Skip to main content
  • Systematic Review
  • Open access
  • Published:

Prognostic and clinicopathological significance of tertiary lymphoid structure in non-small cell lung cancer: a systematic review and meta-analysis

Abstract

Background

Non-small cell lung cancer (NSCLC) is the primary reason for cancer-related deaths globally. Tertiary lymphoid structure (TLS) is an organized collection of immune cells acquired in non-physiological, non-lymphoid tissues. High expression of TLS in tumor tissues is generally associated with better prognosis. This research aimed to investigate the prognostic and clinicopathological significance of TLS in patients with NSCLC.

Methods

A comprehensive literature search was conducted based on Pubmed, EMBASE, and Cochrane Library databases to identify eligible studies published up to December 8, 2023. The prognostic significance and clinicopathological value of TLS in NSCLC were evaluated by calculating the combined hazard ratios (HRs) and odds ratios (ORs) and their 95% confidence intervals (CIs). Following that, additional analyses, including subgroup analysis and sensitivity analysis, were conducted.

Results

This meta-analysis evaluated the prognostic and clinicopathological significance of TLS in 10 studies involving 1,451 patients with NSCLC. The results revealed that the high levels of TLS were strongly associated with better overall survival (OS) (HR = 0.48, 95% CI: 0.35–0.66, p < 0.001), disease-free survival (DFS)/recurrence-free survival (RFS) (HR = 0.37, 95% CI: 0.24–0.54, p < 0.001), and disease-specific survival (DSS) (HR = 0.45, 95% CI: 0.30–0.68, p < 0.001) in NSCLC patients. In addition, the increased expression of TLS was closely related to the Tumor Node Metastasis (TNM) stage of tumors (OR = 0.71, 95% CI: 0.51-1.00, p < 0.05) and neutrophil-lymphocyte ratio (NLR) (OR = 0.33, 95% CI: 0.17–0.62, p < 0.001).

Conclusions

The results revealed that highly expressed TLS is closely associated with a better prognosis in NSCLC patients. TLS may serve as a novel biomarker to predict the prognosis of NSCLC patients and guide the clinical treatment decisions.

Peer Review reports

Introduction

Lung cancer is one of the most prevalent cancers globally, with high death rates in both genders. The majority of lung cancers are attributed to non-small cell lung cancer (NSCLC), causing the most cancer-related deaths and ranking as the second most prevalent cancer globally [1,2,3]. In recent years, despite great progress in multidisciplinary treatment including surgery, radiotherapy, chemotherapy and immunotherapy, the prognosis of patients with NSCLC remains unsatisfactory. Hence, it is crucial to identify significant prognostic biomarkers for NSCLC to improve the clinical management of patients.

Tertiary lymphoid structure (TLS) is an abnormal lymphoid organ that closely resembles the secondary lymphoid organ (SLO) [4]. Under normal circumstances, TLS does not typically occur in the body, instead, it is found in non-lymphoid tissues where chronic inflammation is present [5]. TLS can develop in different pathophysiological conditions such as autoimmune diseases, infectious diseases, and tumors, leading to various effects that are influenced by the environment [6]. Recently, high levels of TLS have been proven to be linked to improved prognosis in various types of cancer [7, 8]. In patients with breast cancer, elevated levels of TLS is strongly related to a positive outlook for their prognosis [9]. In cases of gastrointestinal tumors, TLS can serve as a valuable prognostic indicator for gastrointestinal cancer and help direct the use of cancer immunotherapy [10]. Although numerous studies have explored the significance of TLS in predicting survival outcomes for NSCLC patients, the prognostic and clinicopathological significance of TLS in NSCLC remains controversial. For example, Brunet et al. found that progression-free survival (PFS) in the group of patients with TLS-positive tumors were not significantly different from patients with TLS-negative tumors [11]. However, some other studies have shown that elevated expression of TLS is closely related to better prognosis of patients with resectable NSCLC [12, 13].

In the present study, we conducted a meta-analysis to investigate the prognostic value of intratumoral TLS in patients with NSCLC, with the aim of providing evidence regarding the potential of TLS as a novel prognostic biomarker for NSCLC.

Materials and methods

Protocol and ethics statement

The reports of this systematic review and meta-analysis are in line with the Preferred Reporting Project for Systematic Review and Meta-Analysis (PRISMA) and the Meta-Analysis of Observational Epidemiological Studies (MOOSE) guidelines and statements [14, 15]. This systematic review and meta-analysis protocol has been registered on the PROSPERO website (https://www.crd.york.ac.uk/PROSPERO/) with the registration number CRD42024504484. All data used in this meta-analysis were from published studies, so ethical approval and patient consent were not required for this study.

Databases and search strategy

Two authors (Luyuan Ma and Rongyang Li) independently searched and assessed the availability of studies in each of the three databases: PubMed, EMBASE and the Cochrane Library, up to December 8th, 2023. Medical subject terms (MeSH) in the search strategy include “Tertiary lymphoid structure” and “Pulmonary Neoplasms” and “Prognosis”, and looked up free terms on PubMed. Various possible combinations of keywords and free words are made through two Boolean operators (“AND “and “OR”). The detailed search strategies for all databases are shown in Supplementary Table 1. In addition, we reviewed references in relevant articles for potential studies. Any disagreement between two reviewers is resolved by inviting other reviewers to discuss it.

Study selection and criteria

The primary studies included in this meta-analysis satisfied all of the criteria as follows: (I) This research focuses on individuals diagnosed with NSCLC. (II) Expression level of TLS in tumor tissues was clearly detected. (III) There are clear TLS grouping standards, which divide TLS into high/low expression groups for analysis and research. (IV) The relationship between TLS and survival outcomes or clinicopathological characteristics was evaluated in studies. Meanwhile, we excluded non-compliant studies by using the following criteria: (I) Reviews, meta-analyses, case reports, conference abstracts, letters and comments. (II) Animal experiments or basic research. (III) Studies that don’t have enough data to analyze. (IV) Multiple studies utilizing the same set of samples or participants.

Data extraction and quality assessment

From each of the studies that were included, we extracted the following information: authors, year of publication, country, study design, sample size, treatment, TLS detection methods, TLS location, cut-off criteria of TLS, follow-up time and survival outcomes. In addition, we collected the association of TLS with the age, gender, pathologic staging, smoking, Tumor Node Metastasis (TNM) staging and neutrophil-lymphocyte ratio (NLR) for patients. In the end, we extracted the hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) for overall survival (OS), disease-free survival (DFS)/recurrence-free survival (RFS), disease-specific survival (DSS) from each study. If a study conducts both univariate and multivariate analysis of variance, the results of the multivariate analysis will be used in further meta-analysis.

The quality of the included studies was evaluated using the Newcastle-Ottawa Quality Assessment Scale (NOS) [16]. Studies with scores equal to or higher than 6 points can be used for further meta-analysis. Two authors (Luyuan Ma and Rongyang Li) independently appraised the quality of each study, and all disagreements were resolved by consulting other researchers.

Statistical analysis

The prognostic significance of TLS in patients with NSCLC was evaluated by calculating the aggregated HRs and 95% CIs, and the association between TLS and clinicopathological features in patients was evaluated by the aggregated odds ratio (ORs) and 95% CIs. In cases where studies displayed Kaplan-Meier curves but did not provide HRs or 95% CIs, we determined the HRs and 95% CIs by analyzing the survival curves with Engauge Digitizer V4.1 (Markmitch, Goteborg, Sweden) [17]. To reduce possible bias, a random effects model was used to calculate the overall effect size. The degree of heterogeneity was measured using the Cochrane Q test and I² statistics, where I² values exceeding 50% were deemed to indicate significant heterogeneity. Subgroup analysis was performed to identify the source of heterogeneity. Potential publication bias was assessed by Egger’s and Begg’s test. In order to confirm the stability of the combined results, we conducted a sensitivity analysis to assess how each study influenced the overall estimate by omitting individual studies in turn. A bilateral P value less than 0.05 was deemed statistically significant. All statistical analyses were executed by Stata software (version 15.1; Stata Corp., College Station, Texas, USA) and Review Manager software (RevMan version 5.3, the Nordic Cochrane Center, the Cochrane Collaboration, 2014).

Results

Literature search

Through the literature search scheme, 237 documents with potential research value were retrieved, including 70 PubMed citations, 157 EMBASE citations, 9 Cochrane Library citations, and 1 relevant study yielded from the reference list. After eliminating duplicate publications, there were 171 studies left. By sifting through the titles and abstracts of each study, 31 studies remained. Finally, we carefully read the full text of the remaining articles, and 10 studies with 1,451 patients were included in our meta-analysis. A diagram illustrating the literature search process is shown in Fig. 1.

Fig. 1
figure 1

PRISMA flow diagram of literature search. PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses

Characteristics of the included studies

Table 1 describes the baseline characteristics and methodological assessments of each included study. There are ten retrospective studies published range from 2008 to 2023 from various regions of the globe. Four were published in China, two in Japan, two in America, one in Greece and one in Spain. The sample size of the study ranged from 59 to 490. It must be mentioned that all the eligible studies focused on intratumoral TLS, thus we mainly discuss the influence of intratumoral TLS on the prognosis of NSCLC patients. Of the included studies, patients in eight studies received surgical treatment only, and patients in three studies received neoadjuvant chemoimmunotherapy (NCIT) and surgical treatment. Notably, the study of Sun et al. examined both surgery-only and surgery with NCIT patients in relation to TLS, so we analyzed it as two studies [18]. In these ten studies, four evaluated the correlation between TLS and OS [19,20,21,22], seven evaluated the correlation between TLS and DFS/RFS [12, 13, 18, 19, 22,23,24], two evaluated the correlation between TLS and DSS [19, 25]. The studies that were included had NOS scores ranging from 7 to 9, suggesting that they are of high overall quality. Detailed quality assessments are presented in Supplementary Table 2.

Table 1 Baseline characteristics and methodological assessment of included studies

Prognostic value of TLS in patients with NSCLC

Eight studies involving 670 patients appraised the correlation between intratumoral TLS and DFS/RFS in NSCLC patients [12, 13, 18, 19, 22,23,24]. Pooled results revealed that high level of TLS is significantly associated with more favorable DFS/RFS (HR = 0.37, 95% CI: 0.24–0.54, p < 0.001) (Fig. 2A), with insignificant heterogeneity (I2 = 41.9%, p = 0.099). Subgroup analyses were conducted according to the treatment methods, TLS detection methods, and assessment of TLS cut-off values. The results showed that patients treated with neoadjuvant chemoimmunotherapy and surgery was correlated with better DFS/RFS, patients who used immunohistochemical (IHC) staining to detect TLS and those who used negative and positive TLS grouping had a better prognosis (Table 2, and Supplementary Fig. 1).

Fig. 2
figure 2

Forest plot of the correlation between TLS and (A) disease-free survival/recurrence-free survival, (B) overall survival, and (C) disease-specific survival in non-small cell lung cancer patients. TLS, tertiary lymphoid structures; HR, hazard ratio; CI, confidence interval

Table 2 Subgroup analyses of DFS/RFS in non-small cell lung cancer

Four studies appraised the association between intratumoral TLS and OS in 422 patients [19,20,21,22]. The pooled analysis revealed that high TLS was associated with preferable OS (HR = 0.48, 95% CI: 0.35–0.66, p < 0.001). The heterogeneity of the studies was low (I2 = 0.0%, p = 0.554) (Fig. 2B). Only two studies have appraised the association between intratumoral TLS and DSS in NSCLC patients [19, 25]. The results indicate that high TLS is closely related to batter DSS (HR = 0.45, 95% CI: 0.30–0.68, p < 0.001), with low heterogeneity (I2 = 26.3%, p = 0.244) (Fig. 2C).

Correlation between TLS and clinicopathological characteristics in NSCLC

The correlation analysis and evaluation results between TLS and various clinicopathological features are shown in Table 3. Overall, we examined the patients’ age (elder vs. young), gender (male vs. female), histological type (adenocarcinoma vs. squamous cell carcinoma), tumor size (large vs. small), smoking history (ever vs. never), TNM stage (II-IV vs. I), and NLR levels (high vs. low). After careful investigation, we determined that the relationship between TLS and TNM stage (OR = 0.71, 95% CI: 0.51-1.00, p < 0.05) and NLR level (OR = 0.33, 95% CI: 0.17–0.62, p < 0.001) was significant. TLS did not show any notable correlation with the patient’s age (OR = 1.11, 95% CI: 0.71–1.76, p = 0.64), gender (OR = 0.81, 95% CI: 0.61–1.08, p = 0.15), tumor classification (OR = 0.97, 95% CI: 0.73–1.30, p = 0.85), tumor size(OR = 0.97, 95% CI: 0.55–1.72, p = 0.92), or smoking status(OR = 1.01, 95% CI: 0.67–1.51, p = 0.97) (Fig. 3).

Table 3 Correlations of clinicopathological characteristics in patients with non-small cell lung cancer
Fig. 3
figure 3

Forest plot of the correlation between TLS and clinicopathological characteristics in patients with NSCLC. (A) age; (B) TNM stage; (C) gender; (D) tumor size; (E) smoke; (F) NLR; (G) histological type. TLS, tertiary lymphoid structure; OR, odds ratio; CI, confidence interval; TNM, Tumor Node Metastasis; NLR, Neutrophil-lymphocyte ratio

Sensitive analysis and publication bias

We conducted a sensitivity analysis by excluding the studies one by one. The HRs calculated from the combined results of the remaining studies in each analysis did not go beyond the expected range, as illustrated in Supplementary Fig. 2 and Supplementary Fig. 3. There is no significant difference between the revised overall estimate and the primary combined estimate, indicating that the meta-analysis is reliable. Begg’s and Egger’s tests are employed to identify any potential publication bias. The meta-analysis did not show any clear publication bias on TLS with respect to OS (Egger’s p = 0.369, Begg’s p = 0.308) and DFS/RFS (Egger’s p = 0.117, Begg’s p = 0.117).

Discussion

In the past few years, as researchers have delved deeper into the tumor microenvironment (TME) and the workings of tumor immunotherapy, TLS has emerged as a significant biological structure that hinders tumor growth by stimulating the activation of immune cells near the tumor [4, 26, 27]. Numerous research studies have investigated the significance of TLS in treating individuals with cancers, and the majority indicating that elevated TLS levels are a crucial indicator of a positive prognosis for various solid tumors [28, 29]. However, the prognostic value of TLS in NSCLC remains controversial. This meta-analysis integrated prognostic data and clinical characteristics of 1,451 NSCLC patients from 10 studies and conducted subgroup analysis. Following a thorough quantitative analysis of prognostic data, we determined that elevated levels of TLS were strongly associated with improved OS, DSS, and DFS/RFS. Additionally, high TLS levels were found to be closely linked to the tumor TNM stage and NLR. This meta-analysis represents the most up-to-date and extensive investigation regarding the correlation between TLS and prognosis, as well as relevant clinicopathological characteristics in individuals diagnosed with NSCLC.

Although only three studies in this meta-analysis focused on the relationship between TLS and patient outcomes in those who underwent immunotherapy before surgery, our findings indicated that individuals with increased TLS levels who received immunotherapy before surgery had a more favorable prognosis compared to those who underwent surgery alone [30]. TLS is an essential component of the tumor immune microenvironment (TIME) and includes T cells, B cells, fibroblast reticular cells (FRC) networks, high endothelial venules (HEV), and follicular dendritic cells (FDC) [4, 31, 32]. Within the TME, TLS serves as a site where immune cells can proliferate and interact. This area is primarily made up of an internal region of CD20+ B cells and a surrounding region of CD3+ T cells [33, 34]. Additionally, there is a significant presence of dendritic cells (DC) surrounding the immune cells, all of which congregate in this space to collectively suppress tumor growth. In this cluster of immune cells, DC displays the surface antigen of nearby tumor tissue to T cells via TLS [35, 36]. The activated T cells then produce memory helper T cells and effector memory cytotoxic cells to aid in the destruction of tumor cells through phagocytosis [37, 38]. Furthermore, this cluster supports the activation and growth of B cells, facilitating the development, activation, and growth of memory B cells and plasma cells [39, 40]. These immune cells further contribute to the body’s ability to eliminate tumor cells by generating antibodies. Tumor infiltrating lymphocytes (TILs) are lymphocytes isolated from tumor tissue. It plays a key role in the host antigen-specific tumor immune response [41], and the adoptive immunotherapy approach mediated by TILs has achieved good efficacy in a variety of solid tumors [42, 43]. It has been reported that TLS and TILs play similar roles in the anti-tumor process. However, the study we included found that although there was a certain relationship between the density of TLS and TILs, the joint increase of the two did not have a synergistic effect on the prognosis of the tumor, but were independent of each other [19, 20]. Moreover, Cottrel et al. found that the presence of TLS within the tumor area was consistently associated with cellular apoptosis in patients exhibiting a favorable response to preoperative immune checkpoint inhibitor therapy. Conversely, nonspecific collection of TILs unrelated to the treatment response was also observed [44]. This implies that TLS, rather than TILs, could serve as a more reliable indicator of the therapeutic efficacy for NSCLC patients. Hence, elevated levels of intratumoral TLS could serve as a significant prognostic indicator for NSCLC patients. This further validates the connection between TLS and the immune mechanisms within the tumor microenvironment, highlighting an important area for future investigation.

Currently, there is a lack of consistent criteria globally for choosing TLS detection techniques and determining threshold values, which poses a significant challenge to utilizing TLS as a key prognostic indicator [45]. To identify the most effective approach for assessing TLS, we carried out a subgroup analysis of the studies included. In this meta-analysis, there are variations in how TLS is detected and the cutoff methods used across different studies. According to our analysis results, the use of IHC staining to detect TLS and its grouping by negative or positive results both suggest that patients have a better prognosis. This is probably because these two techniques more accurately reflect the levels of TLS in the patient’s body. Therefore, IHC, along with categorizing TLS as negative or positive, could be potentially used together as a standard method for identifying and assessing TLS. However, due to the limited sample size in the studies included, additional research is required to gather more evidence before it can be established as a universal standard for evaluating.

Nevertheless, this meta-analysis has certain constraints. Primarily, most of the included studies were retrospective cohort studies conducted at a single center, potentially leading to biases such as cohort selection bias that could impact the reliability of the findings. Moreover, variations in the methods used to establish TLS cutoff values among the included studies could result in selection bias and diverse outcomes. Furthermore, certain studies lacked precise prognostic details, prompting us to utilize Engauge Digitizer software to estimate the survival statistics of select studies by analyzing the survival curve. This method may yield results that differ from the original data. Moreover, there are only a few studies that can be used for subgroup analysis, particularly within the immunotherapy subgroup. Merely three immunotherapy studies were incorporated, and the sample size was relatively small, suggesting potential inaccuracies in our assessment of immunotherapy [12, 13, 18]. Finally, our meta-analysis focused solely on the presence of TLS within the tumor itself, rather than its presence outside the tumor, which may not fully represent its impact on tumor prognosis. Given these constraints, it is essential to conduct numerous multi-center prospective studies to validate our findings before implementing them in clinical settings.

Conclusion

TLS plays a crucial role in the treatment of NSCLC. Elevated TLS levels are strongly related to positive survival outcomes such as OS, DSS, and DFS/RFS in NSCLC. Additionally, TLS expression levels are closely associated with certain clinicopathological factors of NSCLC patients. Therefore, TLS has the potential to serve as a biomarker for predicting the prognosis of NSCLC patients and may influence clinical treatment decisions. Nevertheless, further prospective studies are necessary to validate the prognostic significance of TLS in NSCLC patients before its clinical application.

Data availability

All data are generated from public data, which has been shown in the article. The data that support the findings of this study are available on request from the corresponding author.

Abbreviations

NSCLC:

Non-small cell lung cancer

TLS:

Tertiary lymphoid structure

HRs:

Hazard ratios

ORs:

Odds ratios

CIs:

Confidence intervals

OS:

Overall survival

DFS:

Disease-free survival

RFS:

Recurrence-free survival

DSS:

Disease-specific survival

TNM:

Tumor Node Metastasis

NLR:

Neutrophil-lymphocyte ratio

SLO:

Secondary lymphoid organ

MeSH:

Medical subject terms

NOS:

Newcastle-Ottawa Quality Assessment Scale

NCIT:

Neoadjuvant chemoimmunotherapy

IHC:

Immunohistochemical

H-E:

Hematoxylin and eosin

TME:

Tumor microenvironment

TIME:

Tumor immune microenvironment

FRC:

Fibroblast reticular cells

HEV:

High endothelial venules

FDC:

Follicular dendritic cells

DC:

Dendritic cells

TILs:

Tumor infiltrating lymthocytes

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83(5):584 – 94.

  3. de Biase MS, Massip F, Wei TT, Giorgi FM, Stark R, Stone A, et al. Smoking-associated gene expression alterations in nasal epithelium reveal immune impairment linked to lung cancer risk. Genome Med. 2024;16(1):54.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19(6):307–25.

    Article  PubMed  Google Scholar 

  5. Fleig S, Kapanadze T, Bernier-Latmani J, Lill JK, Wyss T, Gamrekelashvili J, et al. Loss of vascular endothelial notch signaling promotes spontaneous formation of tertiary lymphoid structures. Nat Commun. 2022;13(1):2022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sato Y, Silina K, van den Broek M, Hirahara K, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol. 2023;19(8):525–37.

    Article  PubMed  Google Scholar 

  7. Pfannstiel C, Strissel PL, Chiappinelli KB, Sikic D, Wach S, Wirtz RM, et al. The Tumor Immune Microenvironment drives a prognostic relevance that correlates with bladder Cancer subtypes. Cancer Immunol Res. 2019;7(6):923–38.

    Article  CAS  PubMed  Google Scholar 

  8. Munoz-Erazo L, Rhodes JL, Marion VC, Kemp RA. Tertiary lymphoid structures in cancer - considerations for patient prognosis. Cell Mol Immunol. 2020;17(6):570–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang Q, Sun K, Liu R, Song Y, Lv Y, Bi P, et al. Single-cell transcriptome sequencing of B-cell heterogeneity and tertiary lymphoid structure predicts breast cancer prognosis and neoadjuvant therapy efficacy. Clin Transl Med. 2023;13(8):e1346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mao Y, Wang X, Xi L, Dong M, Song P, Miao J, et al. Prediction values of tertiary lymphoid structures in the prognosis of patients with left- and right-sided colon cancer: a multicenter propensity score-matched study. Int J Surg. 2023;109(8):2344–58.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Brunet M, Crombé A, Cousin S, Vanhersecke L, Le Loarer F, Bessede A, et al. Mature tertiary lymphoid structure is a specific biomarker of cancer immunotherapy and does not predict outcome to chemotherapy in non-small-cell lung cancer. Ann Oncol. 2022;33(10):1084–5.

    Article  CAS  PubMed  Google Scholar 

  12. Liu Y, Xiong L, Chen Y, Cai R, Xu X, Wang T, et al. Complete pathological remission and tertiary lymphoid structures are associated with the efficacy of resectable NSCLC receiving neoadjuvant chemoimmunotherapy: a double-center retrospective study. Hum Vaccin Immunother. 2023;19(3):2285902.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Xu F, Zhu H, Dong Y, Li L, Liu N, Yuan S. Combined inflammatory parameters and tertiary lymphoid structure predict prognosis in patients with resectable non-small cell lung cancer treated with neoadjuvant chemoimmunotherapy. Front Immunol. 2023;14:1244256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, Rennie D, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis of Observational studies in Epidemiology (MOOSE) group. JAMA. 2000;283(15):2008–12.

    Article  CAS  PubMed  Google Scholar 

  16. Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol. 2010;25(9):603–5.

    Article  PubMed  Google Scholar 

  17. Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time-to-event data into meta-analysis. Trials. 2007;8:16.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Sun X, Liu W, Sun L, Mo H, Feng Y, Wu X et al. Maturation and abundance of tertiary lymphoid structures are associated with the efficacy of neoadjuvant chemoimmunotherapy in resectable non-small cell lung cancer. J Immunother Cancer. 2022;10(11).

  19. Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 2008;26(27):4410–7.

    Article  CAS  PubMed  Google Scholar 

  20. Giatromanolaki A, Chatzipantelis P, Contrafouris CA, Koukourakis MI. Tertiary lymphoid structures, Immune Response, and Prognostic Relevance in Non-small Cell Lung Cancer. Cancer Invest. 2023;41(1):48–57.

    Article  CAS  PubMed  Google Scholar 

  21. Tang J, Ramis-Cabrer D, Curull V, Wang X, Mateu-Jiménez M, Pijuan L et al. B cells and tertiary lymphoid structures influence survival in Lung Cancer patients with resectable tumors. Cancers (Basel). 2020;12(9).

  22. Tamiya Y, Nakai T, Suzuki A, Mimaki S, Tsuchihara K, Sato K, et al. The impact of tertiary lymphoid structures on clinicopathological, genetic and gene expression characteristics in lung adenocarcinoma. Lung Cancer. 2022;174:125–32.

    Article  PubMed  Google Scholar 

  23. Fukuhara M, Muto S, Inomata S, Yamaguchi H, Mine H, Takagi H, et al. The clinical significance of tertiary lymphoid structure and its relationship with peripheral blood characteristics in patients with surgically resected non-small cell lung cancer: a single-center, retrospective study. Cancer Immunol Immunother. 2022;71(5):1129–37.

    Article  PubMed  Google Scholar 

  24. Yang H, Liu Y, Xu Q, Jiang L, Wen Q, Lin S, et al. The relationship between Immune Microenvironment and Prognosis of Primary Lung Cancer. Anti-Tumor Pharm. 2020;10(3):334–8.

    Google Scholar 

  25. Rakaee M, Kilvaer TK, Jamaly S, Berg T, Paulsen EE, Berglund M, et al. Tertiary lymphoid structure score: a promising approach to refine the TNM staging in resected non-small cell lung cancer. Br J Cancer. 2021;124(10):1680–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Fridman WH, Meylan M, Petitprez F, Sun CM, Italiano A, Sautès-Fridman C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022;19(7):441–57.

    Article  CAS  PubMed  Google Scholar 

  27. Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577(7791):561–5.

    Article  CAS  PubMed  Google Scholar 

  28. Ling Y, Zhong J, Weng Z, Lin G, Liu C, Pan C, et al. The prognostic value and molecular properties of tertiary lymphoid structures in oesophageal squamous cell carcinoma. Clin Transl Med. 2022;12(10):e1074.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang Q, Wu S. Tertiary lymphoid structures are critical for cancer prognosis and therapeutic response. Front Immunol. 2022;13:1063711.

    Article  CAS  PubMed  Google Scholar 

  30. Lv B, Wang Y, Ma D, Cheng W, Liu J, Yong T, et al. Immunotherapy: reshape the Tumor Immune Microenvironment. Front Immunol. 2022;13:844142.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Bejarano L, Jordāo MJC, Joyce JA. Therapeutic targeting of the Tumor Microenvironment. Cancer Discov. 2021;11(4):933–59.

    Article  CAS  PubMed  Google Scholar 

  32. Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375(6576):eabf9419.

    Article  CAS  PubMed  Google Scholar 

  33. Moyron-Quiroz JE, Rangel-Moreno J, Kusser K, Hartson L, Sprague F, Goodrich S, et al. Role of inducible bronchus associated lymphoid tissue (iBALT) in respiratory immunity. Nat Med. 2004;10(9):927–34.

    Article  CAS  PubMed  Google Scholar 

  34. Moyron-Quiroz JE, Rangel-Moreno J, Hartson L, Kusser K, Tighe MP, Klonowski KD, et al. Persistence and responsiveness of immunologic memory in the absence of secondary lymphoid organs. Immunity. 2006;25(4):643–54.

    Article  CAS  PubMed  Google Scholar 

  35. Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF, Sancho D. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24.

    Article  CAS  PubMed  Google Scholar 

  36. Del Prete A, Salvi V, Soriani A, Laffranchi M, Sozio F, Bosisio D, et al. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol Immunol. 2023;20(5):432–47.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Nasr IW, Reel M, Oberbarnscheidt MH, Mounzer RH, Baddoura FK, Ruddle NH, et al. Tertiary lymphoid tissues generate effector and memory T cells that lead to allograft rejection. Am J Transpl. 2007;7(5):1071–9.

    Article  CAS  Google Scholar 

  38. Yin X, Chen S, Eisenbarth SC. Dendritic cell regulation of T Helper cells. Annu Rev Immunol. 2021;39:759–90.

    Article  CAS  PubMed  Google Scholar 

  39. Xia J, Xie Z, Niu G, Lu Z, Wang Z, Xing Y, et al. Single-cell landscape and clinical outcomes of infiltrating B cells in colorectal cancer. Immunology. 2023;168(1):135–51.

    Article  CAS  PubMed  Google Scholar 

  40. Meylan M, Petitprez F, Becht E, Bougoüin A, Pupier G, Calvez A, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity. 2022;55(3):527–e415.

    Article  CAS  PubMed  Google Scholar 

  41. Sidaway P. Efficacy of TILs confirmed. Nat Rev Clin Oncol. 2023;20(2):64.

    Article  PubMed  Google Scholar 

  42. Monberg TJ, Borch TH, Svane IM, Donia M. TIL therapy: facts and hopes. Clin Cancer Res. 2023;29(17):3275–83.

    Article  CAS  PubMed  Google Scholar 

  43. Kumar A, Watkins R, Vilgelm AE. Cell therapy with TILs: training and taming T cells to Fight Cancer. Front Immunol. 2021;12:690499.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cottrell TR, Thompson ED, Forde PM, Stein JE, Duffield AS, Anagnostou V, et al. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann Oncol. 2018;29(8):1853–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Barmpoutis P, Di Capite M, Kayhanian H, Waddingham W, Alexander DC, Jansen M, et al. Tertiary lymphoid structures (TLS) identification and density assessment on H&E-stained digital slides of lung cancer. PLoS ONE. 2021;16(9):e0256907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was funded by National Key Research and Development Program (2021YFC2500905, 2021YFC2500904, and 2021YFC2500900), Natural Science Foundation of Shandong Province (ZR2021LSW006), and and the Taishan Scholar Program of Shandong Province (ts201712087).

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: Luyuan Ma and Hui Tian; Administrative support: Hui Tian and Rongyang Li; Provision of study materials or patients: Luyuan Ma, Rongyang Li, and Xiaomeng Liu; Collection and assembly of data: Luyuan Ma, Rongyang Li, Wenhao Yu, Yi Shen; Data analysis and interpretation: Luyuan Ma, Rongyang Li, Xiaomeng Liu, Zhanpeng Tang. All authors contributed to the article and approved the submitted version.

Corresponding author

Correspondence to Hui Tian.

Ethics declarations

Ethics approval and consent to participate

Ethical approval was not necessary, as this study was a “Systematic Review and Meta-analysis.” There are no individual person’s data and presentations of case reports involved in this article.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Supplementary Material 3: Supplementary Figure 1.

Subgroup analysis of the correlation between TLS in non-small cell lung cancer patients based on (A) the type of tumor treatment, and (B) assessment of TLS cutoff values, and (C) TLS detection methods. TLS, tertiary lymphoid structures; NCIT, neoadjuvant chemoimmunotherapy; IHC, immunohistochemical; H-E, Hematoxylin and eosin; OR, odds ratio; CI, confidence interval

Supplementary Material 4: Supplementary Figure 2.

Sensitivity analysis of (A) disease-free survival/recurrence-free survival, (B) overall survival,(C) disease-specific survival

Supplementary Material 5: Supplementary Figure 3.

Sensitivity analysis of (A) age; (B) TNM stage; (C) gender; (D) tumor size; (E) smoke; (F) NLR; (G) histological type. TNM, Tumor Node Metastasis; NLR, Neutrophil-lymphocyte ratio

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, L., Li, R., Liu, X. et al. Prognostic and clinicopathological significance of tertiary lymphoid structure in non-small cell lung cancer: a systematic review and meta-analysis. BMC Cancer 24, 815 (2024). https://doi.org/10.1186/s12885-024-12587-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12587-x

Keywords