Mechanisms of tyrosine kinase inhibitor resistance in renal cell carcinoma

Patrick L. Sweeney , Yash Suri , Arnab Basu , Vadim S. Koshkin , Arpita Desai

Cancer Drug Resistance ›› 2023, Vol. 6 ›› Issue (4) : 858 -73.

PDF
Cancer Drug Resistance ›› 2023, Vol. 6 ›› Issue (4) :858 -73. DOI: 10.20517/cdr.2023.89
review-article

Mechanisms of tyrosine kinase inhibitor resistance in renal cell carcinoma

Author information +
History +
PDF

Abstract

Renal cell carcinoma (RCC), the most prevalent type of kidney cancer, is a significant cause of cancer morbidity and mortality worldwide. Antiangiogenic tyrosine kinase inhibitors (TKIs), in combination with immune checkpoint inhibitors (ICIs), are among the first-line treatment options for patients with advanced RCC. These therapies target the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase pathway and other kinases crucial to cancer proliferation, survival, and metastasis. TKIs have yielded substantial improvements in progression-free survival (PFS) and overall survival (OS) for patients with advanced RCC. However, nearly all patients eventually progress on these drugs as resistance develops. This review provides an overview of TKI resistance in RCC and explores different mechanisms of resistance, including upregulation of alternative proangiogenic pathways, epithelial-mesenchymal transition (EMT), decreased intracellular drug concentrations due to efflux pumps and lysosomal sequestration, alterations in the tumor microenvironment including bone marrow-derived cells (BMDCs) and tumor-associated fibroblasts (TAFs), and genetic factors such as single nucleotide polymorphisms (SNPs). A comprehensive understanding of these mechanisms opens the door to the development of innovative therapeutic approaches that can effectively overcome TKI resistance, thereby improving outcomes for patients with advanced RCC.

Keywords

Antiangiogenic tyrosine kinase inhibitors / renal cell carcinoma / acquired resistance / sunitinib / tumor microenvironment / immune checkpoint inhibitors

Cite this article

Download citation ▾
Patrick L. Sweeney, Yash Suri, Arnab Basu, Vadim S. Koshkin, Arpita Desai. Mechanisms of tyrosine kinase inhibitor resistance in renal cell carcinoma. Cancer Drug Resistance, 2023, 6(4): 858-73 DOI:10.20517/cdr.2023.89

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Siegel RL,Wagle NS.Cancer statistics, 2023.CA Cancer J Clin2023;73:17-48

[2]

Koul H,Rove KO.Molecular aspects of renal cell carcinoma: a review.Am J Cancer Res2011;1:240-54 PMCID:PMC3180049

[3]

Jin J,Zhang JS.Sunitinib resistance in renal cell carcinoma: from molecular mechanisms to predictive biomarkers.Drug Resist Updat2023;67:100929

[4]

Moch H,Berney DM.The 2022 World Health Organization classification of tumours of the urinary system and male genital organs-part a: renal, penile, and testicular tumours.Eur Urol2022;82:458-68

[5]

Hsieh JJ,Signoretti S.Renal cell carcinoma.Nat Rev Dis Primers2017;3:17009 PMCID:PMC5936048

[6]

Escudier B,Schmidinger M.ESMO Guidelines CommitteeRenal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up.Ann Oncol2016;27:v58-68

[7]

Maher ER,Richard S.von Hippel-Lindau disease: a clinical and scientific review.Eur J Hum Genet2011;19:617-23 PMCID:PMC3110036

[8]

Capitanio U,Bex A.Epidemiology of renal cell carcinoma.Eur Urol2019;75:74-84 PMCID:PMC8397918

[9]

Ljungberg B,Abu-Ghanem Y.European Association of Urology Guidelines on Renal Cell Carcinoma: the 2022 update.Eur Urol2022;82:399-410

[10]

Prins FM,Pronk AA.Renal cell carcinoma: alternative nephron-sparing treatment options for small renal masses, a systematic review.J Endourol2017;31:963-75

[11]

Larroquette M,Domblides C.Adjuvant therapy in renal cell carcinoma: current knowledges and future perspectives.Cancer Treat Rev2021;97:102207

[12]

Rini BI,Figlin RA.The society for immunotherapy of cancer consensus statement on immunotherapy for the treatment of advanced renal cell carcinoma (RCC).J Immunother Cancer2019;7:354 PMCID:PMC6924043

[13]

Barata PC.Treatment of renal cell carcinoma: current status and future directions.CA Cancer J Clin2017;67:507-24

[14]

Liu YF,Wang SY.Immune checkpoint inhibitor-based therapy for advanced clear cell renal cell carcinoma: a narrative review.Int Immunopharmacol2022;110:108900

[15]

Lemmon MA.Cell signaling by receptor tyrosine kinases.Cell2010;141:1117-34 PMCID:PMC2914105

[16]

Du Z.Mechanisms of receptor tyrosine kinase activation in cancer.Mol Cancer2018;17:58 PMCID:PMC5817791

[17]

Mabeta P.The VEGF/VEGFR axis revisited: implications for cancer therapy.Int J Mol Sci2022;23:15585 PMCID:PMC9779738

[18]

Ebrahimi N,Ghaderi H.Receptor tyrosine kinase inhibitors in cancer.Cell Mol Life Sci2023;80:104

[19]

Hartmann JT,Kopp HG.Tyrosine kinase inhibitors - a review on pharmacology, metabolism and side effects.Curr Drug Metab2009;10:470-81

[20]

Sharma R,Myers M,Prithviraj P.Determinants of resistance to VEGF-TKI and immune checkpoint inhibitors in metastatic renal cell carcinoma.J Exp Clin Cancer Res2021;40:186 PMCID:PMC8183071

[21]

Gotink KJ.Anti-angiogenic tyrosine kinase inhibitors: what is their mechanism of action?.Angiogenesis2010;13:1-14 PMCID:PMC2845892

[22]

Rini BI.Resistance to targeted therapy in renal-cell carcinoma.Lancet Oncol2009;10:992-1000

[23]

Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma.Nature2013;499:43-9 PMCID:PMC3771322

[24]

Kim H,Lee SJ,Lee HJ.Loss of von Hippel-Lindau (VHL) tumor suppressor gene function: VHL-HIF pathway and advances in treatments for metastatic renal cell carcinoma (RCC).Int J Mol Sci2021;22:9795 PMCID:PMC8470481

[25]

Meléndez-Rodríguez F,Sanchez-Prieto R.Hypoxia-inducible factor 2-dependent pathways driving von Hippel-Lindau-deficient renal cancer.Front Oncol2018;8:214 PMCID:PMC6002531

[26]

Haider T,Banjare N,Soni V.Drug resistance in cancer: mechanisms and tackling strategies.Pharmacol Rep2020;72:1125-51

[27]

Kelderman S,Haanen JB.Acquired and intrinsic resistance in cancer immunotherapy.Mol Oncol2014;8:1132-9 PMCID:PMC5528612

[28]

Xiang Y,Zhong J,Qin H.Advances in renal cell carcinoma drug resistance models.Front Oncol2022;12:870396 PMCID:PMC9128023

[29]

Ishibashi K,Matsuoka K.Interleukin-6 induces drug resistance in renal cell carcinoma.Fukushima J Med Sci2018;64:103-10 PMCID:PMC6305783

[30]

Rizzo M,Pezzicoli G,Cosmai L.IL-8 and its role as a potential biomarker of resistance to anti-angiogenic agents and immune checkpoint inhibitors in metastatic renal cell carcinoma.Front Oncol2022;12:990568 PMCID:PMC9437355

[31]

Martin D,Gutkind JS.CXCL8/IL8 stimulates vascular endothelial growth factor (VEGF) expression and the autocrine activation of VEGFR2 in endothelial cells by activating NFkappaB through the CBM (Carma3/Bcl10/Malt1) complex.J Biol Chem2009;284:6038-42 PMCID:PMC2649103

[32]

Harmon CS,Figlin RA.Circulating proteins as potential biomarkers of sunitinib and interferon-α efficacy in treatment-naïve patients with metastatic renal cell carcinoma.Cancer Chemother Pharmacol2014;73:151-61 PMCID:PMC3889677

[33]

Tran HT,Zurita AJ.Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials.Lancet Oncol2012;13:827-37

[34]

Akwii RG,Zahra FT.Role of angiopoietin-2 in vascular physiology and pathophysiology.Cells2019;8:471 PMCID:PMC6562915

[35]

He FF,Chen Q.Angiopoietin-Tie signaling in kidney diseases: an updated review.FEBS Lett2019;593:2706-15

[36]

Wang X,Zhang L.The role of angiopoietins as potential therapeutic targets in renal cell carcinoma.Transl Oncol2014;7:188-95 PMCID:PMC4101387

[37]

Deng J,Xia H.A comparison of the prognosis of papillary and clear cell renal cell carcinoma: evidence from a meta-analysis.Medicine2019;98:e16309 PMCID:PMC6635153

[38]

Schöffski P,Escudier B.Crizotinib achieves long-lasting disease control in advanced papillary renal-cell carcinoma type 1 patients with MET mutations or amplification. EORTC 90101 CREATE trial.Eur J Cancer2017;87:147-63

[39]

Marona P,Kotlinowski J,Jura J.C-Met as a key factor responsible for sustaining undifferentiated phenotype and therapy resistance in renal carcinomas.Cells2019;8:272 PMCID:PMC6468372

[40]

Marona P,Kwapisz O.Resistance to tyrosine kinase inhibitors promotes renal cancer progression through MCPIP1 tumor-suppressor downregulation and c-Met activation.Cell Death Dis2022;13:814 PMCID:PMC9500022

[41]

Peltola KJ,Rautiola J.Correlation of c-Met expression and outcome in patients with renal cell carcinoma treated with sunitinib.Clin Genitourin Cancer2017;15:487-94

[42]

Casanovas O,Bergers G.Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors.Cancer Cell2005;8:299-309

[43]

Tsimafeyeu I,Stepanova E,Ta H.Overexpression of fibroblast growth factor receptors FGFR1 and FGFR2 in renal cell carcinoma.Scand J Urol Nephrol2011;45:190-5

[44]

Dessalles CA,Barakat AI.Pericyte mechanics and mechanobiology.J Cell Sci2021;134:jcs240226

[45]

Armulik A,Betsholtz C.Endothelial/pericyte interactions.Circ Res2005;97:512-23

[46]

Cao Z,Zhang G.Tumor cell-mediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis.Biochim Biophys Acta2013;1836:273-86

[47]

Bergers G.Modes of resistance to anti-angiogenic therapy.Nat Rev Cancer2008;8:592-603 PMCID:PMC2874834

[48]

Greenberg JI,Barillas SG.A role for VEGF as a negative regulator of pericyte function and vessel maturation.Nature2008;456:809-13 PMCID:PMC2605188

[49]

Mena AC,Guillén-Ponce C.Understanding the molecular-based mechanism of action of the tyrosine kinase inhibitor: sunitinib.Anticancer Drugs2010;21 Suppl 1:S3-11

[50]

Makhov P,Ghatalia P,Uzzo RG.Resistance to systemic therapies in clear cell renal cell carcinoma: mechanisms and management strategies.Mol Cancer Ther2018;17:1355-64 PMCID:PMC6034114

[51]

Halaby R.Influence of lysosomal sequestration on multidrug resistance in cancer cells.Cancer Drug Resist2019;2:31-42 PMCID:PMC9019175

[52]

Rausch M,Allard PM.Molecular and functional analysis of sunitinib-resistance induction in human renal cell carcinoma cells.Int J Mol Sci2021;22:6467 PMCID:PMC8235637

[53]

Zhitomirsky B.Lysosomal sequestration of hydrophobic weak base chemotherapeutics triggers lysosomal biogenesis and lysosome-dependent cancer multidrug resistance.Oncotarget2015;6:1143-56 PMCID:PMC4359223

[54]

Giuliano S,Dufies M.Resistance to sunitinib in renal clear cell carcinoma results from sequestration in lysosomes and inhibition of the autophagic flux.Autophagy2015;11:1891-904 PMCID:PMC4824581

[55]

de Klerk DJ, Honeywell RJ, Jansen G, Peters GJ. Transporter and lysosomal mediated (Multi)drug resistance to tyrosine kinase inhibitors and potential strategies to overcome resistance.Cancers2018;10:503 PMCID:PMC6315453

[56]

Golle L,Beul K.Bone marrow-derived cells and their conditioned medium induce microvascular repair in uremic rats by stimulation of endogenous repair mechanisms.Sci Rep2017;7:9444 PMCID:PMC5572734

[57]

Mier JW.The tumor microenvironment in renal cell cancer.Curr Opin Oncol2019;31:194-9 PMCID:PMC6467495

[58]

Gabrilovich DI.Myeloid-derived suppressor cells.Cancer Immunol Res2017;5:3-8 PMCID:PMC5426480

[59]

Finke J,Rini B,Ireland J.MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy.Int Immunopharmacol2011;11:856-61 PMCID:PMC3109226

[60]

Susek KH,Alici E.The role of CXC chemokine receptors 1-4 on immune cells in the tumor microenvironment.Front Immunol2018;9:2159 PMCID:PMC6167945

[61]

Elkabets M,Dinarello CA.IL-1β regulates a novel myeloid-derived suppressor cell subset that impairs NK cell development and function.Eur J Immunol2010;40:3347-57

[62]

Ko JS,Rini BI.Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients.Clin Cancer Res2009;15:2148-57

[63]

Kalluri R.The biology and function of fibroblasts in cancer.Nat Rev Cancer2016;16:582-98

[64]

Errarte P,López JI.The role of cancer-associated fibroblasts in renal cell carcinoma. An example of tumor modulation through tumor/non-tumor cell interactions.J Adv Res2020;21:103-8 PMCID:PMC7015466

[65]

Ferrara N.Pathways mediating VEGF-independent tumor angiogenesis.Cytokine Growth Factor Rev2010;21:21-6

[66]

Meads MB,Dalton WS.Environment-mediated drug resistance: a major contributor to minimal residual disease.Nat Rev Cancer2009;9:665-74

[67]

Crawford Y,Yu L.PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment.Cancer Cell2009;15:21-34

[68]

Piva F,Santoni M.Epithelial to mesenchymal transition in renal cell carcinoma: implications for cancer therapy.Mol Diagn Ther2016;20:111-7

[69]

He H.Epithelial-to-mesenchymal transition in renal neoplasms.Adv Anat Pathol2014;21:174-80

[70]

Babaei G,Jaghi NZZ.EMT, cancer stem cells and autophagy; The three main axes of metastasis.Biomed Pharmacother2021;133:110909

[71]

Morel AP,Thomas C,Ansieau S.Generation of breast cancer stem cells through epithelial-mesenchymal transition.PLoS One2008;3:e2888 PMCID:PMC2492808

[72]

Yuan ZX,Zhao G,Fu HL.Targeting strategies for renal cell carcinoma: from renal cancer cells to renal cancer stem cells.Front Pharmacol2016;7:423 PMCID:PMC5103413

[73]

Hammers HJ,Salumbides B.Reversible epithelial to mesenchymal transition and acquired resistance to sunitinib in patients with renal cell carcinoma: evidence from a xenograft study.Mol Cancer Ther2010;9:1525-35

[74]

Amaya GM,Bourgeois DS.Cytochromes P450 1A2 and 3A4 catalyze the metabolic activation of sunitinib.Chem Res Toxicol2018;31:570-84 PMCID:PMC6108078

[75]

Diekstra MH,Lolkema MP.Association analysis of genetic polymorphisms in genes related to sunitinib pharmacokinetics, specifically clearance of sunitinib and SU12662.Clin Pharmacol Ther2014;96:81-9

[76]

Diekstra MH,Swen JJ.Sunitinib-induced hypertension in CYP3A4 rs4646437 A-allele carriers with metastatic renal cell carcinoma.Pharmacogenomics J2017;17:42-6

[77]

van der Veldt AA,Gelderblom H.Genetic polymorphisms associated with a prolonged progression-free survival in patients with metastatic renal cell cancer treated with sunitinib.Clin Cancer Res2011;17:620-9

[78]

Beuselinck B,Lambrechts D.Single-nucleotide polymorphisms associated with outcome in metastatic renal cell carcinoma treated with sunitinib.Br J Cancer2013;108:887-900 PMCID:PMC3590652

[79]

Diekstra MH,Boven E.CYP3A5 and ABCB1 polymorphisms as predictors for sunitinib outcome in metastatic renal cell carcinoma.Eur Urol2015;68:621-9

[80]

Beuselinck B,Van Brussel T.Efflux pump ABCB1 single nucleotide polymorphisms and dose reductions in patients with metastatic renal cell carcinoma treated with sunitinib.Acta Oncol2014;53:1413-22

[81]

Syeda Z, Langden SSS, Munkhzul C, Lee M, Song SJ. Regulatory mechanism of microRNA expression in cancer.Int J Mol Sci2020;21:1723 PMCID:PMC7084905

[82]

Lu L,Wen H.Overexpression of miR-15b promotes resistance to sunitinib in renal cell carcinoma.J Cancer2019;10:3389-96 PMCID:PMC6603409

[83]

Yamaguchi N,Onuma K.Identification of microRNAs involved in resistance to sunitinib in renal cell carcinoma cells.Anticancer Res2017;37:2985-92

[84]

Prior C,Garcia-Donas J.Identification of tissue microRNAs predictive of sunitinib activity in patients with metastatic renal cell carcinoma.PLoS One2014;9:e86263 PMCID:PMC3901669

[85]

Xiao W,Ruan H.Mir-144-3p promotes cell proliferation, metastasis, sunitinib resistance in clear cell renal cell carcinoma by downregulating ARID1A.Cell Physiol Biochem2017;43:2420-33

[86]

Saleeb R,Ding Q.The miR-200 family as prognostic markers in clear cell renal cell carcinoma.Urol Oncol2019;37:955-63

[87]

Duan R,Guo W.EZH2: a novel target for cancer treatment.J Hematol Oncol2020;13:104 PMCID:PMC7385862

[88]

Wang Y,Geng H,Liu Y.Overexpression of YB1 and EZH2 are associated with cancer metastasis and poor prognosis in renal cell carcinomas.Tumour Biol2015;36:7159-66

[89]

Adelaiye-Ogala R,Damayanti NP.EZH2 modifies sunitinib resistance in renal cell carcinoma by kinome reprogramming.Cancer Res2017;77:6651-66 PMCID:PMC5712262

[90]

Qian S,Yang W,Yang Y.The role of BCL-2 family proteins in regulating apoptosis and cancer therapy.Front Oncol2022;12:985363 PMCID:PMC9597512

[91]

Gobé G,Williams G,Buttyan R.Apoptosis and expression of Bcl-2, Bcl-XL, and Bax in renal cell carcinomas.Cancer Invest2002;20:324-32

[92]

Cho H.Targeting HIF2 in clear cell renal cell carcinoma.Cold Spring Harb Symp Quant Biol2016;81:113-21

[93]

Deeks ED.Belzutifan: first approval.Drugs2021;81:1921-7

[94]

Ahmed R.Targeting HIF-2 alpha in renal cell carcinoma.Curr Treat Options Oncol2023;24:1183-98

[95]

Rini BI,Figlin RA.Results from a phase I expansion cohort of the first-in-class oral HIF-2α inhibitor PT2385 in combination with nivolumab in patients with previously treated advanced RCC.JCO2019;37:558

[96]

Choueiri TK.Targeting the HIF2-VEGF axis in renal cell carcinoma.Nat Med2020;26:1519-30

[97]

Huang D,Zhou M.Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma.Cancer Res2010;70:1063-71 PMCID:PMC3719378

[98]

Eroglu Z,Pal SK.Targeting angiopoietin-2 signaling in cancer therapy.Expert Opin Investig Drugs2013;22:813-25

[99]

Semrad TJ,Luo C.Randomized phase 2 study of trebananib (AMG 386) with or without continued anti-vascular endothelial growth factor therapy in patients with renal cell carcinoma who have progressed on bevacizumab, pazopanib, sorafenib, or sunitinib - results of NCI/CTEP protocol 9048.Kidney Cancer2019;3:51-61 PMCID:PMC6400131

[100]

Zhou L,Sun M.Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma.Oncogene2016;35:2687-97 PMCID:PMC4791213

[101]

Welti JC,Powles T.Fibroblast growth factor 2 regulates endothelial cell sensitivity to sunitinib.Oncogene2011;30:1183-93

[102]

Yue S,Chen X.FGFR-TKI resistance in cancer: current status and perspectives.J Hematol Oncol2021;14:23 PMCID:PMC7876795

[103]

Rausch M,Achkhanian J,Nowak-Sliwinska P.Identification of low-dose multidrug combinations for sunitinib-naive and pre-treated renal cell carcinoma.Br J Cancer2020;123:556-67 PMCID:PMC7435198

[104]

Makhov PB,Kutikov A.Modulation of Akt/mTOR signaling overcomes sunitinib resistance in renal and prostate cancer cells.Mol Cancer Ther2012;11:1510-7 PMCID:PMC3491642

[105]

Sekino Y,Liang G.Molecular mechanisms of resistance to tyrosine kinase inhibitor in clear cell renal cell carcinoma.Int J Urol2022;29:1419-28 PMCID:PMC10087189

[106]

Ramesh V,Ceppi P.Targeting EMT in cancer with repurposed metabolic inhibitors.Trends Cancer2020;6:942-50

[107]

Younes A,Patel MR.Safety, tolerability, and preliminary activity of CUDC-907, a first-in-class, oral, dual inhibitor of HDAC and PI3K, in patients with relapsed or refractory lymphoma or multiple myeloma: an open-label, dose-escalation, phase 1 trial.Lancet Oncol2016;17:622-31

[108]

Gotink KJ,Labots M.Lysosomal sequestration of sunitinib: a novel mechanism of drug resistance.Clin Cancer Res2011;17:7337-46 PMCID:PMC4461037

[109]

Mollazadeh S,Hadizadeh F,Arabzadeh S.Structural and functional aspects of P-glycoprotein and its inhibitors.Life Sci2018;214:118-23

[110]

Soto-Vega E,Richaud-Patin Y,Vázquez-Lavista LG.P-glycoprotein activity in renal clear cell carcinoma.Urol Oncol2009;27:363-6

[111]

Liu X.ABC family transporters. In: Liu X, Pan G, editors. Drug transporters in drug disposition, effects and toxicity. Singapore: Springer; 2019. pp. 13-100.

[112]

Callaghan R,Bebawy M.Inhibition of the multidrug resistance P-glycoprotein: time for a change of strategy?.Drug Metab Dispos2014;42:623-31 PMCID:PMC3965902

[113]

Heming C, Muriithi W, Wanjiku Macharia L, Niemeyer Filho P, Moura-Neto V, Aran V. P-glycoprotein and cancer: what do we currently know?.Heliyon2022;8:e11171 PMCID:PMC9618987

[114]

Joosten SC,Aarts MJ.Epigenetics in renal cell cancer: mechanisms and clinical applications.Nat Rev Urol2018;15:430-51

[115]

Tang Y,Gao L.Venetoclax synergizes sunitinib in renal cell carcincoma through inhibition of Bcl-2.Anticancer Agents Med Chem2023;23:2027-34

[116]

Shiravand Y,Kashani SMA.Immune checkpoint inhibitors in cancer therapy.Curr Oncol2022;29:3044-60 PMCID:PMC9139602

[117]

Rassy E,Albiges L.Tyrosine kinase inhibitors and immunotherapy combinations in renal cell carcinoma.Ther Adv Med Oncol2020;12:1758835920907504 PMCID:PMC7081462

AI Summary AI Mindmap
PDF

77

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/