Skip to main content

MINI REVIEW article

Front. Immunol., 03 October 2016
Sec. Inflammation
This article is part of the Research Topic Tertiary Lymphoid Organs (TLOs): powerhouses of disease immunity View all 22 articles

Tertiary Lymphoid Structures in Cancers: Prognostic Value, Regulation, and Manipulation for Therapeutic Intervention

\r\nCatherine Sauts-Fridman,,Catherine Sautès-Fridman1,2,3Myriam Lawand,,Myriam Lawand1,2,3Nicolas A. Giraldo,,Nicolas A. Giraldo1,2,3Hlne Kaplon,,Hélène Kaplon1,2,3Claire Germain,,Claire Germain1,2,3Wolf Herman Fridman,,Wolf Herman Fridman1,2,3Marie-Caroline Dieu-Nosjean,,*\r\nMarie-Caroline Dieu-Nosjean1,2,3*
  • 1INSERM, UMR_S 1138, Team “Cancer, Immune Control and Escape”, Cordeliers Research Center, Paris, France
  • 2UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France
  • 3UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France

Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates that reflect lymphoid neogenesis occurring in tissues at sites of inflammation. They are detected in tumors where they orchestrate local and systemic anti-tumor responses. A correlation has been found between high densities of TLS and prolonged patient’s survival in more than 10 different types of cancer. TLS can be regulated by the same set of chemokines and cytokines that orchestrate lymphoid organogenesis and by regulatory T cells. Thus, TLS offer a series of putative new targets that could be used to develop therapies aiming to increase the anti-tumor immune response.

Introduction

Tertiary lymphoid structures (TLS) are transient ectopic lymphoid organizations that develop after birth in non-lymphoid tissues, in situations of chronic inflammation. They display an overall organization similar to that observed in canonical secondary lymphoid organs (SLOs), such as lymph nodes (LNs), with a T cell-rich area characterized by a T cell and mature DC-Lamp+ dendritic cell (DCs) cluster, a B-cell-rich area composed of a mantle of naïve B cells surrounding an active germinal center (GC) (13), the presence of high endothelial venules (HEVs), a particular type of blood vessels expressing peripheral node addressins (PNAd) and specialized in the extravasation of circulating immune cells, and the secretion of chemokines (CCL19, CCL21, CXCL10, CXCL12, and CXCL13) that are crucial for lymphocyte recruitment and entry into the LN (48). TLS have been detected in the tumor invasive margin and/or in the stroma of most cancers and their densities correlate with a favorable clinical outcome for the patients (Table 1). A series of studies performed by our group in non-small-cell lung cancer (NSCLC) demonstrated that TLS are important sites for the initiation and/or maintenance of the local and systemic T- and B-cell responses against tumors, in accordance with a specific signature of genes related to T and B cell lineage, chemotaxis, Th1 polarization, lymphocyte activation, and effector function associated with TLS presence (Table 2). They represent a privileged area for the recruitment of lymphocytes into tumors and the generation of central-memory T and B cells that circulate and limit cancer progression (5, 9, 10).

TABLE 1
www.frontiersin.org

Table 1. Prognostic value of TLS in primary and metastatic tumors.

TABLE 2
www.frontiersin.org

Table 2. Expression of genes associated with TLS presence in human cancers.

In this mini review, we summarize the available data in the literature regarding the prognostic value of TLS in human cancers, and discuss how these structures are controlled and could be manipulated in order to increase anti-tumor immune responses.

TLS and Prognosis in Cancers

In recent years, numerous publications have assessed the prognosis associated with the presence of TLS in different types of tumors. Several strategies for their quantification have been used. Historically, the first method to measure the densities of TLS was the quantification of mature DCs (DC-Lamp+) within CD3+ T cell aggregates (1, 20). Although relatively challenging due to the relative low number of DC-Lamp+ DCs in some tumors (as compared to other immune populations), our group has described it as the most accurate marker for quantifying TLS (28, 29). Up-to-date, eight publications have found a positive association between increased densities of DC-Lamp+ DCs and prognosis, in several types of tumors, including NSCLC (1, 2, 9), melanoma (20), renal cell carcinoma [RCC (24)], breast (11), and colorectal cancer (15) (Table 1 and Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1. Prognostic value of TLS-associated biomarkers in primary and metastatic cancers. The number of publications studying the impact of mature DCs, TLS-related gene signatures, B-cell aggregates, T cell aggregates, or H&E with regard to prognosis in human cancers is represented (12 cancer types have been included). Blue, orange, and red circles represent an association with good, none, and poor prognosis, respectively. The diameter of the circles represents the total number of tumors (n) that have been analyzed on these studies.

The analysis of expression levels of TLS-related genes gives the possibility to rapidly assess the prognostic impact of these immune aggregates in large retrospective cohorts of tumors. So far, six studies have evaluated the prognostic impact of increased expression of TLS-related genes in cancer. Despite heterogeneity in the TLS-signatures, a significant correlation with good prognosis has been found in melanoma (21), colorectal (3, 18), and gastric (19) cancers (Table 1). Interestingly, TLS found in inflammatory zones from hepatocellular carcinoma (HCC) correlate with increased risk for late recurrence and a trend toward decreased overall survival after HCC resection. This result could reflect an unexpected role for TLS, serving as niche for HCC progenitor cells via local production of Lymphotoxin (LT)-β (25, 30).

Another approach that has been used to estimate the densities of TLS in cancers is the quantification of B-cell aggregates by immunohistochemistry (IHC) (CD20+ B-cell aggregates or islets). The majority of publications measuring CD20+ aggregates (four out of five), accounting for more than 349 analyzed tumors, has determined that increased densities of this population correlate with good prognosis in several neoplasias, such as NSCLC (2), colorectal cancer liver metastasis (27), gastric (19), and oral (22) cancer (Table 1 and Figure 1). Most of the studies quantifying the CD3+ T cell aggregates and immune-cell aggregates (after hematoxylin counterstaining) have also found a positive impact on patient’s prognosis. However, high numbers of B cell or T cell aggregates were found to have no impact on prognosis in biliary tract cancer and in stage III colorectal cancer, respectively. Further studies are needed to investigate whether it reflects that cell aggregates counting is not an accurate method to quantify TLS, or a functional impairment of TLS in these two cancer types (Table 1 and Figure 1).

Overall, despite the heterogeneity of methods used for quantifying TLS, most of the studies have consistently found a correlation between high densities of TLS and prolonged patient’s survival in more than 10 different types of cancer (Table 1). Further efforts should be made to optimize TLS-quantifying methods. Indeed the use of multicolor IHC will facilitate their characterization, by allowing the simultaneous detection of all major cell types and providing an extensive analysis of their cellular complexity.

TLS Neogenesis

The cellular composition and spatial organization of TLS share many similarities with those of SLO. Indeed, an increasing number of studies performed in a large variety of inflammatory disorders, in mice and in humans, suggest that their formation and regulation involve the same set of chemokines than those acting in lymphoid organogenesis.

Positive Regulators

Lymphotoxin, CCL21, and CXCL13 were shown to play a major role during TLS neogenesis, and are related to TLS presence in human tumors (Table 2). In a mouse model of atherosclerosis, the activation of LTβR+ medial smooth muscle cells in the abdominal aorta by LT produced by CD11c+ CD68+ Ly6Clo monocytes leads to the expression of CCL19, CCL21, CXCL13, and CXCL16 chemokines, which in turn trigger the recruitment of lymphocytes to the adventitia and the development of TLS (31). The same observation was made by Thaunat et al. in a rat model of chronic allograft rejection, in which M1-macrophages behaved as LTi cells in diseased arteries by expressing high levels of LTα and TNF-α (32). In human NSCLC, a TLS-related gene signature was identified, including CCL19, CCL21, IL-16, and CXCL13 (5) (Table 2). Interestingly, Matsuda et al. recently suggested in a mouse intrapulmonary tracheal transplant model that lymphoid neogenesis was dependent on spleen tyrosine kinase (Syk)-signaling. Decreased expression of CXCL12, CXCL13, and VEGF-α, lower B-cell recruitment into allograft, and smaller lymphoid aggregate area were observed in Syk-deficient recipient mice as compared to controls (33).

The generation of HEVs is also a critical step in TLS neogenesis. HEV endothelial cells express LTβR, and the continuous engagement of LTβR on HEVs by LT+ CD11c+ DCs is critical for the induction and maintenance of the mature HEV phenotype required for the extravasation of blood lymphocyte into LNs (3437). In addition, CD11c+ DCs can be sources of proangiogenic factors, such as VEGF, favoring the development of HEVs, and ultimately lymphocyte entry into LN (3841). Consistently, LTβ expression correlates with that of HEV-associated chemokines in human breast cancer, and DC-Lamp+ DC density correlates with HEV density, lymphocyte infiltration, and favorable clinical outcome (11). Other cell types were shown to favor the development of HEV. For instance, ectopic expression of CCL21 in the thyroid gives rise first to the recruitment of CD3+ CD4+ T cells followed by DC, and this DC-T cross-talk is required for the local development of both TLS and mature HEV (42). Tumor-infiltrating CD8+ T cells and NK cells were also shown to drive the de novo development of PNAd+ TNFRI+ CCL21+ HEV-like blood vessels through the production of LT and IFN-γ (43).

Th17 cells share many developmental and effector markers with LTi cells, including the nuclear hormone receptor retinoic acid-related orphan receptor γt (RORγt), which promotes not only the production of IL-17 and IL-22 by Th17 cells, LTi cells, and other RORγt+ innate lymphoid cells (ILCs), but also cell membrane expression of LT [reviewed in Ref. (44)]. In mice lungs, the formation of TLS [called here induced-bronchus-associated lymphoid tissues (i-BALT)] following LPS sensitization was dependent of IL-17 production by T cells, including Th17 and γδ T cells (45). This observation was also observed in a mouse experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (46). Similarly, IL-17α-deficient mice exposed to cigarette smoke displayed decreased number of ectopic lymphoid follicles and decreased expression of CXCL12 as compared to wild-type mice in a model of chronic obstructive lung disease (47). It has also been suggested that Th17 cells, and IL-17 and IL-21 secretion by these cells can promote TLS neogenesis within human renal grafts, and are associated with the presence of active GC B cells and fast chronic rejection (48).

Other inflammatory cytokines also seem to promote TLS neogenesis. In rheumatoid arthritis (RA), high protein levels of IL-23 and IL-17F were detected in the synovial fluid of patients displaying ectopic lymphoid follicles, and a positive correlation was observed between CD21L mRNA (as a TLS marker) and IL-23 but also IL-17F, IL-21, and IL-22 mRNAs (49). IL-22 was also proposed to favor TLS induction (50). In a mouse model of virus-induced autoantibody formation in the salivary glands, it was shown that the ligation of IL-22R expressed by epithelial cells and fibroblasts leads to CXCL12 and CXCL13 production, allowing B-cell recruitment and TLS organization. In that case, IL-22 was mainly produced by γδ T cells and to a lesser extent by ILCs and NK cells during the early phase post-infection, and then by αβ T cells later after infection.

Negative Regulators

On the opposite, IL-27, a cytokine known to inhibit effector Th17 responses was recently suggested to negatively regulate the development of ectopic lymphoid-like structures in the synovial tissues of RA patients. While patients having a high density of TLS displayed high synovial levels of IL-17 and IL-21, high levels of IL-27 were observed in patients devoid of any TLS, and IL-27 expression was inversely correlated with CD3+ and CD20+ infiltrates and with synovitis. This observation was confirmed in a mouse model of RA (51).

Among the immune cells infiltrating tumors are regulatory T cells (Tregs), which have been considered in many reports as a marker of poor prognosis in cancer (52, 53). Tregs have been reported to negatively interfere with BALT development. Indeed in CCR7-deficient mice, BALTs developed spontaneously in the absence of infection, an event that is directly reverted by the adoptive transfer of wild-type Tregs but not CCR7−/− Tregs (54). In human breast cancer, Tregs were detected in lymphoid aggregates surrounding tumor nests, and their presence was linked with the poor clinical outcome of patients (55). In mice bearing breast tumors, Treg depletion led to an increased density of HEV within the tumor, facilitated T cell recruitment from the blood, and ultimately induced tumor destruction (56). This observation is in accordance with a human study showing that HEVhigh breast tumors correlated with a high LT-β expression, a high density of tumor-infiltrating mature DC, and a decreased FoxP3+/CD3+ T cell ratio (11).

More recently, a new mechanism involving regulation of TLS formation by Tregs was found, by dampening neutrophilic inflammation (57). The presence of neutrophils seemed to be critical for the neogenesis and the humoral immune function of i-BALT by enhancing B-cell activation and survival, Ig class switching to IgA as well as plasma cell survival (57).

Regulatory T cells have been shown to dampen the effector T cell response promoted within tumor-associated TLS. Treg depletion causes immune-mediated tumor destruction associated with an increased expression of co-stimulatory ligands by DCs and proliferation of T cells in a murine model of lung adenocarcinoma (58). Further studies should be carried out to analyze the prognostic importance of Tregs and their immunosuppressive potential in cancer patients according to their localization.

Altogether, TLS neogenesis and lymphoid organogenesis share many common mechanisms. On the one hand, the production of inflammatory cytokines (LT, IL-17, IL-22, and IL-23) and lymphoid chemokines (CCL21, CXCL12, and CXCL13), HEV development as well as the activation of DCs, B, and effector cells seem to be crucial events leading to TLS neogenesis under inflammatory conditions, such as cancers. On the other hand, the presence of Tregs appears to negatively impact TLS formation and TLS-associated T cell responses.

Manipulation of TLS for a Therapeutic Intervention in Cancer

A series of studies suggest that TLS are sites for generation and maintenance of adaptive anti-tumor responses (10). Therefore, TLS induction could be used as a therapeutic intervention for a better tumor control and prolonged survival of cancer patients. Since LN and TLS share many similarities in terms of cellular composition and organization, deciphering the mechanisms of lymphoid organogenesis enables to first highlight some putative key molecules that can support TLS neogenesis.

Targeting Molecules Involved in Lymphoid Organogenesis

The key cross-talk between LTi cells and lymphoid tissue organizer cells (LTo cells that are cells of mesenchymal origin) occurring during LN development involves several molecules along with RANK and its ligand, which lead to LTβR signaling (59). Therefore, targeting RANK/LT pathway may modulate TLS development through the activation of LTo cells. Currently, antagonists of LTα (Pateclizumab NCT01225393), LTβR (Baminercept, NCT01552681) and RANK signaling (NCT01973569) are under investigation in several inflammatory situations. A special attention should be made in cancer setting where these antagonists might block TLS formation and, hence, reduce survival. The use of agonists might rather present a benefit to cancer patients but no drugs have been developed so far.

Activation of LTβR signaling pathway in LTo cells induces VCAM-1 and ICAM-1 upregulation, and ultimately leukocyte infiltration (60). Because both molecules are known to be induced by inflammation, an ICAM-1 antagonist called Alicaforsen has been tested in autoimmune diseases (NCT00048113, NCT00063830). We can speculate that the development of VCAM-1/ICAM-1 agonists would promote LTi-like cells-LTo clusterings and improve the leukocyte recruitment in order to generate cancer-associated TLS.

IL-7 receptor (CD127) signaling has been reported as a key pathway for TLS neogenesis (61). IL-7 is not only crucial for the survival and proliferation of LTi cells but also for GC formation and Tfh differentiation (62). To date, only one pharmacologic agent (IL-7R) is under investigation in NOD mice to deplete autoreactive T cells and to regulate pro-inflammatory mediators (63).

Altogether, as a counterpart of autoimmune diseases, development of agonist molecules targeting lymphoid organogenesis might be a promising strategy for the initiation and the maintenance of TLS in cancers.

Modulation of Chemokine and Cytokine Networks

Lymphoid chemokines represent a good therapeutic target for the modulation of TLS (Table 2). The CCL19–CCL21/CCR7 and CXCL13/CXCR5 couples are induced after LT-βR signaling during lymphoid genesis (60). They are overexpressed in TLS of melanoma (21), colorectal (3), and lung (5) cancer patients. Using lymphoid chemokines or their agonists could be a promising strategy to induce TLS neogenesis in cancers. For example, CCL21 has been shown to attract circulating naïve T cells and DCs in tumors, and contribute to the control of tumor growth (6466). A Phase I clinical trial is currently under investigation in NSCLC patients receiving intra-tumoral injections of CCL21-transduced autologous DCs (NCT00601094, NCT01574222). It is tempting to speculate that this vaccine therapy would boost TLS formation in tumors associated with an influx of lymphocytes, an effective anti-tumor immune response, and a reduction of tumor burden.

IL-21, which is mainly secreted by Th17 cells and neutrophils, represents also an interesting molecular target. First, this cytokine has been shown to promote TLS neogenesis in lungs after acute LPS exposure and IL-21−/− mice exhibit fewer TLS in allografts than the control group (57). Second, IL-21 can enhance B and plasma cell survival as well as B-cell-dependent immunity, and induce conventional T cells to become refractory to Treg immunosuppression (48, 57, 67). Even if IL-21 can block IL-2 production with deleterious consequences in terms of Treg differentiation, IL-21 can substitute for IL-2 as a T cell growth factor (68). Recombinant IL-21 is currently tested in many clinical trials, alone or in combination with chemotherapy, therapeutic antibodies or tyrosine kinase inhibitors (e.g., NCT00617253, NCT00389285, NCT00095108, NCT01629758, NCT00336986, and NCT01489059). Altogether, it is likely that IL-21 could promote a robust anti-tumor immunity in a TLS-dependent manner.

Conclusion and Perspectives

By facilitating the direct entry of CCR7+ naïve T cells and CXCR5+ B cells into tumors through HEVs, TLS allow T cells to differentiate into effector cells upon contact with mature DCs and B cells to form GC, protected from the immunosuppressive milieu of the tumor microenvironment. Therefore, TLS represent sites for the induction and maintenance of the local and systemic anti-tumor responses, which confer long-term protection against metastasis and, hence, correlate with good prognosis for the patients. Indeed, therapies aiming to increase TLS formation may allow generating anti-tumor responses directly in situ and would be beneficial in patients with high mutational load. TLS may also constitute biomarkers of anti-tumor response in patients undergoing immunotherapies. Thus, TLS induction was observed in cervical cancer patients vaccinated with HPV DNA (69) or with G-VAX (70), and one may speculate that TLS signature could be used to evidence response to therapies that unlock the adaptive immune responses.

Author Contributions

ML, NG, HK, CG, and CSF wrote and revised the paper. WF and MCDN revised the paper.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank all members of the Laboratory “Cancer, Immune control and Escape,” pathologists and clinicians who have participated in these studies, for their help and valuable discussions, as well as cancer patients.

Funding

This work was supported by Institut National de la Santé et de la Recherche Médicale (INSERM), University Paris Descartes, University Pierre et Marie Curie, the Institut National du Cancer (2009-1-PLBIO07-INSERM6-, 2010-1-PLBIO03-INSERM 6-1, 2011-1-PLBIO-06-INSERM 6-1, PLBIO09-088-IDF-KROEMER), CARPEM (CAncer Research for PErsonalized Medicine), the Labex Immuno-Oncology (LAXE62_9UMS872 FRIDMAN), foundation ARC pour la recherche sur le cancer (SL220110603483), Ligue Nationale contre le Cancer (GB/MA/CD/EP-12003), and MedImmune (Gaithersburg, USA, n°11796A10). HK was supported by a grant from La Ligue contre le Cancer.

References

1. 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. doi:10.1200/JCO.2007.15.0284

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, et al. 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 (2014) 189(7):832–44. doi:10.1164/rccm.201309-1611OC

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Coppola D, Nebozhyn M, Khalil F, Dai H, Yeatman T, Loboda A, et al. Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am J Pathol (2011) 179(1):37–45. doi:10.1016/j.ajpath.2011.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Cipponi A, Mercier M, Seremet T, Baurain JF, Théate I, van den Oord J, et al. Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res (2012) 72(16):3997–4007. doi:10.1158/0008-5472.CAN-12-1377

PubMed Abstract | CrossRef Full Text | Google Scholar

5. de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M, et al. Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res (2011) 71(20):6391–9. doi:10.1158/0008-5472.CAN-11-0952

CrossRef Full Text | Google Scholar

6. Martinet L, Garrido I, Girard JP. Tumor high endothelial venules (HEVs) predict lymphocyte infiltration and favorable prognosis in breast cancer. Oncoimmunology (2012) 1(5):789–90. doi:10.4161/onci.19787

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Martinet L, Le Guellec S, Filleron T, Lamant L, Meyer N, Rochaix P, et al. High endothelial venules (HEVs) in human melanoma lesions: major gateways for tumor-infiltrating lymphocytes. Oncoimmunology (2012) 1(6):829–39. doi:10.4161/onci.20492

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Martinet L, Garrido I, Filleron T, Le Guellec S, Bellard E, Fournie JJ, et al. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res (2011) 71(17):5678–87. doi:10.1158/0008-5472.CAN-11-0431

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, 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(3):705–15. doi:10.1158/0008-5472.CAN-13-1342

CrossRef Full Text | Google Scholar

10. Hosmalin A, Sautès-Fridman C, Fougereau M, Yssel H, Fischer A. 50(th) Anniversary of the French Society for Immunology (SFI). Eur J Immunol (2016) 46(7):1545–7. doi:10.1002/eji.201670073

CrossRef Full Text | Google Scholar

11. 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 β-producing dendritic cells in human breast cancer. J Immunol (2013) 191(4):2001–8. doi:10.4049/jimmunol.1300872

CrossRef Full Text | Google Scholar

12. Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest (2013) 123(7):2873–92. doi:10.1172/JCI67428

CrossRef Full Text | Google Scholar

13. Lee HJ, Park IA, Song IH, Shin SJ, Kim JY, Yu JH, et al. Tertiary lymphoid structures: prognostic significance and relationship with tumour-infiltrating lymphocytes in triple-negative breast cancer. J Clin Pathol (2016) 69(5):422–30. doi:10.1136/jclinpath-2015-203089

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Väyrynen JP, Sajanti SA, Klintrup K, Mäkelä J, Herzig KH, Karttunen TJ, et al. Characteristics and significance of colorectal cancer associated lymphoid reaction. Int J Cancer (2014) 134(9):2126–35. doi:10.1002/ijc.28533

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Remark R, Alifano M, Cremer I, Lupo A, Dieu-Nosjean MC, Riquet M, et al. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin. Clin Cancer Res (2013) 19(15):4079–91. doi:10.1158/1078-0432.CCR-12-3847

PubMed Abstract | CrossRef Full Text | Google Scholar

16. McMullen TPW, Lai R, Dabbagh L, Wallace TM, de Gara CJ. Survival in rectal cancer is predicted by T cell infiltration of tumour-associated lymphoid nodules. Clin Exp Immunol (2010) 161(1):81–8. doi:10.1111/j.1365-2249.2010.04147.x

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Di Caro G, Bergomas F, Grizzi F, Doni A, Bianchi P, Malesci A, et al. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin Cancer Res (2014) 20(8):2147–58. doi:10.1158/1078-0432.CCR-13-2590

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity (2013) 39(4):782–95. doi:10.1016/j.immuni.2013.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Hennequin A, Derangère V, Boidot R, Apetoh L, Vincent J, Orry D, et al. Tumor infiltration by Tbet + effector T cells and CD20 + B cells is associated with survival in gastric cancer patients. Oncoimmunology (2016) 5(2):e1054598. doi:10.1080/2162402X.2015.1054598

CrossRef Full Text | Google Scholar

20. Ladányi A, Kiss J, Somlai B, Gilde K, Fejos Z, Mohos A, et al. 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 (2007) 56(9):1459–69. doi:10.1007/s00262-007-0286-3

CrossRef Full Text | Google Scholar

21. Messina JL, Fenstermacher DA, Eschrich S, Qu X, Berglund AE, Lloyd MC, et al. 12-chemokine gene signature identifies lymph node-like structures in melanoma: potential for patient selection for immunotherapy? Sci Rep (2012) 2:765. doi:10.1038/srep00765

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Wirsing AM, Rikardsen OG, Steigen SE, Uhlin-Hansen L, Hadler-Olsen E. Characterisation and prognostic value of tertiary lymphoid structures in oral squamous cell carcinoma. BMC Clin Pathol (2014) 14:38. doi:10.1186/1472-6890-14-38

PubMed Abstract | CrossRef Full Text | Google Scholar

23. 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(11):1782–90. doi:10.1038/bjc.2015.145

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Giraldo NA, Becht E, Pagès F, Skliris G, Verkarre V, Vano Y, et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin Cancer Res (2015) 21(13):3031–40. doi:10.1158/1078-0432.CCR-14-2926

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol (2015) 16(12):1235–44. doi:10.1038/ni.3290

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Goeppert B, Frauenschuh L, Zucknick M, Stenzinger A, Andrulis M, Klauschen F, et al. Prognostic impact of tumour-infiltrating immune cells on biliary tract cancer. Br J Cancer (2013) 109(10):2665–74. doi:10.1038/bjc.2013.610

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Meshcheryakova A, Tamandl D, Bajna E, Stift J, Mittlboeck M, Svoboda M, et al. B cells and ectopic follicular structures: novel players in anti-tumor programming with prognostic power for patients with metastatic colorectal cancer. PLoS One (2014) 9(6):e99008. doi:10.1371/journal.pone.0099008

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Dieu-Nosjean MC, Giraldo NA, Kaplon H, Germain C, Fridman WH, Sautès-Fridman C. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev (2016) 271(1):260–75. doi:10.1111/imr.12405

CrossRef Full Text | Google Scholar

29. Dieu-Nosjean MC, Goc J, Giraldo NA, Sautès-Fridman C, Fridman WH. Tertiary lymphoid structures in cancer and beyond. Trends Immunol (2014) 35(11):571–80. doi:10.1016/j.it.2014.09.006

CrossRef Full Text | Google Scholar

30. Sautès-Fridman C, Fridman WH. TLS in tumors: what lies within. Trends Immunol (2016) 37(1):1–2. doi:10.1016/j.it.2015.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Gräbner R, Lötzer K, Döpping S, Hildner M, Radke D, Beer M, et al. Lymphotoxin beta receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE-/- Mice. J Exp Med (2009) 206(1):233–48. doi:10.1084/jem.20080752

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Thaunat O, Field AC, Dai J, Louedec L, Patey N, Bloch MF, et al. Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response. Proc Natl Acad Sci U S A (2005) 102(41):14723–8. doi:10.1073/pnas.0507223102

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Matsuda Y, Wang X, Oishi H, Guan Z, Saito M, Liu M, et al. Spleen tyrosine kinase modulates fibrous airway obliteration and associated lymphoid neogenesis after transplantation. Am J Transplant (2016) 16(1):342–52. doi:10.1111/ajt.13442

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Browning JL, Allaire N, Ngam-Ek A, Notidis E, Hunt J, Perrin S, et al. Lymphotoxin-beta receptor signaling is required for the homeostatic control of HEV differentiation and function. Immunity (2005) 23(5):539–50. doi:10.1016/j.immuni.2005.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Liao S, Ruddle NH. Synchrony of high endothelial venules and lymphatic vessels revealed by immunization. J Immunol (2006) 177(5):3369–79. doi:10.4049/jimmunol.177.5.3369

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kumar V, Scandella E, Danuser R, Onder L, Nitschké M, Fukui Y, et al. Global lymphoid tissue remodeling during a viral infection is orchestrated by a B cell-lymphotoxin-dependent pathway. Blood (2010) 115(23):4725–33. doi:10.1182/blood-2009-10-250118

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Moussion C, Girard JP. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature (2011) 479(7374):542–6. doi:10.1038/nature10540

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Wendland M, Willenzon S, Kocks J, Davalos-Misslitz AC, Hammerschmidt SI, Schumann K, et al. Lymph node T cell homeostasis relies on steady state homing of dendritic cells. Immunity (2011) 35(6):945–57. doi:10.1016/j.immuni.2011.10.017

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Sozzani S, Rusnati M, Riboldi E, Mitola S, Presta M. Dendritic cell-endothelial cell cross-talk in angiogenesis. Trends Immunol (2007) 28(9):385–92. doi:10.1016/j.it.2007.07.006

CrossRef Full Text | Google Scholar

40. Webster B, Ekland EH, Agle LM, Chyou S, Ruggieri R, Lu TT. Regulation of lymph node vascular growth by dendritic cells. J Exp Med (2006) 203(8):1903–13. doi:10.1084/jem.20052272

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Benahmed F, Chyou S, Dasoveanu D, Chen J, Kumar V, Iwakura Y, et al. Multiple CD11c + cells collaboratively express IL-1β to modulate stromal vascular endothelial growth factor and lymph node vascular-stromal growth. J Immunol (2014) 192(9):4153–63. doi:10.4049/jimmunol.1301765

CrossRef Full Text | Google Scholar

42. Marinkovic T, Garin A, Yokota Y, Fu YX, Ruddle NH, Furtado GC, et al. Interaction of mature CD3 + CD4 + T cells with dendritic cells triggers the development of tertiary lymphoid structures in the thyroid. J Clin Invest (2006) 116(10):2622–32. doi:10.1172/JCI28993

CrossRef Full Text | Google Scholar

43. Peske JD, Thompson ED, Gemta L, Baylis RA, Fu YX, Engelhard VH. Effector lymphocyte-induced lymph node-like vasculature enables naive T-cell entry into tumours and enhanced anti-tumour immunity. Nat Commun (2015) 6:7114. doi:10.1038/ncomms8114

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Grogan JL, Ouyang W. A role for Th17 cells in the regulation of tertiary lymphoid follicles. Eur J Immunol (2012) 42(9):2255–62. doi:10.1002/eji.201242656

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Rangel-Moreno J, Carragher DM, de la Luz Garcia-Hernandez M, Hwang JY, Kusser K, Hartson L, et al. The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat Immunol (2011) 12(7):639–46. doi:10.1038/ni.2053

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Peters A, Pitcher LA, Sullivan JM, Mitsdoerffer M, Acton SE, Franz B, et al. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity (2011) 35(6):986–96. doi:10.1016/j.immuni.2011.10.015

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Roos AB, Sandén C, Mori M, Bjermer L, Stampfli MR, Erjefält JS. IL-17A is elevated in end-stage chronic obstructive pulmonary disease and contributes to cigarette smoke-induced lymphoid neogenesis. Am J Respir Crit Care Med (2015) 191(11):1232–41. doi:10.1164/rccm.201410-1861OC

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Deteix C, Attuil-Audenis V, Duthey A, Patey N, McGregor B, Dubois V, et al. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J Immunol (2010) 184(9):5344–51. doi:10.4049/jimmunol.0902999

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Cañete JD, Celis R, Yeremenko N, Sanmartí R, van Duivenvoorde L, Ramírez J, et al. Ectopic lymphoid neogenesis is strongly associated with activation of the IL-23 pathway in rheumatoid synovitis. Arthritis Res Ther (2015) 17:173. doi:10.1186/s13075-015-0688-0

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Barone F, Nayar S, Campos J, Cloake T, Withers DR, Toellner KM, et al. IL-22 regulates lymphoid chemokine production and assembly of tertiary lymphoid organs. Proc Natl Acad Sci U S A (2015) 112(35):11024–9. doi:10.1073/pnas.1503315112

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Jones GW, Bombardieri M, Greenhill CJ, McLeod L, Nerviani A, Rocher-Ros V, et al. Interleukin-27 inhibits ectopic lymphoid-like structure development in early inflammatory arthritis. J Exp Med (2015) 212(11):1793–802. doi:10.1084/jem.20132307

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer (2012) 12(4):298–306. doi:10.1038/nrc3245

CrossRef Full Text | Google Scholar

53. Becht E, Giraldo NA, Germain C, de Reyniès A, Laurent-Puig P, Zucman-Rossi J, et al. Immune contexture, immunoscore, and malignant cell molecular subgroups for prognostic and theranostic classifications of cancers. Adv Immunol (2016) 130:95–190. doi:10.1016/bs.ai.2015.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Kocks JR, Davalos-Misslitz AC, Hintzen G, Ohl L, Förster R. Regulatory T cells interfere with the development of bronchus-associated lymphoid tissue. J Exp Med (2007) 204(4):723–34. doi:10.1084/jem.20061424

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Gobert M, Treilleux I, Bendriss-Vermare N, Bachelot T, Goddard-Leon S, Arfi V, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Res (2009) 69(5):2000–9. doi:10.1158/0008-5472.CAN-08-2360

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Hindley JP, Jones E, Smart K, Bridgeman H, Lauder SN, Ondondo B, et al. T-cell trafficking facilitated by high endothelial venules is required for tumor control after regulatory T-cell depletion. Cancer Res (2012) 72(21):5473–82. doi:10.1158/0008-5472.CAN-12-1912

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Foo SY, Zhang V, Lalwani A, Lynch JP, Zhuang A, Lam CE, et al. Regulatory T cells prevent inducible BALT formation by dampening neutrophilic inflammation. J Immunol (2015) 194(9):4567–76. doi:10.4049/jimmunol.1400909

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Joshi NS, Akama-Garren EH, Lu Y, Lee DY, Chang GP, Li A, et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity (2015) 43(3):579–90. doi:10.1016/j.immuni.2015.08.006

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Mebius, Reina E. Organogenesis of lymphoid tissues. Nat Rev Immunol (2003) 3(4):292–303. doi:10.1038/nri1054

CrossRef Full Text | Google Scholar

60. Dejardin E, Droin NM, Delhase M, Haas E, Cao Y, Makris C, et al. The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways. Immunity (2002) 17(4):525–35. doi:10.1016/S1074-7613(02)00423-5

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Meier D, Bornmann C, Chappaz S, Schmutz S, Otten LA, Ceredig R, et al. Ectopic lymphoid-organ development occurs through interleukin 7-mediated enhanced survival of lymphoid-tissue-inducer cells. Immunity (2007) 26(5):643–54. doi:10.1016/j.immuni.2007.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Seo YB, Im SJ, Namkoong H, Kim SW, Choi YW, Kang MC, et al. Crucial roles of interleukin-7 in the development of T follicular helper cells and in the induction of humoral immunity. J Virol (2014) 88(16):8998–9009. doi:10.1128/JVI.00534-14

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Lee LF, Logronio K, Tu GH, Zhai W, Ni I, Mei L, et al. Anti-IL-7 Receptor-α reverses established type 1 diabetes in non-obese diabetic mice by modulating effector T-cell function. Proc Natl Acad Sci U S A (2012) 109(31):12674–9. doi:10.1073/pnas.1203795109

CrossRef Full Text | Google Scholar

64. Vicari AP, Ait-Yahia S, Chemin K, Mueller A, Zlotnik A, Caux C. Antitumor effects of the mouse chemokine 6Ckine/SLC through angiostatic and immunological mechanisms. J Immunol (2000) 165(4):1992–2000. doi:10.4049/jimmunol.165.4.1992

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Nomura T, Hasegawa H, Kohno M, Sasaki M, Fujita S. Enhancement of anti-tumor immunity by tumor cells transfected with the secondary lymphoid tissue chemokine EBI-1-ligand chemokine and stromal cell-derived factor-1alpha chemokine genes. Int J Cancer (2001) 91(5):597–606. doi:10.1002/1097-0215(200002)9999:9999<::AID-IJC1107>3.0.CO;2-J

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Yang SC, Batra RK, Hillinger S, Reckamp KL, Strieter RM, Dubinett SM, et al. Intrapulmonary administration of CCL21 gene-modified dendritic cells reduces tumor burden in spontaneous murine bronchoalveolar cell carcinoma. Cancer Res (2006) 66(6):3205–13. doi:10.1158/0008-5472.CAN-05-3619

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Kuchen S, Robbins R, Sims GP, Sheng C, Phillips TM, Lipsky PE, et al. Essential role of IL-21 in B cell activation, expansion, and plasma cell generation during CD4 + T cell-B cell collaboration. J Immunol (2007) 179(9):5886–96. doi:10.4049/jimmunol.179.9.5886

CrossRef Full Text | Google Scholar

68. Attridge K, Wang CJ, Wardzinski L, Kenefeck R, Chamberlain JL, Manzotti C, et al. IL-21 inhibits T cell IL-2 production and impairs treg homeostasis. Blood (2012) 119(20):4656–64. doi:10.1182/blood-2011-10-388546

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Maldonado L, Teague JE, Morrow MP, Jotova I, Wu TC, Wang C, et al. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci Transl Med (2014) 6(221):221ra13. doi:10.1126/scitranslmed.3007323

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, et al. Immunotherapy converts non-immunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res (2014) 2(7):616–31. doi:10.1158/2326-6066.CIR-14-0027

CrossRef Full Text | Google Scholar

Keywords: cancer, tertiary lymphoid structure, tumor microenvironment, chemokine, adaptive immune response

Citation: Sautès-Fridman C, Lawand M, Giraldo NA, Kaplon H, Germain C, Fridman WH and Dieu-Nosjean M-C (2016) Tertiary Lymphoid Structures in Cancers: Prognostic Value, Regulation, and Manipulation for Therapeutic Intervention. Front. Immunol. 7:407. doi: 10.3389/fimmu.2016.00407

Received: 10 August 2016; Accepted: 22 September 2016;
Published: 03 October 2016

Edited by:

Andreas Habenicht, Ludwig Maximilian University of Munich, Germany

Reviewed by:

Jo A. Van Ginderachter, Vrije Universiteit Brussel, Belgium
Yong-Gil Kim, University of Ulsan College of Medicine, South Korea

Copyright: © 2016 Sautès-Fridman, Lawand, Giraldo, Kaplon, Germain, Fridman and Dieu-Nosjean. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Marie-Caroline Dieu-Nosjean, mc.dieu-nosjean@crc.jussieu.fr

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.