AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (3.1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Exploiting the potential of the ubiquitin-proteasome system in overcoming tyrosine kinase inhibitor resistance in chronic myeloid leukemia

Xudong Lia,b,,1Wei Lic,1Yanli ZhangaLinping Xua( )Yongping Songa,c( )
Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China

Peer review under responsibility of Chongqing Medical University.

1 These authors contributed equally to this work.

Show Author Information

Abstract

The advent of tyrosine kinase inhibitors (TKI) targeting BCR-ABL has drastically changed the treatment approach of chronic myeloid leukemia (CML), greatly prolonged the life of CML patients, and improved their prognosis. However, TKI resistance is still a major problem with CML patients, reducing the efficacy of treatment and their quality of life. TKI resistance is mainly divided into BCR-ABL-dependent and BCR-ABL-independent resistance. Now, the main clinical strategy addressing TKI resistance is to switch to newly developed TKIs. However, data have shown that these new drugs may cause serious adverse reactions and intolerance and cannot address all resistance mutations. Therefore, finding new therapeutic targets to overcome TKI resistance is crucial and the ubiquitin-proteasome system (UPS) has emerged as a focus. The UPS mediates the degradation of most proteins in organisms and controls a wide range of physiological processes. In recent years, the study of UPS in hematological malignant tumors has resulted in effective treatments, such as bortezomib in the treatment of multiple myeloma and mantle cell lymphoma. In CML, the components of UPS cooperate or antagonize the efficacy of TKI by directly or indirectly affecting the ubiquitination of BCR-ABL, interfering with CML-related signaling pathways, and negatively or positively affecting leukemia stem cells. Some of these molecules may help overcome TKI resistance and treat CML. In this review, the mechanism of TKI resistance is briefly described, the components of UPS are introduced, existing studies on UPS participating in TKI resistance are listed, and UPS as the therapeutic target and strategies are discussed.

References

1

Kalidas M, Kantarjian H, Talpaz M. Chronic myelogenous leukemia. JAMA. 2001;286(8):895-898.

2

Sawyers CL. Chronic myeloid leukemia. N Engl J Med. 1999;340(17):1330-1340.

3

Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344(14):1031-1037.

4

Lin Q, Mao L, Shao L, et al. Global, regional, and national burden of chronic myeloid leukemia, 1990-2017: a systematic analysis for the global burden of disease study 2017. Front Oncol. 2020;10:580759.

5

Greuber EK, Smith-Pearson P, Wang J, Pendergast AM. Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nat Rev Cancer. 2013;13(8):559-571.

6

Deininger MW, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood. 2000;96(10):3343-3356.

7

Berman E. How I treat chronic-phase chronic myelogenous leukemia. Blood. 2022;139(21):3138-3147.

8

Yeung DT, Shanmuganathan N, Hughes TP. Asciminib: a new therapeutic option in chronic-phase CML with treatment failure. Blood. 2022;139(24):3474-3479.

9

Braun TP, Eide CA, Druker BJ. Response and resistance to BCR-ABL1-targeted therapies. Cancer Cell. 2020;37(4):530-542.

10

Drula R, Iluta S, Gulei, et al. Exploiting the ubiquitin system in myeloid malignancies. From basic research to drug discovery in MDS and AML. Blood Rev. 2022;56:100971.

11

Obeng EA, Carlson LM, Gutman DM, Harrington WJ, Lee KP, Boise LH. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood. 2006;107(12):4907-4916.

12

Reff MJ, Shillingburg A, Shah B, Elder C, Prescott H, Kennerly-Shah J. Front-line use of tyrosine kinase inhibitors in chronic phase chronic myeloid leukemia: practice considerations. J Oncol Pharm Pract. 2020;26(1):156-174.

13

Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol. 2018;93(3):442-459.

14

Kantarjian HM, Shah NP, Cortes JE, et al. Dasatinib or imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: 2-year follow-up from a randomized phase 3 trial (DASISION). Blood. 2012;119(5):1123-1129.

15

Osman AEG, Deininger MW. Chronic myeloid leukemia: modern therapies, current challenges, and future directions. Blood Rev. 2021;49:100825.

16

Meenakshi Sundaram DN, Jiang X, Brandwein JM, Valencia-Serna J, Remant KC, Uludağ H. Current outlook on drug resistance in chronic myeloid leukemia (CML) and potential therapeutic options. Drug Discov Today. 2019;24(7):1355-1369.

17

Dhillon S. Olverembatinib: first approval. Drugs. 2022;82(4):469-475.

18

Qian J, Shi D, Li Z, et al. Updated safety and efficacy results of phase 1 study of olverembatinib (HQP1351), a novel third-generation BCR-ABL tyrosine kinase inhibitor (TKI), in patients with TKI-resistant chronic myeloid leukemia (CML). Blood. 2021;138(suppl 1):311-313.

19

Qian J, Shi D, Li Z, et al. Updated results of pivotal phase 2 trials of olverembatinib (HQP1351) in patients (pts) with tyrosine kinase inhibitor (TKI)-resistant BCR-ABL1T315I-mutated chronic- and accelerated-phase chronic myeloid leukemia (CML-CP and CML-AP). Blood. 2021;138:3598.

20

Chen J, Wang F, Fang J, et al. Dynamic evolution of ponatinib-resistant mutations in BCR-ABL1-positive leukaemias revealed by next-generation sequencing. Br J Haematol. 2020;191(5):e113-e116.

21

Tang C, Schafranek L, Watkins DB, et al. Tyrosine kinase inhibitor resistance in chronic myeloid leukemia cell lines: investigating resistance pathways. Leuk Lymphoma. 2011;52(11):2139-2147.

22

Stetka J, Gursky J, Liñan Velasquez J, et al. Role of DNA damage response in suppressing malignant progression of chronic myeloid leukemia and polycythemia vera: impact of different oncogenes. Cancers (Basel). 2020;12(4):E903.

23

Popp HD, Kohl V, Naumann N, et al. DNA damage and DNA damage response in chronic myeloid leukemia. Int J Mol Sci. 2020;21(4):1177.

24

Xue M, Zeng Z, Wang Q, et al. Mutational profiles during the progression of chronic myeloid leukemia. Blood. 2021;138(suppl 1):3596.

25

Ma D, Liu P, Wang P, Zhou Z, Fang Q, Wang J. PKC-β/Alox5 axis activation promotes Bcr-Abl-independent TKI-resistance in chronic myeloid leukemia. J Cell Physiol. 2021;236(9):6312-6327.

26

Alves R, Gonçalves AC, Rutella S, et al. Resistance to tyrosine kinase inhibitors in chronic myeloid leukemia - from molecular mechanisms to clinical relevance. Cancers (Basel). 2021;13(19):4820.

27

Wagle M, Eiring AM, Wongchenko M, et al. A role for FOXO1 in BCR-ABL1-independent tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Leukemia. 2016;30(7):1493-1501.

28

Sweet K, Atallah E, Radich J, et al. Second treatment free remission after combination therapy with ruxolitinib plus tyrosine kinase inhibitors in chronic phase chronic myeloid leukemia (CML). Blood. 2021;138(suppl 1):2555.

29

Hamilton A, Helgason GV, Schemionek M, et al. Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival. Blood. 2012;119(6):1501-1510.

30

Zhao C, Blum J, Chen A, et al. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007;12(6):528-541.

31

Arrigoni E, Del Re M, Galimberti S, et al. Concise review: chronic myeloid leukemia: stem cell niche and response to pharmacologic treatment. Stem Cells Transl Med. 2018;7(3):305-314.

32

Naka K, Hoshii T, Muraguchi T, et al. TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature. 2010;463(7281):676-680.

33

Li L, Wang L, Li L, et al. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell. 2012;21(2):266-281.

34

Abraham SA, Hopcroft LE, Carrick E, et al. Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells. Nature. 2016;534(7607):341-346.

35

Liu C, Zou W, Nie D, et al. Loss of PRMT7 reprograms glycine metabolism to selectively eradicate leukemia stem cells in CML. Cell Metab. 2022;34(6):818-835.e7.

36

Jin B, Wang C, Li J, et al. Anthelmintic niclosamide disrupts the interplay of p65 and FOXM1/β-catenin and eradicates leukemia stem cells in chronic myelogenous leukemia. Clin Cancer Res. 2017;23:789-803.

37

Naka K, Jomen Y, Ishihara K, et al. Dipeptide species regulate p38MAPK-Smad3 signalling to maintain chronic myelogenous leukaemia stem cells. Nat Commun. 2015;6:8039.

38

Mohd M, Ye H, Winters Amanda C, et al. Lysosomal acid lipase a (LIPA) modulates leukemia stem cell (LSC) response to venetoclax/TKI combination therapy in blast phase chronic myeloid leukemia. Blood. 2021;138(S1):630.

39

Nair RR, Tolentino J, Hazlehurst LA. The bone marrow microenvironment as a sanctuary for minimal residual disease in CML. Biochem Pharmacol. 2010;80(5):602-612.

40

Agarwal P, Li H, Choi K, et al. TNF-α-induced alterations in stromal progenitors enhance leukemic stem cell growth via CXCR2 signaling. Cell Rep. 2021;36(2):109386.

41

Senft D, Qi J, Ronai ZA. Ubiquitin ligases in oncogenic transformation and cancer therapy. Nat Rev Cancer. 2018;18(2):69-88.

42

Yau R, Rape M. The increasing complexity of the ubiquitin code. Nat Cell Biol. 2016;18(6):579-586.

43

Mukhopadhyay D, Riezman H. Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science. 2007;315(5809):201-205.

44

Komander D, Rape M. The ubiquitin code. Annu Rev Biochem. 2012;81:203-229.

45

Behrends C, Harper JW. Constructing and decoding unconventional ubiquitin chains. Nat Struct Mol Biol. 2011;18(5):520-528.

46

Schulman BA, Harper JW. Ubiquitin-like protein activation by E1 enzymes: the apex for downstream signalling pathways. Nat Rev Mol Cell Biol. 2009;10(5):319-331.

47

Dikic I, Wakatsuki S, Walters KJ. Ubiquitin-binding domains - from structures to functions. Nat Rev Mol Cell Biol. 2009;10(10):659-671.

48

Hormaechea-Agulla D, Kim Y, Song MS, Song SJ. New insights into the role of E2s in the pathogenesis of diseases: lessons learned from UBE2O. Mol Cells. 2018;41(3):168-178.

49

Ye Y, Rape M. Building ubiquitin chains: E2 enzymes at work. Nat Rev Mol Cell Biol. 2009;10(11):755-764.

50

van Wijk SJ, Timmers HT. The family of ubiquitin-conjugating enzymes (E2s): deciding between life and death of proteins. FASEB J. 2010;24(4):981-993.

51

Zheng N, Shabek N. Ubiquitin ligases: structure, function, and regulation. Annu Rev Biochem. 2017;86:129-157.

52

Spratt DE, Walden H, Shaw GS. RBR E3 ubiquitin ligases: new structures, new insights, new questions. Biochem J. 2014;458(3):421-437.

53

Pla-Prats C, Thomä NH. Quality control of protein complex assembly by the ubiquitin-proteasome system. Trends Cell Biol. 2022;32(8):696-706.

54

Lange SM, Armstrong LA, Kulathu Y. Deubiquitinases: from mechanisms to their inhibition by small molecules. Mol Cell. 2022;82(1):15-29.

55

van der Veen AG, Ploegh HL. Ubiquitin-like proteins. Annu Rev Biochem. 2012;81:323-357.

56

Hwang JT, Lee A, Kho C. Ubiquitin and ubiquitin-like proteins in cancer, neurodegenerative disorders, and heart diseases. Int J Mol Sci. 2022;23(9):5053.

57

Cappadocia L, Lima CD. Ubiquitin-like protein conjugation: structures, chemistry, and mechanism. Chem Rev. 2018;118(3):889-918.

58

Swatek KN, Komander D. Ubiquitin modifications. Cell Res. 2016;26(4):399-422.

59

Harrigan JA, Jacq X, Martin NM, Jackson SP. Deubiquitylating enzymes and drug discovery: emerging opportunities. Nat Rev Drug Discov. 2018;17(1):57-78.

60

Bhattacharjee S, Nandi S. DNA damage response and cancer therapeutics through the lens of the Fanconi Anemia DNA repair pathway. Cell Commun Signal. 2017;15(1):41.

61

Martín-Vicente M, Medrano LM, Resino S, García-Sastre A, Martínez I. TRIM25 in the regulation of the antiviral innate immunity. Front Immunol. 2017;8:1187.

62

Kattah MG, Malynn BA, Ma A. Ubiquitin-modifying enzymes and regulation of the inflammasome. J Mol Biol. 2017;429(22):3471-3485.

63

do Patrocinio AB, Rodrigues V, Guidi Magalhães L. P53: stability from the ubiquitin-proteasome system and specific 26S proteasome inhibitors. ACS Omega. 2022;7(5):3836-3843.

64

Inui N, Sakai S, Kitagawa M. Molecular pathogenesis of pulmonary fibrosis, with focus on pathways related to TGF-β and the ubiquitin-proteasome pathway. Int J Mol Sci. 2021;22(11):6107.

65

Liu J, Chen H, Kaniskan HÜ, et al. TF-PROTACs enable targeted degradation of transcription factors. J Am Chem Soc. 2021;143(23):8902-8910.

66

Li X, Pu W, Zheng Q, Ai M, Chen S, Peng Y. Proteolysis-targeting chimeras (PROTACs) in cancer therapy. Mol Cancer. 2022;21(1):99.

67

Huang X, Dixit VM. Drugging the undruggables: exploring the ubiquitin system for drug development. Cell Res. 2016;26(4):484-498.

68

Manasanch EE, Orlowski RZ. Proteasome inhibitors in cancer therapy. Nat Rev Clin Oncol. 2017;14(7):417-433.

69

Soucy TA, Smith PG, Milhollen MA, et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature. 2009;458(7239):732-736.

70

Bjorklund CC, Kang J, Amatangelo M, et al. Iberdomide (CC-220) is a potent cereblon E3 ligase modulator with antitumor and immunostimulatory activities in lenalidomide- and pomalidomide-resistant multiple myeloma cells with dysregulated CRBN. Leukemia. 2020;34(4):1197-1201.

71

Shah J, Usmani S, Stadtmauer EA, et al. Oprozomib, pomalidomide, and dexamethasone in patients with relapsed and/or refractory multiple myeloma. Clin Lymphoma Myeloma Leuk. 2019;19(9):570-578.e1.

72

Vogl DT, Martin TG, Vij R, et al. Phase Ⅰ/Ⅱ study of the novel proteasome inhibitor delanzomib (CEP-18770) for relapsed and refractory multiple myeloma. Leuk Lymphoma. 2017;58(8):1872-1879.

73

Hnit SS, Xie C, Yao M, et al. p27(Kip1) signaling: transcriptional and post-translational regulation. Int J Biochem Cell Biol. 2015;68:9-14.

74

Wu T, Gu X, Cui H. Emerging roles of SKP2 in cancer drug resistance. Cells. 2021;10(5):1147.

75

Huang T, Yang L, Wang G, et al. Inhibition of Skp2 sensitizes lung cancer cells to paclitaxel. Onco Targets Ther. 2017;10:439-446.

76

Koga H, Harada M, Ohtsubo M, et al. Troglitazone induces p27Kip1-associated cell-cycle arrest through down-regulating Skp2 in human hepatoma cells. Hepatology. 2003;37(5):1086-1096.

77

Masuda TA, Inoue H, Sonoda H, et al. Clinical and biological significance of S-phase kinase-associated protein 2 (Skp2) gene expression in gastric carcinoma: modulation of malignant phenotype by Skp2 overexpression, possibly via p27 proteolysis. Cancer Res. 2002;62(13):3819-3825.

78

Agarwal A, Bumm TGP, Corbin AS, et al. Absence of SKP2 expression attenuates BCR-ABL-induced myeloproliferative disease. Blood. 2008;112(5):1960-1970.

79

Chen JY, Wang MC, Hung WC. Bcr-Abl-induced tyrosine phosphorylation of Emi1 to stabilize Skp2 protein via inhibition of ubiquitination in chronic myeloid leukemia cells. J Cell Physiol. 2011;226(2):407-413.

80

Bretones G, Acosta JC, Caraballo JM, et al. SKP2 oncogene is a direct MYC target gene and MYC down-regulates p27KIP1 through SKP2 in human leukemia cells. J Biol Chem. 2011;286(11):9815-9825.

81

Gómez-Casares MT, García-Alegria E, López-Jorge CE, et al. MYC antagonizes the differentiation induced by imatinib in chronic myeloid leukemia cells through downregulation of p27(KIP1.). Oncogene. 2013;32(17):2239-2246.

82

Tomiatti V, Istvánffy R, Pietschmann E, et al. Cks1 is a critical regulator of hematopoietic stem cell quiescence and cycling, operating upstream of Cdk inhibitors. Oncogene. 2015;34(33):4347-4357.

83

Iskandarani A, Bhat AA, Siveen KS, et al. Bortezomib-mediated downregulation of S-phase kinase protein-2 (SKP2) causes apoptotic cell death in chronic myelogenous leukemia cells. J Transl Med. 2016;14:69.

84

Chen X, Huang Z, Wu W, Xia R. Inhibition of Skp2 sensitizes chronic myeloid leukemia cells to imatinib. Cancer Manag Res. 2020;12:4777-4787.

85

Sargin B, Choudhary C, Crosetto N, et al. Flt3-dependent transformation by inactivating c-Cbl mutations in AML. Blood. 2007;110(3):1004-1012.

86

Grand FH, Hidalgo-Curtis CE, Ernst T, et al. Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. Blood. 2009;113(24):6182-6192.

87

Caligiuri MA, Briesewitz R, Yu J, et al. Novel c-CBL and CBL-b ubiquitin ligase mutations in human acute myeloid leukemia. Blood. 2007;110(3):1022-1024.

88

Dunbar AJ, Gondek LP, O’Keefe CL, et al. 250K single nucleotide polymorphism array karyotyping identifies acquired uniparental disomy and homozygous mutations, including novel missense substitutions of c-Cbl, in myeloid malignancies. Cancer Res. 2008;68(24):10349-10357.

89

Sanada M, Suzuki T, Shih LY, et al. Gain-of-function of mutated C-CBL tumour suppressor in myeloid neoplasms. Nature. 2009;460(7257):904-908.

90

Papaemmanuil E, Gerstung M, Malcovati L, et al. O-001 Clinical and biological implications of gene mutations in MDS. Leuk Res. 2013;37:S9.

91

Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241-247.

92

Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209-2221.

93

Kohlmann A, Grossmann V, Klein HU, et al. Next-generation sequencing technology reveals a characteristic pattern of molecular mutations in 72.8% of chronic myelomonocytic leukemia by detecting frequent alterations in TET2, CBL, RAS, and RUNX1. J Clin Oncol. 2010;28(24):3858-3865.

94

Loh ML, Sakai DS, Flotho C, et al. Mutations in CBL occur frequently in juvenile myelomonocytic leukemia. Blood. 2009;114(9):1859-1863.

95

Stieglitz E, Taylor-Weiner AN, Chang TY, et al. The genomic landscape of juvenile myelomonocytic leukemia. Nat Genet. 2015;47(11):1326-1333.

96

Toffalini F, Kallin A, Vandenberghe P, et al. The fusion proteins TEL-PDGFRβ and FIP1L1-PDGFRα escape ubiquitination and degradation. Haematologica. 2009;94(8):1085-1093.

97

Hu J, Liu YF, Wu CF, et al. Long-term efficacy and safety of all-trans retinoic acid/arsenic trioxide-based therapy in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci U S A. 2009;106(9):3342-3347.

98

Yin T, Wu YL, Sun HP, et al. Combined effects of As4S4 and imatinib on chronic myeloid leukemia cells and BCR-ABL oncoprotein. Blood. 2004;104(13):4219-4225.

99

Mao JH, Sun XY, Liu JX, et al. As4S4 targets RING-type E3 ligase c-CBL to induce degradation of BCR-ABL in chronic myelogenous leukemia. Proc Natl Acad Sci U S A. 2010;107(50):21683-21688.

100

Nakata Y, Ueda T, Nagamachi A, et al. Acquired expression of CblQ367P in mice induces dysplastic myelopoiesis mimicking chronic myelomonocytic leukemia. Blood. 2017;129(15):2148-2160.

101

Belizaire R, Koochaki SHJ, Udeshi ND, et al. CBL mutations drive PI3K/AKT signaling via increased interaction with LYN and PIK3R1. Blood. 2021;137(16):2209-2220.

102

Gioia R, Leroy C, Drullion C, et al. Quantitative phosphoproteomics revealed interplay between Syk and Lyn in the resistance to nilotinib in chronic myeloid leukemia cells. Blood. 2011;118(8):2211-2221.

103

Gioia R, Trégoat C, Dumas PY, et al. CBL controls a tyrosine kinase network involving AXL, SYK and LYN in nilotinib-resistant chronic myeloid leukaemia. J Pathol. 2015;237(1):14-24.

104

Lv K, Jiang J, Donaghy R, et al. CBL family E3 ubiquitin ligases control JAK2 ubiquitination and stability in hematopoietic stem cells and myeloid malignancies. Genes Dev. 2017;31(10):1007-1023.

105

Kourtis N, Moubarak RS, Aranda-Orgilles B, et al. FBXW7 modulates cellular stress response and metastatic potential through HSF1 post-translational modification. Nat Cell Biol. 2015;17(3):322-332.

106

Ye M, Zhang Y, Zhang X, et al. Targeting FBW7 as a strategy to overcome resistance to targeted therapy in non-small cell lung cancer. Cancer Res. 2017;77(13):3527-3539.

107

Close V, Close W, Kugler SJ, et al. FBXW7 mutations reduce binding of NOTCH1, leading to cleaved NOTCH1 accumulation and target gene activation in CLL. Blood. 2019;133(8):830-839.

108

Cui D, Xiong X, Shu J, Dai X, Sun Y, Zhao Y. FBXW7 confers radiation survival by targeting p53 for degradation. Cell Rep. 2020;30(2):497-509.e4.

109

Matsuoka S, Oike Y, Onoyama I, et al. Fbxw7 acts as a critical fail-safe against premature loss of hematopoietic stem cells and development of T-ALL. Genes Dev. 2008;22(8):986-991.

110

Thompson BJ, Jankovic V, Gao J, et al. Control of hematopoietic stem cell quiescence by the E3 ubiquitin ligase Fbw7. J Exp Med. 2008;205(6):1395-1408.

111

Laurenti E, Wilson A, Trumpp A. Myc’s other life: stem cells and beyond. Curr Opin Cell Biol. 2009;21(6):844-854.

112

Reavie L, Della Gatta G, Crusio K, et al. Regulation of hematopoietic stem cell differentiation by a single ubiquitin ligase-substrate complex. Nat Immunol. 2010;11(3):207-215.

113

Reavie L, Buckley SM, Loizou E, et al. Regulation of c-Myc ubiquitination controls chronic myelogenous leukemia initiation and progression. Cancer Cell. 2013;23(3):362-375.

114

Takeishi S, Matsumoto A, Onoyama I, Naka K, Hirao A, Nakayama KI. Ablation of Fbxw7 eliminates leukemia-initiating cells by preventing quiescence. Cancer Cell. 2013;23(3):347-361.

115

Ballinger CA, Connell P, Wu Y, et al. Identification of CHIP, a novel tetratricopeptide repeat-containing protein that interacts with heat shock proteins and negatively regulates chaperone functions. Mol Cell Biol. 1999;19(6):4535-4545.

116

McDonough H, Patterson C. CHIP: a link between the chaperone and proteasome systems. Cell Stress Chaperones. 2003;8(4):303-308.

117

Murata S, Chiba T, Tanaka K. CHIP: a quality-control E3 ligase collaborating with molecular chaperones. Int J Biochem Cell Biol. 2003;35(5):572-578.

118

Tsukahara F, Maru Y. Bag1 directly routes immature BCR-ABL for proteasomal degradation. Blood. 2010;116(18):3582-3592.

119

Lei H, Jin J, Liu M, et al. Chk1 inhibitors overcome imatinib resistance in chronic myeloid leukemia cells. Leuk Res. 2018;64:17-23.

120

Ru Y, Wang Q, Liu X, et al. The chimeric ubiquitin ligase SH2-U-box inhibits the growth of imatinib-sensitive and resistant CML by targeting the native and T315I-mutant BCR-ABL. Sci Rep. 2016;6:28352.

121

Li K, Li J, Ye M, Jin X. The role of Siah2 in tumorigenesis and cancer therapy. Gene. 2021:146028.

122

Huang J, Lu Z, Xiao Y, et al. Inhibition of Siah2 ubiquitin ligase by vitamin K3 attenuates chronic myeloid leukemia chemo-resistance in hypoxic microenvironment. Med Sci Monit. 2018;24:727-735.

123

Park CS, Lewis AH, Chen TJ, et al. A KLF4-DYRK2-mediated pathway regulating self-renewal in CML stem cells. Blood. 2019;134(22):1960-1972.

124

Deng T, Hu B, Wang X, et al. TRAF6 autophagic degradation by avibirnavirus VP3 inhibits antiviral innate immunity via blocking NFKB/NF-κB activation. Autophagy. 2022;18(12):2781-2798.

125

Zhang JL, Du BB, Zhang DH, et al. OTUB1 alleviates NASH through inhibition of the TRAF6-ASK1 signaling pathways. Hepatology. 2022;75(5):1218-1234.

126

Liu J, Li S, Fei X, et al. Increased alveolar epithelial TRAF6 via autophagy-dependent TRIM37 degradation mediates particulate matter-induced lung metastasis. Autophagy. 2022;18(5):971-989.

127

Muto T, Guillamot M, Yeung J, et al. TRAF6 functions as a tumor suppressor in myeloid malignancies by directly targeting MYC oncogenic activity. Cell Stem Cell. 2022;29(2):298-314.e9.

128

Han SH, Korm S, Han YG, et al. GCA links TRAF6-ULK1-dependent autophagy activation in resistant chronic myeloid leukemia. Autophagy. 2019;15(12):2076-2090.

129

Ali MS, Panuzzo C, Calabrese C, et al. The giant HECT E3 ubiquitin ligase HERC1 is aberrantly expressed in myeloid related disorders and it is a novel BCR-ABL1 binding partner. Cancers (Basel). 2021;13(2):341.

130

Ali MS, Magnati S, Panuzzo C, et al. The downregulation of both giant HERCs, HERC1 and HERC2, is an unambiguous feature of chronic myeloid leukemia, and HERC1 levels are associated with leukemic cell differentiation. J Clin Med. 2022;11(2):324.

131

Bigenzahn JW, Collu GM, Kartnig F, et al. LZTR1 is a regulator of RAS ubiquitination and signaling. Science. 2018;362(6419):1171-1177.

132

Kim SY, Kim HJ, Kim HJ, Kim CH. Non-thermal plasma induces antileukemic effect through mTOR ubiquitination. Cells. 2020;9(3):E595.

133

Yin Z, Huang G, Gu C, Liu Y, Yang J, Fei J. Discovery of berberine that targetedly induces autophagic degradation of both BCR-ABL and BCR-ABL T315I through recruiting LRSAM1 for overcoming imatinib resistance. Clin Cancer Res. 2020;26(15):4040-4053.

134

Paul T, Roy R, Sarkar RD, Sinha S, Biswas N. H2O2 mediated FLIP and XIAP down-regulation involves increased ITCH expression and ERK-Akt crosstalk in imatinib resistant Chronic Myeloid Leukemia cell line K562. Free Radic Biol Med. 2021;166:265-276.

135

Qiu L, Wang J, Chen M, Chen F, Tu W. Exosomal microRNA-146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2-mediated PI3K/Akt axis. Int J Mol Med. 2020;46(2):609-620.

136

Chen X, Chen Y, Zhang M, et al. HucMSC exosomes promoted imatinib-induced apoptosis in K562-R cells via a miR-145a-5p/USP6/GLS1 axis. Cell Death Dis. 2022;13(1):92.

137

Liu M, Zhang Y, Wu Y, et al. IKZF1 selectively enhances homologous recombination repair by interacting with CtIP and USP7 in multiple myeloma. Int J Biol Sci. 2022;18(6):2515-2526.

138

Maat H, Atsma TJ, Hogeling SM, et al. The USP7-TRIM27 axis mediates non-canonical PRC1.1 function and is a druggable target in leukemia. iScience. 2021;24(5):102435.

139

Morotti A, Panuzzo C, Crivellaro S, et al. BCR-ABL disrupts PTEN nuclear-cytoplasmic shuttling through phosphorylation-dependent activation of HAUSP. Leukemia. 2014;28(6):1326-1333.

140

Jiang S, Wang X, He Y, et al. Suppression of USP7 induces BCR-ABL degradation and chronic myelogenous leukemia cell apoptosis. Cell Death Dis. 2021;12(5):456.

141

Jie X, Fong WP, Zhou R, et al. USP9X-mediated KDM4C deubiquitination promotes lung cancer radioresistance by epigenetically inducing TGF-β2 transcription. Cell Death Differ. 2021;28(7):2095-2111.

142

Guan T, Yang X, Liang H, et al. Deubiquitinating enzyme USP9X regulates metastasis and chemoresistance in triple-negative breast cancer by stabilizing Snail1. J Cell Physiol. 2022;237(7):2992-3000.

143

Schwickart M, Huang X, Lill JR, et al. Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature. 2010;463(7277):103-107.

144

Sun H, Kapuria V, Peterson LF, et al. Bcr-Abl ubiquitination and Usp9x inhibition block kinase signaling and promote CML cell apoptosis. Blood. 2011;117(11):3151-3162.

145

Draker R, Sarcinella E, Cheung P. USP10 deubiquitylates the histone variant H2A.Z and both are required for androgen receptor-mediated gene activation. Nucleic Acids Res. 2011;39(9):3529-3542.

146

Faus H, Meyer HA, Huber M, Bahr I, Haendler B. The ubiquitin-specific protease USP10 modulates androgen receptor function. Mol Cell Endocrinol. 2005;245(1–2):138-146.

147

Yuan J, Luo K, Zhang L, Cheville JC, Lou Z. USP10 regulates p53 localization and stability by deubiquitinating p53. Cell. 2010;140(3):384-396.

148

Lin Z, Yang H, Tan C, et al. USP10 antagonizes c-Myc transcriptional activation through SIRT6 stabilization to suppress tumor formation. Cell Rep. 2013;5(6):1639-1649.

149

Deng M, Yang X, Qin B, et al. Deubiquitination and activation of AMPK by USP10. Mol Cell. 2016;61(4):614-624.

150

Weisberg EL, Schauer NJ, Yang J, et al. Inhibition of USP10 induces degradation of oncogenic FLT3. Nat Chem Biol. 2017;13(12):1207-1215.

151

Liao Y, Liu N, Xia X, et al. USP10 modulates the SKP2/Bcr-Abl axis via stabilizing SKP2 in chronic myeloid leukemia. Cell Discov. 2019;5:24.

152

Zhao C, Chen X, Zang D, et al. A novel nickel complex works as a proteasomal deubiquitinase inhibitor for cancer therapy. Oncogene. 2016;35:5916-5927.

153

Lan X, Zhao C, Chen X, et al. Nickel pyrithione induces apoptosis in chronic myeloid leukemia cells resistant to imatinib via both Bcr/Abl-dependent and Bcr/Abl-independent mechanisms. J Hematol Oncol. 2016;9(1):129.

154

Jiang L, He Q, Chen X, et al. Inhibition of proteasomal deubiquitinases USP14 and UCHL5 overcomes tyrosine kinase inhibitor resistance in chronic myeloid leukaemia. Clin Transl Med. 2022;12(9):e1038.

155

Lan X, Hu M, Jiang L, et al. Piperlongumine overcomes imatinib resistance by inducing proteasome inhibition in chronic myelogenous leukemia cells. J Ethnopharmacol. 2023;301:115815.

156

Steelman LS, Pohnert SC, Shelton JG, Franklin RA, Bertrand FE, McCubrey JA. JAK/STAT, Raf/MEK/ERK, PI3K/Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia. 2004;18(2):189-218.

157

Basham B, Sathe M, Grein J, et al. In vivo identification of novel STAT5 target genes. Nucleic Acids Res. 2008;36(11):3802-3818.

158

Turhan AG. STAT5 as a CML target: STATinib therapies? Blood. 2011;117(12):3252-3253.

159

Haetscher N, Feuermann Y, Wingert S, et al. STAT5-regulated microRNA-193b controls haematopoietic stem and progenitor cell expansion by modulating cytokine receptor signalling. Nat Commun. 2015;6:8928.

160

Nie ZY, Yao M, Yang Z, et al. De-regulated STAT5A/miR-202-5p/USP15/caspase-6 regulatory axis suppresses CML cell apoptosis and contributes to imatinib resistance. J Exp Clin Cancer Res. 2020;39(1):17.

161

Shibata N, Ohoka N, Tsuji G, et al. Deubiquitylase USP25 prevents degradation of BCR-ABL protein and ensures proliferation of Ph-positive leukemia cells. Oncogene. 2020;39(19):3867-3878.

162

Yan S, Yue Y, Wang J, et al. LINC00668 promotes tumorigenesis and progression through sponging miR-188-5p and regulating USP47 in colorectal cancer. Eur J Pharmacol. 2019;858:172464.

163

Bufalieri F, Infante P, Bernardi F, et al. ERAP1 promotes Hedgehog-dependent tumorigenesis by controlling USP47-mediated degradation of βTrCP. Nat Commun. 2019;10(1):3304.

164

Yu L, Dong L, Wang Y, et al. Reversible regulation of SATB1 ubiquitination by USP47 and SMURF2 mediates colon cancer cell proliferation and tumor progression. Cancer Lett. 2019;448:40-51.

165

Lei H, Xu HZ, Shan HZ, et al. Targeting USP47 overcomes tyrosine kinase inhibitor resistance and eradicates leukemia stem/progenitor cells in chronic myelogenous leukemia. Nat Commun. 2021;12(1):51.

166

Yang Y, Kitagaki J, Dai RM, et al. Inhibitors of ubiquitin-activating enzyme (E1), a new class of potential cancer therapeutics. Cancer Res. 2007;67(19):9472-9481.

167

Ungermannova D, Parker SJ, Nasveschuk CG, et al. Identification and mechanistic studies of a novel ubiquitin E1 inhibitor. J Biomol Screen. 2012;17(4):421-434.

168

Liu C, Nie D, Li J, et al. Antitumor effects of blocking protein neddylation in T315I-BCR-ABL leukemia cells and leukemia stem cells. Cancer Res. 2018;78(6):1522-1536.

169

Yang S, Zhu XN, Zhang HL, et al. ANP32B-mediated repression of p53 contributes to maintenance of normal and CML stem cells. Blood. 2021;138(24):2485-2498.

170

Magistroni V, Mauri M, D’Aliberti D, et al. De novo UBE2A mutations are recurrently acquired during chronic myeloid leukemia progression and interfere with myeloid differentiation pathways. Haematologica. 2019;104(9):1789-1797.

171

Maletzke S, Salimi A, Vieri M, et al. Combined inhibition of BCR-ABL1 and the proteasome as a potential novel therapeutic approach in BCR-ABL positive acute lymphoblastic leukemia. PLoS One. 2022;17(10):e0268352.

172

Stein R, Smith MR, Chen S, Zalath M, Goldenberg DM. Combining milatuzumab with bortezomib, doxorubicin, or dexamethasone improves responses in multiple myeloma cell lines. Clin Cancer Res. 2009;15(8):2808-2817.

173

Vogl DT, Stadtmauer EA, Richardson PG, et al. Impact of prior therapies on the relative efficacy of bortezomib compared with dexamethasone in patients with relapsed/refractory multiple myeloma. Br J Haematol. 2009;147(4):531-534.

174

Albero MP, Vaquer JM, Andreu EJ, et al. Bortezomib decreases Rb phosphorylation and induces caspase-dependent apoptosis in imatinib-sensitive and-resistant Bcr-Abl1-expressing cells. Oncogene. 2010;29(22):3276-3286.

175

Heaney NB, Pellicano F, Zhang B, et al. Bortezomib induces apoptosis in primitive chronic myeloid leukemia cells including LTC-IC and NOD/SCID repopulating cells. Blood. 2010;115(11):2241-2250.

176

Li QF, Yan J, Zhang K, et al. Bortezomib and sphingosine kinase inhibitor interact synergistically to induces apoptosis in BCR/ABl+ cells sensitive and resistant to STI571 through down-regulation MCL-1. Biochem Biophys Res Commun. 2011;405(1):31-36.

177

Bucur O, Stancu AL, Goganau I, et al. Combination of bortezomib and mitotic inhibitors down-modulate Bcr-Abl and efficiently eliminates tyrosine-kinase inhibitor sensitive and resistant Bcr-Abl-positive leukemic cells. PLoS One. 2013;8(10):e77390.

178

Vij R, Wang M, Kaufman JL, et al. An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma. Blood. 2012;119(24):5661-5670.

179

Katsnelson A. Next-generation proteasome inhibitor approved in multiple myeloma. Nat Biotechnol. 2012;30(11):1011-1012.

180

Crawford LJ, Chan ET, Aujay M, et al. Synergistic effects of proteasome inhibitor carfilzomib in combination with tyrosine kinase inhibitors in imatinib-sensitive and-resistant chronic myeloid leukemia models. Oncogenesis. 2014;3(3):e90.

181

Zhou HJ, Aujay MA, Bennett MK, et al. Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047). J Med Chem. 2009;52(9):3028-3038.

182

Shah JJ, Stadtmauer EA, Abonour R, et al. Carfilzomib, pomalidomide, and dexamethasone for relapsed or refractory myeloma. Blood. 2015;126(20):2284-2290.

183

Ghobrial Irene M, Savona Michael R, Ravi V, et al. Final results from a multicenter, open-label, dose-escalation phase 1b/2 study of single-agent oprozomib in patients with hematologic malignancies. Blood. 2016;128(22):2110.

184

Wang F, Wang X, Li N, et al. Prolonged unfolded protein reaction is involved in the induction of chronic myeloid leukemia cell death upon oprozomib treatment. Cancer Sci. 2021;112(1):133-143.

185

Duncan K, Schäfer G, Vava A, Parker MI, Zerbini LF. Targeting neddylation in cancer therapy. Future Oncol. 2012;8(11):1461-1470.

186

Jiang Y, Jia L. Neddylation pathway as a novel anti-cancer target: mechanistic investigation and therapeutic implication. Anticancer Agents Med Chem. 2015;15(9):1127-1133.

187

Enchev RI, Schulman BA, Peter M. Protein neddylation: beyond cullin-RING ligases. Nat Rev Mol Cell Biol. 2015;16(1):30-44.

188

Huang J, Zhou Y, Thomas GS, et al. NEDD8 inhibition overcomes CKS1B-induced drug resistance by upregulation of p21 in multiple myeloma. Clin Cancer Res. 2015;21(24):5532-5542.

189

Bahjat M, de Wilde G, van Dam T, et al. The NEDD8-activating enzyme inhibitor MLN4924 induces DNA damage in Ph+ leukemia and sensitizes for ABL kinase inhibitors. Cell Cycle. 2019;18(18):2307-2322.

190

Haas AL, Ahrens P, Bright PM, Ankel H. Interferon induces a 15-kilodalton protein exhibiting marked homology to ubiquitin. J Biol Chem. 1987;262(23):11315-11323.

191

Yan M, Luo JK, Ritchie KJ, et al. Ubp43 regulates BCR-ABL leukemogenesis via the type 1 interferon receptor signaling. Blood. 2007;110(1):305-312.

192

Jiang Q, Li Z, Qin Y, et al. Olverembatinib (HQP1351), a well-tolerated and effective tyrosine kinase inhibitor for patients with T315I-mutated chronic myeloid leukemia: results of an open-label, multicenter phase 1/2 trial. J Hematol Oncol. 2022;15(1):1-17.

193

Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022;21(3):181-200.

194

Zhao L, Zhao J, Zhong K, Tong A, Jia D. Targeted protein degradation: mechanisms, strategies and application. Signal Transduct Target Ther. 2022;7(1):113.

195

Lai AC, Toure M, Hellerschmied D, et al. Modular PROTAC design for the degradation of oncogenic BCR-ABL. Angew Chem Int Ed Engl. 2016;55(2):807-810.

196

Burslem GM, Schultz AR, Bondeson DP, et al. Targeting BCR-ABL1 in chronic myeloid leukemia by PROTAC-mediated targeted protein degradation. Cancer Res. 2019;79(18):4744-4753.

197

Burslem GM, Bondeson DP, Crews CM. Scaffold hopping enables direct access to more potent PROTACs with in vivo activity. Chem Commun (Camb). 2020;56(50):6890-6892.

198

Jin YH, Lu MC, Wang Y, et al. Azo-PROTAC: novel light-controlled small-molecule tool for protein knockdown. J Med Chem. 2020;63(9):4644-4654.

199

Yang Y, Gao H, Sun X, et al. Global PROTAC toolbox for degrading BCR-ABL overcomes drug-resistant mutants and adverse effects. J Med Chem. 2020;63(15):8567-8583.

200

Zhao Q, Ren C, Liu L, et al. Discovery of SIAIS178 as an effective BCR-ABL degrader by recruiting von hippel-lindau (VHL) E3 ubiquitin ligase. J Med Chem. 2019;62(20):9281-9298.

201

Shibata N, Miyamoto N, Nagai K, et al. Development of protein degradation inducers of oncogenic BCR-ABL protein by conjugation of ABL kinase inhibitors and IAP ligands. Cancer Sci. 2017;108(8):1657-1666.

202

Shimokawa K, Shibata N, Sameshima T, et al. Targeting the allosteric site of oncoprotein BCR-ABL as an alternative strategy for effective target protein degradation. ACS Med Chem Lett. 2017;8(10):1042-1047.

203

Shibata N, Shimokawa K, Nagai K, et al. Pharmacological difference between degrader and inhibitor against oncogenic BCR-ABL kinase. Sci Rep. 2018;8(1):13549.

204

Chirnomas D, Hornberger KR, Crews CM. Protein degraders enter the clinic - a new approach to cancer therapy. Nat Rev Clin Oncol. 2023;20(4):265-278.

205

Mayor-Ruiz C, Jaeger MG, Bauer S, et al. Plasticity of the cullin-RING ligase repertoire shapes sensitivity to ligand-induced protein degradation. Mol Cell. 2019;75(4):849-858.e8.

206

Ottis P, Palladino C, Thienger P, et al. Cellular resistance mechanisms to targeted protein degradation converge toward impairment of the engaged ubiquitin transfer pathway. ACS Chem Biol. 2019;14(10):2215-2223.

207

Zhang L, Riley-Gillis B, Vijay P, Shen Y. Acquired resistance to BET-PROTACs (proteolysis-targeting chimeras) caused by genomic alterations in core components of E3 ligase complexes. Mol Cancer Ther. 2019;18(7):1302-1311.

208

Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137-148.

209

Biron CA. Interferons alpha and beta as immune regulators: a new look. Immunity. 2001;14(6):661-664.

210

Chin EN, Sulpizio A, Lairson LL. Targeting STING to promote antitumor immunity. Trends Cell Biol. 2023;33(3):189-203.

211

Bonifazi F, de Vivo A, Rosti G, et al. Chronic myeloid leukemia and interferon-alpha: a study of complete cytogenetic responders. Blood. 2001;98(10):3074-3081.

212

Kantarjian HM, O’Brien S, Cortes JE, et al. Complete cytogenetic and molecular responses to interferon-alpha-based therapy for chronic myelogenous leukemia are associated with excellent long-term prognosis. Cancer. 2003;97(4):1033-1041.

213

Angstreich GR, Matsui W, Huff CA, et al. Effects of imatinib and interferon on primitive chronic myeloid leukaemia progenitors. Br J Haematol. 2005;130(3):373-381.

Genes & Diseases
Article number: 101150
Cite this article:
Li X, Li W, Zhang Y, et al. Exploiting the potential of the ubiquitin-proteasome system in overcoming tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Genes & Diseases, 2024, 11(5): 101150. https://doi.org/10.1016/j.gendis.2023.101150

139

Views

0

Downloads

0

Crossref

0

Web of Science

0

Scopus

0

CSCD

Altmetrics

Received: 08 January 2023
Revised: 15 June 2023
Accepted: 01 September 2023
Published: 26 October 2023
© 2023 The Authors.

This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Return