Genetic Alterations Featuring Biological Models to Tailor Clinical Management of Pancreatic Cancer Patients
Abstract
:1. Introduction
2. Genomic Variants of PDAC
2.1. Familial Cancer Syndromes
2.2. Inherited Predisposition Loci
2.3. Somatic Mutations
3. Models of PDAC Research
3.1. Established PDAC Cell Line Cultures
3.2. Primary Cell Lines
3.3. Organ-on-Chip
3.4. Patient Derived Xenografts (PDX)
3.5. Genetically Engineered Mouse Models (GEMM)
4. Organoids
4.1. Development of PDAC Organoids
4.2. Organoids as PDAC Tumour-Drug Response Predictors
4.3. Organoids as Models of Tumour Microenvironment
4.4. Organoids for Biomarker Discovery
4.5. Modelling Human Diseases with CRISPR-Cas9-Modified Organoids
4.6. Advantages/Limitations of Organoids
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Chiorean, E.G.; Coveler, A.L. Pancreatic cancer: Optimizing treatment options, new, and emerging targeted therapies. Drug Des. Dev. Ther. 2015, 9, 3529–3545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10. [Google Scholar] [CrossRef] [PubMed]
- Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Becker, A.E.; Hernandez, Y.G.; Frucht, H.; Lucas, A.L. Pancreatic ductal adenocarcinoma: Risk factors, screening, and early detection. World J. Gastroenterol. 2014, 20, 11182–11198. [Google Scholar] [CrossRef] [PubMed]
- Lynch, S.M.; Vrieling, A.; Lubin, J.H.; Kraft, P.; Mendelsohn, J.B.; Hartge, P.; Canzian, F.; Steplowski, E.; Arslan, A.A.; Gross, M.; et al. Cigarette smoking and pancreatic cancer: A pooled analysis from the pancreatic cancer cohort consortium. Am. J. Epidemiol. 2009, 170, 403–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stolzenberg-Solomon, R.Z.; Amundadottir, L.T. Epidemiology and inherited predisposition for sporadic pancreatic adenocarcinoma. Hematol. Oncol. Clin. N. Am. 2015, 29, 619–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arslan, A.A.; Helzlsouer, K.J.; Kooperberg, C.; Shu, X.-O.; Steplowski, E.; Bueno-de-Mesquita, H.B.; Fuchs, C.S.; Gross, M.D.; Jacobs, E.J.; LaCroix, A.Z. Anthropometric measures, body mass index, and pancreatic cancer: A pooled analysis from the Pancreatic Cancer Cohort Consortium (PanScan). Arch. Intern. Med. 2010, 170, 791–802. [Google Scholar] [CrossRef] [Green Version]
- Elena, J.W.; Steplowski, E.; Yu, K.; Hartge, P.; Tobias, G.S.; Brotzman, M.J.; Chanock, S.J.; Stolzenberg-Solomon, R.Z.; Arslan, A.A.; Bueno-de-Mesquita, H.B. Diabetes and risk of pancreatic cancer: A pooled analysis from the pancreatic cancer cohort consortium. Cancer Causes Control 2013, 24, 13–25. [Google Scholar] [CrossRef]
- Nishio, K.; Kimura, K.; Amano, R.; Yamazoe, S.; Ohrira, G.; Nakata, B.; Hirakawa, K.; Ohira, M. Preoperative predictors for early recurrence of resectable pancreatic cancer. World J. Surg. Oncol. 2017, 15, 16. [Google Scholar] [CrossRef] [Green Version]
- Chand, S.; O’Hayer, K.; Blanco, F.F.; Winter, J.M.; Brody, J.R. The landscape of pancreatic cancer therapeutic resistance mechanisms. Int. J. Biol. Sci. 2016, 12, 273. [Google Scholar] [CrossRef] [Green Version]
- Yao, S.Y.; Ng, A.M.; Cass, C.E.; Baldwin, S.A.; Young, J.D. Nucleobase transport by human equilibrative nucleoside transporter 1 (hENT1). J. Biol. Chem. 2011, 286, 32552–32562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGarrity, T.J.; Amos, C.I.; Baker, M.J. Peutz-Jeghers Syndrome. In GeneReviews(R); Pagon, R.A., Adam, M.P., Ardinger, H.H., Wallace, S.E., Amemiya, A., Bean, L.J.H., Bird, T.D., Ledbetter, N., Mefford, H.C., Smith, R.J.H., et al., Eds.; University of Washington, Seattle University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
- Van Lier, M.G.; Westerman, A.M.; Wagner, A.; Looman, C.W.; Wilson, J.H.; de Rooij, F.W.; Lemmens, V.E.; Kuipers, E.J.; Mathus-Vliegen, E.M.; van Leerdam, M.E. High cancer risk and increased mortality in patients with Peutz-Jeghers syndrome. Gut 2011, 60, 141–147. [Google Scholar] [CrossRef] [PubMed]
- Resta, N.; Pierannunzio, D.; Lenato, G.M.; Stella, A.; Capocaccia, R.; Bagnulo, R.; Lastella, P.; Susca, F.C.; Bozzao, C.; Loconte, D.C.; et al. Cancer risk associated with STK11/LKB1 germline mutations in Peutz-Jeghers syndrome patients: Results of an Italian multicenter study. Dig. Liver Dis. 2013, 45, 606–611. [Google Scholar] [CrossRef] [PubMed]
- Launonen, V. Mutations in the human LKB1/STK11 gene. Hum. Mutat. 2005, 26, 291–297. [Google Scholar] [CrossRef] [PubMed]
- Hezel, A.F.; Gurumurthy, S.; Granot, Z.; Swisa, A.; Chu, G.C.; Bailey, G.; Dor, Y.; Bardeesy, N.; DePinho, R.A. Pancreatic Lkb1 Deletion Leads to Acinar Polarity Defects and Cystic Neoplasms. Mol. Cell. Biol. 2008, 28, 2414–2425. [Google Scholar] [CrossRef] [Green Version]
- Morton, J.P.; Jamieson, N.B.; Karim, S.A.; Athineos, D.; Ridgway, R.A.; Nixon, C.; McKay, C.J.; Carter, R.; Brunton, V.G.; Frame, M.C.; et al. LKB1 haploinsufficiency cooperates with Kras to promote pancreatic cancer through suppression of p21-dependent growth arrest. Gastroenterology 2010, 139, 586–597. [Google Scholar] [CrossRef] [Green Version]
- Schabath, M.B.; Welsh, E.A.; Fulp, W.J.; Chen, L.; Teer, J.K.; Thompson, Z.J.; Engel, B.E.; Xie, M.; Berglund, A.E.; Creelan, B.C.; et al. Differential association of STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma. Oncogene 2016, 35, 3209–3216. [Google Scholar] [CrossRef] [Green Version]
- Whitcomb, D.C.; Gorry, M.C.; Preston, R.A.; Furey, W.; Sossenheimer, M.J.; Ulrich, C.D.; Martin, S.P.; Gates, L.K., Jr.; Amann, S.T.; Toskes, P.P.; et al. Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nat. Genet. 1996, 14, 141–145. [Google Scholar] [CrossRef]
- Koziel, D.; Gluszek, S.; Kowalik, A.; Chlopek, M.; Pieciak, L. Genetic mutations in SPINK1, CFTR, CTRC genes in acute pancreatitis. BMC Gastroenterol. 2015, 15, 70. [Google Scholar] [CrossRef] [Green Version]
- Dhar, P.; Kalghatgi, S.; Saraf, V. Pancreatic Cancer in Chronic Pancreatitis. Indian J. Surg. Oncol. 2015, 6, 57–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hussain, S.P.; Hofseth, L.J.; Harris, C.C. Radical causes of cancer. Nat. Rev. Cancer 2003, 3, 276–285. [Google Scholar] [CrossRef] [PubMed]
- Whitcomb, D.; Greer, J. Germ-Line Mutations, Pancreatic Inflammation, and Pancreatic Cancer. Clin. Gastroenterol. Hepatol. 2009, 7, S29–S34. [Google Scholar] [CrossRef] [PubMed]
- Apte, M.V.; Haber, P.S.; Darby, S.J.; Rodgers, S.C.; McCaughan, G.W.; Korsten, M.A.; Pirola, R.C.; Wilson, J.S. Pancreatic stellate cells are activated by proinflammatory cytokines: Implications for pancreatic fibrogenesis. Gut 1999, 44, 534–541. [Google Scholar] [CrossRef] [Green Version]
- Mews, P.; Phillips, P.; Fahmy, R.; Korsten, M.; Pirola, R.; Wilson, J.; Apte, M. Pancreatic stellate cells respond to inflammatory cytokines: Potential role in chronic pancreatitis. Gut 2002, 50, 535–541. [Google Scholar] [CrossRef]
- Kolodecik, T.; Shugrue, C.; Ashat, M.; Thrower, E.C. Risk factors for pancreatic cancer: Underlying mechanisms and potential targets. Front. Physiol. 2014, 4, 415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yadav, D.; Lowenfels, A.B. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 2013, 144, 1252–1261. [Google Scholar] [CrossRef] [Green Version]
- Mize, D.E.; Bishop, M.; Resse, E.; Sluzevich, J. Familial atypical multiple mole melanoma syndrome. Cancer Syndr. 2009, 15, 352–356. [Google Scholar]
- Gruis, N.A.; van der Velden, P.A.; Sandkuijl, L.A.; Prins, D.E.; Weaver-Feldhaus, J.; Kamb, A.; Bergman, W.; Frants, R.R. Homozygotes for CDKN2 (p16) germline mutation in Dutch familial melanoma kindreds. Nat. Genet. 1995, 10, 351–353. [Google Scholar] [CrossRef] [PubMed]
- Hruban, R.H.; Iacobuzio-Donahue, C.; Wilentz, R.E.; Goggins, M.; Kern, S.E. Molecular pathology of pancreatic cancer. Cancer J. 2001, 7, 251–258. [Google Scholar]
- Tatarian, T.; Winter, J.M. Genetics of Pancreatic Cancer and Its Implications on Therapy. Surg. Clin. N. Am. 2016, 96, 1207–1221. [Google Scholar] [CrossRef] [PubMed]
- Sirivatanauksorn, V.; Sirivatanauksorn, Y.; Lemoine, N.R. Molecular pattern of ductal pancreatic cancer. Langenbecks Arch. Surg. 1998, 383, 105–115. [Google Scholar] [CrossRef] [PubMed]
- De Snoo, F.A.; Bishop, D.T.; Bergman, W.; van Leeuwen, I.; van der Drift, C.; van Nieuwpoort, F.A.; Out-Luiting, C.J.; Vasen, H.F.; ter Huurne, J.A.; Frants, R.R.; et al. Increased risk of cancer other than melanoma in CDKN2A founder mutation (p16-Leiden)-positive melanoma families. Clin. Cancer Res. 2008, 14, 7151–7157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vasen, H.; Ibrahim, I.; Ponce, C.G.; Slater, E.P.; Matthai, E.; Carrato, A.; Earl, J.; Robbers, K.; van Mil, A.M.; Potjer, T.; et al. Benefit of Surveillance for Pancreatic Cancer in High-Risk Individuals: Outcome of Long-Term Prospective Follow-Up Studies From Three European Expert Centers. J. Clin. Oncol. 2016, 34, 2010–2019. [Google Scholar] [CrossRef] [Green Version]
- Kastrinos, F.; Mukherjee, B.; Tayob, N.; Wang, F.; Sparr, J.; Raymond, V.M.; Bandipalliam, P.; Stoffel, E.M.; Gruber, S.B.; Syngal, S. Risk of pancreatic cancer in families with Lynch syndrome. JAMA J. Am. Med. Assoc. 2009, 302, 1790–1795. [Google Scholar] [CrossRef]
- Li, G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008, 18, 85–98. [Google Scholar] [CrossRef] [Green Version]
- Lee, V.; Murphy, A.; Le, D.T.; Diaz, L.A. Mismatch Repair Deficiency and Response to Immune Checkpoint Blockade. Oncologist 2016, 21, 1200. [Google Scholar] [CrossRef] [Green Version]
- Eso, Y.; Shimizu, T.; Takeda, H.; Takai, A.; Marusawa, H. Microsatellite instability and immune checkpoint inhibitors: Toward precision medicine against gastrointestinal and hepatobiliary cancers. J. Gastroenterol. 2020, 55, 15–26. [Google Scholar] [CrossRef] [Green Version]
- Kastrinos, F.; Steyerberg, E.W.; Mercado, R.; Balmana, J.; Holter, S.; Gallinger, S.; Siegmund, K.D.; Church, J.M.; Jenkins, M.A.; Lindor, N.M.; et al. The PREMM(1,2,6) model predicts risk of MLH1, MSH2, and MSH6 germline mutations based on cancer history. Gastroenterology 2011, 140, 73–81. [Google Scholar] [CrossRef] [Green Version]
- Breast Cancer Linkage, C. Cancer risks in BRCA2 mutation carriers. J. Natl. Cancer Inst. 1999, 91, 1310–1316. [Google Scholar] [CrossRef]
- Efthimiou, E.; Crnogorac-Jurcevic, T.; Lemoine, N.R.; Brentnall, T.A. Inherited predisposition to pancreatic cancer. Gut 2001, 48, 143–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, S.; Hruban, R.H.; Kamiyama, M.; Borges, M.; Zhang, X.; Parsons, D.W.; Lin, J.C.-H.; Palmisano, E.; Brune, K.; Jaffee, E.M.; et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 2009, 324, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Slater, E.P.; Langer, P.; Niemczyk, E.; Strauch, K.; Butler, J.; Habbe, N.; Neoptolemos, J.P.; Greenhalf, W.; Bartsch, D.K. PALB2 mutations in European familial pancreatic cancer families. Clin. Genet. 2010, 78, 490–494. [Google Scholar] [CrossRef] [PubMed]
- Villarroel, M.C.; Rajeshkumar, N.V.; Garrido-Laguna, I.; De Jesus-Acosta, A.; Jones, S.; Maitra, A.; Hruban, R.H.; Eshleman, J.R.; Klein, A.; Laheru, D.; et al. Personalizing cancer treatment in the age of global genomic analyses: PALB2 gene mutations and the response to DNA damaging agents in pancreatic cancer. Mol. Cancer Ther. 2011, 10, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, C.; Mazzara, P.F. High-grade pancreatic intraepithelial neoplasia in a patient with familial adenomatous polyposis. Arch. Pathol. Lab. Med. 2005, 129, 1398–1400. [Google Scholar] [PubMed]
- Moussata, D.; Senouci, L.; Berger, F.; Scoazec, J.Y.; Pinson, S.; Walter, T.; Lombard-Bohas, C.; Saurin, J.C. Familial adenomatous polyposis and pancreatic cancer. Pancreas 2015, 44, 512–513. [Google Scholar] [CrossRef]
- Matsumoto, T.; Lida, M.; Kobori, Y.; Mizuno, M.; Nakamura, S.; Hizawa, K.; Yao, T. Genetic predisposition to clinical manifestations in familial adenomatous polyposis with special reference to duodenal lesions. Am. J. Gastroenterol. 2002, 97, 180–185. [Google Scholar] [CrossRef]
- Schneikert, J.; Behrens, J. The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut 2007, 56, 417–425. [Google Scholar] [CrossRef] [Green Version]
- Oshima, M.; Oshima, H.; Kitagawa, K.; Kobayashi, M.; Itakura, C.; Taketo, M. Loss of Apc heterozygosity and abnormal tissue building in nascent intestinal polyps in mice carrying a truncated Apc gene. Proc. Natl. Acad. Sci. USA 1995, 92, 4482–4486. [Google Scholar] [CrossRef] [Green Version]
- Tam, V.; Patel, N.; Turcotte, M.; Bossé, Y.; Paré, G.; Meyre, D. Benefits and limitations of genome-wide association studies. Nat. Rev. Genet. 2019, 20, 467–484. [Google Scholar] [CrossRef]
- Bush, W.S.; Moore, J.H. Genome-wide association studies. PLoS Comput. Biol. 2012, 8, e1002822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robert, F.; Pelletier, J. Exploring the Impact of Single-Nucleotide Polymorphisms on Translation. Front. Genet. 2018, 9, 507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amundadottir, L.; Kraft, P.; Stolzenberg-Solomon, R.Z.; Fuchs, C.S.; Petersen, G.M.; Arslan, A.A.; Bueno-de-Mesquita, H.B.; Gross, M.; Helzlsouer, K.; Jacobs, E.J.; et al. Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat. Genet. 2009, 41, 986–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petersen, G.M.; Amundadottir, L.; Fuchs, C.S.; Kraft, P.; Stolzenberg-Solomon, R.Z.; Jacobs, K.B.; Arslan, A.A.; Bueno-de-Mesquita, H.B.; Gallinger, S.; Gross, M.; et al. A genome-wide association study identifies pancreatic cancer susceptibility loci on chromosomes 13q22.1, 1q32.1 and 5p15.33. Nat. Genet. 2010, 42, 224–228. [Google Scholar] [CrossRef] [Green Version]
- Wolpin, B.M.; Rizzato, C.; Kraft, P.; Kooperberg, C.; Petersen, G.M.; Wang, Z.; Arslan, A.A.; Beane-Freeman, L.; Bracci, P.M.; Buring, J. Genome-wide association study identifies multiple susceptibility loci for pancreatic cancer. Nat. Genet. 2014, 46, 994–1000. [Google Scholar] [CrossRef] [Green Version]
- Childs, E.J.; Mocci, E.; Campa, D.; Bracci, P.M.; Gallinger, S.; Goggins, M.; Li, D.; Neale, R.E.; Olson, S.H.; Scelo, G. Common variation at 2p13. 3, 3q29, 7p13 and 17q25. 1 associated with susceptibility to pancreatic cancer. Nat. Genet. 2015, 47, 911–916. [Google Scholar] [CrossRef]
- Klein, A.P.; Wolpin, B.M.; Risch, H.A.; Stolzenberg-Solomon, R.Z.; Mocci, E.; Zhang, M.; Canzian, F.; Childs, E.J.; Hoskins, J.W.; Jermusyk, A.; et al. Genome-wide meta-analysis identifies five new susceptibility loci for pancreatic cancer. Nat. Commun. 2018, 9, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Wang, Z.; Obazee, O.; Jia, J.; Childs, E.J.; Hoskins, J.; Figlioli, G.; Mocci, E.; Collins, I.; Chung, C.C.; et al. Three new pancreatic cancer susceptibility signals identified on chromosomes 1q32.1, 5p15.33 and 8q24.21. Oncotarget 2016, 7, 66328. [Google Scholar] [CrossRef] [Green Version]
- Obazee, O.; Archibugi, L.; Andriulli, A.; Soucek, P.; Małecka-Panas, E.; Ivanauskas, A.; Johnson, T.; Gazouli, M.; Pausch, T.; Lawlor, R.T.; et al. Germline BRCA2 K3326X and CHEK2 I157T mutations increase risk for sporadic pancreatic ductal adenocarcinoma. Int. J. Cancer 2019, 145, 686–693. [Google Scholar] [CrossRef]
- Ramanan, V.K.; Shen, L.; Moore, J.H.; Saykin, A.J. Pathway analysis of genomic data: Concepts, methods, and prospects for future development. Trends Genet. 2012, 28, 323–332. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Duell, E.J.; Yu, K.; Risch, H.A.; Olson, S.H.; Kooperberg, C.; Wolpin, B.M.; Jiao, L.; Dong, X.; Wheeler, B.; et al. Pathway analysis of genome-wide association study data highlights pancreatic development genes as susceptibility factors for pancreatic cancer. Carcinogenesis 2012, 33, 1384–1390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walsh, N.; Zhang, H.; Hyland, P.; Yang, Q.; Yu, K.; Klein, A.; Stolzenberg-Solomon, R. Agnostic pathway/gene set analysis of genome- wide association data identifies associations for pancreatic cancer. J. Natl. Cancer Inst. 2018, 111, 557–567. [Google Scholar] [CrossRef] [PubMed]
- Campa, D.; Gentiluomo, M.; Obazee, O.; Ballerini, A.; Vodickova, L.; Hegyi, P.; Soucek, P.; Brenner, H.; Milanetto, A.C.; Landi, S.; et al. Genome-wide association study identifies an early onset pancreatic cancer risk locus. Int. J. Cancer 2020. [Google Scholar] [CrossRef] [PubMed]
- Jones, S.; Zhang, X.; Parsons, D.W.; Lin, J.C.-H.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Kamiyama, H.; Jimeno, A.; et al. Core Signaling Pathways in Human Pancreatic Cancers Revealed by Global Genomic Analyses. Science 2008, 321, 1801–1806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cicenas, J.; Kvederaviciute, K.; Meskinyte, I.; Meskinyte-Kausiliene, E.; Skeberdyte, A. KRAS, TP53, CDKN2A, SMAD4, BRCA1, and BRCA2 mutations in pancreatic cancer. Cancers 2017, 9, 42. [Google Scholar] [CrossRef] [Green Version]
- Waddell, N.; Pajic, M.; Patch, A.-M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [Green Version]
- Biankin, A.V.; Waddell, N.; Kassahn, K.S.; Gingras, M.-C.; Muthuswamy, L.B.; Johns, A.L.; Miller, D.K.; Wilson, P.J.; Patch, A.-M.; Wu, J.; et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012, 491, 399–405. [Google Scholar] [CrossRef]
- Witkiewicz, A.K.; McMillan, E.A.; Balaji, U.; Baek, G.; Lin, W.C.; Mansour, J.; Mollaee, M.; Wagner, K.U.; Koduru, P.; Yopp, A.; et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 2015, 6, 6744. [Google Scholar] [CrossRef]
- Rachakonda, P.S.; Bauer, A.S.; Xie, H.; Campa, D.; Rizzato, C.; Canzian, F.; Beghelli, S.; Greenhalf, W.; Costello, E.; Schanne, M.; et al. Somatic mutations in exocrine pancreatic tumors: Association with patient survival. PLoS ONE 2013, 8, e60870. [Google Scholar] [CrossRef] [Green Version]
- Di Magliano, M.P.; Logsdon, C.D. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology 2013, 144, 1220–1229. [Google Scholar] [CrossRef] [Green Version]
- Rowinsky, E.K.; Windle, J.J.; Von Hoff, D.D. Ras protein farnesyltransferase: A strategic target for anticancer therapeutic development. J. Clin. Oncol. 1999, 17, 3631–3652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Cutsem, E.; van de Velde, H.; Karasek, P.; Oettle, H.; Vervenne, W.L.; Szawlowski, A.; Schoffski, P.; Post, S.; Verslype, C.; Neumann, H.; et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J. Clin. Oncol. 2004, 22, 1430–1438. [Google Scholar] [CrossRef] [PubMed]
- Calhoun, E.S.; Kern, S.E. Molecular Genetics of Pancreatic Cancer. In Pancreatic Cancer; Lowy, A.M., Leach, S.D., Philip, P.A., Eds.; Springer US: Boston, MA, USA, 2008; pp. 27–39. ISBN 978-0-387-69252-4. [Google Scholar]
- Hong, S.-M.; Park, J.Y.; Hruban, R.H.; Goggins, M. Molecular signatures of pancreatic cancer. Arch. Pathol. Lab. Med. 2011, 135, 716–727. [Google Scholar] [PubMed]
- Bailey, J.M.; Hendley, A.M.; Lafaro, K.J.; Pruski, M.A.; Jones, N.C.; Alsina, J.; Younes, M.; Maitra, A.; McAllister, F.; Iacobuzio-Donahue, C.A. p53 mutations cooperate with oncogenic Kras to promote adenocarcinoma from pancreatic ductal cells. Oncogene 2016, 35, 4282–4288. [Google Scholar] [CrossRef]
- Tokino, T.; Nakamura, Y. The role of p53-target genes in human cancer. Crit. Rev. Oncol. Hematol. 2000, 33, 1–6. [Google Scholar] [CrossRef]
- Li, D.; Xie, K.; Wolff, R.; Abbruzzese, J.L. Pancreatic cancer. Lancet 2004, 363, 1049–1057. [Google Scholar] [CrossRef]
- Weissmueller, S.; Manchado, E.; Saborowski, M.; Morris IV, J.P.; Wagenblast, E.; Davis, C.A.; Moon, S.H.; Pfister, N.T.; Tschaharganeh, D.F.; Kitzing, T.; et al. Mutant p53 drives pancreatic cancer metastasis through cell-autonomous PDGF receptor β signaling. Cell 2014, 157, 382–394. [Google Scholar] [CrossRef] [Green Version]
- Principe, D.R.; Doll, J.A.; Bauer, J.; Jung, B.; Munshi, H.G.; Bartholin, L.; Pasche, B.; Lee, C.; Grippo, P.J. TGF-β: Duality of function between tumor prevention and carcinogenesis. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef]
- Xia, X.; Wu, W.; Huang, C.; Cen, G.; Jiang, T.; Cao, J.; Huang, K.; Qiu, Z. SMAD4 and its role in pancreatic cancer. Tumour Biol. 2015, 36, 111–119. [Google Scholar] [CrossRef]
- Murphy, S.J.; Hart, S.N.; Lima, J.F.; Kipp, B.R.; Klebig, M.; Winters, J.L.; Szabo, C.; Zhang, L.; Eckloff, B.W.; Petersen, G.M.; et al. Genetic Alterations Associated With Progression From Pancreatic Intraepithelial Neoplasia to Invasive Pancreatic Tumor. Gastroenterology 2013, 145, 1098–1109.e1. [Google Scholar] [CrossRef] [Green Version]
- Grant, R.C.; Selander, I.; Connor, A.A.; Selvarajah, S.; Borgida, A.; Briollais, L.; Petersen, G.M.; Lerner-Ellis, J.; Holter, S.; Gallinger, S. Prevalence of Germline Mutations in Cancer Predisposition Genes in Patients With Pancreatic Cancer. Gastroenterology 2015, 148, 556–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucey, B.P.; Nelson-Rees, W.A.; Hutchins, G.M. Henrietta Lacks, HeLa cells, and cell culture contamination. Arch. Pathol. Lab. Med. 2009, 133, 1463–1467. [Google Scholar] [PubMed]
- Gillet, J.-P.; Varma, S.; Gottesman, M.M. The clinical relevance of cancer cell lines. J. Natl. Cancer Inst. 2013, 105, 452–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nelson-Rees, W.A.; Owens, R.B.; Arnstein, P.; Kniazeff, A.J. Source, alterations, characteristics and use of a new dog cell line (Cf2Th). In Vitro 1976, 12, 665–669. [Google Scholar] [CrossRef] [PubMed]
- Goodspeed, A.; Heiser, L.M.; Gray, J.W.; Costello, J.C. Tumor-Derived Cell Lines as Molecular Models of Cancer Pharmacogenomics. Mol. Cancer Res. 2016, 14, 3–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deer, E.L.; González-Hernández, J.; Coursen, J.D.; Shea, J.E.; Ngatia, J.; Scaife, C.L.; Firpo, M.A.; Mulvihill, S.J. Phenotype and genotype of pancreatic cancer cell lines. Pancreas 2010, 39, 425–435. [Google Scholar] [CrossRef] [Green Version]
- Ben-David, U.; Siranosian, B.; Ha, G.; Tang, H.; Oren, Y.; Hinohara, K.; Strathdee, C.A.; Dempster, J.; Lyons, N.J.; Burns, R.; et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature 2018, 560, 325–330. [Google Scholar] [CrossRef]
- Boonstra, J.J.; van Marion, R.; Beer, D.G.; Lin, L.; Chaves, P.; Ribeiro, C.; Pereira, A.D.; Roque, L.; Darnton, S.J.; Altorki, N.K.; et al. Verification and Unmasking of Widely Used Human Esophageal Adenocarcinoma Cell Lines. JNCI J. Natl. Cancer Inst. 2010, 102, 271–274. [Google Scholar] [CrossRef] [PubMed]
- Horbach, S.P.J.M.; Halffman, W. The ghosts of HeLa: How cell line misidentification contaminates the scientific literature. PLoS ONE 2017, 12, e0186281. [Google Scholar] [CrossRef]
- Oldroyd, N.J.; Urquhart, A.J.; Kimpton, C.P.; Millican, E.S.; Watson, S.K.; Downes, T.; Gill, P.D. A highly discriminating octoplex short tandem repeat polymerase chain reaction system suitable for human individual identification. Electrophoresis 1995, 16, 334–337. [Google Scholar] [CrossRef]
- Reid, Y.; Storts, D.; Riss, T.; Minor, L. Authentication of Human Cell Lines by STR DNA Profiling Analysis. In Assay Guidance Manual; Eli Lilly & Company and the National Center for Advancing Translational Sciences: Bethesda, MD, USA, 2004. [Google Scholar]
- Masters, J.R.; Thomson, J.A.; Daly-Burns, B.; Reid, Y.A.; Dirks, W.G.; Packer, P.; Toji, L.H.; Ohno, T.; Tanabe, H.; Arlett, C.F.; et al. Short tandem repeat profiling provides an international reference standard for human cell lines. Proc. Natl. Acad. Sci. USA 2001, 98, 8012–8017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Janik, K.; Popeda, M.; Peciak, J.; Rosiak, K.; Smolarz, M.; Treda, C.; Rieske, P.; Stoczynska-Fidelus, E.; Ksiazkiewicz, M. Efficient and simple approach to in vitro culture of primary epithelial cancer cells. Biosci. Rep. 2016, 36, e00423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zieba, J.; Ksiazkiewcz, M.; Janik, K.; Banaszczyk, M.; Peciak, J.; Piaskowski, S.; Lipinski, M.; Olczak, M.; Stoczynska-Fidelus, E.; Rieske, P. Sensitivity of neoplastic cells to senescence unveiled under standard cell culture conditions. Anticancer Res. 2015, 35, 2759–2768. [Google Scholar] [PubMed]
- Kodack, D.P.; Farago, A.F.; Dastur, A.; Held, M.A.; Dardaei, L.; Friboulet, L.; von Flotow, F.; Damon, L.J.; Lee, D.; Parks, M.; et al. Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care. Cell Rep. 2017, 21, 3298–3309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Young, E.W.K. Cells, tissues, and organs on chips: Challenges and opportunities for the cancer tumor microenvironment. Integr. Biol. 2013, 5, 1096–1109. [Google Scholar] [CrossRef] [PubMed]
- Caballero, D.; Kaushik, S.; Correlo, V.M.; Oliveira, J.M.; Reis, R.L.; Kundu, S.C. Organ-on-chip models of cancer metastasis for future personalized medicine: From chip to the patient. Biomaterials 2017, 149, 98–115. [Google Scholar] [CrossRef]
- Biselli, E.; Agliari, E.; Barra, A.; Bertani, F.R.; Gerardino, A.; De Ninno, A.; Mencattini, A.; Di Giuseppe, D.; Mattei, F.; Schiavoni, G.; et al. Organs on chip approach: A tool to evaluate cancer-immune cells interactions. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef]
- Beer, M.; Kuppalu, N.; Stefanini, M.; Becker, H.; Schulz, I.; Manoli, S.; Schuette, J.; Schmees, C.; Casazza, A.; Stelzle, M.; et al. A novel microfluidic 3D platform for culturing pancreatic ductal adenocarcinoma cells: Comparison with in vitro cultures and in vivo xenografts. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef]
- Nguyen, D.H.T.; Lee, E.; Alimperti, S.; Norgard, R.J.; Wong, A.; Lee, J.J.K.; Eyckmans, J.; Stanger, B.Z.; Chen, C.S. A biomimetic pancreatic cancer on-chip reveals endothelial ablation via ALK7 signaling. Sci. Adv. 2019, 5, eaav6789. [Google Scholar] [CrossRef] [Green Version]
- Jung, J.; Seol, H.S.; Chang, S. The Generation and Application of Patient-Derived Xenograft Model for Cancer Research. Cancer Res. Treat. 2018, 50, 1. [Google Scholar] [CrossRef] [Green Version]
- Garrido-Laguna, I.; Uson, M.; Rajeshkumar, N.V.; Tan, A.C.; de Oliveira, E.; Karikari, C.; Villaroel, M.C.; Salomon, A.; Taylor, G.; Sharma, R.; et al. Tumor Engraftment in Nude Mice and Enrichment in Stroma- Related Gene Pathways Predict Poor Survival and Resistance to Gemcitabine in Patients with Pancreatic Cancer. Clin. Cancer Res. 2011, 17, 5793–5800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jimeno, A.; Rubio-Viqueira, B.; Rajeshkumar, N.V.; Chan, A.; Solomon, A.; Hidalgo, M. A Fine-Needle Aspirate-Based Vulnerability Assay Identifies Polo-Like Kinase 1 as a Mediator of Gemcitabine Resistance in Pancreatic Cancer. Mol. Cancer Ther. 2010, 9, 311–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Festing, S.; Wilkinson, R. The ethics of animal research. Talking Point on the use of animals in scientific research. EMBO Rep. 2007, 8, 526–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herreros-Villanueva, M.; Hijona, E.; Cosme, A.; Bujanda, L. Mouse models of pancreatic cancer. World J. Gastroenterol. 2012, 18, 1286. [Google Scholar] [CrossRef] [PubMed]
- Schneeberger, V.E.; Allaj, V.; Gardner, E.E.; Poirier, J.T.; Rudin, C.M. Quantitation of murine stroma and selective purification of the human tumor component of patient-derived xenografts for genomic analysis. PLoS ONE 2016, 11, e0160587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kersten, K.; de Visser, K.E.; van Miltenburg, M.H.; Jonkers, J. Genetically engineered mouse models in oncology research and cancer medicine. EMBO Mol. Med. 2017, 9, 137–153. [Google Scholar] [CrossRef]
- Sánchez-Rivera, F.J.; Jacks, T. Applications of the CRISPR–Cas9 system in cancer biology. Nat. Rev. Cancer 2015, 15, 387–393. [Google Scholar] [CrossRef] [Green Version]
- Hingorani, S.R.; Petricoin, E.F.; Maitra, A.; Rajapakse, V.; King, C.; Jacobetz, M.A.; Ross, S.; Conrads, T.P.; Veenstra, T.D.; Hitt, B.A.; et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 2003, 4, 437–450. [Google Scholar] [CrossRef] [Green Version]
- Hingorani, S.R.; Wang, L.; Multani, A.S.; Combs, C.; Deramaudt, T.B.; Hruban, R.H.; Rustgi, A.K.; Chang, S.; Tuveson, D.A. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005, 7, 469–483. [Google Scholar] [CrossRef] [Green Version]
- Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef] [Green Version]
- Frese, K.K.; Neesse, A.; Cook, N.; Bapiro, T.E.; Lolkema, M.P.; Jodrell, D.I.; Tuveson, D.A. nab -Paclitaxel Potentiates Gemcitabine Activity by Reducing Cytidine Deaminase Levels in a Mouse Model of Pancreatic Cancer. Cancer Discov. 2012, 2, 260–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weiswald, L.B.; Bellet, D.; Dangles-Marie, V. Spherical cancer models in tumor biology. Neoplasia 2015, 17, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thoma, C.R.; Zimmermann, M.; Agarkova, I.; Kelm, J.M.; Krek, W. 3D cell culture systems modeling tumor growth determinants in cancer target discovery. Adv. Drug Deliv. Rev. 2014, 69–70, 29–41. [Google Scholar] [CrossRef] [PubMed]
- Zanoni, M.; Piccinini, F.; Arienti, C.; Zamagni, A.; Santi, S.; Polico, R.; Bevilacqua, A.; Tesei, A. 3D tumor spheroid models for in vitro therapeutic screening: A systematic approach to enhance the biological relevance of data obtained. Sci. Rep. 2016, 6, 19103. [Google Scholar] [CrossRef] [PubMed]
- Kim, J. Bin Three-dimensional tissue culture models in cancer biology. Semin. Cancer Biol. 2005, 15, 365–377. [Google Scholar] [CrossRef]
- Smith, S.J.; Wilson, M.; Ward, J.H.; Rahman, C.V.; Peet, A.C.; Macarthur, D.C.; Rose, F.R.A.J.; Grundy, R.G.; Rahman, R. Recapitulation of Tumor Heterogeneity and Molecular Signatures in a 3D Brain Cancer Model with Decreased Sensitivity to Histone Deacetylase Inhibition. PLoS ONE 2012, 7, e52335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loessner, D.; Stok, K.S.; Lutolf, M.P.; Hutmacher, D.W.; Clements, J.A.; Rizzi, S.C. Bioengineered 3D platform to explore cell-ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials 2010, 31, 8494–8506. [Google Scholar] [CrossRef] [Green Version]
- Shaw, K.R.; Wrobel, C.N.; Brugge, J.S. Use of three-dimensional basement membrane cultures to model oncogene-induced changes in mammary epithelial morphogenesis. J. Mammary Gland Biol. Neoplasia 2004, 9, 297–310. [Google Scholar] [CrossRef] [Green Version]
- Anton, D.; Burckel, H.; Josset, E.; Noel, G. Three-dimensional cell culture: A breakthrough in vivo. Int. J. Mol. Sci. 2015, 16, 5517–5527. [Google Scholar] [CrossRef]
- Adcock, A.F.; Trivedi, G.; Edmondson, R.; Spearman, C.; Yang, L. Three-Dimensional (3D) Cell Cultures in Cell-based Assays for in-vitro Evaluation of Anticancer Drugs. J. Anal. Bioanal. Tech. 2015, 6, 247. [Google Scholar] [CrossRef]
- Durand, R.E.; Olive, P.L. Resistance of tumor cells to chemo- and radiotherapy modulated by the three-dimensional architecture of solid tumors and spheroids. Methods Cell Biol. 2001, 64, 211–233. [Google Scholar] [PubMed]
- Sato, T.; Vries, R.G.; Snippert, H.J.; van de Wetering, M.; Barker, N.; Stange, D.E.; van Es, J.H.; Abo, A.; Kujala, P.; Peters, P.J.; et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009, 459, 262–265. [Google Scholar] [CrossRef] [PubMed]
- Lancaster, M.A.; Knoblich, J.A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef] [PubMed]
- Clevers, H. Modeling Development and Disease with Organoids. Cell 2016, 165, 1586–1597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, F.X.; Harrison, L.C. Laminin-1 and epidermal growth factor family members co-stimulate fetal pancreas cell proliferation and colony formation. Differentiation 2005, 73, 45–49. [Google Scholar] [CrossRef]
- Bleijs, M.; Wetering, M.; Clevers, H.; Drost, J. Xenograft and organoid model systems in cancer research. EMBO J. 2019, 38, e101654. [Google Scholar] [CrossRef]
- Masters, J.R.W. Human cancer cell lines: Fact and fantasy. Nat. Rev. Mol. Cell Biol. 2000, 1, 233–236. [Google Scholar] [CrossRef]
- Qiu, W.; Su, G.H. Development of orthotopic pancreatic tumor mouse models. Methods Mol. Biol. 2013, 2013, 215–223. [Google Scholar]
- Tiriac, H.; Bucobo, J.C.; Tzimas, D.; Grewel, S.; Lacomb, J.F.; Rowehl, L.M.; Nagula, S.; Wu, M.; Kim, J.; Sasson, A.; et al. Successful creation of pancreatic cancer organoids by means of EUS-guided fine-needle biopsy sampling for personalized cancer treatment. Gastrointest. Endosc. 2018, 87, 1474–1480. [Google Scholar] [CrossRef]
- Walsh, A.J.; Cook, R.S.; Sanders, M.E.; Arteaga, C.L.; Skala, M.C. Drug response in organoids generated from frozen primary tumor tissues OPEN. Nat. Publ. Group 2015, 6, 18889. [Google Scholar]
- Boj, S.F.; Hwang, C.I.; Baker, L.A.; Chio, I.I.C.; Engle, D.D.; Corbo, V.; Jager, M.; Ponz-Sarvise, M.; Tiriac, H.; Spector, M.S.; et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 2015, 160, 324–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Greggio, C.; De Franceschi, F.; Figueiredo-Larsen, M.; Gobaa, S.; Ranga, A.; Semb, H.; Lutolf, M.; Grapin-Botton, A. Artificial three-dimensional niches deconstruct pancreas development in vitro. Development 2013, 140, 4452–4462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huch, M.; Bonfanti, P.; Boj, S.F.; Sato, T.; Loomans, C.J.M.; van de Wetering, M.; Sojoodi, M.; Li, V.S.W.; Schuijers, J.; Gracanin, A.; et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 2013, 32, 2708–2721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Broutier, L.; Andersson-Rolf, A.; Hindley, C.J.; Boj, S.F.; Clevers, H.; Koo, B.K.; Huch, M. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat. Protoc. 2016, 11, 1724. [Google Scholar] [CrossRef]
- Georgakopoulos, N.; Prior, N.; Angres, B.; Mastrogiovanni, G.; Cagan, A.; Harrison, D.; Hindley, C.J.; Arnes-Benito, R.; Liau, S.S.; Curd, A.; et al. Long-term expansion, genomic stability and in vivo safety of adult human pancreas organoids. BMC Dev. Biol. 2020, 20, 1–20. [Google Scholar] [CrossRef] [Green Version]
- Nelson, S.R.; Zhang, C.; Roche, S.; O’Neill, F.; Swan, N.; Luo, Y.; Larkin, A.M.; Crown, J.; Walsh, N. Modelling of pancreatic cancer biology: Transcriptomic signature for 3D PDX-derived organoids and primary cell line organoid development. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef]
- Hou, S.; Tiriac, H.; Sridharan, B.P.; Scampavia, L.; Madoux, F.; Seldin, J.; Souza, G.R.; Watson, D.; Tuveson, D.; Spicer, T.P. Advanced Development of Primary Pancreatic Organoid Tumor Models for High-Throughput Phenotypic Drug Screening. SLAS Discov. Adv. Life Sci. R D 2018, 23, 574–584. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Holtzinger, A.; Jagan, I.; Begora, M.; Lohse, I.; Ngai, N.; Nostro, C.; Wang, R.; Muthuswamy, L.B.; Crawford, H.C.; et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids. Nat. Med. 2015, 21, 1364. [Google Scholar] [CrossRef]
- Romero-Calvo, I.; Weber, C.R.; Ray, M.; Brown, M.; Kirby, K.; Nandi, R.K.; Long, T.M.; Sparrow, S.M.; Ugolkov, A.; Qiang, W.; et al. Human organoids share structural and genetic features with primary pancreatic adenocarcinoma tumors. Mol. Cancer Res. 2019, 17, 70–83. [Google Scholar] [CrossRef] [Green Version]
- Frappart, P.O.; Walter, K.; Gout, J.; Beutel, A.K.; Morawe, M.; Arnold, F.; Breunig, M.; Barth, T.F.E.; Marienfeld, R.; Schulte, L.; et al. Pancreatic cancer-derived organoids—A disease modeling tool to predict drug response. United Eur. Gastroenterol. J. 2020. [Google Scholar] [CrossRef] [Green Version]
- Tiriac, H.; Belleau, P.; Engle, D.D.; Plenker, D.; Deschênes, A.; Somerville, T.D.D.; Froeling, F.E.M.; Burkhart, R.A.; Denroche, R.E.; Jang, G.-H.; et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018, 8, 1112–1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Von Bernstorff, W.; Voss, M.; Freichel, S.; Schmid, A.; Vogel, I.; Jöhnk, C.; Henne-Bruns, D.; Kremer, B.; Kalthoff, H. Systemic and local immunosuppression in pancreatic cancer patients. Clin. Cancer Res. 2001, 7, 925s–932s. [Google Scholar] [PubMed]
- Tsai, S.; Mcolash, L.; Palen, K.; Johnson, B.; Duris, C.; Yang, Q.; Dwinell, M.B.; Hunt, B.; Evans, D.B.; Gershan, J.; et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer 2018, 18, 335. [Google Scholar] [CrossRef] [PubMed]
- Menon, S.; Shin, S.; Dy, G. Advances in cancer immunotherapy in solid tumors. Cancers 2016, 8, 106. [Google Scholar] [CrossRef] [Green Version]
- Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef]
- Huang, L.; Bockorny, B.; Paul, I.; Akshinthala, D.; Gandarilla, O.; Bose, A.; Sanchez-Gonzalez, V.; Rouse, E.; Lehoux, S.; Pandell, N.; et al. Pancreatic tumor organoids for modeling in vivo drug response and discovering clinically-actionable biomarkers. bioRxiv 2019, 513267. [Google Scholar] [CrossRef]
- Hwang, W.Y.; Fu, Y.; Reyon, D.; Maeder, M.L.; Tsai, S.Q.; Sander, J.D.; Peterson, R.T.; Yeh, J.R.J.; Joung, J.K. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat. Biotechnol. 2013, 31, 227–229. [Google Scholar] [CrossRef]
- Takeda, H.; Kataoka, S.; Nakayama, M.; Ali, M.A.E.; Oshima, H.; Yamamoto, D.; Park, J.-W.; Takegami, Y.; An, T.; Jenkins, N.A.; et al. CRISPR-Cas9–mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes. Proc. Natl. Acad. Sci. USA 2019, 116, 15635–15644. [Google Scholar] [CrossRef] [Green Version]
- Niu, D.; Wei, H.J.; Lin, L.; George, H.; Wang, T.; Lee, I.H.; Zhao, H.Y.; Wang, Y.; Kan, Y.; Shrock, E.; et al. Inactivation of porcine endogenous retrovirus in pigs using CRISPR-Cas9. Science 2017, 357, 1303–1307. [Google Scholar] [CrossRef] [Green Version]
- Yang, H.; Wang, H.; Jaenisch, R. Generating genetically modified mice using CRISPR/Cas-mediated genome engineering. Nat. Protoc. 2014, 9, 1956. [Google Scholar] [CrossRef]
- Driehuis, E.; Clevers, H. CRISPR/Cas 9 genome editing and its applications in organoids. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G257–G265. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.; Snyder, E.R.; Liu, Y.; Gu, X.; Wang, J.; Flowers, B.M.; Kim, Y.J.; Park, S.; Szot, G.L.; Hruban, R.H.; et al. Reconstituting development of pancreatic intraepithelial neoplasia from primary human pancreas duct cells. Nat. Commun. 2017, 8, 14686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seino, T.; Kawasaki, S.; Shimokawa, M.; Tamagawa, H.; Toshimitsu, K.; Fujii, M.; Ohta, Y.; Matano, M.; Nanki, K.; Kawasaki, K.; et al. Human Pancreatic Tumor Organoids Reveal Loss of Stem Cell Niche Factor Dependence during Disease Progression. Cell Stem Cell 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aberle, M.R.; Burkhart, R.A.; Tiriac, H.; Olde Damink, S.W.M.; Dejong, C.H.C.; Tuveson, D.A.; van Dam, R.M. Patient-derived organoid models help define personalized management of gastrointestinal cancer. Br. J. Surg. 2018, 105, e48–e60. [Google Scholar] [CrossRef]
- William, R.; Burch, R. The Principles of Humane Experimental Technique; Russell, W.M.S., Burch, R.L., Eds.; Johns Hopkins: Methuen, MA, USA, 1959. [Google Scholar]
PJS 1 | Pancreatitis | FAMMM 2 | Lynch Syndrome | HBOC 3 | FAP 4 | |
---|---|---|---|---|---|---|
Increased Risk | 132-fold | 69-fold | 13–22-fold | 8.6-fold | 3.5–10-fold | 4.5–6-fold |
Genes | STK11/LKB11 | PRSS1 SPINK1 CFTR | CDNK2A | MLH1 MSH2 MSH6 PMS2 | BRCA1 BRCA2 PALB2 | APC |
Pathways | AMPK/mTOR | Trypsin | Retinoblastoma | Mismatch repair | Homologous recombination repair | Wnt signalling |
Pathway Function | Cell growth Polarity Metabolism | Auto-activation of trypsin | G1 to S-phase checkpoint | Maintenance of genomic stability | Repair of double-strand breaks in DNA | Regulation of gene transcription |
Pathway/Gene Set | Pathway Reference | Study | Pathway p-Value |
---|---|---|---|
Maturity onset diabetes of the young | KEGG | [63] | 5.10 × 10−7 |
Regulation of Beta cell development | REACTOME | [63] | 1.92 × 10−6 |
Breast Cancer 17Q11 Q21 amplicon 1 | NIKOLSKY | [63] | 2.00 × 10−6 |
Role of EGF Receptor Transactivation by GPCRs in Cardiac Hypertrophy | BIOCARTA | [63] | 3.79 × 10−6 |
ATM Pearson Correlation Coefficient (PCC) Network 2 | PUJANA | [63] | 1.25 × 10−5 |
Pancreas development | [62] | 2.0 × 10−6 | |
Heliobacter pylori lacto/neolacto | [62] | 1.6 × 10−5 | |
Hedgehog | [62] | 0.0019 | |
Th1/Th2 immune response | [62] | 0.019 | |
Apoptosis | [62] | 0.023 |
Model | Representativeness of Patient Sample? | Usage | Maintenance | Success Rates Growth Rate | Cost |
---|---|---|---|---|---|
Established Cell Lines | Homogenic [88] Undergo genetic modifications [86] Fail to recapitulate the physiology of tumours | High throughput testing | Low maintenance Fast growing | Fast growing Commercially available | Low cost |
Primary Cell Cultures | Heterogenous [96] Early passage number [96] Representative of original tumour [96] | High throughput testing | Low maintenance Only grow for a limited number of passages [96] | Some commercially available lines Difficult to establish [97] | Low cost |
Organ-on-chip | Heterogenous [101] Allows for the study of the interactions of multiple cell/organ types [98] | Low throughput testing | Medium maintenance | High success rates | Chips are expensive High usage of media and drugs |
Organoids | Heterogenous [127] Tumours retain heterogeneity, genetics, and histological characteristics [142] | High throughput testing | Medium maintenance | Medium growing High success rates Established from small volumes of tumour [132] | Expensive ECM and media |
PDX 1 | Tumours retain heterogeneity, genetics, and histological characteristics [103] Replacement of human stroma with murine stroma [108] Orthotopic tumours in correct microenvironment [104] | In vivo and ex vivo drug testing | High maintenance Requires specialist training, and multiple licenses [106] | Slow growing (up to four months) Medium success rates | Expensive to maintain |
GEMM 2 | Tumours in correct microenvironment [109] Immune cells present [112] | In vivo and ex vivo drug testing Testing of immune targeted therapies | High maintenance Requires specialist training, and multiple licences | High success rates Slow growing—(up to 16 weeks) | Commercially available |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Nelson, S.R.; Walsh, N. Genetic Alterations Featuring Biological Models to Tailor Clinical Management of Pancreatic Cancer Patients. Cancers 2020, 12, 1233. https://doi.org/10.3390/cancers12051233
Nelson SR, Walsh N. Genetic Alterations Featuring Biological Models to Tailor Clinical Management of Pancreatic Cancer Patients. Cancers. 2020; 12(5):1233. https://doi.org/10.3390/cancers12051233
Chicago/Turabian StyleNelson, Shannon R., and Naomi Walsh. 2020. "Genetic Alterations Featuring Biological Models to Tailor Clinical Management of Pancreatic Cancer Patients" Cancers 12, no. 5: 1233. https://doi.org/10.3390/cancers12051233
APA StyleNelson, S. R., & Walsh, N. (2020). Genetic Alterations Featuring Biological Models to Tailor Clinical Management of Pancreatic Cancer Patients. Cancers, 12(5), 1233. https://doi.org/10.3390/cancers12051233