Molecular features of luminal breast cancer defined through spatial and single-cell transcriptomics

Ryohei Yoshitake , Hitomi Mori , Desiree Ha , Xiwei Wu , Jinhui Wang , Xiaoqiang Wang , Kohei Saeki , Gregory Chang , Hyun Jeong Shim , Yin Chan , Shiuan Chen

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (1) : e1548

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (1) : e1548 DOI: 10.1002/ctm2.1548
RESEARCH ARTICLE

Molecular features of luminal breast cancer defined through spatial and single-cell transcriptomics

Author information +
History +
PDF

Abstract

Background: Intratumour heterogeneity is a hallmark of most solid tumours, including breast cancers. We applied spatial transcriptomics and single-cell RNA-sequencing on patient-derived xenografts (PDXs) to profile spatially resolved cell populations within oestrogen receptor-positive (ER+) breast cancer and to elucidate their importance in oestrogen-dependent tumour growth.

Methods: Two PDXs of ‘ER-high’ breast cancers with opposite oestrogen-mediated growth responses were investigated: oestrogen-suppressed GS3 (80-100% ER) and oestrogen-dependent SC31 (40-90% ER) models. The observation was validated via single-cell analyses on an ‘ER-low’ PDX, GS1 (5% ER). The results from our spatial and single-cell analyses were further supported by a public ER+ breast cancer single-cell dataset and protein-based dual immunohistochemistry (IHC) of SC31 examining important luminal cancer markers (i.e., ER, progesterone receptor and Ki67). The translational implication of our findings was assessed by clinical outcome analyses on publicly available cohorts.

Results: Our space-gene-function study revealed four spatially distinct compartments within ER+ breast cancers. These compartments showed functional diversity (oestrogen-responsive, proliferative, hypoxia-induced and inflammation-related). The ‘proliferative’ population, rather than the ‘oestrogen-responsive’ compartment, was crucial for oestrogen-dependent tumour growth, leading to the acquisition of luminal B-like features. The cells expressing typical oestrogen-responsive genes like PGR were not directly linked to oestrogen-dependent proliferation. Dual IHC analyses demonstrated the distinct contribution of the Ki67+ proliferative cells toward oestrogen-mediated growth and their response to a CDK4/6 inhibitor. The gene signatures derived from the proliferative, hypoxia-induced and inflammation-related compartments were significantly correlated with worse clinical outcomes, while patients with the oestrogen-responsive signature showed better prognoses, suggesting that this compartment would not be directly associated with oestrogen-dependent tumour progression.

Conclusions: Our study identified the gene signature in our ‘proliferative’ compartment as an important determinant of luminal cancer subtypes. This ‘proliferative’ cell population is a causative feature of luminal B breast cancer, contributing toward its aggressive behaviours.

Keywords

breast cancer / oestrogen receptor / intratumour heterogeneity / single-cell RNA-sequencing / spatial transcriptomics

Cite this article

Download citation ▾
Ryohei Yoshitake, Hitomi Mori, Desiree Ha, Xiwei Wu, Jinhui Wang, Xiaoqiang Wang, Kohei Saeki, Gregory Chang, Hyun Jeong Shim, Yin Chan, Shiuan Chen. Molecular features of luminal breast cancer defined through spatial and single-cell transcriptomics. Clinical and Translational Medicine, 2024, 14(1): e1548 DOI:10.1002/ctm2.1548

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Perou CM, Sørile T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747-752.

[2]

Sørlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001;98(19):10869-10874.

[3]

The Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61-70.

[4]

Gnant M, Harbeck N, Thomssen C. St. Gallen 2011: summary of the consensus discussion. Breast Care. 2011;6(2):136-141.

[5]

Ades F, Zardavas D, Bozovic-Spasojevic I, et al. Luminal B breast cancer: molecular characterization, clinical management, and future perspectives. J Clin Oncol. 2014;32(25):2794-2803.

[6]

Hanker AB, Sudhan DR, Arteaga CL. Overcoming endocrine resistance in breast cancer. Cancer Cell. 2020;37(4):496-513.

[7]

Iwamoto T, Booser D, Valero V, et al. Estrogen receptor (ER) mRNA and ER-related gene expression in breast cancers that are 1% to 10% ER-positive by immunohistochemistry. J Clin Oncol. 2012;30(7):729-734.

[8]

Allred DC, Carlson RW, Berry DA, et al. NCCN Task Force report: estrogen receptor and progesterone receptor testing in breast cancer by immunohistochemistry. J Natl Compr Canc Netw. 2009;7(6):S1-S21. Suppl_.

[9]

Allison KH, Hammond MEH, Dowsett M, et al. Estrogen and progesterone receptor testing in breast cancer: aSCO/CAP guideline update. J Clin Oncol. 2020;38(12):1346-1366.

[10]

Marusyk A, Janiszewska M, Polyak K. Intratumor heterogeneity: the Rosetta stone of therapy resistance. Cancer Cell. 2020;37(4):471-484.

[11]

Turner KM, Yeo SK, Holm TM, et al. Heterogeneity within molecular subtypes of breast cancer. Am J Physiol Cell Physiol. 2021;321(2):C343-C354.

[12]

Koren S, Bentires-Alj M. Breast tumor heterogeneity: source of fitness, hurdle for therapy. Mol Cell. 2015;60(4):537-546.

[13]

Kanaya N, Somlo G, Wu J, et al. Characterization of patient-derived tumor xenografts (PDXs) as models for estrogen receptor positive (ER+HER2 and ER+HER2+) breast cancers. J Steroid Biochem Mol Biol. 2017;170:65-74.

[14]

Mori H, Saeki K, Chang G, et al. Influence of estrogen treatment on ESR1+ and ESR1 cells in ER+ breast cancer: insights from single-cell analysis of patient-derived xenograft models. Cancers (Basel). 2021;13(24):6375.

[15]

Hsu PY, Wu VS, Kanaya N, et al. Dual mTOR kinase inhibitor MLN0128 sensitize HR+/HER2+ breast cancer patient-derived xenografts to trastuzumab or fulvestrant. Clin Cancer Res. 2018;24(2):395-406.

[16]

Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184(13):3573-3587.e29.

[17]

Korthauer KD, Chu LF, Newton MA, et al. A statistical approach for identifying differential distributions in single-cell RNA-seq experiments. Genome Biol. 2016;17(1):1-15.

[18]

DeTomaso D, Jones MG, Subramaniam M, et al. Functional interpretation of single cell similarity maps. Nat Commun. 2019;10(1):4376.

[19]

Liberzon A, Birger C, Thorvaldsdóttir H, et al. The molecular signatures database hallmark gene set collection. Cell Syst. 2015;1(6):417-425.

[20]

Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847-2849.

[21]

Wu SZ, Al-Eryani G, Roden DL, et al. A single-cell and spatially resolved atlas of human breast cancers. Nat Genet. 2021;53(9):1334-1347.

[22]

Aibar S, González-Blas CB, Moerman T, et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods. 2017;14(11):1083-1086.

[23]

Saeki K, Chang G, Kanaya N, et al. Mammary cell gene expression atlas links epithelial cell remodeling events to breast carcinogenesis. Commun Biol. 2021;4(1):660.

[24]

Bankhead P, Loughrey MB, Fernández JA, et al. QuPath: open source software for digital pathology image analysis. Sci Rep. 2017;7(1):16878.

[25]

Curtis C, Shah SP, Chin SF, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486(7403):346-352.

[26]

Cerami E, Gao J, Dogrusoz U, et al. The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401-404.

[27]

Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1.

[28]

Ramos M, Geistlinger L, Oh S, et al. Multiomic integration of public oncology databases in Bioconductor. JCO Clin Cancer Inform. 2020;4:958-971.

[29]

Sinn BV, Fu C, Lau R, et al. SETER/PR: a robust 18-gene predictor for sensitivity to endocrine therapy for metastatic breast cancer. NPJ Breast Cancer. 2019;5:16.

[30]

Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-Seq data. BMC Bioinformatics. 2013;14:7.

[31]

Hoadley KA, Yau C, Hinoue T, et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell. 2018;173(2):291-304.e6.

[32]

Yoshida GJ. Applications of patient-derived tumor xenograft models and tumor organoids. J Hematol Oncol. 2020;13(1):4.

[33]

Saul D, Kosinsky RL. Spatial transcriptomics herald a new era of transcriptome research. Clin Transl Med. 2023;13(5):e1264.

[34]

Ståhl PL, Salmén F, Vickovic S, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353(6294):78-82.

[35]

Chen R, Qarmali M, Siegal GP, et al. Receptor conversion in metastatic breast cancer: analysis of 390 cases from a single institution. Mod Pathol. 2020;33(12):2499-2506.

[36]

Schwartz GN, Kaufman PA, Giridhar KV, et al. Alternating 17β-Estradiol and aromatase inhibitor therapies is efficacious in postmenopausal women with advanced endocrine-resistant ER+ breast cancer. Clin Cancer Res. 2023;29(15):2767-2773.

[37]

Tentler JJ, Tan AC, Weekes CD, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol. 2012;9(6):338-350.

[38]

Zhou J, Zhu M, Wang Q, et al. SERPINA3-ANKRD11-HDAC3 pathway induced aromatase inhibitor resistance in breast cancer can be reversed by HDAC3 inhibition. Commun Biol. 2023;6(1):695.

[39]

Wang Y, Zhou D, Phung S, et al. SGK3 is an estrogen-inducible kinase promoting estrogen-mediated survival of breast cancer cells. Mol Endocrinol. 2011;25(1):72-82.

[40]

Finlin BS, Gau CL, Murphy GA, et al. RERG is a novel Ras-related, estrogen-regulated and growth-inhibitory gene in breast cancer. J Biol Chem. 2001;276(45):42259-42267.

[41]

Wang Y, Chen S. TXNIP links anticipatory unfolded protein response to estrogen reprogramming glucose metabolism in breast cancer cells. Endocrinology. 2022;163(1):bqab212.

[42]

Cadenas C, Franckenstein D, Schmidt M, et al. Role of thioredoxin reductase 1 and thioredoxin interacting protein in prognosis of breast cancer. Breast Cancer Res. 2010;12(3):R44.

[43]

Habashy HO, Powe DG, Glaab E, et al. RERG (Ras-like, oestrogen-regulated, growth-inhibitor) expression in breast cancer: a marker of ER-positive luminal-like subtype. Breast Cancer Res Treat. 2011;128(2):315-326.

[44]

Xu J, Chen Y, Olopade OI. MYC and breast cancer. Genes Cancer. 2010;1(6):629-640.

[45]

Lu X, Kang Y. Hypoxia and hypoxia-inducible factors: master regulators of metastasis. Clin Cancer Res. 2010;16(24):5928-5935.

[46]

De Nonneville A, Finetti P, Mamessier E, et al. RE: nDRG1 in aggressive breast cancer progression and brain metastasis. J Natl Cancer Inst. 2022;114(7):1046-1047.

[47]

Post AEM, Smid M, Nagelkerke A, et al. Interferon-stimulated genes are involved in cross-resistance to radiotherapy in tamoxifen-resistant breast cancer. Clin Cancer Res. 2018;24(14):3397-3408.

[48]

De Angelis C, Fu X, Cataldo ML, et al. Activation of the IFN signaling pathway is associated with resistance to CDK4/6 inhibitors and immune checkpoint activation in ER-positive breast cancer. Clin Cancer Res. 2021;27(17):4870-4882.

[49]

Fu X, De Angelis C, Schiff R. Interferon signaling in estrogen receptor-positive breast cancer: a revitalized topic. Endocrinology. 2022;163(1):bqab235.

[50]

Uxa S, Castillo-Binder P, Kohler R, et al. Ki-67 gene expression. Cell Death Differ. 2021;28(12):3357-3370.

[51]

Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, a phase II study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR+/HER2 metastatic breast cancer. Clin Cancer Res. 2017;23(17):5218-5224.

[52]

Petrossian K, Kanaya N, Lo C, et al. ERα-mediated cell cycle progression is an important requisite for CDK4/6 inhibitor response in HR+ breast cancer. Oncotarget. 2018;9(45):27736-27751.

[53]

Liu F, Liu X, Powell CA, et al. Initiative of clinical single-cell biomedicine in clinical and translational medicine. Clin Transl Med. 2023;13(1):e1173.

[54]

Rusidzé M, Adlanmérini M, Chantalat E, et al. Estrogen receptor-α signaling in post-natal mammary development and breast cancers. Cell Mol Life Sci. 2021;78(15):5681-5705.

[55]

Platet N, Cathiard AM, Gleizes M, et al. Estrogens and their receptors in breast cancer progression: a dual role in cancer proliferation and invasion. Crit Rev Oncol Hematol. 2004;51(1):55-67.

[56]

Horwitz KB, Sartorius CA. 90 YEARS OF PROGESTERONE: progesterone and progesterone receptors in breast cancer: past, present, future. J Mol Endocrinol. 2020;65(1):T49-T63.

[57]

McBryan J, Howlin J, Napoletano S, et al. Amphiregulin: role in mammary gland development and breast cancer. J Mammary Gland Biol Neoplasia. 2008;13(2):159-169.

[58]

Wang Y, Tzeng YDT, Chang G, et al. Amphiregulin retains ERα expression in acquired aromatase inhibitor resistant breast cancer cells. Endocr Relat Cancer. 2020;27(12):671-683.

[59]

Scabia V, Ayyanan A, De Martino F, et al. Estrogen receptor positive breast cancers have patient specific hormone sensitivities and rely on progesterone receptor. Nat Commun. 2022;13(1):3127.

[60]

Mohammed H, Russell IA, Stark R, et al. Progesterone receptor modulates ERα action in breast cancer. Nature. 2015;523(7560):313-317.

[61]

Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and comparison with the genomic health recurrence score in early breast cancer. J Clin Oncol. 2011;29(32):4273-4278.

[62]

Sistigu A, Yamazaki T, Vacchelli E, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20(11):1301-1309.

[63]

Ariazi EA, Cunliffe HE, Lewis-Wambi JS, et al. Estrogen induces apoptosis in estrogen deprivation-resistant breast cancer through stress responses as identified by global gene expression across time. Proc Natl Acad Sci USA. 2011;108(47):18879-18886.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

197

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/