگاه شهید بهشتی، تهران، ایران

3 استادیار، گروه زیست‌ فن‌آوری پزشکی، دانشکده‌ی پیراپزشکی، دانشگاه علوم پزشکی ایران، تهران، ایران

4 استاد، پژوهشکده‌ی لیزر و پلاسما، دانشگاه شهید بهشتی، تهران، ایران

چکیده

با وجود هزینه‌های هنگفت و زمان بسیاری که در مراحل پیش کارآزمایی‌های بالینی صرف می‌شود، بیش از نیمی از فراورده‌های دارویی به دلیل عدم اثربخشی یا بروز عوارض جانبی از چرخه‌ی تولید حذف می‌گردند. این در حالی ‌است که بروز مقاومت‌های دارویی و تفاوت‌های فردی، در بیماری‌های پیچیده‌ای همچون سرطان و نیز گسترش روزافزون مفهوم پزشکی شخصی، نیاز به ارایه‌ی مدل‌های کارآمد و مؤثر را در بررسی‌های دارویی بیش از پیش نمایان می‌سازد. کشت‌های سلولی، ابزارهای مهمی بررسی‌های دارویی هستند که در مراحل پیش‌ کارآزمایی ‌بالینی به ‌طور گسترده مورد استفاده قرار می‌گیرند. امروزه، مسجل شده است که سهم عظیمی از عدم موفقیت مراحل پیش ‌کارآزمایی‌ بالینی به عدم موفقیت کشت‌های سلولی در شبیه‌سازی شرایط بدن بر می‌گردد. به همین دلیل، امروزه ظهور کشت‌های سه‌ بعدی، پنجره‌ای جدید را به روی مطالعات دارویی گشوده است. مطالعات متعددی نشان می‌دهند که شیوه‌های نوین کشت سلول با توانایی شبیه‌سازی تعاملات سلول-سلول، سلول- ماتریکس و ایجاد شیب غلظت دارو، اکسیژن، متابولیت‌ها و مواد غذایی، مشابه با شرایط بدن، به‌ طور مستقیم یا غیر مستقیم (از طریق تأثیرگذاری بر بیان ژن‌ها و ایجاد فنوتیپ طبیعی)، مطالعات دارویی را قابل اعتمادتر کرده‌ است. به تازگی، برای تطابق بیشتر با بدن، از کشت‌های سه بعدی در تراشه‌های میکروسیالی نیز استفاده می‌شود. ریزتراشه‌ها، به دلیل اندازه‌ی نزدیک به سلول‌ها و عروق بدن، امکان کشت سلول به شیوه‌ی پویا و غیره، شباهت بیشتری با بدن فراهم می‌کنند. در مقاله‌ی پیش‌رو، ضمن مروری بر روش‌های مختلف کشت سه ‌بعدی، کاربردهای آن در مطالعات دارویی نیز مطرح می‌گردد.

کلیدواژه‌ها


عنوان مقاله [English]

Three-Dimensional Cell Cultures in Anticancer Drug Researches: From Traditional Methods to Emerging Microfluidic Technology

نویسندگان [English]

  • Shabnam Shahrivari 1
  • Zeinab Bagheri 2
  • Neda Saraygord-Afshari 3
  • Hamid Latifi 4
1 Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
2 Assistant Professor, Department of Biology Engineering and Nanotechnology, School of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
3 Assistant Professor, Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
4 Professor, Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
چکیده [English]

Despite the expensive and time-consuming pre-clinical assessments, more than 50% of medicines fail to reach manufacturing cycle due to the low therapeutic index or possible side effects. Moreover, the challenge of drug resistance and person-to-person variation in complex diseases, such as cancer, and the growing concept of personalized medicine has also made a strong demand for more reliable pre-clinical drug assessment models. Cell culture is an essential and widely-used model in pre-clinical trials, but today we know that traditional culture methods fail to mimic the real in-vivo microenvironment, and hence they are proven to be responsible for the trails’ failure. It seems that the problem is going to be solved by the advent of three-dimentional (3D)-cell culture systems in which cell-cell and cell-matrix interaction, as well as the gradients of drugs, oxygen, metabolites, and nutrients, are well designed to efficiently mimic natural conditions, and provide more reliable results for drug assessments. A variety of researchers have purposed that this new paradigm, either directly or indirectly (by affecting gene expression and inducing natural phenotypes), affect cell behavior to meet the challenge of pre-clinical and clinical inconsistency. Recently, 3D-cultures on microfluidic platforms are used to provide a higher level of adaptation to living bodies. Due to the comparable similarity between microchips and living cells scales as well as the body vessels, besides the possibility to run a dynamic culture situation with real physical tension, etc. they are more potent to mimic actual in vivo microenvironment. Therefore, here, we are going to review various models of 3D-cell culture systems, and discuss their impact on pharmaceutical researches.

کلیدواژه‌ها [English]

  • Cancer
  • Microfluidic microchips
  • Cell culture
  • Drug discovery
  1. Mohs RC, Greig NH. Drug discovery and development: Role of basic biological research. Alzheimers Dement (NY) 2017; 3(4): 651-7.
  2. Hait WN. Anticancer drug development: the grand challenges. Nat Rev Drug Discov 2010; 9(4): 253-4.
  3. Polson AG, Fuji RN. The successes and limitations of preclinical studies in predicting the pharmacodynamics and safety of cell-surface-targeted biological agents in patients. Br J Pharmacol 2012; 166(5): 1600-2.
  4. Begley CG, Ellis LM. Raise standards for preclinical cancer research. Nature 2012; 483(7391): 531-3.
  5. Jedrzejczak-Silicka M. History of cell culture. In: Gowder SJT, editor. New insights into cell culture technology. London, UK: IntechOpen; 2017.
  6. Larson B. 3D cell culture: A review of current techniques. BioTek [Online]. [cited 2015 Nov 12]; Available from: URL: https://www.biotek.com/ resources/ white-papers/3d-cell-culture-a-review-of-current-techniques/
  7. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 2004; 3(8): 711-5.
  8. Ravi M, Paramesh V, Kaviya SR, Anuradha E, Solomon FD. 3D cell culture systems: advantages and applications. J Cell Physiol 2015; 230(1): 16-26.
  9. Petersen OW, Ronnov-Jessen L, Howlett AR, Bissell MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc Natl Acad Sci USA 1992; 89(19): 9064-8.
  10. Fang Y, Eglen RM. Three-Dimensional Cell Cultures in Drug Discovery and Development. SLAS Discov 2017; 22(5): 456-72.
  11. Souza AG, Silva IBB, Campos-Fernandez E, Barcelos LS, Souza JB, Marangoni K, et al. Comparative Assay of 2D and 3D Cell Culture Models: Proliferation, Gene Expression and Anticancer Drug Response. Curr Pharm Des 2018; 24(15): 1689-94.
  12. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol 2014; 12(4): 207-18.
  13. Chignola R, Schenetti A, Andrighetto G, Chiesa E, Foroni R, Sartoris S, et al. Forecasting the growth of multicell tumour spheroids: implications for the dynamic growth of solid tumours. Cell Prolif 2000; 33(4): 219-29.
  14. Zhang X, Xie Y, Koh CG, James LL. A novel 3-D model for cell culture and tissue engineering. Biomed Microdevices 2009; 11(4): 795-9.
  15. Caron MM, Emans PJ, Coolsen MM, Voss L, Surtel DA, Cremers A, et al. Redifferentiation of dedifferentiated human articular chondrocytes: comparison of 2D and 3D cultures. Osteoarthritis Cartilage 2012; 20(10): 1170-8.
  16. Nunes AS, Costa EC, Barros AS, de Melo-Diogo D, Correia IJ. Establishment of 2D Cell Cultures Derived From 3D MCF-7 Spheroids Displaying a Doxorubicin Resistant Profile. Biotechnol J 2019; 14(4): e1800268.
  17. Nizzero S, Ziemys A, Ferrari M. Transport Barriers and Oncophysics in Cancer Treatment. Trends Cancer 2018; 4(4): 277-80.
  18. Tredan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst 2007; 99(19): 1441-54.
  19. Nourse JL, Pathak MM. How cells channel their stress: Interplay between Piezo1 and the cytoskeleton. Semin Cell Dev Biol 2017; 71: 3-12.
  20. Tyler WJ. The mechanobiology of brain function. Nat Rev Neurosci 2012; 13(12): 867-78.
  21. Lam CR, Wong HK, Nai S, Chua CK, Tan NS, Tan LP. A 3D biomimetic model of tissue stiffness interface for cancer drug testing. Mol Pharm 2014; 11(7): 2016-21.
  22. Wei SC, Fattet L, Tsai JH, Guo Y, Pai VH, Majeski HE, et al. Matrix stiffness drives epithelial-mesenchymal transition and tumour metastasis through a TWIST1-G3BP2 mechanotransduction pathway. Nat Cell Biol 2015; 17(5): 678-88.
  23. Chouaib S, Noman MZ, Kosmatopoulos K, Curran MA. Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene 2017; 36(4): 439-45.
  24. Carrera S, de Verdier PJ, Khan Z, Zhao B, Mahale A, Bowman KJ, et al. Protection of cells in physiological oxygen tensions against DNA damage-induced apoptosis. J Biol Chem 2010; 285(18): 13658-65.
  25. Kinoshita M, Johnson DL, Shatney CH, Lee YL, Mochizuki H. Cancer cells surviving hypoxia obtain hypoxia resistance and maintain anti-apoptotic potential under reoxygenation. Int J Cancer 2001; 91(3): 322-6.
  26. Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther 2006; 5(5): 1275-9.
  27. Dhandayuthapani B, Yoshida Y, Maekawa T, Sakthi Kumar D. Polymeric scaffolds in tissue engineering application: A review. International Journal of Polymer Science 2011; 2011: 290602.
  28. Chevalier E, Chulia D, Pouget C, Viana M. Fabrication of porous substrates: a review of processes using pore forming agents in the biomaterial field. J Pharm Sci 2008; 97(3): 1135-54.
  29. Chan BP, Leong KW. Scaffolding in tissue engineering: general approaches and tissue-specific considerations. Eur Spine J 2008; 17 Suppl 4: 467-79.
  30. Yang S, Leong KF, Du Z, Chua CK. The design of scaffolds for use in tissue engineering. Part II. Rapid prototyping techniques. Tissue Eng 2002; 8(1): 1-11.
  31. Hollister SJ. Porous scaffold design for tissue engineering. Nat Mater 2005; 4(7): 518-24.
  32. El-Sherbiny IM, Yacoub MH. Hydrogel scaffolds for tissue engineering: Progress and challenges. Glob Cardiol Sci Pract 2013; 2013(3): 316-42.
  33. Fan L, Yang H, Yang J, Peng M, Hu J. Preparation and characterization of chitosan/gelatin/PVA hydrogel for wound dressings. Carbohydr Polym 2016; 146: 427-34.
  34. Batorsky A, Liao J, Lund AW, Plopper GE, Stegemann JP. Encapsulation of adult human mesenchymal stem cells within collagen-agarose microenvironments. Biotechnol Bioeng 2005; 92(4): 492-500.
  35. Sutherland RM, Inch WR, McCredie JA, Kruuv J. A multi-component radiation survival curve using an in vitro tumour model. Int J Radiat Biol Relat Stud Phys Chem Med 1970; 18(5): 491-5.
  36. Bjerkvig R. Spheroid Culture in Cancer Research (1991). Taylor & Francis Group; 2017.
  37. Jeppesen M, Hagel G, Glenthoj A, Vainer B, Ibsen P, Harling H, et al. Short-term spheroid culture of primary colorectal cancer cells as an in vitro model for personalizing cancer medicine. PLoS One 2017; 12(9): e0183074.
  38. Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev 2009; 23(5): 537-48.
  39. Ghosh S, Spagnoli GC, Martin I, Ploegert S, Demougin P, Heberer M, et al. Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study. J Cell Physiol 2005; 204(2): 522-31.
  40. Tung YC, Hsiao AY, Allen SG, Torisawa YS, Ho M, Takayama S. High-throughput 3D spheroid culture and drug testing using a 384 hanging drop array. Analyst 2011; 136(3): 473-8.
  41. Close DA, Camarco DP, Shan F, Kochanek SJ, Johnston PA. The Generation of Three-Dimensional Head and Neck Cancer Models for Drug Discovery in 384-Well Ultra-Low Attachment Microplates. Methods Mol Biol 2018; 1683: 355-69.
  42. Langhans SA. Three-Dimensional in Vitro Cell Culture Models in Drug Discovery and Drug Repositioning. Front Pharmacol 2018; 9: 6.
  43. Taromi N, Saraygord-Afshari N. Technology to aid the early detection of cancer: A review on the CTC detection techniques, from conventional methods to the advent of the microfluidic Lab-On-Chip devices. Tehran, Iran: Baraye Farda Publications; 2016. [In Persian].
  44. Bagheri Z, Ehtesabi H, Hallaji Z, Aminoroaya N, Tavana H, Behroodi E, et al. On-chip analysis of carbon dots effect on yeast replicative lifespan. Analytica Chimica Acta 2018; 1033: 119-27.
  45. Shahrivari S, Saraygord Afshari N, Bagheri Z, Arzaghy H. Lab-on-chip circulating tumor cell isolation micro devices: A new window for decoding the evolutionary model of cancer. Proceedings of 9th International Congress of Laboratory and Clinic; 2017 Feb 21-24; Tehran, Iran. [In Persian].
  46. Shahrivari S, Saraygord-Afshari N, Bagheri Z. The emergence of micro-isolator devices for high throughput exosome analysis: A technological leap towards personalized cancer treatment. Proceedings of the 2nd International Personalized Medicine Congress, with Cancer Main Topics; 2018 Jan 13-15; Tehran, Iran. [In Persian].
  47. Shahrivari S, Saraygord-Afshari N, Bagheri Z. Microfluidic 3D cell-culture platforms for drug response monitoring: potential applications for individualized cancer treatment. Proceedings of 1st International Congress on Biomedicine; 2017 Dec 18-21; Tehran, Iran. [In Persian].
  48. Shahrabi Farahani M, Taromi N, Saraygord Afshari N, Farajollahi MM. diagnosis of prostate cancer, from conventional methods towards the new promising CTCS. Alborz University Medical Journal 2016; 5(1): 53-8. [In Persian].
  49. Mintz PJ, Rietz AC, Cardo-Vila M, Ozawa MG, Dondossola E, Do KA, et al. Discovery and horizontal follow-up of an autoantibody signature in human prostate cancer. Proc Natl Acad Sci U S A 2015; 112(8): 2515-20.
  50. Bagheri Z, Ehtesabi H, Latifi H, Samimi A, Salehi Moghaddam M, Roshani S. Droplet-based Microfluidic Synthesis of Fluorescent Carbon Dot Hydrogel for Mercury Ions Detection. Proceedings of the EMBL Microfuidics 2018 Conference; 2018 Jul 15-17; Heidelberg, Germany. 2018.
  51. Damiati S, Kompella UB, Damiati SA, Kodzius R. Microfluidic devices for drug delivery systems and drug screening. Genes (Basel) 2018; 9(2).
  52. Chan HF, Zhang Y, Leong KW. Efficient one-step production of microencapsulated hepatocyte spheroids with enhanced functions. Small 2016; 12(20): 2720-30.
  53. McMillan KS, Boyd M, Zagnoni M. Transitioning from multi-phase to single-phase microfluidics for long-term culture and treatment of multicellular spheroids. Lab Chip 2016; 16(18): 3548-57.
  54. Wong AH, Li H, Jia Y, Mak PI, Martins RPDS, Liu Y, et al. Drug screening of cancer cell lines and human primary tumors using droplet microfluidics. Sci Rep 2017; 7(1): 9109.
  55. Chen Y, Gao D, Liu H, Lin S, Jiang Y. Drug cytotoxicity and signaling pathway analysis with three-dimensional tumor spheroids in a microwell-based microfluidic chip for drug screening. Anal Chim Acta 2015; 898: 85-92.
  56. Fu CY, Tseng SY, Yang SM, Hsu L, Liu CH, Chang HY. A microfluidic chip with a U-shaped microstructure array for multicellular spheroid formation, culturing and analysis. Biofabrication 2014; 6(1): 015009.
  57. Lim W, Park S. A microfluidic spheroid culture device with a concentration gradient generator for high-throughput screening of drug efficacy. Molecules 2018; 23(12).
  58. Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. Br J Pharmacol 2011; 162(6): 1239-49.
  59. Arzaghy H, Saraygord Afshari N, Bagheri Z, Shahrivari S. The Art of metastasis-on-chip platforms to create metastasis models and developing cancer understandings. Proceedings of 9th International Congress of Laboratory and Clinic; 2017 Feb 21-24; Tehran, Iran. [In Persian].
  60. Shin MK, Kim SK, Jung H. Integration of intra- and extravasation in one cell-based microfluidic chip for the study of cancer metastasis. Lab Chip 2011; 11(22): 3880-7.
  61. Huh D, Leslie DC, Matthews BD, Fraser JP, Jurek S, Hamilton GA, et al. A human disease model of drug toxicity-induced pulmonary edema in a lung-on-a-chip microdevice. Sci Transl Med 2012; 4(159): 159ra147.