#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

New Approaches for Chemosensitivity Testing in Malignant Diseases


Authors: Sommerová Lucia;  Michalová Eva;  Hrstka Roman
Authors‘ workplace: RECAMO, Masarykův onkologický ústav, Brno
Published in: Klin Onkol 2018; 31(2): 117-124
Category: Review
doi: https://doi.org/10.14735/amko2018117

Overview

Background:
Due to the irreplaceable role of chemotherapy in cancer treatment, research has focused on improving the efficacies of individual drugs and minimizing, or completely suppressing, their negative side effects. Based on long-term experience and the results of clinical trials, the selection of appropriate treatment is currently based on classical clinical diagnostic criteria, such as tumor size, grade, and the presence or absence of standard markers. However, complications arise due to variability between patients and tumor heterogeneity. Characterization of intratumoral heterogeneity and acquisition of more reliable drug performance indicators should improve personalized therapy. Development and selection of suitable models are therefore important issues in cancer research focused on predicting sensitivity to therapy.

Aim:
This work provides an overview of various chemosensitivity tests that have been previously employed and those that are currently used. Great emphasis is placed on comparing 2D and 3D cell culture models, since their importance and popularity are increasing. Particular attention is paid to in vivo systems, which have significantly improved recently and are tested in clinical trials to predict responses to therapy.

Conclusion:
This work provides a brief overview of chemosensitivity tests, focusing on the importance of individual tests and their application in decision-making and patient stratification to improve the clinical responses of patients and the development of targeted personalized therapy.

Key words:
cell culture techniques – personalized medicine – drug screening – biological models – tumor cell lines – carcinoma – cytotoxicity assays

This work was supported by the project MEYSNPS I-LO1413 and GACR 17-05838S.

The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study.

The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.

Submitted:
21. 9. 2017

Accepted:
20. 12. 2017


Sources

1. Workman P, Brunton VG, Robins DJ. Tyrosine kinase inhibitors. Semin Cancer Biol 1992; 3 (6): 369–381.

2. Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer 2012; 12 (4): 278–287. doi: 10.1038/nrc3236.

3. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest 2015; 125 (9): 3384–3391. doi: 10.1172/JCI80011.

4. Su YZ. Cancer chemosensitivity testing: Review. J Cancer Ther 2014; 5 (7): 672–679. doi: 10.4236/jct.2014.57 076.

5. Black MM, Speer FD. Further observations on the effects of cancer chemotherapeutic agents on the in vitro dehydrogenase activity of cancer tissue. J Natl Cancer Inst 1954; 14 (5): 1147–1158.

6. Kondo T, Imamura T, Ichihashi H. In vitro test for sensitivity of tumor to carcinostatic agents. Gan 1966; 57 (2): 113–121.

7. Ichihashi H, Sasaki S, Kondo T. Colorimetric estimation of succinic dehydrogenase activity by neotetrazolium chloride as a tumor sensitivity test to chemotherapeutic agents. Nagoya J Med Sci 1971; 33 (3): 247–256.

8. Imaizumi M. Study on the sensitivity test of carcinostatic agents by acid phosphatase activity. Nagoya J Med Sci 1972; 34 (4): 315–333.

9. Kondo T, Ichihashi H, Imaizumi M. Predication of the effect of carcinostatic agents on tumor-bearing host by the sensitivity test using acid phosphatase activity in vitro. Gan 1976; 67 (5): 633–639.

10. Volm M, Kaufmann M, Mattern J et al. Possibilities and limits of pre-therapeutic neoplasm sensitivity cytostatics tests under short-term conditions. Schweiz Med Wochenschr 1975; 105 (3): 74–82.

11. Volm M, Faufmann M, Mattern J et al. Sensitivity tests of tumors to cytostatic agents. I. Comparative investigations on transplanted tumors in vivo and in vitro. Z Krebsforsch Klin Onkol Cancer Res Clin Oncol 1975; 83 (2): 85–96.

12. Wiskemann A, Schussmann M, Rothmann D et al. In vitro and in vivo sensitivity of animal and human melanomas to various chemotherapeutical agents. Arch Dermatol Res 1978; 262 (3): 285–299.

13. Roper PR, Drewinko B. Comparison of in vitro methods to determine drug-induced cell lethality. Cancer Res 1976; 36 (7 PT 1): 2182–2188.

14. Weisenthal LM, Dill PL, Kurnick NB et al. Comparison of dye exclusion assays with a clonogenic assay in the determination of drug-induced cytotoxicity. Cancer Res 1983; 43 (1): 258–264.

15. Hamburger AW, Salmon SE. Primary bioassay of human tumor stem cells. Science 1977; 197 (4302): 461–463.

16. Kangas L, Grönroos M, Nieminen AL. Bioluminescence of cellular ATP: a new method for evaluating cytotoxic agents in vitro. Med Biol 1984; 62 (6): 338–343.

17. Sevin BU, Peng ZL, Perras JP et al. Application of an ATP-bioluminescence assay in human tumor chemosensitivity testing. Gynecol Oncol 1988; 31 (1): 191–204.

18. Sevin BU, Perras JP, Averette HE et al. Chemosensitivity testing in ovarian cancer. Cancer 1993; 71 (Suppl 4): 1613–1620.

19. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983; 65 (1–2): 55–63.

20. Carmichael J, De Graff W, Gazdar A et al. Evaluation of a tetrazolium-based semi-automatic colorimetric assay: Assessment of chemosensitivity testing. Cancer Research 1987; 47 (4): 936–942.

21. Slater TF. Studies on a succinate-neotetrazolium reductase system of rat liver. II. points of coupling with the respiratory chain. Biochim Biophys Acta 1963; 77: 365–382.

22. Denizot F, Lang R. Rapid colorimetric assay for cell growth and survival. Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J Immunol Methods 1986; 89 (2): 271–277.

23. Berridge MV, Tan AS, McCoy KD et al. The biochemical and cellular basis of cell proliferation assays that use tetrazolium salts. Biochemica 1996; 4 (1): 15–19.

24. Kiss I, Žaloudík J, Vyzula R et al. Principal clinical indications for in vitro testing of chemoresistance of tumors. Klin Onkol 2000; 13 (Speciál 2): 62–64.

25. Žaloudík J, Hajduch M, Vyzula R et al. Results of chemoresistance MTT in vitro testing in lung and colorectal cacinomas nad soft-tissue sarcomas. Klin Onkol 2000; 13 (Speciál 2): 37–38.

26. Michalova E, Poprach A, Nemeckova I et al. Chemosensitivity prediction in tumor cells ex vivo-difficulties and limitations of the method. Klin Onkol 2008; 21 (3): 93–97.

27. Carrel A. On the permanent life of tissues outside of the organism. J Exp Med 1912; 15 (5): 516–528.

28. Leighton J. A sponge matrix method for tissue culture; formation of organized aggregates of cells in vitro. J Natl Cancer Inst 1951; 12 (3): 545–561.

29. Leighton J, Kline I, Belkin M et al. The similarity in histologic appearance of some human cancer and normal cell strains in sponge-matrix tissue culture. Cancer Res 1957; 17 (5): 359–363.

30. Rotman B, Teplitz C, Dickinson K et al. Individual human tumors in short-term micro-organ cultures: chemosensitivity testing by fluorescent cytoprinting. In vitro Cell Dev Biol 1988; 24 (11): 1137–1146.

31. Vescio RA, Connors KM, Kubota T et al. Correlation of histology and drug response of human tumors grown in native-state three-dimensional histoculture and in nude mice. Proc Natl Acad Sci U S A 1991; 88 (12): 5163–5166.

32. Ashworth A, Balkwill F, Bast RC et al. Opportunities and challenges in ovarian cancer research, a perspective from the 11th Ovarian cancer action/HHMT Forum, Lake Como, March 2007. Gynecol Oncol 2008; 108 (3): 652–657. doi: 10.1016/j.ygyno.2007.11.014.

33. van der Worp HB, Howells DW, Sena ES et al. Can animal models of disease reliably inform human studies? PLoS Med 2010; 7 (3): e1000245. doi: 10.1371/journal.pmed.1000245.

34. Rabacchi SA, Neve RL, Dräger UC. A positional marker for the dorsal embryonic retina is homologous to the high-affinity laminin receptor. Development 1990; 109 (3): 521–531.

35. Hait WN. Anticancer drug development: the grand challenges. Nat Rev Drug Discov 2010; 9 (4): 253–254. doi: 10.1038/nrd3144.

36. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science 2011; 331 (6024): 1559–1564. doi: 10.1126/science.1203543.

37. Mehta G, Hsiao AY, Ingram M et al. Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. J Control Release 2012; 164 (2): 192–204. doi: 10.1016/j.jconrel.2012.04.045.

38. Hirschhaeuser F, Menne H, Dittfeld C et al. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol 2010; 148 (1): 3–15. doi: 10.1016/j.jbiotec.2010.01.012.

39. Hutmacher DW. Biomaterials offer cancer research the third dimension. Nat Mater 2010; 9 (2): 90–93. doi: 10.1038/nmat2619.

40. Hutmacher DW, Loessner D, Rizzi S et al. Can tissue engineering concepts advance tumor biology research? Trends Biotechnol 2010; 28 (3): 125–133. doi: 10.1016/ j.tibtech.2009.12.001.

41. Edmondson R, Broglie JJ, Adcock AF et al. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol 2014; 12 (4): 207–218. doi: 10.1089/adt.2014. 573.

42. Lin RZ, Chang HY. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol J 2008; 3 (9–10): 1172–1184. doi: 10.1002/biot.200700228.

43. Cui X, Hartanto Y, Zhang H. Advances in multicellular spheroids formation. J R Soc Interface 2017; 14 (127). pii: 20160877. doi: 10.1098/rsif.2016.0877.

44. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 2012; 196 (4): 395–406. doi: 10.1083/jcb.201102147.

45. Tibbitt MW, Anseth KS. Dynamic microenvironments: the fourth dimension. Sci Transl Med 2012; 4 (160): 160ps24. doi: 10.1126/scitranslmed.3004804.

46. Bremnes RM, Dønnem T, Al-Saad S et al. The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol 2011; 6 (1): 209–217. doi: 10.1097/JTO.0b013e3181f8a1bd.

47. Gurski LA, Xu X, Labrada LN et al. Hyaluronan (HA) interacting proteins RHAMM and hyaluronidase impact prostate cancer cell behavior and invadopodia formation in 3D HA-based hydrogels. PLoS One 2012; 7 (11): e50075. doi: 10.1371/journal.pone.0050075.

48. Sutherland RM. Cell and environment interactions in tumor microregions: the multicell spheroid model. Science 1988; 240 (4849): 177–184.

49. LaRue KE, Khalil M, Freyer JP. Microenvironmental regulation of proliferation in multicellular spheroids is mediated through differential expression of cyclin-dependent kinase inhibitors. Cancer Res 2004; 64 (5): 1621–1631.

50. Smalley KS, Haass NK, Brafford PA et al. Multiple signaling pathways must be targeted to overcome drug resistance in cell lines derived from melanoma metastases. Mol Cancer Ther 2006; 5 (5): 1136–1144. doi: 10.1158/1535-7163.MCT-06-0084.

51. Zhu H, Luo SF, Wang J et al. Effect of environmental factors on chemoresistance of HepG2 cells by regulating hypoxia-inducible factor-1. Chin Med J (Engl) 2012; 125 (6): 1095–1103.

52. Xu X, Sabanayagam CR, Harrington DA et al. A hydrogel-based tumor model for the evaluation of nanoparticle-based cancer therapeutics. Biomaterials 2014; 35 (10): 3319–3330. doi: 10.1016/j.biomaterials.2013.12.080.

53. Fong EL, Lamhamedi-Cherradi SE, Burdett E et al. Modeling Ewing sarcoma tumors in vitro with 3D scaffolds. Proc Natl Acad Sci U S A 2013; 110 (16): 6500–6505. doi: 10.1073/pnas.1221403110.

54. Loessner D, Stok KS, Lutolf MP et al. Bioengineered 3D platform to explore cell-ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials 2010; 31 (32): 8494–8506. doi: 10.1016/j.biomaterials.2010.07.064.

55. Karlsson H, Fryknäs M, Larsson R et al. Loss of cancer drug activity in colon cancer HCT-116 cells during spheroid formation in a new 3-D spheroid cell culture system. Exp Cell Res 2012; 318 (13): 1577–1585. doi: 10.1016/j.yexcr.2012.03.026.

56. Yamamoto S, Okochi M, Jimbow K et al. Three-dimensional magnetic cell array for evaluation of anti-proliferative effects of chemo-thermo treatment on cancer spheroids. Biotechnology and Bioprocess Engineering 2015; 20 (3): 488–497. doi: 10.1007/s12257-014-0724-y.

57. dit Faute MA, Laurent L, Ploton D et al. Distinctive alterations of invasiveness, drug resistance and cell-cell organization in 3D-cultures of MCF-7, a human breast cancer cell line, and its multidrug resistant variant. Clin Exp Metastasis 2002; 19 (2): 161–168.

58. Kondo J, Endo H, Okuyama H et al. Retaining cell-cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc Natl Acad Sci U S A 2011; 108 (15): 6235–6240. doi: 10.1073/pnas.1015938108.

59. Tung YC, Hsiao AY, Allen SG et al. High-throughput 3D spheroid culture and drug testing using a 384 hanging drop array. Analyst 2011; 136 (3): 473–478. doi: 10.1039/c0an00609b.

60. Raghavan S, Mehta P, Ward MR et al. Personalized medicine-based approach to model patterns of chemoresistance and tumor recurrence using ovarian cancer stem cell spheroids. Clin Cancer Res 2017; 23 (22): 6934–6945. doi: 10.1158/1078-0432.CCR-17-0133.

61. Lei KF, Kao CH, Tsang NM. High throughput and automatic colony formation assay based on impedance measurement technique. Anal Bioanal Chem 2017; 409 (12): 3271–3277. doi: 10.1007/s00216-017-0270-5.

62. Lei KF, Liu TK, Tsang NM. Towards a high throughput impedimetric screening of chemosensitivity of cancer cells suspended in hydrogel and cultured in a paper substrate. Biosens Bioelectron 2018; 100: 355–360. doi: 10.1016/j.bios.2017.09.029.

63. Jeppesen M, Hagel G, Glenthoj A 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. doi: 10.1371/journal.pone.0183074.

64. Kobayashi H, Tanisaka K, Doi O et al. An in vitro chemosensitivity test for solid human tumors using collagen gel droplet embedded cultures. Int J Oncol 1997; 11 (3): 449–455.

65. Hou J, Hong Z, Feng F et al. A novel chemotherapeutic sensitivity-testing system based on collagen gel droplet embedded 3D-culture methods for hepatocellular carcinoma. BMC Cancer 2017; 17 (1): 729. doi: 10.1186/s12885-017-3706-6.

66. Tanigawa N, Yamaue H, Ohyama S et al. Exploratory phase II trial in a multicenter setting to evaluate the clinical value of a chemosensitivity test in patients with gastric cancer (JACCRO-GC 04, Kubota memorial trial). Gastric Cancer 2016; 19 (2): 350–360. doi: 10.1007/s10120-015-0506-z.

67. Kanazawa Y, Yamada T, Fujita I et al. In vitro chemosensitivity test for gastric cancer specimens predicts effectiveness of oxaliplatin and 5-fluorouracil. Anticancer Res 2017; 37 (11): 6401–6405. doi: 10.21873/anticanres.12093.

68. Inoue M, Maeda H, Takeuchi Y et al. Collagen gel droplet-embedded culture drug sensitivity test for adjuvant chemotherapy after complete resection of non-small-cell lung cancer. Surg Today 2017; 48 (4): 380–387. doi: 10.1007/s00595-017-1594-7.

69. Pavía-Jiménez A, Tcheuyap VT, Brugarolas J. Establishing a human renal cell carcinoma tumorgraft platform for preclinical drug testing. Nat Protoc 2014; 9 (8): 1848–1859. doi: 10.1038/nprot.2014.108.

70. Lawson DA, Bhakta NR, Kessenbrock K et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 2015; 526 (7571): 131–135. doi: 10.1038/nature15260.

Labels
Paediatric clinical oncology Surgery Clinical oncology
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#