Targeting Oncogenic Pathways in the Era of Personalized Oncology: A Systemic Analysis Reveals Highly Mutated Signaling Pathways in Cancer Patients and Potential Therapeutic Targets

. 2022 Jan 28 ; 14 (3) : . [epub] 20220128

Status PubMed-not-MEDLINE Jazyk angličtina Země Švýcarsko Médium electronic

Typ dokumentu časopisecké články, přehledy

Perzistentní odkaz   https://www.medvik.cz/link/pmid35158934

Cancer is the second leading cause of death globally. One of the main hallmarks in cancer is the functional deregulation of crucial molecular pathways via driver genetic events that lead to abnormal gene expression, giving cells a selective growth advantage. Driver events are defined as mutations, fusions and copy number alterations that are causally implicated in oncogenesis. Molecular analysis on tissues that have originated from a wide range of anatomical areas has shown that mutations in different members of several pathways are implicated in different cancer types. In recent decades, significant efforts have been made to incorporate this knowledge into daily medical practice, providing substantial insight towards clinical diagnosis and personalized therapies. However, since there is still a strong need for more effective drug development, a deep understanding of the involved signaling mechanisms and the interconnections between these pathways is highly anticipated. Here, we perform a systemic analysis on cancer patients included in the Pan-Cancer Atlas project, with the aim to select the ten most highly mutated signaling pathways (p53, RTK-RAS, lipids metabolism, PI-3-Kinase/Akt, ubiquitination, b-catenin/Wnt, Notch, cell cycle, homology directed repair (HDR) and splicing) and to provide a detailed description of each pathway, along with the corresponding therapeutic applications currently being developed or applied. The ultimate scope is to review the current knowledge on highly mutated pathways and to address the attractive perspectives arising from ongoing experimental studies for the clinical implementation of personalized medicine.

Zobrazit více v PubMed

Sung H., Ferlay J., Siegel R.L., Laversanne M., Soerjomataram I., Jemal A., Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021;71:209–249. doi: 10.3322/caac.21660. PubMed DOI

Goodwin S., McPherson J.D., McCombie W.R. Coming of age: Ten years of next-generation sequencing technologies. Nat. Rev. Genet. 2016;17:333–351. doi: 10.1038/nrg.2016.49. PubMed DOI PMC

Carrasco-Ramiro F., Peiro-Pastor R., Aguado B. Human genomics projects and precision medicine. Gene Ther. 2017;24:551–561. doi: 10.1038/gt.2017.77. PubMed DOI

Imai K., Takaoka A. Comparing antibody and small-molecule therapies for cancer. Nat. Rev. Cancer. 2006;6:714–727. doi: 10.1038/nrc1913. PubMed DOI

O’Connor M.J. Targeting the DNA Damage Response in Cancer. Mol. Cell. 2015;60:547–560. doi: 10.1016/j.molcel.2015.10.040. PubMed DOI

Sharma P., Allison J.P. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell. 2015;161:205–214. doi: 10.1016/j.cell.2015.03.030. PubMed DOI PMC

Parui A.L., Bose K. Cancer Biology and Its Treatment Modalities: A Brief Historical Perspective. In: Bose K., Chaudhari P., editors. Unravelling Cancer Signaling Pathways: A Multidisciplinary Approach. Springer; Singapore: 2019. pp. 1–11. DOI

Martincorena I., Campbell P.J. Somatic mutation in cancer and normal cells. Science. 2016;349:1483–1489. doi: 10.1126/science.aab4082. Erratum in Science 2016, 351, aaf5401. PubMed DOI

Okugawa Y., Grady W.M., Goel A. Epigenetic Alterations in Colorectal Cancer: Emerging Biomarkers. Gastroenterology. 2015;149:1204–1225.e1212. doi: 10.1053/j.gastro.2015.07.011. PubMed DOI PMC

Sever R., Brugge J.S. Signal transduction in cancer. Cold Spring Harb. Perspect. Med. 2015;5:a006098. doi: 10.1101/cshperspect.a006098. PubMed DOI PMC

Baylin S.B., Jones P.A. Epigenetic Determinants of Cancer. Cold Spring Harb. Perspect. Biol. 2016;8 doi: 10.1101/cshperspect.a019505. PubMed DOI PMC

Feinberg A.P., Koldobskiy M.A., Gondor A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat. Rev. Genet. 2016;17:284–299. doi: 10.1038/nrg.2016.13. PubMed DOI PMC

Hanahan D., Weinberg R.A. Hallmarks of cancer: The next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. PubMed DOI

Sanchez-Vega F., Mina M., Armenia J., Chatila W.K., Luna A., La K.C., Dimitriadoy S., Liu D.L., Kantheti H.S., Saghafinia S., et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018;173:321–337.e310. doi: 10.1016/j.cell.2018.03.035. PubMed DOI PMC

Vogelstein B., Kinzler K.W. Cancer genes and the pathways they control. Nat. Med. 2004;10:789–799. doi: 10.1038/nm1087. PubMed DOI

Dempke W.C.M., Fenchel K., Uciechowski P., Chevassut T. Targeting Developmental Pathways: The Achilles Heel of Cancer? Oncology. 2017;93:213–223. doi: 10.1159/000478703. PubMed DOI

Pon J.R., Marra M.A. Driver and passenger mutations in cancer. Annu. Rev. Pathol. 2015;10:25–50. doi: 10.1146/annurev-pathol-012414-040312. PubMed DOI

Stratton M.R., Campbell P.J., Futreal P.A. The cancer genome. Nature. 2009;458:719–724. doi: 10.1038/nature07943. PubMed DOI PMC

Johnson A., Zeng J., Bailey A.M., Holla V., Litzenburger B., Lara-Guerra H., Mills G.B., Mendelsohn J., Shaw K.R., Meric-Bernstam F. The right drugs at the right time for the right patient: The MD Anderson precision oncology decision support platform. Drug Discov. Today. 2015;20:1433–1438. doi: 10.1016/j.drudis.2015.05.013. PubMed DOI PMC

Saltz L.B., Meropol N.J., Loehrer P.J., Sr., Needle M.N., Kopit J., Mayer R.J. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J. Clin. Oncol. 2004;22:1201–1208. doi: 10.1200/JCO.2004.10.182. PubMed DOI

Shaw A.T., Kim D.W., Nakagawa K., Seto T., Crino L., Ahn M.J., De Pas T., Besse B., Solomon B.J., Blackhall F., et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N. Engl. J. Med. 2013;368:2385–2394. doi: 10.1056/NEJMoa1214886. PubMed DOI

Zhan T., Rindtorff N., Betge J., Ebert M.P., Boutros M. CRISPR/Cas9 for cancer research and therapy. Semin. Cancer Biol. 2019;55:106–119. doi: 10.1016/j.semcancer.2018.04.001. PubMed DOI

Khalaf K., Janowicz K., Dyszkiewicz-Konwinska M., Hutchings G., Dompe C., Moncrieff L., Jankowski M., Machnik M., Oleksiewicz U., Kocherova I., et al. CRISPR/Cas9 in Cancer Immunotherapy: Animal Models and Human Clinical Trials. Genes. 2020;11:921. doi: 10.3390/genes11080921. PubMed DOI PMC

Ge Z., Leighton J.S., Wang Y., Peng X., Chen Z., Chen H., Sun Y., Yao F., Li J., Zhang H., et al. Integrated Genomic Analysis of the Ubiquitin Pathway across Cancer Types. Cell Rep. 2018;23:213–226.e213. doi: 10.1016/j.celrep.2018.03.047. PubMed DOI PMC

Knijnenburg T.A., Wang L., Zimmermann M.T., Chambwe N., Gao G.F., Cherniack A.D., Fan H., Shen H., Way G.P., Greene C.S., et al. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep. 2018;23:239–254.e236. doi: 10.1016/j.celrep.2018.03.076. PubMed DOI PMC

Peng X., Chen Z., Farshidfar F., Xu X., Lorenzi P.L., Wang Y., Cheng F., Tan L., Mojumdar K., Du D., et al. Molecular Characterization and Clinical Relevance of Metabolic Expression Subtypes in Human Cancers. Cell Rep. 2018;23:255–269.e254. doi: 10.1016/j.celrep.2018.03.077. PubMed DOI PMC

Seiler M., Peng S., Agrawal A.A., Palacino J., Teng T., Zhu P., Smith P.G., Cancer Genome Atlas Research N., Buonamici S., Yu L. Somatic Mutational Landscape of Splicing Factor Genes and Their Functional Consequences across 33 Cancer Types. Cell Rep. 2018;23:282–296.e284. doi: 10.1016/j.celrep.2018.01.088. PubMed DOI PMC

Thorsson V., Gibbs D.L., Brown S.D., Wolf D., Bortone D.S., Ou Yang T.H., Porta-Pardo E., Gao G.F., Plaisier C.L., Eddy J.A., et al. The Immune Landscape of Cancer. Immunity. 2018;48:812–830.e814. doi: 10.1016/j.immuni.2018.03.023. PubMed DOI PMC

Cerami E., Gao J., Dogrusoz U., Gross B.E., Sumer S.O., Aksoy B.A., Jacobsen A., Byrne C.J., Heuer M.L., Larsson E., et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404. doi: 10.1158/2159-8290.CD-12-0095. PubMed DOI PMC

Vogelstein B., Lane D., Levine A.J. Surfing the p53 network. Nature. 2000;408:307–310. doi: 10.1038/35042675. PubMed DOI

Oren M. Decision making by p53: Life, death and cancer. Cell Death Differ. 2003;10:431–442. doi: 10.1038/sj.cdd.4401183. PubMed DOI

Harris S.L., Levine A.J. The p53 pathway: Positive and negative feedback loops. Oncogene. 2005;24:2899–2908. doi: 10.1038/sj.onc.1208615. PubMed DOI

Jones R.G., Plas D.R., Kubek S., Buzzai M., Mu J., Xu Y., Birnbaum M.J., Thompson C.B. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol. Cell. 2005;18:283–293. doi: 10.1016/j.molcel.2005.03.027. PubMed DOI

Labuschagne C.F., Zani F., Vousden K.H. Control of metabolism by p53–Cancer and beyond. Biochim. Biophys. Acta Rev. Cancer. 2018;1870:32–42. doi: 10.1016/j.bbcan.2018.06.001. PubMed DOI PMC

Levine A.J., Oren M. The first 30 years of p53: Growing ever more complex. Nat. Rev. Cancer. 2009;9:749–758. doi: 10.1038/nrc2723. PubMed DOI PMC

Aubrey B.J., Strasser A., Kelly G.L. Tumor-Suppressor Functions of the TP53 Pathway. Cold Spring Harb. Perspect. Med. 2016;6:a026062. doi: 10.1101/cshperspect.a026062. PubMed DOI PMC

Haronikova L., Olivares-Illana V., Wang L., Karakostis K., Chen S., Fahraeus R. The p53 mRNA: An integral part of the cellular stress response. Nucleic Acids Res. 2019;47:3257–3271. doi: 10.1093/nar/gkz124. PubMed DOI PMC

Muller P.A., Vousden K.H. p53 mutations in cancer. Nat. Cell Biol. 2013;15:2–8. doi: 10.1038/ncb2641. PubMed DOI

Li J., Yang L., Gaur S., Zhang K., Wu X., Yuan Y.C., Li H., Hu S., Weng Y., Yen Y. Mutants TP53 p.R273H and p.R273C but not p.R273G enhance cancer cell malignancy. Hum. Mutat. 2014;35:575–584. doi: 10.1002/humu.22528. PubMed DOI

Sun S., Chen H., Sun L., Wang M., Wu X., Xiao Z.J. Hotspot mutant p53-R273H inhibits KLF6 expression to promote cell migration and tumor metastasis. Cell Death Dis. 2020;11:595. doi: 10.1038/s41419-020-02814-1. PubMed DOI PMC

Schulz-Heddergott R., Stark N., Edmunds S.J., Li J., Conradi L.C., Bohnenberger H., Ceteci F., Greten F.R., Dobbelstein M., Moll U.M. Therapeutic Ablation of Gain-of-Function Mutant p53 in Colorectal Cancer Inhibits Stat3-Mediated Tumor Growth and Invasion. Cancer Cell. 2018;34:298–314.e297. doi: 10.1016/j.ccell.2018.07.004. PubMed DOI PMC

Klemke L., Fehlau C.F., Winkler N., Toboll F., Singh S.K., Moll U.M., Schulz-Heddergott R. The Gain-of-Function p53 R248W Mutant Promotes Migration by STAT3 Deregulation in Human Pancreatic Cancer Cells. Front. Oncol. 2021;11:642603. doi: 10.3389/fonc.2021.642603. PubMed DOI PMC

Yu X., Vazquez A., Levine A.J., Carpizo D.R. Allele-specific p53 mutant reactivation. Cancer Cell. 2012;21:614–625. doi: 10.1016/j.ccr.2012.03.042. PubMed DOI PMC

Grugan K.D., Vega M.E., Wong G.S., Diehl J.A., Bass A.J., Wong K.K., Nakagawa H., Rustgi A.K. A common p53 mutation (R175H) activates c-Met receptor tyrosine kinase to enhance tumor cell invasion. Cancer Biol. Ther. 2013;14:853–859. doi: 10.4161/cbt.25406. PubMed DOI PMC

Parry D., Peters G. Temperature-sensitive mutants of p16CDKN2 associated with familial melanoma. Mol. Cell Biol. 1996;16:3844–3852. doi: 10.1128/MCB.16.7.3844. PubMed DOI PMC

Rutter J.L., Goldstein A.M., Davila M.R., Tucker M.A., Struewing J.P. CDKN2A point mutations D153spl(c.457G>T) and IVS2+1G>T result in aberrant splice products affecting both p16INK4a and p14ARF. Oncogene. 2003;22:4444–4448. doi: 10.1038/sj.onc.1206564. PubMed DOI

Kannengiesser C., Brookes S., del Arroyo A.G., Pham D., Bombled J., Barrois M., Mauffret O., Avril M.F., Chompret A., Lenoir G.M., et al. Functional, structural, and genetic evaluation of 20 CDKN2A germ line mutations identified in melanoma-prone families or patients. Hum. Mutat. 2009;30:564–574. doi: 10.1002/humu.20845. PubMed DOI

Angele S., Jones C., Reis Filho J.S., Fulford L.G., Treilleux I., Lakhani S.R., Hall J. Expression of ATM, p53, and the MRE11-Rad50-NBS1 complex in myoepithelial cells from benign and malignant proliferations of the breast. J. Clin. Pathol. 2004;57:1179–1184. doi: 10.1136/jcp.2004.017434. PubMed DOI PMC

Bailey S.L., Gurley K.E., Hoon-Kim K., Kelly-Spratt K.S., Kemp C.J. Tumor suppression by p53 in the absence of Atm. Mol. Cancer Res. 2008;6:1185–1192. doi: 10.1158/1541-7786.MCR-07-2009. PubMed DOI PMC

Biddlestone-Thorpe L., Sajjad M., Rosenberg E., Beckta J.M., Valerie N.C., Tokarz M., Adams B.R., Wagner A.F., Khalil A., Gilfor D., et al. ATM kinase inhibition preferentially sensitizes p53-mutant glioma to ionizing radiation. Clin. Cancer Res. 2013;19:3189–3200. doi: 10.1158/1078-0432.CCR-12-3408. PubMed DOI PMC

Malbert-Colas L., Ponnuswamy A., Olivares-Illana V., Tournillon A.S., Naski N., Fahraeus R. HDMX folds the nascent p53 mRNA following activation by the ATM kinase. Mol. Cell. 2014;54:500–511. doi: 10.1016/j.molcel.2014.02.035. PubMed DOI

Chwastek J., Jantas D., Lason W. The ATM kinase inhibitor KU-55933 provides neuroprotection against hydrogen peroxide-induced cell damage via a gammaH2AX/p-p53/caspase-3-independent mechanism: Inhibition of calpain and cathepsin D. Int. J. Biochem. Cell Biol. 2017;87:38–53. doi: 10.1016/j.biocel.2017.03.015. PubMed DOI

Karakostis K., Vadivel Gnanasundram S., Lopez I., Thermou A., Wang L., Nylander K., Olivares-Illana V., Fahraeus R. A single synonymous mutation determines the phosphorylation and stability of the nascent protein. J. Mol. Cell Biol. 2019;11:187–199. doi: 10.1093/jmcb/mjy049. PubMed DOI PMC

Hwang S.Y., Kuk M.U., Kim J.W., Lee Y.H., Lee Y.S., Choy H.E., Park S.C., Park J.T. ATM mediated-p53 signaling pathway forms a novel axis for senescence control. Mitochondrion. 2020;55:54–63. doi: 10.1016/j.mito.2020.09.002. PubMed DOI

Easton D.F., Pharoah P.D., Antoniou A.C., Tischkowitz M., Tavtigian S.V., Nathanson K.L., Devilee P., Meindl A., Couch F.J., Southey M., et al. Gene-panel sequencing and the prediction of breast-cancer risk. N. Engl. J. Med. 2015;372:2243–2257. doi: 10.1056/NEJMsr1501341. PubMed DOI PMC

Griffith O.L., Spies N.C., Anurag M., Griffith M., Luo J., Tu D., Yeo B., Kunisaki J., Miller C.A., Krysiak K., et al. The prognostic effects of somatic mutations in ER-positive breast cancer. Nat. Commun. 2018;9:3476. doi: 10.1038/s41467-018-05914-x. PubMed DOI PMC

Nahar R., Zhai W., Zhang T., Takano A., Khng A.J., Lee Y.Y., Liu X., Lim C.H., Koh T.P.T., Aung Z.W., et al. Elucidating the genomic architecture of Asian EGFR-mutant lung adenocarcinoma through multi-region exome sequencing. Nat. Commun. 2018;9:216. doi: 10.1038/s41467-017-02584-z. PubMed DOI PMC

Jette N.R., Kumar M., Radhamani S., Arthur G., Goutam S., Yip S., Kolinsky M., Williams G.J., Bose P., Lees-Miller S.P. ATM-Deficient Cancers Provide New Opportunities for Precision Oncology. Cancers. 2020;12:687. doi: 10.3390/cancers12030687. PubMed DOI PMC

Zhu G., Pan C., Bei J.X., Li B., Liang C., Xu Y., Fu X. Mutant p53 in Cancer Progression and Targeted Therapies. Front. Oncol. 2020;10:595187. doi: 10.3389/fonc.2020.595187. PubMed DOI PMC

Salomao N., Karakostis K., Hupp T., Vollrath F., Vojtesek B., Fahraeus R. What do we need to know and understand about p53 to improve its clinical value? J. Pathol. 2021;254:443–453. doi: 10.1002/path.5677. PubMed DOI

Duffy M.J., Synnott N.C., O’Grady S., Crown J. Targeting p53 for the treatment of cancer. Semin. Cancer Biol. 2020 doi: 10.1016/j.semcancer.2020.07.005. PubMed DOI

Dhillon S. Palbociclib: First global approval. Drugs. 2015;75:543–551. doi: 10.1007/s40265-015-0379-9. PubMed DOI

Kim E.S. Abemaciclib: First Global Approval. Drugs. 2017;77:2063–2070. doi: 10.1007/s40265-017-0840-z. PubMed DOI

Syed Y.Y. Ribociclib: First Global Approval. Drugs. 2017;77:799–807. doi: 10.1007/s40265-017-0742-0. PubMed DOI

Dhillon S. Trilaciclib: First Approval. Drugs. 2021;81:867–874. doi: 10.1007/s40265-021-01508-y. PubMed DOI

Young R.J., Waldeck K., Martin C., Foo J.H., Cameron D.P., Kirby L., Do H., Mitchell C., Cullinane C., Liu W., et al. Loss of CDKN2A expression is a frequent event in primary invasive melanoma and correlates with sensitivity to the CDK4/6 inhibitor PD0332991 in melanoma cell lines. Pigment Cell Melanoma Res. 2014;27:590–600. doi: 10.1111/pcmr.12228. PubMed DOI

Su D., Zhang D., Jin J., Ying L., Han M., Chen K., Li B., Wu J., Xie Z., Zhang F., et al. Identification of predictors of drug sensitivity using patient-derived models of esophageal squamous cell carcinoma. Nat. Commun. 2019;10:5076. doi: 10.1038/s41467-019-12846-7. PubMed DOI PMC

DeMichele A., Clark A.S., Tan K.S., Heitjan D.F., Gramlich K., Gallagher M., Lal P., Feldman M., Zhang P., Colameco C., et al. CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: Phase II activity, safety, and predictive biomarker assessment. Clin. Cancer Res. 2015;21:995–1001. doi: 10.1158/1078-0432.CCR-14-2258. PubMed DOI

Finn R.S., Crown J.P., Lang I., Boer K., Bondarenko I.M., Kulyk S.O., Ettl J., Patel R., Pinter T., Schmidt M., et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): A randomised phase 2 study. Lancet Oncol. 2015;16:25–35. doi: 10.1016/S1470-2045(14)71159-3. PubMed DOI

Arnedos M., Bayar M.A., Cheaib B., Scott V., Bouakka I., Valent A., Adam J., Leroux-Kozal V., Marty V., Rapinat A., et al. Modulation of Rb phosphorylation and antiproliferative response to palbociclib: The preoperative-palbociclib (POP) randomized clinical trial. Ann. Oncol. 2018;29:1755–1762. doi: 10.1093/annonc/mdy202. PubMed DOI

Rose T.L., Chism D.D., Alva A.S., Deal A.M., Maygarden S.J., Whang Y.E., Kardos J., Drier A., Basch E., Godley P.A., et al. Phase II trial of palbociclib in patients with metastatic urothelial cancer after failure of first-line chemotherapy. Br. J. Cancer. 2018;119:801–807. doi: 10.1038/s41416-018-0229-0. PubMed DOI PMC

Toulmonde M., Blay J.Y., Bouche O., Mir O., Penel N., Isambert N., Duffaud F., Bompas E., Esnaud T., Boidot R., et al. Activity and Safety of Palbociclib in Patients with Advanced Gastrointestinal Stromal Tumors Refractory to Imatinib and Sunitinib: A Biomarker-driven Phase II Study. Clin. Cancer Res. 2019;25:4611–4615. doi: 10.1158/1078-0432.CCR-18-3127. PubMed DOI

Mateo J., Carreira S., Sandhu S., Miranda S., Mossop H., Perez-Lopez R., Nava Rodrigues D., Robinson D., Omlin A., Tunariu N., et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N. Engl. J. Med. 2015;373:1697–1708. doi: 10.1056/NEJMoa1506859. PubMed DOI PMC

Schmitt A., Knittel G., Welcker D., Yang T.P., George J., Nowak M., Leeser U., Buttner R., Perner S., Peifer M., et al. ATM Deficiency Is Associated with Sensitivity to PARP1- and ATR Inhibitors in Lung Adenocarcinoma. Cancer Res. 2017;77:3040–3056. doi: 10.1158/0008-5472.CAN-16-3398. PubMed DOI

Lloyd R.L., Wijnhoven P.W.G., Ramos-Montoya A., Wilson Z., Illuzzi G., Falenta K., Jones G.N., James N., Chabbert C.D., Stott J., et al. Combined PARP and ATR inhibition potentiates genome instability and cell death in ATM-deficient cancer cells. Oncogene. 2020;39:4869–4883. doi: 10.1038/s41388-020-1328-y. PubMed DOI PMC

Downward J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer. 2003;3:11–22. doi: 10.1038/nrc969. PubMed DOI

Castellano E., Molina-Arcas M., Krygowska A.A., East P., Warne P., Nicol A., Downward J. RAS signalling through PI3-Kinase controls cell migration via modulation of Reelin expression. Nat. Commun. 2016;7:11245. doi: 10.1038/ncomms11245. PubMed DOI PMC

Imperial R., Toor O.M., Hussain A., Subramanian J., Masood A. Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: Its clinical implications. Semin. Cancer Biol. 2019;54:14–28. doi: 10.1016/j.semcancer.2017.11.016. PubMed DOI

Smith M.J., Neel B.G., Ikura M. NMR-based functional profiling of RASopathies and oncogenic RAS mutations. Proc. Natl. Acad. Sci. USA. 2013;110:4574–4579. doi: 10.1073/pnas.1218173110. PubMed DOI PMC

Cook J.H., Melloni G.E.M., Gulhan D.C., Park P.J., Haigis K.M. The origins and genetic interactions of KRAS mutations are allele- and tissue-specific. Nat. Commun. 2021;12:1808. doi: 10.1038/s41467-021-22125-z. PubMed DOI PMC

Feig L.A., Cooper G.M. Relationship among guanine nucleotide exchange, GTP hydrolysis, and transforming potential of mutated ras proteins. Mol. Cell Biol. 1988;8:2472–2478. doi: 10.1128/mcb.8.6.2472-2478.1988. PubMed DOI PMC

Poulin E.J., Bera A.K., Lu J., Lin Y.J., Strasser S.D., Paulo J.A., Huang T.Q., Morales C., Yan W., Cook J., et al. Tissue-Specific Oncogenic Activity of KRAS(A146T) Cancer Discov. 2019;9:738–755. doi: 10.1158/2159-8290.CD-18-1220. PubMed DOI PMC

Waters A.M., Der C.J. KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb. Perspect. Med. 2018;8:a031435. doi: 10.1101/cshperspect.a031435. PubMed DOI PMC

Davies H., Bignell G.R., Cox C., Stephens P., Edkins S., Clegg S., Teague J., Woffendin H., Garnett M.J., Bottomley W., et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766. PubMed DOI

Cantwell-Dorris E.R., O’Leary J.J., Sheils O.M. BRAFV600E: Implications for carcinogenesis and molecular therapy. Mol. Cancer Ther. 2011;10:385–394. doi: 10.1158/1535-7163.MCT-10-0799. PubMed DOI

Falini B., Martelli M.P., Tiacci E. BRAF V600E mutation in hairy cell leukemia: From bench to bedside. Blood. 2016;128:1918–1927. doi: 10.1182/blood-2016-07-418434. PubMed DOI

Ding L., Getz G., Wheeler D.A., Mardis E.R., McLellan M.D., Cibulskis K., Sougnez C., Greulich H., Muzny D.M., Morgan M.B., et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075. doi: 10.1038/nature07423. PubMed DOI PMC

Upadhyaya M., Kluwe L., Spurlock G., Monem B., Majounie E., Mantripragada K., Ruggieri M., Chuzhanova N., Evans D.G., Ferner R., et al. Germline and somatic NF1 gene mutation spectrum in NF1-associated malignant peripheral nerve sheath tumors (MPNSTs) Hum. Mutat. 2008;29:74–82. doi: 10.1002/humu.20601. PubMed DOI

Brems H., Beert E., de Ravel T., Legius E. Mechanisms in the pathogenesis of malignant tumours in neurofibromatosis type 1. Lancet Oncol. 2009;10:508–515. doi: 10.1016/S1470-2045(09)70033-6. PubMed DOI

Hodis E., Watson I.R., Kryukov G.V., Arold S.T., Imielinski M., Theurillat J.P., Nickerson E., Auclair D., Li L., Place C., et al. A landscape of driver mutations in melanoma. Cell. 2012;150:251–263. doi: 10.1016/j.cell.2012.06.024. PubMed DOI PMC

Dumbrava E.I., Meric-Bernstam F. Personalized cancer therapy-leveraging a knowledge base for clinical decision-making. Cold Spring Harb. Mol. Case Stud. 2018;4:a001578. doi: 10.1101/mcs.a001578. PubMed DOI PMC

Cox A.D., Fesik S.W., Kimmelman A.C., Luo J., Der C.J. Drugging the undruggable RAS: Mission possible? Nat. Rev. Drug Discov. 2014;13:828–851. doi: 10.1038/nrd4389. PubMed DOI PMC

Keeton A.B., Salter E.A., Piazza G.A. The RAS-Effector Interaction as a Drug Target. Cancer Res. 2017;77:221–226. doi: 10.1158/0008-5472.CAN-16-0938. PubMed DOI PMC

Liu P., Wang Y., Li X. Targeting the untargetable KRAS in cancer therapy. Acta Pharm. Sin. B. 2019;9:871–879. doi: 10.1016/j.apsb.2019.03.002. PubMed DOI PMC

Sheridan C. Grail of RAS cancer drugs within reach. Nat. Biotechnol. 2020;38:6–8. doi: 10.1038/s41587-019-0382-x. PubMed DOI

Blair H.A. Sotorasib: First Approval. Drugs. 2021;81:1573–1579. doi: 10.1007/s40265-021-01574-2. PubMed DOI PMC

Riely G.J., Ou S.H.I., Rybkin I., Spira A., Papadopoulos K., Sabari J.K., Johnson M., Heist R.S., Bazhenova L., Barve M., et al. 99O_PR KRYSTAL-1: Activity and preliminary pharmacodynamic (PD) analysis of adagrasib (MRTX849) in patients (Pts) with advanced non–small cell lung cancer (NSCLC) harboring KRASG12C mutation. J. Thorac. Oncol. 2021;16:S751–S752. doi: 10.1016/S1556-0864(21)01941-9. DOI

Flaherty K.T., Yasothan U., Kirkpatrick P. Vemurafenib. Nat. Rev. Drug Discov. 2011;10:811–812. doi: 10.1038/nrd3579. PubMed DOI

Ballantyne A.D., Garnock-Jones K.P. Dabrafenib: First global approval. Drugs. 2013;73:1367–1376. doi: 10.1007/s40265-013-0095-2. PubMed DOI

Dummer R., Ascierto P.A., Gogas H.J., Arance A., Mandala M., Liszkay G., Garbe C., Schadendorf D., Krajsova I., Gutzmer R., et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018;19:603–615. doi: 10.1016/S1470-2045(18)30142-6. PubMed DOI

Zhou D., Mattner J., Cantu C., 3rd, Schrantz N., Yin N., Gao Y., Sagiv Y., Hudspeth K., Wu Y.P., Yamashita T., et al. Lysosomal glycosphingolipid recognition by NKT cells. Science. 2004;306:1786–1789. doi: 10.1126/science.1103440. PubMed DOI

Groux-Degroote S., van Dijk S.M., Wolthoorn J., Neumann S., Theos A.C., De Maziere A.M., Klumperman J., van Meer G., Sprong H. Glycolipid-dependent sorting of melanosomal from lysosomal membrane proteins by lumenal determinants. Traffic. 2008;9:951–963. doi: 10.1111/j.1600-0854.2008.00740.x. PubMed DOI

Wang D., Dubois R.N. Eicosanoids and cancer. Nat. Rev. Cancer. 2010;10:181–193. doi: 10.1038/nrc2809. PubMed DOI PMC

Hishikawa D., Hashidate T., Shimizu T., Shindou H. Diversity and function of membrane glycerophospholipids generated by the remodeling pathway in mammalian cells. J. Lipid Res. 2014;55:799–807. doi: 10.1194/jlr.R046094. PubMed DOI PMC

Gyamfi D., Ofori Awuah E., Owusu S. Chapter 2—Lipid Metabolism: An Overview. In: Patel V.B., editor. The Molecular Nutrition of Fats. Academic Press; Cambridge, MA, USA: 2019. pp. 17–32. DOI

DeBerardinis R.J., Lum J.J., Hatzivassiliou G., Thompson C.B. The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20. doi: 10.1016/j.cmet.2007.10.002. PubMed DOI

Vander Heiden M.G., Cantley L.C., Thompson C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 2009;324:1029–1033. doi: 10.1126/science.1160809. PubMed DOI PMC

Zhao L., Vogt P.K. Helical domain and kinase domain mutations in p110alpha of phosphatidylinositol 3-kinase induce gain of function by different mechanisms. Proc. Natl. Acad. Sci. USA. 2008;105:2652–2657. doi: 10.1073/pnas.0712169105. PubMed DOI PMC

Leontiadou H., Galdadas I., Athanasiou C., Cournia Z. Insights into the mechanism of the PIK3CA E545K activating mutation using MD simulations. Sci. Rep. 2018;8:15544. doi: 10.1038/s41598-018-27044-6. PubMed DOI PMC

Gkeka P., Evangelidis T., Pavlaki M., Lazani V., Christoforidis S., Agianian B., Cournia Z. Investigating the structure and dynamics of the PIK3CA wild-type and H1047R oncogenic mutant. PLoS Comput. Biol. 2014;10:e1003895. doi: 10.1371/journal.pcbi.1003895. PubMed DOI PMC

Ikenoue T., Kanai F., Hikiba Y., Obata T., Tanaka Y., Imamura J., Ohta M., Jazag A., Guleng B., Tateishi K., et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res. 2005;65:4562–4567. doi: 10.1158/0008-5472.CAN-04-4114. PubMed DOI

Kang S., Bader A.G., Vogt P.K. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc. Natl. Acad. Sci. USA. 2005;102:802–807. doi: 10.1073/pnas.0408864102. PubMed DOI PMC

Bader A.G., Kang S., Vogt P.K. Cancer-specific mutations in PIK3CA are oncogenic in vivo. Proc. Natl. Acad. Sci. USA. 2006;103:1475–1479. doi: 10.1073/pnas.0510857103. PubMed DOI PMC

Dogruluk T., Tsang Y.H., Espitia M., Chen F., Chen T., Chong Z., Appadurai V., Dogruluk A., Eterovic A.K., Bonnen P.E., et al. Identification of Variant-Specific Functions of PIK3CA by Rapid Phenotyping of Rare Mutations. Cancer Res. 2015;75:5341–5354. doi: 10.1158/0008-5472.CAN-15-1654. PubMed DOI PMC

Rohrig F., Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat. Rev. Cancer. 2016;16:732–749. doi: 10.1038/nrc.2016.89. PubMed DOI

Snaebjornsson M.T., Janaki-Raman S., Schulze A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab. 2020;31:62–76. doi: 10.1016/j.cmet.2019.11.010. PubMed DOI

Georgescu M.M., Kirsch K.H., Akagi T., Shishido T., Hanafusa H. The tumor-suppressor activity of PTEN is regulated by its carboxyl-terminal region. Proc. Natl. Acad. Sci. USA. 1999;96:10182–10187. doi: 10.1073/pnas.96.18.10182. PubMed DOI PMC

Papa A., Wan L., Bonora M., Salmena L., Song M.S., Hobbs R.M., Lunardi A., Webster K., Ng C., Newton R.H., et al. Cancer-associated PTEN mutants act in a dominant-negative manner to suppress PTEN protein function. Cell. 2014;157:595–610. doi: 10.1016/j.cell.2014.03.027. PubMed DOI PMC

Rashmi R., DeSelm C., Helms C., Bowcock A., Rogers B.E., Rader J.L., Rader J., Grigsby P.W., Schwarz J.K. AKT inhibitors promote cell death in cervical cancer through disruption of mTOR signaling and glucose uptake. PLoS ONE. 2014;9:e92948. doi: 10.1371/journal.pone.0092948. PubMed DOI PMC

Leslie N.R., den Hertog J. Mutant PTEN in Cancer: Worse Than Nothing. Cell. 2014;157:527–529. doi: 10.1016/j.cell.2014.04.008. PubMed DOI

Salmena L., Carracedo A., Pandolfi P.P. Tenets of PTEN tumor suppression. Cell. 2008;133:403–414. doi: 10.1016/j.cell.2008.04.013. PubMed DOI

Choi S.W., Lee Y., Shin K., Koo H., Kim D., Sa J.K., Cho H.J., Shin H.M., Lee S.J., Kim H., et al. Mutation-specific non-canonical pathway of PTEN as a distinct therapeutic target for glioblastoma. Cell Death Dis. 2021;12:374. doi: 10.1038/s41419-021-03657-0. PubMed DOI PMC

Kloosterhof N.K., Bralten L.B., Dubbink H.J., French P.J., van den Bent M.J. Isocitrate dehydrogenase-1 mutations: A fundamentally new understanding of diffuse glioma? Lancet Oncol. 2011;12:83–91. doi: 10.1016/S1470-2045(10)70053-X. PubMed DOI

Clark O., Yen K., Mellinghoff I.K. Molecular Pathways: Isocitrate Dehydrogenase Mutations in Cancer. Clin. Cancer Res. 2016;22:1837–1842. doi: 10.1158/1078-0432.CCR-13-1333. PubMed DOI PMC

Dang L., White D.W., Gross S., Bennett B.D., Bittinger M.A., Driggers E.M., Fantin V.R., Jang H.G., Jin S., Keenan M.C., et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2010;465:966. doi: 10.1038/nature09132. PubMed DOI PMC

Shim E.H., Livi C.B., Rakheja D., Tan J., Benson D., Parekh V., Kho E.Y., Ghosh A.P., Kirkman R., Velu S., et al. L-2-Hydroxyglutarate: An epigenetic modifier and putative oncometabolite in renal cancer. Cancer Discov. 2014;4:1290–1298. doi: 10.1158/2159-8290.CD-13-0696. PubMed DOI PMC

Parker S.J., Metallo C.M. Metabolic consequences of oncogenic IDH mutations. Pharmacol. Ther. 2015;152:54–62. doi: 10.1016/j.pharmthera.2015.05.003. PubMed DOI PMC

Yang H., Ye D., Guan K.L., Xiong Y. IDH1 and IDH2 mutations in tumorigenesis: Mechanistic insights and clinical perspectives. Clin. Cancer Res. 2012;18:5562–5571. doi: 10.1158/1078-0432.CCR-12-1773. PubMed DOI PMC

Reitman Z.J., Jin G., Karoly E.D., Spasojevic I., Yang J., Kinzler K.W., He Y., Bigner D.D., Vogelstein B., Yan H. Profiling the effects of isocitrate dehydrogenase 1 and 2 mutations on the cellular metabolome. Proc. Natl. Acad. Sci. USA. 2011;108:3270–3275. doi: 10.1073/pnas.1019393108. PubMed DOI PMC

Esmaeili M., Hamans B.C., Navis A.C., van Horssen R., Bathen T.F., Gribbestad I.S., Leenders W.P., Heerschap A. IDH1 R132H mutation generates a distinct phospholipid metabolite profile in glioma. Cancer Res. 2014;74:4898–4907. doi: 10.1158/0008-5472.CAN-14-0008. PubMed DOI

Zhou L., Wang Z., Hu C., Zhang C., Kovatcheva-Datchary P., Yu D., Liu S., Ren F., Wang X., Li Y., et al. Integrated Metabolomics and Lipidomics Analyses Reveal Metabolic Reprogramming in Human Glioma with IDH1 Mutation. J. Proteome Res. 2019;18:960–969. doi: 10.1021/acs.jproteome.8b00663. PubMed DOI

Hudes G., Carducci M., Tomczak P., Dutcher J., Figlin R., Kapoor A., Staroslawska E., Sosman J., McDermott D., Bodrogi I., et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 2007;356:2271–2281. doi: 10.1056/NEJMoa066838. PubMed DOI

Motzer R.J., Escudier B., Oudard S., Hutson T.E., Porta C., Bracarda S., Grunwald V., Thompson J.A., Figlin R.A., Hollaender N., et al. Efficacy of everolimus in advanced renal cell carcinoma: A double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008;372:449–456. doi: 10.1016/S0140-6736(08)61039-9. PubMed DOI

Benjamin D., Colombi M., Moroni C., Hall M.N. Rapamycin passes the torch: A new generation of mTOR inhibitors. Nat. Rev. Drug Discov. 2011;10:868–880. doi: 10.1038/nrd3531. PubMed DOI

Blair H.A. Duvelisib: First Global Approval. Drugs. 2018;78:1847–1853. doi: 10.1007/s40265-018-1013-4. PubMed DOI

Markham A. Alpelisib: First Global Approval. Drugs. 2019;79:1249–1253. doi: 10.1007/s40265-019-01161-6. PubMed DOI

Dhillon S. Ivosidenib: First Global Approval. Drugs. 2018;78:1509–1516. doi: 10.1007/s40265-018-0978-3. PubMed DOI PMC

Katso R., Okkenhaug K., Ahmadi K., White S., Timms J., Waterfield M.D. Cellular function of phosphoinositide 3-kinases: Implications for development, homeostasis, and cancer. Annu. Rev. Cell Dev. Biol. 2001;17:615–675. doi: 10.1146/annurev.cellbio.17.1.615. PubMed DOI

Engelman J.A., Luo J., Cantley L.C. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 2006;7:606–619. doi: 10.1038/nrg1879. PubMed DOI

Courtney K.D., Corcoran R.B., Engelman J.A. The PI3K pathway as drug target in human cancer. J. Clin. Oncol. 2010;28:1075–1083. doi: 10.1200/JCO.2009.25.3641. PubMed DOI PMC

Fruman D.A., Chiu H., Hopkins B.D., Bagrodia S., Cantley L.C., Abraham R.T. The PI3K Pathway in Human Disease. Cell. 2017;170:605–635. doi: 10.1016/j.cell.2017.07.029. PubMed DOI PMC

Cheung L.W., Yu S., Zhang D., Li J., Ng P.K., Panupinthu N., Mitra S., Ju Z., Yu Q., Liang H., et al. Naturally occurring neomorphic PIK3R1 mutations activate the MAPK pathway, dictating therapeutic response to MAPK pathway inhibitors. Cancer Cell. 2014;26:479–494. doi: 10.1016/j.ccell.2014.08.017. PubMed DOI PMC

Seton-Rogers S. Signalling: Uncovering new functions of PI3K mutations. Nat. Rev. Cancer. 2014;14:766–767. doi: 10.1038/nrc3863. PubMed DOI

Moukarzel L.A., Da Cruz Paula A., Ferrando L., Hoang T., Sebastiao A.P.M., Pareja F., Park K.J., Jungbluth A.A., Capella G., Pineda M., et al. Clonal relationship and directionality of progression of synchronous endometrial and ovarian carcinomas in patients with DNA mismatch repair-deficiency associated syndromes. Mod. Pathol. 2021;34:994–1007. doi: 10.1038/s41379-020-00721-6. PubMed DOI PMC

Sun M., Hillmann P., Hofmann B.T., Hart J.R., Vogt P.K. Cancer-derived mutations in the regulatory subunit p85alpha of phosphoinositide 3-kinase function through the catalytic subunit p110alpha. Proc. Natl. Acad. Sci. USA. 2010;107:15547–15552. doi: 10.1073/pnas.1009652107. PubMed DOI PMC

Li X., Lau A.Y.T., Ng A.S.N., Aldehaiman A., Zhou Y., Ng P.K.S., Arold S.T., Cheung L.W.T. Cancer-associated mutations in the p85alpha N-terminal SH2 domain activate a spectrum of receptor tyrosine kinases. Proc. Natl. Acad. Sci. USA. 2021;118:e2101751118. doi: 10.1073/pnas.2101751118. PubMed DOI PMC

Song M., Bode A.M., Dong Z., Lee M.H. AKT as a Therapeutic Target for Cancer. Cancer Res. 2019;79:1019–1031. doi: 10.1158/0008-5472.CAN-18-2738. PubMed DOI

Martorana F., Motta G., Pavone G., Motta L., Stella S., Vitale S.R., Manzella L., Vigneri P. AKT Inhibitors: New Weapons in the Fight Against Breast Cancer? Front. Pharmacol. 2021;12:662232. doi: 10.3389/fphar.2021.662232. PubMed DOI PMC

Adamaki M., Zoumpourlis V. Prostate Cancer Biomarkers: From diagnosis to prognosis and precision-guided therapeutics. Pharmacol. Ther. 2021;228:107932. doi: 10.1016/j.pharmthera.2021.107932. PubMed DOI

Deng L., Wang C., Spencer E., Yang L., Braun A., You J., Slaughter C., Pickart C., Chen Z.J. Activation of the IkappaB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain. Cell. 2000;103:351–361. doi: 10.1016/S0092-8674(00)00126-4. PubMed DOI

Pickart C.M. Ubiquitin in chains. Trends Biochem. Sci. 2000;25:544–548. doi: 10.1016/S0968-0004(00)01681-9. PubMed DOI

Spence J., Gali R.R., Dittmar G., Sherman F., Karin M., Finley D. Cell cycle-regulated modification of the ribosome by a variant multiubiquitin chain. Cell. 2000;102:67–76. doi: 10.1016/S0092-8674(00)00011-8. PubMed DOI

Huen M.S., Grant R., Manke I., Minn K., Yu X., Yaffe M.B., Chen J. RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly. Cell. 2007;131:901–914. doi: 10.1016/j.cell.2007.09.041. PubMed DOI PMC

Mukhopadhyay D., Riezman H. Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science. 2007;315:201–205. doi: 10.1126/science.1127085. PubMed DOI

Yumimoto K., Akiyoshi S., Ueo H., Sagara Y., Onoyama I., Ueo H., Ohno S., Mori M., Mimori K., Nakayama K.I. F-box protein FBXW7 inhibits cancer metastasis in a non-cell-autonomous manner. J. Clin. Investig. 2015;125:621–635. doi: 10.1172/JCI78782. PubMed DOI PMC

Yeh C.H., Bellon M., Nicot C. FBXW7: A critical tumor suppressor of human cancers. Mol. Cancer. 2018;17:115. doi: 10.1186/s12943-018-0857-2. PubMed DOI PMC

Close V., Close W., Kugler S.J., Reichenzeller M., Yosifov D.Y., Bloehdorn J., Pan L., Tausch E., Westhoff M.A., Dohner H., et al. FBXW7 mutations reduce binding of NOTCH1, leading to cleaved NOTCH1 accumulation and target gene activation in CLL. Blood. 2019;133:830–839. doi: 10.1182/blood-2018-09-874529. PubMed DOI

Koepp D.M., Schaefer L.K., Ye X., Keyomarsi K., Chu C., Harper J.W., Elledge S.J. Phosphorylation-dependent ubiquitination of cyclin E by the SCFFbw7 ubiquitin ligase. Science. 2001;294:173–177. doi: 10.1126/science.1065203. PubMed DOI

Oberg C., Li J., Pauley A., Wolf E., Gurney M., Lendahl U. The Notch intracellular domain is ubiquitinated and negatively regulated by the mammalian Sel-10 homolog. J. Biol. Chem. 2001;276:35847–35853. doi: 10.1074/jbc.M103992200. PubMed DOI

Yada M., Hatakeyama S., Kamura T., Nishiyama M., Tsunematsu R., Imaki H., Ishida N., Okumura F., Nakayama K., Nakayama K.I. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. EMBO J. 2004;23:2116–2125. doi: 10.1038/sj.emboj.7600217. PubMed DOI PMC

Wei W., Jin J., Schlisio S., Harper J.W., Kaelin W.G., Jr. The v-Jun point mutation allows c-Jun to escape GSK3-dependent recognition and destruction by the Fbw7 ubiquitin ligase. Cancer Cell. 2005;8:25–33. doi: 10.1016/j.ccr.2005.06.005. PubMed DOI

Mao J.H., Kim I.J., Wu D., Climent J., Kang H.C., DelRosario R., Balmain A. FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor suppression. Science. 2008;321:1499–1502. doi: 10.1126/science.1162981. PubMed DOI PMC

Inuzuka H., Shaik S., Onoyama I., Gao D., Tseng A., Maser R.S., Zhai B., Wan L., Gutierrez A., Lau A.W., et al. SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction. Nature. 2011;471:104–109. doi: 10.1038/nature09732. PubMed DOI PMC

Liu X., Wang L., Zhao K., Thompson P.R., Hwang Y., Marmorstein R., Cole P.A. The structural basis of protein acetylation by the p300/CBP transcriptional coactivator. Nature. 2008;451:846–850. doi: 10.1038/nature06546. PubMed DOI

Delvecchio M., Gaucher J., Aguilar-Gurrieri C., Ortega E., Panne D. Structure of the p300 catalytic core and implications for chromatin targeting and HAT regulation. Nat. Struct. Mol. Biol. 2013;20:1040–1046. doi: 10.1038/nsmb.2642. PubMed DOI

Pao G.M., Janknecht R., Ruffner H., Hunter T., Verma I.M. CBP/p300 interact with and function as transcriptional coactivators of BRCA1. Proc. Natl. Acad. Sci. USA. 2000;97:1020–1025. doi: 10.1073/pnas.97.3.1020. PubMed DOI PMC

Chan H.M., Krstic-Demonacos M., Smith L., Demonacos C., La Thangue N.B. Acetylation control of the retinoblastoma tumour-suppressor protein. Nat. Cell Biol. 2001;3:667–674. doi: 10.1038/35083062. PubMed DOI

Grossman S.R. p300/CBP/p53 interaction and regulation of the p53 response. Eur. J. Biochem. 2001;268:2773–2778. doi: 10.1046/j.1432-1327.2001.02226.x. PubMed DOI

Friedrichs N., Jager R., Paggen E., Rudlowski C., Merkelbach-Bruse S., Schorle H., Buettner R. Distinct spatial expression patterns of AP-2alpha and AP-2gamma in non-neoplastic human breast and breast cancer. Mod. Pathol. 2005;18:431–438. doi: 10.1038/modpathol.3800292. PubMed DOI

Salloum R., McConechy M.K., Mikael L.G., Fuller C., Drissi R., DeWire M., Nikbakht H., De Jay N., Yang X., Boue D., et al. Characterizing temporal genomic heterogeneity in pediatric high-grade gliomas. Acta Neuropathol. Commun. 2017;5:78. doi: 10.1186/s40478-017-0479-8. PubMed DOI PMC

Shi D., Pop M.S., Kulikov R., Love I.M., Kung A.L., Grossman S.R. CBP and p300 are cytoplasmic E4 polyubiquitin ligases for p53. Proc. Natl. Acad. Sci. USA. 2009;106:16275–16280. doi: 10.1073/pnas.0904305106. PubMed DOI PMC

Fahraeus R., Olivares-Illana V. MDM2’s social network. Oncogene. 2014;33:4365–4376. doi: 10.1038/onc.2013.410. PubMed DOI

Klein A.M., de Queiroz R.M., Venkatesh D., Prives C. The roles and regulation of MDM2 and MDMX: It is not just about p53. Genes Dev. 2021;35:575–601. doi: 10.1101/gad.347872.120. PubMed DOI PMC

Attar N., Kurdistani S.K. Exploitation of EP300 and CREBBP Lysine Acetyltransferases by Cancer. Cold Spring Harb. Perspect. Med. 2017;7:a026534. doi: 10.1101/cshperspect.a026534. PubMed DOI PMC

Pasqualucci L., Dominguez-Sola D., Chiarenza A., Fabbri G., Grunn A., Trifonov V., Kasper L.H., Lerach S., Tang H., Ma J., et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature. 2011;471:189–195. doi: 10.1038/nature09730. PubMed DOI PMC

Peifer M., Fernandez-Cuesta L., Sos M.L., George J., Seidel D., Kasper L.H., Plenker D., Leenders F., Sun R., Zander T., et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat. Genet. 2012;44:1104–1110. doi: 10.1038/ng.2396. PubMed DOI PMC

Merk D.J., Ohli J., Merk N.D., Thatikonda V., Morrissy S., Schoof M., Schmid S.N., Harrison L., Filser S., Ahlfeld J., et al. Opposing Effects of CREBBP Mutations Govern the Phenotype of Rubinstein-Taybi Syndrome and Adult SHH Medulloblastoma. Dev. Cell. 2018;44:709–724.e706. doi: 10.1016/j.devcel.2018.02.012. PubMed DOI

Mondello P., Tadros S., Teater M., Fontan L., Chang A.Y., Jain N., Yang H., Singh S., Ying H.Y., Chu C.S., et al. Selective Inhibition of HDAC3 Targets Synthetic Vulnerabilities and Activates Immune Surveillance in Lymphoma. Cancer Discov. 2020;10:440–459. doi: 10.1158/2159-8290.CD-19-0116. PubMed DOI PMC

Deng L., Meng T., Chen L., Wei W., Wang P. The role of ubiquitination in tumorigenesis and targeted drug discovery. Signal Transduct. Target. Ther. 2020;5:11. doi: 10.1038/s41392-020-0107-0. PubMed DOI PMC

Lu G., Middleton R.E., Sun H., Naniong M., Ott C.J., Mitsiades C.S., Wong K.K., Bradner J.E., Kaelin W.G., Jr. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science. 2014;343:305–309. doi: 10.1126/science.1244917. PubMed DOI PMC

Stewart A.K. Medicine. How thalidomide works against cancer. Science. 2014;343:256–257. doi: 10.1126/science.1249543. PubMed DOI PMC

Fricker L.D. Proteasome Inhibitor Drugs. Annu. Rev. Pharmacol. Toxicol. 2020;60:457–476. doi: 10.1146/annurev-pharmtox-010919-023603. PubMed DOI

Dick L.R., Fleming P.E. Building on bortezomib: Second-generation proteasome inhibitors as anti-cancer therapy. Drug Discov. Today. 2010;15:243–249. doi: 10.1016/j.drudis.2010.01.008. PubMed DOI

Nelson W.J., Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 2004;303:1483–1487. doi: 10.1126/science.1094291. PubMed DOI PMC

Gattinoni L., Zhong X.S., Palmer D.C., Ji Y., Hinrichs C.S., Yu Z., Wrzesinski C., Boni A., Cassard L., Garvin L.M., et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 2009;15:808–813. doi: 10.1038/nm.1982. PubMed DOI PMC

Petersen C.P., Reddien P.W. Wnt signaling and the polarity of the primary body axis. Cell. 2009;139:1056–1068. doi: 10.1016/j.cell.2009.11.035. PubMed DOI

Spranger S., Gajewski T.F. A new paradigm for tumor immune escape: Beta-catenin-driven immune exclusion. J. Immunother. Cancer. 2015;3:43. doi: 10.1186/s40425-015-0089-6. PubMed DOI PMC

Junge H.J. Ligand-Selective Wnt Receptor Complexes in CNS Blood Vessels: RECK and GPR124 Plugged In. Neuron. 2017;95:983–985. doi: 10.1016/j.neuron.2017.08.026. PubMed DOI

Miyoshi Y., Nagase H., Ando H., Horii A., Ichii S., Nakatsuru S., Aoki T., Miki Y., Mori T., Nakamura Y. Somatic mutations of the APC gene in colorectal tumors: Mutation cluster region in the APC gene. Hum. Mol. Genet. 1992;1:229–233. doi: 10.1093/hmg/1.4.229. PubMed DOI

Fearnhead N.S., Britton M.P., Bodmer W.F. The ABC of APC. Hum. Mol. Genet. 2001;10:721–733. doi: 10.1093/hmg/10.7.721. PubMed DOI

Azzopardi D., Dallosso A.R., Eliason K., Hendrickson B.C., Jones N., Rawstorne E., Colley J., Moskvina V., Frye C., Sampson J.R., et al. Multiple rare nonsynonymous variants in the adenomatous polyposis coli gene predispose to colorectal adenomas. Cancer Res. 2008;68:358–363. doi: 10.1158/0008-5472.CAN-07-5733. PubMed DOI

Pai S.G., Carneiro B.A., Mota J.M., Costa R., Leite C.A., Barroso-Sousa R., Kaplan J.B., Chae Y.K., Giles F.J. Wnt/beta-catenin pathway: Modulating anticancer immune response. J. Hematol. Oncol. 2017;10:101. doi: 10.1186/s13045-017-0471-6. PubMed DOI PMC

Imperial R., Ahmed Z., Toor O.M., Erdogan C., Khaliq A., Case P., Case J., Kennedy K., Cummings L.S., Melton N., et al. Comparative proteogenomic analysis of right-sided colon cancer, left-sided colon cancer and rectal cancer reveals distinct mutational profiles. Mol. Cancer. 2018;17:177. doi: 10.1186/s12943-018-0923-9. PubMed DOI PMC

Ficari F., Cama A., Valanzano R., Curia M.C., Palmirotta R., Aceto G., Esposito D.L., Crognale S., Lombardi A., Messerini L., et al. APC gene mutations and colorectal adenomatosis in familial adenomatous polyposis. Br. J. Cancer. 2000;82:348–353. doi: 10.1054/bjoc.1999.0925. PubMed DOI PMC

Mihalatos M., Danielides I., Belogianni J., Harokopos E., Papadopoulou E., Kalimanis G., Tsiava M., Triantafillidis J.K., Kosmidis P.A., Fountzilas G., et al. Novel mutations of the APC gene in familial adenomatous polyposis in Greek patients. Cancer Genet. Cytogenet. 2003;141:65–70. doi: 10.1016/S0165-4608(02)00723-9. PubMed DOI

Kitagawa M., Hatakeyama S., Shirane M., Matsumoto M., Ishida N., Hattori K., Nakamichi I., Kikuchi A., Nakayama K., Nakayama K. An F-box protein, FWD1, mediates ubiquitin-dependent proteolysis of beta-catenin. EMBO J. 1999;18:2401–2410. doi: 10.1093/emboj/18.9.2401. PubMed DOI PMC

Kikuchi A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 2003;94:225–229. doi: 10.1111/j.1349-7006.2003.tb01424.x. PubMed DOI PMC

Morin P.J., Sparks A.B., Korinek V., Barker N., Clevers H., Vogelstein B., Kinzler K.W. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science. 1997;275:1787–1790. doi: 10.1126/science.275.5307.1787. PubMed DOI

Rubinfeld B., Robbins P., El-Gamil M., Albert I., Porfiri E., Polakis P. Stabilization of beta-catenin by genetic defects in melanoma cell lines. Science. 1997;275:1790–1792. doi: 10.1126/science.275.5307.1790. PubMed DOI

Liu C., Li Y., Semenov M., Han C., Baeg G.H., Tan Y., Zhang Z., Lin X., He X. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 2002;108:837–847. doi: 10.1016/S0092-8674(02)00685-2. PubMed DOI

Rebouissou S., Franconi A., Calderaro J., Letouze E., Imbeaud S., Pilati C., Nault J.C., Couchy G., Laurent A., Balabaud C., et al. Genotype-phenotype correlation of CTNNB1 mutations reveals different ss-catenin activity associated with liver tumor progression. Hepatology. 2016;64:2047–2061. doi: 10.1002/hep.28638. PubMed DOI

Tu J., Park S., Yu W., Zhang S., Wu L., Carmon K., Liu Q.J. The most common RNF43 mutant G659Vfs*41 is fully functional in inhibiting Wnt signaling and unlikely to play a role in tumorigenesis. Sci. Rep. 2019;9:18557. doi: 10.1038/s41598-019-54931-3. PubMed DOI PMC

Hao H.X., Xie Y., Zhang Y., Charlat O., Oster E., Avello M., Lei H., Mickanin C., Liu D., Ruffner H., et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature. 2012;485:195–200. doi: 10.1038/nature11019. PubMed DOI

Koo B.K., Spit M., Jordens I., Low T.Y., Stange D.E., van de Wetering M., van Es J.H., Mohammed S., Heck A.J., Maurice M.M., et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature. 2012;488:665–669. doi: 10.1038/nature11308. PubMed DOI

UniProt C. UniProt: The universal protein knowledgebase in 2021. Nucleic Acids Res. 2021;49:D480–D489. doi: 10.1093/nar/gkaa1100. PubMed DOI PMC

Spit M., Fenderico N., Jordens I., Radaszkiewicz T., Lindeboom R.G., Bugter J.M., Cristobal A., Ootes L., van Osch M., Janssen E., et al. RNF43 truncations trap CK1 to drive niche-independent self-renewal in cancer. EMBO J. 2020;39:e103932. doi: 10.15252/embj.2019103932. PubMed DOI PMC

Anastas J.N., Moon R.T. WNT signalling pathways as therapeutic targets in cancer. Nat. Rev. Cancer. 2013;13:11–26. doi: 10.1038/nrc3419. PubMed DOI

Jung Y.S., Park J.I. Wnt signaling in cancer: Therapeutic targeting of Wnt signaling beyond beta-catenin and the destruction complex. Exp. Mol. Med. 2020;52:183–191. doi: 10.1038/s12276-020-0380-6. PubMed DOI PMC

Hori K., Sen A., Artavanis-Tsakonas S. Notch signaling at a glance. J. Cell Sci. 2013;126:2135–2140. doi: 10.1242/jcs.127308. PubMed DOI PMC

Bray S.J. Notch signalling in context. Nat. Rev. Mol. Cell Biol. 2016;17:722–735. doi: 10.1038/nrm.2016.94. PubMed DOI

Siebel C., Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol. Rev. 2017;97:1235–1294. doi: 10.1152/physrev.00005.2017. PubMed DOI

Pancewicz J., Taylor J.M., Datta A., Baydoun H.H., Waldmann T.A., Hermine O., Nicot C. Notch signaling contributes to proliferation and tumor formation of human T-cell leukemia virus type 1-associated adult T-cell leukemia. Proc. Natl. Acad. Sci. USA. 2010;107:16619–16624. doi: 10.1073/pnas.1010722107. PubMed DOI PMC

Yeh C.H., Bellon M., Pancewicz-Wojtkiewicz J., Nicot C. Oncogenic mutations in the FBXW7 gene of adult T-cell leukemia patients. Proc. Natl. Acad. Sci. USA. 2016;113:6731–6736. doi: 10.1073/pnas.1601537113. PubMed DOI PMC

Oswald F., Tauber B., Dobner T., Bourteele S., Kostezka U., Adler G., Liptay S., Schmid R.M. p300 acts as a transcriptional coactivator for mammalian Notch-1. Mol. Cell Biol. 2001;21:7761–7774. doi: 10.1128/MCB.21.22.7761-7774.2001. PubMed DOI PMC

Wallberg A.E., Pedersen K., Lendahl U., Roeder R.G. p300 and PCAF act cooperatively to mediate transcriptional activation from chromatin templates by notch intracellular domains in vitro. Mol. Cell Biol. 2002;22:7812–7819. doi: 10.1128/MCB.22.22.7812-7819.2002. PubMed DOI PMC

Huang Y.H., Cai K., Xu P.P., Wang L., Huang C.X., Fang Y., Cheng S., Sun X.J., Liu F., Huang J.Y., et al. CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct. Target. Ther. 2021;6:10. doi: 10.1038/s41392-020-00437-8. PubMed DOI PMC

Majumder S., Crabtree J.S., Golde T.E., Minter L.M., Osborne B.A., Miele L. Targeting Notch in oncology: The path forward. Nat. Rev. Drug Discov. 2021;20:125–144. doi: 10.1038/s41573-020-00091-3. PubMed DOI

Pannuti A., Foreman K., Rizzo P., Osipo C., Golde T., Osborne B., Miele L. Targeting Notch to target cancer stem cells. Clin. Cancer Res. 2010;16:3141–3152. doi: 10.1158/1078-0432.CCR-09-2823. PubMed DOI PMC

Takebe N., Nguyen D., Yang S.X. Targeting notch signaling pathway in cancer: Clinical development advances and challenges. Pharmacol. Ther. 2014;141:140–149. doi: 10.1016/j.pharmthera.2013.09.005. PubMed DOI PMC

Malumbres M., Barbacid M. Cell cycle, CDKs and cancer: A changing paradigm. Nat. Rev. Cancer. 2009;9:153–166. doi: 10.1038/nrc2602. PubMed DOI

Matthews H.K., Bertoli C., de Bruin R.A.M. Cell cycle control in cancer. Nat. Rev. Mol. Cell Biol. 2022;23:74–88. doi: 10.1038/s41580-021-00404-3. PubMed DOI

Cowell J.K., Smith T., Bia B. Frequent constitutional C to T mutations in CGA-arginine codons in the RB1 gene produce premature stop codons in patients with bilateral (hereditary) retinoblastoma. Eur. J. Hum. Genet. 1994;2:281–290. doi: 10.1159/000472372. PubMed DOI

Richter S., Vandezande K., Chen N., Zhang K., Sutherland J., Anderson J., Han L., Panton R., Branco P., Gallie B. Sensitive and efficient detection of RB1 gene mutations enhances care for families with retinoblastoma. Am. J. Hum. Genet. 2003;72:253–269. doi: 10.1086/345651. PubMed DOI PMC

Ayari Jeridi H., Bouguila H., Ansperger-Rescher B., Baroudi O., Mdimegh I., Omran I., Charradi K., Bouzayene H., Benammar-Elgaaied A., Lohmann D.R. Genetic testing in Tunisian families with heritable retinoblastoma using a low cost approach permits accurate risk prediction in relatives and reveals incomplete penetrance in adults. Exp. Eye Res. 2014;124:48–55. doi: 10.1016/j.exer.2014.04.013. PubMed DOI

Yu H.A., Suzawa K., Jordan E., Zehir A., Ni A., Kim R., Kris M.G., Hellmann M.D., Li B.T., Somwar R., et al. Concurrent Alterations in EGFR-Mutant Lung Cancers Associated with Resistance to EGFR Kinase Inhibitors and Characterization of MTOR as a Mediator of Resistance. Clin. Cancer Res. 2018;24:3108–3118. doi: 10.1158/1078-0432.CCR-17-2961. PubMed DOI PMC

Trimarchi J.M., Lees J.A. Sibling rivalry in the E2F family. Nat. Rev. Mol. Cell Biol. 2002;3:11–20. doi: 10.1038/nrm714. PubMed DOI

Burkhart D.L., Sage J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat. Rev. Cancer. 2008;8:671–682. doi: 10.1038/nrc2399. PubMed DOI PMC

Xiong Y., Hannon G.J., Zhang H., Casso D., Kobayashi R., Beach D. p21 is a universal inhibitor of cyclin kinases. Nature. 1993;366:701–704. doi: 10.1038/366701a0. PubMed DOI

Waga S., Hannon G.J., Beach D., Stillman B. The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA. Nature. 1994;369:574–578. doi: 10.1038/369574a0. PubMed DOI

Jackson R.J., Adnane J., Coppola D., Cantor A., Sebti S.M., Pledger W.J. Loss of the cell cycle inhibitors p21(Cip1) and p27(Kip1) enhances tumorigenesis in knockout mouse models. Oncogene. 2002;21:8486–8497. doi: 10.1038/sj.onc.1205946. PubMed DOI

Poole A.J., Heap D., Carroll R.E., Tyner A.L. Tumor suppressor functions for the Cdk inhibitor p21 in the mouse colon. Oncogene. 2004;23:8128–8134. doi: 10.1038/sj.onc.1207994. PubMed DOI

Forster K., Obermeier A., Mitina O., Simon N., Warmuth M., Krause G., Hallek M. Role of p21(WAF1/CIP1) as an attenuator of both proliferative and drug-induced apoptotic signals in BCR-ABL-transformed hematopoietic cells. Ann. Hematol. 2008;87:183–193. doi: 10.1007/s00277-007-0400-9. PubMed DOI

Suzuki A., Tsutomi Y., Miura M., Akahane K. Caspase 3 inactivation to suppress Fas-mediated apoptosis: Identification of binding domain with p21 and ILP and inactivation machinery by p21. Oncogene. 1999;18:1239–1244. doi: 10.1038/sj.onc.1202409. PubMed DOI

Yu F., Megyesi J., Safirstein R.L., Price P.M. Identification of the functional domain of p21(WAF1/CIP1) that protects cells from cisplatin cytotoxicity. Am. J. Physiol. Renal Physiol. 2005;289:F514–F520. doi: 10.1152/ajprenal.00101.2005. PubMed DOI

Suski J.M., Braun M., Strmiska V., Sicinski P. Targeting cell-cycle machinery in cancer. Cancer Cell. 2021;39:759–778. doi: 10.1016/j.ccell.2021.03.010. PubMed DOI PMC

Otto T., Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer. 2017;17:93–115. doi: 10.1038/nrc.2016.138. PubMed DOI PMC

Karran P. DNA double strand break repair in mammalian cells. Curr. Opin. Genet. Dev. 2000;10:144–150. doi: 10.1016/S0959-437X(00)00069-1. PubMed DOI

Chen C.C., Feng W., Lim P.X., Kass E.M., Jasin M. Homology-Directed Repair and the Role of BRCA1, BRCA2, and Related Proteins in Genome Integrity and Cancer. Annu. Rev. Cancer Biol. 2018;2:313–336. doi: 10.1146/annurev-cancerbio-030617-050502. PubMed DOI PMC

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. doi: 10.1038/35056049. PubMed DOI

Khanna K.K., Jackson S.P. DNA double-strand breaks: Signaling, repair and the cancer connection. Nat. Genet. 2001;27:247–254. doi: 10.1038/85798. PubMed DOI

Caburet S., Heddar A., Dardillac E., Creux H., Lambert M., Messiaen S., Tourpin S., Livera G., Lopez B.S., Misrahi M. Homozygous hypomorphic BRCA2 variant in primary ovarian insufficiency without cancer or Fanconi anaemia trait. J. Med. Genet. 2020;58:125–134. doi: 10.1136/jmedgenet-2019-106672. PubMed DOI

Sun P., Li Y., Chao X., Li J., Luo R., Li M., He J. Clinical characteristics and prognostic implications of BRCA-associated tumors in males: A pan-tumor survey. BMC Cancer. 2020;20:994. doi: 10.1186/s12885-020-07481-1. PubMed DOI PMC

Lee H. Cycling with BRCA2 from DNA repair to mitosis. Exp. Cell Res. 2014;329:78–84. doi: 10.1016/j.yexcr.2014.10.008. PubMed DOI

Moynahan M.E., Pierce A.J., Jasin M. BRCA2 is required for homology-directed repair of chromosomal breaks. Mol. Cell. 2001;7:263–272. doi: 10.1016/S1097-2765(01)00174-5. PubMed DOI

Tutt A., Bertwistle D., Valentine J., Gabriel A., Swift S., Ross G., Griffin C., Thacker J., Ashworth A. Mutation in Brca2 stimulates error-prone homology-directed repair of DNA double-strand breaks occurring between repeated sequences. EMBO J. 2001;20:4704–4716. doi: 10.1093/emboj/20.17.4704. PubMed DOI PMC

Liu J., Doty T., Gibson B., Heyer W.D. Human BRCA2 protein promotes RAD51 filament formation on RPA-covered single-stranded DNA. Nat. Struct. Mol. Biol. 2010;17:1260–1262. doi: 10.1038/nsmb.1904. PubMed DOI PMC

Rebbeck T.R., Friebel T.M., Friedman E., Hamann U., Huo D., Kwong A., Olah E., Olopade O.I., Solano A.R., Teo S.H., et al. Mutational spectrum in a worldwide study of 29,700 families with BRCA1 or BRCA2 mutations. Hum. Mutat. 2018;39:593–620. doi: 10.1002/humu.23406. PubMed DOI PMC

Scully R., Chen J., Plug A., Xiao Y., Weaver D., Feunteun J., Ashley T., Livingston D.M. Association of BRCA1 with Rad51 in mitotic and meiotic cells. Cell. 1997;88:265–275. doi: 10.1016/S0092-8674(00)81847-4. PubMed DOI

Zhong Q., Chen C.F., Li S., Chen Y., Wang C.C., Xiao J., Chen P.L., Sharp Z.D., Lee W.H. Association of BRCA1 with the hRad50-hMre11-p95 complex and the DNA damage response. Science. 1999;285:747–750. doi: 10.1126/science.285.5428.747. PubMed DOI

Clark S.L., Rodriguez A.M., Snyder R.R., Hankins G.D., Boehning D. Structure-Function Of The Tumor Suppressor BRCA1. Comput. Struct. Biotechnol. J. 2012;1:e201204005. doi: 10.5936/csbj.201204005. PubMed DOI PMC

Christou C.M., Kyriacou K. BRCA1 and Its Network of Interacting Partners. Biology. 2013;2:40–63. doi: 10.3390/biology2010040. PubMed DOI PMC

Bunting S.F., Callen E., Wong N., Chen H.T., Polato F., Gunn A., Bothmer A., Feldhahn N., Fernandez-Capetillo O., Cao L., et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 2010;141:243–254. doi: 10.1016/j.cell.2010.03.012. PubMed DOI PMC

Zhang J., Yan Z., Wang Y., Wang Y., Guo X., Jing J., Dong X., Dong S., Liu X., Yu X., et al. Cancer-associated 53BP1 mutations induce DNA damage repair defects. Cancer Lett. 2021;501:43–54. doi: 10.1016/j.canlet.2020.12.033. PubMed DOI

Panier S., Boulton S.J. Double-strand break repair: 53BP1 comes into focus. Nat. Rev. Mol. Cell Biol. 2014;15:7–18. doi: 10.1038/nrm3719. PubMed DOI

Mao Z., Bozzella M., Seluanov A., Gorbunova V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell Cycle. 2008;7:2902–2906. doi: 10.4161/cc.7.18.6679. PubMed DOI PMC

Yap T.A., Sandhu S.K., Carden C.P., de Bono J.S. Poly(ADP-ribose) polymerase (PARP) inhibitors: Exploiting a synthetic lethal strategy in the clinic. CA Cancer J. Clin. 2011;61:31–49. doi: 10.3322/caac.20095. PubMed DOI

Furgason J.M., Bahassi E.M. Targeting DNA repair mechanisms in cancer. Pharmacol. Ther. 2013;137:298–308. doi: 10.1016/j.pharmthera.2012.10.009. PubMed DOI

Brown J.S., O’Carrigan B., Jackson S.P., Yap T.A. Targeting DNA Repair in Cancer: Beyond PARP Inhibitors. Cancer Discov. 2017;7:20–37. doi: 10.1158/2159-8290.CD-16-0860. PubMed DOI PMC

Helleday T., Petermann E., Lundin C., Hodgson B., Sharma R.A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer. 2008;8:193–204. doi: 10.1038/nrc2342. PubMed DOI

Deeks E.D. Olaparib: First global approval. Drugs. 2015;75:231–240. doi: 10.1007/s40265-015-0345-6. PubMed DOI

Scott L.J. Niraparib: First Global Approval. Drugs. 2017;77:1029–1034. doi: 10.1007/s40265-017-0752-y. PubMed DOI

Syed Y.Y. Rucaparib: First Global Approval. Drugs. 2017;77:585–592. doi: 10.1007/s40265-017-0716-2. PubMed DOI

Hoy S.M. Talazoparib: First Global Approval. Drugs. 2018;78:1939–1946. doi: 10.1007/s40265-018-1026-z. PubMed DOI

Arora S., Balasubramaniam S., Zhang H., Berman T., Narayan P., Suzman D., Bloomquist E., Tang S., Gong Y., Sridhara R., et al. FDA Approval Summary: Olaparib Monotherapy or in Combination with Bevacizumab for the Maintenance Treatment of Patients with Advanced Ovarian Cancer. Oncologist. 2021;26:e164–e172. doi: 10.1002/onco.13551. PubMed DOI PMC

Kwok M., Davies N., Agathanggelou A., Smith E., Oldreive C., Petermann E., Stewart G., Brown J., Lau A., Pratt G., et al. ATR inhibition induces synthetic lethality and overcomes chemoresistance in TP53- or ATM-defective chronic lymphocytic leukemia cells. Blood. 2016;127:582–595. doi: 10.1182/blood-2015-05-644872. PubMed DOI

Menolfi D., Zha S. ATM, ATR and DNA-PKcs kinases-the lessons from the mouse models: Inhibition not equal deletion. Cell Biosci. 2020;10:8. doi: 10.1186/s13578-020-0376-x. PubMed DOI PMC

Topatana W., Juengpanich S., Li S., Cao J., Hu J., Lee J., Suliyanto K., Ma D., Zhang B., Chen M., et al. Advances in synthetic lethality for cancer therapy: Cellular mechanism and clinical translation. J. Hematol. Oncol. 2020;13:118. doi: 10.1186/s13045-020-00956-5. PubMed DOI PMC

David C.J., Manley J.L. Alternative pre-mRNA splicing regulation in cancer: Pathways and programs unhinged. Genes Dev. 2010;24:2343–2364. doi: 10.1101/gad.1973010. PubMed DOI PMC

Dvinge H., Kim E., Abdel-Wahab O., Bradley R.K. RNA splicing factors as oncoproteins and tumour suppressors. Nat. Rev. Cancer. 2016;16:413–430. doi: 10.1038/nrc.2016.51. PubMed DOI PMC

Climente-Gonzalez H., Porta-Pardo E., Godzik A., Eyras E. The Functional Impact of Alternative Splicing in Cancer. Cell Rep. 2017;20:2215–2226. doi: 10.1016/j.celrep.2017.08.012. PubMed DOI

Rahman M.A., Krainer A.R., Abdel-Wahab O. SnapShot: Splicing Alterations in Cancer. Cell. 2020;180:208.e201. doi: 10.1016/j.cell.2019.12.011. PubMed DOI PMC

Legare S., Cavallone L., Mamo A., Chabot C., Sirois I., Magliocco A., Klimowicz A., Tonin P.N., Buchanan M., Keilty D., et al. The Estrogen Receptor Cofactor SPEN Functions as a Tumor Suppressor and Candidate Biomarker of Drug Responsiveness in Hormone-Dependent Breast Cancers. Cancer Res. 2015;75:4351–4363. doi: 10.1158/0008-5472.CAN-14-3475. PubMed DOI

Ariyoshi M., Schwabe J.W. A conserved structural motif reveals the essential transcriptional repression function of Spen proteins and their role in developmental signaling. Genes Dev. 2003;17:1909–1920. doi: 10.1101/gad.266203. PubMed DOI PMC

Hiriart E., Gruffat H., Buisson M., Mikaelian I., Keppler S., Meresse P., Mercher T., Bernard O.A., Sergeant A., Manet E. Interaction of the Epstein-Barr virus mRNA export factor EB2 with human Spen proteins SHARP, OTT1, and a novel member of the family, OTT3, links Spen proteins with splicing regulation and mRNA export. J. Biol. Chem. 2005;280:36935–36945. doi: 10.1074/jbc.M501725200. PubMed DOI

Bartkowiak B., Greenleaf A.L. Expression, purification, and identification of associated proteins of the full-length hCDK12/CyclinK complex. J. Biol. Chem. 2015;290:1786–1795. doi: 10.1074/jbc.M114.612226. PubMed DOI PMC

Ekumi K.M., Paculova H., Lenasi T., Pospichalova V., Bosken C.A., Rybarikova J., Bryja V., Geyer M., Blazek D., Barboric M. Ovarian carcinoma CDK12 mutations misregulate expression of DNA repair genes via deficient formation and function of the Cdk12/CycK complex. Nucleic Acids Res. 2015;43:2575–2589. doi: 10.1093/nar/gkv101. PubMed DOI PMC

Blazek D., Kohoutek J., Bartholomeeusen K., Johansen E., Hulinkova P., Luo Z., Cimermancic P., Ule J., Peterlin B.M. The Cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev. 2011;25:2158–2172. doi: 10.1101/gad.16962311. PubMed DOI PMC

Krajewska M., Dries R., Grassetti A.V., Dust S., Gao Y., Huang H., Sharma B., Day D.S., Kwiatkowski N., Pomaville M., et al. CDK12 loss in cancer cells affects DNA damage response genes through premature cleavage and polyadenylation. Nat. Commun. 2019;10:1757. doi: 10.1038/s41467-019-09703-y. PubMed DOI PMC

Tien J.F., Mazloomian A., Cheng S.G., Hughes C.S., Chow C.C.T., Canapi L.T., Oloumi A., Trigo-Gonzalez G., Bashashati A., Xu J., et al. CDK12 regulates alternative last exon mRNA splicing and promotes breast cancer cell invasion. Nucleic Acids Res. 2017;45:6698–6716. doi: 10.1093/nar/gkx187. PubMed DOI PMC

Cancer Genome Atlas N. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337. doi: 10.1038/nature11252. PubMed DOI PMC

Schneeweiss A., Park-Simon T.-W., Albanell J., Lassen U., Cortés J., Dieras V., May M., Schindler C., Marmé F., Cejalvo J.M., et al. Phase Ib study evaluating safety and clinical activity of the anti-HER3 antibody lumretuzumab combined with the anti-HER2 antibody pertuzumab and paclitaxel in HER3-positive, HER2-low metastatic breast cancer. Investig. New Drugs. 2018;36:848–859. doi: 10.1007/s10637-018-0562-4. PubMed DOI PMC

Duncan R., Bazar L., Michelotti G., Tomonaga T., Krutzsch H., Avigan M., Levens D. A sequence-specific, single-strand binding protein activates the far upstream element of c-myc and defines a new DNA-binding motif. Genes Dev. 1994;8:465–480. doi: 10.1101/gad.8.4.465. PubMed DOI

Hsiao H.H., Nath A., Lin C.Y., Folta-Stogniew E.J., Rhoades E., Braddock D.T. Quantitative characterization of the interactions among c-myc transcriptional regulators FUSE, FBP, and FIR. Biochemistry. 2010;49:4620–4634. doi: 10.1021/bi9021445. PubMed DOI

Jacob A.G., Singh R.K., Mohammad F., Bebee T.W., Chandler D.S. The splicing factor FUBP1 is required for the efficient splicing of oncogene MDM2 pre-mRNA. J. Biol. Chem. 2014;289:17350–17364. doi: 10.1074/jbc.M114.554717. PubMed DOI PMC

Lee S.C., Abdel-Wahab O. Therapeutic targeting of splicing in cancer. Nat. Med. 2016;22:976–986. doi: 10.1038/nm.4165. PubMed DOI PMC

Urbanski L.M., Leclair N., Anczukow O. Alternative-splicing defects in cancer: Splicing regulators and their downstream targets, guiding the way to novel cancer therapeutics. Wiley Interdiscip. Rev. RNA. 2018;9:e1476. doi: 10.1002/wrna.1476. PubMed DOI PMC

Zhang Y., Qian J., Gu C., Yang Y. Alternative splicing and cancer: A systematic review. Signal Transduct. Target. Ther. 2021;6:78. doi: 10.1038/s41392-021-00486-7. PubMed DOI PMC

Elman J.S., Ni T.K., Mengwasser K.E., Jin D., Wronski A., Elledge S.J., Kuperwasser C. Identification of FUBP1 as a Long Tail Cancer Driver and Widespread Regulator of Tumor Suppressor and Oncogene Alternative Splicing. Cell Rep. 2019;28:3435–3449.e3435. doi: 10.1016/j.celrep.2019.08.060. PubMed DOI PMC

Sailo B.L., Banik K., Girisa S., Bordoloi D., Fan L., Halim C.E., Wang H., Kumar A.P., Zheng D., Mao X., et al. FBXW7 in Cancer: What Has Been Unraveled Thus Far? Cancers. 2019;11:246. doi: 10.3390/cancers11020246. PubMed DOI PMC

Wyatt D.W., Feng W., Conlin M.P., Yousefzadeh M.J., Roberts S.A., Mieczkowski P., Wood R.D., Gupta G.P., Ramsden D.A. Essential Roles for Polymerase theta-Mediated End Joining in the Repair of Chromosome Breaks. Mol. Cell. 2016;63:662–673. doi: 10.1016/j.molcel.2016.06.020. PubMed DOI PMC

Feng W., Simpson D.A., Carvajal-Garcia J., Price B.A., Kumar R.J., Mose L.E., Wood R.D., Rashid N., Purvis J.E., Parker J.S., et al. Genetic determinants of cellular addiction to DNA polymerase theta. Nat. Commun. 2019;10:4286. doi: 10.1038/s41467-019-12234-1. PubMed DOI PMC

Najít záznam

Citační ukazatele

Nahrávání dat ...

Možnosti archivace

Nahrávání dat ...