Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2145 2022-05-17 10:01:09 |
2 format correct -13 word(s) 2132 2022-05-18 02:49:48 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Blanco Clemente, M.; , .; Yanguas Casás, N. Tumor Microenvironment Impact on Follicular Lymphomas. Encyclopedia. Available online: https://encyclopedia.pub/entry/23000 (accessed on 05 July 2024).
Blanco Clemente M,  , Yanguas Casás N. Tumor Microenvironment Impact on Follicular Lymphomas. Encyclopedia. Available at: https://encyclopedia.pub/entry/23000. Accessed July 05, 2024.
Blanco Clemente, Mariola, , Natalia Yanguas Casás. "Tumor Microenvironment Impact on Follicular Lymphomas" Encyclopedia, https://encyclopedia.pub/entry/23000 (accessed July 05, 2024).
Blanco Clemente, M., , ., & Yanguas Casás, N. (2022, May 17). Tumor Microenvironment Impact on Follicular Lymphomas. In Encyclopedia. https://encyclopedia.pub/entry/23000
Blanco Clemente, Mariola, et al. "Tumor Microenvironment Impact on Follicular Lymphomas." Encyclopedia. Web. 17 May, 2022.
Tumor Microenvironment Impact on Follicular Lymphomas
Edit

Follicular lymphomas (FL) are neoplasms that resemble normal germinal center (GC) B-cells. Normal GC and neoplastic follicles contain non-neoplastic cells such as T-cells, follicular dendritic cells, cancer associated fibroblasts, and macrophages, which define the tumor microenvironment (TME), which itself is an essential factor in tumor cell survival. The main characteristics of the TME in FL are an increased number of follicular regulatory T-cells (Treg) and follicular helper T-cells (Tfh), M2-polarization of macrophages, and the development of a nodular network by stromal cells that creates a suitable niche for tumor growth. All of them play important roles in tumor angiogenesis, inhibition of apoptosis, and immune evasion, which are key factors in tumor progression and transformation risk. 

tumor microenvironment follicular lymphoma therapeutic targets

1. Quiet Bystanders: TME Impact on FL Development and Prognosis

Understanding the main differences between normal and neoplastic lymph nodes (LNs) is essential. There is now growing evidence that crosstalk between lymphoma cells and TME cells is crucial for disease onset and progression.
In the non-neoplastic setting, B-cells must generate enormous variability in the antigen (Ag) recognition sites of their immunoglobulins (Igs), also known as B-cell receptor (BCR). For this purpose, they developed a mechanism that, in three independent steps (VDJ recombination, somatic hypermutation (SHM), and class switching) enables the production of Igs of almost unlimited specificity, although there is a risk of pathological mutations arising during this process. Naïve B-cells leave the BM after V(D)J recombination and migrate through the bloodstream to secondary LNs. B-cells that have encountered Ag enter the GC, which is formed by the expansion of selected clones within the FDC meshwork [1][2]. The proliferating GC B-cells are known as centroblasts, and they undergo SHM of the Ig variable region (IGV) genes, which alters the antigen affinity of the Ig that will be produced by the cell [3]. On the other hand, when centroblasts stop dividing, they become medium-sized cells called centrocytes, and if the IGV gene mutations have resulted in increased affinity for antigen, they present it to the Tfh cells via the major histocompatibility complex class II (MHCII). Centrocytes initially accumulate among the centroblasts and then migrate to the opposite pole of the GC. This causes the polarization of the GC into a “dark zone”, which contains centroblasts and closely packed centrocytes, and a “light zone”, which contains centrocytes, FDCs, and numerous T-cells. These two zones can be differentiated by the proliferation marker Ki67 as the “dark zone” contains the highly proliferating centroblasts and the “light zone” the low proliferating centrocytes. The selected centrocytes reenter the dark zone, where clonal expansion occurs and cells may cycle between the dark and light zones multiple times [4][5]. Finally, centrocytes will become either plasma B-cells or memory B-cells. The interaction with CD23, which is expressed by FDCs, seems to be important in the differentiation into plasma cells, stimulating class switching. On the other hand, interaction with Tfh cells through the CD40–CD40 ligand appears to be important for generating memory B-cells [6][7].
This GC reaction is highly dependent on the TME. T-cells are predominantly located in the light zone and most are Tfh cells, which are important for selecting B-cells for entry and proliferation within the GC. Treg cells are present in low numbers; they exert negative regulatory effects on both B- and T-cells and are needed to stop the GC reaction, preventing excessive immune responses [8]. FDCs are large cells with delicate nuclear membranes that derive from mesenchymal origin. They have surface complement receptors (CD21) and Fc receptors (CD23) that bind free antigen and antigen–antibody complexes for presentation to B-cells. In normal lymph nodes, the role of FDCs and CAFs is to build a network to support the GC reaction. In contrast, macrophages (CD68+) are typically phagocytic, containing apoptotic debris from B-cells that have failed to express a surface Ig molecule [9]. Macrophages can be polarized into M1 (inflammatory phenotype) or M2 (anti-inflammatory), resulting in distinct cytokine production or T-cell function (Th1 and Th2).
Many of the components of the FL microenvironment mimic those present in normal GCs, but some important differences may contribute to tumor-cell survival. The role of TME in FL can be defined in two directions: supporting tumor growth and suppressing the antitumor immune response [10]. Table 1 recapitulates the main cells involved in the TME of FL.
Table 1. Main cells involved in the tumor microenvironment of follicular lymphoma.

2. B-Cells

The histopathology of FL is closely related to key events in normal B-cell development and differentiation. As previously mentioned, in approximately 85% of FL cases, the malignant B-cells harbor a translocation t(14;18)(q32;q21) that occurs as a consequence of aberrant VDJ recombination in a progenitor B-cell in the BM, in contrast to normal GCs [11][12]. By juxtaposing BCL2 next to the immunoglobulin-heavy chain enhancer, this translocation leads to a constant expression of BCL2, which is observed in almost all FL (≥25% of FL cells) but not in normal GCs, resulting in the inhibition of apoptosis and favoring the selection of these clones in the GCs of secondary lymphoid organs [13]. The t(14;18) translocation in B-cells increases the risk of developing FL, although it is not sufficient and the acquisition of additional mutations is necessary for neoplastic transformation and disease progression, since the t(14;18) translocation has also been observed in GC B-cells of healthy individuals [14]. FL B-cells undergo repeated re-entry into GCs, so this leads to the acquisition of these additional aberrations that enable FL to develop.
Unlike normal GCs, in FL, there is no differentiation into a “dark zone” and a “light zone”, but there is a predominance of centrocytes interspersed with some centroblasts [15]. FL B-cells express many of the antigens that are found on normal GC B-cells and that are associated with interactions with T-cells and dendritic cells such as costimulatory molecules CD80, CD86, CD40/43/44, and CD70 [16]. Furthermore, they express surface Igs that have undergone SHM of the IGV genes and, in approximately 20–50% of cases, have undergone Ig class switching. In a high proportion of FL cases during SHM, new mutations can be acquired. These mutations introduce consensus sequences of unusual mannosylated glycans, which are not seen in normal B-cells and can bind to lectins on FDCs and macrophages, enabling them to survive in the GC environment in the absence of cognate antigen [17][18].

3. T-Cells

In the neoplastic follicles, T-cells are less numerous than in reactive follicles and they are randomly distributed, in contrast to the concentration in the light zone that characterizes normal lymph nodes [19]. CD4+ T-cells (Treg and Tfh) are predominant with a higher CD4:CD8 ratio, Treg cells being more numerous than Tfh cells. Both types of cells are fundamental to providing tumor support and facilitating immune evasion.
FL-associated Tfh lymphocytes are defined by the expression of the chemokine receptor CXCR5, the inducible T-cell co-stimulator (ICOS), the programmed cell death 1 (PD-1), and the transcription repressor BCL6. These cells produce an excess of chemokines (IL-4, IL-17, IL-21, and IFN-γ) and overexpression of CD40L, supporting B-cell viability, inhibiting apoptosis, and influencing the TME. Moreover, Tfh cells expressing IL-4 and CD40L can also induce FL cells to produce CCL17 and CCL22, thereby promoting the active migration of Tregs [7][19].
On the other hand, Treg cells are characterized by the expression of the transcription factor FOXP3 and they have a negative regulatory effect on B- and T-cells, playing a pro-tumor role due to their immunosuppressive activity, which hampers CD8+ T-cell activation [20]. Interestingly, FL patients are particularly rich in CD4+ T-cells that harbor CXCR5high ICOShigh PD1high and FOXP3+/CD4+/CD25+ phenotypes, which are called follicular regulatory T-cells (Tfr). Tfr cells have greater suppressive capacity than Tregs in normal LNs because they upregulate cytotoxic T-lymphocyte antigen 4 (CTLA-4), IL-10, and the glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR). This pattern has been described as being a predictor of shortened overall survival (OS) [21].

4. Stromal Cells, Follicular Dendritic Cells and Macrophages

Stromal cells and macrophages are key factors in the pathogenesis of FL, but their association with prognosis remains controversial.
FDCs and CAFs are stromal cells that phenotypically and functionally differ from their normal counterparts, presenting a niche-based model of oncogenesis attributed to the dynamic coevolution of cancer and stromal cells. FDCs are particular Ag-presenting cells (APCs), and they present intact Ag–Ab complexes on their cell surface, which enable survival and induce the differentiation of FL cells. They can also trigger Tfh recruitment by the production of CXCL13, which interacts with CXCR5. The FDCs of almost all FL express CD21, while only a small subgroup express CD23, which can be expressed by FDCs and B-cells. In contrast, CAFs secrete components of the extracellular matrix such as laminin, fibronectin, and collagen. The nodular network built in the neoplastic setting infiltrates follicles, creating a suitable niche even in extranodal sites and BM, suggesting that they are recruited by the neoplastic cells [7]. The crosstalk between FDCs, CAFs, and FL B-cells is crucial to promote tumor cell survival. They are responsible for B- and T-cell migration thanks to the upregulation of CXCL12, CXCL13, and BAFF signaling as well as adhesion through interactions between adhesive molecules such as VCAM-1 and VLA-4. Through these interactions, stromal cells protect malignant cells against apoptosis induced by chemotherapy or rituximab. They can also induce malignant monocyte recruitment and their differentiation to M2-protumoral macrophages through the secretion of CCL2 and CSF-1 [22][23].
TAMs are typically non-phagocytic, with a low proliferation fraction, and they do not undergo apoptosis. They express CD163+, the Fc fragment of IgG, and C-type lectin domains. These cells can display two different phenotypes: M1- and M2-polarization. This polarization is controlled by tumor cells within the TME and is dynamic throughout cancer development. M1-macrophages are typically tumor-suppressing cells that act in the TME by recruiting CD8+ T-cells and NK cells. These CD8+ and NK cells express high levels of cytokines and chemokines, recruiting other immune cells and favoring the signaling of anti-tumorigenic pathways [24]. On the other hand, the M2-phenotype is associated with tissue remodeling, angiogenesis, and progression, making them an attractive target for therapies [25]. These M2-macrophages secrete immunosuppressive molecules into the TME, suppress T-cell mediated anti-tumor responses and recruit Treg lymphocytes, facilitating tumor proliferation and immune evasion [24][26]. TAMs also play a key role in tumor angiogenesis by secreting VEGFA, which can stimulate the chemotaxis of endothelial cells and macrophages [27]. In addition, they promote the epithelial–mesenchymal transition process, which enables cancer cells to leave the tissue site, favoring the initiation of metastasis [28]. However, TAM’s polarization state is not fixed. At early stages of tumor development, M1-macrophages are predominant, while the M2-phenotype is more frequent in the advanced setting where tumor proliferation and invasion increases [24]. For the time being, the role of TAMs for the prognosis of FL remains unclear. In the pre-rituximab era, some studies have suggested that macrophage infiltration was associated with lower survival, probably due to their M2-polarization. Nevertheless, the addition of rituximab to chemotherapy modified their prognostic impact, and, although the mechanism is still not well-known, it is likely to be associated with antibody-dependent cell-mediated toxicity (ADCC) [29][30]. The antitumoral activity of rituximab is dependent on interactions with the effector cells that have Fc receptors (neutrophils, natural killer cells, and macrophages). Because tissue macrophages are critical for B-cell depletion after anti-CD20 antibody therapy, it is plausible that there is a relationship between TAM content and the efficacy of rituximab [31]. The Finnish and French groups showed that the addition of rituximab to chemotherapy reversed the negative prognostic impact of high macrophage content to favorable, reporting a survival benefit in the rituximab arm [32].

5. Neutrophils

Little is known about the interactions between FL cells and neutrophils although they are important players in the innate immune system. Unlike other hematological cells, they are predominantly present in peripheral blood. In preclinical studies, tumor associated neutrophils (TANs) have demonstrated a reduced cytotoxic effect of chemotherapy through the interaction of CD11b/ICAM-1 with CD44 of malignant B-cells. An increased number of TANs in FL patients has been correlated with poor prognosis and weak treatment responses. These findings suggest that TANs could become a new targeted therapy [33].
In summary, FL B-cells are surrounded by a network of supportive cells that take part in the development of tumor cells, treatment resistance, prognosis, and histological transformation.

References

  1. Federico, M.; Bellei, M.; Marcheselli, L.; Luminari, S.; Lopez-Guillermo, A.; Vitolo, U.; Pro, B.; Pileri, S.; Pulsoni, A.; Soubeyran, P.; et al. Follicular lymphoma international prognostic index 2: A new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. J. Clin. Oncol. 2009, 27, 4555–4562.
  2. Roulland, S.; Kelly, R.S.; Morgado, E.; Sungalee, S.; Solal-Celigny, P.; Colombat, P.; Jouve, N.; Palli, D.; Pala, V.; Tumino, R.; et al. t(14;18) Translocation: A predictive blood biomarker for follicular lymphoma. J. Clin. Oncol. 2014, 32, 1347–1355.
  3. Schwickert, T.A.; Victora, G.D.; Fooksman, D.R.; Kamphorst, A.O.; Mugnier, M.R.; Gitlin, A.D.; Dustin, M.L.; Nussenzweig, M.C. A dynamic T cell-limited checkpoint regulates affinity-dependent B cell entry into the germinal center. J. Exp. Med. 2011, 208, 1243–1252.
  4. MacLennan, I.C. Germinal centers. Annu. Rev. Immunol. 1994, 12, 117–139.
  5. Jacob, J.; Kelsoe, G.; Rajewsky, K.; Weiss, U. Intraclonal generation of antibody mutants in germinal centres. Nature 1991, 354, 389–392.
  6. Durie, F.H.; Foy, T.M.; Masters, S.R.; Laman, J.D.; Noelle, R.J. The role of CD40 in the regulation of humoral and cell-mediated immunity. Immunol. Today 1994, 15, 406–411.
  7. Watanabe, T. The Tumor Microenvironment in Follicular Lymphoma: Its Pro-Malignancy Role with Therapeutic Potential. Int. J. Mol. Sci. 2021, 22, 5352.
  8. Chung, Y.; Tanaka, S.; Chu, F.; Nurieva, R.I.; Martinez, G.J.; Rawal, S.; Wang, Y.-H.; Lim, H.; Reynolds, J.M.; Zhou, X.; et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat. Med. 2011, 17, 983–988.
  9. Wittenbrink, N.; Klein, A.; Weiser, A.A.; Schuchhardt, J.; Or-Guil, M. Is there a typical germinal center? A large-scale immunohistological study on the cellular composition of germinal centers during the hapten-carrier-driven primary immune response in mice. J. Immunol. 2011, 187, 6185–6196.
  10. Gribben, J.G. Implications of the tumor microenvironment on survival and disease response in follicular lymphoma. Curr. Opin. Oncol. 2010, 22, 424–430.
  11. Tsujimoto, Y.; Cossman, J.; Jaffe, E.; Croce, C.M. Involvement of the bcl-2 gene in human follicular lymphoma. Science 1985, 228, 1440–1443.
  12. Küppers, R.; Stevenson, F.K. Critical influences on the pathogenesis of follicular lymphoma. Blood 2018, 131, 2297–2306.
  13. Tsujimoto, Y.; Gorham, J.; Cossman, J.; Jaffe, E.; Croce, C.M. The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining. Science 1985, 229, 1390–1393.
  14. Limpens, J.; Stad, R.; Vos, C.; de Vlaam, C.; de Jong, D.; van Ommen, G.J.; Schuuring, E.; Kluin, P.M. Lymphoma-associated translocation t(14;18) in blood B cells of normal individuals. Blood 1995, 85, 2528–2536.
  15. Victora, G.D.; Dominguez-Sola, D.; Holmes, A.B.; Deroubaix, S.; Dalla-Favera, R.; Nussenzweig, M.C. Identification of human germinal center light and dark zone cells and their relationship to human B-cell lymphomas. Blood 2012, 120, 2240–2248.
  16. Dogan, A.; Du, M.Q.; Aiello, A.; Diss, T.C.; Ye, H.T.; Pan, L.X.; Isaacson, P.G. Follicular lymphomas contain a clonally linked but phenotypically distinct neoplastic B-cell population in the interfollicular zone. Blood 1998, 91, 4708–4714.
  17. Stevenson, F.K.; Stevenson, G.T. Follicular lymphoma and the immune system: From pathogenesis to antibody therapy. Blood 2012, 119, 3659–3667.
  18. Amé-Thomas, P.; Tarte, K. The yin and the yang of follicular lymphoma cell niches: Role of microenvironment heterogeneity and plasticity. Semin. Cancer Biol. 2014, 24, 23–32.
  19. Harris, N.L. Indolent lymphoma: Follicular lymphoma and the microenvironment-insights from the microscope. Hematol. Am. Soc. Hematol. Educ. Program 2014, 2014, 158–162.
  20. Carreras, J.; Lopez-Guillermo, A.; Fox, B.C.; Colomo, L.; Martinez, A.; Roncador, G.; Montserrat, E.; Campo, E.; Banham, A.H. High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood 2006, 108, 2957–2964.
  21. Amé-Thomas, P.; Le Priol, J.; Yssel, H.; Caron, G.; Pangault, C.; Jean, R.; Martin, N.; Marafioti, T.; Gaulard, P.; Lamy, T.; et al. Characterization of intratumoral follicular helper T cells in follicular lymphoma: Role in the survival of malignant B cells. Leukemia 2012, 26, 1053–1063.
  22. Amé-Thomas, P.; Maby-El Hajjami, H.; Monvoisin, C.; Jean, R.; Monnier, D.; Caulet-Maugendre, S.; Guillaudeux, T.; Lamy, T.; Fest, T.; Tarte, K. Human mesenchymal stem cells isolated from bone marrow and lymphoid organs support tumor B-cell growth: Role of stromal cells in follicular lymphoma pathogenesis. Blood 2007, 109, 693–702.
  23. Staiger, A.M.; Duppel, J.; Dengler, M.A.; van der Kuip, H.; Vöhringer, M.C.; Aulitzky, W.E.; Rosenwald, A.; Ott, G.; Horn, H. An analysis of the role of follicular lymphoma-associated fibroblasts to promote tumor cell viability following drug-induced apoptosis. Leuk. Lymphoma 2017, 58, 1922–1930.
  24. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995.
  25. Araujo-Ayala, F.; Pérez-Galán, P.; Campo, E. Vulnerabilities in the tumor and microenvironment in follicular lymphoma. Hematol. Oncol. 2021, 39 (Suppl. S1), 83–87.
  26. Allavena, P.; Mantovani, A. Immunology in the clinic review series; focus on cancer: Tumour-associated macrophages: Undisputed stars of the inflammatory tumour microenvironment. Clin. Exp. Immunol. 2012, 167, 195–205.
  27. Riabov, V.; Gudima, A.; Wang, N.; Mickley, A.; Orekhov, A.; Kzhyshkowska, J. Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front. Physiol. 2014, 5, 75.
  28. Zhu, L.; Fu, X.; Chen, X.; Han, X.; Dong, P. M2 macrophages induce EMT through the TGF-β/Smad2 signaling pathway. Cell Biol. Int. 2017, 41, 960–968.
  29. Kridel, R.; Xerri, L.; Gelas-Dore, B.; Tan, K.; Feugier, P.; Vawda, A.; Canioni, D.; Farinha, P.; Boussetta, S.; Moccia, A.A.; et al. The Prognostic Impact of CD163-Positive Macrophages in Follicular Lymphoma: A Study from the BC Cancer Agency and the Lymphoma Study Association. Clin. Cancer Res. 2015, 21, 3428–3435.
  30. Glas, A.M.; Kersten, M.J.; Delahaye, L.J.M.J.; Witteveen, A.T.; Kibbelaar, R.E.; Velds, A.; Wessels, L.F.A.; Joosten, P.; Kerkhoven, R.M.; Bernards, R.; et al. Gene expression profiling in follicular lymphoma to assess clinical aggressiveness and to guide the choice of treatment. Blood 2005, 105, 301–307.
  31. Taskinen, M.; Karjalainen-Lindsberg, M.-L.; Nyman, H.; Eerola, L.-M.; Leppä, S. A high tumor-associated macrophage content predicts favorable outcome in follicular lymphoma patients treated with rituximab and cyclophosphamide-doxorubicin-vincristine-prednisone. Clin. Cancer Res. 2007, 13, 5784–5789.
  32. Canioni, D.; Salles, G.; Mounier, N.; Brousse, N.; Keuppens, M.; Morchhauser, F.; Lamy, T.; Sonet, A.; Rousselet, M.-C.; Foussard, C.; et al. High numbers of tumor-associated macrophages have an adverse prognostic value that can be circumvented by rituximab in patients with follicular lymphoma enrolled onto the GELA-GOELAMS FL-2000 trial. J. Clin. Oncol. 2008, 26, 440–446.
  33. Hirz, T.; Matera, E.-L.; Chettab, K.; Jordheim, L.P.; Mathé, D.; Evesque, A.; Esmenjaud, J.; Salles, G.; Dumontet, C. Neutrophils protect lymphoma cells against cytotoxic and targeted therapies through CD11b/ICAM-1 binding. Oncotarget 2017, 8, 72818–72834.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 255
Revisions: 2 times (View History)
Update Date: 18 May 2022
1000/1000
Video Production Service