Targeting intracellular proteins with cell type-specific functions for cancer immunotherapy
Madison E. Carelock, Rohan P. Master, Myung-Chul Kim, Zeng Jin, Lei Wang, Chandra K. Maharjan, Nan Hua, Umasankar De, Ryan Kolb, Yufeng Xiao, Daiqing Liao, Guangrong Zheng, Weizhou Zhang
Targeting intracellular proteins with cell type-specific functions for cancer immunotherapy
Immune checkpoint inhibitors (ICIs) use antibodies that block cell surface immune checkpoint proteins with great efficacy in treating immunogenic or “immune hot” tumors such as melanoma, kidney, and lung adenocarcinoma. ICIs have limited response rates to other non-immunogenic cancers. The tumor microenvironment (TME) consists of many cell types that collectively promote tumor progression. Cancer therapeutics are commonly designed to target one molecule in one defined cell type. There is growing evidence that long-term therapeutic responses require the targeting of cancer cells and tumor-promoting populations within the TME. The question remains whether we can identify targetable molecules/pathways that are critical for multiple cell types. Here, we will discuss several molecular targets that may fit a “two or multiple birds, one stone” model, including the B-cell lymphoma-2 (BCL-2) family pro-survival factors, transcriptional factors including signal transducer and activator of transcription 3, the nuclear receptor 4A family (NR4A1, NR4A2, and NR4A3), as well as epigenetic regulators such as bromodomain and extra-terminal (BET) family proteins, histone deacetylase family, SET domain bifurcated histone lysine methyltransferase 1 (SETDB1), and lysine-specific demethylase 1 (LSD1/KDM1A). We will focus on the rationale of these targets in immune modulation, as well as the strategies for targeting these important proteins for cancer therapy.
multifunctional protein targets / immune checkpoint inhibitors / immunotherapy / tumor microenvironment (TME)
[1] |
Sharma P, Siddiqui BA, Anandhan S, et al. The next decade of immune checkpoint therapy. Cancer Discov 2021;11(4):838–857.
CrossRef
Google scholar
|
[2] |
Johnson DB, Nebhan CA, Moslehi JJ, et al. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol 2022;19(4):254–267.
CrossRef
Google scholar
|
[3] |
Zhang C, He S, Zeng Z, et al. Smart nano-PROTACs reprogram tumor microenvironment for activatable photo-metabolic cancer immunotherapy. Angew Chem Int Ed Engl 2022;61(8):e202114957.
CrossRef
Google scholar
|
[4] |
Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol 2021;14(1):45.
CrossRef
Google scholar
|
[5] |
Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 2020;20(1):25–39.
CrossRef
Google scholar
|
[6] |
Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010;140(6):883–899.
CrossRef
Google scholar
|
[7] |
Mao X, Xu J, Wang W, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer 2021;20(1):131.
CrossRef
Google scholar
|
[8] |
Mhaidly R, Mechta-Grigoriou F. Fibroblast heterogeneity in tumor micro-environment: role in immunosuppression and new therapies. Semin Immunol 2020;48:101417.
CrossRef
Google scholar
|
[9] |
Monteran L, Erez N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immunosuppression in the tumor micro-environment. Front Immunol 2019;10:1835.
CrossRef
Google scholar
|
[10] |
De Jaeghere EA, Denys HG, De Wever O. Fibroblasts fuel immune escape in the tumor microenvironment. Trends Cancer 2019;5(11):704–723.
CrossRef
Google scholar
|
[11] |
Yang D, Guo P, He T, et al. Role of endothelial cells in tumor microenvironment. Clin Transl Med 2021;11(6):e450.
CrossRef
Google scholar
|
[12] |
Lee E, Pandey NB, Popel AS. Crosstalk between cancer cells and blood endothelial and lymphatic endothelial cells in tumour and organ microenvironment. Expert Rev Mol Med 2015;17:e3.
CrossRef
Google scholar
|
[13] |
Maishi N, Hida K. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci 2017;108(10):1921–1926.
CrossRef
Google scholar
|
[14] |
Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013;39(1):1–10.
CrossRef
Google scholar
|
[15] |
Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016;16(5):275–287.
CrossRef
Google scholar
|
[16] |
Goc J, Germain C, Vo-Bourgais TK, et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res 2014;74(3):705–715.
CrossRef
Google scholar
|
[17] |
Yost KE, Satpathy AT, Wells DK, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 2019;25(8):1251–1259.
CrossRef
Google scholar
|
[18] |
Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity 2014;41(1):49–61.
CrossRef
Google scholar
|
[19] |
Mantovani A, Allavena P, Marchesi F, et al. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov 2022;21(11):799–820.
CrossRef
Google scholar
|
[20] |
Pittet MJ, Michielin O, Migliorini D. Clinical relevance of tumour-associated macrophages. Nat Rev Clin Oncol 2022;19(6):402–421.
CrossRef
Google scholar
|
[21] |
Allavena P, Anfray C, Ummarino A, et al. Therapeutic manipulation of tumor-associated macrophages: facts and hopes from a clinical and translational perspective. Clin Cancer Res 2021;27(12):3291–3297.
CrossRef
Google scholar
|
[22] |
De Cicco P, Ercolano G, Ianaro A. The new era of cancer immunotherapy: targeting myeloid-derived suppressor cells to overcome immune evasion. Front Immunol 2020;11:1680.
CrossRef
Google scholar
|
[23] |
Tumino N, Fiore PF, Pelosi A, et al. Myeloid derived suppressor cells in tumor microenvironment: interaction with innate lymphoid cells. Semin Immunol 2022;61–64:101668.
CrossRef
Google scholar
|
[24] |
Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol 2021;21(8):485–498.
CrossRef
Google scholar
|
[25] |
Umansky V, Blattner C, Gebhardt C, et al. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines (Basel) 2016;4(4):36.
CrossRef
Google scholar
|
[26] |
Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-derived suppressor cells as a therapeutic target for cancer. Cells 2020;9(3):561.
CrossRef
Google scholar
|
[27] |
Ostrand-Rosenberg S, Fenselau C. Myeloid-derived suppressor cells: immune-suppressive cells that impair antitumor immunity and are sculpted by their environment. J Immunol 2018;200(2):422–431.
CrossRef
Google scholar
|
[28] |
Yang L, DeBusk LM, Fukuda K, et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 2004;6(4):409–421.
CrossRef
Google scholar
|
[29] |
Thornton AM, Shevach EM. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol 2000;164(1):183–190.
CrossRef
Google scholar
|
[30] |
Ahmadzadeh M, Pasetto A, Jia L, et al. Tumor-infiltrating human CD4(+) regulatory T cells display a distinct TCR repertoire and exhibit tumor and neoantigen reactivity. Sci Immunol 2019;4(31):eaao4310.
CrossRef
Google scholar
|
[31] |
Ohue Y, Nishikawa H. Regulatory T (Treg) cells in cancer: can Treg cells be a new therapeutic target? Cancer Sci 2019;110(7):2080–2089.
CrossRef
Google scholar
|
[32] |
Kolb HR, Borcherding N, Zhang W. Understanding and targeting human cancer regulatory T cells to improve therapy. Adv Exp Med Biol 2021;1278:229–256.
CrossRef
Google scholar
|
[33] |
Oft M. IL-10: master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol Res 2014;2(3):194–199.
CrossRef
Google scholar
|
[34] |
Tan W, Zhang W, Strasner A, et al. Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature 2011;470(7335):548–553.
CrossRef
Google scholar
|
[35] |
Chaudhary B, Elkord E. Regulatory T cells in the tumor microenvironment and cancer progression: role and therapeutic targeting. Vaccines (Basel) 2016;4(3):28.
CrossRef
Google scholar
|
[36] |
Kale J, Osterlund EJ, Andrews DW. BCL-2 family proteins: changing partners in the dance towards death. Cell Death Differ 2018;25(1):65–80.
CrossRef
Google scholar
|
[37] |
Chao DT, Korsmeyer SJ. BCL-2 family: regulators of cell death. Annu Rev Immunol 1998;16:395–419.
CrossRef
Google scholar
|
[38] |
Qian S, Wei Z, Yang W, et al. The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front Oncol 2022;12:985363.
CrossRef
Google scholar
|
[39] |
Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100(1):57–70.
CrossRef
Google scholar
|
[40] |
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646–674.
CrossRef
Google scholar
|
[41] |
Yip KW, Reed JC. Bcl-2 family proteins and cancer. Oncogene 2008;27(50):6398–6406.
CrossRef
Google scholar
|
[42] |
Schoenwaelder SM, Jarman KE, Gardiner EE, et al. Bcl-xL–inhibitory BH3 mimetics can induce a transient thrombocytopathy that undermines the hemostatic function of platelets. Blood 2011;118(6):1663–1674.
CrossRef
Google scholar
|
[43] |
Wesarg E, Hoffarth S, Wiewrodt R, et al. Targeting BCL-2 family proteins to overcome drug resistance in non-small cell lung cancer. Int J Cancer 2007;121(11):2387–2394.
CrossRef
Google scholar
|
[44] |
Khan S, Zhang X, Lv D, et al. A selective BCL-X(L) PROTAC degrader achieves safe and potent antitumor activity. Nat Med 2019;25(12):1938–1947.
CrossRef
Google scholar
|
[45] |
Kolb R, De U, Khan S, et al. Proteolysis-targeting chimera against BCL-X(L) destroys tumor-infiltrating regulatory T cells. Nat Commun 2021;12(1):1281.
CrossRef
Google scholar
|
[46] |
Hu X, Bardhan K, Paschall AV, et al. Deregulation of apoptotic factors Bcl-xL and Bax confers apoptotic resistance to myeloid-derived suppressor cells and contributes to their persistence in cancer. J Biol Chem 2013;288(26):19103–19115.
CrossRef
Google scholar
|
[47] |
Micha D, Cummings J, Shoemaker A, et al. Circulating biomarkers of cell death after treatment with the BH-3 mimetic ABT-737 in a preclinical model of small-cell lung cancer. Clin Cancer Res 2008;14(22):7304–7310.
CrossRef
Google scholar
|
[48] |
Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res 2008;68(9):3421–3428.
CrossRef
Google scholar
|
[49] |
Wilson WH, O’Connor OA, Czuczman MS, et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol 2010;11(12):1149–1159.
CrossRef
Google scholar
|
[50] |
Roberts AW, Advani RH, Kahl BS, et al. Phase 1 study of the safety, pharmacokinetics, and antitumour activity of the BCL2 inhibitor navitoclax in combination with rituximab in patients with relapsed or refractory CD20+ lymphoid malignancies. Br J Haematol 2015;170(5):669–678.
CrossRef
Google scholar
|
[51] |
Roberts AW, Seymour JF, Brown JR, et al. Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease. J Clin Oncol 2012;30(5):488–496.
CrossRef
Google scholar
|
[52] |
Cleary JM, Lima CM, Hurwitz HI, et al. A phase I clinical trial of navitoclax, a targeted high-affinity Bcl-2 family inhibitor, in combination with gemcitabine in patients with solid tumors. Invest New Drugs 2014;32(5):937–945.
CrossRef
Google scholar
|
[53] |
Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med 2013;19(2):202–208.
CrossRef
Google scholar
|
[54] |
Diepstraten ST, Anderson MA, Czabotar PE, et al. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat Rev Cancer 2022;22(1):45–64.
CrossRef
Google scholar
|
[55] |
Merino D, Kelly GL, Lessene G, et al. BH3-mimetic drugs: blazing the trail for new cancer medicines. Cancer Cell 2018;34(6):879–891.
CrossRef
Google scholar
|
[56] |
Cang S, Iragavarapu C, Savooji J, et al. ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J Hematol Oncol 2015;8:129.
CrossRef
Google scholar
|
[57] |
Kohlhapp FJ, Haribhai D, Mathew R, et al. Venetoclax increases intratumoral effector t cells and antitumor efficacy in combination with immune checkpoint blockade. Cancer Discov 2021;11(1):68–79.
CrossRef
Google scholar
|
[58] |
Lee JB, Khan DH, Hurren R, et al. Venetoclax enhances T cell-mediated antileukemic activity by increasing ROS production. Blood 2021;138(3):234–245.
CrossRef
Google scholar
|
[59] |
Lee YG, Guruprasad P, Ghilardi G, et al. Modulation of BCL-2 in both T cells and tumor cells to enhance chimeric antigen receptor T-cell immunotherapy against cancer. Cancer Discov 2022;12(10):2372–2391.
CrossRef
Google scholar
|
[60] |
Tao ZF, Hasvold L, Wang L, et al. Discovery of a potent and selective BCL-XL inhibitor with in vivo activity. ACS Med Chem Lett 2014;5(10):1088–1093.
CrossRef
Google scholar
|
[61] |
Wang L, Doherty GA, Judd AS, et al. Discovery of A-1331852, a first-in-class, potent, and orally-bioavailable BCL-X(L) inhibitor. ACS Med Chem Lett 2020;11(10):1829–1836.
CrossRef
Google scholar
|
[62] |
Lessene G, Czabotar PE, Sleebs BE, et al. Structure-guided design of a selective BCL-X(L) inhibitor. Nat Chem Biol 2013;9(6):390–397.
CrossRef
Google scholar
|
[63] |
He Y, Zhang X, Chang J, et al. Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat Commun 2020;11(1):1996.
CrossRef
Google scholar
|
[64] |
Lv D, Pal P, Liu X, et al. Development of a BCL-xL and BCL-2 dual degrader with improved anti-leukemic activity. Nat Commun 2021;12(1):6896.
CrossRef
Google scholar
|
[65] |
Sakamoto KM, Kim KB, Kumagai A, et al. Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc Natl Acad Sci U S A 2001;98(15):8554–8559.
CrossRef
Google scholar
|
[66] |
Schapira M, Calabrese MF, Bullock AN, et al. Targeted protein degradation: expanding the toolbox. Nat Rev Drug Discov 2019;18(12):949–963.
CrossRef
Google scholar
|
[67] |
Tesoriere A, Dinarello A, Argenton F. The roles of post-translational modifications in STAT3 biological activities and functions. Biomedicines 2021;9(8):956.
CrossRef
Google scholar
|
[68] |
Bharadwaj U, Kasembeli MM, Robinson P, et al. Targeting janus kinases and signal transducer and activator of transcription 3 to treat inflammation, fibrosis, and cancer: rationale, progress, and caution. Pharmacol Rev 2020;72(2):486–526.
CrossRef
Google scholar
|
[69] |
Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov 2022;12(1):31–46.
CrossRef
Google scholar
|
[70] |
Zhang W, Zong CS, Hermanto U, et al. RACK1 recruits STAT3 specifically to insulin and insulin-like growth factor 1 receptors for activation, which is important for regulating anchorage-independent growth. Mol Cell Biol 2006;26(2):413–424.
CrossRef
Google scholar
|
[71] |
Dong J, Cheng XD, Zhang WD, et al. Recent update on development of small-molecule STAT3 inhibitors for cancer therapy: from phosphorylation inhibition to protein degradation. J Med Chem 2021;64(13):8884–8915.
CrossRef
Google scholar
|
[72] |
Yuan ZL, Guan YJ, Chatterjee D, et al. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science 2005;307(5707):269–273.
CrossRef
Google scholar
|
[73] |
Zou S, Tong Q, Liu B, et al. Targeting STAT3 in cancer immunotherapy. Mol Cancer 2020;19(1):145.
CrossRef
Google scholar
|
[74] |
Qin J, Shen X, Zhang J, et al. Allosteric inhibitors of the STAT3 signaling pathway. Eur J Med Chem 2020;190:112122.
CrossRef
Google scholar
|
[75] |
Bai L, Zhou H, Xu R, et al. A potent and selective small-molecule degrader of STAT3 achieves complete tumor regression in vivo. Cancer Cell 2019;36(5):498–511.e17.
CrossRef
Google scholar
|
[76] |
Okusaka T, Ueno H, Ikeda M, et al. Phase 1 and pharmacological trial of OPB-31121, a signal transducer and activator of transcription-3 inhibitor, in patients with advanced hepatocellular carcinoma. Hepatol Res 2015;45(13):1283–1291.
CrossRef
Google scholar
|
[77] |
Wong AL, Soo RA, Tan DS, et al. Phase I and biomarker study of OPB-51602, a novel signal transducer and activator of transcription (STAT) 3 inhibitor, in patients with refractory solid malignancies. Ann Oncol 2015;26(5):998–1005.
CrossRef
Google scholar
|
[78] |
Lee M, Hirpara JL, Eu JQ, et al. Targeting STAT3 and oxidative phosphorylation in oncogene-addicted tumors. Redox Biol 2019;25:101073.
CrossRef
Google scholar
|
[79] |
Tweardy DJ. Drugging “undruggable” disease-causing proteins: focus on signal transducer and activator of transcription (Stat) 3. Trans Am Clin Climatol Assoc 2022;132:61–76.
|
[80] |
Wang T, Niu G, Kortylewski M, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med 2004;10(1):48–54.
CrossRef
Google scholar
|
[81] |
Wang Y, Shen Y, Wang S, et al. The role of STAT3 in leading the cross-talk between human cancers and the immune system. Cancer Lett 2018;415:117–128.
CrossRef
Google scholar
|
[82] |
Yang XO, Panopoulos AD, Nurieva R, et al. STAT3 regulates cytokine- mediated generation of inflammatory helper T cells. J Biol Chem 2007;282(13):9358–9363.
CrossRef
Google scholar
|
[83] |
Yang XO, Pappu BP, Nurieva R, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 2008;28(1):29–39.
CrossRef
Google scholar
|
[84] |
Zorn E, Nelson EA, Mohseni M, et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006;108(5):1571–1579.
CrossRef
Google scholar
|
[85] |
Kong LY, Wei J, Sharma AK, et al. A novel phosphorylated STAT3 inhibitor enhances T cell cytotoxicity against melanoma through inhibition of regulatory T cells. Cancer Immunol Immunother 2009;58(7):1023–1032.
CrossRef
Google scholar
|
[86] |
Kong LY, Abou-Ghazal MK, Wei J, et al. A novel inhibitor of signal transducers and activators of transcription 3 activation is efficacious against established central nervous system melanoma and inhibits regulatory T cells. Clin Cancer Res 2008;14(18):5759–5768.
CrossRef
Google scholar
|
[87] |
Hu H, Xiang Y, Li T, et al. Induction of M-MDSCs with IL6/GM-CSF from adherence monocytes and inhibition by WP1066. Exp Ther Med 2022;24(1):487.
CrossRef
Google scholar
|
[88] |
Zhou H, Bai L, Xu R, et al. SD-91 as a potent and selective STAT3 degrader capable of achieving complete and long-lasting tumor regression. ACS Med Chem Lett 2021;12(6):996–1004.
CrossRef
Google scholar
|
[89] |
Safe S, Karki K. The paradoxical roles of orphan nuclear receptor 4A (NR4A) in cancer. Mol Cancer Res 2021;19(2):180–191.
CrossRef
Google scholar
|
[90] |
Wenzl K, Troppan K, Neumeister P, et al. The nuclear orphan receptor NR4A1 and NR4A3 as tumor suppressors in hematologic neoplasms. Curr Drug Targets 2015;16(1):38–46.
CrossRef
Google scholar
|
[91] |
Mullican SE, Zhang S, Konopleva M, et al. Abrogation of nuclear receptors Nr4a3 and Nr4a1 leads to development of acute myeloid leukemia. Nat Med 2007;13(6):730–735.
CrossRef
Google scholar
|
[92] |
Chen C, Li Y, Hou S, et al. Orphan nuclear receptor TR3/Nur77 biologics inhibit tumor growth by targeting angiogenesis and tumor cells. Microvasc Res 2020;128:103934.
CrossRef
Google scholar
|
[93] |
Mohankumar K, Li X, Sridharan S, et al. Nuclear receptor 4A1 (NR4A1) antagonists induce ROS-dependent inhibition of mTOR signaling in endometrial cancer. Gynecol Oncol 2019;154(1):218–227.
CrossRef
Google scholar
|
[94] |
Hedrick E, Li X, Cheng Y, et al. Potent inhibition of breast cancer by bis-indole-derived nuclear receptor 4A1 (NR4A1) antagonists. Breast Cancer Res Treat 2019;177(1):29–40.
CrossRef
Google scholar
|
[95] |
Li XX, Wang ZJ, Zheng Y, et al. Nuclear receptor Nur77 facilitates melanoma cell survival under metabolic stress by protecting fatty acid oxidation. Mol Cell 2018;69(3):480–492.e7.
CrossRef
Google scholar
|
[96] |
Hibino S, Chikuma S, Kondo T, et al. Inhibition of Nr4a receptors enhances antitumor immunity by breaking treg-mediated immune tolerance. Cancer Res 2018;78(11):3027–3040.
CrossRef
Google scholar
|
[97] |
Hedrick E, Lee SO, Doddapaneni R, et al. NR4A1 antagonists inhibit beta1-integrin-dependent breast cancer cell migration. Mol Cell Biol 2016;36(9):1383–1394.
CrossRef
Google scholar
|
[98] |
Hedrick E, Lee SO, Kim G, et al. Nuclear receptor 4A1 (NR4A1) as a drug target for renal cell adenocarcinoma. PLoS One 2015;10(6):e0128308.
CrossRef
Google scholar
|
[99] |
Hedrick E, Lee SO, Doddapaneni R, et al. Nuclear receptor 4A1 as a drug target for breast cancer chemotherapy. Endocr Relat Cancer 2015;22(5):831–840.
CrossRef
Google scholar
|
[100] |
Zhou F, Drabsch Y, Dekker TJ, et al. Nuclear receptor NR4A1 promotes breast cancer invasion and metastasis by activating TGF-beta signalling. Nat Commun 2014;5:3388.
CrossRef
Google scholar
|
[101] |
Lee SO, Li X, Hedrick E, et al. Diindolylmethane analogs bind NR4A1 and are NR4A1 antagonists in colon cancer cells. Mol Endocrinol 2014;28(10):1729–1739.
CrossRef
Google scholar
|
[102] |
Zhao D, Qin L, Bourbon PM, et al. Orphan nuclear transcription factor TR3/Nur77 regulates microvessel permeability by targeting endothelial nitric oxide synthase and destabilizing endothelial junctions. Proc Natl Acad Sci U S A 2011;108(29):12066–12071.
CrossRef
Google scholar
|
[103] |
Li H, Kolluri SK, Gu J, et al. Cytochrome c release and apoptosis induced by mitochondrial targeting of nuclear orphan receptor TR3. Science 2000;289(5482):1159–1164.
CrossRef
Google scholar
|
[104] |
Smith AG, Lim W, Pearen M, et al. Regulation of NR4A nuclear receptor expression by oncogenic BRAF in melanoma cells. Pigment Cell Melanoma Res 2011;24(3):551–563.
CrossRef
Google scholar
|
[105] |
Johannessen CM, Johnson LA, Piccioni F, et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature 2013;504(7478):138–142.
CrossRef
Google scholar
|
[106] |
Zeng H, Qin L, Zhao D, et al. Orphan nuclear receptor TR3/Nur77 regulates VEGF-A-induced angiogenesis through its transcriptional activity. J Exp Med 2006;203(3):719–729.
CrossRef
Google scholar
|
[107] |
Ha CH, Wang W, Jhun BS, et al. Protein kinase D-dependent phosphorylation and nuclear export of histone deacetylase 5 mediates vascular endothelial growth factor-induced gene expression and angiogenesis. J Biol Chem 2008;283(21):14590–14599.
CrossRef
Google scholar
|
[108] |
Ismail H, Mofarrahi M, Echavarria R, et al. Angiopoietin-1 and vascular endothelial growth factor regulation of leukocyte adhesion to endothelial cells: role of nuclear receptor-77. Arterioscler Thromb Vasc Biol 2012;32(7):1707–1716.
CrossRef
Google scholar
|
[109] |
Ye T, Peng J, Liu X, et al. Orphan nuclear receptor TR3/Nur77 differentially regulates the expression of integrins in angiogenesis. Microvasc Res 2019;122:22–33.
CrossRef
Google scholar
|
[110] |
Liu X, Wang Y, Lu H, et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 2019;567(7749):525–529.
CrossRef
Google scholar
|
[111] |
Chen J, Lopez-Moyado IF, Seo H, et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 2019;567(7749):530–534.
CrossRef
Google scholar
|
[112] |
Kim MC, Borcherding N, Ahmed KK, et al. CD177 modulates the function and homeostasis of tumor-infiltrating regulatory T cells. Nat Commun 2021;12(1):5764.
CrossRef
Google scholar
|
[113] |
Oliveira G, Stromhaug K, Cieri N, et al. Landscape of helper and regulatory antitumour CD4(+) T cells in melanoma. Nature 2022;605(7910):532–538.
CrossRef
Google scholar
|
[114] |
Fassett MS, Jiang W, D’Alise AM, et al. Nuclear receptor Nr4a1 modulates both regulatory T-cell (Treg) differentiation and clonal deletion. Proc Natl Acad Sci U S A 2012;109(10):3891–3896.
CrossRef
Google scholar
|
[115] |
Wu L, Chen L. Characteristics of Nur77 and its ligands as potential anti-cancer compounds (Review). Mol Med Rep 2018;18(6):4793–4801.
CrossRef
Google scholar
|
[116] |
Karki K, Wright GA, Mohankumar K, et al. A Bis-indole-derived NR4A1 antagonist induces PD-L1 degradation and enhances antitumor immunity. Cancer Res 2020;80(5):1011–1023.
CrossRef
Google scholar
|
[117] |
Ho TCS, Chan AHY, Ganesan A. Thirty years of HDAC inhibitors: 2020 insight and hindsight. J Med Chem 2020;63(21):12460–12484.
CrossRef
Google scholar
|
[118] |
Li B, Samanta A, Song X, et al. FOXP3 interactions with histone acetyltransferase and class II histone deacetylases are required for repression. Proc Natl Acad Sci U S A 2007;104(11):4571–4576.
CrossRef
Google scholar
|
[119] |
Gryder BE, Pomella S, Sayers C, et al. Histone hyperacetylation disrupts core gene regulatory architecture in rhabdomyosarcoma. Nat Genet 2019;51(12):1714–1722.
CrossRef
Google scholar
|
[120] |
Slingerland M, Guchelaar HJ, Gelderblom H. Histone deacetylase inhibitors: an overview of the clinical studies in solid tumors. Anticancer Drugs 2014;25(2):140–149.
CrossRef
Google scholar
|
[121] |
Gryder BE, Sodji QH, Oyelere AK. Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Future Med Chem 2012;4(4):505–524.
CrossRef
Google scholar
|
[122] |
Li Y, Seto E. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harb Perspect Med 2016;6(10):a026831.
CrossRef
Google scholar
|
[123] |
Hontecillas-Prieto L, Flores-Campos R, Silver A, et al. Synergistic enhancement of cancer therapy using HDAC inhibitors: opportunity for clinical trials. Front Genet 2020;11:578011.
CrossRef
Google scholar
|
[124] |
Shen L, Ciesielski M, Ramakrishnan S, et al. Class I histone deacetylase inhibitor entinostat suppresses regulatory T cells and enhances immunotherapies in renal and prostate cancer models. PLoS One 2012;7(1):e30815.
CrossRef
Google scholar
|
[125] |
Kim K, Skora AD, Li Z, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A 2014;111(32):11774–11779.
CrossRef
Google scholar
|
[126] |
Gameiro SR, Malamas AS, Tsang KY, et al. Inhibitors of histone deacetylase 1 reverse the immune evasion phenotype to enhance T-cell mediated lysis of prostate and breast carcinoma cells. Oncotarget 2016;7(7):7390–7402.
CrossRef
Google scholar
|
[127] |
Truong AS, Zhou M, Krishnan B, et al. Entinostat induces antitumor immune responses through immune editing of tumor neoantigens. J Clin Invest 2021;131(16):e138560.
CrossRef
Google scholar
|
[128] |
Heninger E, Krueger TE, Thiede SM, et al. Inducible expression of cancer-testis antigens in human prostate cancer. Oncotarget 2016;7(51):84359–84374.
CrossRef
Google scholar
|
[129] |
Diaz-Carballo D, Saka S, Acikelli AH, et al. Enhanced antitumoral activity of TLR7 agonists via activation of human endogenous retroviruses by HDAC inhibitors. Commun Biol 2021;4(1):276.
CrossRef
Google scholar
|
[130] |
Banik D, Moufarrij S, Villagra A. Immunoepigenetics combination therapies: an overview of the role of HDACs in cancer immunotherapy. Int J Mol Sci 2019;20(9):2241.
CrossRef
Google scholar
|
[131] |
Tao R, de Zoeten EF, Ozkaynak E, et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 2007;13(11):1299–1307.
CrossRef
Google scholar
|
[132] |
Samanta A, Li B, Song X, et al. TGF-beta and IL-6 signals modulate chromatin binding and promoter occupancy by acetylated FOXP3. Proc Natl Acad Sci U S A 2008;105(37):14023–14027.
CrossRef
Google scholar
|
[133] |
de Zoeten EF, Wang L, Butler K, et al. Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp3(+) T-regulatory cells. Mol Cell Biol 2011;31(10):2066–2078.
CrossRef
Google scholar
|
[134] |
Hamminger P, Rica R, Ellmeier W. Histone deacetylases as targets in autoimmune and autoinflammatory diseases. Adv Immunol 2020;147:1–59.
CrossRef
Google scholar
|
[135] |
Wang L, Liu Y, Han R, et al. FOXP3+ regulatory T cell development and function require histone/protein deacetylase 3. J Clin Invest 2015;125(3):1111–1123.
CrossRef
Google scholar
|
[136] |
Xiong Y, Wang L, Di Giorgio E, et al. Inhibiting the coregulator CoREST impairs Foxp3+ Treg function and promotes antitumor immunity. J Clin Invest 2020;130(4):1830–1842.
CrossRef
Google scholar
|
[137] |
Govindaraj C, Tan P, Walker P, et al. Reducing TNF receptor 2+ regulatory T cells via the combined action of azacitidine and the HDAC inhibitor, panobinostat for clinical benefit in acute myeloid leukemia patients. Clin Cancer Res 2014;20(3):724–735.
CrossRef
Google scholar
|
[138] |
Pili R, Quinn DI, Hammers HJ, et al. Immunomodulation by entinostat in renal cell carcinoma patients receiving high-dose interleukin 2: a multicenter, single-arm, phase I/II trial (NCI-CTEP#7870). Clin Cancer Res 2017;23(23):7199–7208.
CrossRef
Google scholar
|
[139] |
Kim YD, Park SM, Ha HC, et al. HDAC inhibitor, CG-745, enhances the anti-cancer effect of anti-PD-1 immune checkpoint inhibitor by modulation of the immune microenvironment. J Cancer 2020;11(14):4059–4072.
CrossRef
Google scholar
|
[140] |
McCaw TR, Goel N, Brooke DJ, et al. Class I histone deacetylase inhibition promotes CD8 T cell activation in ovarian cancer. Cancer Med 2021;10(2):709–717.
CrossRef
Google scholar
|
[141] |
Chen C, Li X, Zhao H, et al. Discovery of DNA-targeting HDAC inhibitors with potent antitumor efficacy in vivo that trigger antitumor immunity. J Med Chem 2022;65(4):3667–3683.
CrossRef
Google scholar
|
[142] |
Thomas A, Rajan A, Szabo E, et al. A phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study. Clin Cancer Res 2014;20(21):5392–5402.
CrossRef
Google scholar
|
[143] |
Lin J, Elkon J, Ricart B, et al. Phase I study of entinostat in combination with enzalutamide for treatment of patients with metastatic castration-resistant prostate cancer. Oncologist 2021;26(12):e2136–e2142.
CrossRef
Google scholar
|
[144] |
Rosborough BR, Castellaneta A, Natarajan S, et al. Histone deacetylase inhibition facilitates GM-CSF-mediated expansion of myeloid-derived suppressor cells in vitro and in vivo. J Leukoc Biol 2012;91(5):701–709.
CrossRef
Google scholar
|
[145] |
Hashimoto A, Fukumoto T, Zhang R, et al. Selective targeting of different populations of myeloid-derived suppressor cells by histone deacetylase inhibitors. Cancer Immunol Immunother 2020;69(9):1929–1936.
CrossRef
Google scholar
|
[146] |
Wang HF, Ning F, Liu ZC, et al. Histone deacetylase inhibitors deplete myeloid-derived suppressor cells induced by 4T1 mammary tumors in vivo and in vitro. Cancer Immunol Immunother 2017;66(3):355–366.
CrossRef
Google scholar
|
[147] |
Orillion A, Hashimoto A, Damayanti N, et al. Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma. Clin Cancer Res 2017;23(17):5187–5201.
CrossRef
Google scholar
|
[148] |
Bae J, Hideshima T, Tai YT, et al. Histone deacetylase (HDAC) inhibitor ACY241 enhances anti-tumor activities of antigen-specific central memory cytotoxic T lymphocytes against multiple myeloma and solid tumors. Leukemia 2018;32(9):1932–1947.
CrossRef
Google scholar
|
[149] |
Christmas BJ, Rafie CI, Hopkins AC, et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol Res 2018;6(12):1561–1577.
CrossRef
Google scholar
|
[150] |
Lu Z, Zou J, Li S, et al. Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature 2020;579(7798):284–290.
CrossRef
Google scholar
|
[151] |
Sidiropoulos DN, Rafie CI, Jang JK, et al. Entinostat decreases immune suppression to promote antitumor responses in a HER2+ breast tumor microenvironment. Cancer Immunol Res 2022;10(5):656–669.
CrossRef
Google scholar
|
[152] |
Wang Y, Stowe RL, Pinello CE, et al. Identification of histone deacetylase inhibitors with benzoylhydrazide scaffold that selectively inhibit class I histone deacetylases. Chem Biol 2015;22(2):273–284.
CrossRef
Google scholar
|
[153] |
Yue K, Sun S, Jia G, et al. First-in-class hydrazide-based HDAC6 selective inhibitor with potent oral anti-inflammatory activity by attenuating NLRP3 inflammasome activation. J Med Chem 2022;65(18):12140–12162.
CrossRef
Google scholar
|
[154] |
Sun P, Wang J, Khan KS, et al. Development of alkylated hydrazides as highly potent and selective class I histone deacetylase inhibitors with T cell modulatory properties. J Med Chem 2022;65(24):16313-16337.
CrossRef
Google scholar
|
[155] |
Jiang Y, Xu J, Yue K, et al. Potent hydrazide-based HDAC inhibitors with a superior pharmacokinetic profile for efficient treatment of acute myeloid leukemia in vivo. J Med Chem 2022;65(1):285–302.
CrossRef
Google scholar
|
[156] |
Xiao Y, Wang J, Zhao LY, et al. Discovery of histone deacetylase 3 (HDAC3)-specific PROTACs. Chem Commun (Camb) 2020;56(68):9866–9869.
CrossRef
Google scholar
|
[157] |
Wu H, Yang K, Zhang Z, et al. Development of multifunctional histone deacetylase 6 degraders with potent antimyeloma activity. J Med Chem 2019;62(15):7042–7057.
CrossRef
Google scholar
|
[158] |
Yang H, Lv W, He M, et al. Plasticity in designing PROTACs for selective and potent degradation of HDAC6. Chem Commun (Camb) 2019;55(98):14848–14851.
CrossRef
Google scholar
|
[159] |
Cao F, de Weerd S, Chen D, et al. Induced protein degradation of histone deacetylases 3 (HDAC3) by proteolysis targeting chimera (PROTAC). Eur J Med Chem 2020;208:112800.
CrossRef
Google scholar
|
[160] |
Smalley JP, Baker IM, Pytel WA, et al. Optimization of class I histone deacetylase PROTACs reveals that HDAC1/2 degradation is critical to induce apoptosis and cell arrest in cancer cells. J Med Chem 2022;65(7):5642–5659.
CrossRef
Google scholar
|
[161] |
Darwish S, Ghazy E, Heimburg T, et al. Design, Synthesis and biological characterization of histone deacetylase 8 (HDAC8) proteolysis targeting chimeras (PROTACs) with anti-neuroblastoma activity. Int J Mol Sci 2022;23(14):7535.
CrossRef
Google scholar
|
[162] |
Macabuag N, Esmieu W, Breccia P, et al. Developing HDAC4- selective protein degraders to investigate the role of HDAC4 in Huntington’s disease pathology. J Med Chem 2022;65(18):12445–12459.
CrossRef
Google scholar
|
[163] |
Liu Z, Zhang Y, Xiang Y, et al. Small-molecule PROTACs for cancer immunotherapy. Molecules 2022;27(17):5439.
CrossRef
Google scholar
|
[164] |
Tomita Y, Lee MJ, Lee S, et al. The interplay of epigenetic therapy and immunity in locally recurrent or metastatic estrogen receptor-positive breast cancer: correlative analysis of ENCORE 301, a randomized, placebo-controlled phase II trial of exemestane with or without entinostat. Oncoimmunology 2016;5(11):e1219008.
CrossRef
Google scholar
|
[165] |
Taniguchi Y. The bromodomain and extra-terminal domain (BET) family: functional anatomy of BET paralogous proteins. Int J Mol Sci 2016;17(11):1849.
CrossRef
Google scholar
|
[166] |
Shorstova T, Foulkes WD, Witcher M. Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 2021;124(9):1478–1490.
CrossRef
Google scholar
|
[167] |
Waddell AR, Huang H, Liao D. CBP/p300: critical co-activators for nuclear steroid hormone receptors and emerging therapeutic targets in prostate and breast cancers. Cancers (Basel) 2021;13(12):2872.
CrossRef
Google scholar
|
[168] |
Andrieu G, Belkina AC, Denis GV. Clinical trials for BET inhibitors run ahead of the science. Drug Discov Today 2016;19:45–50.
CrossRef
Google scholar
|
[169] |
Lu J, Qian Y, Altieri M, et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem Biol 2015;22(6):755–763.
CrossRef
Google scholar
|
[170] |
Albrecht BK, Gehling VS, Hewitt MC, et al. Identification of a benzoisoxazoloazepine inhibitor (CPI-0610) of the bromodomain and extra-terminal (BET) family as a candidate for human clinical trials. J Med Chem 2016;59(4):1330–1339.
CrossRef
Google scholar
|
[171] |
Gupta V, Mascarenhas J, Kremyanskaya M, et al. MPN-379 matching- adjusted indirect comparison (MAIC) of pelabresib (CPI-0610) in combination with ruxolitinib vs. JAK inhibitor monotherapy in patients with intermediate or high-risk myelofibrosis. Clin Lymphoma Myeloma Leuk 2022;22 Suppl 2:S336.
CrossRef
Google scholar
|
[172] |
Kharenko OA, Patel RG, Calosing C, et al. Combination of ZEN-3694 with CDK4/6 inhibitors reverses acquired resistance to CDK4/6 inhibitors in ER-positive breast cancer. Cancer Gene Ther 2022;29(6):859–869.
CrossRef
Google scholar
|
[173] |
Kim DH, Sun D, Storck WK, et al. BET bromodomain inhibition blocks an AR-repressed, E2F1-activated treatment-emergent neuroendocrine prostate cancer lineage plasticity program. Clin Cancer Res 2021;27(17):4923–4936.
CrossRef
Google scholar
|
[174] |
Aggarwal RR, Schweizer MT, Nanus DM, et al. A phase Ib/IIa study of the pan-BET inhibitor ZEN-3694 in combination with enzalutamide in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res 2020;26(20):5338–5347.
CrossRef
Google scholar
|
[175] |
Wang N, Wu R, Tang D, et al. The BET family in immunity and disease. Signal Transduct Target Ther 2021;6(1):23.
CrossRef
Google scholar
|
[176] |
Riganti C, Lingua MF, Salaroglio IC, Falcomata C, et al. Bromodomain inhibition exerts its therapeutic potential in malignant pleural mesothelioma by promoting immunogenic cell death and changing the tumor immune-environment. Oncoimmunology 2018;7(3):e1398874.
CrossRef
Google scholar
|
[177] |
Wang H, Liu G, Jin X, Song S, et al. BET inhibitor JQ1 enhances anti-tumor immunity and synergizes with PD-1 blockade in CRC. J Cancer 2022;13(7):2126–2137.
CrossRef
Google scholar
|
[178] |
Andrieu GP, Shafran JS, Smith CL, et al. BET protein targeting suppresses the PD-1/PD-L1 pathway in triple-negative breast cancer and elicits anti-tumor immune response. Cancer Lett 2019;465:45–58.
CrossRef
Google scholar
|
[179] |
Jing X, Shao S, Zhang Y, et al. BRD4 inhibition suppresses PD-L1 expression in triple-negative breast cancer. Exp Cell Res 2020;392(2):112034.
CrossRef
Google scholar
|
[180] |
Zhu H, Bengsch F, Svoronos N, et al. BET bromodomain inhibition promotes anti-tumor immunity by suppressing PD-L1 expression. Cell Rep 2016;16(11):2829–2837.
CrossRef
Google scholar
|
[181] |
Adeegbe DO, Liu S, Hattersley MM, et al. BET bromodomain inhibition cooperates with PD-1 blockade to facilitate antitumor response in Kras-mutant non-small cell lung cancer. Cancer Immunol Res 2018;6(10):1234–1245.
CrossRef
Google scholar
|
[182] |
Winter GE, Buckley DL, Paulk J, et al. DRUG DEVELOPMENT. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015;348(6241):1376–1381.
CrossRef
Google scholar
|
[183] |
Zengerle M, Chan KH, Ciulli A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem Biol 2015;10(8):1770–1777.
CrossRef
Google scholar
|
[184] |
Raina K, Lu J, Qian Y, et al. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2016;113(26):7124–7129.
CrossRef
Google scholar
|
[185] |
Bhela IP, Ranza A, Balestrero FC, et al. A versatile and sustainable multicomponent platform for the synthesis of protein degraders: proof-of-concept application to BRD4-degrading PROTACs. J Med Chem 2022;65(22):15282–15299.
CrossRef
Google scholar
|
[186] |
Raina K, Lu J, Qian Y, et al. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2016;113(26):7124–7129.
CrossRef
Google scholar
|
[187] |
Bai L, Zhou B, Yang C-Y, et al. Targeted degradation of BET proteins in triple-negative breast cancersmall-molecule degraders of bet proteins in TNBC. Cancer Res 2017;77(9):2476–2487.
CrossRef
Google scholar
|
[188] |
Zhou B, Hu J, Xu F, et al. Discovery of a small-molecule degrader of bromodomain and extra-terminal (BET) proteins with picomolar cellular potencies and capable of achieving tumor regression. J Med Chem 2018;61(2):462–481.
CrossRef
Google scholar
|
[189] |
Shang E, Wang X, Wen D, et al. Double bromodomain-containing gene Brd2 is essential for embryonic development in mouse. Dev Dyn 2009;238(4):908–917.
CrossRef
Google scholar
|
[190] |
Houzelstein D, Bullock SL, Lynch DE, et al. Growth and early post implantation defects in mice deficient for the bromodomain-containing protein Brd4. Mol Cell Biol 2002;22(11):3794–3802.
CrossRef
Google scholar
|
[191] |
Shi Y, Lan F, Matson C, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 2004;119(7):941–953.
CrossRef
Google scholar
|
[192] |
Metzger E, Wissmann M, Yin N, et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 2005;437(7057):436–439.
CrossRef
Google scholar
|
[193] |
Laurent B, Ruitu L, Murn J, et al. A Specific LSD1/KDM1A isoform regulates neuronal differentiation through H3K9 demethylation. Mol Cell 2015;57(6):957-970.
CrossRef
Google scholar
|
[194] |
Wang J, Telese F, Tan Y, et al. LSD1n is an H4K20 demethylase regulating memory formation via transcriptional elongation control. Nat Neurosci 2015;18(9):1256–1264.
CrossRef
Google scholar
|
[195] |
Kim SA, Zhu J, Yennawar N, et al. Crystal structure of the LSD1/CoREST histone demethylase bound to its nucleosome substrate. Mol Cell 2020;78(5):903–914.e4.
CrossRef
Google scholar
|
[196] |
Rivera C, Lee HG, Lappala A, et al. Unveiling RCOR1 as a rheostat at transcriptionally permissive chromatin. Nat Commun 2022;13(1):1550.
CrossRef
Google scholar
|
[197] |
You A, Tong JK, Grozinger CM, et al. CoREST is an integral component of the CoREST-human histone deacetylase complex. Proc Natl Acad Sci U S A 2001;98(4):1454–1458.
CrossRef
Google scholar
|
[198] |
Song Y, Dagil L, Fairall L, et al. Mechanism of Crosstalk between the LSD1 demethylase and HDAC1 deacetylase in the CoREST complex. Cell Rep 2020;30(8):2699–2711.e8.
CrossRef
Google scholar
|
[199] |
Wu M, Hayward D, Kalin JH, et al. Lysine-14 acetylation of histone H3 in chromatin confers resistance to the deacetylase and demethylase activities of an epigenetic silencing complex. Elife 2018;7:e37231.
CrossRef
Google scholar
|
[200] |
Kalin JH, Wu M, Gomez AV, et al. Targeting the CoREST complex with dual histone deacetylase and demethylase inhibitors. Nat Commun 2018;9(1):53.
CrossRef
Google scholar
|
[201] |
Maksour S, Ooi L, Dottori M. More than a corepressor: the role of CoREST proteins in neurodevelopment. eNeuro 2020;7(2): ENEURO.0337–19.2020.
CrossRef
Google scholar
|
[202] |
Lan F, Collins RE, De Cegli R, et al. Recognition of unmethylated histone H3 lysine 4 links BHC80 to LSD1-mediated gene repression. Nature 2007;448(7154):718–722.
CrossRef
Google scholar
|
[203] |
Wang Z, Zang C, Cui K, et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 2009;138(5):1019–1031.
CrossRef
Google scholar
|
[204] |
Gryder BE, Wu L, Woldemichael GM, et al. Chemical genomics reveals histone deacetylases are required for core regulatory transcription. Nat Commun 2019;10(1):3004.
CrossRef
Google scholar
|
[205] |
Clark EA, Wu F, Chen Y, et al. GR and LSD1/KDM1A-targeted gene activation requires selective targeted gene activation requires selective H3K4me2 demethylation at enhancers. Cell Rep 2019;27(12):3522–3532.e3.
CrossRef
Google scholar
|
[206] |
Kim D, Kim KI, Baek SH. Roles of lysine-specific demethylase 1 (LSD1) in homeostasis and diseases. J Biomed Sci 2021;28(1):41.
CrossRef
Google scholar
|
[207] |
Tu WJ, McCuaig RD, Tan AHY, et al. Targeting nuclear LSD1 to reprogram cancer cells and reinvigorate exhausted t cells via a novel LSD1-EOMES switch. Front Immunol 2020;11:1228.
CrossRef
Google scholar
|
[208] |
Zhao LJ, Li YY, Zhang YT, et al. Lysine demethylase LSD1 delivered via small extracellular vesicles promotes gastric cancer cell stemness. EMBO Rep 2021;22(8):e50922.
CrossRef
Google scholar
|
[209] |
Sheng W, LaFleur MW, Nguyen TH, et al. LSD1 ablation stimulates anti-tumor immunity and enables checkpoint blockade. Cell 2018;174(3):549–563.e19.
CrossRef
Google scholar
|
[210] |
Sheng W, Liu Y, Chakraborty D, et al. Simultaneous inhibition of LSD1 and TGFbeta enables eradication of poorly immunogenic tumors with anti-PD-1 treatment. Cancer Discov 2021:11(8):1970–1981.
CrossRef
Google scholar
|
[211] |
Liu Y, Debo B, Li M, et al. LSD1 inhibition sustains T cell invigoration with a durable response to PD-1 blockade. Nat Commun 2021;12(1):6831.
CrossRef
Google scholar
|
[212] |
Haydn T, Metzger E, Schuele R, et al. Concomitant epigenetic targeting of LSD1 and HDAC synergistically induces mitochondrial apoptosis in rhabdomyosarcoma cells. Cell Death Dis 2017;8(6):e2879.
CrossRef
Google scholar
|
[213] |
Qin Y, Vasilatos SN, Chen L, et al. Inhibition of histone lysine-specific demethylase 1 elicits breast tumor immunity and enhances antitumor efficacy of immune checkpoint blockade. Oncogene 2019;38(3):390–405.
CrossRef
Google scholar
|
[214] |
Nguyen EM, Taniguchi H, Chan JM, et al. Targeting lysine-specific demethylase 1 rescues major histocompatibility complex class I antigen presentation and overcomes programmed death-ligand 1 blockade resistance in SCLC. J Thorac Oncol 2022;17(8):1014-1031.
CrossRef
Google scholar
|
[215] |
Boulding T, McCuaig RD, Tan A, et al. LSD1 activation promotes inducible EMT programs and modulates the tumour microenvironment in breast cancer. Sci Rep 2018;8(1):73.
CrossRef
Google scholar
|
[216] |
Tan AHY, Tu W, McCuaig R, et al. Lysine-specific histone demethylase 1A regulates macrophage polarization and checkpoint molecules in the tumor microenvironment of triple-negative breast cancer. Front Immunol 2019;10:1351.
CrossRef
Google scholar
|
[217] |
Strepkos D, Markouli M, Klonou A, et al. Histone methyltransferase SETDB1: a common denominator of tumorigenesis with therapeutic potential. Cancer Res 2021;81(3):525–534.
CrossRef
Google scholar
|
[218] |
Griffin GK, Wu J, Iracheta-Vellve A, et al. Epigenetic silencing by SETDB1 suppresses tumour intrinsic immunogenicity. Nature 2021;595(7866):309–314.
CrossRef
Google scholar
|
[219] |
Zhang SM, Cai WL, Liu X, et al. KDM5B promotes immune evasion by recruiting SETDB1 to silence retroelements. Nature 2021;598(7882):682–687.
CrossRef
Google scholar
|
[220] |
Hu H, Khodadadi-Jamayran A, Dolgalev I, et al. Targeting the Atf7ip-Setdb1 complex augments antitumor immunity by boosting tumor immunogenicity. Cancer Immunol Res 2021;9(11):1298–1315.
CrossRef
Google scholar
|
[221] |
Pan D, Bao X, Hu M, et al. SETDB1 restrains endogenous retrovirus expression and antitumor immunity during radiotherapy. Cancer Res 2022;82(15):2748–2760.
CrossRef
Google scholar
|
[222] |
Guo Y, Mao X, Xiong L, et al. Structure-guided discovery of a potent and selective cell-active inhibitor of SETDB1 tudor domain. Angew Chem Int Ed Engl 2021;60(16):8760–8765.
CrossRef
Google scholar
|
[223] |
Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51(2):202–206.
CrossRef
Google scholar
|
[224] |
Marcus L, Lemery SJ, Keegan P, et al. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res 2019;25(13):3753–3758.
CrossRef
Google scholar
|
[225] |
Haen SP, Loffler MW, Rammensee HG, et al. Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire. Nat Rev Clin Oncol 2020;17(10):595–610.
CrossRef
Google scholar
|
[226] |
Yang L, Rau R, Goodell MA. DNMT3A in haematological malignancies. Nat Rev Cancer 2015;15(3):152–165.
CrossRef
Google scholar
|
[227] |
de Vos D, van Overveld W. Decitabine: a historical review of the development of an epigenetic drug. Ann Hematol 2005;84 (Suppl 1):3–8.
CrossRef
Google scholar
|
[228] |
Saxena M, van der Burg SH, Melief CJM, et al. Therapeutic cancer vaccines. Nat Rev Cancer 2021;21(6):360–378.
CrossRef
Google scholar
|
[229] |
Kroemer G, Galluzzi L, Kepp O, et al. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013;31:51–72.
CrossRef
Google scholar
|
[230] |
Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20(11):651–668.
CrossRef
Google scholar
|
[231] |
Weichselbaum RR, Liang H, Deng L, et al. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol 2017;14(6):365–379.
CrossRef
Google scholar
|
[232] |
Venkatesulu BP, Mallick S, Lin SH, et al. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit Rev Oncol Hematol 2018;123:42–51.
CrossRef
Google scholar
|
[233] |
Menetrier-Caux C, Ray-Coquard I, Blay JY, et al. Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer 2019;7(1):85.
CrossRef
Google scholar
|
[234] |
Blachere NE, Darnell RB, Albert ML. Apoptotic cells deliver processed antigen to dendritic cells for cross-presentation. PLoS Biol 2005;3(6):e185.
CrossRef
Google scholar
|
[235] |
Galea-Lauri J, Darling D, Mufti G, et al. Eliciting cytotoxic T lymphocytes against acute myeloid leukemia-derived antigens: evaluation of dendritic cell-leukemia cell hybrids and other antigen-loading strategies for dendritic cell-based vaccination. Cancer Immunol Immunother 2002;51(6): 299–310.
CrossRef
Google scholar
|
[236] |
Jenne L, Arrighi JF, Jonuleit H, et al. Dendritic cells containing apoptotic melanoma cells prime human CD8+ T cells for efficient tumor cell lysis. Cancer Res 2000;60(16):4446–4452.
|
[237] |
Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol 2016;39(1):98–106.
CrossRef
Google scholar
|
[238] |
Qureshi OS, Zheng Y, Nakamura K, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 2011;332(6029):600–603.
CrossRef
Google scholar
|
[239] |
ElTanbouly MA, Zhao Y, Nowak E, et al. VISTA is a checkpoint regulator for naive T cell quiescence and peripheral tolerance. Science 2020;367(6475):eaay0524.
CrossRef
Google scholar
|
[240] |
Mackay LK, Rahimpour A, Ma JZ, et al. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol 2013;14(12):1294–1301.
CrossRef
Google scholar
|
[241] |
Beura LK, Wijeyesinghe S, Thompson EA, et al. T cells in nonlymphoid tissues give rise to lymph-node-resident memory T cells. Immunity 2018;48(2):327–338.e5.
CrossRef
Google scholar
|
[242] |
Plebanek MP, Angeloni NL, Vinokour E, et al. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun 2017;8(1):1319.
CrossRef
Google scholar
|
[243] |
Takeda K, Noguchi K, Shi W, et al. Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc Natl Acad Sci U S A 1997;94(8):3801–3804.
CrossRef
Google scholar
|
/
〈 | 〉 |