• Non ci sono risultati.

FUTURE PERSPECTIVES

Novel approaches in pharmacogenetic studies

During the past decades, several studies based on DNA/RNA microarrays have tried to develop predictive signatures to decide the most suitable chemotherapy approach for specific types or subtypes of tumors. Conflicting results and difficulties in replicating the data led to a limited impact on clinical practice, mainly due to: i) lack of sufficiently powered clinical trials; ii) use of sample collection and data analysis methods that are not standardized; iii) intrinsic intratumor heterogeneity.

However, in the last few years, technical advancements in the development of rapid whole-genome studies have initiated a revolution in the field that could potentially change the pharmacogenomic approach to perzonalized treatment in the future. These techniques are represented mainly by massively parallel or next‑generation sequencing (NGS) and GWASs.

During recent decades, thanks to the introduction of advanced systems that may generate millions of short DNA reads (36–150 base pairs), the landscape of DNA sequencing has evolved from f irst-generation sequencing, which was based on DNA sequencing using dideoxynucleotide termination chemistry, to second-generation sequencing or NGS [74]. Noncoding regions of DNA/RNA, in addition to exons, can be analyzed at very high resolution in a short time by NGS with the possibility of identifying any chromosomal rearrangement and variations in copy-number. Furthermore, thanks to the progression in the development of whole-genome amplification, an insight into intratumor heterogeneity has become feasible by performing genetic studies even at the singlecell level [75]. Hopefully, with the diffusion of NGS methodologies in the next few years, we will develop an increasing knowledge of tumor biology.

GWASs are genetic studies that allow the simultaneous evaluation of many polymorphisms, especially common copy-number variations and SNPs, in order to identify existing correlations among any genetic variant and tumor features. In particular, the application of these types of studies in pharmacogenomics aims to find out any possible correlation between polymorphisms and drug response or toxicity. Of note, GWASs allow the investigation of the entire genome independently from preliminary information on chromosomal loci involvement in drug effects. The massive amount of SNPs analyzed with GWASs and the difficulty represented by the definition of real correlations are the limitations of this method, increasing the risk of a high rate of false-positive data; a massive number of samples is necessary to obtain a sufficient statistical power. A recent study has been published by University of Texas MD Anderson researchers (TX, USA), reporting a genome-wide scan of 307,260 polymorphisms in 327 NSCLC patients treated with cisplatin or carboplatin [76]. In this analysis, the rs1878022 variant, which is located within an intron of the gene CMKLR1, has been strongly correlated with shorter survival. The findings of this study were also validated in two other independent cohorts of 315 patients enrolled at the Mayo Clinic (USA) and 420 cases recruited in the Spanish Lung Cancer Group PLATAX clinical trials. Interestingly, the use of this unbiased non-candidate gene-driven approach led to the identification as strong predictive survival factor of a polymorphic variant within an intronic region of a platinum-unrelated gene.

In order to validate these pharmacogenetic findings and use them in the clinical setting, future genomic analysis should use validated and standardized methodology. Moreover, future analysis should be performed on larger cohorts of patients and/or utilize the sharing of data through common websites in order

to create ‘virtual multicentric pharmacogenetic trials’.

Clinical relevance

During recent decades, a number of important genetic determinants have been identified in different tumor types, including NSCLC. The FDA has approved a number of genetic tests and some pharmacogenetic markers have been included in drug labels [102]. However, the application of pharmacogenetic testing in primary care is not as common as predicted. This leads to questions regarding the clinical relevance of pharmacogenetic analyses and on future studies that should successfully bridge the gap from research to clinical practice.

Unfortunately, the clinical relevance of most of the polymorphisms described in the current literature remains controversial because of the exiguous number of (heterogeneous) patients screened, the lack of prospective studies designed to identify specific correlations between gene alterations and drug response or disease prognosis, and the strong variability in analytical analysis, as well as lack of quality controls among different studies.

The development of prospective clinical trials, in which patient treatments selected on the basis of conventional criteria are directly compared with patients given specific treatments suggested by genetic characteristics, might overcome these controversies and reduce the reluctance to transfer the novel knowledge into the clinical routine. Another obstacle to the successful clinical application of the new findings is the lack of association with the main pharmacokinetic/pharmacodynamic parameters as well as functional genomic analysis of the underlying mechanisms of drug sensitivity/resistance. The recent discovery of miRNA has improved our knowledge regarding the control of gene expression, and in the future it may help to clarify the role of the signaling pathways involved in lung cancer oncogenesis. Hopefully, growing evidence relating to miRNA may provide a clue to solve individual drug-resistance problems [77].

In conclusion, despite the intriguing findings of several studies, small sample sizes together with interethnic differences and the retrospective nature of most studies make it difficult to draw any clear conclusions regarding the role of most pharmacogenetic biomarkers in determining the clinical outcome or toxicity in gefitinib and erlotinib treatment. Hopefully, the accurate planning of new prospective trials, the increased knowledge of key mechanisms affecting drug distribution/activity and the use of novel technologies, including genome-wide approaches, may provide essential tools to improve the prospects of pharmacogenetic research, as well as to optimize currently available treatments in selected NSCLC patients.

REFERENCES

1. Galvani E, Peters GJ, Giovannetti E. Thymidylate synthase inhibitors for non-small cell lung cancer. Expert Opin Investig. Drug 20(10), 1343–1356 (2011).

2. Reck M, von Pawel J, Zatloukal P et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer. J. Clin. Oncol. 27(8), 1227–1234 (2009).

3. Scagliotti GV, Parikh P, von Pawel J et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J. Clin. Oncol. 26(21), 3543–3551 (2008).

4 Druker BJ, Talpaz M, Resta DJ et al. Efficacy and safety of a specific inhibitor of the BCR–ABL tyrosine kinase in chronic myeloid leukemia. N. Engl. J. Med. 344(14), 1031–1037 (2001).

5. Baselga J. Targeting tyrosine kinases in cancer: the second wave. Science 312(5777), 1175–1178 (2006).

6. Hoang T, Schiller JH. Advanced NSCLC: from cytotoxic systemic chemotherapy to molecularly targeted therapy. Expert Rev.

Anticancer Ther. 2(4), 393–401 (2002).

7. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science 298(5600), 1912–1934 (2002).

8. Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat. Rev. Cancer 5(5), 341–354 (2005).

9. Suzuki S, Dobashi Y, Sakurai H, Nishikawa K, Hanawa M, Ooi A. Protein overexpression and gene amplification of epidermal growth factor receptor in nonsmall cell lung carcinomas. An immunohistochemical and fluorescence in situ hybridization study.

Cancer 103(6), 1265–1273 (2005).

10. Hirsch FR, Varella-Garcia M, Bunn PA et al. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J. Clin. Oncol. 21(20), 3798–3807 (2003).

11. Kris MG, Natale RB, Herbst RS et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 290(16), 2149–2158 (2003).

12. Lynch TJ, Bell DW, Sordella R et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 350(21), 2129–2139 (2004).

13. Tsao MS, Sakurada A, Cutz JC et al. Erlotinib in lung cancer – molecular and clinical predictors of outcome. N. Engl. J. Med.

353(2), 133–144 (2005).

14. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer 7(3), 169–181 (2007).

15. Danesi R, De Braud F, Fogli S, Di Paolo A, Del Tacca M. Pharmacogenetic determinants of anti-cancer drug activity and toxicity.

Trends Pharmacol. Sci. 22(8), 420–426 (2001).

16. Barker AJ, Gibson KH, Grundy W et al. Studies leading to the identification of ZD1839 (IRESSA): an orally active, selective epidermal growth factor receptor tyrosine kinase inhibitor targeted to the treatment of cancer. Bioorg. Med. Chem. Lett. 11(14), 1911–1914 (2001).

17. Normanno N, Maiello MR, De Luca A. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs): simple drugs with a complex mechanism of action? J. Cell. Physiol. 194(1), 13–19 (2003).

18. Scagliotti GV, Selvaggi G, Novello S, Hirsch FR. The biology of epidermal growth factor receptor in lung cancer. Clin. Cancer Res.

10(12 Pt 2), 4227s–4232s (2004).

19. Kim ES, Hirsh V, Mok T et al. Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): a randomised Phase III trial. Lancet 372(9652), 1809–1818 (2008).

20. Shepherd FA, Rodrigues Pereira J, Ciuleanu T et al. Erlotinib in previously treated non-small cell lung cancer. N. Engl. J. Med.

353(2), 123–132 (2005).

21. Mok TS, Wu YL, Thongprasert S et al. Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 361(10), 947–957 (2009).

22. Helfrich BA, Raben D, Varella-Garcia M et al. Antitumor activity of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor gefitinib (ZD1839, Iressa) in non-small cell lung cancer cell lines correlates with gene copy number and EGFR mutations but not EGFR protein levels. Clin. Cancer Res. 12(23), 7117–7125 (2006).

23. Hartmann JT, Haap M, Kopp HG, Lipp HP. Tyrosine kinase inhibitors – a review on pharmacology, metabolism and side effects.

Curr. Drug Metab. 10(5), 470–481 (2009).

24. Nomura M, Shigematsu H, Li L et al. Polymorphisms, mutations, and amplification of the EGFR gene in non-small cell lung cancers. PLoS Med. 4(4), e125 (2007).

25. Moriai T, Kobrin MS, Hope C, Speck L, Korc M. A variant epidermal growth factor receptor exhibits altered type a transforming growth factor binding and transmembrane signaling. Proc. Natl Acad. Sci. USA 91(21), 10217–10221 (1994).

26. Liu W, Innocenti F, Wu MH et al. A functional common polymorphism in a Sp1 recognition site of the epidermal growth factor receptor gene promoter. Cancer Res. 65(1), 46–53 (2005).

27. Liu G, Gurubhagavatula S, Zhou W et al. Epidermal growth factor receptor polymorphisms and clinical outcomes in non-small-cell lung cancer patients treated with gefitinib. Pharmacogenomics J. 8(2), 129–138 (2008).

28. Ichihara S, Toyooka S, Fujiwara Y et al. The impact of epidermal growth factor receptor gene status on gefitinib-treated Japanese patients with non-small-cell lung cancer. Int. J. Cancer 120(6), 1239–1247 (2007).

29. Gregorc V, Hidalgo M, Spreafico A et al. Germline polymorphisms in EGFR and survival in patients with lung cancer receiving gefitinib. Clin. Pharmacol. Ther. 83(3), 477–484 (2008).

30. Gebhardt F, Bürger H, Brandt B. Modulation of EGFR gene transcription by secondary structures, a polymorphic repetitive sequence and mutations – a link between genetics and epigenetics. Histol. Histopathol. 15(3), 929–936 (2000).

31. Amador ML, Oppenheimer D, Perea S et al. An epidermal growth factor receptor intron 1 polymorphism mediates response to epidermal growth factor receptor inhibitors. Cancer Res. 64(24), 9139–9143 (2004).

32. Gebhardt F, Zänker KS, Brandt B. Modulation of epidermal growth factor receptor gene transcription by a polymorphic dinucleotide repeat in intron 1. J. Biol. Chem. 274(19), 13176–13180 (1999).

33. Buerger H, Gebhardt F, Schmidt H et al. Length and loss of heterozygosity of an intron 1 polymorphic sequence of egfr is related to cytogenetic alterations and epithelial growth factor receptor expression. Cancer Res. 60(4), 854–857 (2000).

34. Han SW, Jeon YK, Lee KH et al. Intron 1 CA dinucleotide repeat polymorphism and mutations of epidermal growth factor receptor and gefitinib responsiveness in non-small-cell lung cancer. Pharmacogenet. Genom. 17(5), 313–319 (2007).

35. Ma F, Sun T, Shi Y et al. Polymorphisms of EGFR predict clinical outcome in advanced non-small-cell lung cancer patients treated with gefitinib. Lung Cancer 66(1), 114–119 (2009).

36. Dubey S, Stephenson P, Levy DE et al. EGFR dinucleotide repeat polymorphism as a prognostic indicator in non-small cell lung cancer. J. Thorac. Oncol. 1(5), 406–412 (2006).

37. Nie Q, Wang Z, Zhang GC et al. The epidermal growth factor receptor intron1 (CA) n microsatellite polymorphism is a potential predictor of treatment outcome in patients with advanced lung cancer treated with gefitinib. Eur. J. Pharmacol. 570(1–3), 175–181 (2007).

38. Tiseo M, Capelletti M, De Palma G et al. Epidermal growth factor receptor intron-1 polymorphism predicts gefitinib outcome in advanced non-small cell lung cancer. J. Thorac. Oncol. 3(10), 1104–1111 (2008).

39. Tiseo M, Rossi G, Capelletti M et al. Predictors of gefitinib outcomes in advanced non-small cell lung cancer (NSCLC): study of a comprehensive panel of molecular markers. Lung Cancer 67(3), 355–360 (2010).

40. Giovannetti E, Zucali PA, Peters GJ et al. Association of polymorphisms in AKT1 and EGFR with clinical outcome and toxicity in non-small cell lung cancer patients treated with gefitinib. Mol. Cancer Ther. 9(3), 581–593 (2010).

41. Liu G, Cheng D, Ding K et al. Pharmacogenetic analysis of BR.21, a placebo-controlled randomized Phase III clinical trial of erlotinib in advanced non-small cell lung cancer. J. Thorac. Oncol. 7(2), 316–322 (2012).

42. Tzeng CWD, Frolov A, Frolova N et al. Pancreatic cancer epidermal growth factor receptor (EGFR) intron 1 polymorphism influences postoperative patient survival and in vitro erlotinib response. Ann. Surg. Oncol. 14(7), 2150–2158 (2007).

43. Frolov A, Liles JS, Kossenkov AV et al. Epidermal growth factor receptor (EGFR) intron 1 polymorphism and clinical outcome in pancreatic adenocarcinoma. Am. J. Surg. 200(3), 398–405 (2010).

44. Cappuzzo F, Magrini E, Ceresoli GL et al. Akt phosphorylation and gefitinib efficacy in patients with advanced non-small-cell lung cancer. J. Natl Cancer Inst. 96(15), 1133–1141 (2004).

45. Cappuzzo F, Ligorio C, Jänne PA et al. Prospective study of gefitinib in epidermal growth factor receptor fluorescence in situ hybridization-positive/phospho-Akt-positive or never smoker patients with advanced non-small-cell lung cancer: the ONCOBELL trial. J. Clin. Oncol. 25(16), 2248–2255 (2007).

46. Kim MS, Jeong EG, Yoo NJ, Lee SH. Mutational analysis of oncogenic AKT E17K mutation in common solid cancers and acute leukaemias. Br. J. Cancer 98(9), 1533–1535 (2008).

47. Harris SL, Gil G, Robins H et al. Detection of functional single-nucleotide polymorphisms that affect apoptosis. Proc. Natl Acad.

Sci. USA 102(45), 16297–16302 (2005).

48. Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA. Convergent evidence for impaired AKT1–GSK3b signaling in schizophrenia. Nat. Genet. 36(2), 131–137 (2004).

49. Hildebrandt MAT, Yang H, Hung MC et al. Genetic variations in the PI3K/PTEN/AKT/mTOR pathway are associated with clinical outcomes in esophageal cancer patients treated with chemoradiotherapy. J. Clin. Oncol. 27(6), 857–871 (2009).

50. Kim MJ, Kang HG, Lee SY et al. AKT1 polymorphisms and survival of early stage non-small cell lung cancer. J. Surg. Oncol.

105(2), 167–174 (2012).

51. van Erp NP, Gelderblom H, Guchelaar HJ. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat. Rev. 35(8), 692–

706 (2012).

52. Lemos C, Jansen G, Peters GJ. Drug transporters: recent advances concerning BCRP and tyrosine kinase inhibitors. Br. J. Cancer 98(5), 857–862 (2008).

53. Li J, Cusatis G, Brahmer J et al. Association of variant ABCG2 and the pharmacokinetics of epidermal growth factor receptor tyrosine kinase inhibitors in cancer patients. Cancer Biol. Ther. 6(3), 432–438 (2007).

54. Lemos C, Giovannetti E, Zucali PA et al. Impact of ABCG2 polymorphisms on the clinical outcome and toxicity of gefitinib in non-small-cell lung cancer patients. Pharmacogenomics 12(2), 159–170 (2011).

55. Olaussen KA, Dunant A, Fouret P et al. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N. Engl. J. Med. 355(10), 983–991 (2006).

56. Isla D, Sarries C, Rosell R et al. Single nucleotide polymorphisms and outcome in docetaxel–cisplatin-treated advanced non-small-cell lung cancer. Ann. Oncol. 15(8), 1194–1203 (2004).

57. Zhou W, Gurubhagavatula S, Liu G et al. Excision repair cross-complementation group 1 polymorphism predicts overall survival in advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Clin. Cancer Res. 10(15), 4939–4943 (2004).

58. Gazdar AF. DNA repair and survival in lung cancer – the two faces of Janus. N. Engl. J. Med. 356(8), 771–773 (2007).

59. Rosell R, Felip E, Taron M et al. Gene expression as a predictive marker of outcome in stage IIB–IIIA–IIIB non-small cell lung cancer after induction gemcitabine-based chemotherapy followed by resectional surgery. Clin. Cancer Res. 10(12 Pt 2), 4215s–

4219s (2004).

60. Masson M, Niedergang C, Schreiber V, Muller S, Menissier-de Murcia J, de Murcia G. XRCC1 is specifically associated with poly(ADP-ribose) polymerase and negatively regulates its activity following DNA damage. Mol. Cell. Biol. 18(6), 3563–3571 (1998).

61. Han JY, Yoon KA, Park JH et al. DNA repair gene polymorphisms and benefit from gefitinib in never-smokers with lung adenocarcinoma. Cancer 117(14), 3201–3208 (2011).

62. Peréz-Soler R, Saltz L. Cutaneous adverse effects with HER1/EGFR-targeted agents: is there a silver lining? J. Clin. Oncol.

23(22), 5235–5246 (2005).

63. Huang CL, Yang CH, Yeh KH et al. EGFR intron 1 dinucleotide repeat polymorphism is associated with the occurrence of skin rash with gefitinib treatment. Lung Cancer 64(3), 346–351 (2009).

64. Rudin CM, Liu W, Desai A et al. Pharmacogenomic and pharmacokinetic determinants of erlotinib toxicity. J. Clin. Oncol. 26(7), 1119–1127 (2008).

65. Wang WS, Chen PM, Chiou TJ et al. Epidermal growth factor receptor R497K polymorphism is a favorable prognostic factor for patients with colorectal carcinoma. Clin. Cancer Res. 13(12), 3597–3604 (2007).

66. Pitari GM, Zingman LV, Hodgson DM et al. Bacterial enterotoxins are associated with resistance to colon cancer. Proc. Natl Acad.

Sci. USA 100(5), 2695–2699 (2003).

67. Cusatis G, Gregorc V, Li J et al. Pharmacogenetics of ABCG2 and adverse reactions to gefitinib. J. Natl Cancer Inst. 98(23), 1739–

1742 (2006).

68. Nyberg F, Barratt BJ, Mushiroda T et al. Interstitial lung disease in gefitinib-treated Japanese patients with non-small-cell lung cancer: genome-wide analysis of genetic data. Pharmacogenomics 12(7), 965–975 (2011).

69. Liu W, Wu X, Zhang W et al. Relationship of EGFR mutations, expression, amplification, and polymorphisms to epidermal growth factor receptor inhibitors in the NCI60 cell lines. Clin. Cancer Res. 13(22 Pt 1), 6788–6795 (2007).

70. Brugger W, Triller N, Blasinska-Morawiec M et al. Prospective molecular marker analyses of EGFR and KRAS from a randomized, placebo-controlled study of erlotinib maintenance therapy in advanced non-small cell lung cancer. J. Clin. Oncol. 29(31), 4113–

4120 (2011).

71. Rodriguez-Antona C, Ingelman-Sundberg M. Cytochrome P450 pharmacogenetics and cancer. Oncogene 25(11), 1679–1691 (2006).

72. Li J, Zhao M, He P, Hidalgo M, Baker SD. Differential metabolism of gefitinib and erlotinib by human cytochrome P450 enzymes.

Clin. Cancer Res. 13(12), 3731–3737 (2007).

73. Hamada A, Sasaki J, Saeki S et al. Association of ABCB1 polymorphisms with erlotinib pharmacokinetics and toxicity in Japanese patients with non-small-cell lung cancer. Pharmacogenomics 13(5), 615–624 (2012).

74. Metzker ML. Sequencing technologies – the next generation. Nat. Rev. Genet. 11(1), 31–46 (2010).

75. Navin N, Kendall J, Troge J et al. Tumour evolution inferred by single-cell sequencing. Nature 472(7341), 90–94 (2011).

76. Wu X, Ye Y, Rosell R et al. Genome-wide association study of survival in non-small cell lung cancer patients receiving platinumbased chemotherapy. J. Natl Cancer Inst. 103(10), 817–825 (2011).

77. Giovannetti E, Erozenci A, Smit J, Danesi R, Peters GJ. Molecular mechanisms underlying the role of microRNAs (miRNAs) in anticancer drug resistance and implications for clinical practice. Crit. Rev. Oncol. Hematol. 81(2), 103–122 (2012).

78. Maemondo M, Inoue A, Kobayashi K et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N. Engl.

J. Med. 362(25), 2380–2388 (2010).

79. Mitsudomi T, Morita S, Yatabe Y et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised Phase 3 trial. Lancet Oncol. 11(2), 121–128 (2010).

Websites

101. National Cancer Institute at the NIH. FDA Approval for Erlotinib Hydrochloride. www.cancer.gov/cancertopics/druginfo/fda-erlotinib-hydrochloride

102. US FDA. Table of Pharmacogenomic Biomarkers in Drug Labels.

www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/uc

Chapter 5

Molecular mechanisms underlying