Abstract
Tertiary lymphoid structures (TLS), formerly recognized as Crohn’s-like structures, serve as crucial biomarkers for evaluating the progression of colorectal cancer (CRC). Understanding their spatial distribution, cellular composition, and interactions within CRC is paramount for comprehending the immune response in the tumor microenvironment (TME). TLS are comprised of a T-cellular compartment and a B-cellular compartment, the latter encompassing follicular dendritic cells (FDCs), high endothelial venules (HEVs), and lymphatic vessels. While T helper cells predominate in cancer TLS, the specific functions of their subpopulations remain inadequately understood. Notably, T follicular helper (Tfh) cells play a central role in the activation of CD8+ T cells, and both Tfh cells and Tfh-associated genes have been linked to enhanced CRC survival. In stage II CRC TLS, an escalation in the number of FoxP3+ T regulatory cells (Tregs) is regarded as a negative prognostic factor. Moreover, within TLS, T lymphocytes shield B lymphocytes from the immunosuppressive effects of the TME. B lymphocyte activation is succeeded by class recombination (CSR) and somatic hypermutation (SHM). Dendritic cells (DCs) constitute a vital cellular component of the TLS T compartment. During steady state and early stages of CRC, specialized antigen-presenting cells such as DCs migrate to regional lymph nodes through afferent lymphatics. They deliver MHC antigen-derived peptide complexes (tumor antigens) to naïve CD4+ and CD8+ T cells, which subsequently infiltrate the tumor site as antigen-specific T cells. Key DC markers studied in TLS include CD83 and DC-LAMP. Research has indicated that the DC-LAMP gene signature in tumor TLS reflects Th1 cell targeting, cytotoxicity, and T cell activation. This review comprehensively outlines the functions performed by distinct cell subsets within tertiary lymphoid structures (TLS) in tumors.
Keywords: Tertiary lymphoid structures (TLS), T helpers, B cell activation, dendritic cells, colorectal cancer
Introduction
Tumors develop a specific microenvironment consisting in immune cells, endothelial cells, cancer-associated stromal cells and recently found lymphocyte aggregations called tertiary lymphoid structures (TLS), that release tumor-promoting or tumor-suppressing factors in the tumor microenvironment (TME), that govern tumor development [1-3]. An immunoscore assessing different lymphocytes has been created, that together with conventional TNM staging AJCC/UICC TNM classification can lead to better determination of tumor prognosis [4,5] and the outcome of chemotherapy, radiotherapy and immunotherapy [6,7].
Colorectal cancer (CRC) and immune biomarkers
CRC is one of the most often diagnosed worldwide [8]. The majority of patients with CRC are usually diagnosed in stage II and III of the disease. Several authors have proposed the use of new immunologic biomarkers that can give information about disease recurrence and the use of adjuvant radio- and chemotherapy [9,10]. The immune cell infiltration has been evaluated mainly in tumor stroma (TS) and the tumor invasive front (IF) [4,11]. In CRC the evaluation of the immune infiltrate has been associated with cancer development and prognosis [11-14]. Tumors usually establish an immunosuppressive microenvironment that inhibits effective immune responses. Classically, it has been accepted that tumor-associated antigens (TAAs) are captured and processed by dendritic cells (DCs) that migrate to regional secondary lymphoid organs (SLO) and to TLS and present them to naïve T cells in paracortex and some TAA are accepted by B cell receptor (BCR) of B cells in the cortex [6,15,16].
Tertiary lymphoid structures and CRC development
One important immune biomarker of CRC development and prognosis is TLS formation [9,17], previously known as Crohn’s-like structures appeared in the vicinity of the tumor [17-19]. According to me and others TLS present in 78.6% of CRC and are positive prognostic factor for stage II CRC [20]. The peritumoral location of TLS in CRC has been associated with worse prognosis and cancer progression [21,22]. In a study of large cohort of CRC patients (n=195) authors have found that higher numbers of TLS present in less advanced tumors (TNM - I+II) [23]. Increased TLS in the IF of CRC in node-negative patients are associated with better prognosis [20].
Therefore, we decided to examine the precise cellular composition of TLS, possible immune cell collaboration in TLS and to delineate the axis of DCs Ag presentation to naïve CD4 and CD8 T cells in T-zone, T follicular helper cells (Tfh) in the T zone and at the border of germinal centers (GC), and B cell activation. We try to represent in schemas the complex efforts of DCs, Tfh cells and B cells to activate CD8+ T cells and send them to the tumor area.
TLS origin and formation
In the recent 20 years, a new lymphoid structure has been investigated in some tumors such as bronchial cancer, CRC, gastric cancer, etc. [9] and has been called tertiary lymphoid structure. TLS has been situated closer to the tumor and mirror SLO organization and function. TLS are formed mainly at the tumor margin or in the tumor stroma [2]. Moreover, TLS are detected at sites of chronic inflammation [20] or autoimmunity [24]. The term TLS has been mentioned first in 1992 by Louis Picker and Eugene Butcher [25]. Authors call these structures extra-lymphoid sites, where lymphocyte precursors (or memory lymphocytes) are re-stimulated by antigen. TLS are structures that arise in non-immune tissues and initiate adaptive immune response [24].
TLS contains immune cells organized as SLOs [26]. The development of SLOs is a genetically programmed process that takes place during embryogenesis. SLOs are lymph nodes (LN), white pulp of the spleen, human appendix, mucosal-associated lymphoid tissue (MALT) that includes also Payer’s patches (PP) and tonsils [27-29]. LNs and PPs develop prenatally, while bronchial-associated lymphoid tissue (BALT), isolated lymphoid follicles in the intestine and gallbladder occur postnatally [30,31]. The latter are induced and preprogrammed during ontogeny but appear after birth [32]. Primary lymphoid organs are bone marrow and thymus. The liver loses its hematopoietic function postnatally and it can form TLS in adult life [33]. Other organs like kidneys, heart, salivary glands, synovium, etc. form lymphoid aggregates in disease, considered as TLS [24].
TLS closely resemble SLO and are structures from single cluster of lymphocytes to structures resembling LNs but without capsule [34]. SLOs are organized by lymphoid tissue inducer (LTi) cells and by lymphoid tissue organizer (LTo) cells that produce CXCL13, CCL19 and CCL21 chemokines that attract recruitment of B and T lymphocytes. Moreover, LTo cells change into follicular dendritic cells (FDCs) and form fibroblastic and reticular cell network in SLO [35]. Morphologists define TLS by the following criteria: TLS have T and B cell compartments; FDCs in B cell zones are associated with enzyme-activation-induced cytidine deaminase (AID) required for initiation of somatic hypermutation (SHM) and immunoglobulin class switching recombination (CSR) [6]. TLS have also high endothelial venules (HEV) and lymphatic vessels that are still not well studied [32]. TLS formation is induced by chronic inflammation even in the absence of LTi cells [36]. Some authors report that T helper 17 (Th17) cells [36,37], B cells [38], DCs [7,39], M1 macrophages [3,40], and T follicular helper cells (Tfh) [29,41] may initiate TLS organization. Therefore, immune cells may substitute for LTi cells in order to stimulate TLS neogenesis [42] (Figure 1).
Figure 1.
Scheme of TLS. Cells in T zones are CD4+ and CD8+ T cells, Tregs and dendritic cells. High endothelial venules (HEVs) carry immune cells i.e. T, B cells DCs, from blood, coming from bone marrow. B cells gather in the germinal centers around Follicular dendritic cells (FDC). TLS have no collagen-rich capsule (adapted after Munoz-Erazo et al., 2020) [29].
Immune infiltrate in TLS
T helper cells predominate in TLS in cancer and Th2 cells and their cytokines mainly present in TLS in advanced CRC [8]. Th17 cells have recently emerged as key factors that trigger TLS development [29,37]. Tfh cells and FDCs in the germinative centres (GCs) of TLS can produce chemokines like CXCL13 and IL-6 that promote B cell activation, their antigen-presentation and the initiation of humoral immune response [3,34].
T helper cells T helper 1 (Th1), T helper 2 (Th2), T helper 17/22 (Th17)/(Th22), T regulatory cells (Tregs) and Tfh cells comprise T cell infiltrate in TME and TLS [43-46]. Th1, Th2 and Tfh cells have been first investigated in SLO and TLS immune responses [46]. Some studies have demonstrated that Th17/Th22 cells participate in shaping of TLS [24]. The presence of Th cell subsets in TLS is reported by a few authors [8,20,47,48].
T follicular helper cells are defined in 1980 and are specialized T cells that provide help to B cells [41,49]. Tfh cells activate other immune cells such as B lymphocytes and CD8+ T cells [45]. Tfh cells stimulate affinity maturation and production of antibodies of B cells. CCR5 chemokine ensures movement of naïve T cells from T cell zone to B cell follicle. Tfh cells express STAT3 and Bcl6 transcription factors (TF) and IL21R, CXCR5, CD28, CD40L, CD84, TCR, ICOS and secrete cytokines like IL-10, IL-21 and IL-6 [49,50]. Bcl6 represses the differentiation and formation of Th1, Th2, and Th17 cells [50]. Recent investigations suggest that Tfh can differentiate into IL-17 producing Tfh cells independent of retinoid-orphan receptor (RORγt) expression [50,51]. In CRC, increased numbers of T helpers in TLS and of Tfh-related genes, such as CXCL13 and IL-21 has been associated with improved survival [8,47]. In SLO naïve T cells (CD4+) activated by TGFβ and IL12/23 change to Tfh cells CXCR5+ PD-1+ Bcl6+ ICOS+ Sox4+ in GCs [52]. In peripheral tissues CXCL13 induces TLS formation and the appearance of T peripheral helper (Tph) cells that are CXCR5- PD-1+ Sox4+ CCR2+ and that target B memory cells [41] (Figure 2).
Figure 2.
Scheme of activation of naïve T cell into Th1, Th2, Th17, Treg and Tfh cells. The connection between activated Tfh and B cell and CD8+ T cell. DCs present antigen to naïve T cells (B-7/CD28). Primed T cell via IL-12 differentiates into Th1 cell; via IL-4 differentiates into Th2; via TGF-β changes into FOXP3+ Tregs; via TGF-β, IL-6 and IL-23 changes into Th17. Activated Tfh cell interacts via CD40/CD40 ligand, and ICOS/ICOSL with B cell. B cell contacts with FDC via BCR and encounters antigens. This triggers their proliferation, differentiation into plasma cell undergoing CSR and antibody secretion (adapted after Kim et al., 2010; Biolegend poster) [78].
Using multiplex immunohistochemistry it is shown that higher Tfh cells and macrophages densities in sixty-seven CRC patients are associated with relapsed patients [8]. The prognostic value in 603 patients with CRC liver metastasis of TLS in the tumor stroma and peritumorally is established. It is shown that Tfh cells prevail in TLS in the tumor [53]. In three endoscopic ortothopic colon-cancer mouse models the densities of Tfh cells decrease with tumor progression [54]. Recently, Tfh cells have emerged as the key cell type, required for the formation of GCs in SLO and TLS [55].
Tregs are detected in CRC TLS [56]. FoxP3+ Tregs are studied in TLS in stage II of CRC and in LNs, where the increased number of Tregs is a negative prognostic factor of overall survival [57]. On the contrary, the increased number of FoxP3+ Tregs in the tumor tissue of stage II CRC has been associated with better survival [58]. Decreased number of FoxP3+ Tregs in tumor stroma of CRC has been found in MSS CRCs [11].
Th17 cells (CD3+ RORγt+) are necessary for the development of TLS [8,29,59]. In fact, Th17+ cells has been dispersed around TLS (my observation). IL-17 is secreted by a lot of cells such as γδ T cells, macrophages and lymphocytes [11]. It has been supposed that Tregs attenuate Th17 cell-dependent pro-inflammatory and tumor-enhancing responses that could be possible explanation for the favourable prognosis in CRC [60] (Figure 3).
Figure 3.
Schema of Tfh cells and CRC promotion. Different cytokines and transcription factors govern T helper appearance in TLS. T follicular regulatory (TFR) cells originating from thymic T-reg cells express CXCR5, PD-1 and ICOS. TFR cells suppresses germinal center B cells and the production of anti-tumor antibodies. Tfh cells via production of IL-21 stimulate Bregs proliferation. Bregs production of TGF-β and IL-10 regulate the immune tolerance in TME promoting tumor development (adapted after Hetta et al., 2020) [46].
Follicular dendritic cells (FDC) contained opsonized antigens on their surface. FDCs are described firs in 1968 by Andras K Szakal and Michel G Hanna [61]. FDCs are cells of mesenchymal origin, that lack phagocytosis, lysosomes or Birbeck granules. They have many fine dendritic processes forming a network in the GC and retain antigen-antibody-complement immune complexes for a long time. They retain mobile B cells and T cells in GC [62]. FDCs are located in the light zone of GCs. These cells have complement receptors (CR1/CR2 - CD35/CD21), immunoglobulin gamma Fc region receptor (FcγRIIB/CD32 and Fc2εRII/CD23) - epsilon Fc region receptor [63]. FDCs are induced in TLS.
Unlike T lymphocytes in T zone of TLS, B lymphocytes has been acknowledged recently as a patent immune cell that organizes anti-tumor immunity [63,64]. Since 1970s, it has been considered that B cells possess only pro-tumor function, while recently it has been shown that B cells take part in anti-tumor immunity [59,65]. B lymphocytes develop in liver during fetal life (B-1a lineage) and secrete “natural IgM antibodies”. In adults, B lymphocytes arise in bone marrow (BM) from common lymphoid progenitor (CLP). These B cells are of B-1b and B-2 lineages [66]. It is proven that B cells in TME, outside TLS can acquire suppressive function and contribute to tumor growth [59,67]. In TLS the mantle of T lymphocytes can physically protect B cells from tumor immunosuppressive molecules [67]. In mature germinal centers of TLS and SLO, there are two zones i.e. dark zone (DZ) in the periphery and light zone (LZ) in the center. Naïve B cells first are concentrated in the DZ, where they contact with Tfh cells coming from T zone, after priming by DCs. Tfh cells through CD40L molecule interact with B cells (CD40 molecule) and promote B cell activation and differentiation [66-68]. In LZ of GC B cells capture antigens through B cell receptor (BCR). Antigens (complement-bound peptides) come from peripheral tissues via lymph and are captured by follicular dendritic cells (FDC) [63]. Later B cells process and present antigen to CD4+ and CD8+ T cells through MHC class II and class I molecules, respectively [69]. So B cells act as APCs such as DCs and promote T cell-mediated immune responses. Activated B cells then undergo class switch recombination (CSR) and generate IgG, IgA and IgE antibodies. B cells also undergo somatic hypermutation (SHM) of the Ig variable region and so ensure antibody diversification and affinity maturation [70,71] (Figure 2).
In TME B cells mainly gather in TLS. In addition, B cells significantly correlate with prognosis of CRC [70]. The role of B lymphocytes in CRC is less explored and in matters of prognostic significance consensus has yet to be reached [72]. The favorable patient prognosis in a large cohort of CRC probes is shown for CD20+ B cells [73]. The neutral and negative impact of CD20+ B lymphocytes on CRC patient’s prognosis has been also detected [74]. Patients with CRCs highly infiltrated by CD20+ B lymphocytes show favorable outcome [72]. However, the function of B lymphocytes in tumor development still remain unclear.
Dendritic cells are professional antigen-presenting cells (APCs), that are a component of TLS [39,75-77]. Tumor-infiltrating macrophages can activate T cells in tumor stroma, while DCs encounter antigen and migrate to TLS or to the draining LNs via various lymph vessels [78], where they prime naïve CD4 and CD8 T lymphocytes [79]. DCs together with B cells present antigen to T cells. At present four main types of DCs are determined i.e. cDC1, cDC2, migratory DC3 (mDC3) and plasmacytoid DCs (pDCs). A fifth DC subset DC5 has been recognized and considered to be inflammatory (DC5) that contained mostly cDC2 state [77,79,80]. The cDC1s have been considered to be main cells in cross-presentation of antigens [76]. The cDC1s are major source of IL-12, activate NK cells and drive CD4 cells towards Th1 responses [76,81]. The cDC2s are the most numerous DC population in tumours. They provide licensing signal to cDC1s, which activate CD8+ T lymphocytes (CTLs). The cDC2s activate CD4+ T cells via MHC class II and provide help to cDC1s to maximize CTL responses [75,76]. CD4+ T lymphocytes primed by DCs are mainly of Th1, Th2, Treg and Tfh phenotypes in TLS and in LNs. Tfh contact with B lymphocytes and activate them to present new antigens (accepted via TCR) to naïve CD4+ and CD8+ T cells or to modify into plasma cells [75] (Figure 4).
Figure 4.
Schema of cDC1 and cDC2 role in anti-tumor immunity. The cDC2 cells activate CD4+ T cells. The cDC1 cells predominantly activate CD8+ T cells. Activated CD4+ T cells provide help to maximize CTL responses. pDCs additionally activate cDC1 through type I interferon and CD4+ T cells (adapted after Noubade et al., 2019; Rossi et al., 2022) [67,69].
The main DCs markers investigated in TLS are CD83 and DC-LAMP [76,77,81]. CD123+ pDCs or BDCA-2 are searched [76,82]. It has been shown that gene signature of DC-LAMPhi TLS tumors presents Th1 orientation, cytotoxicity and T cell activation [81].
When exploring the presence of DCs in CRC, it has been reported that DCs are detected in all CRC specimens [83,84]. Recently, we have shown that CD1a and CD83 DCs are significantly higher in TS and IF of MSI CRC patients [85]. Limited data existed about DCs in TLS of CRC specimens [86]. In early stage of lung cancer DC-Lamp+ DCs home selectively in TLS and are associated with longer survival [87]. DCs from colon and rectal cancers mature differently, while colon cancer inhibits the levels of five DC markers (CD54, CD80, HLA-DR, CD86, and CD83), rectal cancer enhanced the levels of three DC markers (CD80, CD86, and CD83) [88]. Typically, TLS of the periphery of the tumor have more organized and distinct DC/T cell and B cell zones than the intratumoral TLS, which contained mostly B cells [89]. In rectal cancer the neoadjuvant CRT stimulates the recruitment of pDCs in comparison without such therapy [82]. The cDC2 (CD1c+) DC phenotype has been considered to be the most abundant, while other DC subtypes vary in each cancer type [90]. That means that the investigation of DC subtypes in TLS in CRC, will reveal the fine contexture of these immune structures.
TLSs in the invasive front of node negative CRCs have been characterized with increased density of CD3+ T lymphocytes and PNaD+ high endothelial venules (HEVs), CD20+ B lymphocytes and CD21+ FDCs, two chemokines CCL21 and CXCL13 and Lyve-1+ lymph vessels that indicate favorable outcome. Presence of these chemokines indicates that TLSs mediate active recruitment of lymhphocytes and dendritic cells in the tumor. Colon and rectal cancers have different distribution of immune cells and react differently to therapy. Noteworthy, another understudied issue is the impact of standart treatment (surgery, chemotherapy and radiotherapy) on the functional properties and prognostic significance of immune cells in TLS. It will be important to assess B cells, T cells in the context of Th1/Tfh/Treg/Th17 subsets and DCs in TLS as this undoubtedly influences their functional attributes in CRC.
Therefore, TLSs are induced structures by inflammation, autoimmune processes and cancer. They are formed as a site of immune battle next to pathologic area. A more detailed analysis of their immune cell content is needed to clarify the fine mechanisms of anti-tumor cellular and molecular response.
Disclosure of conflict of interest
None.
References
- 1.Kwak Y, Koh J, Kim DW, Kang SB, Kim WH, Lee HS. Immunoscore encompassing CD3+ and CD8+ T cell densities in distant metastasis is a robust prognostic marker for advanced colorectal cancer. Oncotarget. 2016;7:81778–81790. doi: 10.18632/oncotarget.13207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Guo G, Wang Y, Zhou Y, Quan Q, Zhang Y, Wang H, Zhang B, Xia L. Immune cell concentrations among the primary tumor microenvironment in colorectal cancer patients predicted by clinicopathologic characteristics and blood indexes. J Immunother Cancer. 2019;7:179. doi: 10.1186/s40425-019-0656-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kang W, Feng Z, Luo J, He Z, Liu J, Wu J, Rong P. Tertiary lymphoid structures in cancer: the double-edged sword role in antitumor immunity and potential therapeutic induction strategies. Front Immunol. 2021;12:689270. doi: 10.3389/fimmu.2021.689270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zhao Y, Ge X, He J, Cheng Y, Wang Z, Wang J, Sun L. The prognostic value of tumor-infiltrating lymphocytes in colorectal cancer differs by anatomical subsite: a systematic review and meta-analysis. World J Surg Oncol. 2019;17:85. doi: 10.1186/s12957-019-1621-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wang Y, Dong J, Quan Q, Liu S, Chen X, Cai X, Qiu H, Zhang B, Guo G. Immune cell infiltration of the primary tumor microenvironment predicted the treatment outcome of chemotherapy with or without bevacizumab in metastatic colorectal cancer patients. Front Oncol. 2021;10:581051. doi: 10.3389/fonc.2020.581051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Colbeck EJ, Ager A, Gallimore A, Jones GW. Tertiary lymphoid structures in cancer: drivers of antitumor immunity, immunosuppression, or bystander sentinels in disease? Front Immunol. 2017;8:1830. doi: 10.3389/fimmu.2017.01830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mosińska P, Gabryelska A, Zasada M, Fichna J. Dual functional capability of dendritic cells - cytokine-induced killer cells in improving side effects of colorectal cancer therapy. Front Pharmacol. 2017;8:126. doi: 10.3389/fphar.2017.00126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yamaguchi K, Ito M, Ohmura H, Hanamura F, Nakano M, Tsuchihashi K, Nagai S, Ariyama H, Kusaba H, Yamamoto H, Oda Y, Nakamura M, Akashi K, Baba E. Helper T cell-dominant tertiary lymphoid structures are associated with disease relapse of advanced colorectal cancer. Oncoimmunology. 2020;9:1724763. doi: 10.1080/2162402X.2020.1724763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Posch F, Silina K, Leibl S, Mündlein A, Moch H, Siebenhüner A, Samaras P, Riedl J, Stotz M, Szkandera J, Stöger H, Pichler M, Stupp R, van den Broek M, Schraml P, Gerger A, Petrausch U, Winder T. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2017;7:e1378844. doi: 10.1080/2162402X.2017.1378844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chen J, Chen J, Wang L. Tertiary lymphoid structures as unique constructions associated with the organization, education, and function of tumor-infiltrating immunocytes. J Zhejiang Univ Sci B. 2022;23:812–822. doi: 10.1631/jzus.B2200174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gulubova MV, Chonov DC, Ivanova KV, Hristova MK, Krasimirova-Ignatova MM, Vlaykova TI. Intratumoural expression of IL-6/Stat3, IL-17 and Foxp3 immune cells in the immunosuppressive tumour microenvironment of colorectal cancer immune cells-positive for IL-6, Stat3, IL-17 and Foxp3 and colorectal cancer development. Biotechnol Biotechnol Equip. 2022;36:327–38. [Google Scholar]
- 12.Camus M, Tosolini M, Mlecnik B, Pagès F, Kirilovsky A, Berger A, Costes A, Bindea G, Charoentong P, Bruneval P, Trajanoski Z, Fridman WH, Galon J. Coordination of intratumoral immune reaction and human colorectal cancer recurrence. Cancer Res. 2009;69:2685–2693. doi: 10.1158/0008-5472.CAN-08-2654. [DOI] [PubMed] [Google Scholar]
- 13.Pagès F, Kirilovsky A, Mlecnik B, Asslaber M, Tosolini M, Bindea G, Lagorce C, Wind P, Marliot F, Bruneval P, Zatloukal K, Trajanoski Z, Berger A, Fridman WH, Galon J. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J. Clin. Oncol. 2009;27:5944–5951. doi: 10.1200/JCO.2008.19.6147. [DOI] [PubMed] [Google Scholar]
- 14.Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, Lugli A, Zlobec I, Hartmann A, Bifulco C, Nagtegaal ID, Palmqvist R, Masucci GV, Botti G, Tatangelo F, Delrio P, Maio M, Laghi L, Grizzi F, Asslaber M, D’Arrigo C, Vidal-Vanaclocha F, Zavadova E, Chouchane L, Ohashi PS, Hafezi-Bakhtiari S, Wouters BG, Roehrl M, Nguyen L, Kawakami Y, Hazama S, Okuno K, Ogino S, Gibbs P, Waring P, Sato N, Torigoe T, Itoh K, Patel PS, Shukla SN, Wang Y, Kopetz S, Sinicrope FA, Scripcariu V, Ascierto PA, Marincola FM, Fox BA, Pagès F. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 2014;232:199–209. doi: 10.1002/path.4287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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. doi: 10.1038/s41423-020-0457-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Stranford S, Ruddle NH. Follicular dendritic cells, conduits, lymphatic vessels, and high endothelial venules in tertiary lymphoid organs: parallels with lymph node stroma. Front Immunol. 2012;3:350. doi: 10.3389/fimmu.2012.00350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Maoz A, Dennis M, Greenson JK. The Crohn’s-like lymphoid reaction to colorectal cancer-tertiary lymphoid structures with immunologic and potentially therapeutic relevance in colorectal cancer. Front Immunol. 2019;10:1884. doi: 10.3389/fimmu.2019.01884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bergomas F, Grizzi F, Doni A, Pesce S, Laghi L, Allavena P, Mantovani A, Marchesi F. Tertiary intratumor lymphoid tissue in colo-rectal cancer. Cancers (Basel) 2011;4:1–10. doi: 10.3390/cancers4010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kim JH, Kim KJ, Bae JM, Rhee YY, Cho NY, Lee HS, Kang GH. Comparative validation of assessment criteria for Crohn-like lymphoid reaction in colorectal carcinoma. J Clin Pathol. 2015;68:22–8. doi: 10.1136/jclinpath-2014-202603. [DOI] [PubMed] [Google Scholar]
- 20.Di Caro G, Castino GF, Bergomas F, Cortese N, Chiriva-Internati M, Grizzi F, Mantovani A, Marchesi F. Tertiary lymphoid tissue in the tumor microenvironment: from its occurrence to immunotherapeutic implications. Int Rev Immunol. 2015;34:123–33. doi: 10.3109/08830185.2015.1018416. [DOI] [PubMed] [Google Scholar]
- 21.Bento DC, Jones E, Junaid S, Tull J, Williams GT, Godkin A, Ager A, Gallimore A. High endothelial venules are rare in colorectal cancers but accumulate in extra-tumoral areas with disease progression. Oncoimmunology. 2015;4:e974374. doi: 10.4161/2162402X.2014.974374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bery AI, Shepherd HM, Li W, Krupnick AS, Gelman AE, Kreisel D. Role of tertiary lymphoid organs in the regulation of immune responses in the periphery. Cell Mol Life Sci. 2022;79:359. doi: 10.1007/s00018-022-04388-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Markowski AR, Markowska AJ, Ustymowicz W, Pryczynicz A, Guzińska-Ustymowicz K. Simultaneous analysis of tumor-infiltrating immune cells density, tumor budding status, and presence of lymphoid follicles in CRC tissue. Sci Rep. 2022;12:21732. doi: 10.1038/s41598-022-26225-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pipi E, Nayar S, Gardner DH, Colafrancesco S, Smith C, Barone F. Tertiary lymphoid structures: autoimmunity goes local. Front Immunol. 2018;9:1952. doi: 10.3389/fimmu.2018.01952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pimenta EM, Barnes BJ. Role of tertiary lymphoid structures (TLS) in anti-tumor immunity: potential tumor-induced cytokines/chemokines that regulate TLS formation in epithelial-derived cancers. Cancers (Basel) 2014;6:969–997. doi: 10.3390/cancers6020969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol. 2006;7:344–353. doi: 10.1038/ni1330. [DOI] [PubMed] [Google Scholar]
- 27.Jones GW, Hill DG, Jones SA. Understanding immune cells in tertiary lymphoid organ development: it is all starting to come together. Front Immunol. 2016;7:401. doi: 10.3389/fimmu.2016.00401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Honda M, Furuta Y, Naoe H, Sasaki Y. Primary mucosa-associated lymphoid tissue (MALT) lymphoma of the gallbladder and review of the literature. BMJ Case Rep. 2017;2017:bcr2017220161. doi: 10.1136/bcr-2017-220161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Műzes G, Bohusné Barta B, Sipos F. Colitis and colorectal carcinogenesis: the focus on isolated lymphoid follicles. Biomedicines. 2022;10:226. doi: 10.3390/biomedicines10020226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dieu-Nosjean MC, Goc J, Giraldo NA, Sautès-Fridman C, Fridman WH. Tertiary lymphoid structures in cancer and beyond. Trends Immunol. 2014;35:571–80. doi: 10.1016/j.it.2014.09.006. [DOI] [PubMed] [Google Scholar]
- 31.Stefanov IS. Age-dependent distribution and size of lymphatic nodules as components of extrahepatic bile duct-associated lymphatic tissue in domestic swine - a micromorphometric study. Bulg J Vet Med. 2021;26:152–167. [Google Scholar]
- 32.Barone F, Gardner DH, Nayar S, Steinthal N, Buckley CD, Luther SA. Stromal fibroblasts in tertiary lymphoid structures: a novel target in chronic inflammation. Front Immunol. 2016;7:477. doi: 10.3389/fimmu.2016.00477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Goc J, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology. 2013;2:e26836. doi: 10.4161/onci.26836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Katakai T. Marginal reticular cells: a stromal subset directly descended from the lymphoid tissue organizer. Front Immunol. 2012;3:200. doi: 10.3389/fimmu.2012.00200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jones GW, Jones SA. Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues. Immunology. 2016;147:141–51. doi: 10.1111/imm.12554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Grogan JL, Ouyang W. A role for Th17 cells in the regulation of tertiary lymphoid follicles. Eur J Immunol. 2012;42:2255–62. doi: 10.1002/eji.201242656. [DOI] [PubMed] [Google Scholar]
- 37.Hughes CE, Benson RA, Bedaj M, Maffia P. Antigen-presenting cells and antigen presentation in tertiary lymphoid organs. Front Immunol. 2016;7:481. doi: 10.3389/fimmu.2016.00481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.N J, J T, Sl N, Gt B. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies. Oncoimmunology. 2021;10:1900508. doi: 10.1080/2162402X.2021.1900508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gupta YH, Khanom A, Acton SE. Control of dendritic cell function within the tumour microenvironment. Front Immunol. 2022;13:733800. doi: 10.3389/fimmu.2022.733800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zheng X, Weigert A, Reu S, Guenther S, Mansouri S, Bassaly B, Gattenlöhner S, Grimminger F, Pullamsetti S, Seeger W, Winter H, Savai R. Spatial density and distribution of tumor-associated macrophages predict survival in non-small cell lung carcinoma. Cancer Res. 2020;80:4414–4425. doi: 10.1158/0008-5472.CAN-20-0069. [DOI] [PubMed] [Google Scholar]
- 41.Hetta HF, Elkady A, Yahia R, Meshall AK, Saad MM, Mekky MA, Al-Kadmy IMS. T follicular helper and T follicular regulatory cells in colorectal cancer: a complex interplay. J Immunol Methods. 2020;480:112753. doi: 10.1016/j.jim.2020.112753. [DOI] [PubMed] [Google Scholar]
- 42.Tosolini M, Kirilovsky A, Mlecnik B, Fredriksen T, Mauger S, Bindea G, Berger A, Bruneval P, Fridman WH, Pagès F, Galon J. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, Th2, Treg, th17) in patients with colorectal cancer. Cancer Res. 2011;71:1263–71. doi: 10.1158/0008-5472.CAN-10-2907. [DOI] [PubMed] [Google Scholar]
- 43.Cosmi L, Maggi L, Santarlasci V, Liotta F, Annunziato F. T helper cells plasticity in inflammation. Cytometry A. 2014;85:36–42. doi: 10.1002/cyto.a.22348. [DOI] [PubMed] [Google Scholar]
- 44.Cui G. TH9, TH17, and TH22 cell subsets and their main cytokine products in the pathogenesis of colorectal cancer. Front Oncol. 2019;9:1002. doi: 10.3389/fonc.2019.01002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bai Z, Zhou Y, Ye Z, Xiong J, Lan H, Wang F. Tumor-infiltrating lymphocytes in colorectal cancer: the fundamental indication and application on immunotherapy. Front Immunol. 2022;12:808964. doi: 10.3389/fimmu.2021.808964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Mackay CR. Follicular homing T helper (Th) cells and the Th1/Th2 paradigm. J Exp Med. 2000;192:F31–4. doi: 10.1084/jem.192.11.f31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.McMullen TP, 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;161:81–8. doi: 10.1111/j.1365-2249.2010.04147.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hutloff A. T follicular helper-like cells in inflamed non-lymphoid tissues. Front Immunol. 2018;9:1707. doi: 10.3389/fimmu.2018.01707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Teillaud JL, Dieu-Nosjean MC. Tertiary lymphoid structures: an anti-tumor school for adaptive immune cells and an antibody factory to fight cancer? Front Immunol. 2017;8:830. doi: 10.3389/fimmu.2017.00830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Xu K, Yang WY, Nanayakkara GK, Shao Y, Yang F, Hu W, Choi ET, Wang H, Yang X. GATA3, HDAC6, and BCL6 regulate FOXP3+ Treg plasticity and determine Treg conversion into either novel antigen-presenting cell-like Treg or Th1-Treg. Front Immunol. 2018;9:45. doi: 10.3389/fimmu.2018.00045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Rao DA. T cells that help B cells in chronically inflamed tissues. Front Immunol. 2018;9:1924. doi: 10.3389/fimmu.2018.01924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yoshitomi H, Ueno H. Shared and distinct roles of T peripheral helper and T follicular helper cells in human diseases. Cell Mol Immunol. 2021;18:523–527. doi: 10.1038/s41423-020-00529-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang C, Wang XY, Zuo JL, Wang XF, Feng XW, Zhang B, Li YT, Yi CH, Zhang P, Ma XC, Chen ZM, Ma Y, Han JH, Tao BR, Zhang R, Wang TQ, Tong L, Gu W, Wang SY, Zheng XF, Yuan WK, Kan ZJ, Fan J, Hu XY, Li J, Zhang C, Chen JH. 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. doi: 10.1136/jitc-2022-006425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, Angell H, Fredriksen T, Lafontaine L, Berger A, Bruneval P, Fridman WH, Becker C, Pagès F, Speicher MR, Trajanoski Z, Galon J. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–95. doi: 10.1016/j.immuni.2013.10.003. [DOI] [PubMed] [Google Scholar]
- 55.Ioannidou K, Ndiaye DR, Noto A, Fenwick C, Fortis SP, Pantaleo G, Petrovas C, de Leval L. In situ characterization of follicular helper CD4 T cells using multiplexed imaging. Front Immunol. 2021;11:607626. doi: 10.3389/fimmu.2020.607626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schweiger T, Berghoff AS, Glogner C, Glueck O, Rajky O, Traxler D, Birner P, Preusser M, Klepetko W, Hoetzenecker K. Tumor-infiltrating lymphocyte subsets and tertiary lymphoid structures in pulmonary metastases from colorectal cancer. Clin Exp Metastasis. 2016;33:727–39. doi: 10.1007/s10585-016-9813-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hanke T, Melling N, Simon R, Sauter G, Bokemeyer C, Lebok P, Terracciano LM, Izbicki JR, Marx AH. High intratumoral FOXP3+ T regulatory cell (Tregs) density is an independent good prognosticator in nodal negative colorectal cancer. Int J Clin Exp Pathol. 2015;8:8227–35. [PMC free article] [PubMed] [Google Scholar]
- 58.Cui G. Immune battle at the premalignant stage of colorectal cancer: focus on immune cell compositions, functions and cytokine products. Am J Cancer Res. 2020;10:1308–1320. [PMC free article] [PubMed] [Google Scholar]
- 59.Kinker GS, Vitiello GAF, Ferreira WAS, Chaves AS, Cordeiro de Lima VC, Medina TDS. B cell orchestration of anti-tumor immune responses: a matter of cell localization and communication. Front Cell Dev Biol. 2021;9:678127. doi: 10.3389/fcell.2021.678127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ladoire S, Martin F, Ghiringhelli F. Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother. 2011;60:909–18. doi: 10.1007/s00262-011-1046-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Szakal AK, Hanna MG Jr. The ultrastructure of antigen localization and viruslike particles in mouse spleen germinal centers. Exp Mol Pathol. 1968;8:75–89. doi: 10.1016/0014-4800(68)90007-5. [DOI] [PubMed] [Google Scholar]
- 62.El Shikh ME, El Sayed RM, Sukumar S, Szakal AK, Tew JG. Activation of B cells by antigens on follicular dendritic cells. Trends Immunol. 2010;31:205–11. doi: 10.1016/j.it.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fridman WH, Petitprez F, Meylan M, Chen TW, Sun CM, Roumenina LT, Sautès-Fridman C. B cells and cancer: to B or not to B? J Exp Med. 2021;218:e20200851. doi: 10.1084/jem.20200851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Guo FF, Cui JW. The role of tumor-infiltrating B cells in tumor immunity. J Oncol. 2019;2019:2592419. doi: 10.1155/2019/2592419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Peña-Romero AC, Orenes-Piñero E. Dual effect of immune cells within tumour microenvironment: pro- and anti-tumour effects and their triggers. Cancers (Basel) 2022;14:1681. doi: 10.3390/cancers14071681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Germain C, Gnjatic S, Dieu-Nosjean MC. Tertiary lymphoid structure-associated B cells are key players in anti-tumor immunity. Front Immunol. 2015;6:67. doi: 10.3389/fimmu.2015.00067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Qin M, Jin Y, Pan LY. Tertiary lymphoid structure and B-cell-related pathways: a potential target in tumor immunotherapy. Oncol Lett. 2021;22:836. doi: 10.3892/ol.2021.13097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Rossetti RAM, Lorenzi NPC, Yokochi K, Rosa MBSF, Benevides L, Margarido PFR, Baracat EC, Carvalho JP, Villa LL, Lepique AP. B lymphocytes can be activated to act as antigen presenting cells to promote anti-tumor responses. PLoS One. 2018;13:e0199034. doi: 10.1371/journal.pone.0199034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cyster JG, Allen CDC. B cell responses: cell interaction dynamics and decisions. Cell. 2019;177:524–540. doi: 10.1016/j.cell.2019.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Meshcheryakova A, Tamandl D, Bajna E, Stift J, Mittlboeck M, Svoboda M, Heiden D, Stremitzer S, Jensen-Jarolim E, Grünberger T, Bergmann M, Mechtcheriakova D. 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:e99008. doi: 10.1371/journal.pone.0099008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Noubade R, Majri-Morrison S, Tarbell KV. Beyond cDC1: emerging roles of DC crosstalk in cancer immunity. Front Immunol. 2019;10:1014. doi: 10.3389/fimmu.2019.01014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Edin S, Kaprio T, Hagström J, Larsson P, Mustonen H, Böckelman C, Strigård K, Gunnarsson U, Haglund C, Palmqvist R. The prognostic importance of CD20+ B lymphocytes in colorectal cancer and the relation to other immune cell subsets. Sci Rep. 2019;9:19997. doi: 10.1038/s41598-019-56441-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Berntsson J, Nodin B, Eberhard J, Micke P, Jirström K. Prognostic impact of tumour-infiltrating B cells and plasma cells in colorectal cancer. Int J Cancer. 2016;139:1129–39. doi: 10.1002/ijc.30138. [DOI] [PubMed] [Google Scholar]
- 74.Kasajima A, Sers C, Sasano H, Jöhrens K, Stenzinger A, Noske A, Buckendahl AC, Darb-Esfahani S, Müller BM, Budczies J, Lehman A, Dietel M, Denkert C, Weichert W. Down-regulation of the antigen processing machinery is linked to a loss of inflammatory response in colorectal cancer. Hum Pathol. 2010;41:1758–69. doi: 10.1016/j.humpath.2010.05.014. [DOI] [PubMed] [Google Scholar]
- 75.de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M, Cremer I, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res. 2011;71:6391–9. doi: 10.1158/0008-5472.CAN-11-0952. [DOI] [PubMed] [Google Scholar]
- 76.Rossi A, Belmonte B, Carnevale S, Liotti A, De Rosa V, Jaillon S, Piconese S, Tripodo C. Stromal and immune cell dynamics in tumor associated tertiary lymphoid structures and anti-tumor immune responses. Front Cell Dev Biol. 2022;10:933113. doi: 10.3389/fcell.2022.933113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zou J, Zhang Y, Zeng Y, Peng Y, Liu J, Xiao C, Wu F. Tertiary lymphoid structures: a potential biomarker for anti-cancer therapy. Cancers (Basel) 2022;14:5968. doi: 10.3390/cancers14235968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gulinac M, Dikov D, Lichev S, Velikova T. Current concept for tertiary lymphoid structures in urothelial carcinoma of the bladder: a literature review and our experience. Am J Clin Exp Immunol. 2020;9:64–72. [PMC free article] [PubMed] [Google Scholar]
- 79.Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, Cheong C, Liu K, Lee HW, Park CG, Steinman RM, Nussenzweig MC. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315:107–11. doi: 10.1126/science.1136080. [DOI] [PubMed] [Google Scholar]
- 80.Collin M, Bigley V. Human dendritic cell subsets: an update. Immunology. 2018;154:3–20. doi: 10.1111/imm.12888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, de Chaisemartin L, Ouakrim H, Becht E, Alifano M, Validire P, Remark R, Hammond SA, Cremer I, Damotte D, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. 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;74:705–15. doi: 10.1158/0008-5472.CAN-13-1342. [DOI] [PubMed] [Google Scholar]
- 82.Wagner F, Hölig U, Wilczkowski F, Plesca I, Sommer U, Wehner R, Kießler M, Jarosch A, Flecke K, Arsova M, Tunger A, Bogner A, Reißfelder C, Weitz J, Schäkel K, Troost EGC, Krause M, Folprecht G, Bornhäuser M, Bachmann MP, Aust D, Baretton G, Schmitz M. Neoadjuvant radiochemotherapy significantly alters the phenotype of plasmacytoid dendritic cells and 6-Sulfo LacNAc+ monocytes in rectal cancer. Front Immunol. 2019;10:602. doi: 10.3389/fimmu.2019.00602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Nagorsen D, Voigt S, Berg E, Stein H, Thiel E, Loddenkemper C. Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival. J Transl Med. 2007;5:62. doi: 10.1186/1479-5876-5-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gulubova MV, Ananiev JR, Vlaykova TI, Yovchev Y, Tsoneva V, Manolova IM. Role of dendritic cells in progression and clinical outcome of colon cancer. Int J Colorectal Dis. 2012;27:159–69. doi: 10.1007/s00384-011-1334-1. [DOI] [PubMed] [Google Scholar]
- 85.Gulubova M, Aleksandrova E, Yovchev Y, Chonov D, Chilingirov P, Vlaykova T. Microsatellite unstable colorectal cancers are associated with increased CD1a- and CD83-positive dendritic cell infiltration. Biotechnol Biotechnol Equip. 2023;37:2266517–2266536. [Google Scholar]
- 86.Väyrynen JP, Sajanti SA, Klintrup K, Mäkelä J, Herzig KH, Karttunen TJ, Tuomisto A, Mäkinen MJ. Characteristics and significance of colorectal cancer associated lymphoid reaction. Int J Cancer. 2014;134:2126–35. doi: 10.1002/ijc.28533. [DOI] [PubMed] [Google Scholar]
- 87.Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, Rabbe N, Laurans L, Tartour E, de Chaisemartin L, Lebecque S, Fridman WH, Cadranel J. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J. Clin. Oncol. 2008;26:4410–7. doi: 10.1200/JCO.2007.15.0284. [DOI] [PubMed] [Google Scholar]
- 88.Morrissey ME, Byrne R, Nulty C, McCabe NH, Lynam-Lennon N, Butler CT, Kennedy S, O’Toole D, Larkin J, McCormick P, Mehigan B, Cathcart MC, Lysaght J, Reynolds JV, Ryan EJ, Dunne MR, O’Sullivan J. The tumour microenvironment of the upper and lower gastrointestinal tract differentially influences dendritic cell maturation. BMC Cancer. 2020;20:566. doi: 10.1186/s12885-020-07012-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Engelhard VH, Rodriguez AB, Mauldin IS, Woods AN, Peske JD, Slingluff CL Jr. Immune cell infiltration and tertiary lymphoid structures as determinants of antitumor immunity. J Immunol. 2018;200:432–442. doi: 10.4049/jimmunol.1701269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Del Prete A, Sozio F, Barbazza I, Salvi V, Tiberio L, Laffranchi M, Gismondi A, Bosisio D, Schioppa T, Sozzani S. Functional role of dendritic cell subsets in cancer progression and clinical implications. Int J Mol Sci. 2020;21:3930. doi: 10.3390/ijms21113930. [DOI] [PMC free article] [PubMed] [Google Scholar]