Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. World most cancers statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 nations. CA Most cancers J Clin. 2021;71:209–49.
Sandhu S, Moore CM, Chiong E, Beltran H, Bristow RG, Williams SG. Prostate most cancers. Lancet. 2021;398:1075–90.
Davies A, Conteduca V, Zoubeidi A, Beltran H. Organic evolution of castration-resistant prostate most cancers. Eur Urol Focus. 2019;5:147–54.
Fizazi Okay, Scher HI, Molina A, Logothetis CJ, Chi KN, Jones RJ, et al. Abiraterone acetate for remedy of metastatic castration-resistant prostate most cancers: ultimate general survival evaluation of the COU-AA-301 randomised, double-blind, placebo-controlled part 3 research. Lancet Oncol. 2012;13:983–92.
Chi KN, Agarwal N, Bjartell A, Chung BH, Pereira de Santana Gomes AJ, Given R, et al. Apalutamide for metastatic, castration-sensitive prostate most cancers. N Engl J Med. 2019;381:13–24.
Sharma P, Goswami S, Raychaudhuri D, Siddiqui BA, Singh P, Nagarajan A, et al. Immune checkpoint therapy-current views and future instructions. Cell. 2023;186:1652–69.
Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate most cancers. N Engl J Med. 2010;363:411–22.
Beer TM, Kwon ED, Drake CG, Fizazi Okay, Logothetis C, Gravis G, et al. Randomized, double-blind, part iii trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic sufferers with metastatic chemotherapy-naive castration-resistant prostate most cancers. J Clin Oncol. 2017;35:40–47.
Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa Okay, Hoimes CJ, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate most cancers: multicohort, open-label part II KEYNOTE-199 research. J Clin Oncol. 2020;38:395–405.
Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch restore deficiency predicts response of stable tumors to PD-1 blockade. Science. 2017;357:409–13.
Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Evaluation of the prevalence of microsatellite instability in prostate most cancers and response to immune checkpoint blockade. JAMA Oncol. 2019;5:471–8.
Fang B, Wei Y, Zeng H, Li Y, Chen S, Zhang T, et al. Prevalence of mismatch restore genes mutations and scientific exercise of PD-1 remedy in Chinese language prostate most cancers sufferers. Most cancers Immunol Immunother. 2023;72:1541–51.
Obradovic AZ, Dallos MC, Zahurak ML, Partin AW, Schaeffer EM, Ross AE, et al. T-cell infiltration and adaptive treg resistance in response to androgen deprivation with or with out vaccination in localized prostate most cancers. Clin Most cancers Res. 2020;26:3182–92.
Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response fee to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.
Bejarano L, Jordao MJC, Joyce JA. Therapeutic focusing on of the tumor microenvironment. Most cancers Discov. 2021;11:933–59.
Han C, Deng Y, Xu W, Liu Z, Wang T, Wang S, et al. The roles of tumor-associated macrophages in prostate most cancers. J Oncol. 2022;2022:8580043.
Owen JS, Clayton A, Pearson HB. Most cancers-associated fibroblast heterogeneity, activation and performance: implications for prostate most cancers. Biomolecules. 2022;13:67.
Sieminska I, Baran J. Myeloid-derived suppressor cells as key gamers and promising remedy targets in prostate most cancers. Entrance Oncol. 2022;12:862416.
Murray PJ, Wynn TA. Protecting and pathogenic features of macrophage subsets. Nat Rev Immunol. 2011;11:723–37.
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage plasticity, polarization, and performance in well being and illness. J Cell Physiol. 2018;233:6425–40.
Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as remedy targets in oncology. Nat Rev Clin Oncol. 2017;14:399–416.
Huang R, Wang S, Wang N, Zheng Y, Zhou J, Yang B, et al. CCL5 derived from tumor-associated macrophages promotes prostate most cancers stem cells and metastasis through activating beta-catenin/STAT3 signaling. Cell Demise Dis. 2020;11:234.
Lee GT, Kwon SJ, Lee JH, Jeon SS, Jang KT, Choi HY, et al. Macrophages induce neuroendocrine differentiation of prostate most cancers cells through BMP6-IL6 Loop. Prostate. 2011;71:1525–37.
Li XF, Selli C, Zhou HL, Cao J, Wu S, Ma RY, et al. Macrophages promote anti-androgen resistance in prostate most cancers bone illness. J Exp Med. 2023;220:e20221007.
Nonomura N, Takayama H, Nakayama M, Nakai Y, Kawashima A, Mukai M, et al. Infiltration of tumour-associated macrophages in prostate biopsy specimens is predictive of illness development after hormonal remedy for prostate most cancers. BJU Int. 2011;107:1918–22.
Zarif JC, Baena-Del Valle JA, Hicks JL, Heaphy CM, Vidal I, Luo J, et al. Mannose receptor-positive macrophage infiltration correlates with prostate most cancers onset and metastatic castration-resistant illness. Eur Urol Oncol. 2019;2:429–36.
Kfoury Y, Baryawno N, Extreme N, Mei S, Gustafsson Okay, Hirz T, et al. Human prostate most cancers bone metastases have an actionable immunosuppressive microenvironment. Most cancers Cell. 2021;39:1464–78.e1468.
Masetti M, Carriero R, Portale F, Marelli G, Morina N, Pandini M, et al. Lipid-loaded tumor-associated macrophages maintain tumor development and invasiveness in prostate most cancers. J Exp Med. 2022;219:e20210564.
Wang C, Peng G, Huang H, Liu F, Kong DP, Dong KQ, et al. Blocking the suggestions loop between neuroendocrine differentiation and macrophages improves the therapeutic results of enzalutamide (MDV3100) on prostate most cancers. Clin Most cancers Res. 2018;24:708–23.
Spiess PE, Pettaway CA, Vakar-Lopez F, Kassouf W, Wang X, Busby JE, et al. Therapy outcomes of small cell carcinoma of the prostate: a single-center research. Most cancers. 2007;110:1729–37.
Chen X, Tune E. Turning foes to mates: focusing on cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18:99–115.
ChallaSivaKanaka S, Vickman RE, Kakarla M, Hayward SW, Franco OE. Fibroblast heterogeneity in prostate carcinogenesis. Most cancers Lett. 2022;525:76–83.
Biffi G, Tuveson DA. Range and biology of cancer-associated fibroblasts. Physiol Rev. 2021;101:147–76.
Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, et al. Cross-species single-cell evaluation of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Most cancers Discov. 2019;9:1102–23.
Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic most cancers. J Exp Med. 2017;214:579–96.
Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Ideas of extracellular matrix remodelling in tumour development and metastasis. Nat Commun. 2020;11:5120.
Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R, et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce growth of regulatory T cells in pancreatic most cancers. Most cancers Cell. 2022;40:656–73.e657.
Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor development of initiated human prostatic epithelium. Most cancers Res. 1999;59:5002–11.
Jaeschke A, Jacobi A, Lawrence MG, Risbridger GP, Frydenberg M, Williams ED, et al. Most cancers-associated fibroblasts of the prostate promote a compliant and extra invasive phenotype in benign prostate epithelial cells. Mater At the moment Bio. 2020;8:100073.
Bedeschi M, Marino N, Cavassi E, Piccinini F, Tesei A. Most cancers-associated fibroblast: function in prostate most cancers development to metastatic illness and therapeutic resistance. Cells. 2023;12:802.
Gleave M, Hsieh JT, Gao CA, von Eschenbach AC, Chung LW. Acceleration of human prostate most cancers development in vivo by elements produced by prostate and bone fibroblasts. Most cancers Res. 1991;51:3753–61.
Thalmann GN, Rhee H, Sikes RA, Pathak S, Multani A, Zhau HE, et al. Human prostate fibroblasts induce development and confer castration resistance and metastatic potential in LNCaP cells. Eur Urol. 2010;58:162–71.
Giannoni E, Bianchini F, Masieri L, Serni S, Torre E, Calorini L, et al. Reciprocal activation of prostate most cancers cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and most cancers stemness. Most cancers Res. 2010;70:6945–56.
Liu X, Tang J, Peng L, Nie H, Zhang Y, Liu P. Most cancers-associated fibroblasts promote malignant phenotypes of prostate most cancers cells through autophagy : cancer-associated fibroblasts promote prostate most cancers growth. Apoptosis. 2023;28:881–91.
Zhang Z, Karthaus WR, Lee YS, Gao VR, Wu C, Russo JW, et al. Tumor microenvironment-derived NRG1 promotes antiandrogen resistance in prostate most cancers. Most cancers Cell. 2020;38:279–96.e279.
Wang H, Li N, Liu Q, Guo J, Pan Q, Cheng B, et al. Antiandrogen remedy induces stromal cell reprogramming to advertise castration resistance in prostate most cancers. Most cancers Cell. 2023;41:1345–62.e1349.
Solar Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, et al. Therapy-induced harm to the tumor microenvironment promotes prostate most cancers remedy resistance by way of WNT16B. Nat Med. 2012;18:1359–68.
Shan G, Gu J, Zhou D, Li L, Cheng W, Wang Y, et al. Most cancers-associated fibroblast-secreted exosomal miR-423-5p promotes chemotherapy resistance in prostate most cancers by focusing on GREM2 by way of the TGF-beta signaling pathway. Exp Mol Med. 2020;52:1809–22.
Blom S, Erickson A, Ostman A, Rannikko A, Mirtti T, Kallioniemi O, et al. Fibroblast as a important stromal cell sort figuring out prognosis in prostate most cancers. Prostate. 2019;79:1505–13.
Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol. 2018;19:108–19.
Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The character of myeloid-derived suppressor cells within the tumor microenvironment. Developments Immunol. 2016;37:208–20.
Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct goal of HIF-1alpha, and its blockade underneath hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211:781–90.
Wang L, Jia B, Claxton DF, Ehmann WC, Rybka WB, Mineishi S, et al. VISTA is extremely expressed on MDSCs and mediates an inhibition of T cell response in sufferers with AML. Oncoimmunology. 2018;7:e1469594.
Limagne E, Richard C, Thibaudin M, Fumet JD, Truntzer C, Lagrange A, et al. Tim-3/galectin-9 pathway and mMDSC management main and secondary resistances to PD-1 blockade in lung most cancers sufferers. Oncoimmunology. 2019;8:e1564505.
Salminen A. Immunosuppressive community promotes immunosenescence related to growing old and continual inflammatory circumstances. J Mol Med. 2021;99:1553–69.
Ohl Okay, Tenbrock Okay. Reactive oxygen species as regulators of MDSC-mediated immune suppression. Entrance Immunol. 2018;9:2499.
Lu T, Ramakrishnan R, Altiok S, Youn JI, Cheng P, Celis E, et al. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J Clin Make investments. 2011;121:4015–29.
Mojsilovic S, Mojsilovic SS, Bjelica S, Santibanez JF. Reworking development factor-beta1 and myeloid-derived suppressor cells: a cancerous partnership. Dev Dyn. 2022;251:105–24.
Guan Q, Yang B, Warrington RJ, Mink S, Kalicinsky C, Becker AB, et al. Myeloid-derived suppressor cells: roles and relations with Th2, Th17, and Treg cells in bronchial asthma. Allergy. 2019;74:2233–7.
Groth C, Hu X, Weber R, Fleming V, Altevogt P, Utikal J, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) throughout tumour development. Br J Most cancers. 2019;120:16–25.
Garcia AJ, Ruscetti M, Arenzana TL, Tran LM, Bianci-Frias D, Sybert E, et al. Pten null prostate epithelium promotes localized myeloid-derived suppressor cell growth and immune suppression throughout tumor initiation and development. Mol Cell Biol. 2014;34:2017–28.
Zhao D, Cai L, Lu X, Liang X, Li J, Chen P, et al. Chromatin regulator CHD1 remodels the immunosuppressive tumor microenvironment in PTEN-deficient prostate most cancers. Most cancers Discov. 2020;10:1374–87.
Li N, Liu Q, Han Y, Pei S, Cheng B, Xu J, et al. ARID1A loss induces polymorphonuclear myeloid-derived suppressor cell chemotaxis and promotes prostate most cancers development. Nat Commun. 2022;13:7281.
Calcinotto A, Spataro C, Zagato E, Di Mitri D, Gil V, Crespo M, et al. IL-23 secreted by myeloid cells drives castration-resistant prostate most cancers. Nature. 2018;559:363–9.
Consiglio CR, Udartseva O, Ramsey KD, Bush C, Gollnick SO. Enzalutamide, an androgen receptor antagonist, enhances myeloid cell-mediated immune suppression and tumor development. Most cancers Immunol Res. 2020;8:1215–27.
Vuk-Pavlovic S, Bulur PA, Lin Y, Qin R, Szumlanski CL, Zhao X, et al. Immunosuppressive CD14+HLA-DRlow/- monocytes in prostate most cancers. Prostate. 2010;70:443–55.
Idorn M, Kollgaard T, Kongsted P, Sengelov L, Thor Straten P. Correlation between frequencies of blood monocytic myeloid-derived suppressor cells, regulatory T cells and detrimental prognostic markers in sufferers with castration-resistant metastatic prostate most cancers. Most cancers Immunol Immunother. 2014;63:1177–87.
Bronte G, Conteduca V, Landriscina M, Procopio AD. Circulating myeloid-derived suppressor cells and survival in prostate most cancers sufferers: systematic overview and meta-analysis. Prostate Most cancers Prostatic Dis. 2023;26:41–6.
Locati M, Curtale G, Mantovani A. Range, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. 2020;15:123–47.
Singh S, Anshita D, Ravichandiran V. MCP-1: perform, regulation, and involvement in illness. Int Immunopharmacol. 2021;101:107598.
Zhang J, Patel L, Pienta KJ. CC chemokine ligand 2 (CCL2) promotes prostate most cancers tumorigenesis and metastasis. Cytokine Progress Issue Rev. 2010;21:41–8.
Hao Q, Vadgama JV, Wang P. CCL2/CCR2 signaling in most cancers pathogenesis. Cell Commun Sign. 2020;18:82.
Noel M, O’Reilly EM, Wolpin BM, Ryan DP, Bullock AJ, Britten CD, et al. Section 1b research of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) together with nab-paclitaxel/gemcitabine in first-line remedy of metastatic pancreatic ductal adenocarcinoma. Make investments N. Medicine. 2020;38:800–11.
Pienta KJ, Machiels JP, Schrijvers D, Alekseev B, Shkolnik M, Crabb SJ, et al. Section 2 research of carlumab (CNTO 888), a human monoclonal antibody towards CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate most cancers. Make investments N. Medicine. 2013;31:760–8.
Stanley ER, Chitu V. CSF-1 receptor signaling in myeloid cells. Chilly Spring Harb Perspect Biol. 2014;6:a021857.
Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH, Ruttinger D. Colony-stimulating issue 1 receptor (CSF1R) inhibitors in most cancers remedy. J Immunother Most cancers. 2017;5:53.
Kluger HM, Dolled-Filhart M, Rodov S, Kacinski BM, Camp RL, Rimm DL. Macrophage colony-stimulating factor-1 receptor expression is related to poor final result in breast most cancers by massive cohort tissue microarray evaluation. Clin Most cancers Res. 2004;10:173–7.
Li Okay, Xu W, Lu Okay, Wen Y, Xin T, Shen Y, et al. CSF-1R inhibition disrupts the dialog between leukaemia cells and macrophages and delays leukaemia development. J Cell Mol Med. 2020;24:13115–28.
Wang C, Xia B, Wang T, Tian C, Yu Y, Wu X, et al. PD-1, FOXP3, and CSF-1R expression in sufferers with Hodgkin lymphoma and their prognostic worth. Int J Clin Exp Pathol. 2018;11:1923–34.
Ide H, Seligson DB, Memarzadeh S, Xin L, Horvath S, Dubey P, et al. Expression of colony-stimulating issue 1 receptor throughout prostate growth and prostate most cancers development. Proc Natl Acad Sci USA. 2002;99:14404–9.
Richardsen E, Uglehus RD, Due J, Busch C, Busund LT. The prognostic affect of M-CSF, CSF-1 receptor, CD68 and CD3 in prostatic carcinoma. Histopathology. 2008;53:30–38.
Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, et al. Most cancers-associated fibroblasts neutralize the anti-tumor impact of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Most cancers Cell. 2017;32:654–68.e655.
Escamilla J, Schokrpur S, Liu C, Priceman SJ, Moughon D, Jiang Z, et al. CSF1 receptor focusing on in prostate most cancers reverses macrophage-mediated resistance to androgen blockade remedy. Most cancers Res. 2015;75:950–62.
Xu J, Escamilla J, Mok S, David J, Priceman S, West B, et al. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate most cancers. Most cancers Res. 2013;73:2782–94.
Siddiqui BA, Chapin BF, Jindal S, Duan F, Basu S, Yadav SS, et al. Immune and pathologic responses in sufferers with localized prostate most cancers who obtained daratumumab (anti-CD38) or edicotinib (CSF-1R inhibitor). J Immunother Most cancers. 2023;11:e006262.
Mehta AR, Armstrong AJ. Tasquinimod within the remedy of castrate-resistant prostate most cancers – present standing and future prospects. Ther Adv Urol. 2016;8:9–18.
Shen L, Sundstedt A, Ciesielski M, Miles KM, Celander M, Adelaiye R, et al. Tasquinimod modulates suppressive myeloid cells and enhances most cancers immunotherapies in murine fashions. Most cancers Immunol Res. 2015;3:136–48.
Sternberg C, Armstrong A, Pili R, Ng S, Huddart R, Agarwal N, et al. Randomized, double-blind, placebo-controlled part III research of tasquinimod in males with metastatic castration-resistant prostate most cancers. J Clin Oncol. 2016;34:2636–43.
Fadok VA, Voelker DR, Campbell PA, Cohen JJ, Bratton DL, Henson PM. Publicity of phosphatidylserine on the floor of apoptotic lymphocytes triggers particular recognition and removing by macrophages. J Immunol. 1992;148:2207–16.
Yin Y, Huang X, Lynn KD, Thorpe PE. Phosphatidylserine-targeting antibody induces M1 macrophage polarization and promotes myeloid-derived suppressor cell differentiation. Most cancers Immunol Res. 2013;1:256–68.
Jin H, He Y, Zhao P, Hu Y, Tao J, Chen J, et al. Concentrating on lipid metabolism to beat EMT-associated drug resistance through integrin beta3/FAK pathway and tumor-associated macrophage repolarization utilizing legumain-activatable supply. Theranostics. 2019;9:265–78.
Chaudagar Okay, Hieromnimon HM, Khurana R, Labadie B, Hirz T, Mei S, et al. Reversal of lactate and PD-1-mediated macrophage immunosuppression controls development of PTEN/p53-deficient prostate most cancers. Clin Most cancers Res. 2023;29:1952–68.
Chaudagar Okay, Hieromnimon HM, Kelley A, Labadie B, Shafran J, Rameshbabu S, et al. Suppression of tumor cell lactate-generating signaling pathways eradicates murine PTEN/p53-deficient aggressive-variant prostate most cancers through macrophage phagocytosis. Clin Most cancers Res. 2023;29:4930–40.
Doggrell SA. Scientific efficacy and security of zoledronic acid in prostate and breast most cancers. Skilled Rev Anticancer Ther. 2009;9:1211–8.
Rogers TL, Holen I. Tumour macrophages as potential targets of bisphosphonates. J Transl Med. 2011;9:177.
Comito G, Pons Segura C, Taddei ML, Lanciotti M, Serni S, Morandi A, et al. Zoledronic acid impairs stromal reactivity by inhibiting M2-macrophages polarization and prostate cancer-associated fibroblasts. Oncotarget. 2017;8:118–32.
Rimal R, Desai P, Daware R, Hosseinnejad A, Prakash J, Lammers T, et al. Most cancers-associated fibroblasts: origin, perform, imaging, and therapeutic focusing on. Adv Drug Deliv Rev. 2022;189:114504.
Kalluri R. The biology and performance of fibroblasts in most cancers. Nat Rev Most cancers. 2016;16:582–98.
Lopez JI, Errarte P, Erramuzpe A, Guarch R, Cortes JM, Angulo JC, et al. Fibroblast activation protein predicts prognosis in clear cell renal cell carcinoma. Hum Pathol. 2016;54:100–5.
Zou B, Liu X, Zhang B, Gong Y, Cai C, Li P, et al. The expression of FAP in hepatocellular carcinoma cells is induced by hypoxia and correlates with poor scientific outcomes. J Most cancers. 2018;9:3278–86.
Solano-Iturri JD, Beitia M, Errarte P, Calvete-Candenas J, Etxezarraga MC, Loizate A, et al. Altered expression of fibroblast activation protein-alpha (FAP) in colorectal adenoma-carcinoma sequence and in lymph node and liver metastases. Ageing. 2020;12:10337–58.
Pellinen T, Sandeman Okay, Blom S, Turkki R, Hemmes A, Valimaki Okay, et al. Stromal FAP expression is related to MRI visibility and affected person survival in prostate most cancers. Most cancers Res Commun. 2022;2:172–81.
Ji T, Zhao Y, Ding Y, Wang J, Zhao R, Lang J, et al. Transformable peptide nanocarriers for expeditious drug launch and efficient most cancers remedy through cancer-associated fibroblast activation. Angew Chem Int Ed Engl. 2016;55:1050–5.
Lang J, Zhao X, Qi Y, Zhang Y, Han X, Ding Y, et al. Reshaping prostate tumor microenvironment to suppress metastasis through cancer-associated fibroblast inactivation with peptide-assembly-based nanosystem. ACS Nano. 2019;13:12357–71.
Narra Okay, Mullins SR, Lee HO, Strzemkowski-Brun B, Magalong Okay, Christiansen VJ, et al. Section II trial of single agent Val-boroPro (Talabostat) inhibiting Fibroblast Activation Protein in sufferers with metastatic colorectal most cancers. Most cancers Biol Ther. 2007;6:1691–9.
Tomas D, Ulamec M, Hudolin T, Bulimbasic S, Belicza M, Kruslin B. Myofibroblastic stromal response and expression of tenascin-C and laminin in prostate adenocarcinoma. Prostate Most cancers Prostatic Dis. 2006;9:414–9.
Ni WD, Yang ZT, Cui CA, Cui Y, Fang LY, Xuan YH. Tenascin-C is a possible cancer-associated fibroblasts marker and predicts poor prognosis in prostate most cancers. Biochem Biophys Res Commun. 2017;486:607–12.
Chen B, Wang Z, Solar J, Tune Q, He B, Zhang H, et al. A tenascin C focused nanoliposome with navitoclax for particularly eradicating of cancer-associated fibroblasts. Nanomedicine. 2016;12:131–41.
Gil V, Miranda S, Riisnaes R, Gurel B, D’Ambrosio M, Vasciaveo A, et al. HER3 Is an actionable goal in superior prostate most cancers. Most cancers Res. 2021;81:6207–18.
Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Concentrating on CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic most cancers. Proc Natl Acad Sci USA. 2013;110:20212–7.
Heidegger I, Fotakis G, Offermann A, Goveia J, Daum S, Salcher S, et al. Complete characterization of the prostate tumor microenvironment identifies CXCR4/CXCL12 crosstalk as a novel antiangiogenic therapeutic goal in prostate most cancers. Mol Most cancers. 2022;21:132.
Domanska UM, Timmer-Bosscha H, Nagengast WB, Oude Munnink TH, Kruizinga RC, Ananias HJ, et al. CXCR4 inhibition with AMD3100 sensitizes prostate most cancers to docetaxel chemotherapy. Neoplasia. 2012;14:709–18.
Froeling FE, Feig C, Chelala C, Dobson R, Mein CE, Tuveson DA, et al. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-beta-catenin signaling to sluggish tumor development. Gastroenterology. 2011;141:1486–97.e1481-1414.
Sherman MH, Yu RT, Engle DD, Ding N, Atkins AR, Tiriac H, et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic most cancers remedy. Cell. 2014;159:80–93.
Eigentler A, Deal with F, Schanung S, Degen A, Hackl H, Erb HHH, et al. Glucocorticoid remedy influences prostate most cancers cell development and the tumor microenvironment through altered glucocorticoid receptor signaling in prostate fibroblasts. Oncogene 2024;43:235–47.
Shen T, Li Y, Zhu S, Yu J, Zhang B, Chen X, et al. YAP1 performs a key function of the conversion of regular fibroblasts into cancer-associated fibroblasts that contribute to prostate most cancers development. J Exp Clin Most cancers Res. 2020;39:36.
Shen T, Li Y, Wang D, Su Y, Li G, Shang Z, et al. YAP1-TEAD1 mediates the perineural invasion of prostate most cancers cells induced by cancer-associated fibroblasts. Biochim Biophys Acta Mol Foundation Dis. 2022;1868:166540.
Yoshida GJ. Regulation of heterogeneous cancer-associated fibroblasts: the molecular pathology of activated signaling pathways. J Exp Clin Most cancers Res. 2020;39:112.
Peng D, Fu M, Wang M, Wei Y, Wei X. Concentrating on TGF-beta sign transduction for fibrosis and most cancers remedy. Mol Most cancers. 2022;21:104.
Franco OE, Jiang M, Strand DW, Peacock J, Fernandez S, Jackson RS 2nd, et al. Altered TGF-beta signaling in a subpopulation of human stromal cells promotes prostatic carcinogenesis. Most cancers Res. 2011;71:1272–81.
Wen S, Niu Y, Yeh S, Chang C. BM-MSCs promote prostate most cancers development through the conversion of regular fibroblasts to cancer-associated fibroblasts. Int J Oncol. 2015;47:719–27.
Li Okay, Shi H, Zhang B, Ou X, Ma Q, Chen Y, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in most cancers. Sign Transduct Goal Ther. 2021;6:362.
Barry ST, Gabrilovich DI, Sansom OJ, Campbell AD, Morton JP. Therapeutic focusing on of tumour myeloid cells. Nat Rev Most cancers. 2023;23:216–37.
Rivera LB, Bergers G. Intertwined regulation of angiogenesis and immunity by myeloid cells. Developments Immunol. 2015;36:240–9.
Pal SK, Vuong W, Zhang W, Deng J, Liu X, Carmichael C, et al. Scientific and translational evaluation of VEGFR1 as a mediator of the premetastatic area of interest in high-risk localized prostate most cancers. Mol Most cancers Ther. 2015;14:2896–2900.
Yakes FM, Chen J, Tan J, Yamaguchi Okay, Shi Y, Yu P, et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, concurrently suppresses metastasis, angiogenesis, and tumor development. Mol Most cancers Ther. 2011;10:2298–308.
Kwilas AR, Ardiani A, Donahue RN, Aftab DT, Hodge JW. Twin results of a focused small-molecule inhibitor (cabozantinib) on immune-mediated killing of tumor cells and immune tumor microenvironment permissiveness when mixed with a most cancers vaccine. J Transl Med. 2014;12:294.
Dai J, Zhang H, Karatsinides A, Keller JM, Kozloff KM, Aftab DT, et al. Cabozantinib inhibits prostate most cancers development and prevents tumor-induced bone lesions. Clin Most cancers Res. 2014;20:617–30.
Smith M, De Bono J, Sternberg C, Le Moulec S, Oudard S, De Giorgi U, et al. Section III research of cabozantinib in beforehand handled metastatic castration-resistant prostate most cancers: COMET-1. J Clin Oncol. 2016;34:3005–13.
Basch EM, Scholz M, de Bono JS, Vogelzang N, de Souza P, Marx G, et al. Cabozantinib versus mitoxantrone-prednisone in symptomatic metastatic castration-resistant prostate most cancers: a randomized part 3 trial with a main ache endpoint. Eur Urol. 2019;75:929–37.
Madan RA, Karzai FH, Al Harthy M, Petrylak DP, Kim JW, Arlen PM, et al. Cabozantinib plus docetaxel and prednisone in metastatic castration-resistant prostate most cancers. BJU Int. 2021;127:435–44.
Lu X, Horner JW, Paul E, Shang X, Troncoso P, Deng P, et al. Efficient combinatorial immunotherapy for castration-resistant prostate most cancers. Nature. 2017;543:728–32.
Agarwal N, McGregor B, Maughan BL, Dorff TB, Kelly W, Fang B, et al. Cabozantinib together with atezolizumab in sufferers with metastatic castration-resistant prostate most cancers: outcomes from an growth cohort of a multicentre, open-label, part 1b trial (COSMIC-021). Lancet Oncol. 2022;23:899–909.
Agarwal N, Azad A, Carles J, Chowdhury S, McGregor B, Merseburger AS, et al. A part III, randomized, open-label research (CONTACT-02) of cabozantinib plus atezolizumab versus second novel hormone remedy in sufferers with metastatic castration-resistant prostate most cancers. Future Oncol. 2022;18:1185–98.
Agarwal N, Azad A, Carles J, Matsubara N, Oudard S, Saad F, et al. CONTACT-2: Section 3 research of cabozantinib (C) plus atezolizumab (A) vs second novel hormonal remedy (NHT) in sufferers (pts) with metastatic castration-resistant prostate most cancers (mCRPC). J Clin Oncol. 2024;42:18–18.
Bullock Okay, Richmond A. Suppressing MDSC recruitment to the tumor microenvironment by antagonizing CXCR2 to reinforce the efficacy of immunotherapy. Cancers. 2021;13:6293.
Wang G, Lu X, Dey P, Deng P, Wu CC, Jiang S, et al. Concentrating on YAP-dependent MDSC infiltration impairs tumor development. Most cancers Discov. 2016;6:80–95.
Lopez-Bujanda ZA, Haffner MC, Chaimowitz MG, Chowdhury N, Venturini NJ, Patel RA, et al. Castration-mediated IL-8 promotes myeloid infiltration and prostate most cancers development. Nat Most cancers. 2021;2:803–18.
Guo C, Sharp A, Vogl U, Colombo I, Stathis A, Jain S, et al. 454O A part (Ph) I/II trial of the CXCR2 antagonist AZD5069 together with enzalutamide (ENZA) in sufferers (pts) with metastatic castration resistant prostate most cancers (mCRPC). Ann Oncol. 2022;33:S745.
Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and performance. Developments Immunol. 2011;32:19–25.
Trovato R, Fiore A, Sartori S, Cane S, Giugno R, Cascione L, et al. Immunosuppression by monocytic myeloid-derived suppressor cells in sufferers with pancreatic ductal carcinoma is orchestrated by STAT3. J Immunother Most cancers. 2019;7:255.
El-Tanani M, Al Khatib AO, Aladwan SM, Abuelhana A, McCarron PA, Tambuwala MM. Significance of STAT3 signalling in most cancers, metastasis and therapeutic interventions. Cell Sign. 2022;92:110275.
Ma JH, Qin L, Li X. Position of STAT3 signaling pathway in breast most cancers. Cell Commun Sign. 2020;18:33.
Liu Y, Liao S, Bennett S, Tang H, Tune D, Wooden D, et al. STAT3 and its focusing on inhibitors in osteosarcoma. Cell Prolif. 2021;54:e12974.
Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Motahhary M, Saghari S, et al. STAT3 signaling in prostate most cancers development and remedy resistance: an oncogenic pathway with numerous features. Biomed Pharmacother. 2023;158:114168.
Zhu W, Sheng D, Shao Y, Zhang Q, Peng Y. STAT3-regulated LncRNA LINC00160 mediates cell proliferation and cell metabolism of prostate most cancers cells by repressing RCAN1 expression. Mol Cell Biochem. 2022;477:865–75.
Kim C, Lee IH, Hyun HB, Kim JC, Gyawali R, Lee SG, et al. Supercritical fluid extraction of citrus iyo hort. ex tanaka pericarp inhibits development and induces apoptosis by way of abrogation of STAT3 regulated gene merchandise in human prostate most cancers xenograft mouse mannequin. Integr Most cancers Ther. 2017;16:227–43.
Hossain DM, Pal SK, Moreira D, Duttagupta P, Zhang Q, Gained H, et al. TLR9-targeted STAT3 silencing abrogates immunosuppressive exercise of myeloid-derived suppressor cells from prostate most cancers sufferers. Clin Most cancers Res. 2015;21:3771–82.
Moreira D, Adamus T, Zhao X, Su YL, Zhang Z, White SV, et al. STAT3 inhibition mixed with CpG immunostimulation prompts antitumor immunity to eradicate genetically distinct castration-resistant prostate cancers. Clin Most cancers Res. 2018;24:5948–62.
Hellsten R, Lilljebjorn L, Johansson M, Leandersson Okay, Bjartell A. The STAT3 inhibitor galiellalactone inhibits the era of MDSC-like monocytes by prostate most cancers cells and reduces immunosuppressive and tumorigenic elements. Prostate. 2019;79:1611–21.
Orillion A, Hashimoto A, Damayanti N, Shen L, Adelaiye-Ogala R, Arisa S, et al. Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor impact of PD-1 inhibition in murine fashions of lung and renal cell carcinoma. Clin Most cancers Res. 2017;23:5187–201.
Christmas BJ, Rafie CI, Hopkins AC, Scott BA, Ma HS, Cruz KA, et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Most cancers Immunol Res. 2018;6:1561–77.
Bradley D, Rathkopf D, Dunn R, Stadler WM, Liu G, Smith DC, et al. Vorinostat in superior prostate most cancers sufferers progressing on prior chemotherapy (Nationwide Most cancers Institute Trial 6862): trial outcomes and interleukin-6 evaluation: a research by the Division of Protection Prostate Most cancers Scientific Trial Consortium and College of Chicago Section 2 Consortium. Most cancers. 2009;115:5541–9.
Chen Z, Yang X, Chen Z, Li M, Wang W, Yang R, et al. A brand new histone deacetylase inhibitor remodels the tumor microenvironment by deletion of polymorphonuclear myeloid-derived suppressor cells and sensitizes prostate most cancers to immunotherapy. BMC Med. 2023;21:402.
Rodriguez CP, Wu QV, Voutsinas J, Fromm JR, Jiang X, Pillarisetty VG, et al. A part II trial of pembrolizumab and vorinostat in recurrent metastatic head and neck squamous cell carcinomas and salivary gland most cancers. Clin Most cancers Res. 2020;26:837–45.
Ny L, Jespersen H, Karlsson J, Alsen S, Filges S, All-Eriksson C, et al. The PEMDAC part 2 research of pembrolizumab and entinostat in sufferers with metastatic uveal melanoma. Nat Commun. 2021;12:5155.
Hellmann MD, Janne PA, Opyrchal M, Hafez N, Raez LE, Gabrilovich DI, et al. Entinostat plus pembrolizumab in sufferers with metastatic NSCLC beforehand handled with Anti-PD-(L)1 remedy. Clin Most cancers Res. 2021;27:1019–28.
Liu Z, Zhang JY, Yang YJ, Chang Okay, Wang QF, Kong YY, et al. Excessive IL-23+ cells infiltration correlates with worse scientific outcomes and abiraterone effectiveness in sufferers with prostate most cancers. Asian J Androl. 2022;24:147–53.
Teng MW, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, Cooper AM, et al. IL-12 and IL-23 cytokines: from discovery to focused therapies for immune-mediated inflammatory illnesses. Nat Med. 2015;21:719–29.
Feng T, Wei Y, Lee RJ, Zhao L. Liposomal curcumin and its software in most cancers. Int J Nanomed. 2017;12:6027–44.
Tu SP, Jin H, Shi JD, Zhu LM, Suo Y, Lu G, et al. Curcumin induces the differentiation of myeloid-derived suppressor cells and inhibits their interplay with most cancers cells and associated tumor development. Most cancers Prev Res. 2012;5:205–15.
Wang T, Wang J, Jiang H, Ni M, Zou Y, Chen Y, et al. Focused regulation of tumor microenvironment by way of the inhibition of MDSCs by curcumin loaded self-assembled nano-filaments. Mater At the moment Bio. 2022;15:100304.
Goodridge HS, Wolf AJ, Underhill DM. Beta-glucan recognition by the innate immune system. Immunol Rev. 2009;230:38–50.
Albeituni SH, Ding C, Liu M, Hu X, Luo F, Kloecker G, et al. Yeast-derived particulate beta-glucan remedy subverts the suppression of Myeloid-Derived Suppressor Cells (MDSC) by inducing polymorphonuclear MDSC apoptosis and monocytic MDSC differentiation to APC in most cancers. J Immunol. 2016;196:2167–80.
Yu X, Liu R, Gao W, Wang X, Zhang Y. Single-cell omics traces the heterogeneity of prostate most cancers cells and the tumor microenvironment. Cell Mol Biol Lett. 2023;28:38.
Levesque C, Nelson PS. Mobile constituents of the prostate stroma: key contributors to prostate most cancers development and remedy resistance. Chilly Spring Harb Perspect Med. 2018;8:a030510.
Boibessot C, Joncas FH, Park A, Berrehail Z, Pelletier JF, Gris T, et al. Utilizing ex vivo tradition to evaluate dynamic phenotype adjustments in human prostate macrophages following publicity to therapeutic medication. Sci Rep. 2021;11:19299.
Cassetta L, Pollard JW. Concentrating on macrophages: therapeutic approaches in most cancers. Nat Rev Drug Discov. 2018;17:887–904.
Caligiuri G, Tuveson DA. Activated fibroblasts in most cancers: views and challenges. Most cancers Cell. 2023;41:434–49.