Analysis of single nuclear chromatin accessibility reveals unique myeloid populations in human pancreatic ductal adenocarcinoma

Hillary G. Pratt , Li Ma , Sebastian A. Dziadowicz , Sascha Ott , Thomas Whalley , Barbara Szomolay , Timothy D. Eubank , Gangqing Hu , Brian A. Boone

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1595

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1595 DOI: 10.1002/ctm2.1595
RESEARCH ARTICLE

Analysis of single nuclear chromatin accessibility reveals unique myeloid populations in human pancreatic ductal adenocarcinoma

Author information +
History +
PDF

Abstract

Background: A better understanding of the pancreatic ductal adenocarcinoma (PDAC) immune microenvironment is critical to developing new treatments and improving outcomes. Myeloid cells are of particular importance for PDAC progression; however, the presence of heterogenous subsets with different ontogeny and impact, along with some fluidity between them, (infiltrating monocytes vs. tissue-resident macrophages; M1 vs. M2) makes characterisation of myeloid populations challenging. Recent advances in single cell sequencing technology provide tools for characterisation of immune cell infiltrates, and open chromatin provides source and function data for myeloid cells to assist in more comprehensive characterisation. Thus, we explore single nuclear assay for transposase accessible chromatin (ATAC) sequencing (snATAC-Seq), a method to analyse open gene promoters and transcription factor binding, as an important means for discerning the myeloid composition in human PDAC tumours.

Methods: Frozen pancreatic tissues (benign or PDAC) were prepared for snATAC-Seq using 10× Chromium technology. Signac was used for preliminary analysis, clustering and differentially accessible chromatin region identification. The genes annotated in promoter regions were used for Gene Ontology (GO) enrichment and cell type annotation. Gene signatures were used for survival analysis with The Cancer Genome Atlas (TCGA)-pancreatic adenocarcinoma (PAAD) dataset.

Results: Myeloid cell transcription factor activities were higher in tumour than benign pancreatic samples, enabling us to further stratify tumour myeloid populations. Subcluster analysis revealed eight distinct myeloid populations. GO enrichment demonstrated unique functions for myeloid populations, including interleukin-1b signalling (recruited monocytes) and intracellular protein transport (dendritic cells). The identified gene signature for dendritic cells influenced survival (hazard ratio = .63, p = .03) in the TCGA-PAAD dataset, which was unique to PDAC.

Conclusions: These data suggest snATAC-Seq as a method for analysis of frozen human pancreatic tissues to distinguish myeloid populations. An improved understanding of myeloid cell heterogeneity and function is important for developing new treatment targets in PDAC.

Keywords

ATAC-Seq / myeloid / PDAC

Cite this article

Download citation ▾
Hillary G. Pratt, Li Ma, Sebastian A. Dziadowicz, Sascha Ott, Thomas Whalley, Barbara Szomolay, Timothy D. Eubank, Gangqing Hu, Brian A. Boone. Analysis of single nuclear chromatin accessibility reveals unique myeloid populations in human pancreatic ductal adenocarcinoma. Clinical and Translational Medicine, 2024, 14(3): e1595 DOI:10.1002/ctm2.1595

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Fan J-Q, Wang M-F, Chen H-L, Shang D, Das J-K, Song J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol Cancer. 2020;19(1):32.

[2]

Sanford DE, Belt BA, Panni RZ, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res. 2013;19(13):3404-3415.

[3]

Nywening TM, Wang-Gillam A, Sanford DE, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016;17(5):651-662.

[4]

Noel M, O'Reilly EM, Wolpin BM, et al. Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 2020;38(3):800-811.

[5]

Zhu Y, Herndon JM, Sojka DK, et al. Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity. 2017;47(2):323-338.e6.

[6]

Laviron M, Boissonnas A. Ontogeny of tumor-associated macrophages. Front Immunol. 2019;10:1799.

[7]

Fan X, Lu P, Wang H, et al. Integrated single-cell multiomics analysis reveals novel candidate markers for prognosis in human pancreatic ductal adenocarcinoma. Cell Discov. 2022;8(1):13.

[8]

Chen K, Wang Q, Li M, et al. Single-cell RNA-seq reveals dynamic change in tumor microenvironment during pancreatic ductal adenocarcinoma malignant progression. EBioMedicine. 2021;66:103315.

[9]

Li J, Byrne KT, Yan F, et al. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity. 2018;49(1):178-193.e7.

[10]

Zhang D, Wu S, Pan S, et al. Single-cell sequencing reveals heterogeneity between pancreatic adenosquamous carcinoma and pancreatic ductal adenocarcinoma with prognostic value. Front Immunol. 2022;13:972298.

[11]

Werba G, Weissinger D, Kawaler EA, et al. Single-cell RNA sequencing reveals the effects of chemotherapy on human pancreatic adenocarcinoma and its tumor microenvironment. Nat Commun. 2023;14(1):797.

[12]

Yang K, Yang T, Yu J, Li F, Zhao X. Integrated transcriptional analysis reveals macrophage heterogeneity and macrophage-tumor cell interactions in the progression of pancreatic ductal adenocarcinoma. BMC Cancer. 2023;23(1):199.

[13]

Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62(1):112-120.

[14]

Lin W, Noel P, Borazanci EH, et al. Single-cell transcriptome analysis of tumor and stromal compartments of pancreatic ductal adenocarcinoma primary tumors and metastatic lesions. Genome Med. 2020;12(1):80.

[15]

Peng J, Sun BF, Chen CY, et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res. 2019;29(9):725-738.

[16]

Pan Y, Lu F, Fei Q, et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancreatic cancer. J Hematol OncolJ Hematol Oncol. 2019;12(1):124.

[17]

Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr Protoc Mol Biol. 2015;109(1).

[18]

Corces MR, Trevino A, Hamilton EG, et al. Omni-ATAC-seq: improved ATAC-seq protocol. Protoc Exch. 2017.

[19]

Lavin Y, Winter D, Blecher-Gonen R, et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell. 2014;159(6):1312-1326.

[20]

Li Y, He Y, Peng J, et al. Mutant Kras co-opts a proto-oncogenic enhancer network in inflammation-induced metaplastic progenitor cells to initiate pancreatic cancer. Nat Cancer. 2020;2(1):49-65.

[21]

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

[22]

Stuart T, Srivastava A, Madad S, Lareau CA, Satija R. Single-cell chromatin state analysis with Signac. Nat Methods. 2021;18(11):1333-1341.

[23]

Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with harmony. Nat Methods. 2019;16(12):1289-1296.

[24]

Waltman L, Van Eck NJ. A smart local moving algorithm for large-scale modularity-based community detection. Eur Phys J B. 2013;86(11):471.

[25]

Rygiel AM, Unger LS, Sörgel FL, et al. Variants in the pancreatic CUB and zona pellucida-like domains 1 (CUZD1) gene in early-onset chronic pancreatitis—a possible new susceptibility gene. Pancreatology. 2022;22(5):564-571.

[26]

Prévot PP, Simion A, Grimont A, et al. Role of the ductal transcription factors HNF6 and Sox9 in pancreatic acinar-to-ductal metaplasia. Gut. 2012;61(12):1723-1732.

[27]

Buckle A, Nozawa R-S, Kleinjan DA, Gilbert N. Functional characteristics of novel pancreatic Pax6 regulatory elements. Hum Mol Genet. 2018;27(19):3434-3448.

[28]

Katsuta E, Qi Q, Peng X, Hochwald SN, Yan L, Takabe K. Pancreatic adenocarcinomas with mature blood vessels have better overall survival. Sci Rep. 2019;9(1):1310.

[29]

Elyada E, Bolisetty M, Laise P, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019;9(8):1102-1123.

[30]

Bayne LJ, Beatty GL, Jhala N, et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell. 2012;21(6):822-835.

[31]

Poirier SJ, Boudreau LH, Flamand N, Surette ME. LPS induces ALOX5 promoter activation and 5-lipoxygenase expression in human monocytic cells. Prostaglandins Leukot Essent Fatty Acids. 2020;154:102078.

[32]

Awad F, Assrawi E, Jumeau C. Impact of human monocyte and macrophage polarization on NLR expression and NLRP3 inflammasome activation. PLoS One. 2017;12(4):e0175336.

[33]

Kumanogoh A, Marukawa S, Suzuki K, et al. Class IV semaphorin Sema4A enhances T-cell activation and interacts with Tim-2. Nature. 2002;419(6907):629-633.

[34]

Lu N, Li Y, Zhang Z, et al. Human Semaphorin-4A drives Th2 responses by binding to receptor ILT-4. Nat Commun. 2018;9(1):742.

[35]

van Rijn A, Paulis L, te Riet J, et al. Semaphorin 7A promotes chemokine-driven dendritic cell migration. J Immunol. 2016;196(1):459-468.

[36]

Meyer MA, Baer JM, Knolhoff BL, et al. Breast and pancreatic cancer interrupt IRF8-dependent dendritic cell development to overcome immune surveillance. Nat Commun. 2018;9(1):1250.

[37]

Neophytou CM, Pierides C, Christodoulou MI, Costeas P, Kyriakou TC, Papageorgis P. The role of tumor-associated myeloid cells in modulating cancer therapy. Front Oncol. 2020;10:899.

[38]

Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162-174.

[39]

Schep AN, Wu B, Buenrostro JD, Greenleaf WJ. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat Methods. 2017;14(10):975-978.

[40]

Zhou Y, Zhou B, Pache L, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523.

[41]

Dwivedi B, Mumme H, Satpathy S, Bhasin SS, Bhasin M, Survival Genie, a web platform for survival analysis across pediatric and adult cancers. Sci Rep. 2022;12(1):3069.

[42]

Sun K, Mylavarapu C, Crenshaw A, et al. Pancreatic head vs pancreatic body/tail cancer: are they different? World J Gastrointest Oncol. 2022;14(3):716-723.

[43]

Zhang J, Song J, Tang S, et al. Multi-omics analysis reveals the chemoresistance mechanism of proliferating tissue-resident macrophages in PDAC via metabolic adaptation. Cell Rep. 2023;42(6):112620.

[44]

Williams JA. Regulation of normal and adaptive pancreatic growth. Pancreapedia: Exocrine Pancreas Knowledge Base. American Pancreatic Association.

[45]

Hoang CQ, Hale MA, Azevedo-Pouly AC, et al. Transcriptional maintenance of pancreatic acinar identity, differentiation, and homeostasis by PTF1A. Mol Cell Biol. 2016;36(24):3033-3047.

[46]

Cebola I, Rodríguez-Seguí SA, Cho CHH, et al. TEAD and YAP regulate the enhancer network of human embryonic pancreatic progenitors. Nat Cell Biol. 2015;17(5):615-626.

[47]

Eferl R, Wagner EF. AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer. 2003;3(11):859-868.

[48]

Park J, Eisenbarth D, Choi W, et al. YAP and AP-1 cooperate to initiate pancreatic cancer development from ductal cells in mice.

[49]

Wortham M, Sander M. Transcriptional mechanisms of pancreatic β-cell maturation and functional adaptation. Trends Endocrinol Metab. 2021;32(7):474-487.

[50]

De Val S, Black BL. Transcriptional control of endothelial cell development. Dev Cell. 2009;16(2):180-195.

[51]

Wei L, Lin Q, Lu Y, et al. Cancer-associated fibroblasts-mediated ATF4 expression promotes malignancy and gemcitabine resistance in pancreatic cancer via the TGF-β1/SMAD2/3 pathway and ABCC1 transactivation. Cell Death Dis. 2021;12(4):334.

[52]

Bhat NK, Thompson CB, Lindsten T, et al. Reciprocal expression of human ETS1 and ETS2 genes during T-cell activation: regulatory role for the protooncogene ETS1. Proc Natl Acad Sci U S A. 1990;87(10):3723-3727.

[53]

Friedman AD. C/EBPα induces PU.1 and interacts with AP-1 and NF-κB to regulate myeloid development. Blood Cells Mol Dis. 2007;39(3):340-343.

[54]

Song H, Dong M, Zhou J, Sheng W, Li X, Gao W. Expression and prognostic significance of TRPV6 in the development and progression of pancreatic cancer. Oncol Rep. 2018;39(3):1432-1440.

[55]

Wang H, Chen L, Qi L, et al. A single-cell atlas of tumor-infiltrating immune cells in pancreatic ductal adenocarcinoma. Mol Cell Proteomics. 2022;21(8):100258.

[56]

Lkhagvasuren B, Mee-Inta O, Zhao ZW, Hiramoto T, Boldbaatar D, Kuo YM. Pancreas–brain crosstalk. Front Neuroanat. 2021;15:691777.

[57]

Pollard TD, Cooper JA. Actin, a central player in cell shape and movement. Science. 2009;326(5957):1208-1212.

[58]

Qu X, Nyeng P, Xiao F, Dorantes J, Jensen J. Growth factor independence-1 (Gfi1) is required for pancreatic acinar unit formation and centroacinar cell differentiation. Cell Mol Gastroenterol Hepatol. 2015;1(2):233-247.e1.

[59]

Patzek S, Liu Z, De La OS, et al. Loss of Fgf9 in mice leads to pancreatic hypoplasia and asplenia. iScience. 2023;26(4):106500.

[60]

Guillemot F, Zimmer C. From cradle to grave: the multiple roles of fibroblast growth factors in neural development. Neuron. 2011;71(4):574-588.

[61]

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

[62]

Ediger BN, Du A, Liu J, et al. Islet-1 is essential for pancreatic B-cell function. 2014;63.

[63]

Tixi W, Maldonado M, Chang YT, et al. Coordination between ECM and cell-cell adhesion regulates the development of islet aggregation, architecture, and functional maturation. eLife. 2023;12:e90006.

[64]

Rashid M, Olson EC. Delayed cortical development in mice with a neural specific deletion of β1 integrin. Front Neurosci. 2023;17:1158419.

[65]

Du X, Yi X, Zou X, et al. PCDH1, a poor prognostic biomarker and potential target for pancreatic adenocarcinoma metastatic therapy. BMC Cancer. 2023;23(1):1102.

[66]

Ye Z, Yang Y, Wei Y, Li L, Wang X, Zhang J. PCDH1 promotes progression of pancreatic ductal adenocarcinoma via activation of NF-κB signalling by interacting with KPNB1. Cell Death Dis. 2022;13(7):633.

[67]

Tsuji S, Washimi K, Kageyama T, et al. HEG1 is a novel mucin-like membrane protein that serves as a diagnostic and therapeutic target for malignant mesothelioma. Sci Rep. 2017;7(1):45768.

[68]

Long Y, Wu J, Shen Y, et al. CAPG is a novel biomarker for early gastric cancer and is involved in the Wnt/β-catenin signaling pathway. Cell Death Discov. 2024;10(1):15.

[69]

Xiang X, Wang Y, Zhang H, et al. Vasodilator-stimulated phosphoprotein promotes liver metastasis of gastrointestinal cancer by activating a β1-integrin-FAK-YAP1/TAZ signaling pathway. Npj Precis Oncol. 2018;2(1):2.

[70]

Fukumura D, Gohongi T, Kadambi A, et al. Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proc Natl Acad Sci U S A. 2001;98(5):2604-2609.

[71]

Lampson BL, Kendall SD, Ancrile BB, et al. Targeting eNOS in pancreatic cancer. Cancer Res. 2012;72(17):4472-4482.

[72]

Murakami M, Nguyen LT, Zhang ZW, et al. The FGF system has a key role in regulating vascular integrity. J Clin Invest. 2008;118(10):3355-3366.

[73]

Bulle A, Lim KH. Beyond just a tight fortress: contribution of stroma to epithelial-mesenchymal transition in pancreatic cancer. Signal Transduct Target Ther. 2020;5(1):249.

[74]

Alcaraz LB, Mallavialle A, Mollevi C, et al. SPARC in cancer-associated fibroblasts is an independent poor prognostic factor in non-metastatic triple-negative breast cancer and exhibits pro-tumor activity. Int J Cancer. 2023;152(6):1243-1258.

[75]

Shah K, Al-Haidari A, Sun J, Kazi JU. T cell receptor (TCR) signaling in health and disease. Signal Transduct Target Ther. 2021;6(1):412.

[76]

Liu S, Zhou B, Wu L, Sun Y, Chen J, Liu S. Single-cell differential splicing analysis reveals high heterogeneity of liver tumor-infiltrating T cells. Sci Rep. 2021;11(1):5325.

[77]

Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821-832.

[78]

Daley D, Mani VR, Mohan N, et al. NLRP3 signaling drives macrophage-induced adaptive immune suppression in pancreatic carcinoma. J Exp Med. 2017;214(6):1711-1724.

[79]

Trovato R, Fiore A, Sartori S, et al. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3. J Immunother Cancer. 2019;7(1):255.

[80]

Von Moltke J, Trinidad NJ, Moayeri M, et al. Rapid induction of inflammatory lipid mediators by the inflammasome in vivo. Nature. 2012;490(7418):107-111.

[81]

Menon V, Ghaffari S. Transcription factors FOXO in the regulation of homeostatic hematopoiesis. Curr Opin Hematol. 2018;25(4):290-298.

[82]

Brykczynska U, Geigges M, Wiedemann SJ, et al. Distinct transcriptional responses across tissue-resident macrophages to short-term and long-term metabolic challenge. Cell Rep. 2020;30(5):1627-1643.e7.

[83]

Calderon B, Carrero JA, Ferris ST, et al. The pancreas anatomy conditions the origin and properties of resident macrophages. J Exp Med. 2015;212(10):1497-1512.

[84]

Ghislat G, Cheema AS, Baudoin E, et al. NF-κB-dependent IRF1 activation programs cDC1 dendritic cells to drive antitumor immunity. Sci Immunol. 2021;6(61):eabg3570.

[85]

Tailor P, Tamura T, Ozato K. IRF family proteins and type I interferon induction in dendritic cells. Cell Res. 2006;16(2):134-140.

[86]

Kurotaki D, Nakabayashi J, Nishiyama A, et al. Transcription factor IRF8 governs enhancer landscape dynamics in mononuclear phagocyte progenitors. Cell Rep. 2018;22(10):2628-2641.

[87]

Alam Z, Devalaraja S, Li M, et al. Counter regulation of Spic by NF-κB and STAT signaling controls inflammation and iron metabolism in macrophages. Cell Rep. 2020;31(13):107825.

[88]

Ma RY, Black A, Qian BZ. Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol. 2022;43(7):546-563.

[89]

Suzuki E, Williams S, Sato S, Gilkeson G, Watson DK, Zhang XK. The transcription factor Fli-1 regulates monocyte, macrophage and dendritic cell development in mice. Immunology. 2013;139(3):318-327.

[90]

Fontana MF, Baccarella A, Pancholi N, Pufall MA, Herbert DR, Kim CC. JUNB is a key transcriptional modulator of macrophage activation. J Immunol. 2015;194(1):177-186.

[91]

Alshetaiwi H, Pervolarakis N, McIntyre LL, et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci Immunol. 2020;5(44):eaay6017.

[92]

Waight JD, Netherby C, Hensen ML, et al. Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis. J Clin Invest. 2013;123(10):4464-4478.

[93]

Saclier M, Angelini G, Bonfanti C, Mura G, Temponi G, Messina G. Selective ablation of Nfix in macrophages attenuates muscular dystrophy by inhibiting fibro-adipogenic progenitor-dependent fibrosis. J Pathol. 2022;257(3):352-366.

[94]

Adiko AC, Babdor J, Gutiérrez-Martínez E, Guermonprez P, Saveanu L. Intracellular transport routes for MHC I and their relevance for antigen cross-presentation. Front Immunol. 2015;6:335.

[95]

Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, Beatty GL. IFNγ and CCL2 cooperate to redirect tumor-infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma. Cancer Discov. 2016;6(4):400-413.

[96]

Xing J, Weng L, Yuan B, et al. Identification of a role for TRIM29 in the control of innate immunity in the respiratory tract. Nat Immunol. 2016;17(12):1373-1380.

[97]

Masuda Y, Takahashi H, Sato S, et al. TRIM29 regulates the assembly of DNA repair proteins into damaged chromatin. Nat Commun. 2015;6(1):7299.

[98]

Dhara S, Chhangawala S, Chintalapudi H, et al. Pancreatic cancer prognosis is predicted by an ATAC-array technology for assessing chromatin accessibility. Nat Commun. 2021;12(1):3044.

[99]

Hegde S, Krisnawan VE, Herzog BH, et al. Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell. 2020;37(3):289-307.

[100]

Byrne KT, Betts CB, Mick R, et al. Neoadjuvant selicrelumab, an agonist CD40 antibody, induces changes in the tumor microenvironment in patients with resectable pancreatic cancer. Clin Cancer Res. 2021;27(16):4574-4586.

[101]

Ghorbaninezhad F, Masoumi J, Bakhshivand M, et al. CTLA-4 silencing in dendritic cells loaded with colorectal cancer cell lysate improves autologous T cell responses in vitro. Front Immunol. 2022;13:931316.

[102]

Lim TS, Chew V, Sieow JL, et al. PD-1 expression on dendritic cells suppresses CD8 + T cell function and antitumor immunity. OncoImmunology. 2016;5(3):e1085146.

[103]

Mace TA, Ameen Z, Collins A, et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013;73(10):3007-3018.

[104]

Hester R, Mazur PK, McAllister F. Immunotherapy in pancreatic adenocarcinoma: beyond “copy/paste”. Clin Cancer Res. 2021;27(23):6287-6297.

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

243

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/