DNA Damage and Repair in Pancreatic Cancer—The Latest Findings
Abstract
1. Introduction
2. Materials and Methods
3. Pancreatic Cancer
3.1. The Incidence of Pancreatic Cancer
3.2. Pancreatic Carcinogenesis
3.3. Risk Factors for Pancreatic Cancer
3.4. Diagnosis, Treatment, and Survival
4. DNA Damage
4.1. Alkylation of Bases
4.2. Purine and Pyrimidine Dimers
4.3. DNA Adducts
4.4. ICL
4.5. Oxidative DNA Damage
4.6. DNA Strand Breaks
5. A General Overview of the DDR Pathways
5.1. BER
5.2. NER
5.3. NHEJ
5.4. HR
5.5. MMR
6. DDR and PDAC
7. Discussion
8. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Ma, Z.; Zhang, X.; Zhong, W.; Yi, H.; Chen, X.; Zhao, Y.; Ma, Y.; Song, E.; Xu, T. Deciphering early human pancreas development at the single-cell level. Nat. Commun. 2023, 14, 5354. [Google Scholar] [CrossRef]
- Grant, T.; Hua, K.; Singh, A. Molecular Pathogenesis of Pancreatic Cancer. Prog. Mol. Biol. Transl. Sci. 2016, 144, 241–275. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Mostafa, M.E.; Erbarut-Seven, I.; Pehlivanoglu, B.; Adsay, V. Pathologic classification of “pancreatic cancers”: Current concepts and challenges. Chin. Clin. Oncol. 2017, 6, 59. [Google Scholar] [CrossRef] [PubMed]
- Madura, J.A.; Jarman, B.T.; Doherty, M.G.; Yum, M.-N.; Howard, T.J. Adenosquamous carcinoma of the pancreas. Arch. Surg. 1999, 134, 599–603. [Google Scholar] [CrossRef]
- Muraki, T.; Reid, M.D.; Basturk, O.; Jang, K.-T.; Bedolla, G.; Bagci, P.; Mittal, P.; Memis, B.; Katabi, N.; Bandyopadhyay, S.; et al. Undifferentiated Carcinoma with Osteoclastic Giant Cells of the Pancreas: Clinicopathologic Analysis of 38 Cases Highlights a More Protracted Clinical Course Than Currently Appreciated. Am. J. Surg. Pathol. 2016, 40, 1203–1216. [Google Scholar] [CrossRef]
- Stoffel, E.M.; Brand, R.E.; Goggins, M. Pancreatic Cancer: Changing Epidemiology and New Approaches to Risk Assessment, Early Detection, and Prevention. Gastroenterology 2023, 164, 752–765. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hamdi, Y.; Abdeljaoued-Tej, I.; Zatchi, A.A.; Abdelhak, S.; Boubaker, S.; Brown, J.S.; Benkahla, A. Cancer in Africa: The Untold Story. Front. Oncol. 2021, 11, 650117. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Amri, F.; Koulali, H.; Jabi, R.; Zazour, A.; Bouziane, M.; Ismaili, Z.; Kharrasse, G. Pancreatic cancer: Experience from an emerging country in North Africa. J. Cancer Res. Clin. Oncol. 2023, 149, 14297–14302. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Koya, A.I.; Ibrahim, S.A. Carcinogenesis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025; Available online: https://www.ncbi.nlm.nih.gov/books/NBK604463/ (accessed on 2 October 2024).
- Weston, A.; Harris, C.C. Multistage Carcinogenesis. In Holland-Frei Cancer Medicine, 6th ed.; Kufe, D.W., Pollock, R.E., Weichselbaum, R.R., Bast, R.C., Gansler, T.S., Holland, J.F., Frei, E., III, Eds.; BC Decker: Hamilton, ON, USA, 2003; Available online: https://www.ncbi.nlm.nih.gov/books/NBK13982/ (accessed on 20 June 2020).
- Marks, F.; Fürstenberger, G.; Müller-Decker, K. Tumor Promotion as a Target of Cancer Prevention. In Cancer Prevention: Recent Results in Cancer Research; Senn, H.J., Kapp, U., Eds.; Springer: Berlin/Heidelberg, Germany, 2007; Volume 174. [Google Scholar] [CrossRef]
- Salvatore, V.; Teti, G.; Focaroli, S.; Mazzotti, M.C.; Mazzotti, A.; Falconi, M. The tumor microenvironment promotes cancer progression and cell migration. Oncotarget 2016, 8, 9608–9616. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- McGuigan, A.; Kelly, P.; Turkington, R.C.; Jones, C.; Coleman, H.G.; McCain, R.S. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J. Gastroenterol. 2018, 24, 4846–4861. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Brune, K.; Abe, T.; Canto, M.; O’Malley, L.; Klein, A.P.; Maitra, A.; Volkan Adsay, N.; Fishman, E.K.; Cameron, J.L.; Yeo, C.J.; et al. Multifocal neoplastic precursor lesions associated with lobular atrophy of the pancreas in patients having a strong family history of pancreatic cancer. Am. J. Surg. Pathol. 2006, 30, 1067–1076. [Google Scholar] [PubMed] [PubMed Central]
- GBD 2019 Demographics Collaborators. Global age-sex-specific fertility, mortality, healthy life expectancy (HALE), and population estimates in 204 countries and territories, 1950–2019: A comprehensive demographic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1160–1203. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10–27. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Li, Y.; He, Y.; Peng, J.; Su, Z.; Li, Z.; Zhang, B.; Ma, J.; Zhuo, M.; Zou, D.; Liu, X.; et al. Mutant Kras co-opts a proto-oncogenic enhancer network in inflammation-induced metaplastic progenitor cells to initiate pancreatic cancer. Nat. Cancer 2020, 2, 49–65. [Google Scholar] [CrossRef] [PubMed]
- Gentiluomo, M.; Corradi, C.; Arcidiacono, P.G.; Crippa, S.; Falconi, M.; Belfiori, G.; Farinella, R.; Apadula, L.; Lauri, G.; Bina, N.; et al. Role of pancreatic ductal adenocarcinoma risk factors in intraductal papillary mucinous neoplasm progression. Front. Oncol. 2023, 13, 1172606. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Xia, Q.; Li, F.; Min, R.; Sun, S.; Han, Y.X.; Feng, Z.Z.; Li, N. Malignancy risk factors and prognostic variables of pancreatic mucinous cystic neoplasms in Chinese patients. World J. Gastroenterol. 2023, 29, 3119–3132. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Xie, W.; Liang, H.; Guo, Y.; Xiao, S.-Y. Update on mucinous cystic neoplasm of the pancreas: A narrative review. J. Pancreatol. 2021, 4, 115–121. [Google Scholar] [CrossRef]
- Martin, R.C.; McFarland, K.; Ellis, S.; Velanovich, V. Irreversible electroporation therapy in the management of locally advanced pancreatic adenocarcinoma. J. Am. Coll. Surg. 2012, 215, 361–369. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Jung, X.; Hines, O.J.; Eibl, G.; Chen, Y. Obesity and Pancreatic Cancer: Overview of Epidemiology and Potential Prevention by Weight Loss. Pancreas 2018, 47, 158–162. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yuan, C.; Morales-Oyarvide, V.; Babic, A.; Clish, C.B.; Kraft, P.; Bao, Y.; Qian, Z.R.; Rubinson, D.A.; Ng, K.; Giovannucci, E.L.; et al. Cigarette Smoking and Pancreatic Cancer Survival. J. Clin. Oncol. 2017, 35, 1822–1828. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zanini, S.; Renzi, S.; Limongi, A.R.; Bellavite, P.; Giovinazzo, F.; Bermano, G. A review of lifestyle and environment risk factors for pancreatic cancer. Eur. J. Cancer 2021, 145, 53–70. [Google Scholar] [CrossRef] [PubMed]
- Ilic, M.; Ilic, I. Epidemiology of pancreatic cancer. World J. Gastroenterol. 2016, 22, 9694–9705. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Moshayedi, N.; Escobedo, A.L.; Thomassian, S.; Osipov, A.; Hendifar, A.E. Race, sex, age, and geographic disparities in pancreatic cancer incidence. J. Clin. Oncol. 2022, 40, 520. [Google Scholar] [CrossRef]
- Hawksworth, G.; Hales, J.; Martinez, F.; Hynes, A.; Hamilton, A.; Fernandez, V. Pancreatic cancer trends in Europe: Epidemiology and risk factors. Med. Stud. 2019, 35, 164–171. [Google Scholar] [CrossRef]
- Liew, S.Z.H.; Ng, K.W.; Ishak, N.D.B.; Lee, S.Y.; Zhang, Z.; Chiang, J.; Ngeow, J.Y.Y. Geographical, ethnic, and genetic differences in pancreatic cancer predisposition. Chin. Clin. Oncol. 2023, 12, 27. [Google Scholar] [CrossRef]
- Sadr-Azodi, O.; Konings, P.; Brusselaers, N. Menopausal hormone therapy and pancreatic cancer risk in women: A population-based matched cohort study. United Eur. Gastroenterol. 2017, 5, 1123–1128. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Andersson, G.; Borgquist, S.; Jirström, K. Hormonal factors and pancreatic cancer risk in women: The Malmö Diet and Cancer Study. Int. J. Cancer 2018, 143, 52–62. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lee, E.; Horn-Ross, P.L.; Rull, R.P.; Neuhausen, S.L.; Anton-Culver, H.; Ursin, G.; Henderson, K.D.; Bernstein, L. Reproductive factors, exogenous hormones, and pancreatic cancer risk in the CTS. Am. J. Epidemiol. 2013, 178, 1403–1413. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Capasso, M.; Franceschi, M.; Rodriguez-Castro, K.I.; Crafa, P.; Cambiè, G.; Miraglia, C.; Barchi, A.; Nouvenne, A.; Leandro, G.; Meschi, T.; et al. Epidemiology and risk factors of pancreatic cancer. Acta Biomed. 2018, 89, 141–146. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pandol, S.J.; Apte, M.V.; Wilson, J.S.; Gukovskaya, A.S.; Edderkaoui, M. The burning question: Why is smoking a risk factor for pancreatic cancer? Pancreatology 2012, 12, 344–349. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Weissman, S.; Takakura, K.; Eibl, G.; Pandol, S.J.; Saruta, M. The Diverse Involvement of Cigarette Smoking in Pancreatic Cancer Development and Prognosis. Pancreas 2020, 49, 612–620. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Tang, M.S.; Lee, H.W.; Weng, M.W.; Wang, H.T.; Hu, Y.; Chen, L.C.; Park, S.H.; Chan, H.W.; Xu, J.; Wu, X.R.; et al. DNA damage, DNA repair and carcinogenicity: Tobacco smoke versus electronic cigarette aerosol. Mutat. Res. Mol. Mech. Mutagen. 2022, 789, 108409. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yamaguchi, N.H. Smoking, immunity, and DNA damage. Transl. Lung Cancer Res. 2019, 8 (Suppl. S1), S3–S6. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hirano, T. Alcohol consumption and oxidative DNA damage. Int. J. Environ. Res. Public Health 2011, 8, 2895–2906. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Gallina, I.; Duxin, J.P. A safe fix for alcohol-derived DNA damage. Nature 2020, 579, 499–500. [Google Scholar] [CrossRef] [PubMed]
- Anderson, M.A.; Zolotarevsky, E.; Cooper, K.L.; Sherman, S.; Shats, O.; Whitcomb, D.C.; Lynch, H.T.; Ghiorzo, P.; Rubinstein, W.S.; Vogel, K.J.; et al. Alcohol and tobacco lower the age of presentation in sporadic pancreatic cancer in a dose-dependent manner: A multicenter study. Am. J. Gastroenterol. 2012, 107, 1730–1739. [Google Scholar] [CrossRef]
- Genkinger, J.M.; Spiegelman, D.; Anderson, K.E.; Bergkvist, L.; Bernstein, L.; van den Brandt, P.A.; English, D.R.; Freudenheim, J.L.; Fuchs, C.S.; Giles, G.G.; et al. Alcohol intake and pancreatic cancer risk: A pooled analysis of fourteen cohort studies. Cancer Epidemiol. Biomark. Prev. 2009, 18, 765–776. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Madela, F.; Ferndale, L.; Aldous, C. Diagnostic Differentiation between Pancreatitis and Pancreatic Cancer: A Scoping Review. Diagnostics 2024, 14, 290. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Klein, S.; Gastaldelli, A.; Yki-Järvinen, H.; Scherer, P.E. Why does obesity cause diabetes? Cell Metab. 2022, 34, 11–20. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Toledo, F.G.; Chari, S.; Yadav, D. Understanding the Contribution of Insulin Resistance to the Risk of Pancreatic Cancer. Am. J. Gastroenterol. 2021, 116, 669–670. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Włodarczyk, M.; Nowicka, G. Obesity, DNA Damage, and Development of Obesity-Related Diseases. Int. J. Mol. Sci. 2019, 20, 1146. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Valentino, E.; Bellazzo, A.; Di Minin, G.; Sicari, D.; Apollonio, M.; Scognamiglio, G.; Di Bonito, M.; Botti, G.; Del Sal, G.; Collavin, L. Mutant p53 potentiates the oncogenic effects of insulin by inhibiting the tumor suppressor DAB2IP. Proc. Natl. Acad. Sci. USA 2017, 114, 7623–7628. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chang, J.L.; Chen, G.; Ulrich, C.M.; Bigler, J.; King, I.B.; Schwarz, Y.; Li, S.; Li, L.; Potter, J.D.; Lampe, J.W. DNA damage and repair: Fruit and vegetable effects in a feeding trial. Nutr. Cancer 2010, 62, 329–335. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wondmkun, Y.T. Obesity, Insulin Resistance, and Type 2 Diabetes: Associations and Therapeutic Implications. Diabetes Metab. Syndr. Obes. 2020, 13, 3611–3616. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Riondino, S.; Roselli, M.; Palmirotta, R.; Della-Morte, D.; Ferroni, P.; Guadagni, F. Obesity and colorectal cancer: Role of adipokines in tumor initiation and progression. World J. Gastroenterol. 2014, 20, 5177–5190. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Stattin, P.; Lukanova, A.; Biessy, C.; Söderberg, S.; Palmqvist, R.; Kaaks, R.; Olsson, T.; Jellum, E. Obesity and colon cancer: Does leptin provide a link? Int. J. Cancer 2004, 109, 149–152. [Google Scholar] [CrossRef] [PubMed]
- Kanji, Z.S.; Gallinger, S. Diagnosis and management of pancreatic cancer. CMAJ. 2013, 185, 1219–1226. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Bond-Smith, G.; Banga, N.; Hammond, T.M.; Imber, C.J. Pancreatic adenocarcinoma. BMJ 2012, 344, e2476. [Google Scholar] [CrossRef]
- Lee, E.S.; Lee, J.M. Imaging diagnosis of pancreatic cancer: A state-of-the-art review. World J. Gastroenterol. 2014, 20, 7864–7877. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhang, L.; Sanagapalli, S.; Stoita, A. Challenges in diagnosis of pancreatic cancer. World J. Gastroenterol. 2014, 20, 7864–7877. [Google Scholar] [CrossRef] [PubMed]
- Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef] [PubMed]
- Pereira, N.P.; Corrêa, J.R. Pancreatic cancer: Treatment approaches and trends. J. Cancer Metastasis Treat. 2018, 4, 30. [Google Scholar] [CrossRef]
- Weaver, B.A. How Taxol/paclitaxel kills cancer cells. Mol. Biol. Cell 2014, 25, 2677–2681. [Google Scholar] [CrossRef] [PubMed]
- Winter, J.M.; Cameron, J.L.; Campbell, K.A.; Arnold, M.A.; Chang, D.C.; Coleman, J.; Hodgin, M.B.; Sauter, P.K.; Hruban, R.H.; Riall, T.S.; et al. 1423 pancreaticoduodenectomies for pancreatic cancer: A single-institution experience. J. Gastrointest. Surg. 2006, 10, 1199–1210. [Google Scholar] [CrossRef] [PubMed]
- Kolbeinsson, H.; Hoppe, A.; Bayat, A.; Kogelschatz, B.; Mbanugo, C.; Chung, M.; Wolf, A.; Assifi, M.M.; Wright, G.P. Recurrence patterns and postrecurrence survival after curative intent resection for pancreatic ductal adenocarcinoma. Surgery 2021, 169, 649–654. [Google Scholar] [CrossRef] [PubMed]
- Groot, V.P.; Rezaee, N.; Wu, W.; Cameron, J.L.; Fishman, E.K.; Hruban, R.H.; Weiss, M.J.; Zheng, L.; Wolfgang, C.L.; He, J. Patterns, Timing, and Predictors of Recurrence Following Pancreatectomy for Pancreatic Ductal Adenocarcinoma. Ann. Surg. 2018, 267, 936–945. [Google Scholar] [CrossRef]
- Loveday, B.P.; Lipton, L.; Thomson, B.N. Pancreatic cancer: An update on diagnosis and management. Aust. J. Gen. Pract. 2019, 48, 826–831. [Google Scholar] [CrossRef] [PubMed]
- Halbrook, C.J.; Lyssiotis, C.A.; Pasca di Magliano, M.; Maitra, A. Pancreatic cancer: Advances and challenges. Cell 2023, 186, 1729–1754. [Google Scholar] [CrossRef]
- Duan, H.; Li, L.; He, S. Advances and Prospects in the Treatment of Pancreatic Cancer. Int. J. Nanomed. 2023, 18, 3973–3988. [Google Scholar] [CrossRef]
- Gavas, S.; Quazi, S.; Karpiński, T.M. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lindahl, T. Instability and decay of the primary structure of DNA. Nature 1993, 362, 709–715. [Google Scholar] [CrossRef]
- Lindahl, T.; Barnes, D.E. Repair of endogenous DNA damage. Cold Spring Harb. Symp. Quant. Biol. 2000, 65, 127–133. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Rocha, H.; Garcia-Garcia, A.; Panayiotidis, M.I.; Franco, R. DNA damage and autophagy. Mutat. Res. Mol. Mech. Mutagen. 2011, 711, 158–166. [Google Scholar] [CrossRef] [PubMed]
- Tubbs, A.; Nussenzweig, A. Endogenous DNA Damage as a Source of Genomic Instability in Cancer. Cell 2017, 168, 644–656. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Fu, D.; Calvo, J.A.; Samson, L.D. Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat. Rev. Cancer 2012, 12, 104–120. [Google Scholar] [CrossRef]
- Grady, W.M.; Ulrich, C.M. DNA alkylation and DNA methylation: Cooperating mechanisms driving the formation of colorectal adenomas and adenocarcinomas? Gut 2007, 56, 318–320. [Google Scholar] [CrossRef]
- Povey, A.C.; Badawi, A.F.; Cooper, D.P.; Hall, C.N.; Harrison, K.L.; Jackson, P.E.; Lees, N.P.; O’Connor, P.J.; Margison, G.P. DNA alkylation and repair in the large bowel: Animal and human studies. J. Nutr. 2002, 132 (Suppl. S11), 3518S–3521S. [Google Scholar] [CrossRef]
- Delaney, J.C.; Essigmann, J.M. Mutagenesis, genotoxicity, and repair of 1-methyladenine, 3-alkylcytosines, 1-methylguanine, and 3-methylthymine in alkB Escherichia coli. Proc. Natl. Acad. Sci. USA 2004, 101, 14051–14056. [Google Scholar] [CrossRef]
- Tessmer, I.; Margison, G.P. The DNA Alkyltransferase Family of DNA Repair Proteins: Common Mechanisms, Diverse Functions. Int. J. Mol. Sci. 2024, 25, 463. [Google Scholar] [CrossRef]
- Kaina, B.; Ziouta, A.; Ochs, K.; Coquerelle, T. Chromosomal instability, reproductive cell death and apoptosis induced by O6-methylguanine in Mex-, Mex+ and methylation-tolerant mismatch repair compromised cells: Facts and models. Mutat. Res. 1997, 381, 227–241. [Google Scholar] [CrossRef] [PubMed]
- Hung, K.F.; Sidorova, J.M.; Nghiem, P.; Kawasumi, M. The 6-4 photoproduct is the trigger of UV-induced replication blockage and ATR activation. Proc. Natl. Acad. Sci. USA 2020, 117, 12806–12816. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Rastogi, R.P.; Richa; Kumar, A.; Tyagi, M.B.; Sinha, R.P. Molecular mechanisms of ultraviolet radiation-induced DNA damage and repair. J. Nucleic Acids 2010, 2010, 592980. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Douki, T.; Sage, E. Dewar valence isomers, the third type of environmentally relevant DNA photoproducts induced by solar radiation. Photochem. Photobiol. Sci. 2016, 15, 24–30. [Google Scholar] [CrossRef] [PubMed]
- Carell, T.; Epple, R. Repair of UV Light Induced DNA Lesions: A Comparative Study with Model Compounds. Eur. J. Org. Chem. 1998, 1998, 1245–1258. [Google Scholar] [CrossRef]
- Stark, B.; Poon, G.M.; Wyrick, J.J. Molecular mechanism of UV damage modulation in nucleosomes. Comput. Struct. Biotechnol. J. 2022, 20, 5393–5400. [Google Scholar] [CrossRef]
- Kciuk, M.; Marciniak, B.; Mojzych, M.; Kontek, R. Focus on UV-Induced DNA Damage and Repair-Disease Relevance and Protective Strategies. Int. J. Mol. Sci. 2020, 21, 7264. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sinha, R.P.; Häder, D.-P. UV-induced DNA damage and repair: A review. Photochem. Photobiol. Sci. 2002, 1, 225–236. [Google Scholar] [CrossRef]
- Borrego-Soto, G.; Ortiz-López, R.; Rojas-Martínez, A. Ionizing radiation-induced DNA injury and damage detection in patients with breast cancer. Genet. Mol. Biol. 2015, 38, 420–432. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Redon, C.E.; Nakamura, A.J.; Zhang, Y.-W.; Ji, J.J.; Bonner, W.M.; Kinders, R.J.; Parchment, R.E.; Doroshow, J.H.; Pommier, Y. Histone gammaH2AX and poly(ADP-ribose) as clinical pharmacodynamic biomarkers. Clin. Cancer Res. 2010, 16, 4532–4542. [Google Scholar] [CrossRef]
- Aparicio, T.; Baer, R.; Gautier, J. DNA double-strand break repair pathway choice and cancer. DNA Repair 2014, 19, 169–175. [Google Scholar] [CrossRef] [PubMed]
- Brown, L.C.; Mutter, R.W.; Halyard, M.Y. Benefits, risks, and safety of external beam radiation therapy for breast cáncer. Int. J. Womens Health 2015, 7, 449–458. [Google Scholar] [CrossRef] [PubMed]
- Tran, B.; Whiteman, D.C.; Webb, P.M.; Fritschi, L.; Fawcett, J.; Risch, H.A.; Lucas, R.; Pandeya, N.; Schulte, A.; Neale, R.E.; et al. Association between ultraviolet radiation, skin sun sensitivity and risk of pancreatic cancer. Cancer Epidemiol. 2013, 37, 886–892. [Google Scholar] [CrossRef] [PubMed]
- Rajalakshmi, T.R.; AravindhaBabu, N.; Shanmugam, K.T.; Masthan, K.M. DNA adducts-chemical addons. J. Pharm. Bioallied Sci. 2015, 7 (Suppl. S1), S197–S199. [Google Scholar] [CrossRef]
- Peluso, M.; Airoldi, L.; Munnia, A.; Colombi, A.; Veglia, F.; Autrup, H.; Dunning, A.; Garte, S.; Gormally, E.; Malaveille, C.; et al. Bulky DNA adducts, 4-aminobiphenyl-haemoglobin adducts and diet in the European Prospective Investigation into Cancer and Nutrition (EPIC) prospective study. Br. J. Nutr. 2008, 100, 489–495. [Google Scholar] [CrossRef]
- Hashimoto, S.; Anai, H.; Hanada, K. Mechanisms of interstrand DNA crosslink repair and human disorders. Genes Environ. 2016, 38, 1–8. [Google Scholar] [CrossRef]
- Kumar, A.; Pathak, H.; Bhadauria, S.; Sudan, J. Aflatoxin contamination in food crops: Causes, detection, and management: A review. Food Prod. Process. Nutr. 2021, 3, 17. [Google Scholar] [CrossRef]
- Li, D.; Firozi, P.F.; Zhang, W.; Shen, J.; DiGiovanni, J.; Lau, S.; Evans, D.; Friess, H.; Hassan, M.; Abbruzzese, J.L. DNA adducts, genetic polymorphisms, and K-ras mutation in human pancreatic cancer. Mutat. Res. 2001, 513, 37–48. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Abbruzzese, J.L.; Friess, H.; Hittelman, W.N.; Evans, D.B.; Abbruzzese, M.C.; Chiao, P.; Li, D. DNA adducts in human pancreatic tissues and their potential role in carcinogenesis. Cancer Res. 1998, 58, 38–41. [Google Scholar] [PubMed]
- McCabe, K.M.; Olson, S.B.; Moses, R.E. DNA interstrand crosslink repair in mammalian cells. J. Cell Physiol. 2009, 220, 569–573. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Deans, A.J.; West, S.C. DNA interstrand crosslink repair and cancer. Nat. Rev. Cancer 2011, 11, 467–480. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ruggiano, A.; Ramadan, K. DNA-protein crosslink proteases in genome stability. Commun. Biol. 2021, 4, 11. [Google Scholar] [CrossRef]
- Klages-Mundt, N.L.; Li, L. Formation and repair of DNA-protein crosslink damage. Sci. China Life Sci. 2017, 60, 1065–1076. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhang, H.; Xiong, Y.; Chen, J. DNA-protein cross-link repair: What do we know now? Cell Biosci. 2020, 10, 3. [Google Scholar] [CrossRef]
- Teleanu, D.M.; Niculescu, A.G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709, Correction in Nat. Rev. Drug Discov. 2021, 20, 652. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kalisz, O.; Wolski, T.; Gerkowicz, M.; Smorawski, M. Reaktywne formy tlenu (RFT) oraz ich rola w patogenezie niektórych chorób. Ann. Univ. Mariae Curie-Skłodowska Sect. DD Med. Vet. 2007, 62, 87–99. [Google Scholar]
- Burton, G.J.; Jauniaux, E. Oxidative stress. Best Pract. Res. Clin. Obstet. Gynaecol. 2011, 25, 287–299. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Salmon, T.B.; Evert, B.A.; Song, B.; Doetsch, P.W. Biological consequences of oxidative stress-induced DNA damage in Saccharomyces cerevisiae. Nucleic Acids Res. 2004, 32, 3712–3723. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cadet, J.; Davies, K.J.; Medeiros, M.H.; Di Mascio, P.; Wagner, J.R. Formation and repair of oxidatively generated damage in cellular DNA. Free. Radic. Biol. Med. 2017, 107, 13–34. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Di Mascio, P.; Martinez, G.R.; Miyamoto, S.; Ronsein, G.E.; Medeiros, M.H.G.; Cadet, J. Singlet Molecular Oxygen Reactions with Nucleic Acids, Lipids, and Proteins. Chem. Rev. 2019, 119, 2043–2086. [Google Scholar] [CrossRef] [PubMed]
- Ramon, O.; Sauvaigo, S.; Gasparutto, D.; Faure, P.; Favier, A.; Cadet, J. Effects of 8-oxo-7,8-dihydro-2’-deoxyguanosine on the binding of the transcription factor Sp1 to its cognate target DNA sequence (GC box). Free Radic. Res. 1999, 31, 217–229. [Google Scholar] [CrossRef] [PubMed]
- Larsen, E.; Kwon, K.; Coin, F.; Egly, J.M.; Klungland, A. Transcription activities at 8-oxoG lesions in DNA. DNA Repair 2004, 3, 1457–1468. [Google Scholar] [CrossRef] [PubMed]
- Poetsch, A.R. The genomics of oxidative DNA damage, repair, and resulting mutagenesis. Comput. Struct. Biotechnol. J. 2020, 18, 207–219. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ock, C.Y.; Kim, E.H.; Choi, D.J.; Lee, H.J.; Hahm, K.B.; Chung, M.H. 8-Hydroxydeoxyguanosine: Not mere biomarker for oxidative stress, but remedy for oxidative stress-implicated gastrointestinal diseases. World J. Gastroenterol. 2012, 18, 302–308. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Mohamadkhani, A.; Pourshams, A.; Viti, J.; Cellai, F.; Mortazavi, K.; Sharafkhah, M.; Sotoudeh, M.; Malekzadeh, R.; Boffetta, P.; Peluso, M. Pancreatic Cancer is Associated with Peripheral Leukocyte Oxidative DNA Damage. Asian Pac. J. Cancer Prev. 2017, 18, 1349–1355. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Caldecott, K.W. Causes and consequences of DNA single-strand breaks. Trends Biochem. Sci. 2024, 49, 68–78. [Google Scholar] [CrossRef] [PubMed]
- Caldecott, K.W. Mammalian DNA base excision repair: Dancing in the moonlight. DNA Repair 2020, 93, 102921. [Google Scholar] [CrossRef] [PubMed]
- Blasiak, J. Single-Strand Annealing in Cancer. Int. J. Mol. Sci. 2021, 22, 2167. [Google Scholar] [CrossRef]
- Atkinson, J.; Bezak, E.; Le, H.; Kempson, I. DNA Double Strand Break and Response Fluorescent Assays: Choices and Interpretation. Int. J. Mol. Sci. 2024, 25, 2227. [Google Scholar] [CrossRef] [PubMed]
- van Gent, D.C.; Hoeijmakers, J.H.; Kanaar, R. Chromosomal stability and the DNA double-stranded break connection. Nat. Rev. Genet. 2001, 2, 196–206. [Google Scholar] [CrossRef] [PubMed]
- da Silva, M.S. DNA Double-Strand Breaks: A Double-Edged Sword for Trypanosomatids. Front. Cell Dev. Biol. 2021, 9, 669041. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Varga, T.; Aplan, P.D. Chromosomal aberrations induced by double strand DNA breaks. DNA Repair 2005, 4, 1038–1046. [Google Scholar] [CrossRef]
- Li, Y.H.; Wang, X.; Pan, Y.; Lee, D.H.; Chowdhury, D.; Kimmelman, A.C. Inhibition of non-homologous end joining repair impairs pancreatic cancer growth and enhances radiation response. PLoS ONE 2012, 7, e39588. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Huang, R.; Zhou, P.-K. DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 254. [Google Scholar] [CrossRef]
- Nakad, R.; Schumacher, B. DNA Damage Response and Immune Defense: Links and Mechanisms. Front. Genet. 2016, 7, 147. [Google Scholar] [CrossRef]
- Ciccia, A.; Elledge, S.J. The DNA damage response: Making it safe to play with knives. Mol. Cell 2010, 40, 179–204. [Google Scholar] [CrossRef]
- Zhou, B.B.; Elledge, S.J. The DNA damage response: Putting checkpoints in perspective. Nature 2000, 408, 433–439. [Google Scholar] [CrossRef]
- Alhmoud, J.F.; Woolley, J.F.; Al Moustafa, A.E.; Malki, M.I. DNA Damage/Repair Management in Cancers. Cancers 2020, 12, 1050. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pan, M.-R.; Li, K.; Lin, S.-Y.; Hung, W.-C. Connecting the Dots: From DNA Damage and Repair to Aging. Int. J. Mol. Sci. 2016, 17, 685. [Google Scholar] [CrossRef] [PubMed]
- Krokan, H.E.; Bjørås, M. Base excision repair. Cold Spring Harb. Perspect. Biol. 2013, 5, a012583. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Grigory, L.; Hübscher, U. Mammalian Base Excision Repair: The Forgotten Archangel. Nucleic Acids Res. 2013, 41, 3483–3490. [Google Scholar] [CrossRef]
- Woodrick, J.; Gupta, S.; Camacho, S.; Parvathaneni, S.; Choudhury, S.; Cheema, A.; Bai, Y.; Khatkar, P.; Erkizan, H.V.; Sami, F.; et al. A new sub-pathway of long-patch base excision repair involving 5’ gap formation. EMBO J. 2017, 36, 1605–1622. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Grundy, G.J.; Parsons, J.L. Base excision repair and its implications to cancer therapy. Essays Biochem. 2020, 64, 831–843, Correction in Essays Biochem. 2020, 64, 845. [Google Scholar] [CrossRef] [PubMed Central]
- Cappelli, E.; Taylor, R.; Cevasco, M.; Abbondandolo, A.; Caldecott, K.; Frosina, G. Involvement of XRCC1 and DNA ligase III gene products in DNA base excision repair. J. Biol. Chem. 1997, 272, 23970–23975. [Google Scholar] [CrossRef] [PubMed]
- Sattler, U.; Frit, P.; Salles, B.; Calsou, P. Long-patch DNA repair synthesis during base excision repair in mammalian cells. EMBO Rep. 2003, 4, 363–367. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Krasikova, Y.; Rechkunova, N.; Lavrik, O. Nucleotide Excision Repair: From Molecular Defects to Neurological Abnormalities. Int. J. Mol. Sci. 2021, 22, 6220. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kusakabe, M.; Onishi, Y.; Tada, H.; Kurihara, F.; Kusao, K.; Furukawa, M.; Iwai, S.; Yokoi, M.; Sakai, W.; Sugasawa, K. Mechanism and regulation of DNA damage recognition in nucleotide excision repair. Genes Environ. 2019, 41, 2. [Google Scholar] [CrossRef]
- Lee, T.H.; Kang, T.H. DNA Oxidation and Excision Repair Pathways. Int. J. Mol. Sci. 2019, 20, 6092. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Marteijn, J.A.; Lans, H.; Vermeulen, W.; Hoeijmakers, J.H.J. Understanding nucleotide excision repair and its roles in cancer and ageing. Nat. Rev. Mol. Cell Biol. 2014, 15, 465–481. [Google Scholar] [CrossRef]
- Riedl, T.; Hanaoka, F.; Egly, J.M. The comings and goings of nucleotide excision repair factors on damaged DNA. EMBO J. 2003, 22, 5293–5303. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sugasawa, K.; Akagi, J.; Nishi, R.; Iwai, S.; Hanaoka, F. Two-step recognition of DNA damage for mammalian nucleotide excision repair: Directional binding of the XPC complex and DNA strand scanning. Mol. Cell 2009, 36, 642–653. [Google Scholar] [CrossRef] [PubMed]
- Fagbemi, A.F.; Orelli, B.; Schärer, O.D. Regulation of endonuclease activity in human nucleotide excision repair. DNA Repair 2011, 10, 722–729. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Schärer, O.D. Nucleotide excision repair in eukaryotes. Cold Spring Harb. Perspect. Biol. 2013, 5, a012609. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Mocquet, V.; Lainé, J.P.; Riedl, T.; Yajin, Z.; Lee, M.Y.; Egly, J.M. Sequential recruitment of the repair factors during NER: The role of XPG in initiating the resynthesis step. EMBO J. 2008, 27, 155–167. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Vermeulen, W.; Fousteri, M. Mammalian transcription-coupled excision repair. Cold Spring Harb. Perspect. Biol. 2013, 5, a012625. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Duan, M.; Speer, R.M.; Ulibarri, J.; Liu, K.J.; Mao, P. Transcription-coupled nucleotide excision repair: New insights revealed by genomic approaches. DNA Repair 2021, 103, 103126. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Xu, X.; An, H.; Wu, C.; Sang, R.; Wu, L.; Lou, Y.; Yang, X.; Xi, Y. HR repair pathway plays a crucial role in maintaining neural stem cell fate under irradiation stress. Life Sci. Alliance 2023, 6, e202201802. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Stinson, B.M.; Loparo, J.J. Repair of DNA Double-Strand Breaks by the Nonhomologous End Joining Pathway. Annu. Rev. Biochem. 2021, 90, 137–164. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Walker, J.R.; Corpina, R.A.; Goldberg, J. Structure of the Ku heterodimer bound to DNA and its implications for double-strand break repair. Nature 2001, 412, 607–614. [Google Scholar] [CrossRef] [PubMed]
- Davis, A.J.; Chen, D.J. DNA double strand break repair via non-homologous end-joining. Transl. Cancer Res. 2013, 2, 130–143. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Falck, J.; Coates, J.; Jackson, S.P. Conserved modes of recruitment of ATM, ATR and DNA-PKcs to sites of DNA damage. Nature 2005, 434, 605–611. [Google Scholar] [CrossRef] [PubMed]
- Watanabe, G.; Lieber, M.R. Dynamics of the Artemis and DNA-PKcs Complex in the Repair of Double-Strand Breaks. J. Mol. Biol. 2022, 434, 167858. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhao, B.; Watanabe, G.; Lieber, M.R. Polymerase μ in non-homologous DNA end joining: Importance of the order of arrival at a double-strand break in a purified system. Nucleic Acids Res. 2020, 48, 3605–3618. [Google Scholar] [CrossRef]
- Li, X.; Heyer, W.-D. Homologous recombination in DNA repair and DNA damage tolerance. Cell Res. 2008, 18, 99–113. [Google Scholar] [CrossRef]
- van Wilpe, S.; Tolmeijer, S.H.; Koornstra, R.H.T.; de Vries, I.J.M.; Gerritsen, W.R.; Ligtenberg, M.; Mehra, N. Homologous Recombination Repair Deficiency and Implications for Tumor Immunogenicity. Cancers 2021, 13, 2249. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Elbakry, A.; Löbrich, M. Homologous Recombination Subpathways: A Tangle to Resolve. Front. Genet. 2021, 12, 723847. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Krejci, L.; Altmannova, V.; Spirek, M.; Zhao, X. Homologous recombination and its regulation. Nucleic Acids Res. 2012, 40, 5795–5818. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Rahimian, E.; Amini, A.; Alikarami, F.; Pezeshki, S.M.S.; Saki, N.; Safa, M. DNA repair pathways as guardians of the genome: Therapeutic potential and possible prognostic role in hematologic neoplasms. DNA Repair 2020, 96, 102951. [Google Scholar] [CrossRef] [PubMed]
- Kolinjivadi, A.M.; Sannino, V.; de Antoni, A.; Técher, H.; Baldi, G.; Costanzo, V. Moonlighting at replication forks—A new life for homologous recombination proteins BRCA1, BRCA2 and RAD51. FEBS Lett. 2017, 591, 1083–1100. [Google Scholar] [CrossRef] [PubMed]
- Talens, F.; Jalving, M.; Gietema, J.A.; Van Vugt, M.A. Therapeutic targeting and patient selection for cancers with homologous recombination defects. Expert Opin. Drug Discov. 2017, 12, 565–581. [Google Scholar] [CrossRef]
- Li, G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2007, 18, 85–98. [Google Scholar] [CrossRef]
- Hsieh, P.; Yamane, K. DNA mismatch repair: Molecular mechanism, cancer, and ageing. Mech. Ageing Dev. 2008, 129, 391–407. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chakraborty, U.; Alani, E. Understanding how mismatch repair proteins participate in the repair/anti-recombination decision. FEMS Yeast Res. 2016, 16, fow071. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lózsa, R.; Németh, E.; Gervai, J.Z.; Márkus, B.G.; Kollarics, S.; Gyüre, Z.; Tóth, J.; Simon, F.; Szüts, D. DNA mismatch repair protects the genome from oxygen-induced replicative mutagenesis. Nucleic Acids Res. 2023, 51, 11040–11055. [Google Scholar] [CrossRef] [PubMed]
- Shindo, K.; Yu, J.; Suenaga, M.; Fesharakizadeh, S.; Cho, C.; Macgregor-Das, A.; Siddiqui, A.; Witmer, P.D.; Tamura, K.; Song, T.J.; et al. Deleterious Germline Mutations in Patients With Apparently Sporadic Pancreatic Adenocarcinoma. J. Clin. Oncol. 2017, 35, 3382–3390. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.O.; Hochhauser, D.; Arnold, D.; Oh, D.Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hu, C.; Hart, S.N.; Polley, E.C.; Gnanaolivu, R.; Shimelis, H.; Lee, K.Y.; Lilyquist, J.; Na, J.; Moore, R.; Antwi, S.O.; et al. Association Between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. JAMA 2018, 319, 2401–2409. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Waddell, N.; Pajic, M.; Patch, A.M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lord, C.J.; Ashworth, A. BRCAness revisited. Nat. Rev. Cancer 2016, 16, 110–120. [Google Scholar] [CrossRef] [PubMed]
- Laurini, E.; Marson, D.; Fermeglia, A.; Aulic, S.; Fermeglia, M.; Pricl, S. Role of Rad51 and DNA repair in cancer: A molecular perspective. Pharmacol. Ther. 2020, 208, 107492. [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, Erratum in Cancer Res. 2014, 74, 4006. [Google Scholar] [CrossRef] [PubMed]
- Parsa, N. Environmental factors inducing human cancers. Iran. J. Public Health 2012, 41, 1–9. [Google Scholar] [PubMed] [PubMed Central]
- Jones, S.; Zhang, X.; Parsons, D.W.; Lin, J.C.; 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] [PubMed Central]
- Stephens, P.J.; Greenman, C.D.; Fu, B.; Yang, F.; Bignell, G.R.; Mudie, L.J.; Pleasance, E.D.; Lau, K.W.; Beare, D.; Stebbings, L.A.; et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 2011, 144, 27–40. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Perkhofer, L.; Gout, J.; Roger, E.; Kude de Almeida, F.; Baptista Simões, C.; Wiesmüller, L.; Seufferlein, T.; Kleger, A. DNA damage repair as a target in pancreatic cancer: State-of-the-art and future perspectives. Gut 2021, 70, 606–617. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Russell, R.; Perkhofer, L.; Liebau, S.; Lin, Q.; Lechel, A.; Feld, F.M.; Hessmann, E.; Gaedcke, J.; Güthle, M.; Zenke, M.; et al. Loss of ATM accelerates pancreatic cancer formation and epithelial-mesenchymal transition. Nat. Commun. 2015, 6, 7677. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Buchberg, J.; de Stricker, K.; Pfeiffer, P.; Mortensen, M.B.; Detlefsen, S. Mutational profiling of 103 unresectable pancreatic ductal adenocarcinomas using EUS-guided fine-needle biopsy. Endosc. Ultrasound 2024, 13, 154–164. [Google Scholar] [CrossRef]
- Lai, E.; Ziranu, P.; Spanu, D.; Dubois, M.; Pretta, A.; Tolu, S.; Camera, S.; Liscia, N.; Mariani, S.; Persano, M.; et al. BRCA-mutant pancreatic ductal adenocarcinoma. Br. J. Cancer 2021, 125, 1321–1332. [Google Scholar] [CrossRef]
- LaRose, M.; Manji, G.A.; Bates, S.E. Beyond BRCA: Diagnosis and management of homologous recombination repair deficient pancreatic cancer. Semin. Oncol. 2023, 51, 36–44. [Google Scholar] [CrossRef] [PubMed]
- Tadehara, M.; Kato, T.; Adachi, K.; Tamaki, A.; Kesen, Y.; Sakurai, Y.; Ichinoe, M.; Koizumi, W.; Murakumo, Y. Clinicopathological Significance of BRCAness in Resectable Pancreatic Ductal Adenocarcinoma and Its Association With Anticancer Drug Sensitivity in Pancreatic Cancer Cells. Pancreas 2022, 51, 183–189. [Google Scholar] [CrossRef] [PubMed]
- Zhen, D.B.; Rabe, K.G.; Gallinger, S.; Syngal, S.; Schwartz, A.G.; Goggins, M.G.; Hruban, R.H.; Cote, M.L.; McWilliams, R.R.; Roberts, N.J.; et al. BRCA1, BRCA2, PALB2, and CDKN2A mutations in familial pancreatic cancer: A PACGENE study. Genet. Med. 2015, 17, 569–577. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Roberts, N.J.; Norris, A.L.; Petersen, G.M.; Bondy, M.L.; Brand, R.; Gallinger, S.; Kurtz, R.C.; Olson, S.H.; Rustgi, A.K.; Schwartz, A.G.; et al. Whole Genome Sequencing Defines the Genetic Heterogeneity of Familial Pancreatic Cancer. Cancer Discov. 2016, 6, 166–175. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Takai, E.; Yachida, S.; Shimizu, K.; Furuse, J.; Kubo, E.; Ohmoto, A.; Suzuki, M.; Hruban, R.H.; Okusaka, T.; Morizane, C.; et al. Germline mutations in Japanese familial pancreatic cancer patients. Oncotarget 2016, 7, 74227–74235. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yurgelun, M.B.; Chittenden, A.B.; Morales-Oyarvide, V.; Rubinson, D.A.; Dunne, R.F.; Kozak, M.M.; Qian, Z.R.; Welch, M.W.; Brais, L.K.; Da Silva, A.; et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genet. Med. 2019, 21, 213–223. [Google Scholar] [CrossRef]
- 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] [PubMed Central]
- Brand, R.; Borazanci, E.; Speare, V.; Dudley, B.; Karloski, E.; Peters, M.L.B.; Stobie, L.; Bahary, N.; Zeh, H.; Zureikat, A.; et al. Prospective study of germline genetic testing in incident cases of pancreatic adenocarcinoma. Cancer 2018, 124, 3520–3527. [Google Scholar] [CrossRef] [PubMed]
- Stefanoudakis, D.; Frountzas, M.; Schizas, D.; Michalopoulos, N.V.; Drakaki, A.; Toutouzas, K.G. Significance of TP53, CDKN2A, SMAD4 and KRAS in Pancreatic Cancer. Curr. Issues Mol. Biol. 2024, 46, 2827–2844. [Google Scholar] [CrossRef]
- Rowley, M.; Ohashi, A.; Mondal, G.; Mills, L.; Yang, L.; Zhang, L.; Sundsbak, R.; Shapiro, V.; Muders, M.H.; Smyrk, T.; et al. Inactivation of Brca2 promotes Trp53-associated but inhibits KrasG12D-dependent pancreatic cancer development in mice. Gastroenterology 2011, 140, 1303–1313. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Drosos, Y.; Escobar, D.; Chiang, M.Y.; Roys, K.; Valentine, V.; Valentine, M.B.; Rehg, J.E.; Sahai, V.; Begley, L.A.; Ye, J.; et al. ATM-deficiency increases genomic instability and metastatic potential in a mouse model of pancreatic cancer. Sci. Rep. 2017, 7, 11144. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Perkhofer, L.; Schmitt, A.; Romero Carrasco, M.C.; Ihle, M.; Hampp, S.; Ruess, D.A.; Hessmann, E.; Russell, R.; Lechel, A.; Azoitei, N.; et al. ATM Deficiency Generating Genomic Instability Sensitizes Pancreatic Ductal Adenocarcinoma Cells to Therapy-Induced DNA Damage. Cancer Res. 2017, 77, 5576–5590. [Google Scholar] [CrossRef] [PubMed]
- Javle, M.; Shacham-Shmueli, E.; Xiao, L.; Varadhachary, G.; Halpern, N.; Fogelman, D.; Boursi, B.; Uruba, S.; Margalit, O.; Wolff, R.A.; et al. Olaparib Monotherapy for Previously Treated Pancreatic Cancer with DNA Damage Repair Genetic Alterations Other Than Germline BRCA Variants: Findings from 2 Phase 2 Nonrandomized Clinical Trials. JAMA Oncol. 2021, 7, 693–699. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sehdev, A.; Gbolahan, O.; Hancock, B.A.; Stanley, M.; Shahda, S.; Wan, J.; Wu, H.H.; Radovich, M.; O’Neil, B.H. Germline and Somatic DNA Damage Repair Gene Mutations and Overall Survival in Metastatic Pancreatic Adenocarcinoma Patients Treated with FOLFIRINOX. Clin. Cancer Res. 2018, 24, 6204–6211. [Google Scholar] [CrossRef] [PubMed]
- Pishvaian, M.J.; Wang, H.; He, A.R.; Hwang, J.J.; Smaglo, B.G.; Kim, S.S.; Weinberg, B.A.; Weiner, L.M.; Marshall, J.L.; Brody, J.R. A Phase I/II Study of Veliparib (ABT-888) in Combination with 5-Fluorouracil and Oxaliplatin in Patients with Metastatic Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 5092–5101. [Google Scholar] [CrossRef] [PubMed]
- O’Reilly, E.M.; Lee, J.W.; Lowery, M.A.; Capanu, M.; Stadler, Z.K.; Moore, M.J.; Dhani, N.; Kindler, H.L.; Estrella, H.; Maynard, H.; et al. Phase 1 trial evaluating cisplatin, gemcitabine, and veliparib in 2 patient cohorts: Germline BRCA mutation carriers and wild-type BRCA pancreatic ductal adenocarcinoma. Cancer 2018, 124, 1374–1382. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Shroff, R.T.; Hendifar, A.; McWilliams, R.R.; Geva, R.; Epelbaum, R.; Rolfe, L.; Goble, S.; Lin, K.K.; Biankin, A.V.; Giordano, H.; et al. Rucaparib Monotherapy in Patients With Pancreatic Cancer and a Known Deleterious BRCA Mutation. JCO Precis. Oncol. 2018, 2, 1–15. [Google Scholar] [CrossRef]
- Lowery, M.A.; Kelsen, D.P.; Capanu, M.; Smith, S.C.; Lee, J.W.; Stadler, Z.K.; Moore, M.J.; Kindler, H.L.; Golan, T.; Segal, A.; et al. Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. Eur. J. Cancer 2018, 89, 19–26. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Binder, K.A.R.; Mick, R.; O’Hara, M.; Teitelbaum, U.; Karasic, T.; Schneider, C.; O’Dwyer, P.J.; Carpenter, E.; Pantel, A.; Makvandi, M.; et al. Abstract CT234: A Phase II, single arm study of maintenance rucaparib in patients with platinum-sensitive advanced pancreatic cancer and a pathogenic germline or somatic mutation in BRCA1, BRCA2 or PALB2. Cancer Res. 2019, 79 (Suppl. S13), CT234. [Google Scholar] [CrossRef]
- Reiss, K.A.; Mick, R.; Teitelbaum, U.; O’Hara, M.; Schneider, C.; Massa, R.; Karasic, T.; Tondon, R.; Onyiah, C.; Gosselin, M.K.; et al. Niraparib plus nivolumab or niraparib plus ipilimumab in patients with platinum-sensitive advanced pancreatic cancer: A randomised, phase 1b/2 trial. Lancet Oncol. 2022, 23, 1009–1020, Correction in Lancet Oncol. 2022, 23, e446. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cleaver, J. Defective Repair Replication of DNA in Xeroderma Pigmentosum. Nature 1968, 218, 652–656. [Google Scholar] [CrossRef]
- Setlow, R.B.; Carrier, W.L. The disappearance of thymine dimers from DNA: An error-correcting mechanism. Proc. Natl. Acad. Sci. USA 1964, 51, 226–231. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ruze, R.; Song, J.; Yin, X.; Chen, Y.; Xu, R.; Wang, C.; Zhao, Y. Mechanisms of obesity- and diabetes mellitus-related pancreatic carcinogenesis: A comprehensive and systematic review. Signal Transduct. Target. Ther. 2023, 8, 139. [Google Scholar] [CrossRef] [PubMed]
- Pishvaian, M.J.; Blais, E.M.; Brody, J.R.; Lyons, E.; DeArbeloa, P.; Hendifar, A.; Mikhail, S.; Chung, V.; Sahai, V.; Sohal, D.P.S.; et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: A retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol. 2020, 21, 508–518, Correction in Lancet Oncol. 2020, 21, e182. [Google Scholar] [CrossRef] [PubMed]
- Bai, P. Biology of Poly (ADP-Ribose) Polymerases: The Factotums of Cell Maintenance. Mol. Cell 2015, 58, 947–958. [Google Scholar] [CrossRef] [PubMed]
- Kindler, H.L.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.O.; Hochhauser, D.; Arnold, D.; Oh, D.Y.; et al. Overall Survival Results from the POLO Trial: A Phase III Study of Active Maintenance Olaparib Versus Placebo for Germline BRCA-Mutated Metastatic Pancreatic Cancer. J. Clin. Oncol. 2022, 40, 3929–3939, Erratum in J. Clin. Oncol. 2024, 42, 2112. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- de Bono, J.; Ramanathan, R.K.; Mina, L.; Chugh, R.; Glaspy, J.; Rafii, S.; Kaye, S.; Sachdev, J.; Heymach, J.; Smith, D.C.; et al. Phase I, Dose-Escalation, Two-Part Trial of the PARP Inhibitor Talazoparib in Patients with Advanced Germline BRCA1/2 Mutations and Selected Sporadic Cancers. Cancer Discov. 2017, 7, 620–629. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Li, B.; Zhang, Q.; Castaneda, C.; Cook, S. Targeted Therapies in Pancreatic Cancer: A New Era of Precision Medicine. Biomedicines 2024, 12, 2175. [Google Scholar] [CrossRef]
- Weiss, G.J.; Blaydorn, L.; Beck, J.; Bornemann-Kolatzki, K.; Urnovitz, H.; Schütz, E.; Khemka, V. Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Investig. New Drugs 2017, 36, 96–102, Correction in Investig. New Drugs 2019, 37, 797. [Google Scholar] [CrossRef]
- Lecona, E.; Fernandez-Capetillo, O. Targeting ATR in cancer. Nat. Rev. Cancer 2018, 18, 586–595. [Google Scholar] [CrossRef]
- Cimprich, K.A.; Cortez, D. ATR: An essential regulator of genome integrity. Nat. Rev. Mol. Cell Biol. 2008, 9, 616–627. [Google Scholar] [CrossRef]
- Stoof, J.; Harrold, E.; Mariottino, S.; Lowery, M.A.; Walsh, N. DNA Damage Repair Deficiency in Pancreatic Ductal Adenocarcinoma: Preclinical Models and Clinical Perspectives. Front. Cell Dev. Biol. 2021, 9, 749490. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Gorecki, L.; Andrs, M.; Rezacova, M.; Korabecny, J. Discovery of ATR kinase inhibitor berzosertib (VX-970, M6620): Clinical candidate for cancer therapy. Pharmacol. Ther. 2020, 210, 107518. [Google Scholar] [CrossRef] [PubMed]
- Yap, T.A.; Silverman, I.M.; Fontana, E.; Lee, E.; Spigel, D.; Højgaard, M.; Lheureux, S.; Mettu, N.; Carneiro, B.A.; Carter, L.; et al. Abstract CT030: Genomic and pathologic determinants of response to RP-3500, an ataxia telangiectasia and Rad3-related inhibitor (ATRi), in patients (pts) with DNA damage repair (DDR) loss-of-function (LOF) mutant tumors in the Phase 1/2 TRESR trial. Cancer Res. 2022, 82 (Suppl. S12), CT030. [Google Scholar] [CrossRef]
- Foote, K.M.; Nissink, J.W.M.; McGuire, T.; Turner, P.; Guichard, S.; Yates, J.W.T.; Lau, A.; Blades, K.; Heathcote, D.; Odedra, R.; et al. Discovery and Characterization of AZD6738, a Potent Inhibitor of Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Kinase with Application as an Anticancer Agent. J. Med. Chem. 2018, 61, 9889–9907. [Google Scholar] [CrossRef] [PubMed]
- Wengner, A.M.; Siemeister, G.; Lücking, U.; Lefranc, J.; Wortmann, L.; Lienau, P.; Bader, L.P.B.; Bömer, U.; Moosmayer, D.; Eberspächer, U.; et al. The Novel ATR Inhibitor BAY 1895344 Is Efficacious as Monotherapy and Combined with DNA Damage-Inducing or Repair-Compromising Therapies in Preclinical Cancer Models. Mol. Cancer Ther. 2020, 19, 26–38. [Google Scholar] [CrossRef] [PubMed]
- Andrei, A.Z.; Hall, A.; Smith, A.L.; Bascuñana, C.; Malina, A.; Connor, A.; Altinel-Omeroglu, G.; Huang, S.; Pelletier, J.; Huntsman, D.; et al. Increased in vitro and in vivo sensitivity of BRCA2-associated pancreatic cancer to the poly(ADP-ribose) polymerase-1/2 inhibitor BMN 673. Cancer Lett. 2015, 364, 8–16. [Google Scholar] [CrossRef] [PubMed]
Population | Number | ASR 1 World |
---|---|---|
Africa | 18,993 | 2.4 |
Asia | 232,537 | 3.6 |
Europe | 146,477 | 8.0 |
Latin America and the Caribbean | 41,032 | 4.6 |
Northern America | 67,089 | 8.5 |
Oceania | 4864 | 6.2 |
Total | 510,992 | 4.7 |
Type of DNA Damage | Alkylation of Bases | Purine and Pyrimidine Dimers | DNA Adducts | ICL 1 and DPC 2 | Oxidative DNA Damage | DNA Strand Breaks |
---|---|---|---|---|---|---|
Source | Endogenous processes (byproducts of oxidative damage, cellular methylation donors) Environmental compounds (tobacco smoke) | UVR 3, IR 4 (sunlight, X-rays) | Binding with a chemical substance (tobacco smoke, alcohol metabolism, processed food) | Byproduct of cell metabolism Exogenous factors (IR, UV, chemotherapeutics) | Oxidative stress (exposure to increased or decreased oxygen pressure, the action of chemical compounds, radiation, inflammation) | Endogenous DNA metabolic processes (activity of DNA topoisomerase, impaired DNA replication, transcription, and recombination) IR Anticancer drugs (cisplatin) |
Trial | Design | Patients | Treatment | References |
---|---|---|---|---|
NCT02950064 | Phase I, open label | Patients with BRCA1/BRCA2 or other DNA repair mutations with advanced solid tumors (one of the following cancers: pancreatic cancer, castration-resistant prostate cancer, ovarian cancer, triple-negative breast cancer) | BTP-114, novel platinum compound | - |
NCT01489865 | Phase I/II study, single arm | Patients with metastatic PDAC with BRCA/PALB2/FANC mutations or family history | ABT-888 (PARP inhibitor) combined with mFOLFOX6 (oxaliplatin, 5-FU/leucovorin) | [188] |
NCT02184195 | Phase III, randomized | Metastatic PDAC with germline BRCA1/BRCA2, no progression on first-line platinum-based treatment | Olaparib or placebo treatment | [161] |
NCT01585805 | Phase II, randomized | Locally advanced or metastatic PDAC with BRCA1 or PALB2 mutations | Veliparib, gemcitabine, cisplatin (Arm A); gemcitabine, cisplatin (Arm B); placebo (Arm C) | [189] |
NCT02042378 | Phase II, single arm | PDAC with BRCA mutation | Rucaparib (PARP inhibitor) treatment | [190] |
NCT02184195 | Phase II, randomized | PDAC patients with documented mutations in BRCA1/BRCA2 | Olaparib vs. placebo treatment | [191] |
NCT03140670 | Phase II, open-label | Platinum-sensitive advanced PDAC with BRCA1/BRCA2 or PALB2 mutations | Rucaparib (PARP inhibitor) treatment | [192] |
NCT03682289 | Phase II | Advanced solid tumors (including PDAC) with progression | Ceralasertib (ATR Kinase Inhibitor) alone (Arm A), ceralasertib; olaparib, (Arm B), ceralasertib, Durvalumab (Arm C) | - |
NCT03669601 | Phase I, non-randomized (dose escalation) | Inoperable/unresectable locally advanced or metastatic PDAC and other solid tumors | AZD6738 (ATR inhibitor) | - |
NCT03404960 | Phase Ib/II open label | PDAC patients who received prior platinum-based treatment | Niraparib + Nivolumab (Arm A); Niraparib + Iplimumab (Arm B) | [193] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kozłowska, M.; Mik, M.; Nowicki, M.; Śliwińska, A. DNA Damage and Repair in Pancreatic Cancer—The Latest Findings. Int. J. Mol. Sci. 2025, 26, 10106. https://doi.org/10.3390/ijms262010106
Kozłowska M, Mik M, Nowicki M, Śliwińska A. DNA Damage and Repair in Pancreatic Cancer—The Latest Findings. International Journal of Molecular Sciences. 2025; 26(20):10106. https://doi.org/10.3390/ijms262010106
Chicago/Turabian StyleKozłowska, Małgorzata, Michał Mik, Michał Nowicki, and Agnieszka Śliwińska. 2025. "DNA Damage and Repair in Pancreatic Cancer—The Latest Findings" International Journal of Molecular Sciences 26, no. 20: 10106. https://doi.org/10.3390/ijms262010106
APA StyleKozłowska, M., Mik, M., Nowicki, M., & Śliwińska, A. (2025). DNA Damage and Repair in Pancreatic Cancer—The Latest Findings. International Journal of Molecular Sciences, 26(20), 10106. https://doi.org/10.3390/ijms262010106