Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1735 2022-08-09 19:54:12 |
2 format correct Meta information modification 1735 2022-08-12 08:36:29 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Nickoloff, J.A. Targeting Replication Stress Response Pathways to Treat Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/26050 (accessed on 27 July 2024).
Nickoloff JA. Targeting Replication Stress Response Pathways to Treat Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/26050. Accessed July 27, 2024.
Nickoloff, Jac A.. "Targeting Replication Stress Response Pathways to Treat Cancer" Encyclopedia, https://encyclopedia.pub/entry/26050 (accessed July 27, 2024).
Nickoloff, J.A. (2022, August 10). Targeting Replication Stress Response Pathways to Treat Cancer. In Encyclopedia. https://encyclopedia.pub/entry/26050
Nickoloff, Jac A.. "Targeting Replication Stress Response Pathways to Treat Cancer." Encyclopedia. Web. 10 August, 2022.
Targeting Replication Stress Response Pathways to Treat Cancer
Edit

Proliferating cells regularly experience replication stress caused by spontaneous DNA damage that results from endogenous reactive oxygen species, DNA sequences that can assume secondary and tertiary structures, collisions between opposing transcription and replication machineries, and exogenous genotoxic agents. Replication stress often leads to DNA double-strand breaks (DSBs) that can cause genome instability and cell death. The importance of replication stress responses in cells exposed to genotoxic chemo- or radiotherapy has prompted considerable research focused on how tumor cells might be selectively killed by combined treatments with genotoxins and agents targeting DNA damage response (DDR) factors involved in the replication stress response.

DNA replication stress DNA damage response oncogenic stress

1. Targeting Downstream DNA Damage Checkpoint Factors (Chk1, Chk2, Wee1)

Many DDR targets along the DNA damage sensing-signaling-repair continuum have been explored as potential cancer therapeutic targets [1]. In the 1990s and early 2000s, there was considerable excitement about combining genotoxic chemo- or radiotherapy with inhibitors of checkpoint effector kinases. The general idea was that by inhibiting effector checkpoint kinases Chk1, Chk2, or Wee1, cells experiencing genotoxic stress would fail to arrest, and continued cell cycle progression in the face of significant DNA damage would lead to cell death by mitotic catastrophe or other mechanisms. This prompted the early development of Chk1 inhibitors (Chk1i) including UCN-01. Despite success in preclinical studies, in a Phase I trial, UCN-01 had a long half-life but limited bioavailability due to high avidity to plasma α1-acid glycoprotein, and serious side effects were observed when doses were increased to exceed the plasma binding capacity [2]. By 2013, at least 12 additional Chk1i were developed, but like UCN-01, many were cross-inhibitory with other targets (e.g., Chk2, CDK1, VEGFR2, PIM1) and only one, LY2603618, combined with the antifolate antineoplastic drug Pemetredex, reached a Phase I/II trial [3]. The severe side effects of UCN-01 may reflect the rather broad impact that Chk1 has on cellular functions, including replication initiation, replication fork stabilization, cell cycle progression, DNA repair, and apoptosis. Alternatively, the disappointing results with early Chk1i may reflect their lack of specificity.

2. Targeting the Upstream DNA Damage Checkpoint Kinase ATR and Its Activation Partner TopBP1

Given the propensity of serious side effects and limited clinical benefits to date for Chk1i, it may seem counterintuitive that targeting ATR, which acts upstream of Chk1, would have fewer adverse effects on normal tissues. Nevertheless, potent and relatively specific ATRi have been developed and several have been tested in mono- or combination therapy in Phase I/II trials. The ATRi BAY1895344 has shown promising results in a Phase I trial against advanced solid tumors, with ~20% of patients showing partial responses and nearly 40% showing stable disease. Although adverse effects with BAY1895344 were common, including hematologic problems, fatigue, and nausea, these were manageable [4]. The ATRi AZD6738 (Ceralasertib) was well tolerated in combination with carboplatin in a Phase I trial that also showed moderate clinical benefit [5]. Interestingly, in both of these ATRi trials, patients who responded had DDR defects, including loss of ATM. These results should motivate further studies with ATRi and other DNA damage checkpoint inhibitors in patients with DDR defects to further develop personalized therapies.
TopBP1 functions in a variety of cellular processes and has many binding partners [6], including itself, topoisomerase IIβ, p53, and RAD51 [7], and it plays a critical role in ATR signaling to Chk1 [8]. TopBP1 expression is frequently increased in osteosarcoma and other sarcomas, and this correlates with poor prognosis [9]. Interestingly, low or moderate levels of TopBP1 correlate with normal ATR/Chk1 activation during stress, but TopBP1 overexpression has an inhibitory effect on ATR/Chk1 activation [10], which, in theory, would promote the resistance of tumor cells to DNA damage by suppressing apoptosis. Because TopBP1 functions depend on protein–protein interactions, specific inhibitors that block these interactions are of interest. The cell viability dye calcein acetoxymethyl ester (calcein AM) was shown to interfere with TopBP1 oligomerization and p53 binding, resulting in the reactivation of apoptosis and interference with mutant/oncogenic p53 [11]. Calcein AM showed antitumor activity against breast cancer xenografts [11] and it increased the antitumor activity of the topoisomerase inhibitor doxorubicin against lung tumor xenografts [12]. To date no TopBP1 inhibitors have advanced to clinical trials.

3. Targeting Replication Stress Nucleases (CtIP, MUS81, EEPD1, Metnase)

Several nucleases play key roles in the replication stress response [13]. CtIP is a nuclease and key factor in the early steps of DNA end processing at DSBs, functioning with BRCA1 in both end resection to create single-stranded DNA (ssDNA) required for homologous recombination (HR) repair, and in the protection of stressed replication forks [14][15][16]. It thus plays an important role in creating the ssDNA-RPA substrate required for ATR activation [17]. A recent study demonstrated that a peptide mimic that blocks CtIP tetramerization is a specific CtIPi that impairs DSB repair, interferes with stressed fork protection, sensitizes cells to DNA damage and PARP1i, and is cytotoxic to BRCA1-defective cells [18]. CtIP is also a target of a lncRNA (lnc15.2)-encoded micropeptide termed PACMP. PACMP stabilizes CtIP by blocking its ubiquitination, and it also promotes poly(ADP)ribosylation of substrates by PARP1 [19]. siRNA knockdown of lnc15.2 causes hypersensitivity to PARP1i, ATRi, CDK4/6i, the TopoI inhibitor camptothecin, the TopoII inhibitor epirubicin, and radiation, and it has antitumor activity against breast tumor xenografts [19]. Together, these results warrant further studies to test the antitumor effects of CtIPi in preclinical and clinical trials.
Several nucleases that process stressed replication forks are being explored as therapeutic targets. MUS81 cleaves stalled replication forks to initiate HR-mediated fork restart [20], and it has shown promise as a target in preclinical studies [21][22]. EEPD1 also cleaves stalled replication forks [23][24] and given that MUS81 and EEPD1 cleave forks with different polarity [13], combining MUS81i and EEPD1i may be a potent approach to augment the cytotoxic effects of replication stress. Metnase is a nuclease and protein methylase that promotes replication fork restart, but it does not cleave stalled forks [23]. Metnase nuclease is inhibited by the frequently administered antibiotic Ciprofloxacin [25], suggesting a safe and potentially effective means to augment genotoxic cancer therapy, perhaps in combination with drugs targeting other DDR factors.

4. Targeting DSB Repair Proteins (RAD51, PARP1)

RAD51 plays a central role in HR repair of DSBs, and HR plays a critical role in fork protection and the accurate repair and restart of collapsed replication forks. RAD51 is frequently overexpressed in cancer cells [26], thus RAD51 has emerged as a potentially useful target in cancer therapy. RI-1 is a RAD51i that inhibits RAD51-RAD51 interactions that are important for RAD51-ssDNA nucleoprotein filament formation, the key complex that catalyzes strand exchange during HR [27]. RI-1 has been shown by two groups to radiosensitize glioblastoma cells [28][29]   and it enhances the chemosensitivity of glioblastoma cells to genotoxic damage by alkylating agents [30]. The RAD51i B02 interferes with RAD51-mediated strand invasion [31].  B02 sensitizes glioblastoma cells to alkylating agents [30], and it sensitizes multiple myeloma cells to doxorubicin [32]. Tumors with HR defects, such as BRCA1- and BRCA2-defective breast cancer are sensitive to PARP1i because PARP1 promotes the repair of single-strand lesions that block replication forks and therefore require HR to repair/restart these stressed forks [33][34]. The original B02 compound has been modified into a B02-isomer with greater potency, and the HR defect induced by B02-isomer shows strong synergistic sensitization of tumor cells with PARP1i [35].
PARP1 adds poly(ADP)ribose (PAR) groups to numerous target proteins. PARylation of repair factors is seen in response to DNA damage processed by base excision repair, nucleotide excision repair, single-strand break repair, and DSB repair by canonical non-homologous end-joining (NHEJ), alternative NHEJ, and HR [36]. PARP1i induces replication stress by at least two mechanisms: PARP1i increases the load of unrepaired endogenous DNA lesions (analogous to increased lesion load by exogenous genotoxins), and it blocks replication directly by trapping PARP1 on DNA [37]. In addition, recent studies revealed new roles for PARP1 in chromatin remodeling. DNA repair occurs within the chromatin environment, so it is no surprise that proteins that modify or remodel chromatin influence DNA repair [38][39][40][41][42][43][44][45] and replication stress responses [46][47]. PARP1 has emerged has an important factor in chromatin modeling, and PARylation of both repair factors and chromatin promote the recruitment of repair factors to damaged sites [36][48].
Normal cells cope with PARP1i-induced replication stress by marshaling HR to repair and restart collapsed replication forks, but in cancer cells with HR defects, PARP1i are synthetically lethal [49][50]. First exploited in cancer cells with mutant BRCA1 or BRCA2, PARP1i are potentially valuable against tumors with defects in other HR proteins due to inherited germline mutations or sporadic somatic mutations. The PARP1i olaparib was the first targeted therapeutic approved to treat BRCA-defective ovarian cancer, and PARP1i are being explored to treat BRCA-mutant breast, ovarian, prostate, and pancreatic cancers [51][52][53][54]. In 2020 the FDA approved two PARP1i, olaparib and rucaparib to treat metastatic castration-resistant prostate cancer, and it approved olaparib to treat eligible patients with pancreatic cancer. Because PARP1 plays many roles in DNA repair, replication stress responses, and chromatin remodeling, additional uses for PARP1i in cancer monotherapy, combination therapy, and maintenance therapy continue to be explored [55][56][57][58][59].

5. Exploiting Synthetic Lethality of TKIs Targeting Activated Oncogenes and ATM Inhibitors

When cancer cells activate oncogenes that promote growth, an important consequence is oncogenic stress, also known as oncogenic replicative stress [60]. Oncogenic stress reflects dysregulated replication initiation and progression, including mis-timed origin firing. Many drugs targeting activated oncogenes have been developed and brought to clinical practice, including tyrosine kinase inhibitors (TKIs) that target activated HER2/ERBB2, ALK, KRAS, BRAF, and EGFR [61][62][63][64][65][66]. It was recently shown that blocking oncogenic pathways with targeted TKIs causes moderate stress which results in sublethal DSBs induced by caspase-activated DNase (CAD; also known as DFF40 and DFFB) [67]. When fully activated during apoptosis, CAD is responsible for digesting the genome into ~180 bp “ladders” by cleaving linker DNA between nucleosomes [68]. Although cells survive these TKI/CAD-induced DSBs, their repair depends on ATM, thus ATMi kills cells treated with various TKIs targeting activated oncogenes [67]. This effect appears to be general, as ATMi kills tumor cells treated with cognate TKIs targeting different activated oncogenes (EGFR, ALK, KRAS, and BRAF) in different tumor types (lung, pancreatic, melanoma, and acute myeloid leukemia). Interestingly, TKI + ATMi is cytotoxic even in cells that have gained resistance to the cognate TKI, suggesting ATMi may prove beneficial to patients with TKI-resistant tumors [67]. This TKI + ATMi effect represents a novel approach to exploit replication stress (oncogenic stress) and DDR inhibition to selectively target cancer cells.

References

  1. Patrick G. Pilié; Chad Tang; Gordon B. Mills; Timothy A. Yap; State-of-the-art strategies for targeting the DNA damage response in cancer. Nature Reviews Clinical Oncology 2018, 16, 81-104, 10.1038/s41571-018-0114-z.
  2. E Fuse; H Tanii; N Kurata; H Kobayashi; Y Shimada; T Tamura; Y Sasaki; Y Tanigawara; R D Lush; D Headlee; et al.W D FiggS G ArbuckA M SenderowiczE A SausvilleS AkinagaT KuwabaraS Kobayashi Unpredicted clinical pharmacology of UCN-01 caused by specific binding to human alpha1-acid glycoprotein. Cancer Research 1998, 58, 3248-53.
  3. Ruth Thompson; Alan Eastman; The cancer therapeutic potential of Chk1 inhibitors: how mechanistic studies impact on clinical trial design. British Journal of Clinical Pharmacology 2013, 76, 358-369, 10.1111/bcp.12139.
  4. Timothy A. Yap; David S.P. Tan; Angelika Terbuch; Reece Caldwell; Christina Guo; Boon Cher Goh; Valerie Heong; Noor R. Md. Haris; Saira Bashir; Yvette Drew; et al.David S. HongFunda Meric-BernstamGary WilkinsonJoseph HreikiAntje M. WengnerFriedhelm BladtAndreas SchlickerMatthias LudwigYinghui ZhouLi LiuSonal BordiaRuth PlummerEleni LagkadinouJohann S. de Bono First-in-Human Trial of the Oral Ataxia Telangiectasia and RAD3-Related (ATR) Inhibitor BAY 1895344 in Patients with Advanced Solid Tumors. Cancer Discovery 2020, 11, 80-91, 10.1158/2159-8290.cd-20-0868.
  5. Timothy A. Yap; Matthew G. Krebs; Sophie Postel-Vinay; Anthony El-Khouiery; Jean-Charles Soria; Juanita Lopez; Alienor Berges; S.Y. Amy Cheung; Itziar Irurzun-Arana; Andrew Goldwin; et al.Brunella FelicettiGemma N. JonesAlan LauPaul FrewerAndrew J. PierceGlen ClackChristine StephensSimon A. SmithEmma DeanSimon J. Hollingsworth Ceralasertib (AZD6738), an Oral ATR Kinase Inhibitor, in Combination with Carboplatin in Patients with Advanced Solid Tumors: A Phase I Study. Clinical Cancer Research 2021, 27, 5213-5224, 10.1158/1078-0432.ccr-21-1032.
  6. Matthew Day; Antony W. Oliver; Laurence H. Pearl; Phosphorylation-dependent assembly of DNA damage response systems and the central roles of TOPBP1. DNA Repair 2021, 108, 103232, 10.1016/j.dnarep.2021.103232.
  7. Pavel Moudry; Kenji Watanabe; Kamila M. Wolanin; Jirina Bartkova; Isabel E. Wassing; Sugiko Watanabe; Robert Strauss; Rune Troelsgaard Pedersen; Vibe Oestergaard; Michael Lisby; et al.Miguel Andújar-SánchezApolinar Maya-MendozaFumiko EsashiJiri LukasJiri Bartek TOPBP1 regulates RAD51 phosphorylation and chromatin loading and determines PARP inhibitor sensitivity. Journal of Cell Biology 2016, 212, 281-288, 10.1083/jcb.201507042.
  8. Jiadong Wang; Zihua Gong; Junjie Chen; MDC1 collaborates with TopBP1 in DNA replication checkpoint control. Journal of Cell Biology 2011, 193, 267-273, 10.1083/jcb.201010026.
  9. D-P Wu; X-B Yan; L-G Liu; C Tian; K Han; H Zhang; D-L Min; TopBP1 promotes malignant progression and correlates with poor prognosis in osteosarcoma. European review for medical and pharmacological sciences 2017, 21, 4022-4031.
  10. Kang Liu; Joshua D. Graves; Fang-Tsyr Lin; Weei-Chin Lin; Overexpression of TopBP1, a canonical ATR/Chk1 activator, paradoxically hinders ATR/Chk1 activation in cancer. Journal of Biological Chemistry 2020, 296, 100382, 10.1016/j.jbc.2021.100382.
  11. Pinki Chowdhury; Gregory E. Lin; Kang Liu; Yongcheng Song; Fang-Tsyr Lin; Weei-Chin Lin; Targeting TopBP1 at a convergent point of multiple oncogenic pathways for cancer therapy. Nature Communications 2014, 5, 5476, 10.1038/ncomms6476.
  12. Yinxiang Lv; Rongrong Liu; Shangzhi Xie; Xiaoxiao Zheng; Jiayan Mao; Ying Cai; Wei Chen; Calcein-acetoxymethy ester enhances the antitumor effects of doxorubicin in nonsmall cell lung cancer by regulating the TopBP1/p53RR pathway. Anti-Cancer Drugs 2017, 28, 861-868, 10.1097/cad.0000000000000527.
  13. Jac A. Nickoloff; Neelam Sharma; Lynn Taylor; Sage J. Allen; Robert Hromas; Nucleases and Co-Factors in DNA Replication Stress Responses. DNA 2022, 2, 68-85, 10.3390/dna2010006.
  14. Andrés Cruz-García; Ana López-Saavedra; Pablo Huertas; BRCA1 Accelerates CtIP-Mediated DNA-End Resection. Cell Reports 2014, 9, 451-459, 10.1016/j.celrep.2014.08.076.
  15. Nodar Makharashvili; Tanya T. Paull; CtIP: A DNA damage response protein at the intersection of DNA metabolism. DNA Repair 2015, 32, 75-81, 10.1016/j.dnarep.2015.04.016.
  16. Sara Przetocka; Antonio Porro; Hella Bolck; Christina Walker; Aleksandra Lezaja; Anika Trenner; Christine von Aesch; Sarah-Felicitas Himmels; Alan D. D’Andrea; Raphael Ceccaldi; et al.Matthias AltmeyerAlessandro A. Sartori CtIP-Mediated Fork Protection Synergizes with BRCA1 to Suppress Genomic Instability upon DNA Replication Stress. Molecular Cell 2018, 72, 568-582.e6, 10.1016/j.molcel.2018.09.014.
  17. Jun-Hyuk Choi; Laura A. Lindsey-Boltz; Michael Kemp; Aaron C. Mason; Marc S. Wold; Aziz Sancar; Reconstitution of RPA-covered single-stranded DNA-activated ATR-Chk1 signaling. Proceedings of the National Academy of Sciences 2010, 107, 13660-13665, 10.1073/pnas.1007856107.
  18. Anika Kuster; Nour L. Mozaffari; Oliver J. Wilkinson; Jessica L. Wojtaszek; Christina Zurfluh; Sara Przetocka; Dawid Zyla; Christine von Aesch; Mark S. Dillingham; R. Scott Williams; et al.Alessandro A. Sartori A stapled peptide mimetic of the CtIP tetramerization motif interferes with double-strand break repair and replication fork protection. Science Advances 2021, 7, eabc6381, 10.1126/sciadv.abc6381.
  19. Chuanchao Zhang; Bo Zhou; Feng Gu; Hongmei Liu; Honglin Wu; Fuwen Yao; Hui Zheng; Hui Fu; Wei Chong; Shurui Cai; et al.Min HuangXiaolu MaZhifang GuoTingting LiWenyuan DengMeiwen ZhengQiao JiYongliang ZhaoYongjie MaQi-En WangTie-Shan TangCaixia Guo Micropeptide PACMP inhibition elicits synthetic lethal effects by decreasing CtIP and poly(ADP-ribosyl)ation. Molecular Cell 2022, 82, 1297-1312.e8, 10.1016/j.molcel.2022.01.020.
  20. Alessandra Pepe; Stephen C. West; MUS81-EME2 Promotes Replication Fork Restart. Cell Reports 2014, 7, 1048-1055, 10.1016/j.celrep.2014.04.007.
  21. Tao Wang; Peng Zhang; Chengguo Li; Weizhen Liu; Qian Shen; Lei Yang; Gengchen Xie; Jie Bai; Ruidong Li; Kaixiong Tao; et al.Yuping Yin MUS81 Inhibition Enhances the Anticancer Efficacy of Talazoparib by Impairing ATR/CHK1 Signaling Pathway in Gastric Cancer. Frontiers in Oncology 2022, 12, 844135, 10.3389/fonc.2022.844135.
  22. Sisi Chen; Xinwei Geng; Madiha Zahra Syeda; Zhengming Huang; Chao Zhang; Songmin Ying; Human MUS81: A Fence-Sitter in Cancer. Frontiers in Cell and Developmental Biology 2021, 9, 657305, 10.3389/fcell.2021.657305.
  23. Neelam Sharma; Michael C Speed; Christopher P Allen; David G Maranon; Elizabeth Williamson; Sudha Singh; Robert Hromas; Jac A Nickoloff; Distinct roles of structure-specific endonucleases EEPD1 and Metnase in replication stress responses. NAR Cancer 2020, 2, zcaa008, 10.1093/narcan/zcaa008.
  24. Yuehan Wu; Suk-Hee Lee; Elizabeth A. Williamson; Brian L. Reinert; Ju Hwan Cho; Fen Xia; Aruna Shanker Jaiswal; Gayathri Srinivasan; Bhavita Patel; Alexis Brantley; et al.Daohong ZhouLijian ShaoRupak PathakMartin Hauer-JensenSudha SinghKimi KongXaiohua WuHyun-Suk KimTimothy BeissbarthJochen GaedckeSandeep BurmaJac A. NickoloffRobert A. Hromas EEPD1 Rescues Stressed Replication Forks and Maintains Genome Stability by Promoting End Resection and Homologous Recombination Repair. PLOS Genetics 2015, 11, e1005675, 10.1371/journal.pgen.1005675.
  25. Elizabeth A. Williamson; Leah Damiani; Andrei Leitao; Chelin Hu; Helen Hathaway; Tudor Oprea; Larry Sklar; Montaser Shaheen; Julie Bauman; Wei Wang; et al.Jac A. NickoloffSuk-Hee LeeRobert Hromas Targeting the Transposase Domain of the DNA Repair Component Metnase to Enhance Chemotherapy. Cancer Research 2012, 72, 6200-6208, 10.1158/0008-5472.can-12-0313.
  26. Ambber Ward; Kum Kum Khanna; Adrian P. Wiegmans; Targeting homologous recombination, new pre-clinical and clinical therapeutic combinations inhibiting RAD51. Cancer Treatment Reviews 2014, 41, 35-45, 10.1016/j.ctrv.2014.10.006.
  27. Brian Budke; Hillary L. Logan; Jay Kalin; Anna S. Zelivianskaia; William Cameron McGuire; Luke L. Miller; Jeremy M. Stark; Alan P. Kozikowski; Douglas K. Bishop; Philip P. Connell; et al. RI-1: a chemical inhibitor of RAD51 that disrupts homologous recombination in human cells. Nucleic Acids Research 2012, 40, 7347-7357, 10.1093/nar/gks353.
  28. Anaïs Balbous; Ulrich Cortes; Karline Guilloteau; Pierre Rivet; Baptiste Pinel; Mathilde Duchesne; Julie Godet; Odile Boissonnade; Michel Wager; René Jean Bensadoun; et al.Jean-Claude ChomelLucie Karayan-Tapon A radiosensitizing effect of RAD51 inhibition in glioblastoma stem-like cells. BMC Cancer 2016, 16, 604, 10.1186/s12885-016-2647-9.
  29. Harry O. King; Tim Brend; Helen L. Payne; Alexander Wright; Thomas A. Ward; Karan Patel; Teklu Egnuni; Lucy F. Stead; Anjana Patel; Heiko Wurdak; et al.Susan C. Short RAD51 Is a Selective DNA Repair Target to Radiosensitize Glioma Stem Cells. Stem Cell Reports 2016, 8, 125-139, 10.1016/j.stemcr.2016.12.005.
  30. Nancy Berte; Andrea Piée-Staffa; Nadine Piecha; Mengwan Wang; Kerstin Borgmann; Bernd Kaina; Theodora Nikolova; Targeting Homologous Recombination by Pharmacological Inhibitors Enhances the Killing Response of Glioblastoma Cells Treated with Alkylating Drugs. Molecular Cancer Therapeutics 2016, 15, 2665-2678, 10.1158/1535-7163.mct-16-0176.
  31. Fei Huang; Olga M. Mazina; Isaac J. Zentner; Simon Cocklin; Alexander V. Mazin; Inhibition of Homologous Recombination in Human Cells by Targeting RAD51 Recombinase. Journal of Medicinal Chemistry 2012, 55, 3011-3020, 10.1021/jm201173g.
  32. David A. Alagpulinsa; Srinivas Ayyadevara; Robert Joseph Shmookler Reis; A Small-Molecule Inhibitor of RAD51 Reduces Homologous Recombination and Sensitizes Multiple Myeloma Cells to Doxorubicin. Frontiers in Oncology 2014, 4, 289, 10.3389/fonc.2014.00289.
  33. Laura Cortesi; Hope S. Rugo; Christian Jackisch; An Overview of PARP Inhibitors for the Treatment of Breast Cancer. Targeted Oncology 2021, 16, 255-282, 10.1007/s11523-021-00796-4.
  34. Jaydira Del Rivero; Elise C Kohn; PARP Inhibitors: The Cornerstone of DNA Repair-Targeted Therapies. Oncology 2017, 31, 265–273.
  35. Irina Shkundina; Alexander Gall; Alexej Dick; Simon Cocklin; Alexander Mazin; New RAD51 Inhibitors to Target Homologous Recombination in Human Cells. Genes 2021, 12, 920, 10.3390/genes12060920.
  36. John M. Pascal; The comings and goings of PARP-1 in response to DNA damage. DNA Repair 2018, 71, 177-182, 10.1016/j.dnarep.2018.08.022.
  37. Yuqiao Shen; Mika Aoyagi-Scharber; Bing Wang; Trapping Poly(ADP-Ribose) Polymerase. Journal of Pharmacology and Experimental Therapeutics 2015, 353, 446-457, 10.1124/jpet.114.222448.
  38. Suk-Hee Lee; Masahiko Oshige; Stephen T. Durant; Kanwaldeep Kaur Rasila; Elizabeth A. Williamson; Heather Ramsey; Lori Kwan; Jac A. Nickoloff; Robert Hromas; The SET domain protein Metnase mediates foreign DNA integration and links integration to nonhomologous end-joining repair. Proceedings of the National Academy of Sciences 2005, 102, 18075-18080, 10.1073/pnas.0503676102.
  39. Su Wu; Yujiang Shi; Peter Mulligan; Frédérique Gay; Joseph Landry; Huifei Liu; Ju Lu; Hank Qi; Weijia Wang; Jac A Nickoloff; et al.Carl WuYang Shi A YY1–INO80 complex regulates genomic stability through homologous recombination–based repair. Nature Structural & Molecular Biology 2007, 14, 1165-1172, 10.1038/nsmb1332.
  40. Anna L. Chambers; Jessica A. Downs; The RSC and INO80 Chromatin-Remodeling Complexes in DNA Double-Strand Break Repair. Progress in Molecular Biology and Translational Science 2012, 110, 229-261, 10.1016/b978-0-12-387665-2.00009-2.
  41. Yunhe Bao; Xuetong Shen; Chromatin remodeling in DNA double-strand break repair. Current Opinion in Genetics & Development 2007, 17, 126-131, 10.1016/j.gde.2007.02.010.
  42. Thomas Clouaire; Vincent Rocher; Anahita Lashgari; Coline Arnould; Marion Aguirrebengoa; Anna Biernacka; Magdalena Skrzypczak; François Aymard; Bernard Fongang; Norbert Dojer; et al.Jason S. IacovoniMaga RowickaKrzysztof GinalskiJacques CoteGaëlle Legube Comprehensive Mapping of Histone Modifications at DNA Double-Strand Breaks Deciphers Repair Pathway Chromatin Signatures. Molecular Cell 2018, 72, 250-262.e6, 10.1016/j.molcel.2018.08.020.
  43. Brendan D. Price; Alan D. D’Andrea; Chromatin Remodeling at DNA Double-Strand Breaks. Cell 2013, 152, 1344-1354, 10.1016/j.cell.2013.02.011.
  44. Ashby J. Morrison; Genome maintenance functions of the INO80 chromatin remodeller. Philosophical Transactions of the Royal Society B: Biological Sciences 2017, 372, 20160289, 10.1098/rstb.2016.0289.
  45. Louis-Alexandre Fournier; Arun Kumar; Peter C. Stirling; Chromatin as a Platform for Modulating the Replication Stress Response. Genes 2018, 9, 622, 10.3390/genes9120622.
  46. Kenji Shimada; Yukako Oma; Thomas Schleker; Kazuto Kugou; Kunihiro Ohta; Masahiko Harata; Susan M. Gasser; Ino80 Chromatin Remodeling Complex Promotes Recovery of Stalled Replication Forks. Current Biology 2008, 18, 566-575, 10.1016/j.cub.2008.03.049.
  47. Maddalena Mognato; Susanne Burdak-Rothkamm; Kai Rothkamm; Interplay between DNA replication stress, chromatin dynamics and DNA-damage response for the maintenance of genome stability. Mutation Research/Reviews in Mutation Research 2021, 787, 108346, 10.1016/j.mrrev.2020.108346.
  48. Arnab Ray Chaudhuri; André Nussenzweig; The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nature Reviews Molecular Cell Biology 2017, 18, 610-621, 10.1038/nrm.2017.53.
  49. Helen E. Bryant; Niklas Schultz; Huw D. Thomas; Kayan M. Parker; Dan Flower; Elena Lopez; Suzanne Kyle; Mark Meuth; Nicola Curtin; Thomas Helleday; et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005, 434, 913-917, 10.1038/nature03443.
  50. Hannah Farmer; Nuala McCabe; Christopher Lord; Andrew N. J. Tutt; Damian A. Johnson; Tobias B. Richardson; Manuela Santarosa; Krystyna J. Dillon; Ian Hickson; Charlotte Knights; et al.Niall M. B. MartinStephen Philip JacksonGraeme C. M. SmithAlan Ashworth Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005, 434, 917-921, 10.1038/nature03445.
  51. Christopher Lord; Alan Ashworth; BRCAness revisited. Nature Reviews Cancer 2016, 16, 110-120, 10.1038/nrc.2015.21.
  52. Kabir Grewal; Kayanaat Grewal; Imad A. Tabbara; PARP Inhibitors in Prostate Cancer. Anticancer Research 2021, 41, 551-556, 10.21873/anticanres.14807.
  53. Talia Golan; Pascal Hammel; Michele Reni; Eric Van Cutsem; Teresa Macarulla; Michael J. Hall; Joon-Oh Park; Daniel Hochhauser; Dirk Arnold; Do-Youn Oh; et al.Anke Reinacher-SchickGiampaolo TortoraHana AlgülEileen M. O’ReillyDavid McGuinnessKaren Y. CuiKatia SchliengerGershon Y. LockerHedy L. Kindler Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. New England Journal of Medicine 2019, 381, 317-327, 10.1056/nejmoa1903387.
  54. Antonio González-Martín; Ursula A Matulonis; Jacob Korach; Mansoor R Mirza; Kathleen N Moore; Xiaohua Wu; Whitney York; Divya Gupta; Stanislav Lechpammer; Bradley J Monk; et al. Niraparib treatment for patients with BRCA-mutated ovarian cancer: review of clinical data and therapeutic context. Future Oncology 2022, 18, 2505-2536, 10.2217/fon-2022-0206.
  55. Linjie Luo; Khandan Keyomarsi; PARP inhibitors as single agents and in combination therapy: the most promising treatment strategies in clinical trials for BRCA-mutant ovarian and triple-negative breast cancers. Expert Opinion on Investigational Drugs 2022, 31, 607-631, 10.1080/13543784.2022.2067527.
  56. Ranya Barayan; Xiaozhuo Ran; Benjamin H. Lok; PARP inhibitors for small cell lung cancer and their potential for integration into current treatment approaches. Journal of Thoracic Disease 2020, 12, 6240-6252, 10.21037/jtd.2020.03.89.
  57. Arpit Rao; Emmanuel S. Antonarakis; The growing role of rucaparib in contemporary treatment of metastatic prostate cancer: a review of efficacy and guidance for side effect management. Expert Review of Anticancer Therapy 2022, 22, 671-679, 10.1080/14737140.2022.2081154.
  58. Mingyue Xia; Zhigang Guo; Zhigang Hu; The Role of PARP Inhibitors in the Treatment of Prostate Cancer: Recent Advances in Clinical Trials. Biomolecules 2021, 11, 722, 10.3390/biom11050722.
  59. Dalia Kamel; Christopher Gray; Jagdeep Singh Walia; Vikaash Kumar; Jagdeep Walia; PARP Inhibitor Drugs in the Treatment of Breast, Ovarian, Prostate and Pancreatic Cancers: An Update of Clinical Trials. Current Drug Targets 2018, 19, 21-37, 10.2174/1389450118666170711151518.
  60. Stephanie A. Hills; John F.X. Diffley; DNA Replication and Oncogene-Induced Replicative Stress. Current Biology 2014, 24, R435-R444, 10.1016/j.cub.2014.04.012.
  61. Ji Luo; Nicole L. Solimini; Stephen J. Elledge; Principles of Cancer Therapy: Oncogene and Non-oncogene Addiction. Cell 2009, 136, 823-837, 10.1016/j.cell.2009.08.006.
  62. Arthur Aubry; Stéphane Galiacy; Michèle Allouche; Targeting ALK in Cancer: Therapeutic Potential of Proapoptotic Peptides. Cancers 2019, 11, 275, 10.3390/cancers11030275.
  63. Do-Youn Oh; Yung-Jue Bang; HER2-targeted therapies — a role beyond breast cancer. Nature Reviews Clinical Oncology 2019, 17, 33-48, 10.1038/s41571-019-0268-3.
  64. Lisa Mustachio; Anca Chelariu-Raicu; Lorant Szekvolgyi; Jason Roszik; Targeting KRAS in Cancer: Promising Therapeutic Strategies. Cancers 2021, 13, 1204, 10.3390/cancers13061204.
  65. Aubhishek Zaman; Wei Wu; Trever G. Bivona; Targeting Oncogenic BRAF: Past, Present, and Future. Cancers 2019, 11, 1197, 10.3390/cancers11081197.
  66. Elias Da Silva Santos; Karina Alexandre Barros Nogueira; Luiziana Cavalcante Costa Fernandes; Jéssica Roberta Pereira Martins; Alice Vitoria Frota Reis; José De Brito Vieira Neto; Ivanildo José Da Silva Júnior; Claudia Pessoa; Raquel Petrilli; Josimar O. Eloy; et al. EGFR targeting for cancer therapy: Pharmacology and immunoconjugates with drugs and nanoparticles. International Journal of Pharmaceutics 2021, 592, 120082, 10.1016/j.ijpharm.2020.120082.
  67. Moiez Ali; Min Lu; Hazel Xiaohui Ang; Ryan S. Soderquist; Christine E. Eyler; Haley M. Hutchinson; Carolyn Glass; Christopher F. Bassil; Omar M. Lopez; D. Lucas Kerr; et al.Christina J. FalconHelena A. YuAaron N. HataCollin M. BlakelyCaroline E. McCoachTrever G. BivonaKris C. Wood Small-molecule targeted therapies induce dependence on DNA double-strand break repair in residual tumor cells. Science Translational Medicine 2022, 14, eabc7480, 10.1126/scitranslmed.abc7480.
  68. Eui-Jeon Woo; Yeon-Gil Kim; Min-Sung Kim; Won-Deok Han; Sejeong Shin; Howard Robinson; Sam-Yong Park; Byung-Ha Oh; Structural Mechanism for Inactivation and Activation of CAD/DFF40 in the Apoptotic Pathway. Molecular Cell 2004, 14, 531-539, 10.1016/s1097-2765(04)00258-8.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 463
Revisions: 2 times (View History)
Update Date: 12 Aug 2022
1000/1000
Video Production Service