Skip to main content
Annals of Oncology logoLink to Annals of Oncology
editorial
. 2017 Dec 11;29(2):293–295. doi: 10.1093/annonc/mdx780

Pharmacogenomics: time to rethink its role in precision medicine

J A Willis 1, E Vilar 2,3,
PMCID: PMC6248583  PMID: 29236957

The complex genetic landscape of human cancer is evident not only across cancers from different primary sites, but also amongst cancers of the same histopathologic subtype. Understanding the contribution of this genetic landscape to relevant clinical end points such as overall survival (OS), treatment response, and toxicity has helped facilitate the evolution and application of precision clinical oncology [1, 2]. Over the past several years, specific challenges posed by genetic heterogeneity have led to the implementation of novel biomarker-based clinical trial designs for drug development, which have led to improved survival for patients with a wide variety of tumor types [3]. However, whereas many of these successful biomarker-based clinical trials have utilized somatic mutation profiling, relatively fewer studies have harnessed the area of pharmacogenomics and germline variation.

For colorectal cancer (CRC), the role of germline variation in the efficacy and toxicity of cytotoxic chemotherapy has been the subject of widespread investigation [4]. Dihydropyrimidine dehydrogenase (DPYD) gene variation is a well-established example, whereby deleterious single-nucleotide polymorphisms in DPYD have been associated with severe toxicity to 5-fluorouracil (5-FU) therapy [5, 6]. However, despite multiple lines of evidence that specific DPYD variants can reliably predict 5-FU toxicity, a number of issues currently limit pre-treatment DPYD testing from standard clinical practice, namely: regional differences in population allele frequency, technical variation in genotyping methods, and a paucity of large-scale randomized studies [7]. Germline variation in UDP-glucuronosyltransferase 1A1 (UGT1A1) presents a similar example, in which the UGT1A1*28 polymorphism is associated with an increased risk of irinotecan toxicity due to decreased drug metabolism [8–10]. As in the case for DPYD, widespread testing for UGT1A1 polymorphisms in CRC patients remains controversial. It is noteworthy that neither the National Comprehensive Cancer Network (NCCN) [11] nor European Society of Medical Oncology (ESMO) [12] guidelines currently recommend routine clinical testing of DPYD and UGT1A1 polymorphisms. This not only reflects the practical challenges of incorporating germline variability into therapeutic decision-making, but also signifies an opportunity to discover novel germline biomarkers through innovative approaches.

In this issue of Annals of Oncology, Abad and Martinez-Balibrea et al. describe the results of a rigorous multi-center study that examined the feasibility and clinical utility of using germline DNA biomarkers to select front-line chemotherapy for patients with metastatic CRC (mCRC) [13]. Using a randomized, phase II, open-label design, a total of 195 Spain-based patients with mCRC were randomized to receive either standard front-line chemotherapy with XELOX plus bevacizumab (control group, n = 61) or a genotype-driven regimen (experimental group, n = 130) selected based upon germline variants in thymidylate synthetase (TYMS, 1494del6bp) and excision repair 1, endonuclease non-catalytic subunit (ERCC1, c.354T>C). The genotype-driven regimens included XELOX plus bevacizumab, XELIRI plus bevacizumab, FUOX plus bevacizumab, and FUIRI plus bevacizumab. With respect to the efficacy of this approach, no significant difference in progression-free survival (PFS) was observed between the control and experimental group (9.4 versus 10.1 months)—and thus the primary end point of the study was not met. However, modest significant improvements were observed in response rate (control 33% versus experimental 48%) and R0 metastatic resection rate (44% versus 86%). Notably, toxicities also varied between the treatment groups, with significantly lower rates of neuropathy but higher rates of grade 3 diarrhea in the experimental group.

Thymidylate synthetase carries out a critical step in the generation and maintenance of intracellular deoxythymidylate, which in turn is necessary for DNA maintenance. As the primary intracellular target for fluoropyrimidines (such as 5-FU), it has been hypothesized that TYMS gene expression and enzymatic activity are important mediators of treatment efficacy and/or toxicity with fluoropyrimidines-based chemotherapy regimens [14–17]. The TYMS 1494del6bp variant has a global allele frequency of 49% (based on 1000 Genomes project data) and is thought to confer decreased message RNA stability by disruption of 3’ untranslated region [18, 19]. Similarly, ERCC1 is an important component of the nucleotide excision repair pathway, which recognizes DNA adducts formed by platinum-containing agents (e.g. oxaliplatin) and thus is hypothesized to modulate their potency [20–23]. ERCC1 c.354T>C is a common variant (global allele frequency 33%) that is associated with decrease mRNA expression in vitro models [24].

For CRC, the efficacies of 5-FU- and/or oxaliplatin-based treatment regimens have been well-established across (neo)adjuvant and metastatic disease settings. Thus, understanding both extrinsic and intrinsic factors that regulate TYMS and ERCC1 activity is particularly relevant for patients with mCRC. The work by Abad and Martinez-Balibrea et al. contributes additional data on the relative value of TYMS 1494del6bp and ERCC1 c.354T>C as clinically useful biomarkers in the treatment of mCRC patients with standard cytotoxic chemotherapy. Yet, as the primary end point of their study was not met, it would not be reasonable to launch future pharmacogenomic biomarker-driven trials involving solely the determination of TYMS and ERCC1 to guide therapy compared with current standards of practice with the goal to improve OS.

Broadly speaking, Abad and Martinez-Balibrea et al. demonstrate that it is technically and logistically feasible to implement a germline biomarker-driven strategy in CRC. At least two features of their study design were key to its successful implementation: (i) centralized genotyping pipelines and (ii) careful attention to regional variation in genetic substructure. However, their results again highlight important challenges and limitations facing the pharmacogenomics community. The complex interplay between clinical covariates, germline variations, and somatic alterations is likely to be a major determinant of whether a germline biomarker-driven strategy translates into improved PFS or OS. To help advance this strategy further, it is time to rethink the role of pharmacogenomics within the big picture of predictive biomarkers that are available in the space of CRC oncology. In fact, it would be helpful to integrate both worlds: germline and somatic mutation profiling into future large-scale, prospective clinical investigations. This could enable exploration of a complex combinatorial space and broaden applicability—principles that are important not only for cytotoxic therapy, but also the next generation of targeted and biological therapies.

Funding

None declared.

Disclosure

The authors have declared no conflicts of interest.

References

  • 1. Biankin AV, Piantadosi S, Hollingsworth SJ.. Patient-centric trials for therapeutic development in precision oncology. Nature 2015; 526(7573): 361.. [DOI] [PubMed] [Google Scholar]
  • 2. Sleijfer S, Bogaerts J, Siu LL.. Designing transformative clinical trials in the cancer genome era. JCO 2013; 31(15): 1834–1841. [DOI] [PubMed] [Google Scholar]
  • 3. Schwaederle M, Zhao M, Lee JJ. et al. Impact of precision medicine in diverse cancers: a meta-analysis of phase II clinical trials. JCO 2015; 33(32): 3817–3825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Panczyk M. Pharmacogenetics research on chemotherapy resistance in colorectal cancer over the last 20 years. WJG 2014; 20(29): 9775–9827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Mattison LK, Soong R, Diasio RB.. Implications of dihydropyrimidine dehydrogenase on 5-fluorouracil pharmacogenetics and pharmacogenomics. Pharmacogenomics 2002; 3(4): 485–492. [DOI] [PubMed] [Google Scholar]
  • 6. Meulendijks D, Henricks LM, Sonke GS. et al. Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity: a systematic review and meta-analysis of individual patient data. Lancet Oncol 2015; 16(16): 1639–1650. [DOI] [PubMed] [Google Scholar]
  • 7. Ciccolini J. DPD deficiency in patients treated with fluorouracil. Lancet Oncol 2015; 16(16): 1574–1576. [DOI] [PubMed] [Google Scholar]
  • 8. O'Dwyer PJ, Catalano RB.. Uridine diphosphate glucuronosyltransferase (UGT) 1A1 and irinotecan: practical pharmacogenomics arrives in cancer therapy. J Clin Oncol 2006; 24: 4534–4538. [DOI] [PubMed] [Google Scholar]
  • 9. Innocenti F, Undevia SD, Iyer L. et al. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 2004; 22(8): 1382–1388. [DOI] [PubMed] [Google Scholar]
  • 10. Toffoli G, Cecchin E, Gasparini G. et al. Genotype-driven phase I study of irinotecan administered in combination with fluorouracil/leucovorin in patients with metastatic colorectal cancer. JCO 2010; 28(5): 866–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. National Comprehensive Cancer Network. Colon Cancer (Version 3.2017). https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf (27 November 2017, date last accessed).
  • 12. Van Cutsem E, Cervantes A, Adam R. et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 2016; 27(8): 1386–1422. [DOI] [PubMed] [Google Scholar]
  • 13. Abad A, Martínez-Balibrea E, Viéitez JM. et al. Genotype-based selection of treatment for patients with advanced colorectal cancer (SETICC): a pharmacogenetic-based randomized phase II trial. Ann Oncol 2018; 29(2): 439–444. [DOI] [PubMed] [Google Scholar]
  • 14. Johnston PG, Fisher ER, Rockette HE. et al. The role of thymidylate synthase expression in prognosis and outcome of adjuvant chemotherapy in patients with rectal cancer. JCO 1994; 12(12): 2640–2647. [DOI] [PubMed] [Google Scholar]
  • 15. Johnston PG, Lenz H-J, Leichman CG. et al. Thymidylate synthase gene and protein expression correlate and are associated with response to 5-fluorouracil in human colorectal and gastric tumors. Cancer Res 1995; 55: 1407–1412. [PubMed] [Google Scholar]
  • 16. Lenz HJ, Danenberg KD, Leichman CG. et al. p53 and thymidylate synthase expression in untreated stage II colon cancer: associations with recurrence, survival, and site. Clin Cancer Res 1998; 4: 1227–1234. [PubMed] [Google Scholar]
  • 17. Tan BR, Thomas F, Myerson RJ. et al. Thymidylate synthase genotype-directed neoadjuvant chemoradiation for patients with rectal adenocarcinoma. JCO 2011; 29(7): 875–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lurje G, Manegold PC, Ning Y. et al. Thymidylate synthase gene variations: predictive and prognostic markers. Mol Cancer Ther 2009; 8: 1000–1007. [DOI] [PubMed] [Google Scholar]
  • 19. The Genomes Project C. A global reference for human genetic variation. Nature 2015; 526: 68.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Martinez-Balibrea E, Abad A, Aranda E. et al. Pharmacogenetic approach for capecitabine or 5-fluorouracil selection to be combined with oxaliplatin as first-line chemotherapy in advanced colorectal cancer. Eur J Cancer 2008; 44(9): 1229–1237. [DOI] [PubMed] [Google Scholar]
  • 21. Viguier J, Boige V, Miquel C. et al. ERCC1 Codon 118 polymorphism is a predictive factor for the tumor response to oxaliplatin/5-fluorouracil combination chemotherapy in patients with advanced colorectal cancer. Clin Cancer Res 2005; 11: 6212–6217. [DOI] [PubMed] [Google Scholar]
  • 22. de las Peñas R, Sanchez-Ronco M, Alberola V. et al. Polymorphisms in DNA repair genes modulate survival in cisplatin/gemcitabine-treated non-small-cell lung cancer patients. Ann Oncol 2006; 17(4): 668–675. [DOI] [PubMed] [Google Scholar]
  • 23. Tan L-M, Qiu C-F, Zhu T. et al. Genetic polymorphisms and platinum-based chemotherapy treatment outcomes in patients with non-small cell lung cancer: a genetic epidemiology study based meta-analysis. Sci Rep 2017; 7(1): 5593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Yu JJ, Lee KB, Mu C. et al. Comparison of two human ovarian carcinoma cell lines (A2780/CP70 and MCAS) that are equally resistant to platinum, but differ at codon 118 of the ERCC1 gene. Int J Oncol 2000; 16: 555–560. [DOI] [PubMed] [Google Scholar]

Articles from Annals of Oncology are provided here courtesy of Oxford University Press

RESOURCES