Skip to main content
Top
Published in: Molecular Cancer 1/2019

Open Access 01-12-2019 | Review

Tumor microenvironment-driven non-cell-autonomous resistance to antineoplastic treatment

Authors: Yidi Qu, Bo Dou, Horyue Tan, Yibin Feng, Ning Wang, Di Wang

Published in: Molecular Cancer | Issue 1/2019

Login to get access

Abstract

Drug resistance is of great concern in cancer treatment because most effective drugs are limited by the development of resistance following some periods of therapeutic administration. The tumor microenvironment (TME), which includes various types of cells and extracellular components, mediates tumor progression and affects treatment efficacy. TME-mediated drug resistance is associated with tumor cells and their pericellular matrix. Noninherent-adaptive drug resistance refers to a non-cell-autonomous mechanism in which the resistance lies in the treatment process rather than genetic or epigenetic changes, and this mechanism is closely related to the TME. A new concept is therefore proposed in which tumor cell resistance to targeted therapy may be due to non-cell-autonomous mechanisms. However, knowledge of non-cell-autonomous mechanisms of resistance to different treatments is not comprehensive. In this review, we outlined TME factors and molecular events involved in the regulation of non-cell-autonomous resistance of cancer, summarized how the TME contributes to non-cell-autonomous drug resistance in different types of antineoplastic treatment, and discussed the novel strategies to investigate and overcome the non-cell-autonomous mechanism of cancer non-cell-autonomous resistance.
Literature
1.
go back to reference Dugger SA, Platt A, Goldstein DB. Drug development in the era of precision medicine. Nat Rev Drug Discov. 2018;17:183–96.PubMedCrossRef Dugger SA, Platt A, Goldstein DB. Drug development in the era of precision medicine. Nat Rev Drug Discov. 2018;17:183–96.PubMedCrossRef
2.
3.
go back to reference Coussens NP, Braisted JC, Peryea T, Sittampalam GS, Simeonov A, Hall MD. Small-Molecule Screens: A Gateway to Cancer Therapeutic Agents with Case Studies of Food and Drug Administration-Approved Drugs. Pharmacol Rev. 2017;69:479–96.PubMedPubMedCentralCrossRef Coussens NP, Braisted JC, Peryea T, Sittampalam GS, Simeonov A, Hall MD. Small-Molecule Screens: A Gateway to Cancer Therapeutic Agents with Case Studies of Food and Drug Administration-Approved Drugs. Pharmacol Rev. 2017;69:479–96.PubMedPubMedCentralCrossRef
4.
go back to reference Garraway LA. Jã¤Nne PA: Circumventing cancer drug resistance in the era of personalized medicine. Cancer Discovery. 2012;2:214–26.PubMedCrossRef Garraway LA. Jã¤Nne PA: Circumventing cancer drug resistance in the era of personalized medicine. Cancer Discovery. 2012;2:214–26.PubMedCrossRef
5.
go back to reference Corso S, Giordano S. Cell-autonomous and non-cell-autonomous mechanisms of HGF/MET-driven resistance to targeted therapies: from basic research to a clinical perspective. Cancer Discov. 2013;3:978–92.PubMedCrossRef Corso S, Giordano S. Cell-autonomous and non-cell-autonomous mechanisms of HGF/MET-driven resistance to targeted therapies: from basic research to a clinical perspective. Cancer Discov. 2013;3:978–92.PubMedCrossRef
6.
go back to reference Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13:714–26.PubMedCrossRef Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13:714–26.PubMedCrossRef
7.
go back to reference Gottesman MM, Tito F, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58.PubMedCrossRef Gottesman MM, Tito F, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58.PubMedCrossRef
8.
go back to reference Mansoori B, Mohammadi A, Davudian S, Shirjang S, Baradaran B. The Different Mechanisms of Cancer Drug Resistance: A Brief Review. Advanced Pharmaceutical Bulletin. 2017;7:339–48.PubMedPubMedCentralCrossRef Mansoori B, Mohammadi A, Davudian S, Shirjang S, Baradaran B. The Different Mechanisms of Cancer Drug Resistance: A Brief Review. Advanced Pharmaceutical Bulletin. 2017;7:339–48.PubMedPubMedCentralCrossRef
10.
go back to reference Al-Jamal HAN, Jusoh SAM, Ang Cheng Y, Jamaruddin Mat A, Rosline H, Muhammad Farid J. Silencing of suppressor of cytokine signaling-3 due to methylation results in phosphorylation of STAT3 in imatinib resistant BCR-ABL positive chronic myeloid leukemia cells. Asian Pac J Cancer Prev. 2013;15:4555–61.CrossRef Al-Jamal HAN, Jusoh SAM, Ang Cheng Y, Jamaruddin Mat A, Rosline H, Muhammad Farid J. Silencing of suppressor of cytokine signaling-3 due to methylation results in phosphorylation of STAT3 in imatinib resistant BCR-ABL positive chronic myeloid leukemia cells. Asian Pac J Cancer Prev. 2013;15:4555–61.CrossRef
11.
12.
go back to reference Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92:1295–302. Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92:1295–302.
13.
go back to reference Bonanno L, Favaretto A, Rosell R. Platinum Drugs and DNA Repair Mechanisms in Lung Cancer. Anticancer Research. 2014;34:493–501.PubMed Bonanno L, Favaretto A, Rosell R. Platinum Drugs and DNA Repair Mechanisms in Lung Cancer. Anticancer Research. 2014;34:493–501.PubMed
14.
go back to reference Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II. Therapy resistance mediated by cancer stem cells. Semin Cancer Biol. 2018;53:156–67. Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II. Therapy resistance mediated by cancer stem cells. Semin Cancer Biol. 2018;53:156–67.
16.
go back to reference Hanahan D, Coussens L. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell. 2012;21:309–22.CrossRefPubMed Hanahan D, Coussens L. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell. 2012;21:309–22.CrossRefPubMed
18.
19.
go back to reference Guri Y, Hall MN. mTOR Signaling Confers Resistance to Targeted Cancer Drugs. Trends Cancer. 2016;2:688–97.PubMedCrossRef Guri Y, Hall MN. mTOR Signaling Confers Resistance to Targeted Cancer Drugs. Trends Cancer. 2016;2:688–97.PubMedCrossRef
20.
21.
go back to reference Najafi M, Goradel NH, Farhood B, Salehi E, Solhjoo S, Toolee H, Kharazinejad E, Mortezaee K. Tumor microenvironment: Interactions and therapy. J Cell Physiol. 2019;234:5700–21.PubMedCrossRef Najafi M, Goradel NH, Farhood B, Salehi E, Solhjoo S, Toolee H, Kharazinejad E, Mortezaee K. Tumor microenvironment: Interactions and therapy. J Cell Physiol. 2019;234:5700–21.PubMedCrossRef
22.
go back to reference Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, Kong X, Bosenberg MC, Wiesner T, Rosen N, et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature. 2015;520:368–72.PubMedPubMedCentralCrossRef Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, Kong X, Bosenberg MC, Wiesner T, Rosen N, et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature. 2015;520:368–72.PubMedPubMedCentralCrossRef
23.
go back to reference Allen E, Mieville P, Warren CM, Saghafinia S, Li L, Peng MW, Hanahan D. Metabolic Symbiosis Enables Adaptive Resistance to Anti-angiogenic Therapy that Is Dependent on mTOR Signaling. Cell Rep. 2016;15:1144–60.PubMedPubMedCentralCrossRef Allen E, Mieville P, Warren CM, Saghafinia S, Li L, Peng MW, Hanahan D. Metabolic Symbiosis Enables Adaptive Resistance to Anti-angiogenic Therapy that Is Dependent on mTOR Signaling. Cell Rep. 2016;15:1144–60.PubMedPubMedCentralCrossRef
24.
go back to reference Zhang B, Fu D, Xu Q, Cong X, Wu C, Zhong X, Ma Y, Lv Z, Chen F, Han L, et al. The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK1. Nat Commun. 2018;9:1723.PubMedPubMedCentralCrossRef Zhang B, Fu D, Xu Q, Cong X, Wu C, Zhong X, Ma Y, Lv Z, Chen F, Han L, et al. The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK1. Nat Commun. 2018;9:1723.PubMedPubMedCentralCrossRef
25.
go back to reference Krizhanovsky V, Yon M, Dickins R, Hearn SJ, Miething C, Yee H, Zender L, Lowe S. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134:657–67.PubMedPubMedCentralCrossRef Krizhanovsky V, Yon M, Dickins R, Hearn SJ, Miething C, Yee H, Zender L, Lowe S. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134:657–67.PubMedPubMedCentralCrossRef
26.
go back to reference Campisi J, di Fagagna FD. Cellular senescence: when bad things happen to good cells. Nat RevMol Cell Biol. 2007;8:729–40.CrossRef Campisi J, di Fagagna FD. Cellular senescence: when bad things happen to good cells. Nat RevMol Cell Biol. 2007;8:729–40.CrossRef
27.
go back to reference Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, Premsrirut P, Luo W, Chicas A, Lee CS, et al. Control of the senescence-associated secretory phenotype by NF-kappaB promotes senescence and enhances chemosensitivity. Genes Dev. 2011;25:2125–36.PubMedPubMedCentralCrossRef Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, Premsrirut P, Luo W, Chicas A, Lee CS, et al. Control of the senescence-associated secretory phenotype by NF-kappaB promotes senescence and enhances chemosensitivity. Genes Dev. 2011;25:2125–36.PubMedPubMedCentralCrossRef
29.
go back to reference Salony SX, Alves CP, Dey-Guha I, Ritsma L, Boukhali M, Lee JH, Chowdhury J, Ross KN, Haas W. AKT Inhibition Promotes Nonautonomous Cancer Cell Survival. Mol Cancer Ther. 2016;15:142–53. Salony SX, Alves CP, Dey-Guha I, Ritsma L, Boukhali M, Lee JH, Chowdhury J, Ross KN, Haas W. AKT Inhibition Promotes Nonautonomous Cancer Cell Survival. Mol Cancer Ther. 2016;15:142–53.
30.
go back to reference Kim BH, Yi EH, Ye SK. Signal transducer and activator of transcription 3 as a therapeutic target for cancer and the tumor microenvironment. Arch Pharm Res. 2016;39:1085–99.PubMedCrossRef Kim BH, Yi EH, Ye SK. Signal transducer and activator of transcription 3 as a therapeutic target for cancer and the tumor microenvironment. Arch Pharm Res. 2016;39:1085–99.PubMedCrossRef
31.
go back to reference Samavati L, Rastogi R, Du W, Huttemann M, Fite A, Franchi L. STAT3 tyrosine phosphorylation is critical for interleukin 1 beta and interleukin-6 production in response to lipopolysaccharide and live bacteria. Mol Immunol. 2009;46:1867–77.PubMedCrossRef Samavati L, Rastogi R, Du W, Huttemann M, Fite A, Franchi L. STAT3 tyrosine phosphorylation is critical for interleukin 1 beta and interleukin-6 production in response to lipopolysaccharide and live bacteria. Mol Immunol. 2009;46:1867–77.PubMedCrossRef
32.
go back to reference Wang LL, Zhang F, Cui JY, Chen L, Chen YT, Liu BW. CAFs enhance paclitaxel resistance by inducing EMT through the IL-6/JAK2/STAT3 pathway. Oncology Reports. 2018;39:2081–90.PubMedPubMedCentral Wang LL, Zhang F, Cui JY, Chen L, Chen YT, Liu BW. CAFs enhance paclitaxel resistance by inducing EMT through the IL-6/JAK2/STAT3 pathway. Oncology Reports. 2018;39:2081–90.PubMedPubMedCentral
33.
go back to reference Jeong SK, Kim JS, Lee CG, Park YS, Kim SD, Yoon SO, Han DH, Lee KY, Jeong MH, Jo WS. Tumor associated macrophages provide the survival resistance of tumor cells to hypoxic microenvironmental condition through IL-6 receptor-mediated signals. Immunobiology. 2017;222:55–65.PubMedCrossRef Jeong SK, Kim JS, Lee CG, Park YS, Kim SD, Yoon SO, Han DH, Lee KY, Jeong MH, Jo WS. Tumor associated macrophages provide the survival resistance of tumor cells to hypoxic microenvironmental condition through IL-6 receptor-mediated signals. Immunobiology. 2017;222:55–65.PubMedCrossRef
34.
go back to reference Yin Y, Yao S, Hu Y, Feng Y, Li M, Bian Z, Zhang J, Qin Y, Qi X, Zhou L, et al. The Immune-microenvironment Confers Chemoresistance of Colorectal Cancer through Macrophage-Derived IL6. Clin Cancer Res. 2017;23:7375–87.PubMedCrossRef Yin Y, Yao S, Hu Y, Feng Y, Li M, Bian Z, Zhang J, Qin Y, Qi X, Zhou L, et al. The Immune-microenvironment Confers Chemoresistance of Colorectal Cancer through Macrophage-Derived IL6. Clin Cancer Res. 2017;23:7375–87.PubMedCrossRef
35.
go back to reference Nagathihalli NS, Castellanos JA, Shi CJ, Beesetty Y, Reyzer ML, Caprioli R, Chen X, Walsh AJ, Skala MC, Moses HL, Merchant NB. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology. 2015;149:1932–+.PubMedCrossRef Nagathihalli NS, Castellanos JA, Shi CJ, Beesetty Y, Reyzer ML, Caprioli R, Chen X, Walsh AJ, Skala MC, Moses HL, Merchant NB. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology. 2015;149:1932–+.PubMedCrossRef
36.
go back to reference Dorayappan KDP, Wanner R, Wallbillich JJ, Saini U, Zingarelli R, Suarez AA, Cohn DE, Selvendiran K. Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: a novel mechanism linking STAT3/Rab proteins. Oncogene. 2018;37:3806–21.PubMedPubMedCentralCrossRef Dorayappan KDP, Wanner R, Wallbillich JJ, Saini U, Zingarelli R, Suarez AA, Cohn DE, Selvendiran K. Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: a novel mechanism linking STAT3/Rab proteins. Oncogene. 2018;37:3806–21.PubMedPubMedCentralCrossRef
37.
go back to reference Li MM, Li M, Yin T, Shi HS, Wen Y, Zhang BL, Chen MH, Xu GC, Ren KX, Wei YQ. Targeting of cancer-associated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol Med Rep. 2016;13:2476–84.PubMedPubMedCentralCrossRef Li MM, Li M, Yin T, Shi HS, Wen Y, Zhang BL, Chen MH, Xu GC, Ren KX, Wei YQ. Targeting of cancer-associated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol Med Rep. 2016;13:2476–84.PubMedPubMedCentralCrossRef
38.
go back to reference Billottet C, Tuefferd M, Gentien D, Rapinat A, Thiery JP, Broet P, Jouanneau J. Modulation of several waves of gene expression during FGF-1 induced epithelial-mesenchymal transition of carcinoma cells. J Cell Biochem. 2008;104:826–39.PubMedCrossRef Billottet C, Tuefferd M, Gentien D, Rapinat A, Thiery JP, Broet P, Jouanneau J. Modulation of several waves of gene expression during FGF-1 induced epithelial-mesenchymal transition of carcinoma cells. J Cell Biochem. 2008;104:826–39.PubMedCrossRef
39.
go back to reference Che Y, Wang J, Li Y, Lu Z, Huang J, Sun S, Mao S, Lei Y, Zang R, Sun N, He J. Cisplatin-activated PAI-1 secretion in the cancer-associated fibroblasts with paracrine effects promoting esophageal squamous cell carcinoma progression and causing chemoresistance. Cell Death Dis. 2018;9:759.PubMedPubMedCentralCrossRef Che Y, Wang J, Li Y, Lu Z, Huang J, Sun S, Mao S, Lei Y, Zang R, Sun N, He J. Cisplatin-activated PAI-1 secretion in the cancer-associated fibroblasts with paracrine effects promoting esophageal squamous cell carcinoma progression and causing chemoresistance. Cell Death Dis. 2018;9:759.PubMedPubMedCentralCrossRef
40.
go back to reference Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben-Porath I, Onder TT, Wang ZC, Richardson AL, Weinberg RA, Orimo A. Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci U S A. 2010;107:20009–14.PubMedPubMedCentralCrossRef Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben-Porath I, Onder TT, Wang ZC, Richardson AL, Weinberg RA, Orimo A. Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci U S A. 2010;107:20009–14.PubMedPubMedCentralCrossRef
41.
go back to reference Castells M, Milhas D, Gandy C, Thibault B, Rafii A, Delord JP, Couderc B. Microenvironment mesenchymal cells protect ovarian cancer cell lines from apoptosis by inhibiting XIAP inactivation. Cell Death Dis. 2013;4:e887.PubMedPubMedCentralCrossRef Castells M, Milhas D, Gandy C, Thibault B, Rafii A, Delord JP, Couderc B. Microenvironment mesenchymal cells protect ovarian cancer cell lines from apoptosis by inhibiting XIAP inactivation. Cell Death Dis. 2013;4:e887.PubMedPubMedCentralCrossRef
42.
go back to reference Xu H, Zhou Y, Li W, Zhang B, Zhang H, Zhao S, Zheng P, Wu H, Yang J. Tumor-derived mesenchymal-stem-cell-secreted IL-6 enhances resistance to cisplatin via the STAT3 pathway in breast cancer. Oncol Lett. 2018;15:9142–50.PubMedPubMedCentral Xu H, Zhou Y, Li W, Zhang B, Zhang H, Zhao S, Zheng P, Wu H, Yang J. Tumor-derived mesenchymal-stem-cell-secreted IL-6 enhances resistance to cisplatin via the STAT3 pathway in breast cancer. Oncol Lett. 2018;15:9142–50.PubMedPubMedCentral
43.
go back to reference Skolekova S, Matuskova M, Bohac M, Toro L, Durinikova E, Tyciakova S, Demkova L, Gursky J, Kucerova L. Cisplatin-induced mesenchymal stromal cells-mediated mechanism contributing to decreased antitumor effect in breast cancer cells. Cell Commun Signal. 2016;14:7. Skolekova S, Matuskova M, Bohac M, Toro L, Durinikova E, Tyciakova S, Demkova L, Gursky J, Kucerova L. Cisplatin-induced mesenchymal stromal cells-mediated mechanism contributing to decreased antitumor effect in breast cancer cells. Cell Commun Signal. 2016;14:7.
44.
go back to reference Samuel P, Mulcahy LA, Furlong F, McCarthy HO, Brooks SA, Fabbri M, Pink RC, Carter DRF. Cisplatin induces the release of extracellular vesicles from ovarian cancer cells that can induce invasiveness and drug resistance in bystander cells. Philos Trans R Soc Lond B Biol Sci. 2018;373:20170065.PubMedCrossRef Samuel P, Mulcahy LA, Furlong F, McCarthy HO, Brooks SA, Fabbri M, Pink RC, Carter DRF. Cisplatin induces the release of extracellular vesicles from ovarian cancer cells that can induce invasiveness and drug resistance in bystander cells. Philos Trans R Soc Lond B Biol Sci. 2018;373:20170065.PubMedCrossRef
45.
go back to reference Bliss SA, Sinha G, Sandiford OA, Williams LM, Engelberth DJ, Guiro K, Isenalumhe LL, Greco SJ, Ayer S, Bryan M, et al. Mesenchymal Stem Cell-Derived Exosomes Stimulate Cycling Quiescence and Early Breast Cancer Dormancy in Bone Marrow. Cancer Res. 2016;76:5832–44.PubMedCrossRef Bliss SA, Sinha G, Sandiford OA, Williams LM, Engelberth DJ, Guiro K, Isenalumhe LL, Greco SJ, Ayer S, Bryan M, et al. Mesenchymal Stem Cell-Derived Exosomes Stimulate Cycling Quiescence and Early Breast Cancer Dormancy in Bone Marrow. Cancer Res. 2016;76:5832–44.PubMedCrossRef
46.
go back to reference Sung PL, Jan YH, Lin SC, Huang CC, Lin H, Wen KC, Chao KC, Lai CR, Wang PH, Chuang CM, et al. Periostin in tumor microenvironment is associated with poor prognosis and platinum resistance in epithelial ovarian carcinoma. Oncotarget. 2016;7:4036–47.PubMed Sung PL, Jan YH, Lin SC, Huang CC, Lin H, Wen KC, Chao KC, Lai CR, Wang PH, Chuang CM, et al. Periostin in tumor microenvironment is associated with poor prognosis and platinum resistance in epithelial ovarian carcinoma. Oncotarget. 2016;7:4036–47.PubMed
47.
go back to reference Mhawech-Fauceglia P, Yan L, Sharifian M, Ren X, Liu S, Kim G, Gayther SA, Pejovic T, Lawrenson K. Stromal Expression of Fibroblast Activation Protein Alpha (FAP) Predicts Platinum Resistance and Shorter Recurrence in patients with Epithelial Ovarian Cancer. Cancer Microenviron. 2015;8:23–31.PubMedCrossRef Mhawech-Fauceglia P, Yan L, Sharifian M, Ren X, Liu S, Kim G, Gayther SA, Pejovic T, Lawrenson K. Stromal Expression of Fibroblast Activation Protein Alpha (FAP) Predicts Platinum Resistance and Shorter Recurrence in patients with Epithelial Ovarian Cancer. Cancer Microenviron. 2015;8:23–31.PubMedCrossRef
48.
go back to reference Dijkgraaf EM, Heusinkveld M, Tummers B, Vogelpoel LT, Goedemans R, Jha V, Nortier JW, Welters MJ, Kroep JR, van der Burg SH. Chemotherapy alters monocyte differentiation to favor generation of cancer-supporting M2 macrophages in the tumor microenvironment. Cancer Res. 2013;73:2480–92.PubMedCrossRef Dijkgraaf EM, Heusinkveld M, Tummers B, Vogelpoel LT, Goedemans R, Jha V, Nortier JW, Welters MJ, Kroep JR, van der Burg SH. Chemotherapy alters monocyte differentiation to favor generation of cancer-supporting M2 macrophages in the tumor microenvironment. Cancer Res. 2013;73:2480–92.PubMedCrossRef
49.
go back to reference Perrotta C, Cervia D, Di Renzo I, Moscheni C, Bassi MT, Campana L, Martelli C, Catalani E, Giovarelli M, Zecchini S, et al. Nitric Oxide Generated by Tumor-Associated Macrophages Is Responsible for Cancer Resistance to Cisplatin and Correlated With Syntaxin 4 and Acid Sphingomyelinase Inhibition. Front Immunol. 2018;9:1186.PubMedPubMedCentralCrossRef Perrotta C, Cervia D, Di Renzo I, Moscheni C, Bassi MT, Campana L, Martelli C, Catalani E, Giovarelli M, Zecchini S, et al. Nitric Oxide Generated by Tumor-Associated Macrophages Is Responsible for Cancer Resistance to Cisplatin and Correlated With Syntaxin 4 and Acid Sphingomyelinase Inhibition. Front Immunol. 2018;9:1186.PubMedPubMedCentralCrossRef
50.
go back to reference Kitange GJ, Carlson BL, Schroeder MA, Grogan PT, Lamont JD, Decker PA, Wu W, James CD, Sarkaria JN. Induction of MGMT expression is associated with temozolomide resistance in glioblastoma xenografts. Neuro-oncology. 2009;11:281–91.PubMedPubMedCentralCrossRef Kitange GJ, Carlson BL, Schroeder MA, Grogan PT, Lamont JD, Decker PA, Wu W, James CD, Sarkaria JN. Induction of MGMT expression is associated with temozolomide resistance in glioblastoma xenografts. Neuro-oncology. 2009;11:281–91.PubMedPubMedCentralCrossRef
51.
go back to reference Chen WL, Wang CC, Lin YJ, Wu CP, Hsieh CH. Cycling hypoxia induces chemoresistance through the activation of reactive oxygen species-mediated B-cell lymphoma extra-long pathway in glioblastoma multiforme. J Transl Med. 2015;13:389.PubMedPubMedCentralCrossRef Chen WL, Wang CC, Lin YJ, Wu CP, Hsieh CH. Cycling hypoxia induces chemoresistance through the activation of reactive oxygen species-mediated B-cell lymphoma extra-long pathway in glioblastoma multiforme. J Transl Med. 2015;13:389.PubMedPubMedCentralCrossRef
52.
go back to reference Ge X, Pan MH, Wang L, Li W, Jiang C, He J, Abouzid K, Liu LZ, Shi Z, Jiang BH. Hypoxia-mediated mitochondria apoptosis inhibition induces temozolomide treatment resistance through miR-26a/Bad/Bax axis. Cell Death Dis. 2018;9:1128.PubMedPubMedCentralCrossRef Ge X, Pan MH, Wang L, Li W, Jiang C, He J, Abouzid K, Liu LZ, Shi Z, Jiang BH. Hypoxia-mediated mitochondria apoptosis inhibition induces temozolomide treatment resistance through miR-26a/Bad/Bax axis. Cell Death Dis. 2018;9:1128.PubMedPubMedCentralCrossRef
53.
go back to reference Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials. 2019;198:122–34. Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials. 2019;198:122–34.
54.
go back to reference Chen W, Wang D, Du X, He Y, Chen S, Shao Q, Ma C, Huang B, Chen A, Zhao P, et al. Glioma cells escaped from cytotoxicity of temozolomide and vincristine by communicating with human astrocytes. Med Oncol. 2015;32:43.PubMedCrossRef Chen W, Wang D, Du X, He Y, Chen S, Shao Q, Ma C, Huang B, Chen A, Zhao P, et al. Glioma cells escaped from cytotoxicity of temozolomide and vincristine by communicating with human astrocytes. Med Oncol. 2015;32:43.PubMedCrossRef
55.
go back to reference Orimo A, Weinberg RA. Stromal fibroblasts in cancer - A novel tumor-promoting cell type. Cell Cycle. 2006;5:1597–601.PubMedCrossRef Orimo A, Weinberg RA. Stromal fibroblasts in cancer - A novel tumor-promoting cell type. Cell Cycle. 2006;5:1597–601.PubMedCrossRef
56.
go back to reference Udagawa T, Wood M. Tumor-stromal cell interactions and opportunities for therapeutic intervention. Curr Opin Pharmacol. 2010;10:369–74.PubMedCrossRef Udagawa T, Wood M. Tumor-stromal cell interactions and opportunities for therapeutic intervention. Curr Opin Pharmacol. 2010;10:369–74.PubMedCrossRef
57.
go back to reference Wei L, Ye H, Li G, Lu Y, Zhou Q, Zheng S, Lin Q, Liu Y, Li Z, Chen R. Cancer-associated fibroblasts promote progression and gemcitabine resistance via the SDF-1/SATB-1 pathway in pancreatic cancer. Cell Death Dis. 2018;9:1065.PubMedPubMedCentralCrossRef Wei L, Ye H, Li G, Lu Y, Zhou Q, Zheng S, Lin Q, Liu Y, Li Z, Chen R. Cancer-associated fibroblasts promote progression and gemcitabine resistance via the SDF-1/SATB-1 pathway in pancreatic cancer. Cell Death Dis. 2018;9:1065.PubMedPubMedCentralCrossRef
58.
go back to reference Lee J, Yakubov B, Ivan C, Jones DR, Caperell-Grant A, Fishel M, Cardenas H, Matei D. Tissue transglutaminase activates cancer-associated fibroblasts and contributes to gemcitabine resistance in pancreatic cancer. Neoplasia. 2016;18:689–98.PubMedPubMedCentralCrossRef Lee J, Yakubov B, Ivan C, Jones DR, Caperell-Grant A, Fishel M, Cardenas H, Matei D. Tissue transglutaminase activates cancer-associated fibroblasts and contributes to gemcitabine resistance in pancreatic cancer. Neoplasia. 2016;18:689–98.PubMedPubMedCentralCrossRef
59.
go back to reference Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, Shlomi T, Gil Z. Transfer of miRNA in macrophages-derived exosomes induces drug resistance of pancreatic adenocarcinoma. Cancer Res. 2018;78:5287–99. Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, Shlomi T, Gil Z. Transfer of miRNA in macrophages-derived exosomes induces drug resistance of pancreatic adenocarcinoma. Cancer Res. 2018;78:5287–99.
60.
go back to reference Bruchard M, Mignot G, Derangere V, Chalmin F, Chevriaux A, Vegran F, Boireau W, Simon B, Ryffel B, Connat JL, et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19:57–64.PubMedCrossRef Bruchard M, Mignot G, Derangere V, Chalmin F, Chevriaux A, Vegran F, Boireau W, Simon B, Ryffel B, Connat JL, et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19:57–64.PubMedCrossRef
61.
go back to reference Mak PY, Mak DH, Mu H, Shi Y, Ruvolo P, Ruvolo V, Jacamo R, Burks JK, Wei W, Huang X, et al. Apoptosis repressor with caspase recruitment domain is regulated by MAPK/PI3K and confers drug resistance and survival advantage to AML. Apoptosis. 2014;19:698–707.PubMedPubMedCentralCrossRef Mak PY, Mak DH, Mu H, Shi Y, Ruvolo P, Ruvolo V, Jacamo R, Burks JK, Wei W, Huang X, et al. Apoptosis repressor with caspase recruitment domain is regulated by MAPK/PI3K and confers drug resistance and survival advantage to AML. Apoptosis. 2014;19:698–707.PubMedPubMedCentralCrossRef
62.
go back to reference OR E, Dhami SPS, Baev DV, Ortutay C, Halpin-McCormick A, Morrell R, Santocanale C, Samali A, Quinn J, O'Dwyer ME, Szegezdi E. Repression of Mcl-1 expression by the CDC7/CDK9 inhibitor PHA-767491 overcomes bone marrow stroma-mediated drug resistance in AML. Sci Rep. 2018;8:15752.PubMedPubMedCentralCrossRef OR E, Dhami SPS, Baev DV, Ortutay C, Halpin-McCormick A, Morrell R, Santocanale C, Samali A, Quinn J, O'Dwyer ME, Szegezdi E. Repression of Mcl-1 expression by the CDC7/CDK9 inhibitor PHA-767491 overcomes bone marrow stroma-mediated drug resistance in AML. Sci Rep. 2018;8:15752.PubMedPubMedCentralCrossRef
63.
go back to reference Piya S, Andreeff M, Borthakur G. Targeting autophagy to overcome chemoresistance in acute myleogenous leukemia. Autophagy. 2017;13:214–5.PubMedCrossRef Piya S, Andreeff M, Borthakur G. Targeting autophagy to overcome chemoresistance in acute myleogenous leukemia. Autophagy. 2017;13:214–5.PubMedCrossRef
64.
go back to reference Sironi S, Wagner M, Kuett A, Drolle H, Polzer H, Spiekermann K, Rieger C, Fiegl M. Microenvironmental hypoxia regulates FLT3 expression and biology in AML. Sci Rep. 2015;5:17550.PubMedPubMedCentralCrossRef Sironi S, Wagner M, Kuett A, Drolle H, Polzer H, Spiekermann K, Rieger C, Fiegl M. Microenvironmental hypoxia regulates FLT3 expression and biology in AML. Sci Rep. 2015;5:17550.PubMedPubMedCentralCrossRef
65.
go back to reference Hoellenriegel J, Zboralski D, Maasch C, Rosin NY, Wierda WG, Keating MJ, Kruschinski A, Burger JA. The Spiegelmer NOX-A12, a novel CXCL12 inhibitor, interferes with chronic lymphocytic leukemia cell motility and causes chemosensitization. Blood. 2014;123:1032–9.PubMedPubMedCentralCrossRef Hoellenriegel J, Zboralski D, Maasch C, Rosin NY, Wierda WG, Keating MJ, Kruschinski A, Burger JA. The Spiegelmer NOX-A12, a novel CXCL12 inhibitor, interferes with chronic lymphocytic leukemia cell motility and causes chemosensitization. Blood. 2014;123:1032–9.PubMedPubMedCentralCrossRef
66.
go back to reference Rosich L, Saborit-Villarroya I, Lopez-Guerra M, Xargay-Torrent S, Montraveta A, Aymerich M, Villamor N, Campo E, Perez-Galan P, Roue G, Colomer D. The phosphatidylinositol-3-kinase inhibitor NVP-BKM120 overcomes resistance signals derived from microenvironment by regulating the Akt/FoxO3a/Bim axis in chronic lymphocytic leukemia cells. Haematologica. 2013;98:1739–47.PubMedPubMedCentralCrossRef Rosich L, Saborit-Villarroya I, Lopez-Guerra M, Xargay-Torrent S, Montraveta A, Aymerich M, Villamor N, Campo E, Perez-Galan P, Roue G, Colomer D. The phosphatidylinositol-3-kinase inhibitor NVP-BKM120 overcomes resistance signals derived from microenvironment by regulating the Akt/FoxO3a/Bim axis in chronic lymphocytic leukemia cells. Haematologica. 2013;98:1739–47.PubMedPubMedCentralCrossRef
67.
go back to reference Trimarco V, Ave E, Facco M, Chiodin G, Frezzato F, Martini V, Gattazzo C, Lessi F, Giorgi CA, Visentin A, et al. Cross-talk between chronic lymphocytic leukemia (CLL) tumor B cells and mesenchymal stromal cells (MSCs): implications for neoplastic cell survival. Oncotarget. 2015;6:42130–49.PubMedPubMedCentralCrossRef Trimarco V, Ave E, Facco M, Chiodin G, Frezzato F, Martini V, Gattazzo C, Lessi F, Giorgi CA, Visentin A, et al. Cross-talk between chronic lymphocytic leukemia (CLL) tumor B cells and mesenchymal stromal cells (MSCs): implications for neoplastic cell survival. Oncotarget. 2015;6:42130–49.PubMedPubMedCentralCrossRef
68.
go back to reference Zhang W, Trachootham D, Liu J, Chen G, Pelicano H, Garcia-Prieto C, Lu W, Burger JA, Croce CM, Plunkett W, et al. Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat Cell Biol. 2012;14:276–86.PubMedPubMedCentralCrossRef Zhang W, Trachootham D, Liu J, Chen G, Pelicano H, Garcia-Prieto C, Lu W, Burger JA, Croce CM, Plunkett W, et al. Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat Cell Biol. 2012;14:276–86.PubMedPubMedCentralCrossRef
69.
go back to reference Tu B, Zhu J, Liu S, Wang L, Fan Q, Hao Y, Fan C, Tang TT. Mesenchymal stem cells promote osteosarcoma cell survival and drug resistance through activation of STAT3. Oncotarget. 2016;7:48296–308. Tu B, Zhu J, Liu S, Wang L, Fan Q, Hao Y, Fan C, Tang TT. Mesenchymal stem cells promote osteosarcoma cell survival and drug resistance through activation of STAT3. Oncotarget. 2016;7:48296–308.
70.
go back to reference Yang H, Zheng Y, Zhang Y, Cao Z, Jiang Y. Mesenchymal stem cells derived from multiple myeloma patients protect against chemotherapy through autophagy-dependent activation of NF-κB signaling. Leuk Res. 2017;60:82–8.PubMedCrossRef Yang H, Zheng Y, Zhang Y, Cao Z, Jiang Y. Mesenchymal stem cells derived from multiple myeloma patients protect against chemotherapy through autophagy-dependent activation of NF-κB signaling. Leuk Res. 2017;60:82–8.PubMedCrossRef
71.
go back to reference Ramachandran IR, Condamine T, Lin C, Herlihy SE, Garfall A, Vogl DT, Gabrilovich DI, Nefedova Y. Bone marrow PMN-MDSCs and neutrophils are functionally similar in protection of multiple myeloma from chemotherapy. Cancer Letters. 2016;371:117–24.PubMedCrossRef Ramachandran IR, Condamine T, Lin C, Herlihy SE, Garfall A, Vogl DT, Gabrilovich DI, Nefedova Y. Bone marrow PMN-MDSCs and neutrophils are functionally similar in protection of multiple myeloma from chemotherapy. Cancer Letters. 2016;371:117–24.PubMedCrossRef
72.
go back to reference Hsu YL, Hung JY, Tsai EM, Wu CY, Ho YW, Jian SF, Yen MC, Chang WA, Hou MF, Kuo PL. Benzyl butyl phthalate increases the chemoresistance to doxorubicin/cyclophosphamide by increasing breast cancer-associated dendritic cell-derived CXCL1/GROalpha and S100A8/A9. Oncol Rep. 2015;34:2889–900.PubMedCrossRef Hsu YL, Hung JY, Tsai EM, Wu CY, Ho YW, Jian SF, Yen MC, Chang WA, Hou MF, Kuo PL. Benzyl butyl phthalate increases the chemoresistance to doxorubicin/cyclophosphamide by increasing breast cancer-associated dendritic cell-derived CXCL1/GROalpha and S100A8/A9. Oncol Rep. 2015;34:2889–900.PubMedCrossRef
73.
go back to reference Mendoza-Rodriguez M, Arevalo Romero H, Fuentes-Panana EM, Ayala-Sumuano JT, Meza I. IL-1beta induces up-regulation of BIRC3, a gene involved in chemoresistance to doxorubicin in breast cancer cells. Cancer Lett. 2017;390:39–44.PubMedCrossRef Mendoza-Rodriguez M, Arevalo Romero H, Fuentes-Panana EM, Ayala-Sumuano JT, Meza I. IL-1beta induces up-regulation of BIRC3, a gene involved in chemoresistance to doxorubicin in breast cancer cells. Cancer Lett. 2017;390:39–44.PubMedCrossRef
74.
go back to reference Zhang L, Hannay JA, Liu J, Das P, Zhan M, Nguyen T, Hicklin DJ, Yu D, Pollock RE, Lev D. Vascular endothelial growth factor overexpression by soft tissue sarcoma cells: implications for tumor growth, metastasis, and chemoresistance. Cancer Res. 2006;66:8770–8.PubMedCrossRef Zhang L, Hannay JA, Liu J, Das P, Zhan M, Nguyen T, Hicklin DJ, Yu D, Pollock RE, Lev D. Vascular endothelial growth factor overexpression by soft tissue sarcoma cells: implications for tumor growth, metastasis, and chemoresistance. Cancer Res. 2006;66:8770–8.PubMedCrossRef
75.
go back to reference Cheteh EH, Augsten M, Rundqvist H, Bianchi J, Sarne V, Egevad L, Bykov VJ, Östman A, Wiman KG. Human cancer-associated fibroblasts enhance glutathione levels and antagonize drug-induced prostate cancer cell death. Cell Death Dis. 2017;8:e2848.PubMedPubMedCentralCrossRef Cheteh EH, Augsten M, Rundqvist H, Bianchi J, Sarne V, Egevad L, Bykov VJ, Östman A, Wiman KG. Human cancer-associated fibroblasts enhance glutathione levels and antagonize drug-induced prostate cancer cell death. Cell Death Dis. 2017;8:e2848.PubMedPubMedCentralCrossRef
76.
77.
go back to reference Li X, Shepard HM, Cowell JA, Zhao C, Osgood RJ, Rosengren S, Blouw B, Garrovillo SA, Pagel MD, Whatcott CJ, et al. Parallel Accumulation of Tumor Hyaluronan, Collagen and Other Drivers of Tumor Progression. Clin Cancer Res. 2018;24:4798–807. Li X, Shepard HM, Cowell JA, Zhao C, Osgood RJ, Rosengren S, Blouw B, Garrovillo SA, Pagel MD, Whatcott CJ, et al. Parallel Accumulation of Tumor Hyaluronan, Collagen and Other Drivers of Tumor Progression. Clin Cancer Res. 2018;24:4798–807.
78.
go back to reference Gangoda L, Keerthikumar S, Fonseka P, Edgington LE, Ang CS, Ozcitti C, Bogyo M, Parker BS, Mathivanan S. Inhibition of cathepsin proteases attenuates migration and sensitizes aggressive N-Myc amplified human neuroblastoma cells to doxorubicin. Oncotarget. 2015;6:11175–90.PubMedPubMedCentralCrossRef Gangoda L, Keerthikumar S, Fonseka P, Edgington LE, Ang CS, Ozcitti C, Bogyo M, Parker BS, Mathivanan S. Inhibition of cathepsin proteases attenuates migration and sensitizes aggressive N-Myc amplified human neuroblastoma cells to doxorubicin. Oncotarget. 2015;6:11175–90.PubMedPubMedCentralCrossRef
79.
go back to reference Joyce MH, Lu C, James ER, Hegab R, Allen SC, Suggs LJ, Brock A. Phenotypic basis for matrix stiffness-dependent chemoresistance of breast cancer cells to doxorubicin. Front Oncol. 2018;8:337.PubMedPubMedCentralCrossRef Joyce MH, Lu C, James ER, Hegab R, Allen SC, Suggs LJ, Brock A. Phenotypic basis for matrix stiffness-dependent chemoresistance of breast cancer cells to doxorubicin. Front Oncol. 2018;8:337.PubMedPubMedCentralCrossRef
80.
go back to reference Ebata T, Mitsui Y, Sugimoto W, Maeda M, Araki K, Machiyama H, Harada I, Sawada Y, Fujita H, Hirata H, Kawauchi K. Substrate Stiffness Influences Doxorubicin-Induced p53 Activation via ROCK2 Expression. Biomed Res Int. 2017;2017:5158961.PubMedPubMedCentralCrossRef Ebata T, Mitsui Y, Sugimoto W, Maeda M, Araki K, Machiyama H, Harada I, Sawada Y, Fujita H, Hirata H, Kawauchi K. Substrate Stiffness Influences Doxorubicin-Induced p53 Activation via ROCK2 Expression. Biomed Res Int. 2017;2017:5158961.PubMedPubMedCentralCrossRef
81.
go back to reference Lopes-Rodrigues V, Di Luca A, Sousa D, Seca H, Meleady P, Henry M, Lima RT, O'Connor R, Vasconcelos MH. Multidrug resistant tumour cells shed more microvesicle-like EVs and less exosomes than their drug-sensitive counterpart cells. Biochim Biophys Acta. 2016;1860:618–27.PubMedCrossRef Lopes-Rodrigues V, Di Luca A, Sousa D, Seca H, Meleady P, Henry M, Lima RT, O'Connor R, Vasconcelos MH. Multidrug resistant tumour cells shed more microvesicle-like EVs and less exosomes than their drug-sensitive counterpart cells. Biochim Biophys Acta. 2016;1860:618–27.PubMedCrossRef
82.
go back to reference Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, Ji MH, Hu Q, Luo Z, Wu JZ, Tang JH. Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One. 2014;9:e95240.PubMedPubMedCentralCrossRef Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, Ji MH, Hu Q, Luo Z, Wu JZ, Tang JH. Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One. 2014;9:e95240.PubMedPubMedCentralCrossRef
83.
go back to reference Huber RM, Lucas JM, Gomez-Sarosi LA, Coleman I, Zhao S, Coleman R. Nelson PS: DNA damage induces GDNF secretion in the tumor microenvironment with paracrine effects promoting prostate cancer treatment resistance. Oncotarget. 2015;6:2134–47.PubMedCrossRef Huber RM, Lucas JM, Gomez-Sarosi LA, Coleman I, Zhao S, Coleman R. Nelson PS: DNA damage induces GDNF secretion in the tumor microenvironment with paracrine effects promoting prostate cancer treatment resistance. Oncotarget. 2015;6:2134–47.PubMedCrossRef
84.
go back to reference Yu T, Yang G, Hou Y, Tang X, Wu C, Wu XA, Guo L, Zhu Q, Luo H, Du YE, et al. Cytoplasmic GPER translocation in cancer-associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene. 2017;36:2131–45.PubMedCrossRef Yu T, Yang G, Hou Y, Tang X, Wu C, Wu XA, Guo L, Zhu Q, Luo H, Du YE, et al. Cytoplasmic GPER translocation in cancer-associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene. 2017;36:2131–45.PubMedCrossRef
85.
go back to reference Milito AD, Fais S. Tumor acidity, chemoresistance and proton pump inhibitors. Future Oncology. 2005;1:779–86.PubMedCrossRef Milito AD, Fais S. Tumor acidity, chemoresistance and proton pump inhibitors. Future Oncology. 2005;1:779–86.PubMedCrossRef
86.
go back to reference Mahoney BP, Raghunand N, Baggett B, Gillies RJ. Tumor acidity, ion trapping and chemotherapeutics I. Acid pH affects the distribution of chemotherapeutic agents in vitro. Biochem Pharmacol. 2003;66:1207–18.PubMedCrossRef Mahoney BP, Raghunand N, Baggett B, Gillies RJ. Tumor acidity, ion trapping and chemotherapeutics I. Acid pH affects the distribution of chemotherapeutic agents in vitro. Biochem Pharmacol. 2003;66:1207–18.PubMedCrossRef
87.
go back to reference Fan S, Niu Y, Tan N, Wu Z, Wang Y, You H, Ke R, Song J, Shen Q, Wang W, et al. LASS2 enhances chemosensitivity of breast cancer by counteracting acidic tumor microenvironment through inhibiting activity of V-ATPase proton pump. Oncogene. 2013;32:1682–90.PubMedCrossRef Fan S, Niu Y, Tan N, Wu Z, Wang Y, You H, Ke R, Song J, Shen Q, Wang W, et al. LASS2 enhances chemosensitivity of breast cancer by counteracting acidic tumor microenvironment through inhibiting activity of V-ATPase proton pump. Oncogene. 2013;32:1682–90.PubMedCrossRef
88.
go back to reference Bar-Natan M, Stroopinsky D, Luptakova K, Coll MD, Apel A, Rajabi H, Pyzer AR, Palmer K, Reagan MR, Nahas MR, et al. Bone marrow stroma protects myeloma cells from cytotoxic damage via induction of the oncoprotein MUC1. Br J Haematol. 2017;176(6):929–38.PubMedPubMedCentralCrossRef Bar-Natan M, Stroopinsky D, Luptakova K, Coll MD, Apel A, Rajabi H, Pyzer AR, Palmer K, Reagan MR, Nahas MR, et al. Bone marrow stroma protects myeloma cells from cytotoxic damage via induction of the oncoprotein MUC1. Br J Haematol. 2017;176(6):929–38.PubMedPubMedCentralCrossRef
89.
go back to reference De Beule N, De Veirman K, Maes K, De Bruyne E, Menu E, Breckpot K, De Raeve H, Van Rampelbergh R, Van Ginderachter JA, Schots R, et al. Tumour-associated macrophage-mediated survival of myeloma cells through STAT3 activation. J Pathol. 2017;241:534–46.PubMedCrossRef De Beule N, De Veirman K, Maes K, De Bruyne E, Menu E, Breckpot K, De Raeve H, Van Rampelbergh R, Van Ginderachter JA, Schots R, et al. Tumour-associated macrophage-mediated survival of myeloma cells through STAT3 activation. J Pathol. 2017;241:534–46.PubMedCrossRef
90.
go back to reference Santoni M, Massari F, Amantini C, Nabissi M, Maines F, Burattini L, Berardi R, Santoni G, Montironi R, Tortora G, Cascinu S. Emerging role of tumor-associated macrophages as therapeutic targets in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother. 2013;62:1757–68.PubMedCrossRef Santoni M, Massari F, Amantini C, Nabissi M, Maines F, Burattini L, Berardi R, Santoni G, Montironi R, Tortora G, Cascinu S. Emerging role of tumor-associated macrophages as therapeutic targets in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother. 2013;62:1757–68.PubMedCrossRef
91.
go back to reference Shree T, Olson OC, Elie BT, Kester JC, Garfall AL, Simpson K, Bell-McGuinn KM, Zabor EC, Brogi E, Joyce JA. Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer. Genes Dev. 2011;25:2465–79.PubMedPubMedCentralCrossRef Shree T, Olson OC, Elie BT, Kester JC, Garfall AL, Simpson K, Bell-McGuinn KM, Zabor EC, Brogi E, Joyce JA. Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer. Genes Dev. 2011;25:2465–79.PubMedPubMedCentralCrossRef
92.
go back to reference Cosse JP, Ronvaux M, Ninane N, Raes MJ. Michiels C: Hypoxia-induced decrease in p53 protein level and increase in c-jun DNA binding activity results in cancer cell resistance to etoposide. Neoplasia. 2009;11:976–86.PubMedPubMedCentralCrossRef Cosse JP, Ronvaux M, Ninane N, Raes MJ. Michiels C: Hypoxia-induced decrease in p53 protein level and increase in c-jun DNA binding activity results in cancer cell resistance to etoposide. Neoplasia. 2009;11:976–86.PubMedPubMedCentralCrossRef
93.
go back to reference Sermeus A, Rebucci M, Fransolet M, Flamant L, Desmet D, Delaive E, Arnould T, Michiels C. Differential effect of hypoxia on etoposide-induced DNA damage response and p53 regulation in different cell types. J Cell Physiol. 2013;228:2365–76.PubMedCrossRef Sermeus A, Rebucci M, Fransolet M, Flamant L, Desmet D, Delaive E, Arnould T, Michiels C. Differential effect of hypoxia on etoposide-induced DNA damage response and p53 regulation in different cell types. J Cell Physiol. 2013;228:2365–76.PubMedCrossRef
94.
go back to reference Dudley AC, Shih SC, Cliffe AR, Hida K, Klagsbrun M. Attenuated p53 activation in tumour-associated stromal cells accompanies decreased sensitivity to etoposide and vincristine. Br J Cancer. 2008;99:118–25.PubMedPubMedCentralCrossRef Dudley AC, Shih SC, Cliffe AR, Hida K, Klagsbrun M. Attenuated p53 activation in tumour-associated stromal cells accompanies decreased sensitivity to etoposide and vincristine. Br J Cancer. 2008;99:118–25.PubMedPubMedCentralCrossRef
95.
go back to reference Shin K, Klosterhoff BS, Han B. Characterization of Cell-Type-Specific Drug Transport and Resistance of Breast Cancers Using Tumor-Microenvironment-on-Chip. Mol Pharm. 2016;13:2214–23.PubMedPubMedCentralCrossRef Shin K, Klosterhoff BS, Han B. Characterization of Cell-Type-Specific Drug Transport and Resistance of Breast Cancers Using Tumor-Microenvironment-on-Chip. Mol Pharm. 2016;13:2214–23.PubMedPubMedCentralCrossRef
96.
go back to reference Hazlehurst LA, Damiano JS, Buyuksal I, Pledger WJ, Dalton WS. Adhesion to fibronectin via beta1 integrins regulates p27kip1 levels and contributes to cell adhesion mediated drug resistance (CAM-DR). Oncogene. 2000;19:4319–27.PubMedCrossRef Hazlehurst LA, Damiano JS, Buyuksal I, Pledger WJ, Dalton WS. Adhesion to fibronectin via beta1 integrins regulates p27kip1 levels and contributes to cell adhesion mediated drug resistance (CAM-DR). Oncogene. 2000;19:4319–27.PubMedCrossRef
97.
98.
go back to reference Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, Fehrenbacher L, Wolter JM, Paton V, Shak S, Lieberman G, Slamon DJ. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol. 1999;17:2639–48.PubMedCrossRef Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, Fehrenbacher L, Wolter JM, Paton V, Shak S, Lieberman G, Slamon DJ. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol. 1999;17:2639–48.PubMedCrossRef
99.
go back to reference Park S, Jiang ZJ, Mortenson ED, Deng LF, Radkevich-Brown O, Yang XM, Sattar H, Wang Y, Brown NK, Greene M, et al. The Therapeutic Effect of Anti-HER2/neu Antibody Depends on Both Innate and Adaptive Immunity. Cancer Cell. 2010;18:160–70.PubMedPubMedCentralCrossRef Park S, Jiang ZJ, Mortenson ED, Deng LF, Radkevich-Brown O, Yang XM, Sattar H, Wang Y, Brown NK, Greene M, et al. The Therapeutic Effect of Anti-HER2/neu Antibody Depends on Both Innate and Adaptive Immunity. Cancer Cell. 2010;18:160–70.PubMedPubMedCentralCrossRef
100.
go back to reference Duong MN, Cleret A, Matera EL, Chettab K, Mathé D, Valsesia-Wittmann S, Clémenceau B, Dumontet C. Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity. Breast Cancer Res. 2015;17:57.PubMedPubMedCentralCrossRef Duong MN, Cleret A, Matera EL, Chettab K, Mathé D, Valsesia-Wittmann S, Clémenceau B, Dumontet C. Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity. Breast Cancer Res. 2015;17:57.PubMedPubMedCentralCrossRef
101.
go back to reference Saito S, Morishima K, Ui T, Hoshino H, Matsubara D, Ishikawa S, Aburatani H, Fukayama M, Hosoya Y, Sata N, et al. The role of HGF/MET and FGF/FGFR in fibroblast-derived growth stimulation and lapatinib-resistance of esophageal squamous cell carcinoma. BMC Cancer. 2015;15:82.PubMedPubMedCentralCrossRef Saito S, Morishima K, Ui T, Hoshino H, Matsubara D, Ishikawa S, Aburatani H, Fukayama M, Hosoya Y, Sata N, et al. The role of HGF/MET and FGF/FGFR in fibroblast-derived growth stimulation and lapatinib-resistance of esophageal squamous cell carcinoma. BMC Cancer. 2015;15:82.PubMedPubMedCentralCrossRef
102.
go back to reference Marusyk A, Tabassum DP, Janiszewska M, Place AE, Trinh A, Rozhok AI, Pyne S, Guerriero JL, Shu S, Ekram M, et al. Spatial Proximity to Fibroblasts Impacts Molecular Features and Therapeutic Sensitivity of Breast Cancer Cells Influencing Clinical Outcomes. Cancer Res. 2016;76(22):6495–506.PubMedPubMedCentralCrossRef Marusyk A, Tabassum DP, Janiszewska M, Place AE, Trinh A, Rozhok AI, Pyne S, Guerriero JL, Shu S, Ekram M, et al. Spatial Proximity to Fibroblasts Impacts Molecular Features and Therapeutic Sensitivity of Breast Cancer Cells Influencing Clinical Outcomes. Cancer Res. 2016;76(22):6495–506.PubMedPubMedCentralCrossRef
103.
go back to reference Zoeller JJ, Bronson RT, Selfors LM, Mills GB, Brugge JS. Niche-localized tumor cells are protected from HER2-targeted therapy via upregulation of an anti-apoptotic program in vivo. NPJ Breast Cancer. 2017;3:18.PubMedPubMedCentralCrossRef Zoeller JJ, Bronson RT, Selfors LM, Mills GB, Brugge JS. Niche-localized tumor cells are protected from HER2-targeted therapy via upregulation of an anti-apoptotic program in vivo. NPJ Breast Cancer. 2017;3:18.PubMedPubMedCentralCrossRef
104.
go back to reference Ono M, Kuwano M. Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs. Clin Cancer Res. 2006;12:7242–51.PubMedCrossRef Ono M, Kuwano M. Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs. Clin Cancer Res. 2006;12:7242–51.PubMedCrossRef
105.
go back to reference Li SQ, Schmitz KR, Jeffrey PD, Wiltzius JJW, Kussie P, Ferguson KM. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell. 2005;7:301–11.PubMedCrossRef Li SQ, Schmitz KR, Jeffrey PD, Wiltzius JJW, Kussie P, Ferguson KM. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell. 2005;7:301–11.PubMedCrossRef
106.
go back to reference Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, Erfan J, Zabolotnyy D, Kienzer HR, Cupissol D, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359:1116–27.PubMedCrossRef Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, Erfan J, Zabolotnyy D, Kienzer HR, Cupissol D, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359:1116–27.PubMedCrossRef
107.
go back to reference Johansson AC, Ansell A, Jerhammar F, Lindh MB, Grénman R, Munck-Wikland E, Östman A, Roberg K. Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol Cancer Res. 2012;10:1158–68.CrossRefPubMed Johansson AC, Ansell A, Jerhammar F, Lindh MB, Grénman R, Munck-Wikland E, Östman A, Roberg K. Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol Cancer Res. 2012;10:1158–68.CrossRefPubMed
108.
go back to reference Apicella M, Giannoni E, Fiore S, Ferrari KJ, Fernandez-Perez D, Isella C, Granchi C, Minutolo F, Sottile A, Comoglio PM, et al. Increased Lactate Secretion by Cancer Cells Sustains Non-cell-autonomous Adaptive Resistance to MET and EGFR Targeted Therapies. Cell Metabolism. 2018;28:848–+ Apicella M, Giannoni E, Fiore S, Ferrari KJ, Fernandez-Perez D, Isella C, Granchi C, Minutolo F, Sottile A, Comoglio PM, et al. Increased Lactate Secretion by Cancer Cells Sustains Non-cell-autonomous Adaptive Resistance to MET and EGFR Targeted Therapies. Cell Metabolism. 2018;28:848–+
109.
go back to reference Mueller KL, Madden JM, Zoratti GL, Kuperwasser C, List K, Boerner JL. Fibroblast-secreted hepatocyte growth factor mediates epidermal growth factor receptor tyrosine kinase inhibitor resistance in triple-negative breast cancers through paracrine activation of Met. Breast Cancer Res. 2012;14:R104.PubMedPubMedCentralCrossRef Mueller KL, Madden JM, Zoratti GL, Kuperwasser C, List K, Boerner JL. Fibroblast-secreted hepatocyte growth factor mediates epidermal growth factor receptor tyrosine kinase inhibitor resistance in triple-negative breast cancers through paracrine activation of Met. Breast Cancer Res. 2012;14:R104.PubMedPubMedCentralCrossRef
110.
go back to reference Yoshida T, Ishii G, Goto K, Neri S, Hashimoto H, Yoh K, Niho S, Umemura S, Matsumoto S, Ohmatsu H, et al. Podoplanin-positive cancer-associated fibroblasts in the tumor microenvironment induce primary resistance to EGFR-TKIs in lung adenocarcinoma with EGFR mutation. Clin Cancer Res. 2015;21:642–51.PubMedCrossRef Yoshida T, Ishii G, Goto K, Neri S, Hashimoto H, Yoh K, Niho S, Umemura S, Matsumoto S, Ohmatsu H, et al. Podoplanin-positive cancer-associated fibroblasts in the tumor microenvironment induce primary resistance to EGFR-TKIs in lung adenocarcinoma with EGFR mutation. Clin Cancer Res. 2015;21:642–51.PubMedCrossRef
111.
go back to reference Vaquero J, Lobe C, Tahraoui S, Claperon A, Mergey M, Merabtene F, Wendum D, Coulouarn C, Housset C, Desbois-Mouthon C, et al. The IGF2/IR/IGF1R Pathway in Tumor Cells and Myofibroblasts Mediates Resistance to EGFR Inhibition in Cholangiocarcinoma. Clin Cancer Res. 2018;24:4282–96.PubMedCrossRef Vaquero J, Lobe C, Tahraoui S, Claperon A, Mergey M, Merabtene F, Wendum D, Coulouarn C, Housset C, Desbois-Mouthon C, et al. The IGF2/IR/IGF1R Pathway in Tumor Cells and Myofibroblasts Mediates Resistance to EGFR Inhibition in Cholangiocarcinoma. Clin Cancer Res. 2018;24:4282–96.PubMedCrossRef
112.
go back to reference Kharaziha P, Rodriguez P, Li Q, Rundqvist H, Björklund AC, Augsten M, Ullén A, Egevad L, Wiklund P, Nilsson S, et al. Targeting of distinct signaling cascades and cancer-associated fibroblasts define the efficacy of Sorafenib against prostate cancer cells. Cell Death Dis. 2012;3:e262.PubMedPubMedCentralCrossRef Kharaziha P, Rodriguez P, Li Q, Rundqvist H, Björklund AC, Augsten M, Ullén A, Egevad L, Wiklund P, Nilsson S, et al. Targeting of distinct signaling cascades and cancer-associated fibroblasts define the efficacy of Sorafenib against prostate cancer cells. Cell Death Dis. 2012;3:e262.PubMedPubMedCentralCrossRef
113.
go back to reference Chen Y, Huang Y, Reiberger T, Duyverman AM, Huang P, Samuel R, Hiddingh L, Roberge S, Koppel C, Lauwers GY, et al. Differential effects of sorafenib on liver versus tumor fibrosis mediated by stromal-derived factor 1 alpha/C-X-C receptor type 4 axis and myeloid differentiation antigen-positive myeloid cell infiltration in mice. Hepatology. 2014;59:1435–47.PubMedCrossRef Chen Y, Huang Y, Reiberger T, Duyverman AM, Huang P, Samuel R, Hiddingh L, Roberge S, Koppel C, Lauwers GY, et al. Differential effects of sorafenib on liver versus tumor fibrosis mediated by stromal-derived factor 1 alpha/C-X-C receptor type 4 axis and myeloid differentiation antigen-positive myeloid cell infiltration in mice. Hepatology. 2014;59:1435–47.PubMedCrossRef
114.
go back to reference Chen Y, Ramjiawan RR, Reiberger T, Ng MR, Hato T, Huang Y, Ochiai H, Kitahara S, Unan EC, Reddy TP, et al. CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology. 2015;61:1591–602.PubMedCrossRef Chen Y, Ramjiawan RR, Reiberger T, Ng MR, Hato T, Huang Y, Ochiai H, Kitahara S, Unan EC, Reddy TP, et al. CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology. 2015;61:1591–602.PubMedCrossRef
115.
go back to reference Song Y, Kim SH, Kim KM, Choi EK, Kim J, Seo HR. Activated hepatic stellate cells play pivotal roles in hepatocellular carcinoma cell chemoresistance and migration in multicellular tumor spheroids. Sci Rep. 2016;6:36750.PubMedPubMedCentralCrossRef Song Y, Kim SH, Kim KM, Choi EK, Kim J, Seo HR. Activated hepatic stellate cells play pivotal roles in hepatocellular carcinoma cell chemoresistance and migration in multicellular tumor spheroids. Sci Rep. 2016;6:36750.PubMedPubMedCentralCrossRef
116.
go back to reference Khawar IA, Park JK, Jung ES, Lee MA, Chang S, Kuh HJ. Three Dimensional Mixed-Cell Spheroids Mimic Stroma-Mediated Chemoresistance and Invasive Migration in hepatocellular carcinoma. Neoplasia. 2018;20:800–12.PubMedPubMedCentralCrossRef Khawar IA, Park JK, Jung ES, Lee MA, Chang S, Kuh HJ. Three Dimensional Mixed-Cell Spheroids Mimic Stroma-Mediated Chemoresistance and Invasive Migration in hepatocellular carcinoma. Neoplasia. 2018;20:800–12.PubMedPubMedCentralCrossRef
117.
go back to reference Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, Fan J, Cao Y, Dai Z, Zhou J. Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology. 2016;150:1646–+. Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, Fan J, Cao Y, Dai Z, Zhou J. Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology. 2016;150:1646–+.
118.
go back to reference van Oosterwijk JG, Buelow DR, Drenberg CD, Vasilyeva A, Li L, Shi L, Wang YD, Finkelstein D, Shurtleff SA, Janke LJ, et al. Hypoxia-induced upregulation of BMX kinase mediates therapeutic resistance in acute myeloid leukemia. J Clin Invest. 2018;128:369–80. van Oosterwijk JG, Buelow DR, Drenberg CD, Vasilyeva A, Li L, Shi L, Wang YD, Finkelstein D, Shurtleff SA, Janke LJ, et al. Hypoxia-induced upregulation of BMX kinase mediates therapeutic resistance in acute myeloid leukemia. J Clin Invest. 2018;128:369–80.
119.
go back to reference Tang L, Zeng J, Geng PY, Fang CN, Wang Y, Sun MJ, Wang CS, Wang J, Yin PY, Hu CX, et al. Global metabolic profiling identifies a pivotal role of proline and hydroxyproline metabolism in supporting hypoxic response in hepatocellular carcinoma. Clin Cancer Res. 2018;24:474–85.PubMedCrossRef Tang L, Zeng J, Geng PY, Fang CN, Wang Y, Sun MJ, Wang CS, Wang J, Yin PY, Hu CX, et al. Global metabolic profiling identifies a pivotal role of proline and hydroxyproline metabolism in supporting hypoxic response in hepatocellular carcinoma. Clin Cancer Res. 2018;24:474–85.PubMedCrossRef
120.
go back to reference Xing MZ, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, et al. Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. Jama-J Am Med Assoc. 2013;309:1493–501.CrossRef Xing MZ, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, et al. Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. Jama-J Am Med Assoc. 2013;309:1493–501.CrossRef
121.
go back to reference Ruzzolini J, Peppicelli S, Andreucci E, Bianchini F, Margheri F, Laurenzana A, Fibbi G, Pimpinelli N, Calorini L. Everolimus selectively targets vemurafenib resistant BRAF(V600E) melanoma cells adapted to low pH. Cancer Lett. 2017;408:43–54.PubMedCrossRef Ruzzolini J, Peppicelli S, Andreucci E, Bianchini F, Margheri F, Laurenzana A, Fibbi G, Pimpinelli N, Calorini L. Everolimus selectively targets vemurafenib resistant BRAF(V600E) melanoma cells adapted to low pH. Cancer Lett. 2017;408:43–54.PubMedCrossRef
122.
go back to reference Kaur A, Webster MR, Marchbank K, Behera R, Ndoye A, Kugel CH, Dang VM, Appleton J, O'Connell MP, Cheng P, et al. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature. 2016;532:250–4.PubMedPubMedCentralCrossRef Kaur A, Webster MR, Marchbank K, Behera R, Ndoye A, Kugel CH, Dang VM, Appleton J, O'Connell MP, Cheng P, et al. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature. 2016;532:250–4.PubMedPubMedCentralCrossRef
123.
go back to reference Prete A, Lo AS, Sadow PM, Bhasin SS, Antonello ZA, Vodopivec DM, Ullas S, Sims JN, Clohessy JG, Dvorak AM, et al. Pericytes elicit resistance to vemurafenib and sorafenib therapy in thyroid carcinoma via the TSP-1/TGFβ1 axis. Clin Cancer Res. 2018;24:6078–97. Prete A, Lo AS, Sadow PM, Bhasin SS, Antonello ZA, Vodopivec DM, Ullas S, Sims JN, Clohessy JG, Dvorak AM, et al. Pericytes elicit resistance to vemurafenib and sorafenib therapy in thyroid carcinoma via the TSP-1/TGFβ1 axis. Clin Cancer Res. 2018;24:6078–97.
124.
go back to reference Guerrouahen BS, Pasquier J, Kaoud NA, Maleki M, Beauchamp MC, Yasmeen A, Ghiabi P, Lis R, Vidal F, Saleh A, et al. Akt-activated endothelium constitutes the niche for residual disease and resistance to bevacizumab in ovarian cancer. Mol Cancer Ther. 2014;13:3123–36.PubMedCrossRef Guerrouahen BS, Pasquier J, Kaoud NA, Maleki M, Beauchamp MC, Yasmeen A, Ghiabi P, Lis R, Vidal F, Saleh A, et al. Akt-activated endothelium constitutes the niche for residual disease and resistance to bevacizumab in ovarian cancer. Mol Cancer Ther. 2014;13:3123–36.PubMedCrossRef
125.
go back to reference Castro BA, Flanigan P, Jahangiri A, Hoffman D, Chen W, Kuang R, De Lay M, Yagnik G, Wagner JR, Mascharak S, et al. Macrophage migration inhibitory factor downregulation: a novel mechanism of resistance to anti-angiogenic therapy. Oncogene. 2017;36:3749–59. Castro BA, Flanigan P, Jahangiri A, Hoffman D, Chen W, Kuang R, De Lay M, Yagnik G, Wagner JR, Mascharak S, et al. Macrophage migration inhibitory factor downregulation: a novel mechanism of resistance to anti-angiogenic therapy. Oncogene. 2017;36:3749–59.
126.
go back to reference Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al. Exosome-Transmitted lncARSR Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA. Cancer Cell. 2016;29:653–68.PubMedCrossRef Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al. Exosome-Transmitted lncARSR Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA. Cancer Cell. 2016;29:653–68.PubMedCrossRef
127.
go back to reference Finke J, Ko J, Rini B, Rayman P, Ireland J, Cohen P. MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy ☆. Int Immunopharmacol. 2011;11:856–61.PubMedCrossRef Finke J, Ko J, Rini B, Rayman P, Ireland J, Cohen P. MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy ☆. Int Immunopharmacol. 2011;11:856–61.PubMedCrossRef
128.
go back to reference Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, Fuh G, Gerber HP, Ferrara N. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25:911–20.PubMedCrossRef Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, Fuh G, Gerber HP, Ferrara N. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25:911–20.PubMedCrossRef
129.
go back to reference Piao Y, Liang J, Holmes L, Zurita AJ, Henry V, Heymach JV, de Groot JF. Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro-Oncol. 2012;14:1379–92.PubMedPubMedCentralCrossRef Piao Y, Liang J, Holmes L, Zurita AJ, Henry V, Heymach JV, de Groot JF. Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro-Oncol. 2012;14:1379–92.PubMedPubMedCentralCrossRef
130.
go back to reference Achyut BR, Shankar A, Iskander AS, Ara R, Angara K, Zeng P, Knight RA, Scicli AG, Arbab AS. Bone marrow derived myeloid cells orchestrate antiangiogenic resistance in glioblastoma through coordinated molecular networks. Cancer Lett. 2015;369:416–26.PubMedPubMedCentralCrossRef Achyut BR, Shankar A, Iskander AS, Ara R, Angara K, Zeng P, Knight RA, Scicli AG, Arbab AS. Bone marrow derived myeloid cells orchestrate antiangiogenic resistance in glioblastoma through coordinated molecular networks. Cancer Lett. 2015;369:416–26.PubMedPubMedCentralCrossRef
131.
go back to reference Yan D, Kowal J, Akkari L, Schuhmacher AJ, Huse JT, West BL, Joyce JA. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene. 2017;36:6049–58.PubMedPubMedCentralCrossRef Yan D, Kowal J, Akkari L, Schuhmacher AJ, Huse JT, West BL, Joyce JA. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene. 2017;36:6049–58.PubMedPubMedCentralCrossRef
132.
go back to reference Xu X, Zhang X, Liu Y, Yang L, Huang S, Lu L, Wang S, Guo Q, Zhao L. BM microenvironmental protection of CML cells from imatinib through Stat5/NF-kappa B signaling and reversal by Wogonin. Oncotarget. 2016;7:24436–54.PubMedPubMedCentral Xu X, Zhang X, Liu Y, Yang L, Huang S, Lu L, Wang S, Guo Q, Zhao L. BM microenvironmental protection of CML cells from imatinib through Stat5/NF-kappa B signaling and reversal by Wogonin. Oncotarget. 2016;7:24436–54.PubMedPubMedCentral
133.
go back to reference Liu P, Ma D, Yu Z, Zhe N, Ren M, Wang P, Yu M, Huang J, Fang Q, Wang J. Overexpression of heme oxygenase-1 in bone marrow stromal cells promotes microenvironment-mediated imatinib resistance in chronic myeloid leukemia. Biomed Pharmacother. 2017;91:21–30.PubMedCrossRef Liu P, Ma D, Yu Z, Zhe N, Ren M, Wang P, Yu M, Huang J, Fang Q, Wang J. Overexpression of heme oxygenase-1 in bone marrow stromal cells promotes microenvironment-mediated imatinib resistance in chronic myeloid leukemia. Biomed Pharmacother. 2017;91:21–30.PubMedCrossRef
134.
go back to reference Zhao X, Lwin T, Silva A, Shah B, Tao J, Fang B, Zhang L, Fu K, Bi C, Li J, et al. Unification of de novo and acquired ibrutinib resistance in mantle cell lymphoma. Nat Commun. 2017;8:14920.PubMedPubMedCentralCrossRef Zhao X, Lwin T, Silva A, Shah B, Tao J, Fang B, Zhang L, Fu K, Bi C, Li J, et al. Unification of de novo and acquired ibrutinib resistance in mantle cell lymphoma. Nat Commun. 2017;8:14920.PubMedPubMedCentralCrossRef
135.
go back to reference Boissard F, Fournie JJ, Quillet-Mary A, Ysebaert L, Poupot M. Nurse-like cells mediate ibrutinib resistance in chronic lymphocytic leukemia patients. Blood Cancer J. 2015;5:e355.PubMedPubMedCentralCrossRef Boissard F, Fournie JJ, Quillet-Mary A, Ysebaert L, Poupot M. Nurse-like cells mediate ibrutinib resistance in chronic lymphocytic leukemia patients. Blood Cancer J. 2015;5:e355.PubMedPubMedCentralCrossRef
136.
go back to reference Zhang Z, Yu X, Wang Z, Wu P, Huang J. Anthracyclines potentiate anti-tumor immunity: A new opportunity for chemoimmunotherapy. Cancer Lett. 2015;369:331–5.PubMedCrossRef Zhang Z, Yu X, Wang Z, Wu P, Huang J. Anthracyclines potentiate anti-tumor immunity: A new opportunity for chemoimmunotherapy. Cancer Lett. 2015;369:331–5.PubMedCrossRef
137.
go back to reference Xu M, Liu M, Du X, Li S, Li H, Li X, Li Y, Wang Y, Qin Z, Fu YX, Wang S. Intratumoral delivery of IL-21 Overcomes Anti-Her2/Neu resistance through shifting tumor-associated macrophages from M2 to M1 phenotype. J Immunol. 2015;194:4997–5006.PubMedCrossRef Xu M, Liu M, Du X, Li S, Li H, Li X, Li Y, Wang Y, Qin Z, Fu YX, Wang S. Intratumoral delivery of IL-21 Overcomes Anti-Her2/Neu resistance through shifting tumor-associated macrophages from M2 to M1 phenotype. J Immunol. 2015;194:4997–5006.PubMedCrossRef
138.
go back to reference Lam C, Ferguson ID, Mariano MC, Lin YT, Murnane M, Liu H, Smith GA, Wong SW, Taunton J, Liu JO, et al. Repurposing tofacitinib as an anti-myeloma therapeutic to reverse growth-promoting effects of the bone marrow microenvironment. Haematologica. 2018;103:1218–28. Lam C, Ferguson ID, Mariano MC, Lin YT, Murnane M, Liu H, Smith GA, Wong SW, Taunton J, Liu JO, et al. Repurposing tofacitinib as an anti-myeloma therapeutic to reverse growth-promoting effects of the bone marrow microenvironment. Haematologica. 2018;103:1218–28.
139.
go back to reference Hu K, Miao L, Goodwin TJ, Li J, Liu Q, Huang L. Quercetin remodels the tumor microenvironment to improve the permeation, retention, and antitumor effects of nanoparticles. ACS Nano. 2017;11:4916–25. Hu K, Miao L, Goodwin TJ, Li J, Liu Q, Huang L. Quercetin remodels the tumor microenvironment to improve the permeation, retention, and antitumor effects of nanoparticles. ACS Nano. 2017;11:4916–25.
140.
go back to reference Amini MA, Abbasi AZ, Cai P, Lip H, Gordijo CR, Li J, Chen B, Zhang L, Rauth AM, Wu XY. Combining Tumor Microenvironment Modulating Nanoparticles with Doxorubicin to Enhance Chemotherapeutic Efficacy and Boost Antitumor Immunity. J Natl Cancer Inst. 2019;111. Amini MA, Abbasi AZ, Cai P, Lip H, Gordijo CR, Li J, Chen B, Zhang L, Rauth AM, Wu XY. Combining Tumor Microenvironment Modulating Nanoparticles with Doxorubicin to Enhance Chemotherapeutic Efficacy and Boost Antitumor Immunity. J Natl Cancer Inst. 2019;111.
141.
go back to reference Fang J, Zhang S, Xue X, Zhu X, Song S, Wang B, Jiang L, Qin M, Liang H, Gao L. Quercetin and doxorubicin co-delivery using mesoporous silica nanoparticles enhance the efficacy of gastric carcinoma chemotherapy. Int J Nanomedicine. 2018;13:5113–26.PubMedPubMedCentralCrossRef Fang J, Zhang S, Xue X, Zhu X, Song S, Wang B, Jiang L, Qin M, Liang H, Gao L. Quercetin and doxorubicin co-delivery using mesoporous silica nanoparticles enhance the efficacy of gastric carcinoma chemotherapy. Int J Nanomedicine. 2018;13:5113–26.PubMedPubMedCentralCrossRef
142.
go back to reference Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920–6. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920–6.
143.
go back to reference Lampis A, Carotenuto P, Vlachogiannis G, Cascione L, Hedayat S, Burke R, Clarke P, Bosma E, Simbolo M, Scarpa A, et al. MIR21 drives resistance to heat shock protein 90 inhibition in cholangiocarcinoma. Gastroenterology. 2018;154:1066–+.PubMedCrossRef Lampis A, Carotenuto P, Vlachogiannis G, Cascione L, Hedayat S, Burke R, Clarke P, Bosma E, Simbolo M, Scarpa A, et al. MIR21 drives resistance to heat shock protein 90 inhibition in cholangiocarcinoma. Gastroenterology. 2018;154:1066–+.PubMedCrossRef
145.
go back to reference Mazzarella L, Curigliano G. A new approach to assess drug sensitivity in cells for novel drug discovery. Expert Opin Drug Discov. 2018;13:339–46.PubMedCrossRef Mazzarella L, Curigliano G. A new approach to assess drug sensitivity in cells for novel drug discovery. Expert Opin Drug Discov. 2018;13:339–46.PubMedCrossRef
146.
go back to reference Cloosen S, Gratama J, van Leeuwen EBM, Senden-Gijsbers bLMG, Oving EBH, von Mensdorff-Pouilly S, Tarp MA, Mandel U, Clausen H, Germeraad WTV, Bos GMJ. Cancer specific Mucin-1 glycoforms are expressed on multiple myeloma. Br J Haematol. 2006;135:513–6.PubMedCrossRef Cloosen S, Gratama J, van Leeuwen EBM, Senden-Gijsbers bLMG, Oving EBH, von Mensdorff-Pouilly S, Tarp MA, Mandel U, Clausen H, Germeraad WTV, Bos GMJ. Cancer specific Mucin-1 glycoforms are expressed on multiple myeloma. Br J Haematol. 2006;135:513–6.PubMedCrossRef
147.
go back to reference Yin L, Ahmad R, Kosugi M, Kufe T, Vasir B, Avigan D, Kharbanda S, Kufe D. Survival of human multiple myeloma cells is dependent on MUC1 C-terminal transmembrane subunit oncoprotein function. Mol Pharmacol. 2010;78:166–74.PubMedPubMedCentralCrossRef Yin L, Ahmad R, Kosugi M, Kufe T, Vasir B, Avigan D, Kharbanda S, Kufe D. Survival of human multiple myeloma cells is dependent on MUC1 C-terminal transmembrane subunit oncoprotein function. Mol Pharmacol. 2010;78:166–74.PubMedPubMedCentralCrossRef
148.
go back to reference Kharbanda A, Rajabi H, Jin C, Tchaicha J, Kikuchi E, Wong K-K, Kufe D. Targeting the oncogenic MUC1-C protein inhibits mutant EGFR-mediated signaling and survival in non-small cell lung cancer cells. Clin Cancer Re. 2014;20:5423–34.CrossRef Kharbanda A, Rajabi H, Jin C, Tchaicha J, Kikuchi E, Wong K-K, Kufe D. Targeting the oncogenic MUC1-C protein inhibits mutant EGFR-mediated signaling and survival in non-small cell lung cancer cells. Clin Cancer Re. 2014;20:5423–34.CrossRef
149.
go back to reference Emeagi PU, Maenhout S, Dang N, Heirman C, Thielemans K, Breckpot K. Downregulation of Stat3 in melanoma: reprogramming the immune microenvironment as an anticancer therapeutic strategy. Gene Therapy. 2013;20:1085–92.PubMedCrossRef Emeagi PU, Maenhout S, Dang N, Heirman C, Thielemans K, Breckpot K. Downregulation of Stat3 in melanoma: reprogramming the immune microenvironment as an anticancer therapeutic strategy. Gene Therapy. 2013;20:1085–92.PubMedCrossRef
150.
go back to reference Takahashi S. Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol. 2011;4:13. Takahashi S. Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol. 2011;4:13.
151.
go back to reference Buchner M, Baer C, Prinz G, Dierks C, Burger M, Zenz T, Stilgenbauer S, Jumaa H, Veelken H, Zirlik K. Spleen tyrosine kinase inhibition prevents chemokine- and integrin-mediated stromal protective effects in chronic lymphocytic leukemia. Blood. 2010;115:4497–506.PubMedCrossRef Buchner M, Baer C, Prinz G, Dierks C, Burger M, Zenz T, Stilgenbauer S, Jumaa H, Veelken H, Zirlik K. Spleen tyrosine kinase inhibition prevents chemokine- and integrin-mediated stromal protective effects in chronic lymphocytic leukemia. Blood. 2010;115:4497–506.PubMedCrossRef
152.
go back to reference Koch H, Wilhelm M, Ruprecht B, Beck S, Frejno M, Klaeger S, Kuster B. Phosphoproteome profiling reveals molecular mechanisms of growth-factor-mediated kinase inhibitor resistance in EGFR-overexpressing cancer cells. J Proteome Res. 2016;15:4490–504.PubMedCrossRef Koch H, Wilhelm M, Ruprecht B, Beck S, Frejno M, Klaeger S, Kuster B. Phosphoproteome profiling reveals molecular mechanisms of growth-factor-mediated kinase inhibitor resistance in EGFR-overexpressing cancer cells. J Proteome Res. 2016;15:4490–504.PubMedCrossRef
153.
go back to reference Murakami A, Takahashi F, Nurwidya F, Kobayashi I, Minakata K, Hashimoto M, Nara T, Kato M, Tajima K, Shimada N, et al. Hypoxia increases gefitinib-resistant lung cancer stem cells through the activation of insulin-like growth factor 1 receptor. Plos One. 2014;9:e86459. Murakami A, Takahashi F, Nurwidya F, Kobayashi I, Minakata K, Hashimoto M, Nara T, Kato M, Tajima K, Shimada N, et al. Hypoxia increases gefitinib-resistant lung cancer stem cells through the activation of insulin-like growth factor 1 receptor. Plos One. 2014;9:e86459.
154.
go back to reference Lee J, Condello S, Yakubov B, Emerson R, Caperell-Grant A, Hitomi K, Xie J, Matei D. Tissue transglutaminase mediated tumor-stroma interaction promotes pancreatic cancer progression. Clin Cancer Res. 2015;21:4482–93. Lee J, Condello S, Yakubov B, Emerson R, Caperell-Grant A, Hitomi K, Xie J, Matei D. Tissue transglutaminase mediated tumor-stroma interaction promotes pancreatic cancer progression. Clin Cancer Res. 2015;21:4482–93.
155.
go back to reference Guo L, Zheng J, Yu T, Liu Y, Duo L. Elevated expression of SATB1 is involved in pancreatic tumorigenesis and is associated with poor patient survival. Mol Med Rep. 2017;16:8842–8.PubMedPubMedCentralCrossRef Guo L, Zheng J, Yu T, Liu Y, Duo L. Elevated expression of SATB1 is involved in pancreatic tumorigenesis and is associated with poor patient survival. Mol Med Rep. 2017;16:8842–8.PubMedPubMedCentralCrossRef
156.
go back to reference Weizman N, Krelin Y, Shabtay-Orbach A, Amit M, Binenbaum Y, Wong RJ, Gil Z. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene. 2014;33:3812–9.CrossRefPubMed Weizman N, Krelin Y, Shabtay-Orbach A, Amit M, Binenbaum Y, Wong RJ, Gil Z. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene. 2014;33:3812–9.CrossRefPubMed
157.
go back to reference Hamada S, Masamune A, Miura S, Satoh K, Shimosegawa T. MiR-365 induces gemcitabine resistance in pancreatic cancer cells by targeting the adaptor protein SHC1 and pro-apoptotic regulator BAX. Cellular Signalling. 2014;26:179–85.PubMedCrossRef Hamada S, Masamune A, Miura S, Satoh K, Shimosegawa T. MiR-365 induces gemcitabine resistance in pancreatic cancer cells by targeting the adaptor protein SHC1 and pro-apoptotic regulator BAX. Cellular Signalling. 2014;26:179–85.PubMedCrossRef
158.
go back to reference Grugan KD, Miller CG, Yao Y, Michaylira CZ, Ohashi S, Klein-Szanto AJ, Diehl A, Herlyn M, Han M, Nakagawa H, Rustgi AK. Fibroblast-secreted hepatocyte growth factor plays a functional role in esophageal squamous cell carcinoma invasion. Proc Natl Acad Sci U S A. 2010;107:11026–31.PubMedPubMedCentralCrossRef Grugan KD, Miller CG, Yao Y, Michaylira CZ, Ohashi S, Klein-Szanto AJ, Diehl A, Herlyn M, Han M, Nakagawa H, Rustgi AK. Fibroblast-secreted hepatocyte growth factor plays a functional role in esophageal squamous cell carcinoma invasion. Proc Natl Acad Sci U S A. 2010;107:11026–31.PubMedPubMedCentralCrossRef
159.
go back to reference Siddiquee KAZ, Turkson J. STAT3 as a target for inducing apoptosis in solid and hematological tumors. Cell Res. 2008;18:254–67.CrossRef Siddiquee KAZ, Turkson J. STAT3 as a target for inducing apoptosis in solid and hematological tumors. Cell Res. 2008;18:254–67.CrossRef
160.
go back to reference Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem. 2000;275:36803–10.PubMedCrossRef Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem. 2000;275:36803–10.PubMedCrossRef
161.
go back to reference Zhou S-L, Zhou Z-J, Hu Z-Q, Li X, Huang X-W, Wang Z, Fan J, Dai Z, Zhou J. CXCR2/CXCL5 axis contributes to epithelial–mesenchymal transition of HCC cells through activating PI3K/Akt/GSK-3β/Snail signaling. Cancer Lett. 2015;358:124–35.PubMedCrossRef Zhou S-L, Zhou Z-J, Hu Z-Q, Li X, Huang X-W, Wang Z, Fan J, Dai Z, Zhou J. CXCR2/CXCL5 axis contributes to epithelial–mesenchymal transition of HCC cells through activating PI3K/Akt/GSK-3β/Snail signaling. Cancer Lett. 2015;358:124–35.PubMedCrossRef
162.
go back to reference Zhou SL, Dai Z, Zhou ZJ, Chen Q, Wang Z, Xiao YS, Hu ZQ, Huang XY, Yang GH, Shi YH, et al. CXCL5 contributes to tumor metastasis and recurrence of intrahepatic cholangiocarcinoma by recruiting infiltrative intratumoral neutrophils. Carcinogenesis. 2014;35:597–605.PubMedCrossRef Zhou SL, Dai Z, Zhou ZJ, Chen Q, Wang Z, Xiao YS, Hu ZQ, Huang XY, Yang GH, Shi YH, et al. CXCL5 contributes to tumor metastasis and recurrence of intrahepatic cholangiocarcinoma by recruiting infiltrative intratumoral neutrophils. Carcinogenesis. 2014;35:597–605.PubMedCrossRef
163.
go back to reference Hsieh CH, Lee CH, Liang JA, Yu CY, Shyu WC. Cycling hypoxia increases U87 glioma cell radioresistance via ROS induced higher and long-term HIF-1 signal transduction activity. Oncol Rep. 2010;24:1629–36.PubMedCrossRef Hsieh CH, Lee CH, Liang JA, Yu CY, Shyu WC. Cycling hypoxia increases U87 glioma cell radioresistance via ROS induced higher and long-term HIF-1 signal transduction activity. Oncol Rep. 2010;24:1629–36.PubMedCrossRef
164.
go back to reference Gielen PR, Aftab Q, Ma N, Chen VC, Hong XT, Lozinsky S, Naus CC. Sin WC: Connexin43 confers Temozolomide resistance in human glioma cells by modulating the mitochondrial apoptosis pathway. Neuropharmacology. 2013;75:539–48.PubMedCrossRef Gielen PR, Aftab Q, Ma N, Chen VC, Hong XT, Lozinsky S, Naus CC. Sin WC: Connexin43 confers Temozolomide resistance in human glioma cells by modulating the mitochondrial apoptosis pathway. Neuropharmacology. 2013;75:539–48.PubMedCrossRef
165.
go back to reference Xu GY, Li JY. Differential expression of PDGFRB and EGFR in microvascular proliferation in glioblastoma. Tumor Biol. 2016;37:10577–86.CrossRef Xu GY, Li JY. Differential expression of PDGFRB and EGFR in microvascular proliferation in glioblastoma. Tumor Biol. 2016;37:10577–86.CrossRef
Metadata
Title
Tumor microenvironment-driven non-cell-autonomous resistance to antineoplastic treatment
Authors
Yidi Qu
Bo Dou
Horyue Tan
Yibin Feng
Ning Wang
Di Wang
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2019
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-019-0992-4

Other articles of this Issue 1/2019

Molecular Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine