The impact of molecular classification based on the transcriptome of pancreatic cancer: from bench to bedside

Chinese Journal of Academic Radiology - Tập 3 - Trang 67-75 - 2020
Yan Deng1, Ting Zhou1, Jia-long Wu1, Yong Chen2, Cheng-yi Shen3, Mei Zeng4, Tianwu Chen1, Xiao-Ming Zhang1
1Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
2Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
3Sichuan Key Laboratory of Medical Imaging, Department of Pathophysiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
4Biology Group, North Sichuan Medical College, Nanchong, China

Tóm tắt

Pancreatic cancer is a malignancy with a 5-year overall survival rate of less than 10% and is the third leading cause of death among cancers. Total resection is the only potentially curative treatment. Unfortunately, less than 20% of patients are candidates for surgery. High-throughput molecular analysis has revealed the intra- and inter-heterogeneity of pancreatic cancer, leading to a challenge for optimizing clinical treatment to reduce the morbidity and mortality. Currently, the AJCC TNM stage is associated with the outcome of patients with pancreatic cancer, but this clinicopathological factor cannot always predict the prognosis. Owing to the complex nature of this disease, a long-term goal includes the effective identification of different subgroups of patients with prognostic and predictive outcomes to enable precision medicine. Evidence has made it clear that information extracted from the transcriptome is promising and plays an important role in clinical decision-making. In this review, we simply summaries the molecular subtypes and its molecular features based on the transcriptome of pancreatic cancer. Besides, we provide a brief discussion on the impact of molecular classification on prognosis and therapy based on the transcriptome of pancreatic cancer. In addition, the challenges and opportunities for translation from bench to bedside will also be discussed.

Tài liệu tham khảo

Rahib L, et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21. Oettle H, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310(14):1473–81. Lewis R, et al. A contemporary analysis of survival for resected pancreatic ductal adenocarcinoma. HPB (Oxford). 2013;15(1):49–60. Gerlinger M, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–92. Maurer C, et al. Experimental microdissection enables functional harmonisation of pancreatic cancer subtypes. Gut. 2019;68(6):1034–43. Puleo F, et al. Stratification of pancreatic ductal adenocarcinomas based on tumor and microenvironment features. Gastroenterology. 2018;155(6):1999–2013.e3. Bailey P, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531(7592):47–52. Moffitt RA, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47(10):1168–78. Collisson EA, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17(4):500–3. Von Hoff DD, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703. Vaccaro V, Sperduti I, Milella M. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;365(8):768–9 (author reply 769). Gourgou-Bourgade S, et al. Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol. 2013;31(1):23–9. Saka B, et al. Pancreatic ductal adenocarcinoma is spread to the peripancreatic soft tissue in the majority of resected cases, rendering the AJCC T-stage protocol (7th edition) inapplicable and insignificant: a size-based staging system (pT1: ≤ 2, pT2: %3e 2–≤ 4, pT3: %3e 4 cm) is more valid and clinically relevant. Ann Surg Oncol. 2016;23(6):2010–8. Garcea G, et al. Survival following curative resection for pancreatic ductal adenocarcinoma. A systematic review of the literature. JOP. 2008;9(2):99–132. de Jong MC, et al. Re-evaluating the impact of tumor size on survival following pancreaticoduodenectomy for pancreatic adenocarcinoma. J Surg Oncol. 2011;103(7):656–62. Abdel-Rahman O. Evaluation of the 8th AJCC staging system for pathologically versus clinically staged pancreatic adenocarcinoma: a time to revisit a dogma? Hepatobiliary Pancreat Dis Int. 2018;17(1):64–9. Pongprasobchai S, et al. Long-term survival and prognostic indicators in small (%3c or = 2 cm) pancreatic cancer. Pancreatology. 2008;8(6):587–92. Garcea G, et al. Tumour characteristics predictive of survival following resection for ductal adenocarcinoma of the head of pancreas. Eur J Surg Oncol. 2007;33(7):892–7. Rochefort MM, et al. Impact of tumor grade on pancreatic cancer prognosis: validation of a novel TNMG staging system. Ann Surg Oncol. 2013;20(13):4322–9. Konstantinidis IT, et al. Does the mechanism of lymph node invasion affect survival in patients with pancreatic ductal adenocarcinoma? J Gastrointest Surg. 2010;14(2):261–7. Delpero JR, et al. Prognostic value of resection margin involvement after pancreaticoduodenectomy for ductal adenocarcinoma: updates from a French prospective multicenter study. Ann Surg. 2017;266(5):787–96. Delpero JR, et al. Pancreaticoduodenectomy for pancreatic ductal adenocarcinoma: a French multicentre prospective evaluation of resection margins in 150 evaluable specimens. HPB (Oxford). 2014;16(1):20–33. Badea L, et al. Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepatogastroenterology. 2008;55(88):2016–27. Decker K, et al. Gata6 is an important regulator of mouse pancreas development. Dev Biol. 2006;298(2):415–29. Kwei KA, et al. Genomic profiling identifies GATA6 as a candidate oncogene amplified in pancreatobiliary cancer. PLoS Genet. 2008;4(5):e1000081. Xuan S, et al. Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis. J Clin Invest. 2012;122(10):3516–28. Carrasco M, et al. GATA4 and GATA6 control mouse pancreas organogenesis. J Clin Invest. 2012;122(10):3504–15. Singh A, et al. A gene expression signature associated with "K-Ras addiction" reveals regulators of EMT and tumor cell survival. Cancer Cell. 2009;15(6):489–500. Birnbaum DJ, et al. Prognostic value of PDL1 expression in pancreatic cancer. Oncotarget. 2016;7(44):71198–21010. Engelmann D, Putzer BM. Emerging from the shade of p53 mutants: N-terminally truncated variants of the p53 family in EMT signaling and cancer progression. Sci Signal. 2014;7(345):re9. von Figura G, et al. Nr5a2 maintains acinar cell differentiation and constrains oncogenic Kras-mediated pancreatic neoplastic initiation. Gut. 2014;63(4):656–64. Hale MA, et al. The nuclear hormone receptor family member NR5A2 controls aspects of multipotent progenitor cell formation and acinar differentiation during pancreatic organogenesis. Development. 2014;141(16):3123–33. Takahashi K, et al. Pancreatic tumor microenvironment confers highly malignant properties on pancreatic cancer cells. Oncogene. 2018;37(21):2757–72. Waghray M, et al. Deciphering the role of stroma in pancreatic cancer. Curr Opin Gastroenterol. 2013;29(5):537–43. Yee NS. Immunotherapeutic approaches in pancreatic adenocarcinoma: current status and future perspectives. Curr Mol Pharmacol. 2016;9(3):231–41. Heinemann V, et al. Tumour–stroma interactions in pancreatic ductal adenocarcinoma: rationale and current evidence for new therapeutic strategies. Cancer Treat Rev. 2014;40(1):118–28. Froeling FE, et al. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-beta-catenin signaling to slow tumor progression. Gastroenterology. 2011;141(4):1486–97, 1497.e1–14. Ozdemir BC, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2015;28(6):831–3. Rhim AD, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25(6):735–47. Cohen SJ, et al. Fibroblast activation protein and its relationship to clinical outcome in pancreatic adenocarcinoma. Pancreas. 2008;37(2):154–8. Raphael BJ, et al. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32(2):185–203.e13. Birnbaum DJ, et al. A 25-gene classifier predicts overall survival in resectable pancreatic cancer. BMC Med. 2017;15(1):170. Newhook TE, et al. A thirteen-gene expression signature predicts survival of patients with pancreatic cancer and identifies new genes of interest. PLoS ONE. 2014;9(9):e105631. Haider S, et al. A multi-gene signature predicts outcome in patients with pancreatic ductal adenocarcinoma. Genome Med. 2014;6(12):105–105. Janky R, et al. Prognostic relevance of molecular subtypes and master regulators in pancreatic ductal adenocarcinoma. BMC Cancer. 2016;16:632. Birnbaum DJ, et al. Validation and comparison of the molecular classifications of pancreatic carcinomas. Mol Cancer. 2017;16(1):168. Bertucci F, et al. Prognostic value of molecular subtypes in pancreatic cancer. Pancreas. 2017;46(4):e29–e31. Duconseil P, et al. Transcriptomic analysis predicts survival and sensitivity to anticancer drugs of patients with a pancreatic adenocarcinoma. Am J Pathol. 2015;185(4):1022–32. Moore MJ, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6. Yu S, et al. Modified PNA-PCR method: a convenient and accurate method to screen plasma KRAS mutations of cancer patients. Cancer Biol Ther. 2012;13(5):314–20. Birnbaum DJ, et al. Molecular classification as prognostic factor and guide for treatment decision of pancreatic cancer. Biochim Biophys Acta. 2018;1869(2):248–55. Singh S, Hasselluhn MC, Neesse A. A tangled tale of molecular subtypes in pancreatic cancer. Gut. 2019;68(6):953–4. Aung KL, et al. Genomics-driven precision medicine for advanced pancreatic cancer: early results from the COMPASS trial. Clin Cancer Res. 2018;24(6):1344–54. Er JL, Goh PN, Lee CY, et al. Identification of inhibitors synergizing gemcitabine sensitivity in the squamous subtype of pancreatic ductal adenocarcinoma (PDAC). Apoptosis. 2018;23(5–6):343–55. Bian B, et al. Gene expression profiling of patient-derived pancreatic cancer xenografts predicts sensitivity to the BET bromodomain inhibitor JQ1: implications for individualized medicine efforts. EMBO Mol Med. 2017;9(4):482–97. Taneja SS. Re: safety and activity of anti-PD-L1 antibody in patients with advanced cancer. J Urol. 2012;188(6):2148–9. Burr ML, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017;549(7670):101–5. Koido S, et al. Current immunotherapeutic approaches in pancreatic cancer. Clin Dev Immunol. 2011;2011:267539. Lee JJ, et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad Sci USA. 2014;111(30):E3091–E31003100. Beatty GL, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331(6024):1612–6. Olive KP, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324(5933):1457–61. Jiang H, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22(8):851–60. Humphrey ES, Su SP, Nagrial AM, et al. Resolution of novel pancreatic ductal adenocarcinoma subtypes by global phosphotyrosine profiling. Mol Cell Proteomics. 2016;15(8):2671–85. Huang S, Chaudhary K, Garmire LX. More is better: recent progress in multi-omics data integration methods. Front Genet. 2017;8:84. Robertson AG, et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell. 2017;171(3):540–556.e25. Wang H, et al. Integrating omic data with a multiplex network-based approach for the identification of cancer subtypes. IEEE Trans Nanobiosci. 2016;15(4):335–42. Zhang Z, et al. Molecular subtyping of serous ovarian cancer based on multi-omics data. Sci Rep. 2016;6:26001. Sturgeon CM, et al. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for use of tumor markers in liver, bladder, cervical, and gastric cancers. Clin Chem. 2010;56(6):e1–48. Kuhlmann L, et al. Identification and validation of novel subtype-specific protein biomarkers in pancreatic ductal adenocarcinoma. Pancreas. 2017;46(3):311–22. Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than pictures, they are data. Radiology. 2016;278(2):563–77. Yin Q, et al. Integrative radiomics expression predicts molecular subtypes of primary clear cell renal cell carcinoma. Clin Radiol. 2018;73(9):782–91. Ma W, et al. Breast cancer molecular subtype prediction by mammographic radiomic features. Acad Radiol. 2018;26(2):196–201. Attiyeh MA, et al. CT radiomics associations with genotype and stromal content in pancreatic ductal adenocarcinoma. Abdom Radiol (New York). 2019;44(9):3148–57. Kaissis GA, et al. Image-based molecular phenotyping of pancreatic ductal adenocarcinoma. J Clin Med. 2020;9(3):724. Sandrasegaran K, et al. CT texture analysis of pancreatic cancer. Eur Radiol. 2019;29(3):1067–73. Yue Y, et al. Identifying prognostic intratumor heterogeneity using pre- and post-radiotherapy 18F-FDG PET images for pancreatic cancer patients. J Gastrointest Oncol. 2017;8(1):127–38.