Full Text:  <414>

Summary:  <88>

CLC number: 

On-line Access: 2023-06-13

Received: 2022-11-27

Revision Accepted: 2023-02-27

Crosschecked: 2023-09-13

Cited: 0

Clicked: 537

Citations:  Bibtex RefMan EndNote GB/T7714

 ORCID:

Weidong WANG

https://orcid.org/0000-0003-2257-0314

-   Go to

Article info.
Open peer comments

Journal of Zhejiang University SCIENCE B

Accepted manuscript available online (unedited version)


Detection and quantitative analysis of tumor-associated tertiary lymphoid structures


Author(s):  Man YANG, Yurou CHE, Kezhen LI, Zengyi FANG, Simin LI, Mei WANG, Yiyao ZHANG, Zhu XU, Liping LUO, Chuan WU, Xin LAI, Weidong WANG

Affiliation(s):  Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China; more

Corresponding email(s):  wwdwyl@sina.com

Key Words:  Tertiary lymphoid structure; Tumor microenvironment; Chemokine signature; Spatial omics; Artificial intelligence; Radiomics


Share this article to: More <<< Previous Paper|Next Paper >>>

Man YANG, Yurou CHE, Kezhen LI, Zengyi FANG, Simin LI, Mei WANG, Yiyao ZHANG, Zhu XU, Liping LUO, Chuan WU, Xin LAI, Weidong WANG. Detection and quantitative analysis of tumor-associated tertiary lymphoid structures[J]. Journal of Zhejiang University Science B,in press.Frontiers of Information Technology & Electronic Engineering,in press.https://doi.org/10.1631/jzus.B2200605

@article{title="Detection and quantitative analysis of tumor-associated tertiary lymphoid structures",
author="Man YANG, Yurou CHE, Kezhen LI, Zengyi FANG, Simin LI, Mei WANG, Yiyao ZHANG, Zhu XU, Liping LUO, Chuan WU, Xin LAI, Weidong WANG",
journal="Journal of Zhejiang University Science B",
year="in press",
publisher="Zhejiang University Press & Springer",
doi="https://doi.org/10.1631/jzus.B2200605"
}

%0 Journal Article
%T Detection and quantitative analysis of tumor-associated tertiary lymphoid structures
%A Man YANG
%A Yurou CHE
%A Kezhen LI
%A Zengyi FANG
%A Simin LI
%A Mei WANG
%A Yiyao ZHANG
%A Zhu XU
%A Liping LUO
%A Chuan WU
%A Xin LAI
%A Weidong WANG
%J Journal of Zhejiang University SCIENCE B
%P 779-795
%@ 1673-1581
%D in press
%I Zhejiang University Press & Springer
doi="https://doi.org/10.1631/jzus.B2200605"

TY - JOUR
T1 - Detection and quantitative analysis of tumor-associated tertiary lymphoid structures
A1 - Man YANG
A1 - Yurou CHE
A1 - Kezhen LI
A1 - Zengyi FANG
A1 - Simin LI
A1 - Mei WANG
A1 - Yiyao ZHANG
A1 - Zhu XU
A1 - Liping LUO
A1 - Chuan WU
A1 - Xin LAI
A1 - Weidong WANG
J0 - Journal of Zhejiang University Science B
SP - 779
EP - 795
%@ 1673-1581
Y1 - in press
PB - Zhejiang University Press & Springer
ER -
doi="https://doi.org/10.1631/jzus.B2200605"


Abstract: 
Tumor-associated tertiary lymphoid structures (TLSs) are ectopic lymphoid formations within tumor tissue, with mainly B and T cell populations forming the organic aggregates. The presence of TLSs in tumors has been strongly associated with patient responsiveness to immunotherapy regimens and improving tumor prognosis. Researchers have been motivated to actively explore TLSs due to their bright clinical application prospects. Various studies have attempted to decipher TLSs regarding their formation mechanism, structural composition, induction generation, predictive markers, and clinical utilization. Meanwhile, the scientific approaches to qualitative and quantitative descriptions are crucial for TLS studies. In terms of detection, hematoxylin and eosin (H&E), multiplex immunohistochemistry (mIHC), multiplex immunofluorescence (mIF), and 12-chemokine gene signature have been the top approved methods. However, no standard methods exist for the quantitative analysis of TLSs, such as absolute TLS count, analysis of TLS constituent cells, structural features, TLS spatial location, density, and maturity. This study reviews the latest research progress on TLS detection and quantification, proposes new directions for TLS assessment, and addresses issues for the quantitative application of TLSs in the clinic.

肿瘤相关的三级淋巴结构的检测和量化分析

杨曼1,2,车雨柔1,2,李柯臻1,3,方曾怡1,2,李思敏1,3,王梅1,2,张艺耀1,2,徐祝1,3,罗丽萍1,2,吴川1,2,赖昕1,2,王卫东1,2,3
1电子科技大学附属肿瘤医院,放射肿瘤学四川省重点实验室,四川省肿瘤临床医学研究中心,四川省肿瘤医院放疗科,中国成都市,610000
2电子科技大学医学院,中国成都市,610000
3西南医科大学临床医学院肿瘤科,中国泸州市,646000
摘要:肿瘤相关的三级淋巴结构(TLS),主要由B细胞和T细胞群体有机聚集形成于肿瘤组织范围内的异位淋巴组织。在肿瘤中,TLS的存在与免疫治疗的反应性和肿瘤预后密切相关。因其具有广阔的临床应用前景,研究者一直在积极探索。许多研究都试图破译TLS的形成机制、结构组成、诱导生成、预测标记物和临床利用。与此同时,用科学的方法定性和定量描述TLS,对其研究至关重要。在检测方面,苏木精-伊红染色法(H&E)染色、多重免疫组化、多重免疫荧光和12趋化因子基因特征是被认可的方法。然而在TLS的定量分析方面,例如TLS的绝对计数、组成TLS的细胞分析、TLS的结构特征、空间位置、密度、成熟度等,目前尚无标准方法。本研究回顾了TLS检测和定量分析的最新研究进展,提出了TLS评估的新方向,并解决了TLS在临床上的定量应用问题。

关键词组:三级淋巴结构(TLS);肿瘤微环境;趋化因子特征;空间转录组学;人工智能;影像组学

Darkslateblue:Affiliate; Royal Blue:Author; Turquoise:Article

Reference

[1]AbelsE, PantanowitzL, AeffnerF, et al., 2019. Computational pathology definitions, best practices, and recommendations for regulatory guidance: a white paper from the Digital Pathology Association. J Pathol, 249(3):286-294.

[2]AgerA, 2017. High endothelial venules and other blood vessels: critical regulators of lymphoid organ development and function. Front Immunol, 8:45.

[3]AhmedA, KöhlerS, KlotzR, et al., 2022. Tertiary lymphoid structures and their association to immune phenotypes and circulatory IL2 levels in pancreatic ductal adenocarcinoma. Oncoimmunology, 11:2027148.

[4]AmariaRN, ReddySM, TawbiHA, et al., 2018. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med, 24(11):1649-1654.

[5]AnthimopoulosM, ChristodoulidisS, EbnerL, et al., 2016. Lung pattern classification for interstitial lung diseases using a deep convolutional neural network. IEEE Trans Med Imaging, 35(5):1207-1216.

[6]BarmpoutisP, di CapiteM, KayhanianH, et al., 2021. Tertiary lymphoid structures (TLS) identification and density assessment on H&E-stained digital slides of lung cancer. PLoS ONE, 16(9):e0256907.

[7]BénézechC, LuuNT, WalkerJA, et al., 2015. Inflammation-induced formation of fat-associated lymphoid clusters. Nat Immunol, 16(8):819-828.

[8]BenzerdjebN, DartiguesP, KepenekianV, et al., 2021. Tertiary lymphoid structures in epithelioid malignant peritoneal mesothelioma are associated with neoadjuvant chemotherapy, but not with prognosis. Virchows Arch, 479(4):765-772.

[9]BoivinG, KalambadenP, FagetJ, et al., 2018. Cellular composition and contribution of tertiary lymphoid structures to tumor immune infiltration and modulation by radiation therapy. Front Oncol, 8:256.

[10]BramanNM, EtesamiM, PrasannaP, et al., 2017. Intratumoral and peritumoral radiomics for the pretreatment prediction of pathological complete response to neoadjuvant chemotherapy based on breast DCE-MRI. Breast Cancer Res, 19:57.

[11]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.

[12]CabritaR, LaussM, SannaA, et al., 2020. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature, 577(7791):561-565.

[13]CadizF, GormazJG, BurottoM, 2018. Breast cancer staging: is TNM ready to evolve? J Glob Oncol, 4:1-3.

[14]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.

[15]ChanHP, SamalaRK, HadjiiskiLM, et al., 2020. Deep learning in medical image analysis. In: Lee G, Fujita H (Eds.), Deep Learning in Medical Image Analysis: Challenges and Applications. Springer, Cham, p.3-21.

[16]ChaurioRA, AnadonCM, CostichTL, et al., 2022. TGF-‍‍β- mediated silencing of genomic organizer SATB1 promotes Tfh cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity, 55(1):115-128.e9.

[17]ClubbJHA, KudlingTV, HeiniöC, et al., 2022. Adenovirus encoding tumor necrosis factor alpha and interleukin 2 induces a tertiary lymphoid structure signature in immune checkpoint inhibitor refractory head and neck cancer. Front Immunol, 13:794251.

[18]ColbeckEJ, AgerA, GallimoreA, et al., 2017. Tertiary lymphoid structures in cancer: drivers of antitumor immunity, immunosuppression, or bystander sentinels in disease? Front Immunol, 8:1830.

[19]CoppolaD, NebozhynM, KhalilF, et al., 2011. Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am J Pathol, 179(1):37-45.

[20]CoudrayN, OcampoPS, SakellaropoulosT, et al., 2018. Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat Med, 24(10):1559-1567.

[21]CuiM, ZhangDY, 2021. Artificial intelligence and computational pathology. Lab Invest, 101(4):412-422.

[22]DaumS, HagenH, NaismithE, et al., 2021. The role of anti-angiogenesis in the treatment landscape of non-small cell lung cancer ‒ new combinational approaches and strategies of neovessel inhibition. Front Cell Dev Biol, 8:610903.

[23]de ChaisemartinL, GocJ, DamotteD, et al., 2011. Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res, 71(20):6391-6399.

[24]de SilvaNS, KleinU, 2015. Dynamics of B cells in germinal centres. Nat Rev Immunol, 15(3):137-148.

[25]DeguchiS, TanakaH, SuzukiS, et al., 2022. Clinical relevance of tertiary lymphoid structures in esophageal squamous cell carcinoma. BMC Cancer, 22:699.

[26]DelvecchioFR, FinchamREA, SpearS, et al., 2021. Pancreatic cancer chemotherapy is potentiated by induction of tertiary lymphoid structures in mice. Cell Mol Gastroenterol Hepatol, 12(5):1543-1565.

[27]DeteixC, Attuil-AudenisV, DutheyA, et al., 2010. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J Immunol, 184(9):5344-5351.

[28]di CaroG, BergomasF, GrizziF, et al., 2014. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin Cancer Res, 20(8):2147-2158.

[29]Dieu-NosjeanMC, AntoineM, DanelC, et al., 2008. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol, 26(27):4410-4417.

[30]Dieu-NosjeanMC, GocJ, GiraldoNA, et al., 2014. Tertiary lymphoid structures in cancer and beyond. Trends Immunol, 35(11):571-580.

[31]Dieu-NosjeanMC, GiraldoNA, KaplonH, et al., 2016. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev, 271(1):260-275.

[32]DingGY, MaJQ, YunJP, et al., 2022. Distribution and density of tertiary lymphoid structures predict clinical outcome in intrahepatic cholangiocarcinoma. J Hepatol, 76(3):608-618.

[33]DraytonDL, LiaoS, MounzerRH, et al., 2006. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol, 7(4):344-353.

[34]EberlG, MarmonS, SunshineMJ, et al., 2004. An essential function for the nuclear receptor RORγt in the generation of fetal lymphoid tissue inducer cells. Nat Immunol, 5(1):64-73.

[35]FinkinS, YuanDT, SteinI, et al., 2015. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol, 16(12):1235-1244.

[36]FridmanWH, ZitvogelL, Sautès-FridmanC, et al., 2017. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol, 14(12):717-734.

[37]FurtadoGC, MarinkovicT, MartinAP, et al., 2007. Lymphotoxin β receptor signaling is required for inflammatory lymphangiogenesis in the thyroid. Proc Natl Acad Sci USA, 104(12):5026-5031.

[38]Gago da GraçaC, van BaarsenLGM, MebiusRE, 2021. Tertiary lymphoid structures: diversity in their development, composition, and role. J Immunol, 206(2):273-281.

[39]GalonJ, CostesA, Sanchez-CaboF, et al., 2006. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science, 313(5795):1960-1964.

[40]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.

[41]GirardJP, MoussionC, FörsterR, 2012. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol, 12(11):762-773.

[42]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.

[43]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.

[44]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.

[45]GuptaS, ZugazagoitiaJ, Martinez-MorillaS, et al., 2020. Digital quantitative assessment of PD-L1 using digital spatial profiling. Lab Invest, 100(10):1311-1317.

[46]HattoriA, TakamochiK, OhS, et al., 2019. New revisions and current issues in the eighth edition of the TNM classification for non-small cell lung cancer. Jpn J Clin Oncol, 49(1):3-11.

[47]HelminkBA, ReddySM, GaoJJ, et al., 2020. B cells and tertiary lymphoid structures promote immunotherapy response. Nature, 577(7791):549-555.

[48]HorewegN, WorkelHH, LoieroD, et al., 2022. Tertiary lymphoid structures critical for prognosis in endometrial cancer patients. Nat Commun, 13:1373.

[49]HoytCC, 2021. Multiplex immunofluorescence and multispectral imaging: forming the basis of a clinical test platform for immuno-oncology. Front Mol Biosci, 8:674747.

[50]JonesGW, HillDG, JonesSA, 2016. Understanding immune cells in tertiary lymphoid organ development: it is all starting to come together. Front Immunol, 7:401.

[51]KangWD, FengZC, LuoJW, et al., 2021. Tertiary lymphoid structures in cancer: the double-edged sword role in antitumor immunity and potential therapeutic induction strategies. Front Immunol, 12:689270.

[52]KoenigA, ThaunatO, 2016. Lymphoid neogenesis and tertiary lymphoid organs in transplanted organs. Front Immunol, 7:646.

[53]LadányiA, KissJ, SomlaiB, et al., 2007. Density of DC-LAMP+ mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol Immunother, 56(9):1459-1469.

[54]LaidlawBJ, CysterJG, 2021. Transcriptional regulation of memory B cell differentiation. Nat Rev Immunol, 21(4):209-220.

[55]LiH, WangJJ, LiuHL, et al., 2020. Existence of intratumoral tertiary lymphoid structures is associated with immune cells infiltration and predicts better prognosis in early-stage hepatocellular carcinoma. Aging, 12(4):3451-3472.

[56]LiH, LiuHL, FuHY, et al., 2021. Peritumoral tertiary lymphoid structures correlate with protective immunity and improved prognosis in patients with hepatocellular carcinoma. Front Immunol, 12:648812.

[57]LiPX, LiangY, ZengBZ, et al., 2022. Preoperative prediction of intra-tumoral tertiary lymphoid structures based on CT in hepatocellular cancer. Eur J Radiol, 151:110309.

[58]LiQX, LiuXQ, WangDK, et al., 2020. Prognostic value of tertiary lymphoid structure and tumour infiltrating lymphocytes in oral squamous cell carcinoma. Int J Oral Sci, 12:24.

[59]LiR, BerglundA, ZempL, et al., 2021. The 12-CK score: global measurement of tertiary lymphoid structures. Front Immunol, 12:694079.

[60]LiRT, HuangX, YangWM, et al., 2022. Tertiary lymphoid structures favor outcome in resected esophageal squamous cell carcinoma. J Pathol Clin Res, 8(5):422-435.

[61]LiaoS, RuddleNH, 2006. Synchrony of high endothelial venules and lymphatic vessels revealed by immunization. J Immunol, 177(5):3369-3379.

[62]LiuX, TsangJYS, HlaingT, et al., 2017. Distinct tertiary lymphoid structure associations and their prognostic relevance in HER2 positive and negative breast cancers. Oncologist, 22(11):1316-1324.

[63]LochnerM, OhnmachtC, PresleyL, et al., 2011. Microbiota-induced tertiary lymphoid tissues aggravate inflammatory disease in the absence of RORγt and LTi cells. J Exp Med, 208(1):125-134.

[64]LucchesiD, BombardieriM, 2013. The role of viruses in autoreactive B cell activation within tertiary lymphoid structures in autoimmune diseases. J Leukoc Biol, 94(6):1191-1199.

[65]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.

[66]ManzoA, BombardieriM, HumbyF, et al., 2010. Secondary and ectopic lymphoid tissue responses in rheumatoid arthritis: from inflammation to autoimmunity and tissue damage/remodeling. Immunol Rev, 233(1):267-285.

[67]MarliotF, ChenXY, KirilovskyA, et al., 2020a. Analytical validation of the Immunoscore and its associated prognostic value in patients with colon cancer. J Immunother Cancer, 8(1):e000272.

[68]MarliotF, LafontaineL, GalonJ, 2020b. Immunoscore assay for the immune classification of solid tumors: technical aspects, improvements and clinical perspectives. Methods Enzymol, 636:109-128.

[69]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.

[70]MartinetL, FilleronT, le GuellecS, et al., 2013. High endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated with lymphotoxin β‍-producing dendritic cells in human breast cancer. J Immunol, 191(4):2001-2008.

[71]MeierD, BornmannC, ChappazS, et al., 2007. Ectopic lymphoid-organ development occurs through interleukin 7-mediated enhanced survival of lymphoid-tissue-inducer cells. Immunity, 26(5):643-654.

[72]MessinaJL, FenstermacherDA, EschrichS, et al., 2012. 12-Chemokine gene signature identifies lymph node-like structures in melanoma: potential for patient selection for immunotherapy? Sci Rep, 2:765.

[73]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.

[74]MuellerCG, NayarS, CamposJ, et al., 2018. Molecular and cellular requirements for the assembly of tertiary lymphoid structures. In: Owens B, Lakins M (Eds.), Stromal Immunology. Springer, Cham, p.55-72.

[75]NakamuraM, MagaraT, KanoS, et al., 2022. Tertiary lymphoid structures and chemokine landscape in virus-positive and virus-negative Merkel cell carcinoma. Front Oncol, 12:811586.

[76]NawazS, YuanYY, 2016. Computational pathology: exploring the spatial dimension of tumor ecology. Cancer Lett, 380(1):296-303.

[77]NeytK, PerrosF, GeurtsvanKesselCH, et al., 2012. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol, 33(6):297-305.

[78]PagèsF, MlecnikB, MarliotF, et al., 2018. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet, 391(10135):2128-2139.

[79]PetersA, PitcherLA, SullivanJM, et al., 2011. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity, 35(6):986-996.

[80]PetitprezF, de ReynièsA, KeungEZ, et al., 2020. B cells are associated with survival and immunotherapy response in sarcoma. Nature, 577(7791):556-560.

[81]PimentaEM, BarnesBJ, 2014. Role of Tertiary lymphoid structures (TLS) in anti-tumor immunity: potential tumor-induced cytokines/chemokines that regulate TLS formation in epithelial-derived cancers. Cancers, 6(2):969-997.

[82]PipiE, NayarS, GardnerDH, et al., 2018. Tertiary lymphoid structures: autoimmunity goes local. Front Immunol, 9:1952.

[83]PitzalisC, JonesGW, BombardieriM, et al., 2014. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol, 14(7):447-462.

[84]PoschF, SilinaK, LeiblS, et al., 2018. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. OncoImmunology, 7(2):e1378844.

[85]PrabhakaranS, RizkVT, MaZJ, et al., 2017. Evaluation of invasive breast cancer samples using a 12-chemokine gene expression score: correlation with clinical outcomes. Breast Cancer Res, 19:71.

[86]QinM, HamanishiJ, UkitaM, et al., 2022. Tertiary lymphoid structures are associated with favorable survival outcomes in patients with endometrial cancer. Cancer Immunol Immunother, 71(6):1431-1442.

[87]RakaeeM, KilvaerTK, JamalyS, et al., 2021. Tertiary lymphoid structure score: a promising approach to refine the TNM staging in resected non-small cell lung cancer. Br J Cancer, 124(10):1680-1689.

[88]Ramos-CasalsM, BrahmerJR, CallahanMK, et al., 2020. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers, 6:38.

[89]RandolphGJ, BalaS, RahierJF, et al., 2016. Lymphoid aggregates remodel lymphatic collecting vessels that serve mesenteric lymph nodes in Crohn disease. Am J Pathol, 186(12):3066-3073.

[90]RenFP, XieM, GaoJ, et al., 2022. Tertiary lymphoid structures in lung adenocarcinoma: characteristics and related factors. Cancer Med, 11(15):2969-2977.

[91]RodriguezAB, EngelhardVH, 2020. Insights into tumor-associated tertiary lymphoid structures: novel targets for antitumor immunity and cancer immunotherapy. Cancer Immunol Res, 8(11):1338-1345.

[92]RodriguezAB, PeskeJD, WoodsAN, et al., 2021. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep, 36(3):109422.

[93]Sautès-FridmanC, PetitprezF, CalderaroJ, et al., 2019. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer, 19(6):307-325.

[94]SchumacherTN, ThommenDS, 2022. Tertiary lymphoid structures in cancer. Science, 375(6576):eabf9419.

[95]SiliņaK, SoltermannA, AttarFM, et al., 2018a. 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.

[96]SiliņaK, BurkhardtC, CasanovaR, et al., 2018b. A quantitative pathology approach to analyze the development of human cancer-associated tertiary lymphoid structures. In: Dieu-Nosjean MC (Ed.), Tertiary Lymphoid Structures: Methods and Protocols. Humana Press, New York, p.71-86.

[97]SofopoulosM, FortisSP, VaxevanisCK, et al., 2019. The prognostic significance of peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol Immunother, 68(11):1733-1745.

[98]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.

[99]TangJ, Ramis-CabrerD, CurullV, et al., 2020. B cells and tertiary lymphoid structures influence survival in lung cancer patients with resectable tumors. Cancers, 12(9):2644.

[100]ThaunatO, PateyN, CaligiuriG, et al., 2010. Chronic rejection triggers the development of an aggressive intragraft immune response through recapitulation of lymphoid organogenesis. J Immunol, 185(1):717-728.

[101]TokunagaR, NakagawaS, SakamotoY, et al., 2020. 12-Chemokine signature, a predictor of tumor recurrence in colorectal cancer. Int J Cancer, 147(2):532-541.

[102]TrajkovskiG, OgnjenovicL, KaradzovZ, et al., 2018. Tertiary lymphoid structures in colorectal cancers and their prognostic value. Open Access Maced J Med Sci, 6(10):1824-1828.

[103]TumehPC, HarviewCL, YearleyJH, et al., 2014. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature, 515(7528):568-571.

[104]VaidyaP, BeraK, PatilPD, et al., 2020. Novel, non-invasive imaging approach to identify patients with advanced non-small cell lung cancer at risk of hyperprogressive disease with immune checkpoint blockade. J Immunother Cancer, 8(2):e001343.

[105]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.

[106]VanherseckeL, BrunetM, GuéganJP, et al., 2021. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat Cancer, 2(8):794-802.

[107]van HoorenL, VaccaroA, RamachandranM, et al., 2021. Agonistic CD40 therapy induces tertiary lymphoid structures but impairs responses to checkpoint blockade in glioma. Nat Commun, 12:4127.

[108]van RijthovenM, BalkenholM, SiliņaK, et al., 2021. HookNet: multi-resolution convolutional neural networks for semantic segmentation in histopathology whole-slide images. Med Image Anal, 68:101890.

[109]VanTM, BlankCU, 2019. A user’s perspective on GeoMxTM digital spatial profiling. Immunooncol Technol, 1:11-18.

[110]VondenhoffMF, GreuterM, GoverseG, et al., 2009. LTβR signaling induces cytokine expression and up-regulates lymphangiogenic factors in lymph node anlagen. J Immunol, 182(9):5439-5445.

[111]WangC, HuangZX, ZhangM, et al., 2021. Prognostic value of tertiary lymphoid structures in early clinical stage oral tongue squamous cell carcinoma. J Oral Pathol Med, 50(8):776-784.

[112]WangDY, SalemJE, CohenJV, et al., 2018. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol, 4(12):1721-1728.

[113]WangJ, FosterA, ChinR, et al., 2002. The complementation of lymphotoxin deficiency with LIGHT, a newly discovered TNF family member, for the restoration of secondary lymphoid structure and function. Eur J Immunol, 32(7):1969-1979.

[114]WangYC, ZhouSH, YangF, et al., 2019. Treatment-related adverse events of PD-1 and PD-L1 inhibitors in clinical trials: a systematic review and meta-analysis. JAMA Oncol, 5(7):1008-1019.

[115]WernerF, WagnerC, SimonM, et al., 2021. A standardized analysis of tertiary lymphoid structures in human melanoma: disease progression- and tumor site-associated changes with germinal center alteration. Front Immunol, 12:675146.

[116]WuYC, ChengYF, WangXD, et al., 2022. Spatial omics: navigating to the golden era of cancer research. Clin Transl Med, 12(1):e696.

[117]XuWH, MaCG, LiuWR, et al., 2022. Prognostic value, DNA variation and immunologic features of a tertiary lymphoid structure-related chemokine signature in clear cell renal cell carcinoma. Cancer Immunol Immunother, 71(8):1923-1935.

[118]XuY, SuGH, MaD, et al., 2021. Technological advances in cancer immunity: from immunogenomics to single-cell analysis and artificial intelligence. Signal Transduct Target Ther, 6:312.

[119]ZempL, BerglundAE, DhillonJ, et al., 2021. The prognostic and predictive implications of the 12-chemokine score in muscle invasive bladder cancer. J Clin Oncol, 39(S6):466.

[120]ZhangJ, WuZR, ZhangX, et al., 2020. Machine learning: an approach to preoperatively predict PD-1/PD-L1 expression and outcome in intrahepatic cholangiocarcinoma using MRI biomarkers. ESMO Open, 5(6):e000910.

[121]ZhangY, LiJ, YangF, et al., 2022. Relationship and prognostic significance of IL-33, PD-1/PD-L1, and tertiary lymphoid structures in cervical cancer. J Leukoc Biol, 112(6):‍1591-1603.

[122]ZhouL, XuB, LiuYS, et al., 2021. Tertiary lymphoid structure signatures are associated with survival and immunotherapy response in muscle-invasive bladder cancer. OncoImmunology, 10(1):1915574.

[123]ZhouQ, ZhouZY, ChenCM, et al., 2019. Grading of hepatocellular carcinoma using 3D SE-DenseNet in dynamic enhanced MR images. Comput Biol Med, 107:47-57.

Open peer comments: Debate/Discuss/Question/Opinion

<1>

Please provide your name, email address and a comment





Journal of Zhejiang University-SCIENCE, 38 Zheda Road, Hangzhou 310027, China
Tel: +86-571-87952783; E-mail: cjzhang@zju.edu.cn
Copyright © 2000 - 2024 Journal of Zhejiang University-SCIENCE