CLC number:
On-line Access: 2024-08-27
Received: 2023-10-17
Revision Accepted: 2024-05-08
Crosschecked: 2022-10-13
Cited: 0
Clicked: 1508
Citations: Bibtex RefMan EndNote GB/T7714
Jing CHEN, Jian CHEN, Lie WANG. Tertiary lymphoid structures as unique constructions associated with the organization, education, and function of tumor-infiltrating immunocytes[J]. Journal of Zhejiang University Science B,in press.Frontiers of Information Technology & Electronic Engineering,in press.https://doi.org/10.1631/jzus.B2200174 @article{title="Tertiary lymphoid structures as unique constructions associated with the organization, education, and function of tumor-infiltrating immunocytes", %0 Journal Article TY - JOUR
三级淋巴结构--影响肿瘤浸润免疫细胞编排、调控及功能的独特结构1浙江大学医学院附属第二医院胃肠外科,中国杭州市,310009 2浙江大学医学院免疫研究所及附属第一医院骨髓移植中心,中国杭州市,310003 3浙江大学医学中心良渚实验室,中国杭州市,311121 4浙江大学癌症中心,中国杭州市,310058 概要:三级淋巴结构主要在包括肿瘤在内的慢性炎症部位的非淋巴组织中发展形成。这一异位形成的淋巴结构主要包含趋化因子招募而来的B细胞、T细胞以及起支持作用的树突状细胞(DC)。成熟的三级淋巴结构有利于适应性免疫细胞的发育与互作,进而增强其抗肿瘤免疫应答。但该抗肿瘤效应取决于三级淋巴结构的具体定位以及成熟阶段。目前有越来越多的研究表明三级淋巴结构的抗肿瘤机制将有助于新型免疫疗法的探索。现有的治疗策略研究方案包括联合瘤内三级淋巴结构与药物治疗或诱导瘤内三级淋巴结构的形成,从而实现提升肿瘤治疗疗效和改善肿瘤患者预后的目的。 关键词组: Darkslateblue:Affiliate; Royal Blue:Author; Turquoise:Article
Reference[1]AhrendsT, SpanjaardA, PilzeckerB, et al., 2017. CD4+ T cell help confers a cytotoxic T cell effector program including coinhibitory receptor downregulation and increased tissue invasiveness. Immunity, 47(5):848-861.e5. ![]() [2]AllenCDC, OkadaT, TangHL, et al., 2007. Imaging of germinal center selection events during affinity maturation. Science, 315(5811):528-531. ![]() [3]AllenE, JabouilleA, RiveraLB, et al., 2017. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med, 9(385):eaak9679. ![]() [4]AloisiF, Pujol-BorrellR, 2006. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol, 6(3):205-217. ![]() [5]AmariaRN, ReddySM, TawbiHA, et al., 2018. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med, 24(11):1649-1654. ![]() [6]AnselKM, NgoVN, HymanPL, et al., 2000. A chemokine-driven positive feedback loop organizes lymphoid fol ![]() [7]licles. Nature, 406(6793):309-314. ![]() [8]AsrirA, TardiveauC, CoudertJ, et al., 2022. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell, 40(3):318-334.e9. ![]() [9]BaroneF, GardnerDH, NayarS, et al., 2016. Stromal fibroblasts in tertiary lymphoid structures: a novel target in chronic inflammation. Front Immunol, 7:477. ![]() [10]BénézechC, WhiteA, MaderE, et al., 2010. Ontogeny of stromal organizer cells during lymph node development. J Immunol, 184(8):4521-4530. ![]() [11]BénézechC, LuuNT, WalkerJA, et al., 2015. Inflammation-induced formation of fat-associated lymphoid clusters. Nat Immunol, 16(8):819-828. ![]() [12]BennettSRM, CarboneFR, KaramalisF, et al., 1997. Induction of a CD8+ cytotoxic T lymphocyte response by cross-priming requires cognate CD4+ T cell help. J Exp Med, 186(1):65-70. ![]() [13]BerekC, BergerA, ApelM, 1991. Maturation of the immune response in germinal centers. Cell, 67(6):1121-1129. ![]() [14]BindeaG, MlecnikB, TosoliniM, et al., 2013. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity, 39(4):782-795. ![]() [15]BraunA, WorbsT, MoschovakisGL, et al., 2011. Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat Immunol, 12(9):879-887. ![]() [16]BuisseretL, GaraudS, de WindA, et al., 2017. Tumor-infiltrating lymphocyte composition, organization and PD-1/PD-L1 expression are linked in breast cancer. OncoImmunology, 6(1):e1257452. ![]() [17]CabritaR, LaussM, SannaA, et al., 2020. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature, 577(7791):561-565. ![]() [18]CalderaroJ, PetitprezF, BechtE, et al., 2019. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J Hepatol, 70(1):58-65. ![]() [19]CarmiY, SpitzerMH, LindeIL, et al., 2015. Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity. Nature, 521(7550):99-104. ![]() [20]CarregaP, LoiaconoF, di CarloE, et al., 2015. NCR+ILC3 concentrate in human lung cancer and associate with intratumoral lymphoid structures. Nat Commun, 6:8280. ![]() [21]CastellinoF, HuangAY, Altan-BonnetG, et al., 2006. Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4+ T cell-dendritic cell interaction. Nature, 440(7086):890-895. ![]() [22]CastinoGF, CorteseN, CaprettiG, et al., 2016. Spatial distribution of B cells predicts prognosis in human pancreatic adenocarcinoma. OncoImmunology, 5(4):e1085147. ![]() [23]ChoiYS, KageyamaR, EtoD, et al., 2011. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity, 34(6):932-946. ![]() [24]CilloAR, KurtenCHL, TabibT, et al., 2020. Immune landscape of viral- and carcinogen-driven head and neck cancer. Immunity, 52(1):183-199.e9. ![]() [25]ColbeckEJ, JonesE, HindleyJP, et al., 2017. Treg depletion licenses T cell-driven HEV neogenesis and promotes tumor destruction. Cancer Immunol Res, 5(11):1005-1015. ![]() [26]CuiC, WangJW, FagerbergE, et al., 2021. Neoantigen-driven B cell and CD4 T follicular helper cell collaboration promotes anti-tumor CD8 T cell responses. Cell, 184(25):6101-6118.e13. ![]() [27]DaiSY, ZengH, LiuZP, et al., 2021. Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer, 9(2):e001823. ![]() [28]DejardinE, DroinNM, DelhaseM, et al., 2002. The lymphotoxin-β receptor induces different patterns of gene expression via two NF-κB pathways. Immunity, 17(4):525-535. ![]() [29]DenkertC, von MinckwitzG, Darb-EsfahaniS, et al., 2018. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol, 19(1):40-50. ![]() [30]DepilS, DuchateauP, GruppSA, et al., 2020. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov, 19(3):185-199. ![]() [31]de TogniP, GoellnerJ, RuddleNH, et al., 1994. Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science, 264(5159):703-707. ![]() [32]di PucchioT, ChatterjeeB, Smed-SörensenA, et al., 2008. Direct proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility complex class I. Nat Immunol, 9(5):551-557. ![]() [33]DraytonDL, LiaoS, MounzerRH, et al., 2006. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol, 7(4):344-353. ![]() [34]EickhoffS, BrewitzA, GernerMY, et al., 2015. Robust anti-viral immunity requires multiple distinct T cell-dendritic cell interactions. Cell, 162(6):1322-1337. ![]() [35]FurtadoGC, PacerME, BongersG, et al., 2014. TNFα-dependent development of lymphoid tissue in the absence of RORγt+ lymphoid tissue inducer cells. Mucosal Immunol, 7(3):602-614. ![]() [36]GarnerH, deVisser KE, 2020. Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat Rev Immunol, 20(8):483-497. ![]() [37]GeeMH, HanA, LofgrenSM, et al., 2018. Antigen identification for orphan T cell receptors expressed on tumor-infiltrating lymphocytes. Cell, 172(3):549-563.e16. ![]() [38]GermainC, GnjaticS, TamzalitF, et al., 2014. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med, 189(7):832-844. ![]() [39]GeurtsvankesselCH, WillartMAM, BergenIM, et al., 2009. Dendritic cells are crucial for maintenance of tertiary lymphoid structures in the lung of influenza virus-infected mice. J Exp Med, 206(11):2339-2349. ![]() [40]GiraldoNA, Sanchez-SalasR, PeskeJD, et al., 2019. The clinical role of the TME in solid cancer. Br J Cancer, 120(1):45-53. ![]() [41]GocJ, GermainC, Vo-BourgaisTKD, et al., 2014. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res, 74(3):705-715. ![]() [42]GoenkaR, BarnettLG, SilverJS, et al., 2011. Cutting edge: dendritic cell-restricted antigen presentation initiates the follicular helper T cell program but cannot complete ultim ![]() [43]ate effector differentiation. J Immunol, 187(3):1091-1095. ![]() [44]GroeneveldCS, FontugneJ, CabelL, et al., 2021. Tertiary lymphoid structures marker CXCL13 is associated with better survival for patients with advanced-stage bladder cancer treated with immunotherapy. Eur J Cancer, 148:181-189. ![]() [45]Gu-TrantienC, LoiS, GaraudS, et al., 2013. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest, 123(7):2873-2892. ![]() [46]GuedjK, Khallou-LaschetJ, ClementM, et al., 2014. M1 macrophages act as LTβR-independent lymphoid tissue inducer cells during atherosclerosis-related lymphoid neogenesis. Cardiovasc Res, 101(3):434-443. ![]() [47]GunnMD, NgoVN, AnselKM, et al., 1998. A B-cell-homing chemokine made in lymphoid follicles activates Burkitt’s lymphoma receptor-1. Nature, 391(6669):799-803. ![]() [48]HalleS, DujardinHC, BakocevicN, et al., 2009. Induced bronchus-associated lymphoid tissue serves as a general priming site for T cells and is maintained by dendritic cells. J Exp Med, 206(12):2593-2601. ![]() [49]HeJJ, XiongXX, YangH, et al., 2022. Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response. Cell Res, 32(6):530-542. ![]() [50]HelminkBA, ReddySM, GaoJJ, et al., 2020. B cells and tertiary lymphoid structures promote immunotherapy response. Nature, 577(7791):549-555. ![]() [51]HinshawDC, ShevdeLA, 2019. The tumor microenvironment innately modulates cancer progression. Cancer Res, 79(18):4557-4566. ![]() [52]HiraokaN, InoY, Yamazaki-ItohR, et al., 2015. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer, 112(11):1782-1790. ![]() [53]HodiFS, O'DaySJ, McDermottDF, et al., 2010. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med, 363(8):711-723. ![]() [54]HorJL, WhitneyPG, ZaidA, et al., 2015. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity, 43(3):554-565. ![]() [55]JansenCS, ProkhnevskaN, MasterVA, et al., 2019. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature, 576(7787):465-470. ![]() [56]JanssenEM, LemmensEE, WolfeT, et al., 2003. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature, 421(6925):852-856. ![]() [57]JiangXG, 2020. Lymphatic vasculature in tumor metastasis and immunobiology. J Zhejiang Univ-Sci B (Biomed & Biotechnol), 21(1):3-11. ![]() [58]JinKF, CaoYF, GuY, et al., 2021. Poor clinical outcomes and immunoevasive contexture in CXCL13+CD8+ T cells enriched gastric cancer patients. OncoImmunology, 10(1):1915560. ![]() [59]Johansson-PercivalA, HeB, LiZJ, et al., 2017. De novo induction of intratumoral lymphoid structures and vessel normalization enhances immunotherapy in resistant tumors. Nat Immunol, 18(11):1207-1217. ![]() [60]JoshiNS, Akama-GarrenEH, LuYS, et al., 2015. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity, 43(3):579-590. ![]() [61]KatoY, ZaidA, DaveyGM, et al., 2015. Targeting antigen to Clec9A primes follicular Th cell memory responses capable of robust recall. J Immunol, 195(3):1006-1014. ![]() [62]KerfootSM, YaariG, PatelJR, et al., 2011. Germinal center B cell and T follicular helper cell development initiates in the interfollicular zone. Immunity, 34(6):947-960. ![]() [63]KinkerGS, VitielloGAF, FerreiraWAS, et al., 2021. B cell orchestration of anti-tumor immune responses: a matter of cell localization and communication. Front Cell Dev Biol, 9:678127. ![]() [64]KrishnaswamyJK, GowthamanU, ZhangBY, et al., 2017. Migratory CD11b+ conventional dendritic cells induce T follicular helper cell-dependent antibody responses. Sci Immunol, 2(18):eaam9169. ![]() [65]KroegerDR, MilneK, NelsonBH, 2016. Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer. Clin Cancer Res, 22(12):3005-3015. ![]() [66]LeeJM, LeeMH, GaronE, et al., 2017. Phase I trial of intratumoral injection of CCL21 gene-modified dendritic cells in lung cancer elicits tumor-specific immune responses and CD8+ T-cell infiltration. Clin Cancer Res, 23(16):4556-4568. ![]() [67]LeeY, ChinRK, ChristiansenP, et al., 2006. Recruitment and activation of naive T cells in the islets by lymphotoxin β receptor-dependent tertiary lymphoid structure. Immunity, 25(3):499-509. ![]() [68]LeiX, LeiY, LiJK, et al., 2020. Immune cells within the tumor microenvironment: biological functions and roles in cancer immunotherapy. Cancer Lett, 470:126-133. ![]() [69]LiHJ, van der LeunAM, YofeI, et al., 2019. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell, 176(4):775-789.e18. ![]() [70]LiJP, WuCY, ChenMY, et al., 2021. PD-1+CXCR5-CD4+ Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer, 9(7):e002101. ![]() [71]LochnerM, OhnmachtC, PresleyL, et al., 2011. Microbiota-induced tertiary lymphoid tissues aggravate inflammatory disease in the absence of RORγ T and LTi cells. J Exp Med, 208(1):125-134. ![]() [72]LutherSA, LopezT, BaiW, et al., 2000. BLC expression in pancreatic islets causes B cell recruitment and lymphotoxin-dependent lymphoid neogenesis. Immunity, 12(5):471-481. ![]() [73]LutherSA, BidgolA, HargreavesDC, et al., 2002. Differing activities of homeostatic chemokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell recruitment and lymphoid neogenesis. J Immunol, 169(1):424-433. ![]() [74]LutherSA, AnselKM, CysterJG, 2003. Overlapping roles of CXCL13, interleukin 7 receptor α, and CCR7 ligands in lymph node development. J Exp Med, 197(9):1191-1198. ![]() [75]LutzER, WuAA, BigelowE, et al., 2014. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res, 2(7):616-631. ![]() [76]MaldonadoL, TeagueJE, MorrowMP, et al., 2014. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci Transl Med, 6(221):221ra213. ![]() [77]MartinetL, GarridoI, FilleronT, et al., 2011. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res, 71(17):5678-5687. ![]() [78]MatsubaraS, SekiM, SuzukiS, et al., 2019. Tertiary lymphoid organs in the inflammatory myopathy associated with PD-1 inhibitors. J Immunother Cancer, 7(1):256. ![]() [79]MebiusRE, RennertP, WeissmanIL, 1997. Developing lymph nodes collect CD4+CD3-LTβ+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity, 7(4):493-504. ![]() [80]MeylanM, PetitprezF, BechtE, et al., 2022. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity, 55(3):527-541.e5. ![]() [81]MorcretteG, HirschTZ, BadourE, et al., 2019. APC germline hepatoblastomas demonstrate cisplatin-induced intratumor tertiary lymphoid structures. OncoImmunology, 8(6):e1583547. ![]() [82]MounzerRH, SvendsenOS, BalukP, et al., 2010. Lymphotoxin-alpha contributes to lymphangiogenesis. Blood, 116(12):2173-2182. ![]() [83]Munoz-ErazoL, RhodesJL, MarionVC, et al., 2020. Tertiary lymphoid structures in cancer—considerations for patient prognosis. Cell Mol Immunol, 17(6):570-575. ![]() [84]NgoVN, KornerH, GunnMD, et al., 1999. Lymphotoxin α/β and tumor necrosis factor are required for stromal cell expression of homing chemokines in B and T cell areas of the spleen. J Exp Med, 189(2):403-412. ![]() [85]NiogretJ, BergerH, RebeC, et al., 2021. Follicular helper-T cells restore CD8+-dependent antitumor immunity and anti-PD-L1/PD-1 efficacy. J Immunother Cancer, 9(6):e002157. ![]() [86]NoëlG, FontsaML, GaraudS, et al., 2021. Functional Th1-oriented T follicular helper cells that infiltrate human breast cancer promote effective adaptive immunity. J Clin Invest, 131(19): e139905. ![]() [87]Overacre-DelgoffeAE, BumgarnerHJ, CilloAR, et al., 2021. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity, 54(12):2812-2824.e4. ![]() [88]PardollDM, 2012. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer, 12(4):252-264. ![]() [89]PeskeJD, ThompsonED, GemtaL, et al., 2015. Effector lymphocyte-induced lymph node-like vasculature enables naive T-cell entry into tumours and enhanced anti-tumour immunity. Nat Commun, 6:7114. ![]() [90]PetersA, PitcherLA, SullivanJM, et al., 2011. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity, 35(6):986-996. ![]() [91]PetitprezF, de ReynièsA, KeungEZ, et al., 2020. B cells are associated with survival and immunotherapy response in sarcoma. Nature, 577(7791):556-560. ![]() [92]PitzalisC, JonesGW, BombardieriM, et al., 2014. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol, 14(7):447-462. ![]() [93]PoschF, SilinaK, LeiblS, et al., 2018. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. OncoImmunology, 7(2):e1378844. ![]() [94]RidgeJP, di RosaF, MatzingerP, 1998. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature, 393(6684):474-478. ![]() [95]RodriguezAB, PeskeJD, WoodsAN, et al., 2021. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep, 36(3):109422. ![]() [96]RuffinAT, CilloAR, TabibT, et al., 2021. B cell signatures and tertiary lymphoid structures contribute to outcome in head and neck squamous cell carcinoma. Nat Commun, 12:3349. ![]() [97]Sautès-FridmanC, PetitprezF, CalderaroJ, et al., 2019. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer, 19(6):307-325. ![]() [98]SchmittN, MoritaR, BourderyL, et al., 2009. Human dendritic cells induce the differentiation of interleukin-21-producing T follicular helper-like cells through interleukin-12. Immunity, 31(1):158-169. ![]() [99]SchoenbergerSP, ToesREM, van der VoortEIH, et al., 1998. T-cell help for cytotoxic T lymphocytes is mediated by CD40‒CD40L interactions. Nature, 393(6684):480-483. ![]() [100]ShalapourS, LinXJ, BastianIN, et al., 2017. Inflammation-induced IgA+ cells dismantle anti-liver cancer immunity. Nature, 551(7680):340-345. ![]() [101]ShedlockDJ, ShenH, 2003. Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science, 300(5617):337-339. ![]() [102]SiliņaK, SoltermannA, AttarFM, et al., 2018. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res, 78(5):1308-1320. ![]() [103]SongIH, HeoSH, BangWS, et al., 2017. Predictive value of tertiary lymphoid structures assessed by high endothelial venule counts in the neoadjuvant setting of triple-negative breast cancer. Cancer Res Treat, 49(2):399-407. ![]() [104]SuzukiG, SawaH, KobayashiY, et al., 1999. Pertussis toxin-sensitive signal controls the trafficking of thymocytes across the corticomedullary junction in the thymus. J Immunol, 162(10):5981-5985. ![]() [105]ThommenDS, KoelzerVH, HerzigP, et al., 2018. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med, 24(7):994-1004. ![]() [106]TranE, RobbinsPF, LuYC, et al., 2016. T-cell transfer therapy targeting mutant KRAS in cancer. N Engl J Med, 375(23):2255-2262. ![]() [107]van de PavertSA, OlivierBJ, GoverseG, et al., 2009. Chemokine CXCL13 is essential for lymph node initiation and is induced by retinoic acid and neuronal stimulation. Nat Immunol, 10(11):1193-1199. ![]() [108]van DijkN, Gil-JimenezA, SilinaK, et al., 2020. Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: the NABUCCO trial. Nat Med, 26(12):1839-1844. ![]() [109]Veiga-FernandesH, ColesMC, FosterKE, et al., 2007. Tyrosine kinase receptor RET is a key regulator of Peyer’s Patch organogenesis. Nature, 446(7135):547-551. ![]() [110]WeinsteinAM, ChenL, BrzanaEA, et al., 2017. Tbet and IL-36γ cooperate in therapeutic DC-mediated promotion of ectopic lymphoid organogenesis in the tumor microenvironment. OncoImmunology, 6(6):e1322238. ![]() [111]WorkelHH, LubbersJM, ArnoldR, et al., 2019. A transcriptionally distinct CXCL13+CD103+CD8+ T-cell population is associated with B-cell recruitment and neoantigen load in human cancer. Cancer Immunol Res, 7(5):784-796. ![]() [112]WoutersMCA, NelsonBH, 2018. Prognostic significance of tumor-infiltrating B cells and plasma cells in human cancer. Clin Cancer Res, 24(24):6125-6135. ![]() [113]WuSZ, RodenDL, WangCF, et al., 2020. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. EMBO J, 39(19):e104063. ![]() [114]WykesM, PomboA, JenkinsC, et al., 1998. Dendritic cells interact directly with naive B lymphocytes to transfer antigen and initiate class switching in a primary T-dependent response. J Immunol, 161(3):1313-1319. ![]() [115]YangSC, BatraRK, HillingerS, et al., 2006. Intrapulmonary administration of CCL21 gene-modified dendritic cells reduces tumor burden in spontaneous murine bronchoalveolar cell carcinoma. Cancer Res, 66(6):3205-3213. ![]() [116]YehCH, FinneyJ, OkadaT, et al., 2022. Primary germinal center-resident T follicular helper cells are a physiologically distinct subset of CXCR5hiPD-1hi T follicular helper cells. Immunity, 55(2):272-289.e7. ![]() [117]YoshidaH, NaitoA, InoueJ, et al., 2002. Different cytokines induce surface lymphotoxin-αβ on IL-7 receptor-α cells that differentially engender lymph nodes and Peyer’s patches. Immunity, 17(6):823-833. ![]() Journal of Zhejiang University-SCIENCE, 38 Zheda Road, Hangzhou
310027, China
Tel: +86-571-87952783; E-mail: cjzhang@zju.edu.cn Copyright © 2000 - 2025 Journal of Zhejiang University-SCIENCE |
Open peer comments: Debate/Discuss/Question/Opinion
<1>