Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity

This information is current as Victor H. Engelhard, Anthony B. Rodriguez, Ileana S. of September 25, 2021. Mauldin, Amber N. Woods, J. David Peske and Craig L. Slingluff, Jr. J Immunol 2018; 200:432-442; ; doi: 10.4049/jimmunol.1701269

http://www.jimmunol.org/content/200/2/432 Downloaded from

References This article cites 171 articles, 71 of which you can access for free at: http://www.jimmunol.org/content/200/2/432.full#ref-list-1 http://www.jimmunol.org/

Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication by guest on September 25, 2021

*average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2018 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Th eJournal of Brief Reviews Immunology

Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity Victor H. Engelhard,*,† Anthony B. Rodriguez,*,† Ileana S. Mauldin,*,‡ Amber N. Woods,*,† J. David Peske,*,†,1 and Craig L. Slingluff, Jr.*,‡ Limited representation of intratumoral immune cells is a support infiltration of these effectors. The desired approach major barrier to tumor control. However, simply en- would promote a sustained increase in functional intratumoral hancing immune responses in tumor-draining lymph effector cells. nodes or through adoptive transfer may not overcome An interesting alternative is to promote development of an the limited ability of tumor vasculature to support ef- immune response inside the tumor, circumventing the limi- fector infiltration. An alternative is to promote a sus- tations of dendritic cell (DC) trafficking from tumor to LN tained immune response intratumorally. This idea has and of effector cells trafficking in the reverse direction. This Downloaded from gained traction with the observation that many tumors idea originated in studies from the early 2000s, in which are associated with tertiary lymphoid structures (TLS), tumors were engineered to support naive T cell infiltration (11, which organizationally resemble lymph nodes. These 12). However, it has recently gained additional importance peri- and intratumoral structures are usually, but not with the observation that many tumors are associated with tertiary lymphoid structures (TLS). TLS, which resemble LN,

always, associated with positive prognoses in patients. http://www.jimmunol.org/ were initially described in conjunction with chronic and Preclinical and clinical data support a role for TLS in pathogen-driven immune responses. However, they are now modulating immunity in the tumor microenvironment. recognized as a common feature in juxtaposition to tumors However, there appear to be varied functions of TLS, and are often associated with a positive prognosis in patients. potentially based on their structure or location in rela- In this article, we summarize the current state of knowledge tion to the tumor or the origin or location of the tumor about the significance of tumor-associated TLS. We place itself. Understanding more about TLS development, them in the context of immune infiltration into tumors more composition, and function may offer new therapeutic generally, as well as in the context of what is known about

opportunities to modulate antitumor immunity. The the development of conventional LN and inflammation- by guest on September 25, 2021 Journal of Immunology, 2018, 200: 432–442. associated TLS. Finally, we point to the issues that still need to be addressed to harness them for therapeutic pur- poses. ubstantial intratumoral representation of T lymphocytes, either spontaneously or after vaccination or adoptive Tumor-associated vasculature and control of T cell infiltration into tumors therapy, is generally well correlated with immune- S Infiltration of tumors by exogenously activated effectors. The entry of mediated control of human cancers (1–5). Importantly, the leukocytes, including T and B cells, into lymphoid and subset of patients who responds clinically to new-generation nonlymphoid tissues is controlled by sequential engagement immunotherapies is those in whom an immunological infil- of homing receptors (HR) (selectins, receptors, trate is evident prior to treatment (6–10). Thus, enhancing the and integrins) that act with corresponding ligands on representation of intratumoral immune effectors holds the vascular endothelial cells to enable capture, rolling, firm promise of improving clinical outcomes. However, simply adhesion, and extravasation (13–16). During differentiation, enhancing the immune response in the tumor-draining lymph effector T cells acquire the ability to enter peripheral tissues, node (LN) or by infusing large numbers of tumor-reactive including tumors, by upregulating HR that bind to cognate T cells through adoptive transfer may not overcome the limi- ligands expressed on inflamed vasculature. HR expression on tations of the tumor vasculature and microenvironment to activated CD8 T cells is determined by the secondary

*Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Address correspondence and reprint requests to Dr. Victor H. Engelhard, Carter Immu- VA 22908; †Department of Microbiology, Immunology, and Cancer Biology, Univer- nology Center, University of Virginia, Box 801386, Charlottesville, VA 22908. E-mail sity of Virginia School of Medicine, Charlottesville, VA 22908; and ‡Department of address: [email protected] Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908 Abbreviations used in this article: DC, dendritic cell; FDC, follicular DC; HEV, high 1Current address: Department of Pathology, Johns Hopkins University, Baltimore, MD. endothelial venule; HR, homing receptor; LN, lymph node; LTa, -a; LTa b , lymphotoxin-a b ;LTi,lymphoidtissueinducer;LTbR, lymphotoxin-b ORCIDs: 0000-0002-1741-0925 (V.H.E.); 0000-0001-6314-6881 (D.P.). 1 2 1 2 receptor; PNAd, peripheral node addressin; SLO, secondary lymphoid organ; Received for publication September 6, 2017. Accepted for publication October 19, TFH,follicularhelperT;TIL,tumorinfiltrating lymphocyte; TLS, tertiary 2017. lymphoid structure; Treg, regulatory T cell.

This work was supported by United States Public Health Service Grants CA78400 and Ó CA181794 (to V.H.E.) and Training Grants GM007267 (to J.D.P.) and AI007496 (to Copyright 2018 by The American Association of Immunologists, Inc. 0022-1767/18/$35.00 A.B.R. and A.N.W.). J.D.P. was the recipient of a Farrow Fellowship, and A.B.R. was the recipient of a Wagner Fellowship.

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1701269 The Journal of Immunology 433 lymphoid organ (SLO) in which priming occurs (17–21). development of tumor-associated blood vessels that express Tissue-specific and inflammation-induced expression of PNAd and CCL21 (55). PNAd and CCL21 were coex- different HR ligands, in conjunction with the patterns of pressed on ∼5% of blood vessels in tumors growing in HR expressed by T cells, determines which tissues are multiple anatomical locations (55). The primary source of infiltrated. CCL21 was intratumoral gp38+ fibroblasts and, to a lesser Although the requirements for entry of effector T cells and extent, CD31+ blood endothelial cells. It was also shown other leukocytes into inflamed peripheral tissues, particularly that preventing this influx of naive T cells diminished tumor skin and gut, have been well established, the requirements for control, indicating that it represents an important compo- entry into tumors remain inadequately defined. Several studies nent of the immune response in unmanipulated tumors. have unambiguously identified individual HR that mediate Mechanisms driving PNAd and CCL21 expression on tumor- T cell infiltration into some tumors (22–27), whereas others associated vasculature. In adult LN, HEV morphology and ex- have shown correlations between individual HR or HR li- pression of required for biosynthesis of PNAd are gands and T cell infiltrates (28–34). We recently completed a maintained by continuous engagement of lymphotoxin-a1b2 comprehensive analysis of the HR that mediate entry of CD8 (LTa1b2) expressed on DC with the lymphotoxin-b receptor T cell effectors into B16 melanoma and Lewis lung carcinoma (LTbR) expressed on blood endothelial cells (56, 57). and demonstrated that HR ligand expression on tumor- However, the genes for PNAd biosynthesis can also be in- associated vasculature varies with anatomical location of the duced in cultured endothelial cells and monocytes by TNF-a tumor (35). This also determines the ability of T cells acti- (58, 59). LTbR signaling does not control expression of Downloaded from vated in different SLO to enter tumors growing in different CCL21 in adult LN (56). Developmentally, CCL21 locations. Consistent with other work (25, 29, 32, 33, 36– expression can be induced by signals from LTa1b2; from 41), we also found that HR ligand expression on tumor soluble homotrimeric LTa3, which engages TNFR 1 and 2 vasculature is often low. This is consistent with the low in- (TNFR) but not LTbR (60–63); and from TNF-a (64, 65) filtration of adoptively transferred effector T cells observed in and another TNFR family member, CD30 (66). However, it several studies (42–45). Thus, one opportunity to improve is not clear whether these signals regulate the expression of http://www.jimmunol.org/ cancer immunotherapy is to identify and manipulate the ex- CCL21 directly or the development of CCL21-expressing pression of HR and HR ligands to enhance infiltration of cells. Overall, these results point to the central importance CD8 T cell effectors into tumors. This approach has been of LTa1b2–LTbR engagement in driving the development explored by transducing adoptively transferred T cells to ex- and maintenance of PNAd expression on blood endothelial press various chemokine receptors and has resulted in en- cells, whereas control of CCL21 expression remains uncertain. hanced infiltration and tumor control (46–49). In tumors, the density of intratumoral DC (67) and regulatory Naive T cells enter tumors through LN-like vasculature and enhance T cell (Treg) depletion (68) was correlated with development of + + tumor control. Naive T cells enter LN based on their expression PNAd CCL21 vasculature, but a cause and effect relationship by guest on September 25, 2021 of L-selectin and CCR7, which bind to peripheral node was not established. Indeed, in murine models of melanoma and addressin (PNAd) and the CCL19/CCL21, lung carcinoma, development of PNAd+ CCL21+ vasculature respectively. The latter are expressed on specialized LN was not induced by DC or LTbR signaling (55). Instead, in- blood vessels called high endothelial venules (HEV). Because duction of both molecules depended on CD8 T cell effectors naive T cells do not generally enter peripheral tissue, it had that had infiltrated tumors at an earlier point. In i.p. tumors, been assumed that all intratumoral lymphocytes are effectors PNAd expression depended on CD8 T cell effectors secreting that differentiate in tumor-draining LN and home to the LTa3, which acted through TNFR expressed on endothelial tumor thereafter. However, naive T cells were shown to cells and fibroblasts. CCL21, but not PNAd, also depended on infiltrate tumors that had been genetically modified to secrete IFN-g secreted by CD8 T cell effectors, which acted through lymphotoxin-a (LTa) (11, 50) or LIGHT (12). Tumors IFN-g receptors on fibroblasts and endothelial cells. Control of expressing LTa accumulated activated T cells in the absence PNAd and CCL21 in s.c. tumors was more complex, in that of SLO (50). Similar results were obtained by intratumoral CD8 T cell effectors and NK cells could independently induce injection of CCL21 or CCL21-transduced DC (51–53). It expression of these molecules. Although expression depends on was suggested that naive T cells infiltrated through a LN-like signaling through TNFR, IFN-g seems to act redundantly with vasculature based on expression of CCL21 (12) or expression of another unidentified modulator to induce CCL21. These results CCL21 and PNAd and the accumulation of CD62L+ Tcells demonstrate that the mechanisms controlling development of (11), although this was not shown. In all of these models, the PNAd+ CCL21+ vasculature in tumors are distinct from those infiltration of naive T cells, in conjunction with their identified in SLO and also differ between s.c. and i.p. tumors. It differentiation to become effectors, was shown to promote remains to be determined whether these or additional mecha- tumor destruction. These studies established that tumors nisms operate in human tumors growing in different anatomical could be engineered to serve as a surrogate site for naive locations. T cell recruitment and their activation to become mediators of improved antitumor immunity. TLS More recently, we showed that adoptively transferred naive TLS are lymphoid aggregates that frequently develop at sites of CD8 T cells can enter unmanipulated tumors and subse- chronic infection, autoimmune disease, and allograft rejection quently differentiate into functional effectors (54, 55). This (69–78). These structures have considerable morphological, also occurs in the absence of SLO (54). We also showed cellular, and molecular similarity to SLO, particularly LN. directly that naive CD8 T cell entry into tumors was en- Inflammation-associated TLS are associated with blood vascular tirely dependent on L-selectin and CCR7, as well as on the endothelial cells that express PNAd and CCL21, similar to that 434 BRIEF REVIEWS: TUMOR INFILTRATES AND TERTIARY LYMPHOID STRUCTURES of the tumors described above, and often have a characteristic and other investigators (114, 115) have also identified TLS “puffy” morphology. TLS contain B cells, T cells, and DC, in murine tumors. which are typically organized into two distinct functional One significant consideration is how tumor-associated TLS compartments: a B cell follicle primarily composed of naive are distinguished from otherwise localized immune cell infil- B cells, surrounding a germinal center composed of highly trates. In addition to the histological documentation of lym- proliferative B cells, and a T cell area mainly composed of phoid aggregates, which are typically located peritumorally, T cells and DC (77, 79). In SLO, this microarchitecture is most human studies have relied largely or exclusively on the presence of only some TLS-associated elements, including orchestrated by the homeostatic chemokines CCL19, CCL21, + CXCL13, and CXCL12 (80–83). These chemokines are also PNAd HEV (105, 106), mature DC (67, 91–93), B cells found in TLS (84–88). Cells resembling fibroblastic reticular (99, 106), or follicular helper T (TFH) cells (102). A cells and follicular DC (FDC) have also been reported in well- signature that includes CCL19, CCL21, and CXCL13, the homeostatic chemokines that likely organize TLS, also has developed TLS (63, 75, 84, 85, 87), suggesting that they may been used (67, 96, 102, 106, 109, 111). Although it seems be the source of these chemokines and function as organizers of likely that the structures identified by all groups are indeed TLS. However, this remains to be demonstrated directly. TLS, this points to the possibility of significant unappreciated Identification and categorization of tumor-associated TLS. Well- heterogeneity in structure or composition that may arise in organized TLS have been described in association with connection with different tumor types, anatomical locations, several tumor types in humans, and two recent reviews have and primary or metastatic lesions. Downloaded from provided comprehensive summaries of this information (89, In this regard, it was shown that colorectal metastases to the 90). The initial description in non–small cell lung cancers lung were associated with well-developed TLS in humans, (91) documented large peritumoral aggregates of immune whereas renal cell carcinoma metastases to the same organ were cells containing distinct T/DC and B/FDC zones, the latter associated with poorly developed TLS (110). In human of which also contained germinal centers. This group also melanoma, it was found that TLS frequently developed in documented the preferential expression of several homeostatic metastatic lesions yet were largely absent from primary tu- http://www.jimmunol.org/ chemokines and HR ligands in these TLS (92), which mors, despite the presence of a PNAd+ vasculature (95). + correlated with the presence of larger numbers of CD4 Similarly, in the transplantable murine models that we have + CD62L T cells (92, 93). Subsequent studies have largely studied, TLS form around PNAd+ vasculature in all i.p. tu- replicated these findings in a number of tumor types in mors, but not s.c. tumors, despite the presence of comparable humans, although the range of cellular and molecular LN-like vasculature (55, 116) (Fig. 1). The TLS found in markers examined has been variable (67, 94–113). We (55) these murine i.p. tumors also lack discrete B and T cell areas. by guest on September 25, 2021

FIGURE 1. Intratumoral TLS with a nonclassical organization are found in association with PNAd+ vasculature in i.p. (B–D) but not s.c. (A) murine B16-OVA melanoma. Scale bars, 50 mm. The Journal of Immunology 435

Instead, T cells and DC are distributed within large B cell antiviral immunity through activation of naive T cells (71, aggregates that, in turn, are organized within a reticular net- 72). In keeping with this, tumor-associated TLS are usually a work of podoplanin+ fibroblasts (55, 116). In these i.p. favorable prognostic indicator for patient survival (reviewed tumor-associated TLS, it appears that the same podoplanin+ in Refs. 89, 90), although there are exceptions (110, 118). fibroblasts make CCL21 and CXCL13, which may explain Several studies have pointed to the relationship among the lack of discrete B and T cell areas. It remains to be de- the densities of HEV, CD4 or TFH cells, B cells, and termined how this difference in organization relative to more mature DC in defining TLS, as well as to a relationship conventional SLO and TLS affects the nature of the immune among the densities of TLS, levels of intratumoral T and response. B cells, and a TH1/cytotoxic immune profile among tumor A second categorization of tumor-associated TLS is their infiltrating lymphocytes (TIL) (92, 93, 96, 97, 99, 112, anatomical relationship to the tumor mass. Almost all studies 119). These observations are most often interpreted to mean in humans have identified peritumoral TLS (summarized in that tumor-associated TLS are sites for generating useful Ref. 89), and TLS in this location have also been described in antitumor immune responses in newly entering naive T and a genetically engineered mouse model (115). A smaller Bcells. number of human studies have identified tumors with intra- An alternative possibility is that TLS are simply proxies for tumoral TLS, usually as a subset of the tumors with peritumoral more robust intratumoral CD8 T cell effector activity. Because structures, although a largely intratumoral representation has also intratumoral CD8 T cells have been implicated most often as been reported in germ cell tumors (117; summarized in Ref. 89). positive prognostic factors for cancer patient survival, the Downloaded from TheTLSinthetransplantablemurinemodelsthatwehaveused prognostic value of TLS needs to be evaluated through mul- are exclusively intratumoral (55, 116). In studies of human tivariate analysis capable of distinguishing independent effects. melanoma metastases using seven-color immunofluorescence, we One recent study concluded that TLS did have independent have identified TLS in 38% of metastatic melanomas (I.S. value after controlling for specific TIL variables (97). Another Mauldin and C.L. Slingluff, Jr., unpublished observations). study established that a subset of individuals with low TLS-

They appear in three locations: stroma surrounding tumor associated DC density were at great risk for death, despite the http://www.jimmunol.org/ nests (extratumoral), at the periphery of the tumor nests presence of a high density of CD8 effectors (93). Interest- (peritumoral), and intratumoral. In general, TLS are more ingly,thepresenceofTLSintriple-negativebreastcancer organized with discrete B cell and DC/T cell zones in peritu- added to the positive prognostic value of moderate levels of moral and extratumoral locations (Fig. 2A–D) than within the TIL, but it had no impact if there were high levels of TIL tumor nests (Fig. 2E, 2F). However, some peritumoral TLS also present (105). At the other extreme, TLS had positive show an intermixing of T cells within B cell aggregates (Fig. 2C). prognosticvalueinMerkelcellcarcinoma,whereasTcell FOXP3+ putative Treg are prevalent in the T cell area. We have infiltration and several other immunological parameters did + also detected intratumoral PNAd vasculature that is not asso- not (119). Similarly, neither CD8 nor TH17 TIL was as- by guest on September 25, 2021 ciated with well-organized aggregates, although there are diffuse sociated with a positive prognosis in gastric cancer patients; distributions of lymphocytes and DC in the general vicinity however, Tbet+ cells, CD20 B cells organized into TLS, and (Fig.2F).Animportantsetofquestionsiswhethertheselevelsof coordinate expression of TH1 and B cell genes were associ- TLS organization in different locations are formed by distinct ated with a positive prognosis (103). Curiously, however, mechanisms, have distinct immunological properties, or have DC–Lamp+ cell density, another TLS marker, did not have distinct prognostic values in relation to patient survival and re- prognostic value. sponse to immunotherapy. Conversely, a limited number of studies have shown that Prognostic significance and immunological impact of tumor- tumor-associated TLS are a negative prognostic factor for associated TLS. TLS are often found in lungs in response to cancer patient survival. The presence of TLS in colorectal acute infection (77, 78) where they lead to enhanced local cancer has been associated with more advanced disease (118),

FIGURE 2. Multispectral imaging of TLS in human melanoma metastases localized outside the tumor area (A), peritumorally at the peripheral edge of tumor (B–D), or intratumorally (E and F). Artifactual tissue creases due to sectioning can be seen in (B) and (C). Scale bars, 100 mm. 436 BRIEF REVIEWS: TUMOR INFILTRATES AND TERTIARY LYMPHOID STRUCTURES whereas TLS in breast cancer was associated with a higher Mechanisms that control the development of TLS. The develop- tumor grade, low representation of intratumoral immune ment of conventional LN depends on interaction between cells, and a high frequency of LN metastasis (120). These CD4+ CD3neg lymphoid tissue inducer (LTi) cells expressing studies are seemingly at odds with studies suggesting positive LTa1b2 and RANKL [now known to be group 3 innate associations with patient survival in each of these diseases (67, lymphoid cells (133)] and mesenchymal lymphoid tissue or- 97, 102, 105–107, 110, 112) and remain to be fully recon- ganizer cells expressing LTbR (134–139). This leads to ex- ciled. An intriguing study showed a negative impact of DC- pression of CCL19, CCL21, and CXCL13, which attract Lamp and CD8 markers on survival of patients with renal cell additional LTi cells, and the recruitment and positioning of T carcinoma metastases to lung but a positive impact on pa- and B cells (81, 140–142). In keeping with this, mice tients with colorectal cancer metastases to the same organ deficient in LTa,LTb,orLTbR fail to develop all or most (110). In murine lung adenocarcinoma, it was shown that LN (143–146). The formation of FDC networks and TLS serve as a site for recruitment of Treg that restrain an- germinal centers as an element of LN development depends titumor immune responses (115). It was also shown that TLS on engagement of TNFR, as well as LTbR (147). LTbR can function as niches for malignant hepatocellular progenitor signaling does not control expression of CCL21 in adult cells (98). Interestingly, in human lung transplants, TLS have LN (56), although it does maintain CXCL13 expression been associated with a lower probability of graft rejection, in adult spleen (148), presumably through interaction of + whereas some other organ transplants are more likely to be LTa1b2-expressing B cells with LTbR FDC (149, 150). rejected when TLS are identified (121–123). Thus, in cancers A key question is whether the formation of TLS in general, Downloaded from and in transplanted organs, immune rejection may be en- and tumor-associated TLS in particular, occurs through hanced or reduced by the presence of TLS. This raises the pathways analogous to those controlling conventional LN possibility that some TLS are dominated by Treg and support development. Transgenic expression of LTa or LTb in tolerance, whereas others support immune rejection. It will be variousorgansleadstotheformation of organized lym- important to understand details of the composition and lo- phoidaggregatestypicallyidentifiedasTLS,althoughthey cation of TLS and which is more relevant for immune re- are not driven by an immune response (60–63). Transgenic http://www.jimmunol.org/ jection. expression of CCL21 also leads to formation of such A characteristic feature of all tumor-associated TLS in structures, although PNAd expression still depends on humans and mice is the presence of a large number of LTbR signaling (151, 152), and this operates through B lymphocytes, often accompanied by TFH cells. In recent mature CD4 T cells rather than LTi cells (153). Impor- years, the consensus of the field has become that CD8 tantly, blockade of LTbR signaling prevents immune re- T cells are the most important determinant of antitumor sponse–drivenTLSformationinthyroid,lung,salivary immunity; however, these observations suggest that intra- gland, aorta, and heart (71, 88, 154–157). These studies

tumoral B cells may play an important role(s). On the suggest that development of LN and TLS is mechanistically by guest on September 25, 2021 positive side, several studies have suggested that intra- similar. tumoral B cells can capture and present tumor Ags to In contrast, TLS have been shown to depend on TNF-a in T cells (124–126). Perhaps even more heretical (in the aorta, fat, and intestine (158–160), IL-22 in salivary gland current environment) have been suggestions that intra- (84), and IL-17A in lung and meninges (86, 88, 161). IL-6 tumoral Ab responses can be an effective component of and IL-23 also contribute to TLS development through IL- antitumor immunity (127, 128). On the negative side, 17A–dependent and -independent pathways (162–164). Al- many studies have shown that B cells can play a suppressive though the exact mechanisms of action remain to be fully role in antitumor immunity, and regulatory B cells have defined, TNF-a induces CCL21 (64, 65), CXCL13 (148), been found to accumulate in several tumor models (129– and enzymes necessary for PNAd synthesis (58, 59), whereas 132). The differences in prognostic significance of tumor- IL-22 and IL-17A induce CXCL12 and CXCL13 in TLS- associated TLS may lie, at least in part, in the distinct associated stromal cells (84, 85, 88, 165, 166). These cyto- functional attributes of the B cell compartment in different kines sometimes induce TLS independent of LTbR signaling tumors. However, the ways in which tumors may regulate (158, 160), but in other instances they act cooperatively, ei- these functional attributes remain to be discovered. ther by inducing distinct aspects of TLS structure (84, 86) or Collectively, these studies point to a complex interplay in by acting at the TLS-induction or maintenance phases (88). which the prognostic value of TIL and TLS depends at the very Importantly, different pathogens can induce lung-associated least on tumor type. It seems likely that different tumor types TLS by distinct cellular and molecular pathways (85). The may drive qualitatively distinct immune responses, distin- involvement of additional in the formation and/or guishable by their effector and regulatory mechanisms. Such maintenance of many LTbR-dependent TLS remains to be responses may also evolve over time. In some tumors, or at evaluated. certain times, these may be determined largely within con- Aside from the involvement of distinct molecular signaling ventional tumor-draining SLO, whereas in other circum- pathways in TLS formation, a variety of inducing cells, in lieu stances, they may be determined within tumor-associated TLS. of LTi cells, have been identified. This includes DC (71, 72, Evidence demonstrating such a temporal evolution has recently 167) and naive B cells (168), both expressing LTa1b2; been reported (94). This suggests that TLS “quality” might macrophages (158–160), TH17 cells (86, 88, 161), NKT cells also confer, or result from, distinct kinds of immunity or (158), gd T cells (85), and multiple populations of IL-22– immune regulation. Genetically engineered mouse models secreting adaptive and innate lymphoid cells (84). Overall, it (115) provide a valuable opportunity to evaluate these vari- should be expected that distinct molecular signals, conveyed ables systematically. by immune cells with pleiotropic regulatory signatures, will The Journal of Immunology 437 induce TLS with important differences in the immune re- which are important for the development and survival of sponses that they support. B cells (170–172) and maintaining the integrity of germinal In this context, the mechanisms driving TLS formation in centers (173, 174). This suggests that elements of the i.p. tumors remain almost entirely unknown. Analogous to the tumor microenvironment induce proliferation and differen- overexpression models described above, transgenic expression of tiation of podoplanin+ cells, which act as scaffolding for TLS LTa or LIGHT in tumors (11, 169) or intratumoral admin- formation. Although our work establishes that CCL21 re- istration of CCL21 (52, 53) establishes that these pathways can cruits naive T cells into i.p. and s.c. tumors (55), it remains to lead to TLS formation. In transplantable murine tumors, TLS be directly determined whether and how these remaining develop in i.p., but not in s.c., tumors (55, 116), suggesting that molecules impact B cell recruitment and the formation of the mechanisms controlling their development depend on an- TLS in i.p. tumors and what mechanism is responsible for atomical environmental factors and providing an opportunity to changes in their expression in tumor-associated podoplanin+ gain insight into these mechanisms through comparative anal- fibroblasts (Fig. 3). ysis. Because PNAd+ CCL21+ vasculature develops comparably in tumors growing in both locations, this also points to distinct Conclusions mechanisms controlling these two features, as well as to this Tumor-associated TLS have been frequently correlated with vasculature in providing a nucleation site for TLS formation. improved patient survival and in murine models have generally Compared with s.c. tumors, i.p. tumors showed a 10-fold been associated with the development of improved immune- + increase in the number of B cells relative to CD31 vascular mediated tumor control. However, tumor-associated TLS have Downloaded from endothelial cells, whereas the numbers of T cells and DC were also been identified as a locus of Treg accumulation, and they similar (116). Interestingly, i.p. tumors also showed a 5-fold characteristically contain large numbers of B cells, which exert relative increase in podoplanin+ fibroblasts, and these cells a regulatory role in several tumor models. Much remains to be formed a reticular network surrounding the PNAd+ CCL21+ done to fully understand the immune mechanisms that are vasculature, which was coextensive with the B cells (55, 116). activated within these structures and whether these vary based + Although perivascular podoplanin cells were evident in s.c. on the origin of the tumor, the oncogenic drivers leading to its http://www.jimmunol.org/ tumors, they had not expanded to form reticular networks. formation, or its anatomical location as a primary tumor or Also, podoplanin+ cells in i.p. tumors expressed significantly metastatic lesion. To do so will require application of the full higher levels of CXCL13 and the B cell survival factors BAFF range of technologies that have been applied in previous and APRIL (A.B. Rodriguez, J.D. Peske, S. Cyranowski, G. studies, together with a more comprehensive analysis of Parriott, and V.H. Engelhard, manuscript in preparation), molecules that mark various immune mechanisms. Advances by guest on September 25, 2021

FIGURE 3. Model for the formation of nonclassical and classical tumor-associated TLS. 438 BRIEF REVIEWS: TUMOR INFILTRATES AND TERTIARY LYMPHOID STRUCTURES in multispectral imaging of single sections, and entire tu- novo antitumor immune responses? As new immune thera- mors after tissue clearing, and advances in small-scale RNA peutic modalities (e.g., IDO inhibitors, myeloid cell depletion) sequencing combined with laser capture dissection of TLS come into the clinical landscape, these questions will only get create new opportunities for comprehensive assessment of broader. cellular composition and functional state of individual TLS in Overall, the work accomplished to date clearly establishes the a variety of contexts. This work is also important as a means to importance of tumor-associated TLS as an aspect of immu- more clearly establish the prognostic value of TLS, indepen- nological control of tumors. However, a substantial amount of dent of other immune factors. work remains to be done to fully understand how they are The pathways that drive the formation of PNAD+ CCL21+ important, as well as how to begin to use them as an element vasculature have been uncovered in a limited number of of our developing armamentarium of immunological cancer murine tumor models, but the factors that drive formation control strategies. and/or maintenance of tumor-associated TLS remain almost completely unknown. Studies in a variety of inflammation- Disclosures driven TLS models have pointed to a number of molecular The authors have no financial conflicts of interest. and cellular mediators that work in collaboration with or in lieu of more classical mechanisms that underlie the formation References of SLO. This heterogeneity has been shown to reflect, at least 1. Page`s, F., A. Berger, M. Camus, F. Sanchez-Cabo, A. Costes, R. Molidor, in part, the nature of the inflammatory stimulus. One might B. Mlecnik, A. Kirilovsky, M. Nilsson, D. Damotte, et al. 2005. Effector memory Downloaded from T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med. 353: also imagine a role for anatomical location, reflecting the 2654–2666. extent to which peripheral tissues are invested with different 2. Galon, J., A. Costes, F. Sanchez-Cabo, A. Kirilovsky, B. Mlecnik, C. Lagorce- Page`s, M. Tosolini, M. Camus, A. Berger, P. Wind, et al. 2006. Type, density, and subsets of myeloid derived cells, innate lymphoid cells, or location of immune cells within human colorectal tumors predict clinical outcome. tissue-resident memory cells. The impact of the pre-existing Science 313: 1960–1964. or early-stage microenvironment in proximity to tumors 3. Sato, E., S. H. Olson, J. Ahn, B. Bundy, H. Nishikawa, F. Qian, A. A. Jungbluth, D. Frosina, S. Gnjatic, C. Ambrosone, et al. 2005. Intraepithelial CD8+ tumor- on TLS formation is clear, but the exact drivers and how infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated http://www.jimmunol.org/ they vary based on tumor origin or location remain to be with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA 102: 18538– 18543. determined. These drivers are likely to also influence the 4.Erdag,G.,J.T.Schaefer,M.E.Smolkin,D.H.Deacon,S.M.Shea, composition and function of TLS, as mentioned above. Un- L. T. Dengel, J. W. Patterson, and C. L. Slingluff, Jr. 2012. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are as- derstanding these driver mechanisms will rely heavily on ap- sociated with clinical outcome in metastatic melanoma. Cancer Res. 72: 1070– propriate transplantable and genetically induced mouse 1080. 5. Angell, H., and J. Galon. 2013. From the immune contexture to the immuno- models, in which they can be directly tested through knock- score: the role of prognostic and predictive immune markers in cancer. Curr. Opin. out or blockade approaches. Their validity in human settings Immunol. 25: 261–267. 6. Gajewski, T. F., J. Louahed, and V. G. Brichard. 2010. Gene signature in mela- will be more difficult to establish, because it will depend by guest on September 25, 2021 noma associated with clinical activity: a potential clue to unlock cancer immu- largely on correlative studies to establish the presence or ab- notherapy. Cancer J. 16: 399–403. sence of various cells or signaling molecules. However, as 7. Ulloa-Montoya, F., J. Louahed, B. Dizier, O. Gruselle, B. Spiessens, F. F. Lehmann, S. Suciu, W. H. Kruit, A. M. Eggermont, J. Vansteenkiste, and argued above, incisive multiparameter correlative studies are V. G. Brichard. 2013. Predictive gene signature in MAGE-A3 antigen-specific key to understanding the prognostic significance of TLS in cancer immunotherapy. J. Clin. Oncol. 31: 2388–2395. 8. Ji, R.-R., S. D. Chasalow, L. Wang, O. Hamid, H. Schmidt, J. Cogswell, humans. Together with work in mouse models, they will S. Alaparthy, D. Berman, M. Jure-Kunkel, N. O. Siemers, et al. 2012. An provide the basis for moving forward to manipulate TLS immune-active tumor microenvironment favors clinical response to ipilimumab. incidence, location, composition, and function. Cancer Immunol. Immunother. 61: 1019–1031. 9. Herbst, R. S., J.-C. Soria, M. Kowanetz, G. D. Fine, O. Hamid, M. S. Gordon, Based on the preponderance of evidence, it seems highly J. A. Sosman, D. F. McDermott, J. D. Powderly, S. N. Gettinger, et al. 2014. desirable, in most settings, to increase the number of TLS as a Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563–567. way to promote an ongoing immune response in immediate 10. Gajewski, T. F., H. Schreiber, and Y.-X. Fu. 2013. Innate and adaptive immune proximity to tumor cells, avoiding the need for two-way cells in the tumor microenvironment. Nat. Immunol. 14: 1014–1022. 11. Schrama, D., P. thor Straten, W. H. Fischer, A. D. McLellan, E. B. Bro¨cker, cellular traffic between tumor and tumor-draining SLO. R. A. Reisfeld, and J. C. Becker. 2001. Targeting of lymphotoxin-a to the tumor One open question is whether this is best achieved via elicits an efficient immune response associated with induction of peripheral intratumoral or peritumoral TLS. Related to this is the lymphoid-like tissue. Immunity 14: 111–121. 12. Yu, P., Y. Lee, W. Liu, R. K. Chin, J. Wang, Y. Wang, A. Schietinger, M. Philip, question of whether regulatory mechanisms that often occur H. Schreiber, and Y. X. Fu. 2004. Priming of naive T cells inside tumors leads to within the tumor microenvironment are more prevalent eradication of established tumors. Nat. Immunol. 5: 141–149. 13. Butcher, E. C., and L. J. Picker. 1996. Lymphocyte homing and homeostasis. within, or in proximity to, TLS. A second set of questions Science 272: 60–66. concerns TLS functionality. It remains to be established how 14. von Andrian, U. H., and C. R. Mackay. 2000. T-cell function and migration. Two sides of the same coin. N. Engl. J. Med. 343: 1020–1034. TLS and tumor-draining SLO compare as sites for develop- 15. Sackstein, R. 2005. The lymphocyte homing receptors: gatekeepers of the multi- ment of tumor-specific CD8 T cells, either numerically or over step paradigm. Curr. Opin. Hematol. 12: 444–450. 16. Ley, K., C. Laudanna, M. I. Cybulsky, and S. Nourshargh. 2007. Getting to the time. The presence of large numbers of B cells in TLS has site of inflammation: the leukocyte adhesion cascade updated. Nat. Rev. Immunol. brought renewed focus on the positive and negative roles that 7: 678–689. B cells might play in antitumor immunity, but this has yet to be 17. Mora, J. R., M. R. Bono, N. Manjunath, W. Weninger, L. L. Cavanagh, M. Rosemblatt, and U. H. Von Andrian. 2003. Selective imprinting of gut- clearly tied to the activity of B cells within TLS. Finally, but homing T cells by Peyer’s patch dendritic cells. Nature 424: 88–93. more immediately in the current clinical environment, there 18. Mora, J. R., G. Cheng, D. Picarella, M. Briskin, N. Buchanan, and U. H. von Andrian. 2005. Reciprocal and dynamic control of CD8 T cell homing by den- are open questions about whether the presence of TLS, either dritic cells from skin- and gut-associated lymphoid tissues. J. Exp. Med. 201: 303– pre- or posttreatment, has prognostic value in responses to 316. 19. Johansson-Lindbom, B., and W. W. Agace. 2007. Generation of gut-homing checkpoint blockade therapy. Does checkpoint blockade en- T cells and their localization to the small intestinal mucosa. Immunol. Rev. 215: hance TLS formation or enhance TLS function as a site for de 226–242. The Journal of Immunology 439

20. Sigmundsdottir, H., and E. C. Butcher. 2008. Environmental cues, dendritic cells of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and and the programming of tissue-selective lymphocyte trafficking. Nat. Immunol. 9: peripheral blood lymphocytes in patients with metastatic melanoma. J. Natl. Cancer 981–987. Inst. 81: 1709–1717. 21. Ferguson, A. R., and V. H. Engelhard. 2010. CD8 T cells activated in distinct 43. Fisher, B., B. S. Packard, E. J. Read, J. A. Carrasquillo, C. S. Carter, lymphoid organs differentially express adhesion and coexpress multiple S. L. Topalian, J. C. Yang, P. Yolles, S. M. Larson, and S. A. Rosenberg. 1989. chemokine receptors. J. Immunol. 184: 4079–4086. Tumor localization of adoptively transferred indium-111 labeled tumor 22. Yamada, M., K. Yanaba, M. Hasegawa, Y. Matsushita, M. Horikawa, K. Komura, infiltrating lymphocytes in patients with metastatic melanoma. J. Clin. Oncol. 7: T. Matsushita, A. Kawasuji, T. Fujita, K. Takehara, et al. 2006. Regulation of local 250–261. and metastatic host-mediated anti-tumour mechanisms by L-selectin and inter- 44. Economou, J. S., A. S. Belldegrun, J. Glaspy, E. M. Toloza, R. Figlin, J. Hobbs, cellular adhesion molecule-1. Clin. Exp. Immunol. 143: 216–227. N. Meldon, R. Kaboo, C. L. Tso, A. Miller, et al. 1996. In vivo trafficking of 23. Sasaki, K., X. Zhu, C. Vasquez, F. Nishimura, J. E. Dusak, J. Huang, M. Fujita, adoptively transferred -2 expanded tumor-infiltrating lymphocytes and A. Wesa, D. M. Potter, P. R. Walker, et al. 2007. Preferential expression of very peripheral blood lymphocytes. Results of a double gene marking trial. J. Clin. late antigen-4 on type 1 CTL cells plays a critical role in trafficking into central Invest. 97: 515–521. nervous system tumors. Cancer Res. 67: 6451–6458. 45. Kershaw, M. H., J. A. Westwood, L. L. Parker, G. Wang, Z. Eshhar, 24. Buckanovich, R. J., A. Facciabene, S. Kim, F. Benencia, D. Sasaroli, K. Balint, S. A. Mavroukakis, D. E. White, J. R. Wunderlich, S. Canevari, L. Rogers-Freezer, D. Katsaros, A. O’Brien-Jenkins, P. A. Gimotty, and G. Coukos. 2008. Endo- et al. 2006. A phase I study on adoptive immunotherapy using gene-modified thelin B receptor mediates the endothelial barrier to T cell homing to tumors and T cells for ovarian cancer. Clin. Cancer Res. 12: 6106–6115. disables immune therapy. Nat. Med. 14: 28–36. 46. Craddock, J. A., A. Lu, A. Bear, M. Pule, M. K. Brenner, C. M. Rooney, and 25. Fisher, D. T., Q. Chen, J. J. Skitzki, J. B. Muhitch, L. Zhou, M. M. Appenheimer, A. E. Foster. 2010. Enhanced tumor trafficking of GD2 chimeric antigen receptor T. D. Vardam, E. L. Weis, J. Passanese, W.-C. Wang, et al. 2011. IL-6 trans- T cells by expression of the chemokine receptor CCR2b. J. Immunother. 33: 780– signaling licenses mouse and human tumor microvascular gateways for trafficking 788. of cytotoxic T cells. J. Clin. Invest. 121: 3846–3859. 47. Di Stasi, A., B. De Angelis, C. M. Rooney, L. Zhang, A. Mahendravada, 26. Bose, A., J. L. Taylor, S. Alber, S. C. Watkins, J. A. Garcia, B. I. Rini, J. S. Ko, A. E. Foster, H. E. Heslop, M. K. Brenner, G. Dotti, and B. Savoldo. 2009. P. A. Cohen, J. H. Finke, and W. J. Storkus. 2011. Sunitinib facilitates the ac- T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting tivation and recruitment of therapeutic anti-tumor immunity in concert with CD30 have improved homing and antitumor activity in a Hodgkin tumor model. specific vaccination. Int. J. Cancer 129: 2158–2170. Blood 113: 6392–6402. Downloaded from 27. Mikucki, M. E., D. T. Fisher, J. Matsuzaki, J. J. Skitzki, N. B. Gaulin, 48. Peng, W., Y. Ye, B. A. Rabinovich, C. Liu, Y. Lou, M. Zhang, M. Whittington, J. B. Muhitch, A. W. Ku, J. G. Frelinger, K. Odunsi, T. F. Gajewski, et al. 2015. Y. Yang, W. W. Overwijk, G. Lize´e, and P. Hwu. 2010. Transduction of tumor- Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell specific T cells with CXCR2 chemokine receptor improves migration to tumor and trafficking across tumour vascular checkpoints. Nat. Commun. 6: 7458. antitumor immune responses. Clin. Cancer Res. 16: 5458–5468. 28. Kunz, M., A. Toksoy, M. Goebeler, E. Engelhardt, E. Bro¨cker, and R. Gillitzer. 49. Moon, E. K., C. Carpenito, J. Sun, L.-C. S. Wang, V. Kapoor, J. Predina, 1999. Strong expression of the lymphoattractant C-X-C chemokine Mig is asso- D. J. Powell, Jr., J. L. Riley, C. H. June, and S. M. Albelda. 2011. Expression of a ciated with heavy infiltration of T cells in human malignant melanoma. J. Pathol. functional CCR2 receptor enhances tumor localization and tumor eradication by

189: 552–558. retargeted human T cells expressing a mesothelin-specific chimeric antibody re- http://www.jimmunol.org/ 29. Garbi, N., B. Arnold, S. Gordon, G. J. Ha¨mmerling, and R. Ganss. 2004. CpG ceptor. Clin. Cancer Res. 17: 4719–4730. motifs as proinflammatory factors render autochthonous tumors permissive for 50. Schrama, D., H. Voigt, A. O. Eggert, R. Xiang, H. Zhou, T. N. M. Schumacher, infiltration and destruction. J. Immunol. 172: 5861–5869. M. H. Andersen, P. thor Straten, R. A. Reisfeld, and J. C. Becker. 2008. Im- 30. Hensbergen, P. J., P. G. Wijnands, M. W. Schreurs, R. J. Scheper, R. Willemze, munological tumor destruction in a murine melanoma model by targeted LTalpha and C. P. Tensen. 2005. The CXCR3 targeting chemokine CXCL11 has potent independent of secondary lymphoid tissue. Cancer Immunol. Immunother. 57: 85– antitumor activity in vivo involving attraction of CD8+ T lymphocytes but not 95. inhibition of angiogenesis. J. Immunother. 28: 343–351. 51. Kirk, C. J., D. Hartigan-O’Connor, B. J. Nickoloff, J. S. Chamberlain, 31. Musha, H., H. Ohtani, T. Mizoi, M. Kinouchi, T. Nakayama, K. Shiiba, M. Giedlin, L. Aukerman, and J. J. Mule´. 2001. T cell-dependent antitumor K. Miyagawa, H. Nagura, O. Yoshie, and I. Sasaki. 2005. Selective infiltration of immunity mediated by secondary lymphoid tissue chemokine: augmentation of CCR5+CXCR3+ T lymphocytes in human colorectal carcinoma. Int. J. Cancer dendritic cell-based immunotherapy. Cancer Res. 61: 2062–2070. 116: 949–956. 52. Kirk, C. J., D. Hartigan-O’Connor, and J. J. Mule´. 2001. The dynamics of the

32. Clark, R. A., S. J. Huang, G. F. Murphy, I. G. Mollet, D. Hijnen, M. Muthukuru, T-cell antitumor response: chemokine-secreting dendritic cells can prime tumor- by guest on September 25, 2021 C. F. Schanbacher, V. Edwards, D. M. Miller, J. E. Kim, et al. 2008. Human reactive T cells extranodally. Cancer Res. 61: 8794–8802. squamous cell carcinomas evade the immune response by down-regulation of 53. Turnquist, H. R., X. Lin, A. E. Ashour, M. A. Hollingsworth, R. K. Singh, vascular E-selectin and recruitment of regulatory T cells. J. Exp. Med. 205: 2221– J. E. Talmadge, and J. C. Solheim. 2007. CCL21 induces extensive intratumoral 2234. immune cell infiltration and specific anti-tumor cellular immunity. Int. J. Oncol. 33. Quezada, S. A., K. S. Peggs, T. R. Simpson, Y. Shen, D. R. Littman, and 30: 631–639. J. P. Allison. 2008. Limited tumor infiltration by activated T effector cells restricts 54. Thompson, E. D., H. L. Enriquez, Y.-X. Fu, and V. H. Engelhard. 2010. Tumor the therapeutic activity of regulatory T cell depletion against established mela- masses support naive T cell infiltration, activation, and differentiation into effec- noma. J. Exp. Med. 205: 2125–2138. tors. J. Exp. Med. 207: 1791–1804. 34. Lohr, J., T. Ratliff, A. Huppertz, Y. Ge, C. Dictus, R. Ahmadi, S. Grau, 55. Peske, J. D., E. D. Thompson, L. Gemta, R. A. Baylis, Y.-X. Fu, and N. Hiraoka, V. Eckstein, R. C. Ecker, et al. 2011. Effector T-cell infiltration V. H. Engelhard. 2015. Effector lymphocyte-induced lymph node-like vasculature positively impacts survival of glioblastoma patients and is impaired by tumor- enables naive T-cell entry into tumours and enhanced anti-tumour immunity. Nat. derived TGF-b. Clin. Cancer Res. 17: 4296–4308. Commun. 6: 7114. 35. Woods, A. N., A. L. Wilson, N. Srinivasan, J. Zeng, A. Dutta, J. D. Peske, 56. Browning, J. L., N. Allaire, A. Ngam-Ek, E. Notidis, J. Hunt, S. Perrin, and E. F. Tewalt, R. K. Gregg, A. R. Ferguson, and V. H. Engelhard. 2017. Ana- R. A. Fava. 2005. Lymphotoxin-beta receptor signaling is required for the ho- tomical location and effector cytokines determine homing receptor ligand ex- meostatic control of HEV differentiation and function. Immunity 23: 539–550. pression on tumor-associated vasculature and the entry of effector CD8 T-cells. 57. Moussion, C., and J.-P. Girard. 2011. Dendritic cells control lymphocyte entry to Cancer Immunol. Res. DOI: 10.1158/2326-6066.CIR-17-0190. lymph nodes through high endothelial venules. Nature 479: 542–546. 36. Weishaupt, C., K. N. Munoz, E. Buzney, T. S. Kupper, and R. C. Fuhlbrigge. 2007. T-cell distribution and adhesion receptor expression in metastatic mela- 58. Pablos, J. L., B. Santiago, D. Tsay, M. S. Singer, G. Palao, M. Galindo, and noma. Clin. Cancer Res. 13: 2549–2556. S. D. Rosen. 2005. A HEV-restricted sulfotransferase is expressed in rheumatoid 37. Dengel, L. T., A. G. Norrod, B. L. Gregory, E. Clancy-Thompson, arthritis synovium and is induced by lymphotoxin-a/b and TNF-a in cultured M. D. Burdick, R. M. Strieter, C. L. Slingluff, Jr., and D. W. Mullins. 2010. endothelial cells. BMC Immunol. 6: 6. induce CXCR3-cognate chemokine production by human metastatic 59. Tjew, S. L., K. L. Brown, R. Kannagi, and P. Johnson. 2005. Expression of melanoma. J. Immunother. 33: 965–974. N-acetylglucosamine 6-O-sulfotransferases (GlcNAc6STs)-1 and -4 in human monocytes: GlcNAc6ST-1 is implicated in the generation of the 6-sulfo 38. Enarsson, K., E. Johnsson, C. Lindholm, A. Lundgren, Q. Pan-Hammarstro¨m, N-acetyllactosamine/Lewis x epitope on CD44 and is induced by TNF-a. E. Stro¨mberg, P. Bergin, E.-L. Baunge, A.-M. Svennerholm, and M. Quiding- Glycobiology 15: 7C–13C. Ja¨rbrink. 2006. Differential mechanisms for T lymphocyte recruitment in normal 60. Sacca, R., C. A. Cuff, W. Lesslauer, and N. H. Ruddle. 1998. Differential activities and neoplastic human gastric mucosa. Clin. Immunol. 118: 24–34. of secreted lymphotoxin-a3 and membrane lymphotoxin-a1b2 in lymphotoxin- 39. Dirkx, A. E. M., M. G. A. oude Egbrink, M. J. E. Kuijpers, S. T. van der Niet, induced inflammation: critical role of TNF receptor 1 signaling. J. Immunol. 160: V. V. T. Heijnen, J. C. A. Bouma-ter Steege, J. Wagstaff, and A. W. Griffioen. 485–491. 2003. Tumor angiogenesis modulates leukocyte-vessel wall interactions in vivo by reducing endothelial adhesion molecule expression. Cancer Res. 63: 2322–2329. 61. Cuff, C. A., J. Schwartz, C. M. Bergman, K. S. Russell, J. R. Bender, and 40. Yoong, K. F., G. McNab, S. G. Hubscher,€ and D. H. Adams. 1998. Vascular N. H. Ruddle. 1998. Lymphotoxin a3 induces chemokines and adhesion mole- adhesion -1 and ICAM-1 support the adhesion of tumor-infiltrating cules: insight into the role of LT a in inflammation and lymphoid organ devel- lymphocytes to tumor endothelium in human hepatocellular carcinoma. opment. J. Immunol. 161: 6853–6860. J. Immunol. 160: 3978–3988. 62. Cuff, C. A., R. Sacca, and N. H. Ruddle. 1999. Differential induction of adhesion 41. Harlin, H., Y. Meng, A. C. Peterson, Y. Zha, M. Tretiakova, C. Slingluff, molecule and chemokine expression by LTalpha3 and LTalphabeta in inflamma- M. McKee, and T. F. Gajewski. 2009. Chemokine expression in melanoma me- tion elucidates potential mechanisms of mesenteric and peripheral lymph node tastases associated with CD8+ T-cell recruitment. Cancer Res. 69: 3077–3085. development. J. Immunol. 162: 5965–5972. 42.Griffith,K.D.,E.J.Read,J.A.Carrasquillo,C.S.Carter,J.C.Yang,B.Fisher, 63. Drayton, D. L., X. Ying, J. Lee, W. Lesslauer, and N. H. Ruddle. 2003. Ectopic P. Aebersold, B. S. Packard, M. Y. Yu, and S. A. Rosenberg. 1989. In vivo distribution LT abdirects lymphoid organ neogenesis with concomitant expression of peripheral 440 BRIEF REVIEWS: TUMOR INFILTRATES AND TERTIARY LYMPHOID STRUCTURES

node addressin and a HEV-restricted sulfotransferase. J. Exp. Med. 197: 1153– 2011. The development of inducible bronchus-associated lymphoid tissue depends 1163. on IL-17. Nat. Immunol. 12: 639–646. 64. Johnson, L. A., and D. G. Jackson. 2010. Inflammation-induced secretion of 89. Hiraoka, N., Y. Ino, and R. Yamazaki-Itoh. 2016. Tertiary lymphoid organs in CCL21 in lymphatic endothelium is a key regulator of integrin-mediated dendritic cancer tissues. Front. Immunol. 7: 244. cell transmigration. Int. Immunol. 22: 839–849. 90. Saute`s-Fridman, C., M. Lawand, N. A. Giraldo, H. Kaplon, C. Germain, 65. Wiede, F., K. Vana, L. M. Sedger, A. Lechner, and H. Ko¨rner. 2007. TNF- W. H. Fridman, and M.-C. Dieu-Nosjean. 2016. Tertiary lymphoid structures in dependent overexpression of CCL21 is an underlying cause of progressive lym- cancers: prognostic value, regulation, and manipulation for therapeutic interven- phoaccumulation in generalized lymphoproliferative disorder. Eur. J. Immunol. 37: tion. Front. Immunol. 7: 407. 351–357. 91. Dieu-Nosjean, M.-C., M. Antoine, C. Danel, D. Heudes, M. Wislez, V. Poulot, 66. Bekiaris, V., F. Gaspal, M.-Y. Kim, D. R. Withers, F. M. McConnell, N. Rabbe, L. Laurans, E. Tartour, L. de Chaisemartin, et al. 2008. Long-term G. Anderson, and P. J. L. Lane. 2009. CD30 is required for CCL21 expression and survival for patients with non-small-cell lung cancer with intratumoral lymphoid CD4 T cell recruitment in the absence of lymphotoxin signals. J. Immunol. 182: structures. J. Clin. Oncol. 26: 4410–4417. 4771–4775. 92. de Chaisemartin, L., J. Goc, D. Damotte, P. Validire, P. Magdeleinat, M. Alifano, 67. Martinet, L., T. Filleron, S. Le Guellec, P. Rochaix, I. Garrido, and J.-P. Girard. I. Cremer, W.-H. Fridman, C. Saute`s-Fridman, and M.-C. Dieu-Nosjean. 2011. 2013. High endothelial venule blood vessels for tumor-infiltrating lymphocytes are Characterization of chemokines and adhesion molecules associated with T cell associated with lymphotoxin b–producing dendritic cells in human breast cancer. presence in tertiary lymphoid structures in human lung cancer. Cancer Res. 71: J. Immunol. 191: 2001–2008. 6391–6399. 68. Hindley, J. P., E. Jones, K. Smart, H. Bridgeman, S. N. Lauder, B. Ondondo, 93. Goc, J., C. Germain, T. K. D. Vo-Bourgais, A. Lupo, C. Klein, S. Knockaert, S. Cutting, K. Ladell, K. K. Wynn, D. Withers, et al. 2012. T-cell trafficking L. de Chaisemartin, H. Ouakrim, E. Becht, M. Alifano, et al. 2014. Dendritic cells facilitated by high endothelial venules is required for tumor control after regulatory in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune T-cell depletion. Cancer Res. 72: 5473–5482. contexture and license the positive prognostic value of infiltrating CD8+ T cells. 69. Drayton, D. L., S. Liao, R. H. Mounzer, and N. H. Ruddle. 2006. Lymphoid Cancer Res. 74: 705–715. organ development: from ontogeny to neogenesis. Nat. Immunol. 7: 344–353. 94. Bindea, G., B. Mlecnik, M. Tosolini, A. Kirilovsky, M. Waldner, A. C. Obenauf, 70. Carragher, D. M., J. Rangel-Moreno, and T. D. Randall. 2008. Ectopic lymphoid H. Angell, T. Fredriksen, L. Lafontaine, A. Berger, et al. 2013. Spatiotemporal tissues and local immunity. Semin. Immunol. 20: 26–42. dynamics of intratumoral immune cells reveal the immune landscape in human 71. GeurtsvanKessel, C. H., M. A. Willart, I. M. Bergen, L. S. van Rijt, F. Muskens, cancer. Immunity 39: 782–795. Downloaded from D. Elewaut, A. D. Osterhaus, R. Hendriks, G. F. Rimmelzwaan, and 95. Cipponi, A., M. Mercier, T. Seremet, J.-F. Baurain, I. The´ate, J. van den Oord, B. N. Lambrecht. 2009. Dendritic cells are crucial for maintenance of tertiary M. Stas, T. Boon, P. G. Coulie, and N. van Baren. 2012. Neogenesis of lymphoid lymphoid structures in the lung of influenza virus-infected mice. J. Exp. Med. 206: structures and antibody responses occur in human melanoma metastases. Cancer 2339–2349. Res. 72: 3997–4007. 72. Halle, S., H. C. Dujardin, N. Bakocevic, H. Fleige, H. Danzer, S. Willenzon, 96. Coppola, D., M. Nebozhyn, F. Khalil, H. Dai, T. Yeatman, A. Loboda, and Y. Suezer, G. Ha¨mmerling, N. Garbi, G. Sutter, et al. 2009. Induced bronchus- J. J. Mule´. 2011. Unique ectopic lymph node-like structures present in human associated lymphoid tissue serves as a general priming site for T cells and is primary colorectal carcinoma are identified by immune gene array profiling. Am. J. maintained by dendritic cells. J. Exp. Med. 206: 2593–2601. Pathol. 179: 37–45. http://www.jimmunol.org/ 73. Hayasaka, H., K. Taniguchi, S. Fukai, and M. Miyasaka. 2010. Neogenesis and 97. Di Caro, G., F. Bergomas, F. Grizzi, A. Doni, P. Bianchi, A. Malesci, L. Laghi, development of the high endothelial venules that mediate lymphocyte trafficking. P. Allavena, A. Mantovani, and F. Marchesi. 2014. Occurrence of tertiary lym- Cancer Sci. 101: 2302–2308. phoid tissue is associated with T-cell infiltration and predicts better prognosis in 74. van de Pavert, S. A., and R. E. Mebius. 2010. New insights into the development early-stage colorectal cancers. Clin. Cancer Res. 20: 2147–2158. of lymphoid tissues. Nat. Rev. Immunol. 10: 664–674. 98. Finkin, S., D. Yuan, I. Stein, K. Taniguchi, A. Weber, K. Unger, J. L. Browning, 75. Link, A., D. L. Hardie, S. Favre, M. R. Britschgi, D. H. Adams, M. Sixt, N. Goossens, S. Nakagawa, G. Gunasekaran, et al. 2015. Ectopic lymphoid J. G. Cyster, C. D. Buckley, and S. A. Luther. 2011. Association of T-zone re- structures function as microniches for tumor progenitor cells in hepatocellular ticular networks and conduits with ectopic lymphoid tissues in mice and humans. carcinoma. Nat. Immunol. 16: 1235–1244. Am. J. Pathol. 178: 1662–1675. 99. Germain, C., S. Gnjatic, F. Tamzalit, S. Knockaert, R. Remark, J. Goc, 76. Huang, H.-Y., and S. A. Luther. 2012. Expression and function of interleukin-7 in A. Lepelley, E. Becht, S. Katsahian, G. Bizouard, et al. 2014. Presence of B cells in

secondary and tertiary lymphoid organs. Semin. Immunol. 24: 175–189. by guest on September 25, 2021 tertiary lymphoid structures is associated with a protective immunity in patients 77. Neyt, K., F. Perros, C. H. GeurtsvanKessel, H. Hammad, and B. N. Lambrecht. with lung cancer. Am. J. Respir. Crit. Care Med. 189: 832–844. 2012. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 100. Giraldo, N. A., E. Becht, F. Page`s, G. Skliris, V. Verkarre, Y. Vano, A. Mejean, 33: 297–305. N. Saint-Aubert, L. Lacroix, I. Natario, et al. 2015. Orchestration and prognostic 78. Stranford, S., and N. H. Ruddle. 2012. Follicular dendritic cells, conduits, lym- significance of immune checkpoints in the microenvironment of primary and phatic vessels, and high endothelial venules in tertiary lymphoid organs: parallels metastatic renal cell cancer. Clin. Cancer Res. 21: 3031–3040. with lymph node stroma. Front. Immunol. 3: 350. 101. Goeppert, B., L. Frauenschuh, M. Zucknick, A. Stenzinger, M. Andrulis, 79. Jones, G. W., D. G. Hill, and S. A. Jones. 2016. Understanding immune cells in F. Klauschen, K. Joehrens, A. Warth, M. Renner, A. Mehrabi, et al. 2013. tertiary lymphoid organ development: it is all starting to come together. Front. Prognostic impact of tumour-infiltrating immune cells on biliary tract cancer. Br. Immunol. 7: 401. J. Cancer 109: 2665–2674. 80. Legler, D. F., M. Loetscher, R. S. Roos, I. Clark-Lewis, M. Baggiolini, and 102. Gu-Trantien, C., S. Loi, S. Garaud, C. Equeter, M. Libin, A. de Wind, M. Ravoet, B. Moser. 1998. B cell–attracting chemokine 1, a human CXC chemokine H. Le Buanec, C. Sibille, G. Manfouo-Foutsop, et al. 2013. CD4+ follicular helper expressed in lymphoid tissues, selectively attracts B lymphocytes via BLR1/ T cell infiltration predicts breast cancer survival. J. Clin. Invest. 123: 2873–2892. CXCR5. J. Exp. Med. 187: 655–660. 103. Hennequin, A., V. Derange`re, R. Boidot, L. Apetoh, J. Vincent, D. Orry, 81. Ansel, K. M., V. N. Ngo, P. L. Hyman, S. A. Luther, R. Fo¨rster, J. D. Sedgwick, J. Fraisse, S. Causeret, F. Martin, L. Arnould, et al. 2015. Tumor infiltration by J. L. Browning, M. Lipp, and J. G. Cyster. 2000. A chemokine-driven positive Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer feedback loop organizes lymphoid follicles. Nature 406: 309–314. patients. OncoImmunology 5: e1054598. 82. Luther, S. A., H. L. Tang, P. L. Hyman, A. G. Farr, and J. G. Cyster. 2000. 104. Hiraoka, N., Y. Ino, R. Yamazaki-Itoh, Y. Kanai, T. Kosuge, and K. Shimada. Coexpression of the chemokines ELC and SLC by T zone stromal cells and de- 2015. Intratumoral tertiary lymphoid organ is a favourable prognosticator in pa- letion of the ELC gene in the plt/plt mouse. Proc. Natl. Acad. Sci. USA 97: 12694– tients with pancreatic cancer. Br. J. Cancer 112: 1782–1790. 12699. 105. Lee, H. J., I. A. Park, I. H. Song, S.-J. Shin, J. Y. Kim, J. H. Yu, and G. Gong. 83. Allen, C. D. C., K. M. Ansel, C. Low, R. Lesley, H. Tamamura, N. Fujii, and 2016. Tertiary lymphoid structures: prognostic significance and relationship with J. G. Cyster. 2004. Germinal center dark and zone organization is mediated tumour-infiltrating lymphocytes in triple-negative breast cancer. J. Clin. Pathol. by CXCR4 and CXCR5. Nat. Immunol. 5: 943–952. 69: 422–430. 84. Barone, F., S. Nayar, J. Campos, T. Cloake, D. R. Withers, K.-M. Toellner, 106. Martinet, L., I. Garrido, T. Filleron, S. Le Guellec, E. Bellard, J.-J. Fournie, Y. Zhang, L. Fouser, B. Fisher, S. Bowman, et al. 2015. IL-22 regulates lymphoid P. Rochaix, and J.-P. Girard. 2011. Human solid tumors contain high endothelial chemokine production and assembly of tertiary lymphoid organs. Proc. Natl. Acad. venules: association with T- and B-lymphocyte infiltration and favorable prognosis Sci. USA 112: 11024–11029. in breast cancer. Cancer Res. 71: 5678–5687. 85. Fleige, H., S. Ravens, G. L. Moschovakis, J. Bo¨lter, S. Willenzon, G. Sutter, 107. McMullen, T. P. W., R. Lai, L. Dabbagh, T. M. Wallace, and C. J. de Gara. 2010. S. Ha¨ussler, U. Kalinke, I. Prinz, and R. Fo¨rster. 2014. IL-17–induced CXCL12 Survival in rectal cancer is predicted by T cell infiltration of tumour-associated recruits B cells and induces follicle formation in BALT in the absence of differ- lymphoid nodules. Clin. Exp. Immunol. 161: 81–88. entiated FDCs. J. Exp. Med. 211: 643–651. 108. Meshcheryakova, A., D. Tamandl, E. Bajna, J. Stift, M. Mittlboeck, M. Svoboda, 86. Pikor, N. B., J. L. Astarita, L. Summers-Deluca, G. Galicia, J. Qu, L. A. Ward, D. Heiden, S. Stremitzer, E. Jensen-Jarolim, T. Grunberger,€ et al. 2014. B cells S. Armstrong, C. X. Dominguez, D. Malhotra, B. Heiden, et al. 2015. Integration and ectopic follicular structures: novel players in anti-tumor programming with of Th17- and lymphotoxin-derived signals initiates meningeal-resident stromal cell prognostic power for patients with metastatic colorectal cancer. PLoS One 9: remodeling to propagate neuroinflammation. Immunity 43: 1160–1173. e99008. 87. Rangel-Moreno, J., J. E. Moyron-Quiroz, L. Hartson, K. Kusser, and 109. Messina, J. L., D. A. Fenstermacher, S. Eschrich, X. Qu, A. E. Berglund, T. D. Randall. 2007. Pulmonary expression of CXC chemokine ligand 13, CC M. C. Lloyd, M. J. Schell, V. K. Sondak, J. S. Weber, and J. J. Mule´. 2012. 12- chemokine ligand 19, and CC chemokine ligand 21 is essential for local immunity Chemokine gene signature identifies lymph node-like structures in melanoma: to influenza. Proc. Natl. Acad. Sci. USA 104: 10577–10582. potential for patient selection for immunotherapy? Sci. Rep. 2: 765. 88. Rangel-Moreno, J., D. M. Carragher, M. de la Luz Garcia-Hernandez, 110. Remark, R., M. Alifano, I. Cremer, A. Lupo, M.-C. Dieu-Nosjean, M. Riquet, J. Y. Hwang, K. Kusser, L. Hartson, J. K. Kolls, S. A. Khader, and T. D. Randall. L. Crozet, H. Ouakrim, J. Goc, A. Cazes, et al. 2013. Characteristics and clinical The Journal of Immunology 441

impacts of the immune environments in colorectal and renal cell carcinoma lung 137. Withers, D. R. 2011. Lymphoid tissue inducer cells. Curr. Biol. 21: R381–R382. metastases: influence of tumor origin. Clin. Cancer Res. 19: 4079–4091. 138. Lane, P. J. L., F. M. Gaspal, F. M. McConnell, D. R. Withers, and G. Anderson. 111. Sakai, Y., H. Hoshino, R. Kitazawa, and M. Kobayashi. 2014. High endothelial 2012. Lymphoid tissue inducer cells: pivotal cells in the evolution of CD4 im- venule-like vessels and lymphocyte recruitment in testicular seminoma. Andrology munity and tolerance? Front. Immunol. 3: 24. 2: 282–289. 139. Fletcher, A. L., S. E. Acton, and K. Knoblich. 2015. Lymph node fibroblastic 112. Va¨yrynen, J. P., S. A. Sajanti, K. Klintrup, J. Ma¨kela¨, K.-H. Herzig, reticular cells in health and disease. Nat. Rev. Immunol. 15: 350–361. T. J. Karttunen, A. Tuomisto, and M. J. Ma¨kinen. 2014. Characteristics and 140. Nakano, H., and M. D. Gunn. 2001. Gene duplications at the chemokine locus significance of colorectal cancer associated lymphoid reaction. Int. J. Cancer 134: on mouse 4: multiple strain-specific haplotypes and the deletion of 2126–2135. secondary lymphoid-organ chemokine and EBI-1 ligand chemokine genes in the 113. Wirsing, A. M., O. G. Rikardsen, S. E. Steigen, L. Uhlin-Hansen, and E. Hadler- plt mutation. J. Immunol. 166: 361–369. Olsen. 2014. Characterisation and prognostic value of tertiary lymphoid structures 141. Ohl, L., G. Henning, S. Krautwald, M. Lipp, S. Hardtke, G. Bernhardt, O. Pabst, in oral squamous cell carcinoma. BMC Clin. Pathol. 14: 38. and R. Fo¨rster. 2003. Cooperating mechanisms of CXCR5 and CCR7 in devel- 114. Bergomas, F., F. Grizzi, A. Doni, S. Pesce, L. Laghi, P. Allavena, A. Mantovani, opment and organization of secondary lymphoid organs. J. Exp. Med. 197: 1199– and F. Marchesi. 2011. Tertiary intratumor lymphoid tissue in colo-rectal cancer. 1204. Cancers 4: 1–10. 142. Luther, S. A., K. M. Ansel, and J. G. Cyster. 2003. Overlapping roles of CXCL13, 115. Joshi, N. S., E. H. Akama-Garren, Y. Lu, D.-Y. Lee, G. P. Chang, A. Li, receptor a, and CCR7 ligands in lymph node development. J. Exp. M. DuPage, T. Tammela, N. R. Kerper, A. F. Farago, et al. 2015. Regulatory Med. 197: 1191–1198. T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell 143. De Togni, P., J. Goellner, N. H. Ruddle, P. R. Streeter, A. Fick, S. Mariathasan, responses. Immunity 43: 579–590. S. C. Smith, R. Carlson, L. P. Shornick, J. Strauss-Schoenberger, et al. 1994. 116. Rodriguez, A. B., J. D. Peske, and V. H. Engelhard. Identification and charac- Abnormal development of peripheral lymphoid organs in mice deficient in lym- terization of tertiary lymphoid structures in murine melanoma. Methods Mol. Biol. photoxin. Science 264: 703–707. In press. 144. Alimzhanov, M. B., D. V. Kuprash, M. H. Kosco-Vilbois, A. Luz, 117. Willis, S. N., S. S. Mallozzi, S. J. Rodig, K. M. Cronk, S. L. McArdel, T. Caron, R. L. Turetskaya, A. Tarakhovsky, K. Rajewsky, S. A. Nedospasov, and K. Pfeffer. G. S. Pinkus, L. Lovato, K. L. Shampain, D. E. Anderson, et al. 2009. The mi- 1997. Abnormal development of secondary lymphoid tissues in lymphotoxin croenvironment of germ cell tumors harbors a prominent antigen-driven humoral b-deficient mice. Proc. Natl. Acad. Sci. USA 94: 9302–9307. Downloaded from response. J. Immunol. 182: 3310–3317. 145. Koni, P. A., R. Sacca, P. Lawton, J. L. Browning, N. H. Ruddle, and R. A. Flavell. 118. Bento, D. C., E. Jones, S. Junaid, J. Tull, G. T. Williams, A. Godkin, A. Ager, and 1997. Distinct roles in lymphoid organogenesis for alpha and beta A. Gallimore. 2015. High endothelial venules are rare in colorectal cancers but revealed in lymphotoxin beta-deficient mice. Immunity 6: 491–500. accumulate in extra-tumoral areas with disease progression. OncoImmunology 4: 146. Futterer,€ A., K. Mink, A. Luz, M. H. Kosco-Vilbois, and K. Pfeffer. 1998. The e974374. lymphotoxin b receptor controls organogenesis and affinity maturation in pe- 119. Behr, D. S., W. K. Peitsch, C. Hametner, F. Lasitschka, R. Houben, K. Scho¨nhaar, ripheral lymphoid tissues. Immunity 9: 59–70. J. Michel, C. Dollt, M. Goebeler, A. Marx, et al. 2014. Prognostic value of im- 147. Kuprash, D. V., M. B. Alimzhanov, A. V. Tumanov, A. O. Anderson, K. Pfeffer, mune cell infiltration, tertiary lymphoid structures and PD-L1 expression in and S. A. Nedospasov. 1999. TNF and lymphotoxin b cooperate in the mainte-

Merkel cell carcinomas. Int. J. Clin. Exp. Pathol. 7: 7610–7621. nance of secondary lymphoid tissue microarchitecture but not in the development http://www.jimmunol.org/ 120. Figenschau, S. L., S. Fismen, K. A. Fenton, C. Fenton, and E. S. Mortensen. 2015. of lymph nodes. J. Immunol. 163: 6575–6580. Tertiary lymphoid structures are associated with higher tumor grade in primary 148. Ngo, V. N., H. Korner, M. D. Gunn, K. N. Schmidt, D. S. Riminton, operable breast cancer patients. BMC Cancer 15: 101. M. D. Cooper, J. L. Browning, J. D. Sedgwick, and J. G. Cyster. 1999. Lym- 121. Hsao, H.-M., W. Li, A. E. Gelman, A. S. Krupnick, and D. Kreisel. 2015. The photoxin alpha/beta and are required for stromal cell ex- role of lymphoid neogenesis in allografts. Am. J. Transplant. 16: 1079–1085. pression of homing chemokines in B and T cell areas of the spleen. J. Exp. Med. 122. Koenig, A., and O. Thaunat. 2016. Lymphoid neogenesis and tertiary lymphoid 189: 403–412. organs in transplanted organs. Front. Immunol. 7: 646. 149. Tumanov, A., D. Kuprash, M. Lagarkova, S. Grivennikov, K. Abe, A. Shakhov, 123. Li, W., A. C. Bribriesco, R. G. Nava, A. A. Brescia, A. Ibricevic, J. H. Spahn, L. Drutskaya, C. Stewart, A. Chervonsky, and S. Nedospasov. 2002. Distinct role S. L. Brody, J. H. Ritter, A. E. Gelman, A. S. Krupnick, et al. 2012. Lung of surface lymphotoxin expressed by B cells in the organization of secondary transplant acceptance is facilitated by early events in the graft and is associated with lymphoid tissues. Immunity 17: 239–250. lymphoid neogenesis. Mucosal Immunol. 5: 544–554.

150. Tumanov, A. V., S. I. Grivennikov, A. A. Kruglov, Y. V. Shebzukhov, by guest on September 25, 2021 124. Bruno, T. C., P. Ebner, B. Moore, O. Squalls, K. Waugh, E. B. Eruslanov, S. E. P. Koroleva, Y. Piao, C. Y. Cui, D. V. Kuprash, and S. A. Nedospasov. 2010. Singhal, J. Mitchell, W. Franklin, D. Merrick, et al. 2017. Antigen-presenting Cellular source and molecular form of TNF specify its distinct functions in or- tumor B cells impact the phenotype of CD4 tumor infiltrating T cells in lung ganization of secondary lymphoid organs. Blood 116: 3456–3464. cancer patients. J. Immunol. 198 (Suppl. 1): 130.26. 125. Coughlin, C. M., B. A. Vance, S. A. Grupp, and R. H. Vonderheide. 2004. RNA- 151. Luther, S. A., T. Lopez, W. Bai, D. Hanahan, and J. G. Cyster. 2000. BLC ex- transfected CD40-activated B cells induce functional T-cell responses against viral pression in pancreatic islets causes B cell recruitment and lymphotoxin-dependent and tumor antigen targets: implications for pediatric immunotherapy. Blood 103: lymphoid neogenesis. Immunity 12: 471–481. 2046–2054. 152. Luther, S. A., A. Bidgol, D. C. Hargreaves, A. Schmidt, Y. Xu, J. Paniyadi, 126. Yuseff, M.-I., P. Pierobon, A. Reversat, and A.-M. Lennon-Dume´nil. 2013. How M. Matloubian, and J. G. Cyster. 2002. Differing activities of homeostatic che- B cells capture, process and present antigens: a crucial role for cell polarity. Nat. mokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell re- Rev. Immunol. 13: 475–486. cruitment and lymphoid neogenesis. J. Immunol. 169: 424–433. 127. Germain, C., S. Gnjatic, and M.-C. Dieu-Nosjean. 2015. Tertiary lymphoid 153. Marinkovic, T., A. Garin, Y. Yokota, Y.-X. Fu, N. H. Ruddle, G. C. Furtado, and structure-associated B cells are key players in anti-tumor immunity. Front. S. A. Lira. 2006. Interaction of mature CD3+CD4+ T cells with dendritic cells Immunol. 6: 67. triggers the development of tertiary lymphoid structures in the thyroid. J. Clin. 128. Nelson, B. H. 2010. CD20+ B cells: the other tumor-infiltrating lymphocytes. J. Invest. 116: 2622–2632. Immunol. 185: 4977–4982. 154. Furtado, G. C., T. Marinkovic, A. P. Martin, A. Garin, B. Hoch, W. Hubner, 129. Affara, N. I., B. Ruffell, T. R. Medler, A. J. Gunderson, M. Johansson, B. K. Chen, E. Genden, M. Skobe, and S. A. Lira. 2007. Lymphotoxin beta re- S. Bornstein, E. Bergsland, M. Steinhoff, Y. Li, Q. Gong, et al. 2014. B cells ceptor signaling is required for inflammatory lymphangiogenesis in the thyroid. regulate macrophage phenotype and response to chemotherapy in squamous car- Proc. Natl. Acad. Sci. USA 104: 5026–5031. cinomas. Cancer Cell 25: 809–821. 155. Gatumu, M. K., K. Skarstein, A. Papandile, J. L. Browning, R. A. Fava, and 130. Schioppa, T., R. Moore, R. G. Thompson, E. C. Rosser, H. Kulbe, A. I. Bolstad. 2009. Blockade of lymphotoxin-beta receptor signaling reduces as- S. Nedospasov, C. Mauri, L. M. Coussens, and F. R. Balkwill. 2011. B regulatory pects of Sjo¨gren’s syndrome in salivary glands of non-obese diabetic mice. Arthritis cells and the tumor-promoting actions of TNF-a during squamous carcinogenesis. Res. Ther. 11: R24. Proc. Natl. Acad. Sci. USA 108: 10662–10667. 156. Gra¨bner, R., K. Lo¨tzer, S. Do¨pping, M. Hildner, D. Radke, M. Beer, 131. Schwartz, M., Y. Zhang, and J. D. Rosenblatt. 2016. B cell regulation of the anti- R. Spanbroek, B. Lippert, C. A. Reardon, G. S. Getz, et al. 2009. Lymphotoxin b tumor response and role in carcinogenesis. J. Immunother. Cancer 4: 40. receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia 2 2 132. Shah, S., A. A. Divekar, S. P. Hilchey, H. M. Cho, C. L. Newman, S. U. Shin, of aged ApoE / mice. J. Exp. Med. 206: 233–248. H. Nechustan, P. M. Challita-Eid, B. M. Segal, K. H. Yi, and J. D. Rosenblatt. 157. Motallebzadeh, R., S. Rehakova, T. M. Conlon, T. S. Win, C. J. Callaghan, 2005. Increased rejection of primary tumors in mice lacking B cells: inhibition of M. Goddard, E. M. Bolton, N. H. Ruddle, J. A. Bradley, and G. J. Pettigrew. anti-tumor CTL and TH1 responses by B cells. Int. J. Cancer 117: 574– 2012. Blocking lymphotoxin signaling abrogates the development of ectopic 586. lymphoid tissue within cardiac allografts and inhibits effector antibody responses. 133. Sawa, S., M. Cherrier, M. Lochner, N. Satoh-Takayama, H. J. Fehling, F. Langa, FASEB J. 26: 51–62. J. P. Di Santo, and G. Eberl. 2010. Lineage relationship analysis of RORgammat+ 158. Be´ne´zech, C., N.-T. Luu, J. A. Walker, A. A. Kruglov, Y. Loo, K. Nakamura, innate lymphoid cells. Science 330: 665–669. Y. Zhang, S. Nayar, L. H. Jones, A. Flores-Langarica, et al. 2015. Inflammation- 134. Kim, D., R. E. Mebius, J. D. MacMicking, S. Jung, T. Cupedo, Y. Castellanos, induced formation of fat-associated lymphoid clusters. Nat. Immunol. 16: 819– J. Rho, B. R. Wong, R. Josien, N. Kim, et al. 2000. Regulation of peripheral 828. lymph node genesis by the tumor necrosis factor family member TRANCE. J. Exp. 159. Furtado, G. C., M. E. Pacer, G. Bongers, C. Be´ne´zech, Z. He, L. Chen, Med. 192: 1467–1478. M. C. Berin, G. Kollias, J. H. Caaman˜o, and S. A. Lira. 2014. TNFa-dependent 135. Mebius, R. E. 2003. Organogenesis of lymphoid tissues. Nat. Rev. Immunol. 3: development of lymphoid tissue in the absence of RORgt+ lymphoid tissue in- 292–303. ducer cells. Mucosal Immunol. 7: 602–614. 136. Randall, T. D., D. M. Carragher, and J. Rangel-Moreno. 2008. Development of 160. Guedj, K., J. Khallou-Laschet, M. Clement, M. Morvan, A.-T. Gaston, secondary lymphoid organs. Annu. Rev. Immunol. 26: 627–650. G. Fornasa, J. Dai, M. Gervais-Taurel, G. Eberl, J.-B. Michel, et al. 2014. M1 442 BRIEF REVIEWS: TUMOR INFILTRATES AND TERTIARY LYMPHOID STRUCTURES

macrophages act as LTbR-independent lymphoid tissue inducer cells during 167. Muniz, L. R., M. E. Pacer, S. A. Lira, and G. C. Furtado. 2011. A critical role for atherosclerosis-related lymphoid neogenesis. Cardiovasc. Res. 101: 434–443. dendritic cells in the formation of lymphatic vessels within tertiary lymphoid 161. Peters, A., L. A. Pitcher, J. M. Sullivan, M. Mitsdoerffer, S. E. Acton, B. Franz, structures. J. Immunol. 187: 828–834. K. Wucherpfennig, S. Turley, M. C. Carroll, R. A. Sobel, et al. 2011. Th17 cells 168. McDonald, K. G., J. S. McDonough, and R. D. Newberry. 2005. Adaptive induce ectopic lymphoid follicles in central nervous system tissue inflammation. immune responses are dispensable for isolated lymphoid follicle formation: Immunity 35: 986–996. antigen-naive, lymphotoxin-sufficient B lymphocytes drive the formation of mature 162. Can˜ete, J. D., R. Celis, N. Yeremenko, R. Sanmartı´, L. van Duivenvoorde, isolated lymphoid follicles. J. Immunol. 174: 5720–5728. J. Ramı´rez, I. Blijdorp, C. M. Garcı´a-Herrero, J. L. Pablos, and D. L. Baeten. 169. Kim, H.-J., T. Kammertoens, M. Janke, O. Schmetzer, Z. Qin, C. Berek, and 2015. Ectopic lymphoid neogenesis is strongly associated with activation of the IL- T. Blankenstein. 2004. Establishment of early lymphoid organ infrastructure in 23 pathway in rheumatoid synovitis. Arthritis Res. Ther. 17: 173. transplanted tumors mediated by local production of lymphotoxin a and in the 163. Khader, S. A., L. Guglani, J. Rangel-Moreno, R. Gopal, B. A. Junecko, combined absence of functional B and T cells. J. Immunol. 172: 4037–4047. J. J. Fountain, C. Martino, J. E. Pearl, M. Tighe, Y. Lin, et al. 2011. IL-23 is 170. Mackay, F., P. Schneider, P. Rennert, and J. Browning. 2003. BAFF AND APRIL: required for long-term control of Mycobacterium tuberculosis and B cell follicle a tutorial on B cell survival. Annu. Rev. Immunol. 21: 231–264. formation in the infected lung. J. Immunol. 187: 5402–5407. 171. Schiemann, B., J. L. Gommerman, K. Vora, T. G. Cachero, S. Shulga-Morskaya, 164. Goya, S., H. Matsuoka, M. Mori, H. Morishita, H. Kida, Y. Kobashi, T. Kato, M. Dobles, E. Frew, and M. L. Scott. 2001. An essential role for BAFF in the Y. Taguchi, T. Osaki, I. Tachibana, et al. 2003. Sustained interleukin-6 signalling normal development of B cells through a BCMA-independent pathway. Science leads to the development of lymphoid organ-like structures in the lung. J. Pathol. 293: 2111–2114. 200: 82–87. 172. Varfolomeev, E., F. Kischkel, F. Martin, D. Seshasayee, H. Wang, D. Lawrence, 165. Hsu, H.-C., P. Yang, J. Wang, Q. Wu, R. Myers, J. Chen, J. Yi, T. Guentert, C. Olsson, L. Tom, S. Erickson, D. French, et al. 2004. APRIL-deficient mice A. Tousson, A. L. Stanus, et al. 2008. -producing T helper cells and have normal immune system development. Mol. Cell. Biol. 24: 997–1006. interleukin 17 orchestrate autoreactive germinal center development in autoim- 173. Kalled, S. L. 2006. Impact of the BAFF/BR3 axis on B cell survival, germinal mune BXD2 mice. Nat. Immunol. 9: 166–175. center maintenance and antibody production. Semin. Immunol. 18: 290–296. 166. Gopal, R., J. Rangel-Moreno, S. Slight, Y. Lin, H. F. Nawar, B. A. Fallert Junecko, 174. Vora, K. A., L. C. Wang, S. P. Rao, Z.-Y. Liu, G. R. Majeau, A. H. Cutler, T. A. Reinhart, J. Kolls, T. D. Randall, T. D. Connell, and S. A. Khader. 2013. P. S. Hochman, M. L. Scott, and S. L. Kalled. 2003. Cutting edge: germinal Interleukin-17–dependent CXCL13 mediates mucosal vaccine-induced immunity centers formed in the absence of B cell-activating factor belonging to the TNF against tuberculosis. Mucosal Immunol. 6: 972–984. family exhibit impaired maturation and function. J. Immunol. 171: 547–551. Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021