Predictive and Prognostic markers in the treatment of metastatic colorectal cancer (mCRC) – personalized medicine at work –




This article clarifies prognostic and predictive markers in the treatment of colorectal cancer. Multiple chemotherapeutic drugs are approved for metastatic colorectal cancer (mCRC), but available guidelines are often not helpful in directing drug selections. It would be desirable to define patient populations before chemotherapy by biomarkers that predict outcome and toxicities. RAS mutational evaluation remains the only established biomarker analysis in the treatment of mCRC. BRAF mutant tumors are associated with poor outcome. Chemotherapeutic combination therapies still remain the most active treatments in the armamentarium, and future trials should address the need to prospectively investigate and validate biomarkers.










5-FU remains the most used and most active drug in the treatment of mCRC. The antimetabolite 5-FU is a pyrimidine analogue that inhibits thymidylate synthase (TS) irreversibly. The successive lack of thymidine triphosphate inhibits DNA replication, resulting in cell death of rapid proliferative cells. With the exception of the monotherapeutic use of regorafenib and anti-EGFR antibodies cetuximab or panitumumab in further-line treatment, all regimens contain 5-FU or one of the orally available 5-FU prodrugs capecitabine, UFT (tegafur/uracil), or S1 (tegafur/gimeracil/oteracil). 5-FU is metabolized more than 80% in the liver by dihydropyrimidine dehydrogenase (DPD). TS and DPD have been extensively investigated in regard to their predictive value for 5-FU treatment.


A relative DPD deficiency with a higher possibility of grade 3/4 toxicity exists in 3% to 5% of patients, but a complete DPD deficiency causing serious adverse events is rare, with a frequency of 0.1%. Single-nucleotide polymorphisms (SNPs) within the DPD are responsible for the variability of DPD activity and 5-FU toxicity. Several studies demonstrated a correlation between DPD tumor expression and chemotherapeutic efficacy, and recently it has been shown that SNPs within the DPD may also predict outcome. However, as of now DPD is predictive for 5-FU toxicity rather than its efficacy.


TS is the primary target of 5-FU and has been extensively studied as a predictor for 5-FU efficacy. High intratumor TS expression was correlated with shorter OS in a large meta-analysis of 20 studies in both adjuvant and metastatic CRC. Cutoff differences and unresolved challenges to implement sound cross-center stable measurements of intratumor RNA expression levels make a clinical implementation difficult. SNPs to the TS gene are another approach to investigate TS as a predictive marker. SNPs are easy, inexpensive, and quick to access. Several SNPs in the TS gene have been described to be of potentially predictive value, but validation studies are lacking. To distinguish between the predictive and possible prognostic value of TS expression a control arm not containing 5-FU would be necessary, which is impossible to find in the treatment of mCRC.


No validated predictive marker for the use of 5-FU has been established as yet. DPD deficiency remains to be predictive for toxicity, although routine measurement of DPD is not current practice.


Oxaliplatin


Oxaliplatin forms inter-DNA and intra-DNA cross-links, which disrupt DNA replication and result in cell death. Oxaliplatin has essentially no single-agent activity in mCRC but is used in a variety of combinations with 5-FU (FUFOX, FOLFOX) or irinotecan (IrOx). DNA cross-links can be removed by the base excision repair system. One of the key components representing the rate-limiting process of the excision repair system is the excision repair cross-complementing group 1 (ERCC1) enzyme. The data on ERCC1 intratumor expression and efficacy in oxaliplatin-based chemotherapy is contradictory. In one first-line study low ERCC1 expression was associated with longer survival ; however, the CAIRO study investigating 506 primary tumor samples in patients treated with oxaliplatin in second and third line after CAPIRI or capecitabine first-line therapy was unable to confirm this observation, possibly because of changes in ERCC1 expression during first-line therapy. SNPs in ERCC1 have been tested for their predictive value but, again because of the lack of validation studies and controls, none has made it into clinical practice. The importance of intratumor ERCC1 levels for oxaliplatin efficacy and resistance is underlined by data showing that ERCC1 and DPD is upregulated in CRC samples after oxaliplatin-based first-line therapy for mCRC.


Recently the randomized phase II MAVERICC trial ( NCT01374425 ), prospectively testing ERCC1 expression as a biomarker for FOLFIRI or FOLFOX first-line treatment of mCRC, has completed recruitment. With a planned recruitment of 360 patients, this will be the first study prospectively stratifying for a biomarker in the treatment of mCRC.


Detoxification of oxaliplatin depends on glutathione S -transferase family members. SNPs have been shown to be of predictive value for oxaliplatin-dependent neurotoxicity and efficacy, but again validation studies are lacking so the value for clinical decision making is unclear.


Irinotecan


Irinotecan is hydrolyzed to SN38, which is a topoisomerase 1 (TOPO1) inhibitor. Inhibition of TOPO1 leads to inhibition of DNA replication and transcription by inducing single- and double-strand breaks and DNA fragmentation. SN38 is detoxified by uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Genetic variants of UGT1A1 are associated with enzyme activity and irinotecan toxicity in regimens using higher doses of irinotecan. Irinotecan has shown single-agent activity after 5-FU failure, but is usually administered in combination with anti-EGFR therapy in further therapy lines or with 5-FU in first-line and second-line regimens. Defined by the mechanism of action and detoxification, biomarker development has been focused on TOPO1 and UGT1A1.


In UGT1A1, owing to the known toxicity issues, research on the predictive value has focused on SNPs. However, published results have been controversial, and it is hypothesized that an SNP signature might be predictive for irinotecan use.


The data on TOPO1 expression for prediction of irinotecan efficacy are controversial. The United Kingdom–based FOCUS trial tested 822 samples successfully for TOPO1 expression by immunohistochemistry. A high TOPO1 expression was associated with a significantly longer survival in both irinotecan-treated and oxaliplatin-treated patients, with a higher predictive value for irinotecan-treated patients. These data were confirmed in a smaller trial using fluorescence in situ hybridization, but the association with oxaliplatin use is not understood and a prospective trial is needed.


Aprataxin (APTX) is a gene involved in the repair of DNA strand breaks as caused by irinotecan. The immunohistochemical expression of APTX has suggested it to be of predictive value for irinotecan treatment in CRC, but no confirmation study has been conducted thus far ( Table 2 ).



Table 2

Overview of important predictive and prognostic factors in the treatment of metastatic colorectal cancer (mCRC)




















































































Pathway Marker Value Clinical Conclusion
EGFR


  • RAS mutation in KRAS and NRAS




    • Exon 2 (codons 12, 13)



    • Exon 3 (codons 59, 61)



    • Exon 4 (codons 117, 146)


Negative predictive for the use of cetuximab and panitumumab Established biomarker in the treatment of mCRC
EGFR BRAF mutation Negative prognostic value
Possible negative predictive value for cetuximab and panitumumab
FOLFOXIRI may be discussed in a certain subpopulation
EGFR PIK3CA mutation Negative predictive value for the use of cetuximab and panitumumab is discussed but not validated Not a validated biomarker, no clinical conclusion
EGFR PTEN loss
PTEN mutations
Negative predictive value has been discussed for both mutations and loss of expression Not a validated biomarker, no clinical conclusion
EGFR AREG
EREG
Higher expression levels are associated with increased response to anti-EGFR therapy with cetuximab Methodological problems in measuring expression levels and cutoff calculations are ongoing. Not to be used in clinical practice
VEGF VEGF-A plasma levels Higher VEGF-A plasma levels predict increase in bevacizumab efficacy Methodological problems. Not to be used in clinical practice
VEGF IL-8 Bevacizumab predictor as a VEGF-A independent proangiogenic factor Methodological problems. Not to be used in clinical practice
VEGF SNP Predictive VEGF SNPs and VEGFR SNPs for bevacizumab. Easy to measure but not validated. Not to be used in clinical practice
TS TS expression Predictive for 5-FU. Higher TS expression correlated significant in adjuvant and mCRC with shorter OS Methodological problems. Not to be used in clinical practice. A 5-FU–free control arm is difficult to create
DPD DPD deficiency Complete deficiency is rare (0.1%) but even relative deficiency due to polymorphisms (3%–5%) are predictive for 5-FU toxicity Screening is not recommended
BER ERCC1 expression level ERCC1 expression levels are predictive for oxaliplatin response in first-line treatment. Data on second and third-line treatment was inconclusive. Results from a prospective randomized phase II trial are awaited. Not to be used in clinical practice as yet
GSTP1 SNP SNPs have been predictive for oxaliplatin induced neurotoxicity Easy to measure but not validated. Not to be used in clinical practice
TOPO1 TOPO1 expression Higher TOPO1 expression has been associated with longer OS in irinotecan and oxaliplatin-treated patients Methodological problems. Not to be used in clinical practice
UGT1A1 UGT1A1 SNPs Genetic variations of UGT1A1 are predictive for irinotecan toxicity Test is recommended in the USA. Regimen with lower irinotecan dosing do not have UGT1A1-related toxicity problems
Aprataxin APTX expression APTX is involved in repairing irinotecan-related DNA strand breaks Not validated in a trial with a higher number of patients. Not to be used in clinical practice

Abbreviations: 5-FU, 5-fluorouracil; APTX, aprataxin; AREG, amphiregulin; DPD, dihydropyrimidine dehydrogenase; EGFR, epidermal growth factor receptor; EREG, epiregulin; FOLFOXIRI, leucovorin, 5-FU, oxaliplatin, irinotecan; IL-8, interleukin-8; OS, overall survival; PTEN, phosphatase and tensin homologue; SNP, single-nucleotide polymorphism; TPO1, topoisomerase 1; TS, thymidylate synthase; UGT1A1, uridine diphosphate glucuronosyltransferase 1A1; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor.


Summary


Extended RAS analysis, including mutations of KRAS exons 2, 3, and 4 and NRAS exons 2, 3, and 4, defines the subpopulation of patients that most likely benefit from anti-EGFR treatment. RAS currently remains the only established predictive biomarker in the treatment of mCRC. It only has negative predictive value, but should be assessed at first diagnosis of metastatic disease. BRAF mutations are the second biomarker that may be tested for. Patients with an excellent performance status and a BRAF mutant tumor can be considered for FOLFOXIRI therapy, as this has shown remarkable outcomes in a small phase II trial in patients bearing a BRAF mutant tumor. Several recent studies have suggested that gene-expression levels of EREG and AREG may be predictive biomarkers for EGFR inhibitors, although the validation and standardization of the technologies have limited the implementation of these biomarkers in the clinic. The mechanisms of action of anti-VEGF are too diversified, and no predictive factor has yet been identified or validated. New approaches looking for predictive proteins in the plasma are promising, helping to define the population that benefits from anti-VEGF treatment. New techniques such as NGS-based sequencing and whole-genome sequencing are becoming more widely used and affordable, and may help in the near future to detect novel biomarkers and guide treatment decisions. Although biomarker development during the past years has focused on targeted drugs, the most active substances in mCRC treatment remain cytotoxic chemotherapeutic drugs. To maximize the benefit/toxicity ratio, personalize treatment, and prolong OS, more efforts should be focused on biomarker development for 5-FU, oxaliplatin, and irinotecan.


Re-biopsies of tumor tissue and highly sensitive methods enabling clinicians to detect tumor changes during therapy in the blood will change concepts of treatment. The knowledge of resistance mechanisms and changes in tumor biology during therapy will help clinicians to make biologically rational decisions in the treatment sequence of mCRC.


The authors declare no conflict of interest.


Funding: Supported by the 5P30CA014089-27S1 grant, and the Daniel Butler Research Fund . S. Stintzing is supported by a postdoctoral fellowship from the German Cancer Aid (Mildred-Scheel Foundation , grant number 110422). S. Stremitzer is a recipient of an Erwin Schrödinger fellowship. A. Sebio is a recipient of a Rio Hortega Research Grant from the Insituto de Salud Carlos III ( CM11/00102 ).



References



  1. 1. Siegel R., Ma J., Zou Z., et al: Cancer statistics, 2014. CA Cancer J Clin 2014; 64: pp. 9-29

  2. 2. Van Cutsem E., Kohne C.H., Lang I., et al: Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 2011; 29: pp. 2011-2019

  3. 3. Bennouna J., Sastre J., Arnold D., et al: Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol 2013; 14: pp. 29-37

  4. 4. O’Connell J.B., Maggard M.A., and Ko C.Y.: Colon cancer survival rates with the new American Joint Committee on Cancer sixth edition staging. J Natl Cancer Inst 2004; 96: pp. 1420-1425

  5. 5. Tournigand C., Andre T., Achille E., et al: FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol 2004; 22: pp. 229-237

  6. 6. Saltz L.B., Clarke S., Diaz-Rubio E., et al: Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2008; 26: pp. 2013-2019

  7. 7. Cassidy J., Clarke S., Diaz-Rubio E., et al: XELOX vs FOLFOX-4 as first-line therapy for metastatic colorectal cancer: NO16966 updated results. Br J Cancer 2011; 105: pp. 58-64

  8. 8. Hurwitz H., Fehrenbacher L., Novotny W., et al: Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350: pp. 2335-2342

  9. 9. Bokemeyer C., Bondarenko I., Makhson A., et al: Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol 2009; 27: pp. 663-671

  10. 10. Douillard J.Y., Siena S., Cassidy J., et al: Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol 2010; 28: pp. 4697-4705

  11. 11. Stintzing S., and Lenz H.J.: Protein kinase inhibitors in metastatic colorectal cancer. Let’s pick patients, tumors, and kinase inhibitors to piece the puzzle together! Expert Opin Pharmacother 2013; 14: pp. 2203-2220

  12. 12. Grothey A., Van Cutsem E., Sobrero A., et al: Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013; 381: pp. 303-312

  13. 13. Joulain F., Proskorovsky I., Allegra C., et al: Mean overall survival gain with aflibercept plus FOLFIRI vs placebo plus FOLFIRI in patients with previously treated metastatic colorectal cancer. Br J Cancer 2013; 109: pp. 1735-1743

  14. 14. Falcone A., Ricci S., Brunetti I., et al: Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol 2007; 25: pp. 1670-1676

  15. 15. Schmoll H.J., Van Cutsem E., Stein A., et al: ESMO Consensus Guidelines for management of patients with colon and rectal cancer. A personalized approach to clinical decision making. Ann Oncol 2012; 23: pp. 2479-2516

  16. 16. Heinemann V., Stintzing S., Kirchner T., et al: Clinical relevance of EGFR- and KRAS-status in colorectal cancer patients treated with monoclonal antibodies directed against the EGFR. Cancer Treat Rev 2009; 35: pp. 262-271

  17. 17. Van Cutsem E., Peeters M., Siena S., et al: Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol 2007; 25: pp. 1658-1664

  18. 18. Jonker D.J., O’Callaghan C.J., Karapetis C.S., et al: Cetuximab for the treatment of colorectal cancer. N Engl J Med 2007; 357: pp. 2040-2048

  19. 19. Cunningham D., Humblet Y., Siena S., et al: Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 2004; 351: pp. 337-345

  20. 20. Lievre A., Bachet J.B., Le Corre D., et al: KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 2006; 66: pp. 3992-3995

  21. 21. Stintzing S., Fischer von Weikersthal L., Decker T., et al: FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer-subgroup analysis of patients with KRAS: mutated tumours in the randomised German AIO study KRK-0306. Ann Oncol 2012; 23: pp. 1693-1699

  22. 22. Chen C.C., Er T.K., Liu Y.Y., et al: Computational analysis of KRAS mutations: implications for different effects on the KRAS p.G12D and p.G13D mutations. PLoS One 2013; 8: pp. e55793

  23. 23. Irahara N., Baba Y., Nosho K., et al: NRAS mutations are rare in colorectal cancer. Diagn Mol Pathol 2010; 19: pp. 157-163

  24. 24. Douillard J.Y., Oliner K.S., Siena S., et al: Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 2013; 369: pp. 1023-1034

  25. 25. Malumbres M., and Barbacid M.: RAS oncogenes: the first 30 years. Nat Rev Cancer 2003; 3: pp. 459-465

  26. 26. Tejpar S., Lenz H.J., Kohne C.H., et al: Effect of KRAS and NRAS mutations on treatment outcomes in patients with metastatic colorectal cancer (mCRC) treated first-line with cetuximab plus FOLFOX4: New results from the OPUS study. J Clin Oncol 2014; 32:

  27. 27. Heinemann V., von Weikersthal L.F., Decker T., et al: FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol 2014; 15: pp. 1065-1075

  28. 28. Brodowicz T., Vrbanec D., Kaczirek K., et al: FOLFOX4 plus cetuximab administered weekly or every two weeks in first-line treatment of patients with KRAS and NRAS wild-type (wt) metastatic colorectal cancer (mCRC). J Clin Oncol 2014; 32:

  29. 29. Schubbert S., Shannon K., and Bollag G.: Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer 2007; 7: pp. 295-308

  30. 30. Zuo Z., Chen S.S., Chandra P.K., et al: Application of COLD-PCR for improved detection of KRAS mutations in clinical samples. Mod Pathol 2009; 22: pp. 1023-1031

  31. 31. Anderson S.M.: Laboratory methods for KRAS mutation analysis. Expert Rev Mol Diagn 2011; 11: pp. 635-642

  32. 32. Janne P.A., Borras A.M., Kuang Y., et al: A rapid and sensitive enzymatic method for epidermal growth factor receptor mutation screening. Clin Cancer Res 2006; 12: pp. 751-758

  33. 33. Li M., Diehl F., Dressman D., et al: BEAMing up for detection and quantification of rare sequence variants. Nat Methods 2006; 3: pp. 95-97

  34. 34. Domagala P., Hybiak J., Sulzyc-Bielicka V., et al: KRAS mutation testing in colorectal cancer as an example of the pathologist’s role in personalized targeted therapy: a practical approach. Pol J Pathol 2012; 63: pp. 145-164

  35. 35. Lee S., Brophy V.H., Cao J., et al: Analytical performance of a PCR assay for the detection of KRAS mutations (codons 12/13 and 61) in formalin-fixed paraffin-embedded tissue samples of colorectal carcinoma. Virchows Arch 2012; 460: pp. 141-149

  36. 36. Carotenuto P., Roma C., Rachiglio A.M., et al: Detection of KRAS mutations in colorectal carcinoma patients with an integrated PCR/sequencing and real-time PCR approach. Pharmacogenomics 2010; 11: pp. 1169-1179

  37. 37. Basso M., Strippoli A., Orlandi A., et al: KRAS mutational status affects oxaliplatin-based chemotherapy independently from basal mRNA ERCC-1 expression in metastatic colorectal cancer patients. Br J Cancer 2013; 108: pp. 115-120

  38. 38. Lin Y.L., Liau J.Y., Yu S.C., et al: KRAS mutation is a predictor of oxaliplatin sensitivity in colon cancer cells. PLoS One 2012; 7: pp. e50701

  39. 39. Schmiegel W., Reinacher-Schick A., Arnold D., et al: Capecitabine/irinotecan or capecitabine/oxaliplatin in combination with bevacizumab is effective and safe as first-line therapy for metastatic colorectal cancer: a randomized phase II study of the AIO colorectal study group. Ann Oncol 2013; 24: pp. 1580-1587

  40. 40. Lin J.S., Webber E.M., Senger C.A., et al: Systematic review of pharmacogenetic testing for predicting clinical benefit to anti-EGFR therapy in metastatic colorectal cancer. Am J Cancer Res 2011; 1: pp. 650-662

  41. 41. Sahin I.H., Kazmi S.M., Yorio J.T., et al: Rare though not mutually exclusive: a report of three cases of concomitant KRAS and BRAF mutation and a review of the literature. J Cancer 2013; 4: pp. 320-322

  42. 42. Tol J., Nagtegaal I.D., and Punt C.J.: BRAF mutation in metastatic colorectal cancer. N Engl J Med 2009; 361: pp. 98-99

  43. 43. Richman S.D., Seymour M.T., Chambers P., et al: KRAS and BRAF mutations in advanced colorectal cancer are associated with poor prognosis but do not preclude benefit from oxaliplatin or irinotecan: results from the MRC FOCUS trial. J Clin Oncol 2009; 27: pp. 5931-5937

  44. 44. Price T.J., Hardingham J.E., Lee C.K., et al: Impact of KRAS and BRAF gene mutation status on outcomes from the phase III AGITG MAX trial of capecitabine alone or in combination with bevacizumab and mitomycin in advanced colorectal cancer. J Clin Oncol 2011; 29: pp. 2675-2682

  45. 45. Loupakis F., Cremolini C., Salvatore L., et al: FOLFOXIRI plus bevacizumab as first-line treatment in BRAF mutant metastatic colorectal cancer. Eur J Cancer 2014; 50: pp. 57-63

  46. 46. Shen Y., Wang J., Han X., et al: Effectors of epidermal growth factor receptor pathway: the genetic profiling of KRAS, BRAF, PIK3CA, NRAS mutations in colorectal cancer characteristics and personalized medicine. PLoS One 2013; 8: pp. e81628

  47. 47. Seymour M.T., Brown S.R., Middleton G., et al: Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced colorectal cancer (PICCOLO): a prospectively stratified randomised trial. Lancet Oncol 2013; 14: pp. 749-759

  48. 48. Stintzing S., and Lenz H.J.: A small cog in a big wheel: PIK3CA mutations in colorectal cancer. J Natl Cancer Inst 2013; 105: pp. 1775-1776

  49. 49. Janku F., Lee J.J., Tsimberidou A.M., et al: PIK3CA mutations frequently coexist with RAS and BRAF mutations in patients with advanced cancers. PLoS One 2011; 6: pp. e22769

  50. 50. Liao X., Lochhead P., Nishihara R., et al: Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med 2012; 367: pp. 1596-1606

  51. 51. Kothari N., Kim R.D., Gibbs P., et al: Regular aspirin (ASA) use and survival in patients with PIK3CA-mutated metastatic colorectal cancer (CRC). J Clin Oncol 2014; 32:

  52. 52. De Roock W., De Vriendt V., Normanno N., et al: KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer. Lancet Oncol 2011; 12: pp. 594-603

  53. 53. Britten C.D.: PI3K and MEK inhibitor combinations: examining the evidence in selected tumor types. Cancer Chemother Pharmacol 2013; 71: pp. 1395-1409

  54. 54. Yang Z.Y., Wu X.Y., Huang Y.F., et al: Promising biomarkers for predicting the outcomes of patients with KRAS wild-type metastatic colorectal cancer treated with anti-epidermal growth factor receptor monoclonal antibodies: a systematic review with meta-analysis. Int J Cancer 2013; 133: pp. 1914-1925

  55. 55. Majek O., Gondos A., Jansen L., et al: Sex differences in colorectal cancer survival: population-based analysis of 164,996 colorectal cancer patients in Germany. PLoS One 2013; 8: pp. e68077

  56. 56. Molinari F., and Frattini M.: Functions and regulation of the PTEN gene in colorectal cancer. Front Oncol 2013; 3: pp. 326

  57. 57. Sood A., McClain D., Maitra R., et al: PTEN gene expression and mutations in the PIK3CA gene as predictors of clinical benefit to anti-epidermal growth factor receptor antibody therapy in patients with KRAS wild-type metastatic colorectal cancer. Clin Colorectal Cancer 2012; 11: pp. 143-150

  58. 58. Danielsen S.A., Lind G.E., Bjornslett M., et al: Novel mutations of the suppressor gene PTEN in colorectal carcinomas stratified by microsatellite instability- and TP53 mutation-status. Hum Mutat 2008; 29: pp. E252-E262

  59. 59. Sartore-Bianchi A., Martini M., Molinari F., et al: PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res 2009; 69: pp. 1851-1857

  60. 60. Loupakis F., Pollina L., Stasi I., et al: PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. J Clin Oncol 2009; 27: pp. 2622-2629

  61. 61. Tol J., Dijkstra J.R., Klomp M., et al: Markers for EGFR pathway activation as predictor of outcome in metastatic colorectal cancer patients treated with or without cetuximab. Eur J Cancer 2010; 46: pp. 1997-2009

  62. 62. Khambata-Ford S., Garrett C.R., Meropol N.J., et al: Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J Clin Oncol 2007; 25: pp. 3230-3237

  63. 63. Jacobs B., De Roock W., Piessevaux H., et al: Amphiregulin and epiregulin mRNA expression in primary tumors predicts outcome in metastatic colorectal cancer treated with cetuximab. J Clin Oncol 2009; 27: pp. 5068-5074

  64. 64. Jonker D.J., Karapetis C.S., Harbison C., et al: Epiregulin gene expression as a biomarker of benefit from cetuximab in the treatment of advanced colorectal cancer. Br J Cancer 2014; 110: pp. 648-655

  65. 65. Stintzing S., Jung A., Kapaun C., et al: Amphiregulin and epiregulin expression predicts treatment efficacy in metastatic colorectal cancer (mCRC) patients treated with cetuximab plus CAPIRI or CAPOX: analysis of the German AIO CRC Group trial: KRK-0204. Ann Oncol 2011; 22: pp. v10-v18

  66. 66. Lieu C.H., Tran H., Jiang Z.Q., et al: The association of alternate VEGF ligands with resistance to anti-VEGF therapy in metastatic colorectal cancer. PLoS One 2013; 8: pp. e77117

  67. 67. Vasudev N.S., and Reynolds A.R.: Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 2014; 17: pp. 471-494

  68. 68. Formica V., Palmirotta R., Del Monte G., et al: Predictive value of VEGF gene polymorphisms for metastatic colorectal cancer patients receiving first-line treatment including fluorouracil, irinotecan, and bevacizumab. Int J Colorectal Dis 2011; 26: pp. 143-151

  69. 69. Kopetz S., Hoff P.M., Morris J.S., et al: Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance. J Clin Oncol 2010; 28: pp. 453-459

  70. 70. Gianni L., Romieu G.H., Lichinitser M., et al: AVEREL: a randomized phase III trial evaluating bevacizumab in combination with docetaxel and trastuzumab as first-line therapy for HER2-positive locally recurrent/metastatic breast cancer. J Clin Oncol 2013; 31: pp. 1719-1725

  71. 71. Longley D.B., Harkin D.P., and Johnston P.G.: 5-Fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer 2003; 3: pp. 330-338

  72. 72. Walther A., Johnstone E., Swanton C., et al: Genetic prognostic and predictive markers in colorectal cancer. Nat Rev Cancer 2009; 9: pp. 489-499

  73. 73. Diasio R.B., and Johnson M.R.: The role of pharmacogenetics and pharmacogenomics in cancer chemotherapy with 5-fluorouracil. Pharmacology 2000; 61: pp. 199-203

  74. 74. Koopman M., Venderbosch S., van Tinteren H., et al: Predictive and prognostic markers for the outcome of chemotherapy in advanced colorectal cancer, a retrospective analysis of the phase III randomised CAIRO study. Eur J Cancer 2009; 45: pp. 1999-2006

  75. 75. Vallbohmer D., Yang D.Y., Kuramochi H., et al: DPD is a molecular determinant of capecitabine efficacy in colorectal cancer. Int J Oncol 2007; 31: pp. 413-418

  76. 76. Teh L.K., Hamzah S., Hashim H., et al: Potential of dihydropyrimidine dehydrogenase genotypes in personalizing 5-fluorouracil therapy among colorectal cancer patients. Ther Drug Monit 2013; 35: pp. 624-630

  77. 77. Popat S., Matakidou A., and Houlston R.S.: Thymidylate synthase expression and prognosis in colorectal cancer: a systematic review and meta-analysis. J Clin Oncol 2004; 22: pp. 529-536

  78. 78. Pullarkat S.T., Stoehlmacher J., Ghaderi V., et al: Thymidylate synthase gene polymorphism determines response and toxicity of 5-FU chemotherapy. Pharmacogenomics J 2001; 1: pp. 65-70

  79. 79. Kweekel D.M., Gelderblom H., and Guchelaar H.J.: Pharmacology of oxaliplatin and the use of pharmacogenomics to individualize therapy. Cancer Treat Rev 2005; 31: pp. 90-105

  80. 80. Grothey A., and Goldberg R.M.: A review of oxaliplatin and its clinical use in colorectal cancer. Expert Opin Pharmacother 2004; 5: pp. 2159-2170

  81. 81. Fischer von Weikersthal L., Schalhorn A., Stauch M., et al: Phase III trial of irinotecan plus infusional 5-fluorouracil/folinic acid versus irinotecan plus oxaliplatin as first-line treatment of advanced colorectal cancer. Eur J Cancer 2011; 47: pp. 206-214

  82. 82. Shirota Y., Stoehlmacher J., Brabender J., et al: ERCC1 and thymidylate synthase mRNA levels predict survival for colorectal cancer patients receiving combination oxaliplatin and fluorouracil chemotherapy. J Clin Oncol 2001; 19: pp. 4298-4304

  83. 83. Ruzzo A., Graziano F., Loupakis F., et al: Pharmacogenetic profiling in patients with advanced colorectal cancer treated with first-line FOLFOX-4 chemotherapy. J Clin Oncol 2007; 25: pp. 1247-1254

  84. 84. Baba H., Watanabe M., Okabe H., et al: Upregulation of ERCC1 and DPD expressions after oxaliplatin-based first-line chemotherapy for metastatic colorectal cancer. Br J Cancer 2012; 107: pp. 1950-1955

  85. 85. Kanai M., Yoshioka A., Tanaka S., et al: Associations between glutathione S-transferase pi Ile105Val and glyoxylate aminotransferase Pro11Leu and Ile340Met polymorphisms and early-onset oxaliplatin-induced neuropathy. Cancer Epidemiol 2010; 34: pp. 189-193

  86. 86. Lamas M.J., Duran G., Balboa E., et al: Use of a comprehensive panel of biomarkers to predict response to a fluorouracil-oxaliplatin regimen in patients with metastatic colorectal cancer. Pharmacogenomics 2011; 12: pp. 433-442

  87. 87. Stoehlmacher J., Park D.J., Zhang W., et al: Association between glutathione S-transferase P1, T1, and M1 genetic polymorphism and survival of patients with metastatic colorectal cancer. J Natl Cancer Inst 2002; 94: pp. 936-942

  88. 88. Lamas M.J., Duran G., Balboa E., et al: The value of genetic polymorphisms to predict toxicity in metastatic colorectal patients with irinotecan-based regimens. Cancer Chemother Pharmacol 2012; 69: pp. 1591-1599

  89. 89. Cunningham D., Pyrhonen S., James R.D., et al: Randomised trial of irinotecan plus supportive care versus supportive care alone after fluorouracil failure for patients with metastatic colorectal cancer. Lancet 1998; 352: pp. 1413-1418

  90. 90. Douillard J.Y., Cunningham D., Roth A.D., et al: Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomised trial. Lancet 2000; 355: pp. 1041-1047

  91. 91. Cecchin E., Innocenti F., D’Andrea M., et al: Predictive role of the UGT1A1, UGT1A7, and UGT1A9 genetic variants and their haplotypes on the outcome of metastatic colorectal cancer patients treated with fluorouracil, leucovorin, and irinotecan. J Clin Oncol 2009; 27: pp. 2457-2465

  92. 92. Suenaga M., Fuse N., Yamaguchi T., et al: Pharmacokinetics, safety, and efficacy of FOLFIRI plus bevacizumab in Japanese colorectal cancer patients with UGT1A1 gene polymorphisms. J Clin Pharmacol 2014; 54: pp. 495-502

  93. 93. Braun M.S., Richman S.D., Quirke P., et al: Predictive biomarkers of chemotherapy efficacy in colorectal cancer: results from the UK MRC FOCUS trial. J Clin Oncol 2008; 26: pp. 2690-2698

  94. 94. Nygard S.B., Christensen I.J., Nielsen S.L., et al: Assessment of the topoisomerase I gene copy number as a predictive biomarker of objective response to irinotecan in metastatic colorectal cancer. Scand J Gastroenterol 2014; 49: pp. 84-91

  95. 95. Moreira M.C., Barbot C., Tachi N., et al: The gene mutated in ataxia-ocular apraxia 1 encodes the new HIT/Zn-finger protein aprataxin. Nat Genet 2001; 29: pp. 189-193

  96. 96. Dopeso H., Mateo-Lozano S., Elez E., et al: Aprataxin tumor levels predict response of colorectal cancer patients to irinotecan-based treatment. Clin Cancer Res 2010; 16: pp. 2375-2382

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Mar 1, 2017 | Posted by in HEMATOLOGY | Comments Off on Predictive and Prognostic markers in the treatment of metastatic colorectal cancer (mCRC) – personalized medicine at work –

Full access? Get Clinical Tree

Get Clinical Tree app for offline access