[en] [en] BACKGROUND: Epidermal growth factor receptor (EGFR) is an essential target for cancer treatment. However, EGFR inhibitor erlotinib showed limited clinical benefit in pancreatic cancer therapy. Here, we showed the underlying mechanism of tumor microenvironment suppressing the sensitivity of EGFR inhibitor through the pancreatic stellate cell (PSC).
METHODS: The expression of alpha-smooth muscle actin (α-SMA) and hypoxia marker in human pancreatic cancer tissues were detected by immunohistochemistry, and their correlation with overall survival was evaluated. Human immortalized PSC was constructed and used to investigate the potential effect on pancreatic cancer cell lines in hypoxia and normoxia. Luciferase reporter assay and Chromatin immunoprecipitation were performed to explore the potential mechanisms in vitro. The combined inhibition of EGFR and Met was evaluated in an orthotopic xenograft mouse model of pancreatic cancer.
FINDINGS: We found that high expression levels of α-SMA and hypoxia markers are associated with poor prognosis of pancreatic cancer patients. Mechanistically, we demonstrated that hypoxia induced the expression and secretion of HGF in PSC via transcription factor HIF-1α. PSC-derived HGF activates Met, the HGF receptor, suppressing the sensitivity of pancreatic cancer cells to EGFR inhibitor in a KRAS-independent manner by activating the PI3K-AKT pathway. Furthermore, we found that the combination of EGFR inhibitor and Met inhibitor significantly suppressed tumor growth in an orthotopic xenograft mouse model.
INTERPRETATION: Our study revealed a previously uncharacterized HIF1α-HGF-Met-PI3K-AKT signaling axis between PSC and cancer cells and indicated that EGFR inhibition plus Met inhibition might be a promising strategy for pancreatic cancer treatment.
FUNDING: This study was supported by The National Natural Science Foundation of China.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
Shi, Xiuhui ✱; Université de Liège - ULiège > Département de pharmacie ; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
Wang, Min ✱; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Zhang, Yuqing ✱; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
Guo, Xingjun; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Liu, Mingyang; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
Zhou, Zhijun; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
Zhao, Yan; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
He, Ruizhi; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Gao, Yang; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Liu, Yuhui; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Pan, Shutao; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Zhou, Min; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Zhao, Chunle; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Yin, Taoyuan; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Li, Xu; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Wang, Hebin; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Yang, Jingxuan; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
Zhu, Feng; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Li, Min ✱; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA. Electronic address: Min-Li@ouhsc.edu
Qin, Renyi ✱; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. Electronic address: ryqin@tjh.tjmu.edu.cn
Siegel, R.L., Miller, K.D., Fuchs, H.E., Jemal, A., Cancer statistics, 2021. CA Cancer J Clin 71:1 (2021), 7–33.
Erkan, M., Reiser-Erkan, C., Michalski, C.W., et al. Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia 11:5 (2009), 497–508.
Vonderheide, R.H., Bayne, L.J., Inflammatory networks and immune surveillance of pancreatic carcinoma. Curr Opin Immunol 25:2 (2013), 200–205.
Sherman, M.H., Yu, R.T., Tseng, T.W., et al. Stromal cues regulate the pancreatic cancer epigenome and metabolome. Proc Natl Acad Sci U S A 114:5 (2017), 1129–1134.
Carstens, J.L., Correa de Sampaio, P., Yang, D., et al. Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat Commun, 8, 2017, 15095.
Fan, Y.F., Shang, W.T., Lu, G.H., et al. Decreasing hyaluronic acid combined with drug-loaded nanoprobes improve the delivery and efficacy of chemotherapeutic drugs for pancreatic cancer. Cancer Lett 523 (2021), 1–9.
Endo, S., Nakata, K., Ohuchida, K., et al. Autophagy is required for activation of pancreatic stellate cells, associated with pancreatic cancer progression and promotes growth of pancreatic tumors in mice. Gastroenterology, 152(6), 2017, 1492.
Neesse, A., Bauer, C.A., Ohlund, D., et al. Stromal biology and therapy in pancreatic cancer: ready for clinical translation?. Gut 68:1 (2019), 159–171.
Cai, W., Sun, X., Jin, F., et al. PERK-eIF2alpha-ERK1/2 axis drives mesenchymal-endothelial transition of cancer-associated fibroblasts in pancreatic cancer. Cancer Lett 515 (2021), 86–95.
Kuninty, P.R., Bansal, R., De Geus, S.W.L., et al. ITGA5 inhibition in pancreatic stellate cells attenuates desmoplasia and potentiates efficacy of chemotherapy in pancreatic cancer. Sci Adv, 5(9), 2019, eaax2770.
Lee, J.W., Stone, M.L., Porrett, P.M., et al. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature 567:7747 (2019), 249–252.
Tape, C.J., Ling, S., Dimitriadi, M., et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell 165:4 (2016), 910–920.
Sousa, C.M., Biancur, D.E., Wang, X., et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 536:7617 (2016), 479–483.
Auciello, F.R., Bulusu, V., Oon, C., et al. A stromal lysolipid-autotaxin signaling axis promotes pancreatic tumor progression. Cancer Discov 9:5 (2019), 617–627.
Shi, Y., Gao, W., Lytle, N.K., et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature 569:7754 (2019), 131–135.
Ide, T., Kitajima, Y., Miyoshi, A., et al. Tumor-stromal cell interaction under hypoxia increases the invasiveness of pancreatic cancer cells through the hepatocyte growth factor/c-Met pathway. Int J Cancer 119:12 (2006), 2750–2759.
Tang, H.W., Weng, J.H., Lee, W.X., et al. mTORC1-chaperonin CCT signaling regulates m(6)A RNA methylation to suppress autophagy. Proc Natl Acad Sci U S A, 118(10), 2021, e2021945118.
McAleese, C.E., Choudhury, C., Butcher, N.J., Minchin, R.F., Hypoxia-mediated drug resistance in breast cancers. Cancer Lett 502 (2021), 189–199.
Ye, L., Jin, K., Liao, Z., et al. Hypoxia-reprogrammed regulatory group 2 innate lymphoid cells promote immunosuppression in pancreatic cancer. EBioMedicine, 79, 2022, 104016.
Chen, J., Bao, Y., Song, Y., et al. Hypoxia-alleviated nanoplatform to enhance chemosensitivity and sonodynamic effect in pancreatic cancer. Cancer Lett 520 (2021), 100–108.
Li, X., Lee, Y., Kang, Y., et al. Hypoxia-induced autophagy of stellate cells inhibits expression and secretion of lumican into microenvironment of pancreatic ductal adenocarcinoma. Cell Death Differ 26:2 (2019), 382–393.
Joosten, S.P.J., Mizutani, T., Spaargaren, M., Clevers, H., Pals, S.T., MET signaling overcomes epidermal growth factor receptor inhibition in normal and colorectal cancer stem cells causing drug resistance. Gastroenterology 157:4 (2019), 1153–1155.e1.
Cascone, T., Xu, L., Lin, H.Y., et al. The HGF/c-MET pathway is a driver and biomarker of VEGFR-inhibitor resistance and vascular remodeling in non-small cell lung cancer. Clin Cancer Res 23:18 (2017), 5489–5501.
Straussman, R., Morikawa, T., Shee, K., et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487:7408 (2012), 500–504.
Apicella, M., Giannoni, E., Fiore, S., et al. Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to MET and EGFR targeted therapies. Cell Metab 28:6 (2018), 848–865.e6.
Mori, S., Akita, H., Kobayashi, S., et al. Inhibition of c-MET reverses radiation-induced malignant potential in pancreatic cancer. Cancer Lett 512 (2021), 51–59.
Yu, F., Lin, Y., Zhan, T., Chen, L., Guo, S., HGF expression induced by HIF-1alpha promote the proliferation and tube formation of endothelial progenitor cells. Cell Biol Int 39:3 (2015), 310–317.
Gluck, A.A., Orlando, E., Leiser, D., et al. Identification of a MET-eIF4G1 translational regulation axis that controls HIF-1alpha levels under hypoxia. Oncogene 37:30 (2018), 4181–4196.
Yu, S., Li, A., Liu, Q., et al. Recent advances of bispecific antibodies in solid tumors. J Hematol Oncol, 10(1), 2017, 155.
Tsao, M.S., Sakurada, A., Cutz, J.C., et al. Erlotinib in lung cancer - molecular and clinical predictors of outcome. N Engl J Med 353:2 (2005), 133–144.
Karapetis, C.S., Khambata-Ford, S., Jonker, D.J., et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 359:17 (2008), 1757–1765.
Liu, Q., Yu, S., Zhao, W., Qin, S., Chu, Q., Wu, K., EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer, 17(1), 2018, 53.
da Cunha Santos, G., Dhani, N., Tu, D., et al. Molecular predictors of outcome in a phase 3 study of gemcitabine and erlotinib therapy in patients with advanced pancreatic cancer: National Cancer Institute of Canada Clinical Trials Group Study PA.3. Cancer 116:24 (2010), 5599–5607.
Yan, H.H., Jung, K.H., Lee, J.E., et al. ANGPTL4 accelerates KRAS(G12D)-induced acinar to ductal metaplasia and pancreatic carcinogenesis. Cancer Lett 519 (2021), 185–198.
Chang, W.H., Nguyen, T.T., Hsu, C.H., et al. KRAS-dependent cancer cells promote survival by producing exosomes enriched in Survivin. Cancer Lett 517 (2021), 66–77.
Ardito, C.M., Gruner, B.M., Takeuchi, K.K., et al. EGF receptor is required for KRAS-induced pancreatic tumorigenesis. Cancer Cell 22:3 (2012), 304–317.
Moore, M.J., Goldstein, D., Hamm, J., et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 25:15 (2007), 1960–1966.
Xiong, H.Q., Rosenberg, A., LoBuglio, A., et al. Cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor, in combination with gemcitabine for advanced pancreatic cancer: a multicenter phase II trial. J Clin Oncol 22:13 (2004), 2610–2616.
Shi, X., Yang, J., Liu, M., et al. Circular RNA ANAPC7 inhibits tumor growth and muscle wasting via PHLPP2-AKT-TGF-beta signaling axis in pancreatic cancer. Gastroenterology 162:7 (2022), 2004–20017.e2.
Yang, J., Zhang, Z., Zhang, Y., et al. ZIP4 promotes muscle wasting and cachexia in mice with orthotopic pancreatic tumors by stimulating RAB27B-regulated release of extracellular vesicles from cancer cells. Gastroenterology 156:3 (2019), 722–734.e6.
Charan, J., Kantharia, N.D., How to calculate sample size in animal studies?. J Pharmacol Pharmacother 4:4 (2013), 303–306.
Sun, Q., Zhang, B., Hu, Q., et al. The impact of cancer-associated fibroblasts on major hallmarks of pancreatic cancer. Theranostics 8:18 (2018), 5072–5087.
Intlekofer Andrew, M., Dematteo Raymond, G., Venneti, S., et al. Hypoxia induces production of L-2-hydroxyglutarate. Cell Metab 22:2 (2015), 304–311.
Patel, M.B., Pothula, S.P., Xu, Z., et al. The role of the hepatocyte growth factor/c-MET pathway in pancreatic stellate cell-endothelial cell interactions: antiangiogenic implications in pancreatic cancer. Carcinogenesis 35:8 (2014), 1891–1900.
Khan, A., Fornes, O., Stigliani, A., et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res 46:D1 (2018), D260–d6.
Li, X., Wang, Z., Ma, Q., et al. Sonic hedgehog paracrine signaling activates stromal cells to promote perineural invasion in pancreatic cancer. Clin Cancer Res 20:16 (2014), 4326–4338.
Scaltriti, M., Baselga, J., The epidermal growth factor receptor pathway: a model for targeted therapy. Clin Cancer Res 12:18 (2006), 5268–5272.
Ozdemir, B.C., Pentcheva-Hoang, T., Carstens, J.L., et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25:6 (2014), 719–734.
Rhim, A.D., Oberstein, P.E., Thomas, D.H., et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25:6 (2014), 735–747.
Schnittert, J., Bansal, R., Prakash, J., Targeting pancreatic stellate cells in cancer. Trends Cancer 5:2 (2019), 128–142.
Iwamoto, C., Ohuchida, K., Shinkawa, T., et al. Bone marrow-derived macrophages converted into cancer-associated fibroblast-like cells promote pancreatic cancer progression. Cancer Lett 512 (2021), 15–27.
Neesse, A., Algul, H., Tuveson, D.A., Gress, T.M., Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut 64:9 (2015), 1476–1484.
Ohlund, D., Handly-Santana, A., Biffi, G., et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med 214:3 (2017), 579–596.
Zhu, Y., Zhang, H., Han, X., et al. STAT3 mediated upregulation of C-MET signaling acts as a compensatory survival mechanism upon EGFR family inhibition in chemoresistant breast cancer cells. Cancer Lett 519 (2021), 328–342.
Rajadurai, C.V., Havrylov, S., Zaoui, K., et al. Met receptor tyrosine kinase signals through a cortactin-Gab1 scaffold complex, to mediate invadopodia. J Cell Sci 125:12 (2012), 2940–2953.
Gu, Z., Du, Y., Zhao, X., Wang, C., Tumor microenvironment and metabolic remodeling in gemcitabine-based chemoresistance of pancreatic cancer. Cancer Lett 521 (2021), 98–108.
Pennacchietti, S., Michieli, P., Galluzzo, M., Mazzone, M., Giordano, S., Comoglio, P.M., Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 3:4 (2003), 347–361.
McDonald, P.C., Chafe, S.C., Brown, W.S., et al. Regulation of pH by carbonic anhydrase 9 mediates survival of pancreatic cancer cells with activated KRAS in response to hypoxia. Gastroenterology 157:3 (2019), 823–837.
Xu, Z., Pang, T.C.Y., Liu, A.C., et al. Targeting the HGF/c-MET pathway in advanced pancreatic cancer: a key element of treatment that limits primary tumour growth and eliminates metastasis. Br J Cancer 122:10 (2020), 1486–1495.
Liu, M., Zhang, Y., Yang, J., et al. ZIP4 increases expression of transcription factor ZEB1 to PROMOTE INTegrin alpha3beta1 signaling and inhibit expression of the gemcitabine transporter ENT1 in pancreatic cancer cells. Gastroenterology 158:3 (2020), 679–692e1.
Shukla, S.K., Purohit, V., Mehla, K., et al. MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer. Cancer Cell, 32(3), 2017, 392.
Yu, X., Liu, W., Wang, Z., et al. CD73 induces gemcitabine resistance in pancreatic ductal adenocarcinoma: a promising target with non-canonical mechanisms. Cancer Lett 519 (2021), 289–303.
Lee, J.E., Kang, Y.W., Jung, K.H., et al. Intracellular KRAS-specific antibody enhances the anti-tumor efficacy of gemcitabine in pancreatic cancer by inducing endosomal escape. Cancer Lett 507 (2021), 97–111.
Wilson, T.R., Fridlyand, J., Yan, Y., et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 487:7408 (2012), 505–509.
Navas, C., Hernandez-Porras, I., Schuhmacher, A.J., Sibilia, M., Guerra, C., Barbacid, M., EGF receptor signaling is essential for k-ras oncogene-driven pancreatic ductal adenocarcinoma. Cancer Cell 22:3 (2012), 318–330.
Yang, L., Lin, S., Kang, Y., et al. Rhein sensitizes human pancreatic cancer cells to EGFR inhibitors by inhibiting STAT3 pathway. J Exp Clin Cancer Res, 38(1), 2019, 31.
Fu, J., Su, X., Li, Z., et al. HGF/c-MET pathway in cancer: from molecular characterization to clinical evidence. Oncogene 40:28 (2021), 4625–4651.