Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer
Abstract
:Simple Summary
Abstract
1. Introduction
2. Origins and Characteristics of Persisters
2.1. Bacterial Persisters
2.2. Persisters in Cancer
2.3. Origin of Drug-Tolerant Persister Cells
3. Drug-Tolerant Persister Cells: Mechanisms of Drug Tolerance and Therapeutic Vulnerabilities
3.1. Epigenetic Reprogramming/Plasticity
Tumor Type | Target Oncogene | Targeted Therapy | Model System | Mechanism of Drug Tolerance | Susceptibility | Reference |
---|---|---|---|---|---|---|
Epigenetic programming/plasticity | ||||||
NSCLC | EGFR | Gefitinib | PC9 | Repressed chromatin state (KDM5 upregulation; H3K9 methylation-dependent LINE-1 elements) | HDAC (trichostatin) | [23,24] |
Erlotinib | PC9 | Cell fate and lineage plasticity (increased methylation on K116 of Jarid2, with attendant stabilization and recruitment to chromatin of the PRC2 complex) | EZH2 (EPZ-6438) | [86] | ||
EGFR, ALK | Erlotinib, crizotinib | PC9 and xenograft, H3122 | Dynamic transcriptional responses—remodeling of enhancers | CDK7/12 (THZ1) | [77] | |
Breast | HER2, EGFR, PI3K | Lapatanib | SKBR3, EVSA-T | Repressed chromatin state (KDM5 upregulation) | KDM5 (CPI-455) | [39] |
HER2, EGFR | Lapatinib | SKBR | Cell fate and lineage plasticity (increased methylation on K116 of Jarid2, stabilization, and recruitment to chromatin of the PRC2 complex) | EZH2 (EPZ-6438) | [86] | |
MEK | Trametinib | SUM-159PT, HCC1806; xenografts | Transcriptional adaptation | BET (JQ1 and I-BET151) | [74] | |
MEK and PI3K/mTOR | Trametinib and AZD6244; BEZ235 and PI103 | HCC1143, SUM149PT | Transcriptional adaptation | BET inhibitor (JQ1) | [66] | |
Melanoma | BRAF, c-Raf-1 | AZ628 | Hs888, M14 | Repressed chromatin state (KDM5 upregulation) | KDM5 (CPI-455) | [39] |
BRAF | Vemurafenib, bortezomib | WM3734 and xenograft | Repressed chromatin state (H3K4-demethylase JARID1B/KDM5B/PLU-1) | Mitochondrial enzyme (oligomycin, Bz-423, rotenone and phenformin, antimycin A, oligomycin) | [60] | |
BRAF, MEK | Dabrafenib, trametinib | PDX | Cell fate and lineage plasticity (EMT) | RXR (HX531) | [48] | |
RAF, MEK, ERK | Vemurafenib, selumetinib, ERK inhibitor | A375, WM266.4 | Cell fate and lineage plasticity (high MITF expression) | HDAC (panobinostat, vorinostat, or entinostat); forskolin and IBMX (FSK/I) | [82] | |
Glioblastoma | RTK | Dasatinib | GSC6 and GSC8 | Adaptive chromatin remodeling (KDM3, KDM6 upregulation) | KDM (GSKJ4) | [45] |
Colon cancer | BRAF, c-Raf-1 | Vemurafenib | Colo205 | Repressed chromatin state—KDM5 upregulation | KDM5 (CPI-455) | [39] |
Bladder carcinoma | FGFR | BGJ398 | RT112 and xenograft | Dynamic transcriptional response (remodeling of enhancers) | CDK7/12 (THZ1) | [77] |
Basal cell carcinoma | Hedgehog (Smoothened) | Vismodegib | Ptch1–Trp53 mouse model | Cell identity switch (Wnt pathway activation and reprogramming of super-enhancers) | Wnt (function-blocking anti-LRP6 antibody) | [46] |
Activation of bypass and alternative signaling pathways | ||||||
NSCLC | EGFR | Erlotinib | HCC827 and xenograft | AXL kinase signaling | AXL (MP-470 or XL-880) | [87] |
HCC827, HCC4006, and xenograft | Notch3-dependent β-catenin signaling | β-catenin (XAV939, ICG-001) | [88] | |||
H1650 and xenograft | TGF-B-IL-6-STAT3 axis | Innate immune (12-O-tetradecanoyl- and LPS) | [89] | |||
HCC827, PDX | NF-kB signaling | NF-kB (PBS-1086) | [90] | |||
HCC827 and HCC4006 | YAP/FOXM1 axis | CDK (dinaciclib, alvocidib), SAC components (volasertib, ON-01910, ispinesib, SB743921, AMG-900, alisertib) | [91] | |||
Osimertinib | PC9 | AXL kinase signaling | AXL (NPS1034) | [92] | ||
EGF816 | PDCL | FGFR signaling | FGFR (BGJ398) | [93] | ||
Osimertinib, rociletinib | PC9, HCC827, HCC4006, and NCI-H1975 | AURKA signaling | AURKA (MLN8237) | [94] | ||
EGFR, MEK | Selumetinib | PC9, HCC827, HCC2935 | FGF-JAK kinase-Stat3 signaling | JAK (ponatinib/ruxolitinib) FGF (PD173074) inhibitors | [67] | |
Osimertinib, trametinib | PC9, HCC827, HCC4006; xenograft | YAP/TEAD/SLUG signaling | YAP (XAV939) | [56] | ||
Melanoma | BRAF, MAPK | Vemurafenib, trametinib | PDX | AXL signaling | AXL antibody–drug conjugate (AXL-107-MMAE) | [80] |
BRAF, MEK | Dabrafenib, trametinib | PDX | RXR signaling | RXR (HX531) | [48] | |
Colon cancer | BRAF | Vemurafenib | VACO432, SNU- C5, HT29, KM20, WiDr; xenografts | EGFR-mediated feedback activation | EGFR (cetuximab, gefitinib, or erlotinib) | [95] |
Colorectal cancer | BRAF | Vemurafenib | HT-29 and WiDr; xenografts | EGFR-mediated feedback activation | EGFR (erlotinib) | [96] |
BCC | Hedgehog (Smoothened) | Vismodegib | Xenograft | Wnt signaling | Wnt (LGK-974) | [47] |
Glioblastoma | RTK | Dasatinib | GSC6 and GSC8 | Notch signaling | KDM (GSKJ4) | [45] |
Various cancers | KRAS | ARS-1620, AMG 510 | KRAS G12C cell lines, xenograft | RAS pathway feedback reactivation | SHP2 (SHP099) | [97] |
Metabolic reprogramming | ||||||
NSCLC | EGFR | Erlotinib | PC9 | ALDH upregulation to maintain ROS levels | ALDH (disulfiram) | [43] |
Melanoma | BRAF | Vemurafenib | Primary cells | Bioenergetic metabolism (mitochondrial respiratory chain) | Mitochondrial enzymes (oligomycin and Bz-423, rotenone and phenformin, antimycin A, and oligomycin) | [60] |
BRAF, MEK | Vemurafenib, dabrafenib, trametinib | A375 | Lipid hydroperoxidase (GPX4) pathway | GPX4 (RSL3) | [40] | |
Gastric | MET | Crizotinib | GTL-16, MKN-45 | ALDH upregulation to maintain ROS levels | ALDH (disulfiram) | [43] |
Breast | HER2 | Lapatinib | BT474 | Lipid hydroperoxidase (GPX4) pathway | GPX4 (RSL3) | [40] |
Interactions with the tumor microenvironment | ||||||
Lung | EGFR | CL-387785 | Cancer tissue-originated spheroids (CTOS) | MIG6/ERRFI1/RALT/Gene33 induction by hypoxia | PI3K (LY294002), MEK (trametinib) | [98] |
Melanoma | BRAF | Vemurafenib | 928MEL and 624MEL | Stromal secretion of HGF | MET (GDC-0712) | [16] |
SK-MEL-5, SK-MEL-28, G-361 | Stromal cell secretion of HGF | MAPK (PD184352), PI3K–AKT (crizotinib, MK-2206) | [99] | |||
5555 and 4434 | Activation of MAFs | FAK (PF573228, PF562271, FAKi14) | [100] |
3.2. Activation of Bypass and Alternative Signaling Pathways
3.3. Suppression of Apoptosis
3.4. Metabolic Reprogramming
3.5. Interactions with the Tumor Microenvironment
3.6. Adaptive Mutagenesis
4. Persisters in the Clinic
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating Mutations in the Epidermal Growth Factor Receptor Underlying Responsiveness of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef]
- Soda, M.; Choi, Y.L.; Enomoto, M.; Takada, S.; Yamashita, Y.; Ishikawa, S.; Fujiwara, S.; Watanabe, H.; Kurashina, K.; Hatanaka, H.; et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007, 448, 561–566. [Google Scholar] [CrossRef]
- Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
- Mok, T.; Camidge, D.R.; Gadgeel, S.M.; Rosell, R.; Dziadziuszko, R.; Kim, D.W.; Perol, M.; Ou, S.I.; Ahn, J.S.; Shaw, A.T.; et al. Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study. Ann. Oncol. 2020, 31, 1056–1064. [Google Scholar] [CrossRef] [PubMed]
- Hyman, D.M.; Puzanov, I.; Subbiah, V.; Faris, J.E.; Chau, I.; Blay, J.Y.; Wolf, J.; Raje, N.S.; Diamond, E.L.; Hollebecque, A.; et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N. Engl. J. Med. 2015, 373, 726–736. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Grob, J.J.; Stroyakovskiy, D.; Karaszewska, B.; Hauschild, A.; Levchenko, E.; Chiarion Sileni, V.; Schachter, J.; Garbe, C.; Bondarenko, I.; et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N. Engl. J. Med. 2019, 381, 626–636. [Google Scholar] [CrossRef]
- Paez, J.G. EGFR Mutations in Lung Cancer: Correlation with Clinical Response to Gefitinib Therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef] [Green Version]
- Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef]
- Chatterjee, N.; Bivona, T.G. Polytherapy and Targeted Cancer Drug Resistance. Trends Cancer 2019, 5, 170–182. [Google Scholar] [CrossRef]
- Yoda, S.; Dagogo-Jack, I.; Hata, A.N. Targeting oncogenic drivers in lung cancer: Recent progress, current challenges and future opportunities. Pharmacology 2019, 193, 20–30. [Google Scholar] [CrossRef]
- Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Jänne, P.A.; Kocher, O.; Meyerson, M.; Johnson, B.E.; Eck, M.J.; Tenen, D.G.; Halmos, B. EGFRMutation and Resistance of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef] [PubMed]
- Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and Histological Evolution of Lung Cancers Acquiring Resistance to EGFR Inhibitors. Sci. Transl. Med. 2011, 3, 75ra26. [Google Scholar] [CrossRef] [Green Version]
- Yun, C.-H.; Mengwasser, K.E.; Toms, A.V.; Woo, M.S.; Greulich, H.; Wong, K.-K.; Meyerson, M.; Eck, M.J. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl. Acad. Sci. USA 2008, 105, 2070–2075. [Google Scholar] [CrossRef] [Green Version]
- Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.-M.; Zhao, X.; Christensen, J.; et al. MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef]
- Turke, A.B.; Zejnullahu, K.; Wu, Y.L.; Song, Y.; Dias-Santagata, D.; Lifshits, E.; Toschi, L.; Rogers, A.; Mok, T.; Sequist, L.; et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell 2010, 17, 77–88. [Google Scholar] [CrossRef] [Green Version]
- Wilson, T.R.; Fridlyand, J.; Yan, Y.; Penuel, E.; Burton, L.; Chan, E.; Peng, J.; Lin, E.; Wang, Y.; Sosman, J.; et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 2012, 487, 505–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schoenfeld, A.J.; Chan, J.M.; Kubota, D.; Sato, H.; Rizvi, H.; Daneshbod, Y.; Chang, J.C.; Paik, P.K.; Offin, M.; Arcila, M.E.; et al. Tumor Analyses Reveal Squamous Transformation and Off-Target Alterations As Early Resistance Mechanisms to First-line Osimertinib in EGFR-Mutant Lung Cancer. Clin. Cancer Res. 2020, 26, 2654–2663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oser, M.G.; Niederst, M.J.; Sequist, L.V.; Engelman, J.A. Transformation from non-small-cell lung cancer to small-cell lung cancer: Molecular drivers and cells of origin. Lancet Oncol. 2015, 16, e165–e172. [Google Scholar] [CrossRef] [Green Version]
- Niederst, M.J.; Hu, H.; Mulvey, H.E.; Lockerman, E.L.; Garcia, A.R.; Piotrowska, Z.; Sequist, L.V.; Engelman, J.A. The Allelic Context of the C797S Mutation Acquired upon Treatment with Third-Generation EGFR Inhibitors Impacts Sensitivity to Subsequent Treatment Strategies. Clin. Cancer Res. 2015, 21, 3924–3933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhang, H.E.; Ruddy, D.A.; Krishnamurthy Radhakrishna, V.; Caushi, J.X.; Zhao, R.; Hims, M.M.; Singh, A.P.; Kao, I.; Rakiec, D.; Shaw, P.; et al. Studying clonal dynamics in response to cancer therapy using high-complexity barcoding. Nat. Med. 2015, 21, 440–448. [Google Scholar] [CrossRef] [PubMed]
- Su, K.Y.; Chen, H.Y.; Li, K.C.; Kuo, M.L.; Yang, J.C.; Chan, W.K.; Ho, B.C.; Chang, G.C.; Shih, J.Y.; Yu, S.L.; et al. Pretreatment epidermal growth factor receptor (EGFR) T790M mutation predicts shorter EGFR tyrosine kinase inhibitor response duration in patients with non-small-cell lung cancer. J. Clin. Oncol. 2012, 30, 433–440. [Google Scholar] [CrossRef] [Green Version]
- Ye, X.; Zhu, Z.Z.; Zhong, L.; Lu, Y.; Sun, Y.; Yin, X.; Yang, Z.; Zhu, G.; Ji, Q. High T790M detection rate in TKI-naive NSCLC with EGFR sensitive mutation: Truth or artifact? J. Thorac. Oncol. 2013, 8, 1118–1120. [Google Scholar] [CrossRef] [Green Version]
- Sharma, S.V.; Lee, D.Y.; Li, B.; Quinlan, M.P.; Takahashi, F.; Maheswaran, S.; McDermott, U.; Azizian, N.; Zou, L.; Fischbach, M.A.; et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 2010, 141, 69–80. [Google Scholar] [CrossRef] [Green Version]
- Guler, G.D.; Tindell, C.A.; Pitti, R.; Wilson, C.; Nichols, K.; KaiWai Cheung, T.; Kim, H.J.; Wongchenko, M.; Yan, Y.; Haley, B.; et al. Repression of Stress-Induced LINE-1 Expression Protects Cancer Cell Subpopulations from Lethal Drug Exposure. Cancer Cell 2017, 32, 221–237. [Google Scholar] [CrossRef] [PubMed]
- Hata, A.N.; Niederst, M.J.; Archibald, H.L.; Gomez-Caraballo, M.; Siddiqui, F.M.; Mulvey, H.E.; Maruvka, Y.E.; Ji, F.; Bhang, H.E.; Krishnamurthy Radhakrishna, V.; et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 2016, 22, 262–269. [Google Scholar] [CrossRef] [PubMed]
- Ramirez, M.; Rajaram, S.; Steininger, R.J.; Osipchuk, D.; Roth, M.A.; Morinishi, L.S.; Evans, L.; Ji, W.; Hsu, C.H.; Thurley, K.; et al. Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells. Nat. Commun. 2016, 7, 10690. [Google Scholar] [CrossRef]
- Neel, D.S.; Bivona, T.G. Resistance is futile: Overcoming resistance to targeted therapies in lung adenocarcinoma. NPJ Precis. Oncol. 2017, 1. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D. Rethinking the war on cancer. Lancet 2014, 383, 558–563. [Google Scholar] [CrossRef]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [Green Version]
- Balaban, N.Q. Bacterial Persistence as a Phenotypic Switch. Science 2004, 305, 1622–1625. [Google Scholar] [CrossRef] [Green Version]
- Holden, D.W. Persisters unmasked. Science 2015, 347, 30–32. [Google Scholar] [CrossRef] [PubMed]
- Fisher, R.A.; Gollan, B.; Helaine, S. Persistent bacterial infections and persister cells. Nat. Rev. Microbiol. 2017, 15, 453–464. [Google Scholar] [CrossRef] [PubMed]
- Hobby, G.L.; Meyer, K.; Chaffee, E. Observations on the Mechanism of Action of Penicillin. Proc. Soc. Exp. Biol. Med. 1942, 50, 281–285. [Google Scholar] [CrossRef]
- Bigger, J. Treatment of staphylococcal infections with penicillin by intermittent sterilisation. Lancet 1944, 244, 497–500. [Google Scholar] [CrossRef]
- Fan, W.; Tang, Z.; Yin, L.; Morrison, B.; Hafez-Khayyata, S.; Fu, P.; Huang, H.; Bagai, R.; Jiang, S.; Kresak, A.; et al. MET-independent lung cancer cells evading EGFR kinase inhibitors are therapeutically susceptible to BH3 mimetic agents. Cancer Res. 2011, 71, 4494–4505. [Google Scholar] [CrossRef] [Green Version]
- Rothenberg, S.M.; Concannon, K.; Cullen, S.; Boulay, G.; Turke, A.B.; Faber, A.C.; Lockerman, E.L.; Rivera, M.N.; Engelman, J.A.; Maheswaran, S.; et al. Inhibition of mutant EGFR in lung cancer cells triggers SOX2-FOXO6-dependent survival pathways. Elife 2015, 4. [Google Scholar] [CrossRef]
- Kobayashi, I.; Takahashi, F.; Nurwidya, F.; Nara, T.; Hashimoto, M.; Murakami, A.; Yagishita, S.; Tajima, K.; Hidayat, M.; Shimada, N.; et al. Oct4 plays a crucial role in the maintenance of gefitinib-resistant lung cancer stem cells. Biochem. Biophys. Res. Commun. 2016, 473, 125–132. [Google Scholar] [CrossRef]
- Murakami, A.; Takahashi, F.; Nurwidya, F.; Kobayashi, I.; Minakata, K.; Hashimoto, M.; Nara, T.; Kato, M.; Tajima, K.; Shimada, N.; et al. Hypoxia increases gefitinib-resistant lung cancer stem cells through the activation of insulin-like growth factor 1 receptor. PLoS ONE 2014, 9, e86459. [Google Scholar] [CrossRef]
- Vinogradova, M.; Gehling, V.S.; Gustafson, A.; Arora, S.; Tindell, C.A.; Wilson, C.; Williamson, K.E.; Guler, G.D.; Gangurde, P.; Manieri, W.; et al. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat. Chem. Biol. 2016, 12, 531–538. [Google Scholar] [CrossRef]
- Hangauer, M.J.; Viswanathan, V.S.; Ryan, M.J.; Bole, D.; Eaton, J.K.; Matov, A.; Galeas, J.; Dhruv, H.D.; Berens, M.E.; Schreiber, S.L.; et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 2017, 551, 247–250. [Google Scholar] [CrossRef] [Green Version]
- Ravindran Menon, D.; Das, S.; Krepler, C.; Vultur, A.; Rinner, B.; Schauer, S.; Kashofer, K.; Wagner, K.; Zhang, G.; Bonyadi Rad, E.; et al. A stress-induced early innate response causes multidrug tolerance in melanoma. Oncogene 2015, 34, 4448–4459. [Google Scholar] [CrossRef] [Green Version]
- Moody, S.E.; Schinzel, A.C.; Singh, S.; Izzo, F.; Strickland, M.R.; Luo, L.; Thomas, S.R.; Boehm, J.S.; Kim, S.Y.; Wang, Z.C.; et al. PRKACA mediates resistance to HER2-targeted therapy in breast cancer cells and restores anti-apoptotic signaling. Oncogene 2015, 34, 2061–2071. [Google Scholar] [CrossRef] [Green Version]
- Raha, D.; Wilson, T.R.; Peng, J.; Peterson, D.; Yue, P.; Evangelista, M.; Wilson, C.; Merchant, M.; Settleman, J. The cancer stem cell marker aldehyde dehydrogenase is required to maintain a drug-tolerant tumor cell subpopulation. Cancer Res. 2014, 74, 3579–3590. [Google Scholar] [CrossRef] [Green Version]
- Knoechel, B.; Roderick, J.E.; Williamson, K.E.; Zhu, J.; Lohr, J.G.; Cotton, M.J.; Gillespie, S.M.; Fernandez, D.; Ku, M.; Wang, H.; et al. An epigenetic mechanism of resistance to targeted therapy in T cell acute lymphoblastic leukemia. Nat. Genet. 2014, 46, 364–370. [Google Scholar] [CrossRef]
- Liau, B.B.; Sievers, C.; Donohue, L.K.; Gillespie, S.M.; Flavahan, W.A.; Miller, T.E.; Venteicher, A.S.; Hebert, C.H.; Carey, C.D.; Rodig, S.J.; et al. Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. Cell Stem Cell 2017, 20, 233–246.e237. [Google Scholar] [CrossRef] [Green Version]
- Biehs, B.; Dijkgraaf, G.J.P.; Piskol, R.; Alicke, B.; Boumahdi, S.; Peale, F.; Gould, S.E.; de Sauvage, F.J. A cell identity switch allows residual BCC to survive Hedgehog pathway inhibition. Nature 2018, 562, 429–433. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Danes, A.; Larsimont, J.C.; Liagre, M.; Munoz-Couselo, E.; Lapouge, G.; Brisebarre, A.; Dubois, C.; Suppa, M.; Sukumaran, V.; Del Marmol, V.; et al. A slow-cycling LGR5 tumour population mediates basal cell carcinoma relapse after therapy. Nature 2018, 562, 434–438. [Google Scholar] [CrossRef] [PubMed]
- Rambow, F.; Rogiers, A.; Marin-Bejar, O.; Aibar, S.; Femel, J.; Dewaele, M.; Karras, P.; Brown, D.; Chang, Y.H.; Debiec-Rychter, M.; et al. Toward Minimal Residual Disease-Directed Therapy in Melanoma. Cell 2018, 174, 843–855.e819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brauner, A.; Fridman, O.; Gefen, O.; Balaban, N.Q. Distinguishing between resistance, tolerance and persistence to antibiotic treatment. Nat. Rev. Microbiol. 2016, 14, 320–330. [Google Scholar] [CrossRef] [PubMed]
- Kochanowski, K.; Morinishi, L.; Altschuler, S.; Wu, L. Drug persistence - from antibiotics to cancer therapies. Curr Opin. Syst. Biol. 2018, 10, 1–8. [Google Scholar] [CrossRef]
- Salgia, R.; Kulkarni, P. The Genetic/Non-genetic Duality of Drug ’Resistance’ in Cancer. Trends Cancer 2018, 4, 110–118. [Google Scholar] [CrossRef] [PubMed]
- Viale, A.; Pettazzoni, P.; Lyssiotis, C.A.; Ying, H.; Sánchez, N.; Marchesini, M.; Carugo, A.; Green, T.; Seth, S.; Giuliani, V.; et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014, 514, 628–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Li, Y.; Yu, T.S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vallette, F.M.; Olivier, C.; Lezot, F.; Oliver, L.; Cochonneau, D.; Lalier, L.; Cartron, P.F.; Heymann, D. Dormant, quiescent, tolerant and persister cells: Four synonyms for the same target in cancer. Biochem. Pharm. 2019, 162, 169–176. [Google Scholar] [CrossRef] [Green Version]
- Banelli, B.; Carra, E.; Barbieri, F.; Wurth, R.; Parodi, F.; Pattarozzi, A.; Carosio, R.; Forlani, A.; Allemanni, G.; Marubbi, D.; et al. The histone demethylase KDM5A is a key factor for the resistance to temozolomide in glioblastoma. Cell Cycle 2015, 14, 3418–3429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kurppa, K.J.; Liu, Y.; To, C.; Zhang, T.; Fan, M.; Vajdi, A.; Knelson, E.H.; Xie, Y.; Lim, K.; Cejas, P.; et al. Treatment-Induced Tumor Dormancy through YAP-Mediated Transcriptional Reprogramming of the Apoptotic Pathway. Cancer Cell 2020, 37, 104–122.e112. [Google Scholar] [CrossRef]
- Ewald, J.A.; Desotelle, J.A.; Wilding, G.; Jarrard, D.F. Therapy-induced senescence in cancer. J. Natl. Cancer Inst. 2010, 102, 1536–1546. [Google Scholar] [CrossRef] [Green Version]
- Gupta, P.B.; Fillmore, C.M.; Jiang, G.; Shapira, S.D.; Tao, K.; Kuperwasser, C.; Lander, E.S. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 2011, 146, 633–644. [Google Scholar] [CrossRef] [Green Version]
- Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [Green Version]
- Roesch, A.; Vultur, A.; Bogeski, I.; Wang, H.; Zimmermann, K.M.; Speicher, D.; Korbel, C.; Laschke, M.W.; Gimotty, P.A.; Philipp, S.E.; et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 2013, 23, 811–825. [Google Scholar] [CrossRef] [Green Version]
- Shaffer, S.M.; Dunagin, M.C.; Torborg, S.R.; Torre, E.A.; Emert, B.; Krepler, C.; Beqiri, M.; Sproesser, K.; Brafford, P.A.; Xiao, M.; et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 2017, 546, 431–435. [Google Scholar] [CrossRef] [Green Version]
- Trumpp, A.; Wiestler, O.D. Mechanisms of Disease: Cancer stem cells--targeting the evil twin. Nat. Clin. Pr. Oncol. 2008, 5, 337–347. [Google Scholar] [CrossRef] [PubMed]
- Hinohara, K.; Wu, H.J.; Vigneau, S.; McDonald, T.O.; Igarashi, K.J.; Yamamoto, K.N.; Madsen, T.; Fassl, A.; Egri, S.B.; Papanastasiou, M.; et al. KDM5 Histone Demethylase Activity Links Cellular Transcriptomic Heterogeneity to Therapeutic Resistance. Cancer Cell 2018, 34, 939–953.e939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goldman, A.; Majumder, B.; Dhawan, A.; Ravi, S.; Goldman, D.; Kohandel, M.; Majumder, P.K.; Sengupta, S. Temporally sequenced anticancer drugs overcome adaptive resistance by targeting a vulnerable chemotherapy-induced phenotypic transition. Nat. Commun. 2015, 6, 6139. [Google Scholar] [CrossRef] [Green Version]
- Glasspool, R.M.; Teodoridis, J.M.; Brown, R. Epigenetics as a mechanism driving polygenic clinical drug resistance. Br. J. Cancer 2006, 94, 1087–1092. [Google Scholar] [CrossRef] [Green Version]
- Risom, T.; Langer, E.M.; Chapman, M.P.; Rantala, J.; Fields, A.J.; Boniface, C.; Alvarez, M.J.; Kendsersky, N.D.; Pelz, C.R.; Johnson-Camacho, K.; et al. Differentiation-state plasticity is a targetable resistance mechanism in basal-like breast cancer. Nat. Commun. 2018, 9, 3815. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.J.; Zhuang, G.; Cao, Y.; Du, P.; Kim, H.J.; Settleman, J. Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell 2014, 26, 207–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boumahdi, S.; de Sauvage, F.J. The great escape: Tumour cell plasticity in resistance to targeted therapy. Nat. Rev. Drug. Discov. 2020, 19, 39–56. [Google Scholar] [CrossRef] [PubMed]
- Pfister, S.X.; Ashworth, A. Marked for death: Targeting epigenetic changes in cancer. Nat. Rev. Drug Discov. 2017, 16, 241–263. [Google Scholar] [CrossRef]
- Flavahan, W.A.; Gaskell, E.; Bernstein, B.E. Epigenetic plasticity and the hallmarks of cancer. Science 2017, 357. [Google Scholar] [CrossRef] [Green Version]
- Staberg, M.; Rasmussen, R.D.; Michaelsen, S.R.; Pedersen, H.; Jensen, K.E.; Villingshøj, M.; Skjoth-Rasmussen, J.; Brennum, J.; Vitting-Seerup, K.; Poulsen, H.S.; et al. Targeting glioma stem-like cell survival and chemoresistance through inhibition of lysine-specific histone demethylase KDM2B. Mol. Oncol. 2018, 12, 406–420. [Google Scholar] [CrossRef]
- Roesch, A.; Fukunaga-Kalabis, M.; Schmidt, E.C.; Zabierowski, S.E.; Brafford, P.A.; Vultur, A.; Basu, D.; Gimotty, P.; Vogt, T.; Herlyn, M. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 2010, 141, 583–594. [Google Scholar] [CrossRef] [Green Version]
- Bradner, J.E.; Hnisz, D.; Young, R.A. Transcriptional Addiction in Cancer. Cell 2017, 168, 629–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zawistowski, J.S.; Bevill, S.M.; Goulet, D.R.; Stuhlmiller, T.J.; Beltran, A.S.; Olivares-Quintero, J.F.; Singh, D.; Sciaky, N.; Parker, J.S.; Rashid, N.U.; et al. Enhancer Remodeling during Adaptive Bypass to MEK Inhibition Is Attenuated by Pharmacologic Targeting of the P-TEFb Complex. Cancer Discov. 2017, 7, 302–321. [Google Scholar] [CrossRef] [Green Version]
- Zanconato, F.; Battilana, G.; Forcato, M.; Filippi, L.; Azzolin, L.; Manfrin, A.; Quaranta, E.; Di Biagio, D.; Sigismondo, G.; Guzzardo, V.; et al. Transcriptional addiction in cancer cells is mediated by YAP/TAZ through BRD4. Nat. Med. 2018, 24, 1599–1610. [Google Scholar] [CrossRef]
- Kwiatkowski, N.; Zhang, T.; Rahl, P.B.; Abraham, B.J.; Reddy, J.; Ficarro, S.B.; Dastur, A.; Amzallag, A.; Ramaswamy, S.; Tesar, B.; et al. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014, 511, 616–620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rusan, M.; Li, K.; Li, Y.; Christensen, C.L.; Abraham, B.J.; Kwiatkowski, N.; Buczkowski, K.A.; Bockorny, B.; Chen, T.; Li, S.; et al. Suppression of Adaptive Responses to Targeted Cancer Therapy by Transcriptional Repression. Cancer Discov. 2018, 8, 59–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kemper, K.; De Goeje, P.L.; Peeper, D.S.; Van Amerongen, R. Phenotype Switching: Tumor Cell Plasticity as a Resistance Mechanism and Target for Therapy. Cancer Res. 2014, 74, 5937–5941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boshuizen, J.; Koopman, L.A.; Krijgsman, O.; Shahrabi, A.; Van Den Heuvel, E.G.; Ligtenberg, M.A.; Vredevoogd, D.W.; Kemper, K.; Kuilman, T.; Song, J.-Y.; et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat. Med. 2018, 24, 203–212. [Google Scholar] [CrossRef] [PubMed]
- Hoek, K.S.; Eichhoff, O.M.; Schlegel, N.C.; Döbbeling, U.; Kobert, N.; Schaerer, L.; Hemmi, S.; Dummer, R. In vivo Switching of Human Melanoma Cells between Proliferative and Invasive States. Cancer Res. 2008, 68, 650–656. [Google Scholar] [CrossRef] [Green Version]
- Johannessen, C.M.; Johnson, L.A.; Piccioni, F.; Townes, A.; Frederick, D.T.; Donahue, M.K.; Narayan, R.; Flaherty, K.T.; Wargo, J.A.; Root, D.E.; et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature 2013, 504, 138–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, H.; Hugo, W.; Kong, X.; Hong, A.; Koya, R.C.; Moriceau, G.; Chodon, T.; Guo, R.; Johnson, D.B.; Dahlman, K.B.; et al. Acquired Resistance and Clonal Evolution in Melanoma during BRAF Inhibitor Therapy. Cancer Discov. 2014, 4, 80–93. [Google Scholar] [CrossRef] [Green Version]
- Muller, J.; Krijgsman, O.; Tsoi, J.; Robert, L.; Hugo, W.; Song, C.; Kong, X.; Possik, P.A.; Cornelissen-Steijger, P.D.; Geukes Foppen, M.H.; et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 2014, 5, 5712. [Google Scholar] [CrossRef] [PubMed]
- Dravis, C.; Chung, C.Y.; Lytle, N.K.; Herrera-Valdez, J.; Luna, G.; Trejo, C.L.; Reya, T.; Wahl, G.M. Epigenetic and Transcriptomic Profiling of Mammary Gland Development and Tumor Models Disclose Regulators of Cell State Plasticity. Cancer Cell 2018, 34, 466–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pham, V.; Pitti, R.; Tindell, C.A.; Cheung, T.K.; Masselot, A.; Stephan, J.P.; Guler, G.D.; Wilson, C.; Lill, J.; Arnott, D.; et al. Proteomic Analyses Identify a Novel Role for EZH2 in the Initiation of Cancer Cell Drug Tolerance. J. Proteome Res. 2020, 19, 1533–1547. [Google Scholar] [CrossRef]
- Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; Abdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef]
- Arasada, R.R.; Shilo, K.; Yamada, T.; Zhang, J.; Yano, S.; Ghanem, R.; Wang, W.; Takeuchi, S.; Fukuda, K.; Katakami, N.; et al. Notch3-dependent beta-catenin signaling mediates EGFR TKI drug persistence in EGFR mutant NSCLC. Nat. Commun. 2018, 9, 3198. [Google Scholar] [CrossRef]
- Yao, Z.; Fenoglio, S.; Gao, D.C.; Camiolo, M.; Stiles, B.; Lindsted, T.; Schlederer, M.; Johns, C.; Altorki, N.; Mittal, V.; et al. TGF- IL-6 axis mediates selective and adaptive mechanisms of resistance to molecular targeted therapy in lung cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 15535–15540. [Google Scholar] [CrossRef] [Green Version]
- Blakely, C.M.; Pazarentzos, E.; Olivas, V.; Asthana, S.; Yan, J.J.; Tan, I.; Hrustanovic, G.; Chan, E.; Lin, L.; Neel, D.S.; et al. NF-kappaB-activating complex engaged in response to EGFR oncogene inhibition drives tumor cell survival and residual disease in lung cancer. Cell Rep. 2015, 11, 98–110. [Google Scholar] [CrossRef] [Green Version]
- Nilsson, M.B.; Sun, H.; Robichaux, J.; Pfeifer, M.; Mcdermott, U.; Travers, J.; Diao, L.; Xi, Y.; Tong, P.; Shen, L.; et al. A YAP/FOXM1 axis mediates EMT-associated EGFR inhibitor resistance and increased expression of spindle assembly checkpoint components. Sci. Transl. Med. 2020, 12, eaaz4589. [Google Scholar] [CrossRef]
- Taniguchi, H.; Yamada, T.; Wang, R.; Tanimura, K.; Adachi, Y.; Nishiyama, A.; Tanimoto, A.; Takeuchi, S.; Araujo, L.H.; Boroni, M.; et al. AXL confers intrinsic resistance to osimertinib and advances the emergence of tolerant cells. Nat. Commun. 2019, 10, 259. [Google Scholar] [CrossRef]
- Raoof, S.; Mulford, I.J.; Frisco-Cabanos, H.; Nangia, V.; Timonina, D.; Labrot, E.; Hafeez, N.; Bilton, S.J.; Drier, Y.; Ji, F.; et al. Targeting FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer. Oncogene 2019, 38, 6399–6413. [Google Scholar] [CrossRef]
- Shah, K.N.; Bhatt, R.; Rotow, J.; Rohrberg, J.; Olivas, V.; Wang, V.E.; Hemmati, G.; Martins, M.M.; Maynard, A.; Kuhn, J.; et al. Aurora kinase A drives the evolution of resistance to third-generation EGFR inhibitors in lung cancer. Nat. Med. 2019, 25, 111–118. [Google Scholar] [CrossRef]
- Prahallad, A.; Sun, C.; Huang, S.; Di Nicolantonio, F.; Salazar, R.; Zecchin, D.; Beijersbergen, R.L.; Bardelli, A.; Bernards, R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012, 483, 100–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corcoran, R.B.; Ebi, H.; Turke, A.B.; Coffee, E.M.; Nishino, M.; Cogdill, A.P.; Brown, R.D.; Della Pelle, P.; Dias-Santagata, D.; Hung, K.E.; et al. EGFR-Mediated Reactivation of MAPK Signaling Contributes to Insensitivity of BRAF-Mutant Colorectal Cancers to RAF Inhibition with Vemurafenib. Cancer Discov. 2012, 2, 227–235. [Google Scholar] [CrossRef] [Green Version]
- Ryan, M.B.; De La Cruz, F.F.; Phat, S.; Myers, D.T.; Wong, E.; Shahzade, H.A.; Hong, C.B.; Corcoran, R.B. Vertical Pathway Inhibition Overcomes Adaptive Feedback Resistance to KRASG12C Inhibition. Clin. Cancer Res. 2020, 26, 1633–1643. [Google Scholar] [CrossRef] [Green Version]
- Endo, H.; Okami, J.; Okuyama, H.; Nishizawa, Y.; Imamura, F.; Inoue, M. The induction of MIG6 under hypoxic conditions is critical for dormancy in primary cultured lung cancer cells with activating EGFR mutations. Oncogene 2017, 36, 2824–2834. [Google Scholar] [CrossRef] [PubMed]
- Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; Davis, A.; Mongare, M.M.; Gould, J.; Frederick, D.T.; et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012, 487, 500–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirata, E.; Girotti, M.R.; Viros, A.; Hooper, S.; Spencer-Dene, B.; Matsuda, M.; Larkin, J.; Marais, R.; Sahai, E. Intravital imaging reveals how BRAF inhibition generates drug-tolerant microenvironments with high integrin beta1/FAK signaling. Cancer Cell 2015, 27, 574–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishak, C.A.; Classon, M.; De Carvalho, D.D. Deregulation of Retroelements as an Emerging Therapeutic Opportunity in Cancer. Trends Cancer 2018, 4, 583–597. [Google Scholar] [CrossRef]
- Goel, S.; Decristo, M.J.; Watt, A.C.; Brinjones, H.; Sceneay, J.; Li, B.B.; Khan, N.; Ubellacker, J.M.; Xie, S.; Metzger-Filho, O.; et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 2017, 548, 471–475. [Google Scholar] [CrossRef]
- Sequist, L.V.; Han, J.-Y.; Ahn, M.-J.; Cho, B.C.; Yu, H.; Kim, S.-W.; Yang, J.C.-H.; Lee, J.S.; Su, W.-C.; Kowalski, D.; et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: Interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef]
- Sun, C.; Wang, L.; Huang, S.; Heynen, G.J.J.E.; Prahallad, A.; Robert, C.; Haanen, J.; Blank, C.; Wesseling, J.; Willems, S.M.; et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature 2014, 508, 118–122. [Google Scholar] [CrossRef]
- Corcoran, R.B.; André, T.; Atreya, C.E.; Schellens, J.H.M.; Yoshino, T.; Bendell, J.C.; Hollebecque, A.; Mcree, A.J.; Siena, S.; Middleton, G.; et al. Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAFV600E-Mutant Colorectal Cancer. Cancer Discov. 2018, 8, 428–443. [Google Scholar] [CrossRef] [Green Version]
- Xue, J.Y.; Zhao, Y.; Aronowitz, J.; Mai, T.T.; Vides, A.; Qeriqi, B.; Kim, D.; Li, C.; De Stanchina, E.; Mazutis, L.; et al. Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature 2020, 577, 421–425. [Google Scholar] [CrossRef] [PubMed]
- Mainardi, S.; Mulero-Sánchez, A.; Prahallad, A.; Germano, G.; Bosma, A.; Krimpenfort, P.; Lieftink, C.; Steinberg, J.D.; De Wit, N.; Gonçalves-Ribeiro, S.; et al. SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo. Nat. Med. 2018, 24, 961–967. [Google Scholar] [CrossRef] [PubMed]
- Ruess, D.A.; Heynen, G.J.; Ciecielski, K.J.; Ai, J.; Berninger, A.; Kabacaoglu, D.; Görgülü, K.; Dantes, Z.; Wörmann, S.M.; Diakopoulos, K.N.; et al. Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase. Nat. Med. 2018, 24, 954–960. [Google Scholar] [CrossRef] [PubMed]
- Wong, G.S.; Zhou, J.; Liu, J.B.; Wu, Z.; Xu, X.; Li, T.; Xu, D.; Schumacher, S.E.; Puschhof, J.; Mcfarland, J.; et al. Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition. Nat. Med. 2018, 24, 968–977. [Google Scholar] [CrossRef]
- Nabhan, A.N.; Brownfield, D.G.; Harbury, P.B.; Krasnow, M.A.; Desai, T.J. Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 2018, 359, 1118–1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Desai, T.J.; Brownfield, D.G.; Krasnow, M.A. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature 2014, 507, 190–194. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Zhu, H.; Liao, Y.; Wu, W.; Liu, L.; Liu, L.; Wu, Y.; Sun, F.; Lin, H.W. Inhibition of Wnt/beta-catenin pathway reverses multi-drug resistance and EMT in Oct4(+)/Nanog(+) NSCLC cells. Biomed. Pharm. 2020, 127, 110225. [Google Scholar] [CrossRef]
- Eberl, M.; Mangelberger, D.; Swanson, J.B.; Verhaegen, M.E.; Harms, P.W.; Frohm, M.L.; Dlugosz, A.A.; Wong, S.Y. Tumor Architecture and Notch Signaling Modulate Drug Response in Basal Cell Carcinoma. Cancer Cell 2018, 33, 229–243.e224. [Google Scholar] [CrossRef]
- Bivona, T.G.; Hieronymus, H.; Parker, J.; Chang, K.; Taron, M.; Rosell, R.; Moonsamy, P.; Dahlman, K.; Miller, V.A.; Costa, C.; et al. FAS and NF-kappaB signalling modulate dependence of lung cancers on mutant EGFR. Nature 2011, 471, 523–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gong, K.; Guo, G.; Panchani, N.; Bender, M.E.; Gerber, D.E.; Minna, J.D.; Fattah, F.; Gao, B.; Peyton, M.; Kernstine, K.; et al. EGFR inhibition triggers an adaptive response by co-opting antiviral signaling pathways in lung cancer. Nat. Cancer 2020, 1, 394–409. [Google Scholar] [CrossRef] [PubMed]
- Hata, A.N.; Engelman, J.A.; Faber, A.C. The BCL2 Family: Key Mediators of the Apoptotic Response to Targeted Anticancer Therapeutics. Cancer Discov. 2015, 5, 475–487. [Google Scholar] [CrossRef] [Green Version]
- Song, K.-A.; Niederst, M.J.; Lochmann, T.L.; Hata, A.N.; Kitai, H.; Ham, J.; Floros, K.V.; Hicks, M.A.; Hu, H.; Mulvey, H.E.; et al. Epithelial-to-Mesenchymal Transition Antagonizes Response to Targeted Therapies in Lung Cancer by Suppressing BIM. Clin. Cancer Res. 2018, 24, 197–208. [Google Scholar] [CrossRef] [Green Version]
- Lue, H.W.; Podolak, J.; Kolahi, K.; Cheng, L.; Rao, S.; Garg, D.; Xue, C.H.; Rantala, J.K.; Tyner, J.W.; Thornburg, K.L.; et al. Metabolic reprogramming ensures cancer cell survival despite oncogenic signaling blockade. Genes Dev. 2017, 31, 2067–2084. [Google Scholar] [CrossRef]
- Viswanathan, V.S.; Ryan, M.J.; Dhruv, H.D.; Gill, S.; Eichhoff, O.M.; Seashore-Ludlow, B.; Kaffenberger, S.D.; Eaton, J.K.; Shimada, K.; Aguirre, A.J.; et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 2017, 547, 453–457. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
- Keeratichamroen, S.; Lirdprapamongkol, K.; Svasti, J. Mechanism of ECM-induced dormancy and chemoresistance in A549 human lung carcinoma cells. Oncol. Rep. 2018, 39, 1765–1774. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Li, Q.; Yamada, T.; Matsumoto, K.; Matsumoto, I.; Oda, M.; Watanabe, G.; Kayano, Y.; Nishioka, Y.; Sone, S.; et al. Crosstalk to Stromal Fibroblasts Induces Resistance of Lung Cancer to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. Clin. Cancer Res. 2009, 15, 6630–6638. [Google Scholar] [CrossRef] [Green Version]
- Mendez-Blanco, C.; Fondevila, F.; Garcia-Palomo, A.; Gonzalez-Gallego, J.; Mauriz, J.L. Sorafenib resistance in hepatocarcinoma: Role of hypoxia-inducible factors. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Russo, M.; Crisafulli, G.; Sogari, A.; Reilly, N.M.; Arena, S.; Lamba, S.; Bartolini, A.; Amodio, V.; Magrì, A.; Novara, L.; et al. Adaptive mutability of colorectal cancers in response to targeted therapies. Science 2019, 366, 1473–1480. [Google Scholar] [CrossRef] [PubMed]
- Bivona, T.G.; Doebele, R.C. A framework for understanding and targeting residual disease in oncogene-driven solid cancers. Nat. Med. 2016, 22, 472–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karrison, T.G.; Ferguson, D.J.; Meier, P. Dormancy of Mammary Carcinoma After Mastectomy. JNCI J. Natl. Cancer Inst. 1999, 91, 80–85. [Google Scholar] [CrossRef]
- Osisami, M.; Keller, E.T. Mechanisms of Metastatic Tumor Dormancy. J. Clin. Med. 2013, 2, 136–150. [Google Scholar] [CrossRef]
- Grassberger, C.; McClatchy, D., 3rd; Geng, C.; Kamran, S.C.; Fintelmann, F.; Maruvka, Y.E.; Piotrowska, Z.; Willers, H.; Sequist, L.V.; Hata, A.N.; et al. Patient-Specific Tumor Growth Trajectories Determine Persistent and Resistant Cancer Cell Populations during Treatment with Targeted Therapies. Cancer Res. 2019, 79, 3776–3788. [Google Scholar] [CrossRef]
- Piotrowska, Z.; Niederst, M.J.; Karlovich, C.A.; Wakelee, H.A.; Neal, J.W.; Mino-Kenudson, M.; Fulton, L.; Hata, A.N.; Lockerman, E.L.; Kalsy, A.; et al. Heterogeneity Underlies the Emergence of EGFRT790 Wild-Type Clones Following Treatment of T790M-Positive Cancers with a Third-Generation EGFR Inhibitor. Cancer Discov. 2015, 5, 713–722. [Google Scholar] [CrossRef] [Green Version]
- Maynard, A.; McCoach, C.E.; Rotow, J.K.; Harris, L.; Haderk, F.; Kerr, D.L.; Yu, E.A.; Schenk, E.L.; Tan, W.; Zee, A.; et al. Therapy-Induced Evolution of Human Lung Cancer Revealed by Single-Cell RNA Sequencing. Cell 2020, 182, 1232–1251. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cabanos, H.F.; Hata, A.N. Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer. Cancers 2021, 13, 2666. https://doi.org/10.3390/cancers13112666
Cabanos HF, Hata AN. Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer. Cancers. 2021; 13(11):2666. https://doi.org/10.3390/cancers13112666
Chicago/Turabian StyleCabanos, Heidie Frisco, and Aaron N. Hata. 2021. "Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer" Cancers 13, no. 11: 2666. https://doi.org/10.3390/cancers13112666