Unveiling the impact of tertiary lymphoid structures on immunotherapeutic responses of clear cell renal cell carcinoma

Wenhao Xu, Jiahe Lu, Xi Tian, Shiqi Ye, Shiyin Wei, Jun Wang, Aihetaimujiang Anwaier, Yuanyuan Qu, Wangrui Liu, Kun Chang, Hailiang Zhang, Dingwei Ye

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (1) : e461. DOI: 10.1002/mco2.461
ORIGINAL ARTICLE

Unveiling the impact of tertiary lymphoid structures on immunotherapeutic responses of clear cell renal cell carcinoma

Author information +
History +

Abstract

Tertiary lymphoid structures (TLS) are organized aggregates of immune cells that form under pathological conditions. However, the predictive value of TLS in clear cell renal cell carcinoma (ccRCC) for immunotherapies remains unclear. We comprehensively assessed the implications for prognosis and immunological responses of the TLS spatial and maturation heterogeneity in 655 ccRCC patients. A higher proportion of early-TLS was found in peritumoral TLS, while intratumoral TLS mainly comprised secondary follicle-like TLS (SFL-TLS), indicating markedly better survival. Notably, presence of TLS, especially intratumoral TLS and SFL-TLS, significantly correlated with better survival and objective reflection rate for ccRCC patients receiving anti-Programmed Cell Death Protein-1 (PD-1)/Programmed Cell Death-Ligand-1 (PD-L1) immunotherapies. In peritumoral TLS cluster, primary follicle-like TLS, the proportion of tumor-associated macrophages, and Treg infiltration in the peritumoral regions increased prominently, suggesting an immunosuppressive tumor microenvironment. Interestingly, spatial transcriptome annotation and multispectral fluorescence showed that an abundance of mature plasma cells within mature TLS has the capacity to produce IgA and IgG, which demonstrate significantly higher objective response rates and a superior prognosis for ccRCC patients subjected to immunotherapy. In conclusion, this study revealed the implications of TLS spatial and maturation heterogeneity on the immunological status and clinical responses, allowing the improvement of precise immunotherapies of ccRCC.

Keywords

clear cell renal cell carcinoma / immune checkpoint inhibitors / tertiary lymphoid structures / tumor heterogeneity / tumor microenvironment

Cite this article

Download citation ▾
Wenhao Xu, Jiahe Lu, Xi Tian, Shiqi Ye, Shiyin Wei, Jun Wang, Aihetaimujiang Anwaier, Yuanyuan Qu, Wangrui Liu, Kun Chang, Hailiang Zhang, Dingwei Ye. Unveiling the impact of tertiary lymphoid structures on immunotherapeutic responses of clear cell renal cell carcinoma. MedComm, 2024, 5(1): e461 https://doi.org/10.1002/mco2.461

References

[1]
Renshaw AA. Subclassification of renal cell neoplasms: an update for the practising pathologist. Histopathology. 2002;41:283-300.
[2]
Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373:1119-1132.
[3]
Xu W, Anwaier A, Liu W, et al. The unique genomic landscape and prognostic mutational signature of Chinese clear cell renal cell carcinoma. J Natl Cancer Center. 2022.
[4]
Zheng R, Zhang S, Zeng H, et al. Cancer incidence and mortality in China, 2016. J Natl Cancer Center. 2022;2(1):1-9.
[5]
George S, Rini BI, Hammers HJ. Emerging role of combination immunotherapy in the first-line treatment of advanced renal cell carcinoma: a review. JAMA Oncol. 2019;5:411-421.
[6]
Rini BI, Signoretti S, Choueiri TK, et al. Long-term outcomes with nivolumab plus ipilimumab versus sunitinib in first-line treatment of patients with advanced sarcomatoid renal cell carcinoma. J Immunother Cancer. 2022;10:e005445.
[7]
Kim MC, Jin Z, Kolb R, et al. Updates on immunotherapy and immune landscape in renal clear cell carcinoma. Cancers (Basel). 2021;13:5856.
[8]
Qu Y, Wu X, Anwaier A, et al. Proteogenomic characterization of MiT family translocation renal cell carcinoma. Nat Commun. 2022;13:7494.
[9]
Qu Y, Feng J, Wu X, et al. A proteogenomic analysis of clear cell renal cell carcinoma in a Chinese population. Nat Commun. 2022;13:2052.
[10]
Xie Y, Sahin M, Sinha S, et al. SETD2 loss perturbs the kidney cancer epigenetic landscape to promote metastasis and engenders actionable dependencies on histone chaperone complexes. Nat Cancer. 2022;3:188-202.
[11]
Xu W, Anwaier A, Ma C, et al. Prognostic immunophenotyping clusters of clear cell renal cell carcinoma defined by the unique tumor immune microenvironment. Front Cell Dev Biol. 2021;9:785410.
[12]
Xu W, Liu WR, Xu Y, et al. Hexokinase 3 dysfunction promotes tumorigenesis and immune escape by upregulating monocyte/macrophage infiltration into the clear cell renal cell carcinoma microenvironment. Int J Biol Sci. 2021;17:2205-2222.
[13]
Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375:eabf9419.
[14]
Fridman WH, Meylan M, Pupier G, Calvez A, Hernandez I, Sautès-Fridman C. Tertiary lymphoid structures and B cells: an intratumoral immunity cycle. Immunity. 2023;56:2254-2269.
[15]
Meylan M, Petitprez F, Lacroix L, et al. Early hepatic lesions display immature tertiary lymphoid structures and show elevated expression of immune inhibitory and immunosuppressive molecules. Clin Cancer Res. 2020;26:4381-4389.
[16]
Tietscher S, Wagner J, Anzeneder T, et al. A comprehensive single-cell map of T cell exhaustion-associated immune environments in human breast cancer. Nat Commun. 2023;14:98.
[17]
Zhang H, AbdulJabbar K, Moore DA, et al. Spatial positioning of immune hotspots reflects the interplay between B and T cells in lung squamous cell carcinoma. Cancer Res. 2023;83:1410-1425.
[18]
Meylan M, Petitprez F, Becht E, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity. 2022;55:527-541.e525.
[19]
Bruni D, Angell HK, Galon J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020;20:662-680.
[20]
Chevrier S, Levine JH, Zanotelli VRT, et al. An immune atlas of clear cell renal cell carcinoma. Cell. 2017;169:736-749.e718.
[21]
Geissler K, Fornara P, Lautenschlager C, Holzhausen HJ, Seliger B, Riemann D. Immune signature of tumor infiltrating immune cells in renal cancer. Oncoimmunology. 2015;4:e985082.
[22]
Jansen CS, Prokhnevska N, Master VA, et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature. 2019;576:465-470.
[23]
Giraldo NA, Becht E, Pages F, et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin Cancer Res. 2015;21:3031-3040.
[24]
Vanhersecke L, Brunet M, Guegan JP, et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat Cancer. 2021;2:794-802.
[25]
Masuda T, Tanaka N, Takamatsu K, et al. Unique characteristics of tertiary lymphoid structures in kidney clear cell carcinoma: prognostic outcome and comparison with bladder cancer. J Immunother Cancer. 2022;10:e003883.
[26]
Calderaro J, Petitprez F, Becht E, et al. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J Hepatol. 2019;70:58-65.
[27]
Willis SN, Mallozzi SS, Rodig SJ, et al. The microenvironment of germ cell tumors harbors a prominent antigen-driven humoral response. J Immunol. 2009;182:3310-3317.
[28]
Silina K, Soltermann A, Attar FM, et al. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. 2018;78:1308-1320.
[29]
Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2018;7:e1378844.
[30]
Domblides C, Rochefort J, Riffard C, et al. Tumor-associated tertiary lymphoid structures: from basic and clinical knowledge to therapeutic manipulation. Front Immunol. 2021;12:698604.
[31]
Munoz-Erazo L, Rhodes JL, Marion VC, Kemp RA. Tertiary lymphoid structures in cancer—considerations for patient prognosis. Cell Mol Immunol. 2020;17:570-575.
[32]
Sautes-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307-325.
[33]
Qian CN, Huang D, Wondergem B, Teh BT. Complexity of tumor vasculature in clear cell renal cell carcinoma. Cancer. 2009;115:2282-2289.
[34]
Vano YA, Elaidi R, Bennamoun M, et al. Nivolumab, nivolumab-ipilimumab, and VEGFR-tyrosine kinase inhibitors as first-line treatment for metastatic clear-cell renal cell carcinoma (BIONIKK): a biomarker-driven, open-label, non-comparative, randomised, phase 2 trial. Lancet Oncol. 2022;23:612-624.
[35]
White K, Connor K, Meylan M, et al. Identification, validation and biological characterisation of novel glioblastoma tumour microenvironment subtypes: implications for precision immunotherapy. Ann Oncol. 2023;34:300-314.
[36]
Chalabi M, Fanchi LF, Dijkstra KK, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26:566-576.
[37]
Hiraoka N, Ino Y, Yamazaki-Itoh R, Kanai Y, Kosuge T, Shimada K. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer. 2015;112:1782-1790.
[38]
Giatromanolaki A, Chatzipantelis P, Contrafouris CA, Koukourakis MI. Tertiary lymphoid structures, immune response, and prognostic relevance in non-small cell lung cancer. Cancer Invest. 2022;1–10:48-57.
[39]
Sofopoulos M, Fortis SP, Vaxevanis CK, et al. The prognostic significance of peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol Immunother. 2019;68:1733-1745.
[40]
Zhang FP, Zhu K, Zhu TF, et al. Intra-tumoral secondary follicle-like tertiary lymphoid structures are associated with a superior prognosis of overall survival of perihilar cholangiocarcinoma. Cancers (Basel). 2022;14:6107.
[41]
Helmink BA, Reddy SM, Gao J, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549-555.
[42]
Magen A, Hamon P, Fiaschi N, et al. Intratumoral dendritic cell-CD4(+) T helper cell niches enable CD8(+) T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat Med. 2023;29:1389-1399.
[43]
Gaglia G, Burger ML, Ritch CC, et al. Lymphocyte networks are dynamic cellular communities in the immunoregulatory landscape of lung adenocarcinoma. Cancer Cell. 2023;41:871-886.e810.
[44]
Luther SA, Bidgol A, Hargreaves DC, et al. Differing activities of homeostatic chemokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell recruitment and lymphoid neogenesis. J Immunol. 2002;169:424-433.
[45]
Furtado GC, Pacer ME, Bongers G, et al. TNFα-dependent development of lymphoid tissue in the absence of RORγt⁺ lymphoid tissue inducer cells. Mucosal Immunol. 2014;7:602-614.
[46]
Braun DA, Street K, Burke KP, et al. Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell. 2021;39:632-648.e638.
[47]
Cai J, Qi Q, Qian X, et al. The role of PD-1/PD-L1 axis and macrophage in the progression and treatment of cancer. J Cancer Res Clin Oncol. 2019;145:1377-1385.
[48]
Murakami T, Tanaka N, Takamatsu K, et al. Multiplexed single-cell pathology reveals the association of CD8 T-cell heterogeneity with prognostic outcomes in renal cell carcinoma. Cancer Immunol Immunother. 2021;70:3001-3013.
[49]
Krishna C, DiNatale RG, Kuo F, et al. Single-cell sequencing links multiregional immune landscapes and tissue-resident T cells in ccRCC to tumor topology and therapy efficacy. Cancer Cell. 2021;39:662-677.e666.
[50]
Carlisle JW, Jansen CS, Cardenas MA, et al. Clinical outcome following checkpoint therapy in renal cell carcinoma is associated with a burst of activated CD8 T cells in blood. J Immunother Cancer. 2022;10:e004803.
[51]
Bill R, Wirapati P, Messemaker M, et al. CXCL9:sPP1 macrophage polarity identifies a network of cellular programs that control human cancers. Science. 2023;381:515-524.
[52]
Johansson-Percival A, He B, Li ZJ, et al. De novo induction of intratumoral lymphoid structures and vessel normalization enhances immunotherapy in resistant tumors. Nat Immunol. 2017;18:1207-1217.
[53]
van Hooren L, Vaccaro A, Ramachandran M, et al. Agonistic CD40 therapy induces tertiary lymphoid structures but impairs responses to checkpoint blockade in glioma. Nat Commun. 2021;12:4127.
[54]
Xu W, Anwaier A, Liu W, et al. Genomic alteration of MTAP/CDKN2A predicts sarcomatoid differentiation and poor prognosis and modulates response to immune checkpoint blockade in renal cell carcinoma. Front Immunol. 2022;13:953721.
[55]
van Dijk N, Gil-Jimenez A, Silina K, et al. Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: the NABUCCO trial. Nat Med. 2020;26:1839-1844.
[56]
Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561-565.
[57]
Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573-3587.e3529.
[58]
Becht E, Giraldo NA, Lacroix L, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17:218.
[59]
Xu W, Wu Y, Liu W, et al. Tumor-associated macrophage-derived chemokine CCL5 facilitates the progression and immunosuppressive tumor microenvironment of clear cell renal cell carcinoma. Int J Biol Sci. 2022;18:4884-4900.
[60]
Ju WT, Xia RH, Zhu DW, et al. A pilot study of neoadjuvant combination of anti-PD-1 camrelizumab and VEGFR2 inhibitor apatinib for locally advanced resectable oral squamous cell carcinoma. Nat Commun. 2022;13:5378.
[61]
Zhang C, Wang XY, Zuo JL, et al. Localization and density of tertiary lymphoid structures associate with molecular subtype and clinical outcome in colorectal cancer liver metastases. J Immunother Cancer. 2023;11:e006425.
[62]
Goc J, Germain C, Vo-Bourgais TK, et al. 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. 2014;74:705-715.
[63]
Martinet L, Filleron T, Le Guellec S, Rochaix P, Garrido I, Girard JP. High endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated with lymphotoxin beta-producing dendritic cells in human breast cancer. J Immunol. 2013;191:2001-2008.

RIGHTS & PERMISSIONS

2024 2024 The Authors. MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.
PDF

Accesses

Citations

Detail

Sections
Recommended

/