Chimeric antigen receptor-based cell therapy for treating urological tumors

Huidi Tang , Linpei Guo , Wen Zhang , Dongqi Tang

Current Urology ›› 2025, Vol. 19 ›› Issue (6) : 363 -375.

PDF (562KB)
Current Urology ›› 2025, Vol. 19 ›› Issue (6) : 363 -375. DOI: 10.1097/CU9.0000000000000305
Reviews
research-article

Chimeric antigen receptor-based cell therapy for treating urological tumors

Author information +
History +
PDF (562KB)

Abstract

Urological tumors represent a significant global health challenge, with conventional therapies often proving insufficient to control disease progression. Recent breakthroughs in cellular immunotherapy, particularly in chimeric antigen receptor (CAR)-T cell, CAR-natural killer cell, and CAR-macrophage therapies, have demonstrated remarkable potential for treating these malignancies. Ongoing research is actively refining CAR-based strategies to enhance their precision in targeting tumor-associated antigens. This review comprehensively summarizes the applications of CAR cell therapy in the following 3 major urological tumors: renal cell carcinoma, bladder cancer, and prostate cancer. Furthermore, we analyzed the current advantages and limitations of these approaches and propose potential strategies for optimization focused on CAR-T cells. This review will provide future directions in this field and contribute to the development of more effective treatments for patients with urological cancer.

Keywords

Chimeric antigen receptor / Urological tumors / CAR cell therapy / Clinical application

Cite this article

Download citation ▾
Huidi Tang, Linpei Guo, Wen Zhang, Dongqi Tang. Chimeric antigen receptor-based cell therapy for treating urological tumors. Current Urology, 2025, 19(6): 363-375 DOI:10.1097/CU9.0000000000000305

登录浏览全文

4963

注册一个新账户 忘记密码

Acknowledgments

None.

Statement of ethics

Not applicable.

Conflict of interest statement

DT is an editor-in-chief of Current Urology and confirms no involvement in any stage of this article’s review process, ensuring unbiased editorial decision making. The other authors declare no conflicts of interest.

Funding source

This work was supported by the Special Fund for the Taishan Scholars Project (tsqn202211324) and the Key Research and Development Program of Shandong Province (2021CXGC011101).

Author contributions

HT: Data acquisition, study design, and manuscript preparation;

LG: Study design, manuscript review, and editing;

WZ: Study concept and design, manuscript review, and editing;

DT: Project supervision, study concept and design, manuscript review, and editing.

Data availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1]

Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024. CA Cancer J Clin 2024; 74(1):12-49.

[2]

Pisignano G, Michael DC, Visal TH, Pirlog R, Ladomery M, Calin GA. Going circular: History, present, and future of circRNAs in cancer. Oncogene 2023; 42(38):2783-2800.

[3]

Malmström PU, Sylvester RJ, Crawford DE, et al. An individual patient data meta-analysis of the long-term outcome of randomised studies comparing intravesical mitomycin C versus bacillus Calmette-Guérin for non-muscle-invasive bladder cancer. Eur Urol 2009; 56(2):247-256.

[4]

Carosella ED, Ploussard G, LeMaoult J, Desgrandchamps F. A systematic review of immunotherapy in urologic cancer: Evolving roles for targeting of CTLA-4, PD-1/PD-L1, and HLA-G. Eur Urol 2015; 68(2):267-279.

[5]

Huang X, Li X, Ma L, Liu C. Management and nursing strategies for different patterns of adverse events in patients with urological cancer treated with immune checkpoint inhibitors. Curr Urol 2024; 18(3):212-217.

[6]

Wu ZS, Wu S. The era of personalized treatments: Updates on immunotherapy within urothelial of bladder cancer. Curr Urol 2022; 16(3):117-120.

[7]

June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 2018; 359(6382):1361-1365.

[8]

Lee JK, Bangayan NJ, Chai T, et al. Systemic surfaceome profiling identifies target antigens for immune-based therapy in subtypes of advanced prostate cancer. Proc Natl Acad Sci U S A 2018; 115(19):E4473-E4482.

[9]

Hubert RS, Vivanco I, Chen E, et al. STEAP: A prostate-specific cell-surface antigen highly expressed in human prostate tumors. Proc Natl Acad Sci U S A 1999; 96(25):14523-14528.

[10]

Bhatia V, Kamat NV, Pariva TE, et al. Targeting advanced prostate cancer with STEAP1 chimeric antigen receptor T cell and tumor-localized IL-12 immunotherapy. Nat Commun 2023; 14(1):2041.

[11]

Zhang G, Wang Y, Lu S, et al. Molecular understanding and clinical outcomes of CAR T cell therapy in the treatment of urological tumors. Cell Death Dis 2024; 15(5):359.

[12]

Li J, Li W, Huang K, Zhang Y, Kupfer G, Zhao Q. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: Lessons learned and strategies for moving forward. J Hematol Oncol 2018; 11(1):22.

[13]

Kong JC, Sa'ad MA, Vijayan HM, et al. Chimeric antigen receptor-natural killer cell therapy: Current advancements and strategies to overcome challenges. Front Immunol 2024;15:1384039.

[14]

Zhang Y, Wallace DL, de Lara CM, et al. In vivo kinetics of human natural killer cells: The effects of ageing and acute and chronic viral infection. Immunology 2007; 121(2):258-265.

[15]

Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J. CAR-NK cells: A promising cellular immunotherapy for cancer. EBioMedicine 2020;59:102975.

[16]

Zhou X, Liu X, Huang L. Macrophage-mediated tumor cell phagocytosis: Opportunity for nanomedicine intervention. Adv Funct Mater 2021; 31(5).

[17]

He C, Mansilla-Soto J, Khanra N, et al. CD19 CAR antigen engagement mechanisms and affinity tuning. Sci Immunol 2023; 8(81):eadf1426.

[18]

Chan W, Cao YM, Zhao X, et al. TCR ligand potency differentially impacts PD-1 inhibitory effects on diverse signaling pathways. J Exp Med 2023; 220(12):e20231242.

[19]

Kagoya Y, Tanaka S, Guo T, et al. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat Med 2018; 24(3):352-359.

[20]

Wang Y, Suarez ER, Kastrunes G, et al. Evolution of cell therapy for renal cell carcinoma. Mol Cancer 2024; 23(1):8.

[21]

Panowski SH, Srinivasan S, Tan N, et al. Preclinical development and evaluation of allogeneic CAR T cells targeting CD70 for the treatment of renal cell carcinoma. Cancer Res 2022; 82(14):2610-2624.

[22]

Suarez ER, Chang de K, Sun J, et al. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget 2016; 7(23):34341-34355.

[23]

Mori JI, Adachi K, Sakoda Y, et al. Anti-tumor efficacy of human anti-c-met CAR-T cells against papillary renal cell carcinoma in an orthotopic model. Cancer Sci 2021; 112(4):1417-1428.

[24]

Tanaka M, Siemann DW. Gas6/Axl signaling pathway in the tumor immune microenvironment. Cancers (Basel) 2020; 12(7).

[25]

Hwang A, Mehra V, Chhetri J, Ali S, Tran M, Roddie C. Current treatment options for renal cell carcinoma: Focus on cell-based immunotherapy. Cancers (Basel) 2024; 16(6):1209.

[26]

Guan Y, Chambers CB, Tabatabai T, et al. Renal cell tumors convert natural killer cells to a proangiogenic phenotype. Oncotarget 2020; 11(26):2571-2585.

[27]

Schönfeld K, Sahm C, Zhang C, et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol Ther 2015; 23(2):330-338.

[28]

Kremer V, Ligtenberg MA, Zendehdel R, et al. Genetic engineering of human NK cells to express CXCR2 improves migration to renal cell carcinoma. J Immunother Cancer 2017; 5(1):73.

[29]

Fiore PF, Di Matteo S, Tumino N, et al. Interleukin-15 and cancer: Some solved and many unsolved questions. J Immunother Cancer 2020; 8(2):e001428.

[30]

Marofi F, Motavalli R, Safonov VA, et al. CAR T cells in solid tumors: Challenges and opportunities. Stem Cell Res Ther 2021; 12(1):81.

[31]

Boroughs AC, Larson RC, Choi BD, et al. Chimeric antigen receptor costimulation domains modulate human regulatory T cell function. JCI Insight 2019; 5(8):e126194.

[32]

Good Z, Spiegel JY, Sahaf B, et al. Post-infusion CAR T(Reg) cells identify patients resistant to CD19-CAR therapy. Nat Med 2022; 28(9):1860-1871.

[33]

Wang D, Ning H, Wu H, et al. Construction and evaluation of a novel prognostic risk model of aging-related genes in bladder cancer. Curr Urol 2023; 17(4):236-245.

[34]

Tripathi A, Khaki AR, Grivas P. Perioperative immunotherapy in muscle-invasive bladder bancer. Eur Urol Oncol 2021; 4(2):131-133.

[35]

Parriott G, Deal K, Crean S, Richardson E, Nylen E, Barber A. T-cells expressing a chimeric-PD1-Dap10-CD3zeta receptor reduce tumour burden in multiple murine syngeneic models of solid cancer. Immunology 2020; 160(3):280-294.

[36]

Yu L, Li Z, Mei H, et al. Patient-derived organoids of bladder cancer recapitulate antigen expression profiles and serve as a personal evaluation model for CAR-T cells in vitro. Clin Transl Immunology 2021; 10(2):e1248.

[37]

Grunewald CM, Haist C, König C, et al. Epigenetic priming of bladder cancer cells with decitabine increases cytotoxicity of human EGFR and CD44v6 CAR engineered T-cells. Front Immunol 2021;12:782448.

[38]

Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, Kurzrock R. HER2 expression status in diverse cancers: Review of results from 37,992 patients. Cancer Metastasis Rev 2015; 34(1):157-164.

[39]

Ma XY, Cai HZ, Xu WZ, Xu ZC, Yu B. Advances of HER2-targeted therapy for bladder cancer. Chinese Journal of Surgical Oncology 2024; 16(06):614-619.

[40]

Ding M, Lin J, Qin C, et al. Novel CAR-T cells specifically targeting SIA-CIgG demonstrate effective antitumor efficacy in bladder cancer. Adv Sci (Weinh) 2024; 11(40):e2400156.

[41]

Khazamipour N, Oo HZ, Al-Nakouzi N, et al. Transient CAR T cells with specificity to oncofetal glycosaminoglycans in solid tumors. EMBO Mol Med 2024; 16(11):2775-2794.

[42]

Shen JJ, Dai DP, Zhao WX, et al. A novel co-receptor with mutated TIGIT to enhance PSCA CAR-T therapy for bladder cancer. J Clin Oncol 2024; 42(16):e14574.

[43]

Jiang Y, Sun X, Song X, et al. Patient-derived bladder cancer organoid model to predict sensitivity and feasibility of tailored precision therapy. Curr Urol 2023; 17(4):221-228.

[44]

Bahramloo M, Shahabi SA, Kalarestaghi H, et al. CAR-NK cell therapy in AML: Current treatment, challenges, and advantage. Biomed Pharmacother 2024;177:117024.

[45]

Chen Y, Yu Z, Tan X, et al. CAR-macrophage: A new immunotherapy candidate against solid tumors. Biomed Pharmacother 2021;139:111605.

[46]

Gillessen S, Attard G, Beer TM, et al. Management of patients with advanced prostate cancer:Report of the advanced prostate cancer consensus conference 2019. Eur Urol 2020; 77(4):508-547.

[47]

Zhang M, Wang H, Wang M, et al. EphA2 specific chimeric antigen receptor engineered T cells for the treatment of prostate cancer. Transl Oncol 2024;50:102111.

[48]

Kloss CC, Lee J, Zhang A, et al. Dominant-negative TGF-β receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol Ther 2018; 26(7):1855-1866.

[49]

Narayan V, Barber-Rotenberg JS, Jung IY, et al. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: A phase 1 trial. Nat Med 2022; 28(4):724-734.

[50]

Priceman SJ, Gerdts EA, Tilakawardane D, et al. Co-stimulatory signaling determines tumor antigen sensitivity and persistence of CAR T cells targeting PSCA+ metastatic prostate cancer. Onco Targets Ther 2018; 7(2):e1380764.

[51]

Dorff TB, Blanchard S, Martirosyan H, et al. Final results from phase I study of PSCA-targeted chimeric antigen receptor (CAR) T cells in patients with metastatic castration resistant prostate cancer (mCRPC). J Clin Oncol 2023; 41(16):1.

[52]

Dorff TB, Blanchard MS, Adkins LN, et al. PSCA-CAR T cell therapy in metastatic castration-resistant prostate cancer: A phase 1 trial. Nat Med 2024; 30(6):1636-1644.

[53]

Stein MN, Dumbrava EE, Teply BA, et al. PSCA-targeted BPX-601 CAR T cells with pharmacological activation by rimiducid in metastatic pancreatic and prostate cancer: A phase 1 dose escalation trial. Nat Commun 2024; 15(1):10743.

[54]

Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ. Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol 2015; 16(1):1.

[55]

He C, Zhou Y, Li Z, et al. Co-expression of IL-7 improves NKG2D-based CAR T cell therapy on prostate cancer by enhancing the expansion and inhibiting the apoptosis and exhaustion. Cancers (Basel) 2020; 12(7):1969.

[56]

Wang F, Wu L, Yin L, Shi H, Gu Y, Xing N. Combined treatment with anti-PSMA CAR NK-92 cell and anti-PD-L1 monoclonal antibody enhances the antitumour efficacy against castration-resistant prostate cancer. Clin Transl Med 2022; 12(6):e901.

[57]

Chen Y, Song Y, Du W, Gong L, Chang H, Zou Z. Tumor-associated macrophages: An accomplice in solid tumor progression. J Biomed Sci 2019; 26(1):78.

[58]

Shi Y, Fan X, Deng H, et al. Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcγ receptors on macrophages. J Immunol 2015; 194(9):4379-4386.

[59]

Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 2010; 142(5):699-713.

[60]

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

[61]

Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J Clin Invest 2015; 125(9):3356-3364.

[62]

Feng PH, Lee KY, Chang YL, et al. CD14(+)S100A9(+) monocytic myeloid-derived suppressor cells and their clinical relevance in non-small cell lung cancer. Am J Respir Crit Care Med 2012; 186(10):1025-1036.

[63]

Sevko A, Sade-Feldman M, Kanterman J, et al. Cyclophosphamide promotes chronic inflammation-dependent immunosuppression and prevents antitumor response in melanoma. J Invest Dermatol 2013; 133(6):1610-1619.

[64]

Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother 2009; 58(1):49-59.

[65]

Nagaraj S, Youn JI, Weber H, et al. Anti-inflammatory triterpenoid blocks immune suppressive function of MDSCs and improves immune response in cancer. Clin Cancer Res 2010; 16(6):1812-1823.

[66]

Serafini P, Meckel K, Kelso M, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med 2006; 203(12):2691-2702.

[67]

Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 2005; 5(4):263-274.

[68]

Mandapathil M, Szczepanski MJ, Szajnik M, et al. Adenosine and prostaglandin E2 cooperate in the suppression of immune responses mediated by adaptive regulatory T cells. J Biol Chem 2010; 285(36):27571-27580.

[69]

Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest 2007; 117(5):1175-1183.

[70]

Ma Y, Kepp O, Ghiringhelli F, et al. Chemotherapy and radiotherapy: Cryptic anticancer vaccines. Semin Immunol 2010; 22(3):113-124.

[71]

Strauss L, Bergmann C, Gooding W, Johnson JT, Whiteside TL. The frequency and suppressor function of CD4 + CD25highFoxp3+ T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clin Cancer Res 2007; 13(21):6301-6311.

[72]

Bughda R, Dimou P, D'Souza RR, Klampatsa A. Fibroblast activation protein (FAP)-targeted CAR-T cells: Launching an attack on tumor stroma. Immunotargets Ther 2021;10:313-323.

[73]

Liu T, Han C, Wang S, et al. Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy. J Hematol Oncol 2019; 12(1):86.

[74]

Busek P, Mateu R, Zubal M, Kotackova L, Sedo A. Targeting fibroblast activation protein in cancer - Prospects and caveats. Front Biosci (Landmark Ed) 2018; 23(10):1933-1968.

[75]

Garin-Chesa P, Old LJ, Rettig WJ. Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers. Proc Natl Acad Sci U S A 1990; 87(18):7235-7239.

[76]

Huang Y, Wang S, Kelly T. Seprase promotes rapid tumor growth and increased microvessel density in a mouse model of human breast cancer. Cancer Res 2004; 64(8):2712-2716.

[77]

Chen H, Yang WW, Wen QT, Xu L, Chen M. TGF-beta induces fibroblast activation protein expression; fibroblast activation protein expression increases the proliferation, adhesion, and migration of HO-8910 PM [corrected]. Exp Mol Pathol 2009; 87(3):189-194.

[78]

Kakarla S, Chow KK, Mata M, et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol Ther 2013; 21(8):1611-1620.

[79]

Cai Z, Ang X, Xu Z, et al. A pan-cancer study of PD-1 and CTLA-4 as therapeutic targets. Transl Cancer Res 2021; 10(9):3993-4001.

[80]

Hou AJ, Chen LC, Chen YY. Navigating CAR-T cells through the solid-tumour microenvironment. Nat Rev Drug Discov 2021; 20(7):531-550.

[81]

Dahmani A, Delisle JS. TGF-β in T cell biology: Implications for cancer immunotherapy. Cancers (Basel) 2018; 10(6):194.

[82]

Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limón P. The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol 2010; 10(8):554-567.

[83]

Nizard M, Roussel H, Diniz MO, et al. Induction of resident memory T cells enhances the efficacy of cancer vaccine. Nat Commun 2017;8:15221.

[84]

Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol 2001;19:683-765.

[85]

Salkeni MA, Naing A. Interleukin-10 in cancer immunotherapy: From bench to bedside. Trends Cancer 2023; 9(9):716-725.

[86]

Gabrilovich DI, Chen HL, Girgis KR, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 1996; 2(10):1096-1103.

[87]

Ohm JE, Gabrilovich DI, Sempowski GD, et al. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood 2003; 101(12):4878-4886.

[88]

Patel SA, Nilsson MB, Le X, Cascone T, Jain RK, Heymach JV. Molecular mechanisms and future implications of VEGF/VEGFR in cancer therapy. Clin Cancer Res 2023; 29(1):30-39.

[89]

Jain RK. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science 2005; 307(5706):58-62.

[90]

Mishra A, Verma M. Epigenetic and genetic regulation of PDCD1 gene in cancer immunology. Methods Mol Biol 2018;1856:247-254.

[91]

Yang Z, Su W, Wei X, et al. HIF-1α drives resistance to ferroptosis in solid tumors by promoting lactate production and activating SLC1A1. Cell Rep 2023; 42(8):112945.

[92]

Huber V, Camisaschi C, Berzi A, et al. Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 2017;43:74-89.

[93]

De Leo A, Ugolini A, Yu X, et al. Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma. Immunity 2024; 57(5):1105-1123.e8.

[94]

Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol Res 2016; 4(6):498-508.

[95]

Frey NV, Gill S, Hwang WT, et al. CART22-65s co-administered with huCART19 in adult patients with relapsed or refractory ALL. Blood 2021;138.

[96]

Qin H, Ramakrishna S, Nguyen S, et al. Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol Ther Oncolytics 2018;11:127-137.

[97]

Bos TJ, De Bruyne E, Van Lint S, Heirman C, Vanderkerken K. Large double copy vectors are functional but show a size-dependent decline in transduction efficiency. J Biotechnol 2010; 150(1):37-40.

[98]

Bielamowicz K, Fousek K, Byrd TT, et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro Oncol 2018; 20(4):506-518.

[99]

Hu B, Ren J, Luo Y, et al. Augmentation of antitumor immunity by human and mouse CAR T cells secreting IL-18. Cell Rep 2017; 20(13):3025-3033.

[100]

Laurent SA, Hoffmann FS, Kuhn PH, et al. γ-Secretase directly sheds the survival receptor BCMA from plasma cells. Nat Commun 2015;6:7333.

[101]

Chapuis AG, Ragnarsson GB, Nguyen HN, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med 2013; 5(174):174ra27.

[102]

Nielsen M, Lundegaard C, Blicher T, et al. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PloS One 2007; 2(8):e796.

[103]

Krishnan S, Suarez-Martinez AD, Bagher P, et al. Microvascular dysfunction and kidney disease: Challenges and opportunities? Microcirculation 2021; 28(3):e12661.

[104]

Singleton DC, Macann A, Wilson WR. Therapeutic targeting of the hypoxic tumour microenvironment. Nat Rev Clin Oncol 2021; 18(12):751-772.

[105]

Pallone TL, Edwards A, Mattson DL. Renal medullary circulation. Compr Physiol 2012; 2(1):97-140.

[106]

Afsar B, Afsar RE, Dagel T, et al. Capillary rarefaction from the kidney point of view. Clin Kidney J 2018; 11(3):295-301.

[107]

Bhargava P, Schnellmann RG. Mitochondrial energetics in the kidney. Nat Rev Nephrol 2017; 13(10):629-646.

[108]

Tanabe K, Wada J, Sato Y. Targeting angiogenesis and lymphangiogenesis in kidney disease. Nat Rev Nephrol 2020; 16(5):289-303.

[109]

Logue OC, McGowan JW, George EM, Bidwell GL 3rd. Therapeutic angiogenesis by vascular endothelial growth factor supplementation for treatment of renal disease. Curr Opin Nephrol Hypertens 2016; 25(5):404-409.

[110]

Beyer AM, Durand MJ, Hockenberry J, Gamblin TC, Phillips SA, Gutterman DD. An acute rise in intraluminal pressure shifts the mediator of flow-mediated dilation from nitric oxide to hydrogen peroxide in human arterioles. Am J Physiol Heart Circ Physiol 2014; 307(11):H1587-H1593.

[111]

Schrimpf C, Duffield JS. Mechanisms of fibrosis: The role of the pericyte. Curr Opin Nephrol Hypertens 2011; 20(3):297-305.

[112]

Iliescu R, Fernandez SR, Kelsen S, Maric C, Chade AR. Role of renal microcirculation in experimental renovascular disease. Nephrol Dial Transplant 2010; 25(4):1079-1087.

[113]

Parsons CL. The role of the urinary epithelium in the pathogenesis of interstitial cystitis/prostatitis/urethritis. Urology 2007; 69(4 Suppl):9-16.

[114]

Hurst RE, Zebrowski R. Identification of proteoglycans present at high density on bovine and human bladder luminal surface. J Urol 1994; 152(5 Pt1):1641-1645.

[115]

Parsons CL. Prostatitis, interstitial cystitis, chronic pelvic pain, and urethral syndrome share a common pathophysiology: Lower urinary dysfunctional epithelium and potassium recycling. Urology 2003; 62(6):976-982.

[116]

Parsons CL, Bautista SL, Stein PC, Zupkas P. Cyto-injury factors in urine: A possible mechanism for the development of interstitial cystitis. J Urol 2000; 164(4):1381-1384.

[117]

Parsons CL, Greenspan C, Mulholland SG. The primary antibacterial defense mechanism of the bladder. Invest Urol 1975; 13(1):72-78.

[118]

Parsons CL, Pollen JJ, Anwar H, Stauffer C, Schmidt JD. Antibacterial activity of bladder surface mucin duplicated in the rabbit bladder by exogenous glycosaminoglycan (sodium pentosanpolysulfate). Infect Immun 1980; 27(3):876-881.

[119]

das Neves J, Sverdlov Arzi R, Sosnik A. Molecular and cellular cues governing nanomaterial-mucosae interactions: From nanomedicine to nanotoxicology. Chem Soc Rev 2020; 49(14):5058-5100.

[120]

Otani T, Furuse M. Tight junction structure and function revisited. Trends Cell Biol 2020; 30(10):805-817.

[121]

Stephan SB, Taber AM, Jileaeva I, Pegues EP, Sentman CL, Stephan MT. Biopolymer implants enhance the efficacy of adoptive T-cell therapy. Nat Biotechnol 2015; 33(1):97-101.

[122]

Liu Y, Liu Z, Li T, Ye G. Ultrasonic sonoporation can enhance the prostate permeability. Med Hypotheses 2010; 74(3):449-451.

[123]

Shenoi MM, Iltis I, Choi J, et al. Nanoparticle delivered vascular disrupting agents (VDAs): Use of TNF-alpha conjugated gold nanoparticles for multimodal cancer therapy. Mol Pharm 2013; 10(5):1683-1694.

[124]

Gao W, Wang Z, Lv L, et al. Photodynamic therapy induced enhancement of tumor vasculature permeability using an upconversion nanoconstruct for improved intratumoral nanoparticle delivery in deep tissues. Theranostics 2016; 6(8):1131-1144.

[125]

Bellone M, Mondino A, Corti A. Vascular targeting, chemotherapy and active immunotherapy: Teaming up to attack cancer. Trends Immunol 2008; 29(5):235-241.

[126]

Sakai N, Chiba H, Fujita H, et al. Expression patterns of claudin family of tight-junction proteins in the mouse prostate. Histochem Cell Biol 2007; 127(4):457-462.

AI Summary AI Mindmap
PDF (562KB)

294

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/