Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets

Jun Jiang , Donghui Han , Jiawei Wang , Weihong Wen , Rui Zhang , Weijun Qin

MedComm ›› 2024, Vol. 5 ›› Issue (10) : e761

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (10) : e761 DOI: 10.1002/mco2.761
REVIEW

Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets

Author information +
History +
PDF

Abstract

Neuroendocrine transdifferentiation (NEtD), also commonly referred to as lineage plasticity, emerges as an acquired resistance mechanism to molecular targeted therapies in multiple cancer types, predominately occurs in metastatic epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer treated with EGFR tyrosine kinase inhibitors and metastatic castration-resistant prostate cancer treated with androgen receptor targeting therapies. NEtD tumors are the lethal cancer histologic subtype with unfavorable prognosis and limited treatment. A comprehensive understanding of molecular mechanism underlying targeted-induced plasticity could greatly facilitate the development of novel therapies. In the past few years, increasingly elegant studies indicated that NEtD tumors share key the convergent genomic and phenotypic characteristics irrespective of their site of origin, but also embrace distinct change and function of molecular mechanisms. In this review, we provide a comprehensive overview of the current understanding of molecular mechanism in regulating the NEtD, including genetic alterations, DNA methylation, histone modifications, dysregulated noncoding RNA, lineage-specific transcription factors regulation, and other proteomic alterations. We also provide the current management of targeted therapies in clinical and preclinical practice.

Keywords

acquired resistance / lineage plasticity / neuroendocrine prostate cancer / neuroendocrine transdifferentiation / small cell lung cancer

Cite this article

Download citation ▾
Jun Jiang, Donghui Han, Jiawei Wang, Weihong Wen, Rui Zhang, Weijun Qin. Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets. MedComm, 2024, 5(10): e761 DOI:10.1002/mco2.761

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Beltran H, Rickman DS, Park K, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 2011; 1(6): 487-495.

[2]

Wang Q, Gümüş ZH, Colarossi C, et al. SCLC: epidemiology, risk factors, genetic susceptibility, molecular pathology, screening, and early detection. J Thorac Oncol. 2023; 18(1): 31-46.

[3]

Quintanal-Villalonga Á, Chan JM, Yu HA, et al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat Rev Clin Oncol. 2020; 17(6): 360-371.

[4]

Balanis NG, Sheu KM, Esedebe FN, et al. Pan-cancer convergence to a small-cell neuroendocrine phenotype that shares susceptibilities with hematological malignancies. Cancer Cell. 2019; 36(1): 17-34. e17.

[5]

Aggarwal R, Huang J, Alumkal JJ, et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. J Clin Oncol. 2018; 36(24): 2492-2503.

[6]

Marcoux N, Gettinger SN, O’Kane G, et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. J Clin Oncol. 2019; 37(4): 278-285.

[7]

Wang Y, Wang Y, Ci X, et al. Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol. 2021; 18(10): 581-596.

[8]

Rickman DS, Beltran H, Demichelis F, Rubin MA. Biology and evolution of poorly differentiated neuroendocrine tumors. Nat Med. 2017; 23(6): 1-10.

[9]

Offin M, Chan JM, Tenet M, et al. Concurrent RB1 and TP53 alterations define a subset of EGFR-mutant lung cancers at risk for histologic transformation and inferior clinical outcomes. J Thorac Oncol. 2019; 14(10): 1784-1793.

[10]

Park JW, Lee JK, Sheu KM, et al. Reprogramming normal human epithelial tissues to a common, lethal neuroendocrine cancer lineage. Science. 2018; 362(6410): 91-95.

[11]

Patel AS, Yoo S, Kong R, et al. Prototypical oncogene family Myc defines unappreciated distinct lineage states of small cell lung cancer. Sci Adv. 2021; 7(5): eabc2578.

[12]

Baine MK, Hsieh MS, Lai WV, et al. SCLC subtypes defined by ASCL1, NEUROD1, POU2F3, and YAP1: a comprehensive immunohistochemical and histopathologic characterization. J Thorac Oncol. 2020; 15(12): 1823-1835.

[13]

Gay CM, Stewart CA, Park EM, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell. 2021; 39(3): 346-360.

[14]

Cejas P, Xie Y, Font-Tello A, et al. Subtype heterogeneity and epigenetic convergence in neuroendocrine prostate cancer. Nat Commun. 2021; 12(1): 5775.

[15]

Chen CC, Tran W, Song K, et al. Temporal evolution reveals bifurcated lineages in aggressive neuroendocrine small cell prostate cancer trans-differentiation. Cancer Cell. 2023; 41(12): 2066-2082.

[16]

Han M, Li F, Zhang Y, et al. FOXA2 drives lineage plasticity and KIT pathway activation in neuroendocrine prostate cancer. Cancer Cell. 2022; 40(11): 1306-1323. e1308.

[17]

Nouruzi S, Ganguli D, Tabrizian N, et al. ASCL1 activates neuronal stem cell-like lineage programming through remodeling of the chromatin landscape in prostate cancer. Nat Commun. 2022; 13(1): 2282.

[18]

Joung J, Ma S, Tay T, et al. A transcription factor atlas of directed differentiation. Cell. 2023; 186(1): 209-229. e226.

[19]

Mu P, Zhang Z, Benelli M, et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53-and RB1-deficient prostate cancer. Science. 2017; 355(6320): 84-88.

[20]

Ku SY, Rosario S, Wang Y, et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science. 2017; 355(6320): 78-83.

[21]

Niederst MJ, Sequist LV, Poirier JT, et al. RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer. Nat Commun. 2015; 6: 6377.

[22]

Quigley DA, Dang HX, Zhao SG, et al. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell. 2018; 174(3): 758-769. e759.

[23]

Sivakumar S, Moore JA, Montesion M, et al. Integrative analysis of a large real-world cohort of small cell lung cancer identifies distinct genetic subtypes and insights into histologic transformation. Cancer Discov. 2023; 13(7): 1572-1591.

[24]

Chan JM, Zaidi S, Love JR, et al. Lineage plasticity in prostate cancer depends on JAK/STAT inflammatory signaling. Science. 2022; 377(6611): 1180-1191.

[25]

Chang MT, Penson A, Desai NB, et al. Small-cell carcinomas of the bladder and lung are characterized by a convergent but distinct pathogenesis. Clin Cancer Res. 2018; 24(8): 1965-1973.

[26]

Lázaro S, Pérez-Crespo M, Lorz C, et al. Differential development of large-cell neuroendocrine or small-cell lung carcinoma upon inactivation of 4 tumor suppressor genes. Proc Natl Acad Sci USA. 2019; 116(44): 22300-22306.

[27]

Armenia J, Wankowicz SAM, Liu D, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet. 2018; 50(5): 645-651.

[28]

Wang S, Garcia AJ, Wu M, Lawson DA, Witte ON, Wu H. Pten deletion leads to the expansion of a prostatic stem/progenitor cell subpopulation and tumor initiation. Proc Natl Acad Sci USA. 2006; 103(5): 1480-1485.

[29]

Kwon OJ, Zhang L, Jia D, Xin L. Sox2 is necessary for androgen ablation-induced neuroendocrine differentiation from Pten null Sca-1(+) prostate luminal cells. Oncogene. 2021; 40(1): 203-214.

[30]

Zhang L, Liu C, Zhang B, et al. PTEN loss expands the histopathologic diversity and lineage plasticity of lung cancers initiated by Rb1/Trp53 deletion. J Thorac Oncol. 2023; 18(3): 324-338.

[31]

Quintanal-Villalonga A, Durani V, Sabet A, et al. Exportin 1 inhibition prevents neuroendocrine transformation through SOX2 down-regulation in lung and prostate cancers. Sci Transl Med. 2023; 15(707): eadf7006.

[32]

Quintanal-Villalonga A, Taniguchi H, Zhan YA, et al. Multiomic analysis of lung tumors defines pathways activated in neuroendocrine transformation. Cancer Discov. 2021; 11(12): 3028-3047.

[33]

Gardner EE, Earlie EM, Li K, et al. Lineage-specific intolerance to oncogenic drivers restricts histological transformation. Science. 2024; 383(6683): eadj1415.

[34]

Dang CV. MYC on the path to cancer. Cell. 2012; 149(1): 22-35.

[35]

Peifer M, Fernández-Cuesta L, Sos ML, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012; 44(10): 1104-1110.

[36]

Kim DW, Wu N, Kim YC, et al. Genetic requirement for Mycl and efficacy of RNA Pol I inhibition in mouse models of small cell lung cancer. Genes Dev. 2016; 30(11): 1289-1299.

[37]

Ireland AS, Micinski AM, Kastner DW, et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell. 2020; 38(1): 60-78.

[38]

Lee JK, Phillips JW, Smith BA, et al. N-Myc drives neuroendocrine prostate cancer initiated from human prostate epithelial cells. Cancer Cell. 2016; 29(4): 536-547.

[39]

Dardenne E, Beltran H, Benelli M, et al. N-Myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell. 2016; 30(4): 563-577.

[40]

Berger A, Brady NJ, Bareja R, et al. N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest. 2019; 129(9): 3924-3940.

[41]

Unno K, Chalmers ZR, Pamarthy S, et al. Activated ALK cooperates with N-Myc via Wnt/β-catenin signaling to induce neuroendocrine prostate cancer. Cancer Res. 2021; 81(8): 2157-2170.

[42]

Chen WY, Thuy Dung PV, Yeh HL, et al. Targeting PKLR/MYCN/ROMO1 signaling suppresses neuroendocrine differentiation of castration-resistant prostate cancer. Redox Biol. 2023; 62: 102686.

[43]

Beltran H, Prandi D, Mosquera JM, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016; 22(3): 298-305.

[44]

Gritsina G, Fong KW, Lu X, et al. Chemokine receptor CXCR7 activates Aurora Kinase A and promotes neuroendocrine prostate cancer growth. J Clin Invest. 2023; 133(15): e166248.

[45]

Montagnoli A, Moll J, Colotta F. Targeting cell division cycle 7 kinase: a new approach for cancer therapy. Clin Cancer Res. 2010; 16(18): 4503-4508.

[46]

Quintanal-Villalonga A, Kawasaki K, Redin E, et al. CDC7 inhibition impairs neuroendocrine transformation in lung and prostate tumors through MYC degradation. Signal Transduct Target Ther. 2024; 9(1): 189.

[47]

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5): 646-674.

[48]

Ray Chaudhuri A, Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol. 2017; 18(10): 610-621.

[49]

Demény MA, Virág L. The PARP enzyme family and the hallmarks of cancer part 1. Cell intrinsic hallmarks. Cancers (Basel). 2021; 13(9): 2042.

[50]

Zhang W, Liu B, Wu W, et al. Targeting the MYCN-PARP-DNA damage response pathway in neuroendocrine prostate cancer. Clin Cancer Res. 2018; 24(3): 696-707.

[51]

Dias MP, Moser SC, Ganesan S, Jonkers J. Understanding and overcoming resistance to PARP inhibitors in cancer therapy. Nat Rev Clin Oncol. 2021; 18(12): 773-791.

[52]

Liu B, Li L, Yang G, et al. PARP inhibition suppresses GR-MYCN-CDK5-RB1-E2F1 signaling and neuroendocrine differentiation in castration-resistant prostate cancer. Clin Cancer Res. 2019; 25(22): 6839-6851.

[53]

Shen J, Zhao W, Ju Z, et al. PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness. Cancer Res. 2019; 79(2): 311-319.

[54]

Li Q, Qian W, Zhang Y, Hu L, Chen S, Xia Y. A new wave of innovations within the DNA damage response. Signal Transduct Target Ther. 2023; 8(1): 338.

[55]

Thomas A, Takahashi N, Rajapakse VN, et al. Therapeutic targeting of ATR yields durable regressions in small cell lung cancers with high replication stress. Cancer Cell. 2021; 39(4): 566-579.

[56]

Yin Y, Xu L, Chang Y, et al. N-Myc promotes therapeutic resistance development of neuroendocrine prostate cancer by differentially regulating miR-421/ATM pathway. Mol Cancer. 2019; 18(1): 11.

[57]

George J, Lim JS, Jang SJ, et al. Comprehensive genomic profiles of small cell lung cancer. Nature. 2015; 524(7563): 47-53.

[58]

Kim B, Zhang S, Huang Y, et al. CRACD loss induces neuroendocrine cell plasticity of lung adenocarcinoma. Cell Rep. 2024; 43(6): 114286.

[59]

Cyrta J, Augspach A, De Filippo MR, et al. Role of specialized composition of SWI/SNF complexes in prostate cancer lineage plasticity. Nat Commun. 2020; 11(1): 5549.

[60]

Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014; 505(7484): 495-501.

[61]

Concepcion CP, Ma S, LaFave LM, et al. Smarca4 inactivation promotes lineage-specific transformation and early metastatic features in the lung. Cancer Discov. 2022; 12(2): 562-585.

[62]

Redin E, Sridhar H, Zhan YA, et al. SMARCA4 controls state plasticity in small cell lung cancer through regulation of neuroendocrine transcription factors and REST splicing. J Hematol Oncol. 2024; 17(1): 58.

[63]

Ge R, Wang Z, Montironi R, et al. Epigenetic modulations and lineage plasticity in advanced prostate cancer. Ann Oncol. 2020; 31(4): 470-479.

[64]

Song X, Lan Y, Zheng X, et al. Targeting drug-tolerant cells: a promising strategy for overcoming acquired drug resistance in cancer cells. Med Comm (2020). 2023; 4(5): e342.

[65]

Das PM, Singal R. DNA methylation and cancer. J Clin Oncol. 2004; 22(22): 4632-4642.

[66]

Nishiyama A, Nakanishi M. Navigating the DNA methylation landscape of cancer. Trends Genet. 2021; 37(11): 1012-1027.

[67]

Yamada Y, Venkadakrishnan VB, Mizuno K, et al. Targeting DNA methylation and B7-H3 in RB1-deficient and neuroendocrine prostate cancer. Sci Transl Med. 2023; 15(722): eadf6732.

[68]

Cong B, Zhang Q, Cao X. The function and regulation of TET2 in innate immunity and inflammation. Protein Cell. 2021; 12(3): 165-173.

[69]

Kim MR, Wu MJ, Zhang Y, Yang JY, Chang CJ. TET2 directs mammary luminal cell differentiation and endocrine response. Nat Commun. 2020; 11(1): 4642.

[70]

Xu Y, Yang Y, Wang Z, et al. ZNF397 deficiency triggers TET2-driven lineage plasticity and AR-targeted therapy resistance in prostate cancer. Cancer Discov. 2024; 14(8): 1496-1521.

[71]

Littau VC, Burdick CJ, Allfrey VG, Mirsky SA. The role of histones in the maintenance of chromatin structure. Proc Natl Acad Sci USA. 1965; 54(4): 1204-1212.

[72]

Hyun K, Jeon J, Park K, Kim J. Writing, erasing and reading histone lysine methylations. Exp Mol Med. 2017; 49(4): e324.

[73]

Husmann D, Gozani O. Histone lysine methyltransferases in biology and disease. Nat Struct Mol Biol. 2019; 26(10): 880-889.

[74]

Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat Med. 2016; 22(2): 128-134.

[75]

Zhang Y, Zheng D, Zhou T, et al. Androgen deprivation promotes neuroendocrine differentiation and angiogenesis through CREB-EZH2-TSP1 pathway in prostate cancers. Nat Commun. 2018; 9(1): 4080.

[76]

Mahadevan NR, Knelson EH, Wolff JO, et al. Intrinsic immunogenicity of small cell lung carcinoma revealed by its cellular plasticity. Cancer Discov. 2021; 11(8): 1952-1969.

[77]

Cañadas I, Thummalapalli R, Kim JW, et al. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med. 2018; 24(8): 1143-1150.

[78]

Souza BK, Freire NH, Jaeger M, et al. EHMT2/G9a as an epigenetic target in pediatric and adult brain tumors. Int J Mol Sci. 2021; 22(20): 11292.

[79]

Yang C, Ma S, Zhang J, et al. EHMT2-mediated transcriptional reprogramming drives neuroendocrine transformation in non-small cell lung cancer. Proc Natl Acad Sci USA. 2024; 121(23): e2317790121.

[80]

Augert A, Zhang Q, Bates B, et al. Small cell lung cancer exhibits frequent inactivating mutations in the histone methyltransferase KMT2D/MLL2: cALGB 151111 (Alliance). J Thorac Oncol. 2017; 12(4): 704-713.

[81]

Bellone S, Jeong K, Halle MK, et al. Integrated mutational landscape analysis of poorly differentiated high-grade neuroendocrine carcinoma of the uterine cervix. Proc Natl Acad Sci USA. 2024; 121(17): e2321898121.

[82]

Manni W, Jianxin X, Weiqi H, Siyuan C, Huashan S. JMJD family proteins in cancer and inflammation. Signal Transduct Target Ther. 2022; 7(1): 304.

[83]

Kumaraswamy A, Duan Z, Flores D, et al. LSD1 promotes prostate cancer reprogramming by repressing TP53 signaling independently of its demethylase function. JCI Insight. 2023; 8(15): e167440.

[84]

Augert A, Eastwood E, Ibrahim AH, et al. Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci Signal. 2019; 12(567): eaau2922.

[85]

Park DE, Cheng J, McGrath JP, et al. Merkel cell polyomavirus activates LSD1-mediated blockade of non-canonical BAF to regulate transformation and tumorigenesis. Nat Cell Biol. 2020; 22(5): 603-615.

[86]

Sehrawat A, Gao L, Wang Y, et al. LSD1 activates a lethal prostate cancer gene network independently of its demethylase function. Proc Natl Acad Sci USA. 2018; 115(18): E4179-e4188.

[87]

Huang J, Sengupta R, Espejo AB, et al. p53 is regulated by the lysine demethylase LSD1. Nature. 2007; 449(7158): 105-108.

[88]

Shi Y, Lan F, Matson C, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004; 119(7): 941-953.

[89]

Rudin CM, Balli D, Lai WV, et al. Clinical benefit from immunotherapy in patients with SCLC is associated with tumor capacity for antigen presentation. J Thorac Oncol. 2023; 18(9): 1222-1232.

[90]

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.

[91]

Hiatt JB, Sandborg H, Garrison SM, et al. Inhibition of LSD1 with bomedemstat sensitizes small cell lung cancer to immune checkpoint blockade and T-cell killing. Clin Cancer Res. 2022; 28(20): 4551-4564.

[92]

Oser MG, Sabet AH, Gao W, et al. The KDM5A/RBP2 histone demethylase represses NOTCH signaling to sustain neuroendocrine differentiation and promote small cell lung cancer tumorigenesis. Genes Dev. 2019; 33(23-24): 1718-1738.

[93]

Du C, Lv C, Feng Y, Yu S. Activation of the KDM5A/miRNA-495/YTHDF2/m6A-MOB3B axis facilitates prostate cancer progression. J Exp Clin Cancer Res. 2020; 39(1): 223.

[94]

Wang L, Shilatifard A. UTX mutations in human cancer. Cancer Cell. 2019; 35(2): 168-176.

[95]

Duplaquet L, Li Y, Booker MA, et al. KDM6A epigenetically regulates subtype plasticity in small cell lung cancer. Nat Cell Biol. 2023; 25(9): 1346-1358.

[96]

Liu Q, Pang J, Wang LA, et al. Histone demethylase PHF8 drives neuroendocrine prostate cancer progression by epigenetically upregulating FOXA2. J Pathol. 2021; 253(1): 106-118.

[97]

Huang YH, Cai K, Xu PP, et al. CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct Target Ther. 2021; 6(1): 10.

[98]

Song Y, Li L, Ou Y, et al. Identification of genomic alterations in oesophageal squamous cell cancer. Nature. 2014; 509(7498): 91-95.

[99]

Ianculescu I, Wu DY, Siegmund KD, Stallcup MR. Selective roles for cAMP response element-binding protein binding protein and p300 protein as coregulators for androgen-regulated gene expression in advanced prostate cancer cells. J Biol Chem. 2012; 287(6): 4000-4013.

[100]

Jia D, Augert A, Kim DW, et al. Crebbp loss drives small cell lung cancer and increases sensitivity to HDAC inhibition. Cancer Discov. 2018; 8(11): 1422-1437.

[101]

Furlan T, Kirchmair A, Sampson N, et al. MYC-mediated ribosomal gene expression sensitizes enzalutamide-resistant prostate cancer cells to EP300/CREBBP inhibitors. Am J Pathol. 2021; 191(6): 1094-1107.

[102]

Zhao Z, Szczepanski AP, Tsuboyama N, et al. PAX9 determines epigenetic state transition and cell fate in cancer. Cancer Res. 2021; 81(18): 4696-4708.

[103]

Oh S, Koh J, Kim TM, et al. Transcriptomic heterogeneity of EGFR-mutant non-small cell lung cancer evolution towards small cell lung cancer. Clin Cancer Res. 2024.

[104]

Donati B, Lorenzini E, Ciarrocchi A. BRD4 and Cancer: going beyond transcriptional regulation. Mol Cancer. 2018; 17(1): 164.

[105]

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.

[106]

Fan L, Gong Y, He Y, et al. TRIM59 is suppressed by androgen receptor and acts to promote lineage plasticity and treatment-induced neuroendocrine differentiation in prostate cancer. Oncogene. 2023; 42(8): 559-571.

[107]

Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019; 574(7779): 575-580.

[108]

Wang D, Du G, Chen X, et al. Zeb1-controlled metabolic plasticity enables remodeling of chromatin accessibility in the development of neuroendocrine prostate cancer. Cell Death Differ. 2024; 31(6): 779-791.

[109]

He Y, Ji Z, Gong Y, et al. Numb/Parkin-directed mitochondrial fitness governs cancer cell fate via metabolic regulation of histone lactylation. Cell Rep. 2023; 42(2): 112033.

[110]

Li F, Si W, Xia L, et al. Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer. 2024; 23(1): 90.

[111]

Mirzaei S, Paskeh MDA, Okina E, et al. Molecular landscape of LncRNAs in prostate cancer: a focus on pathways and therapeutic targets for intervention. J Exp Clin Cancer Res. 2022; 41(1): 214.

[112]

Raveh E, Matouk IJ, Gilon M, Hochberg A. The H19 Long non-coding RNA in cancer initiation, progression and metastasis—a proposed unifying theory. Mol Cancer. 2015; 14: 184.

[113]

Ramnarine VR, Alshalalfa M, Mo F, et al. The long noncoding RNA landscape of neuroendocrine prostate cancer and its clinical implications. Gigascience. 2018; 7(6): giy050.

[114]

Singh N, Ramnarine VR, Song JH, et al. The long noncoding RNA H19 regulates tumor plasticity in neuroendocrine prostate cancer. Nat Commun. 2021; 12(1): 7349.

[115]

Huarte M, Guttman M, Feldser D, et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 2010; 142(3): 409-419.

[116]

Luo J, Wang K, Yeh S, et al. LncRNA-p21 alters the antiandrogen enzalutamide-induced prostate cancer neuroendocrine differentiation via modulating the EZH2/STAT3 signaling. Nat Commun. 2019; 10(1): 2571.

[117]

Bhagirath D, Liston M, Patel N, et al. MicroRNA determinants of neuroendocrine differentiation in metastatic castration-resistant prostate cancer. Oncogene. 2020; 39(49): 7209-7223.

[118]

Lovnicki J, Gan Y, Feng T, et al. LIN28B promotes the development of neuroendocrine prostate cancer. J Clin Invest. 2020; 130(10): 5338-5348.

[119]

Fernandes RC, Toubia J, Townley S, et al. Post-transcriptional gene regulation by microRNA-194 promotes neuroendocrine transdifferentiation in prostate cancer. Cell Rep. 2021; 34(1): 108585.

[120]

Lambert SA, Jolma A, Campitelli LF, et al. The human transcription factors. Cell. 2018; 172(4): 650-665.

[121]

Chen H, Thiagalingam A, Chopra H, et al. Conservation of the Drosophila lateral inhibition pathway in human lung cancer: a hairy-related protein (HES-1) directly represses achaete-scute homolog-1 expression. Proc Natl Acad Sci USA. 1997; 94(10): 5355-5360.

[122]

Sriuranpong V, Borges MW, Ravi RK, et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res. 2001; 61(7): 3200-3205.

[123]

Trigo J, Subbiah V, Besse B, et al. Lurbinectedin as second-line treatment for patients with small-cell lung cancer: a single-arm, open-label, phase 2 basket trial. Lancet Oncol. 2020; 21(5): 645-654.

[124]

Costanzo F, Martínez Diez M, Santamaría Nuñez G, et al. Promoters of ASCL1-and NEUROD1-dependent genes are specific targets of lurbinectedin in SCLC cells. EMBO Mol Med. 2022; 14(4): e14841.

[125]

Huang YH, Klingbeil O, He XY, et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Genes Dev. 2018; 32(13-14): 915-928.

[126]

Wu XS, He XY, Ipsaro JJ, et al. OCA-T1 and OCA-T2 are coactivators of POU2F3 in the tuft cell lineage. Nature. 2022; 607(7917): 169-175.

[127]

Szczepanski AP, Tsuboyama N, Watanabe J, Hashizume R, Zhao Z, Wang L. POU2AF2/C11orf53 functions as a coactivator of POU2F3 by maintaining chromatin accessibility and enhancer activity. Sci Adv. 2022; 8(40): eabq2403.

[128]

Baine MK, Febres-Aldana CA, Chang JC, et al. POU2F3 in SCLC: clinicopathologic and genomic analysis with a focus on its diagnostic utility in neuroendocrine-low SCLC. J Thorac Oncol. 2022; 17(9): 1109-1121.

[129]

Chakraborty S, Coleman C, Manoj P, et al. De novo and histologically transformed small-cell lung cancer is sensitive to lurbinectedin treatment through the modulation of EMT and NOTCH signaling pathways. Clin Cancer Res. 2023; 29(17): 3526-3540.

[130]

Brady NJ, Bagadion AM, Singh R, et al. Temporal evolution of cellular heterogeneity during the progression to advanced AR-negative prostate cancer. Nat Commun. 2021; 12(1): 3372.

[131]

Yu X, Gupta A, Wang Y, et al. Foxa1 and Foxa2 interact with the androgen receptor to regulate prostate and epididymal genes differentially. Ann N Y Acad Sci. 2005; 1061: 77-93.

[132]

Chiaverotti T, Couto SS, Donjacour A, et al. Dissociation of epithelial and neuroendocrine carcinoma lineages in the transgenic adenocarcinoma of mouse prostate model of prostate cancer. Am J Pathol. 2008; 172(1): 236-246.

[133]

Qi J, Nakayama K, Cardiff RD, et al. Siah2-dependent concerted activity of HIF and FoxA2 regulates formation of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell. 2010; 18(1): 23-38.

[134]

Wang Z, Townley SL, Zhang S, et al. FOXA2 rewires AP-1 for transcriptional reprogramming and lineage plasticity in prostate cancer. Nat Commun. 2024; 15(1): 4914.

[135]

Rotinen M, You S, Yang J, et al. ONECUT2 is a targetable master regulator of lethal prostate cancer that suppresses the androgen axis. Nat Med. 2018; 24(12): 1887-1898.

[136]

Guo H, Ci X, Ahmed M, et al. ONECUT2 is a driver of neuroendocrine prostate cancer. Nat Commun. 2019; 10(1): 278.

[137]

Yachida S, Totoki Y, Noë M, et al. Comprehensive genomic profiling of neuroendocrine carcinomas of the gastrointestinal system. Cancer Discov. 2022; 12(3): 692-711.

[138]

Lin SC, Chou YT, Jiang SS, et al. Epigenetic switch between SOX2 and SOX9 regulates cancer cell plasticity. Cancer Res. 2016; 76(23): 7036-7048.

[139]

Domenici G, Aurrekoetxea-Rodríguez I, Simões BM, et al. A Sox2-Sox9 signalling axis maintains human breast luminal progenitor and breast cancer stem cells. Oncogene. 2019; 38(17): 3151-3169.

[140]

Olsen RR, Ireland AS, Kastner DW, et al. ASCL1 represses a SOX9(+) neural crest stem-like state in small cell lung cancer. Genes Dev. 2021; 35(11-12): 847-869.

[141]

Liu Y, Zhuo S, Zhou Y, et al. Yap-Sox9 signaling determines hepatocyte plasticity and lineage-specific hepatocarcinogenesis. J Hepatol. 2022; 76(3): 652-664.

[142]

Deng S, Wang C, Wang Y, et al. Ectopic JAK-STAT activation enables the transition to a stem-like and multilineage state conferring AR-targeted therapy resistance. Nat Cancer. 2022; 3(9): 1071-1087.

[143]

Lo UG, Chen YA, Cen J, et al. The driver role of JAK-STAT signalling in cancer stemness capabilities leading to new therapeutic strategies for therapy-and castration-resistant prostate cancer. Clin Transl Med. 2022; 12(8): e978.

[144]

Shue YT, Drainas AP, Li NY, et al. A conserved YAP/Notch/REST network controls the neuroendocrine cell fate in the lungs. Nat Commun. 2022; 13(1): 2690.

[145]

Flores-Morales A, Bergmann TB, Lavallee C, et al. Proteogenomic characterization of patient-derived xenografts highlights the role of rest in neuroendocrine differentiation of castration-resistant prostate cancer. Clin Cancer Res. 2019; 25(2): 595-608.

[146]

Hong D, Knelson EH, Li Y, et al. Plasticity in the absence of NOTCH uncovers a RUNX2-dependent pathway in small cell lung cancer. Cancer Res. 2022; 82(2): 248-263.

[147]

Zhang X, Huang CT, Chen J, et al. Pax6 is a human neuroectoderm cell fate determinant. Cell Stem Cell. 2010; 7(1): 90-100.

[148]

Jing N, Du X, Liang Y, et al. PAX6 promotes neuroendocrine phenotypes of prostate cancer via enhancing MET/STAT5A-mediated chromatin accessibility. J Exp Clin Cancer Res. 2024; 43(1): 144.

[149]

George J, Walter V, Peifer M, et al. Integrative genomic profiling of large-cell neuroendocrine carcinomas reveals distinct subtypes of high-grade neuroendocrine lung tumors. Nat Commun. 2018; 9(1): 1048.

[150]

Koshkin VS, Garcia JA, Reynolds J, et al. Transcriptomic and protein analysis of small-cell bladder cancer (SCBC) identifies prognostic biomarkers and DLL3 as a relevant therapeutic target. Clin Cancer Res. 2019; 25(1): 210-221.

[151]

Puca L, Gavyert K, Sailer V, et al. Delta-like protein 3 expression and therapeutic targeting in neuroendocrine prostate cancer. Sci Transl Med. 2019; 11(484): eaav0891.

[152]

Hsu EC, Rice MA, Bermudez A, et al. Trop2 is a driver of metastatic prostate cancer with neuroendocrine phenotype via PARP1. Proc Natl Acad Sci USA. 2020; 117(4): 2032-2042.

[153]

Gray JE, Heist RS, Starodub AN, et al. Therapy of small cell lung cancer (SCLC) with a topoisomerase-i-inhibiting antibody-drug conjugate (ADC) targeting trop-2, sacituzumab govitecan. Clin Cancer Res. 2017; 23(19): 5711-5719.

[154]

Jing N, Zhang K, Chen X, et al. ADORA2A-driven proline synthesis triggers epigenetic reprogramming in neuroendocrine prostate and lung cancers. J Clin Invest. 2023; 133(24): e168670.

[155]

Chen WY, Wen YC, Lin SR, et al. Nerve growth factor interacts with CHRM4 and promotes neuroendocrine differentiation of prostate cancer and castration resistance. Commun Biol. 2021; 4(1): 22.

[156]

Wen YC, Tram VTN, Chen WH, et al. CHRM4/AKT/MYCN upregulates interferon alpha-17 in the tumor microenvironment to promote neuroendocrine differentiation of prostate cancer. Cell Death Dis. 2023; 14(5): 304.

[157]

Yasumizu Y, Rajabi H, Jin C, et al. MUC1-C regulates lineage plasticity driving progression to neuroendocrine prostate cancer. Nat Commun. 2020; 11(1): 338.

[158]

Bishop JL, Thaper D, Vahid S, et al. The master neural transcription factor BRN2 is an androgen receptor-suppressed driver of neuroendocrine differentiation in prostate cancer. Cancer Discov. 2017; 7(1): 54-71.

[159]

Ferone G, Song JY, Krijgsman O, et al. FGFR1 oncogenic activation reveals an alternative cell of origin of SCLC in Rb1/p53 mice. Cell Rep. 2020; 30(11): 3837-3850. e3833.

[160]

Nachmias B, Schimmer AD. Targeting nuclear import and export in hematological malignancies. Leukemia. 2020; 34(11): 2875-2886.

[161]

Liu S, Chai T, Garcia-Marques F, et al. UCHL1 is a potential molecular indicator and therapeutic target for neuroendocrine carcinomas. Cell Rep Med. 2024; 5(2): 101381.

[162]

Beltran H, Oromendia C, Danila DC, et al. A phase II trial of the aurora kinase A inhibitor alisertib for patients with castration-resistant and neuroendocrine prostate cancer: efficacy and biomarkers. Clin Cancer Res. 2019; 25(1): 43-51.

[163]

Owonikoko TK, Niu H, Nackaerts K, et al. Randomized phase II study of paclitaxel plus alisertib versus paclitaxel plus placebo as second-line therapy for SCLC: primary and correlative biomarker analyses. J Thorac Oncol. 2020; 15(2): 274-287.

[164]

Lim KH, Opyrchal M, Acharya A, et al. Phase 1 study combining alisertib with nab-paclitaxel in patients with advanced solid malignancies. Eur J Cancer. 2021; 154: 102-110.

[165]

Krebs MG, Delord JP, Jeffry Evans TR, et al. Olaparib and durvalumab in patients with relapsed small cell lung cancer (MEDIOLA): an open-label, multicenter, phase 1/2, basket study. Lung Cancer. 2023; 180: 107216.

[166]

Thomas A, Vilimas R, Trindade C, et al. Durvalumab in combination with olaparib in patients with relapsed SCLC: results from a phase II study. J Thorac Oncol. 2019; 14(8): 1447-1457.

[167]

Farago AF, Yeap BY, Stanzione M, et al. Combination olaparib and temozolomide in relapsed small-cell lung cancer. Cancer Discov. 2019; 9(10): 1372-1387.

[168]

de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020; 382(22): 2091-2102.

[169]

Antonarakis ES, Park SH, Goh JC, et al. Pembrolizumab plus olaparib for patients with previously treated and biomarker-unselected metastatic castration-resistant prostate cancer: the randomized, open-label, phase III KEYLYNK-010 trial. J Clin Oncol. 2023; 41(22): 3839-3850.

[170]

Abida W, Campbell D, Patnaik A, et al. Rucaparib for the treatment of metastatic castration-resistant prostate cancer associated with a DNA damage repair gene alteration: final results from the phase 2 TRITON2 study. Eur Urol. 2023; 84(3): 321-330.

[171]

Takahashi N, Hao Z, Villaruz LC, et al. Berzosertib plus topotecan vs topotecan alone in patients with relapsed small cell lung cancer: a randomized clinical trial. JAMA Oncol. 2023; 9(12): 1669-1677.

[172]

Bauer TM, Besse B, Martinez-Marti A, et al. Phase I, open-label, dose-escalation study of the safety, pharmacokinetics, pharmacodynamics, and efficacy of GSK2879552 in relapsed/refractory SCLC. J Thorac Oncol. 2019; 14(10): 1828-1838.

[173]

Noce B, Di Bello E, Fioravanti R, Mai A. LSD1 inhibitors for cancer treatment: focus on multi-target agents and compounds in clinical trials. Front Pharmacol. 2023; 14: 1120911.

[174]

Ferrari AC, Alumkal JJ, Stein MN, et al. Epigenetic therapy with panobinostat combined with bicalutamide rechallenge in castration-resistant prostate cancer. Clin Cancer Res. 2019; 25(1): 52-63.

[175]

Saunders LR, Bankovich AJ, Anderson WC, et al. A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci Transl Med. 2015; 7(302): 302ra136.

[176]

Rudin CM, Pietanza MC, Bauer TM, et al. Rovalpituzumab tesirine, a DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung cancer: a first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol. 2017; 18(1): 42-51.

[177]

Udagawa H, Akamatsu H, Tanaka K, et al. Phase I safety and pharmacokinetics study of rovalpituzumab tesirine in Japanese patients with advanced, recurrent small cell lung cancer. Lung Cancer. 2019; 135: 145-150.

[178]

Morgensztern D, Besse B, Greillier L, et al. Efficacy and safety of rovalpituzumab tesirine in third-line and beyond patients with DLL3-expressing, relapsed/refractory small-cell lung cancer: results from the phase II TRINITY study. Clin Cancer Res. 2019; 25(23): 6958-6966.

[179]

Blackhall F, Jao K, Greillier L, et al. Efficacy and safety of rovalpituzumab tesirine compared with topotecan as second-line therapy in DLL3-High SCLC: results from the phase 3 TAHOE study. J Thorac Oncol. 2021; 16(9): 1547-1558.

[180]

Malhotra J, Nikolinakos P, Leal T, et al. A phase 1–2 study of rovalpituzumab tesirine in combination with nivolumab plus or minus ipilimumab in patients with previously treated extensive-stage SCLC. J Thorac Oncol. 2021; 16(9): 1559-1569.

[181]

Johnson ML, Zvirbule Z, Laktionov K, et al. Rovalpituzumab tesirine as a maintenance therapy after first-line platinum-based chemotherapy in patients with extensive-stage-SCLC: results from the phase 3 MERU study. J Thorac Oncol. 2021; 16(9): 1570-1581.

[182]

Giffin MJ, Cooke K, Lobenhofer EK, et al. AMG 757, a half-life extended, DLL3-targeted bispecific T-cell engager, shows high potency and sensitivity in preclinical models of small-cell lung cancer. Clin Cancer Res. 2021; 27(5): 1526-1537.

[183]

Paz-Ares L, Champiat S, Lai WV, et al. Tarlatamab, a first-in-class DLL3-targeted bispecific T-cell engager, in recurrent small-cell lung cancer: an open-label, phase I study. J Clin Oncol. 2023; 41(16): 2893-2903.

[184]

Jaspers JE, Khan JF, Godfrey WD, et al. IL-18-secreting CAR T cells targeting DLL3 are highly effective in small cell lung cancer models. J Clin Invest. 2023; 133(9): e166028.

[185]

Shiba-Ishii A, Takemura N, Kawai H, Matsubara D. Histologic transformation of non-small-cell lung cancer in response to tyrosine kinase inhibitors: current knowledge of genetic changes and molecular mechanisms. Cancer Sci. 2024; 115(7): 2138-2146.

[186]

Li F, Yuan Q, Di W, et al. ERG orchestrates chromatin interactions to drive prostate cell fate reprogramming. J Clin Invest. 2020; 130(11): 5924-5941.

[187]

Feng M, Matoso A, Epstein G, et al. Identification of lineage-specific transcriptional factor-defined molecular subtypes in small cell bladder cancer. Eur Urol. 2023.

[188]

Zheng X, Zhuge J, Bezerra SM, et al. High frequency of TERT promoter mutation in small cell carcinoma of bladder, but not in small cell carcinoma of other origins. J Hematol Oncol. 2014; 7: 47.

[189]

Rudin CM, Poirier JT, Byers LA, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer. 2019; 19(5): 289-297.

[190]

Simpson KL, Stoney R, Frese KK, et al. A biobank of small cell lung cancer CDX models elucidates inter-and intratumoral phenotypic heterogeneity. Nat Cancer. 2020; 1(4): 437-451.

[191]

Owonikoko TK, Dwivedi B, Chen Z, et al. YAP1 expression in SCLC defines a distinct subtype with T-cell-inflamed phenotype. J Thorac Oncol. 2021; 16(3): 464-476.

[192]

Qu S, Fetsch P, Thomas A, et al. Molecular subtypes of primary SCLC tumors and their associations with neuroendocrine and therapeutic markers. J Thorac Oncol. 2022; 17(1): 141-153.

[193]

Chan JM, Quintanal-Villalonga Á, Gao VR, et al. Signatures of plasticity, metastasis, and immunosuppression in an atlas of human small cell lung cancer. Cancer Cell. 2021; 39(11): 1479-1496. e1418.

[194]

Ishioka K, Yasuda H, Hamamoto J, et al. Upregulation of FGF9 in lung adenocarcinoma transdifferentiation to small cell lung cancer. Cancer Res. 2021; 81(14): 3916-3929.

[195]

Ku SY, Wang Y, Garcia MM, et al. Notch signaling suppresses neuroendocrine differentiation and alters the immune microenvironment in advanced prostate cancer. J Clin Invest. 2024.

[196]

Pearson JD, Huang K, Pacal M, et al. Binary pan-cancer classes with distinct vulnerabilities defined by pro-or anti-cancer YAP/TEAD activity. Cancer Cell. 2021; 39(8): 1115-1134. e1112.

[197]

Wu Q, Guo J, Liu Y, et al. YAP drives fate conversion and chemoresistance of small cell lung cancer. Sci Adv. 2021; 7(40): eabg1850.

[198]

Rubin MA, Bristow RG, Thienger PD, Dive C, Imielinski M. Impact of lineage plasticity to and from a neuroendocrine phenotype on progression and response in prostate and lung cancers. Mol Cell. 2020; 80(4): 562-577.

[199]

Wu H, Yu Z, Liu Y, et al. Genomic characterization reveals distinct mutation landscapes and therapeutic implications in neuroendocrine carcinomas of the gastrointestinal tract. Cancer Commun (Lond). 2022; 42(12): 1367-1386.

[200]

Sanchez-Vega F, Mina M, Armenia J, et al. Oncogenic signaling pathways in the cancer genome atlas. Cell. 2018; 173(2): 321-337.

[201]

Bradley CA. SCNC phenotypic convergence reveals shared vulnerabilities. Nat Rev Cancer. 2019; 19(10): 543.

[202]

Altorki NK, Markowitz GJ, Gao D, et al. The lung microenvironment: an important regulator of tumour growth and metastasis. Nat Rev Cancer. 2019; 19(1): 9-31.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

127

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/