Tumor Microenvironment in Thymic Epithelial Tumors: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Apostolos Agrafiotis.

The tumor microenvironment (TME) is defined as the environment surrounding a tumor. There is a close, dynamic relation and interaction between the tumor and its neighboring microenvironment. There are some particularities of the thymus itself and of the TME of thymic epithelial tumors that hinder the routine use of targeted therapies, including immune checkpoint inhibitors. The understanding of the unique characteristics of the TME of thymic epithelial tumors could possibly result in the development of novel promising therapies.

  • thymus
  • tumor microenvironment
  • thymoma
  • thymic carcinoma

1. The Particularities of TME of TET and the Limitations in the Use of Immunotherapy

Surgical resection is considered as the standard treatment for early-stage thymomas, whereas radiation therapy and chemotherapy are reserved for advanced or recurrent tumors and for patients who are not candidates for surgical resection [8,9][1][2]. However, as immunotherapy has shown promising results in other solid tumors, it has gained the interest of researchers who investigate its efficacy in thymic epithelial TETtumors (TET) [11][3]. Indeed, there are clinical trials that evaluate the use of these agents in the treatment of TET, revealing encouraging preliminary clinical outcomes [12,13][4][5]. What is interesting in the case of TET is the key role of the thymus gland in the development of adaptive immune responses, where the maturation of T-cells takes place [9,11][2][3]. For that reason, there are severe side-effects (auto-immune toxicity) that have been observed in patients with TET receiving immune checkpoint inhibitors (ICI) [9[2][3],11], and they constitute an important obstacle that limit widespread and routine use. Immune-related adverse events generally represent an acceptable morbidity, but, in some cases (ranging from 0.36% to 1.23% determined by the type of ICI used), they can be fatal. Fatal toxicity is less frequent with anti-programmed death 1 (anti-PD-1) receptor and anti-programmed death ligand 1 (anti-PD-L1), followed by anti-Cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4), and more frequent with their combined administration (anti-PD1/PDL1 and anti-CTLA-4) [14][6].
There are studies that have identified PD-L1 expression in the majority of TET [15,16][7][8]. Padda et al. examined 69 TET and found 68% of cases with PD-L1 overexpression [16][8]. Katsuya et al. examined a series consisting of 141 TET and discovered high PD-L1 expression in 70% of thymic carcinoma specimens, but only in 23% of thymomas [17][9]. Similarly, Yokoyama et al. observed increased PD-L1 expression in 80% of thymic carcinoma specimens [18][10]. However, there is controversy as far as survival is concerned, and, therefore, the prognostic influence and benefit of PD-L1 is debated. Padda et al. demonstrated that increased PD-L1 expression resulted in decreased overall survival (OS), whereas Katsuya et al. described no association between PD-L1 expression and OS [16,17][8][9]. Bedekovics et al. found that 69% of thymomas and 43% of thymic carcinomas in their series were highly positive (positive tumor cells ≥50% or positive immune cells ≥10%) [15][7].
As already mentioned, TET are characterized by histological heterogeneity, and it is observed that most patients are refractory to immunotherapy because of primary or acquired resistance [19,20][11][12]. Contrary to standard cytotoxic therapy, the therapeutic action of immunotherapy agents occurs by triggering the anti-tumor immune reaction, which is based on the immunoregulative process that occurs between cancer cells and the tumor microenvironment (TME) [19][11]. Therefore, to help guide the selection of patients who could benefit from this kind of treatment and optimize its outcomes, more specific biomarkers and more pertinent predictive elements for identifying these patients who will potentially respond to these therapeutic modalities are required [19][11]. Su et al. established an immune-related long noncoding RNAs (IRLs) classifier to accurately identify the response of patients suffering from TET. According to the authors, long non-coding RNAs (LncRNAs) can modulate the immune response by managing the homeostasis, TME, anti-inflammatory factors, and immune cell function [19][11].
Another issue that needs to be taken into account is the absence of standard actionable mutations, which is the main reason for the lack of effective targeted therapies in TET [18,19][10][11]. However, high expression of PD-L1 on tumor cells and abundant CD8+ lymphocytes provide a strong basis for the administration of ICI, which target the PD-1/PD-L1 pathway in the case of TET. Two phase II trials evaluated the efficacy of pembrolizumab (PD-1 inhibitor) in patients with recurrent TET. Giaccone et al. investigated thymic carcinoma cases and reported a response rate of 22.5%. Cho et al. evaluated patients with thymomas and thymic carcinomas, and they reported response rates of 28.6% and 19.2%, respectively. The incidence of adverse events was elevated in thymoma compared to thymic carcinoma (71.4% v 15.4%, respectively) [12,13,21][4][5][13]. Both trials identified an association of high PD-L1 expression with enhanced oncological response, but did not associate PD-L1 expression to adverse events. For those reasons, the development of pertinent biomarkers that could select patients eligible for this treatment is of paramount importance.

2. The Impact of PD-L1 Expression and the Presence of Different Cell Types in the TME of TET

PD-L1 is an immunomodulatory glycoprotein expressed on antigen-presenting cells. PD-L1 binds to the PD-1 receptor, which is expressed on the surface of T cells, and plays an important role in immune response modulation through the suppression of cytokine production, T-cell proliferation, and T-cell adhesion [11,22,23,24,25,26][3][14][15][16][17][18]. In general, the TME of subtypes A, AB, B1, and B2 thymomas is infiltrated mainly by immature T-cells (CD4+ CD8+), which means that their immune status simulates the normal thymocytes [27][19]. In the case of advanced WHO histological types (B3 thymomas and thymic carcinomas), there are fully differentiated cytotoxic T-cells. On the contrary, the role of B-cells is largely unknown. They are present in a minority of thymomas, but their role is clearer in the development of autoimmune conditions [28][20].
Arbour et al. investigated the expression of PD-L1 in the membrane of tumor cells, CD3+ and CD8+ tumor-infiltrating lymphocytes (TILs), co-stimulatory such as CD137, glucocorticoid-induced TNFR-related protein (GITR) and inducible T cell costimulator (ICOS), and co-inhibitory immune checkpoint molecules (PD-1, CTLA-4, and T cell immunoglobulin and mucin-domain-containing protein, TIM-3) in tissue samples from 23 patients with TET (12 thymomas and 11 thymic carcinomas) [11][3]. PD-L1 positivity (≥25% of tumor membrane expression) was frequent in TET (65%), more common in thymomas compared to thymic carcinomas, and associated with longer OS compared to PD-L1 negative tumors [median OS (mOS) 87 months versus 29 months]. The difference in PD-L1 positivity between primary and metastatic tumors was not significant. All tumors contained TILs. TIM-3 was expressed in all TET. Moderate and high TIM-3 expression was observed in 73% of cases. All TET expressed GITR. Moderate and high expression was observed less frequently (52% of cases). Expression of ICOS and CTLA-4 was observed in 91% of cases. Their frequency of moderate and high expression was comparable to those of GITR. In cases where CD3+ TILs (IHC2 or 3 compared to IHC1) were moderately or highly present, then the OS was increased (mOS 80 months versus 43 months). In contrast, the presence of CD8+ TILs did not impact the OS. According to the authors, PD-L1 expression could be considered as a surrogate marker for an “inflamed” TME, which results in an effective host response to the tumor and prolonged survival because of native anti-tumor immunity [11][3].
Inhibitors of PD-1/PD-L1, such as pembrolizumab and nivolumab, act by decreasing the immune escape of tumor cells. Rajan et al. studied the treatment of advanced-stage thymomas by administrating the PD-L1 antibody avelumab. The study demonstrated a contrast of immune cell subgroups between patients that responded to treatment compared to those that did not. More specifically, patients with a response to treatment had decreased proportions of regulatory dendritic cells, T cells, B cells, and natural killer cells prior to treatment [26][18].
Yokoyama et al. analyzed 44 cases of thymomas. They demonstrated that high PD-L1 expression was statistically associated with advanced Masaoka stages III/IV and WHO histological types (type B2 or B3 thymoma). Disease-free survival after complete resection in high PD-L1 expression was significantly worse than in low PD-L1 expression, although there was no significant difference in OS. According to the authors, high PD-L1 expression was qualified as an independent risk factor for recurrence [22][14].

3. Tumor Mutational Burden (TMB) and Immune Infiltration

Tumor mutational burden (TMB) and immune infiltration are critical elements of cancerogenesis [34][21]. TMB is defined as the amount of gene mutation that occurs in the genome of a cancer cell. Consequently, TMB can be a marker of the capacity of tumors to produce new antigens [34][21]. An elevated TMB is responsible for the production of more antigens, and, therefore, the capacity of the tumor to provoke an immune response is increased. As a result, the T cell response and anti-tumor response is stronger, and, thus, the tumor is more suitable for immunotherapy treatment, and the benefit from ICI treatment is more probable [35,36][22][23]. TMB could eventually predict the effect of immunotherapy. In the study of Wang et al., the TET samples were classified according to the median value into low- and high-TMB groups. It was demonstrated that there is a significant association between TMB and more advanced clinical stage (Masaoka-Koga), more advanced pathological type, and increased patient age. In contrast, no notable difference in terms of PD-L1 expression was observed between the two groups. The long-term prognosis of high-TMB patients was significantly lower compared to the low-TMB group. The low- and high-TMB groups also presented significant dissimilarities in terms of infiltration levels of plasma cells, activated NK cells, macrophages, resting mast cells, activated mast cells, neutrophils, T cells CD4 naive, regulatory T cells, naive B cells, and B cell memory [37][24]. Increased CD8+ and CD20+ in tumor islets and in stroma that surrounds cancer cells, decreased CD204+ in tumor islets, high Forkhead box P3+ (FOXP3+), increased presence of CD8+/CD204+, and high CD20+/CD204+ in stroma are considered predictive factors of tumor recurrence in the study of Sato et al., who investigated the evolution of thymic carcinomas [38][25].

4. Tumor-Associated Macrophages (TAMs)

As already mentioned, the TME contains malignant cells, immune cells, mesenchymal cells, and extracellular matrix (ECM) with different functions, and plays a crucial role in the different stages of tumor growth, invasion, and metastasis. Tumor-associated macrophages (TAMs) are a subtype of immune cells that infiltrate tumor tissues, and, thus, they are abundantly present in the TME. In the study of Wang et al., the cluster with a high TMB had increased macrophage presence [34][21]. M1 is considered to be capable of eliminating bacteria and tumor cells, and can release many different proinflammatory cytokines. In contrast, M2 inhibits the immune response and promotes angiogenesis and tissue remodeling [39][26]. Thymic carcinoma hosts an increased proportion of CD163+ compared to thymomas. It is also demonstrated that TAMs can secrete different factors, such as epidermal growth factor (EGF) and transforming growth factor beta 1 (TGF-B1), that promote cell proliferation and survival [40][27]. Consequently, TAMs are necessary for the growth of many tumor types. In addition, TAMs have a major role in tumor invasiveness because they stimulate the overexpression of proteolytic enzymes, plasmin, matrix metalloproteinases (MMPs), urokinase-type plasminogen activator (uPA), and their receptors. As a result, tumor cells are capable of degrading the extracellular matrix, destroying the basement membrane, and invading the surrounding tissue [41][28]. Other studies have demonstrated that the activation of the phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) signal transduction pathway can result in M2 polarization of macrophages, while inhibition of the same pathway leads to M1 polarization [42,43][29][30]. Therefore, the associations among TAMs, TMB, and long-term prognosis could be the basis of further investigation.

5. The Heat Shock Protein 27 and 70 (HSP27 and 70) Expression

Heat Shock Proteins (HSP) have anti-apoptotic properties and, thus, effectuate a key action in tumor initiation, survival, and metastasis [44,45,46,47,48][31][32][33][34][35]. These are proteins that exist in almost all types of nucleated cells, and they are expressed in many types of malignancies, such as colon cancer, prostate cancer, hepatocellular carcinoma, lung cancer, and oral squamous cell carcinoma. The overexpression of HSP in cancer cells also accounts for resistance to chemotherapy, metastatic potential, and poor survival [44,45,46,47,48,49][31][32][33][34][35][36]. Janik et al. investigated the expression of HSP27 and 70 in 101 patients with TET [49][36]. HSP27 and 70 were both expressed in endothelial cells, fibroblasts, adipocytes, and macrophages, but were absent in lymphocytes. The strongest HSP70 expression in cells of the TME was observed in macrophages, especially in type AB thymomas. In contrast, the lowest HSP70 expression in fibroblasts was found in type B3 thymomas, and it was significantly weaker compared to fibroblasts of the TME in type AB thymomas. HSP70 expression in the cytoplasm of TET was higher than in stromal cells. However, the expression of HSP27 in stromal cells was significantly higher compared to HSP70.

6. Fibronectin

Fibronectin (FN) is an attractive target of the stroma. It is a protein that is profusely expressed on blood neo-vessels of cancer tissues [51][37]. FN is considered to be undetectable in adult healthy tissues, apart from specific conditions, such as tissue remodeling and repair, fibrosis, and cell migration [52,53][38][39]. The extra-domain B fibronectin (ED-B FN) is abundantly expressed in TET, with a predominant expression in the stromal cells of the thymoma microenvironment [53][39]. Petrini et al. selected 11 patients with recurrent TET enrolled in a prospective theragnostic phase I/II trials with radretumab, an ED-B FN specific recombinant human antibody. Radretumab radioimmunotherapy (R-RIT) was administered to eligible patients. High ED-B FN expression of the target in the peripherical microenvironment was confirmed by immunohistochemistry. The target was more abundantly expressed in B3 thymomas. According to the authors, thymomas induce stromal cells to shift FN production to the ED-B subtype, which is a crucial step for tumor progression and spread. However, despite the demonstration of the target expression, the R-RIT proved to be inefficacious in terms of objective responses. As the authors advocate, one of the major obstacles in developing a TME-specific treatment strategy is the presence of tumor heterogeneity [53][39].

7. SOX (SRY-Related High-Mobility Group Box) 9

SOX9 is an HMG-box transcription factor in the SOX family of proteins, and it plays a key role in tumor development and progression, including tumor initiation, TME regulation, metastasis, and drug resistance [54,55,56,57][40][41][42][43]. Yuan et al. used the immunohistochemistry method to demonstrate that SOX9 was profusely expressed in the epithelial thymic cells, especially in the epithelial cells of Hassall’s corpuscles. Moreover, SOX9 was largely expressed in the nuclei of TET tumor cells, and, according to the authors, it could be a diagnostic marker for thymomas [54][40]. Additionally, high expression of SOX9 was observed in 70% of type A, 50% of type AB, 22.22% of type B2 thymomas, and 45% of thymic carcinomas cases. Moreover, it was revealed that patients with increased SOX9 expression had shorter mOS. The authors advocate that SOX9 expression is correlated with the histological type of thymomas, and, consequently, it could be a dismal prognostic marker for thymomas. In conclusion, the authors suggest that SOX9 expression might be associated with an immune suppressive microenvironment of thymomas.

References

  1. Meng, F.; Wang, S.; Zhang, J.; Yan, Y.; Wang, C.; Yang, C.; Guan, Z.; Wang, C. Alteration in gene expression profiles of thymoma: Genetic differences and potential novel targets. Thorac. Cancer 2019, 10, 1129–1135.
  2. Hou, X.; Lin, S.; Liu, Y.; Wang, K.; Yu, Z.; Jia, J.; Yu, J.; Zheng, W.; Bai, J.; Chang, L.; et al. Analysis of the tumor microen-vironment and mutation burden identifies prognostic features in thymic epithelial tumors. Am. J. Cancer Res. 2022, 12, 2387–2396.
  3. Arbour, K.C.; Naidoo, J.; Steele, K.E.; Ni, A.; Moreira, A.L.; Rekhtman, N.; Robbins, P.B.; Karakunnel, J.; Rimner, A.; Huang, J.; et al. Expression of PD-L1 and other immunotherapeutic targets in thymic epithelial tumors. PLoS ONE 2017, 12, e0182665.
  4. Giaccone, G.; Thompson, J.; McGuire, C.; Manning, M.; Kallakury, B.; Chahine, J.J.; Subramaniam, D.S.; Liu, S.V.; Gibney, G.T.; Kim, C.; et al. Pembrolizumab in patients with recurrent thymic carcinoma: Results of a phase II study. J. Clin. Oncol. 2017, 35, 8573.
  5. Giaccone, G.; Kim, C.; Thompson, J.; McGuire, C.; Kallakury, B.; Chahine, J.J.; Manning, M.; Mogg, R.; Blumenschein, W.M.; Tan, M.T.; et al. Pembrolizumab in patients with thymic carcinoma: A single-arm, single-centre, phase 2 study. Lancet Oncol. 2018, 19, 347–355.
  6. Tateo, V.; Manuzzi, L.; De Giglio, A.; Parisi, C.; Lamberti, G.; Campana, D.; Pantaleo, M. Immunobiology of Thymic Epithelial Tumors: Implications for Immunotherapy with Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2020, 21, 9056.
  7. Bedekovics, J.; Beke, L.; Mokanszki, A.; Szilagyi, S.; Mehes, G. Programmed Death-ligand 1 (PD-L1) Expression in Thymic Epithelial Tumors. Appl. Immunohistochem. Mol. Morphol. 2020, 28, 1–9.
  8. Padda, S.K.; Riess, J.W.; Schwartz, E.J.; Tian, L.; Kohrt, H.E.; Neal, J.W.; West, R.B.; Wakelee, H.A. Diffuse High Intensity PD–L1 Staining in Thymic Epithelial Tumors. J. Thorac. Oncol. 2015, 10, 500–508.
  9. Katsuya, Y.; Fujita, Y.; Horinouchi, H.; Ohe, Y.; Watanabe, S.-I.; Tsuta, K. Immunohistochemical status of PD-L1 in thymoma and thymic carcinoma. Lung Cancer 2015, 88, 154–159.
  10. Yokoyama, S.; Miyoshi, H.; Nakashima, K.; Shimono, J.; Hashiguchi, T.; Mitsuoka, M.; Takamori, S.; Akagi, Y.; Ohshima, K. Prognostic Value of Programmed Death Ligand 1 and Programmed Death 1 Expression in Thymic Carcinoma. Clin. Cancer Res. 2016, 22, 4727–4734.
  11. Su, Y.; Ou, Y.; Chen, Y.; Ma, X. Construction of immune-related LncRNAs classifier to predict prognosis and immunotherapy response in thymic epithelial tumors. Biosci. Rep. 2022, 42, BSR20220317.
  12. Katsuya, Y.; Horinouchi, H.; Seto, T.; Umemura, S.; Hosomi, Y.; Satouchi, M.; Nishio, M.; Kozuki, T.; Hida, T.; Sukigara, T.; et al. Single-arm, multicentre, phase II trial of nivolumab for unresectable or recurrent thymic carcinoma: PRIMER study. Eur. J. Cancer 2019, 113, 78–86.
  13. Cho, J.; Kim, H.S.; Ku, B.M.; Choi, Y.-L.; Cristescu, R.; Han, J.; Sun, J.-M.; Lee, S.-H.; Ahn, J.S.; Park, K.; et al. Pembrolizumab for Patients With Refractory or Relapsed Thymic Epithelial Tumor: An Open-Label Phase II Trial. J. Clin. Oncol. 2019, 37, 2162–2170.
  14. Yokoyama, S.; Miyoshi, H.; Nishi, T.; Hashiguchi, T.; Mitsuoka, M.; Takamori, S.; Akagi, Y.; Kakuma, T.; Ohshima, K. Clinicopathologic and Prognostic Implications of Programmed Death Ligand 1 Expression in Thymoma. Ann. Thorac. Surg. 2016, 101, 1361–1369.
  15. Chen, H.F.; Wu, L.X.; Li, X.F.; Zhu, Y.C.; Pan, W.W.; Wang, W.X.; Xu, C.W.; Huang, J.H.; Wu, M.H.; Du, K.Q. PD-L1 Ex-pression Level in Different Thymoma Stages and Thymic Carcinoma: A Meta-Analysis. Tumori 2020, 106, 306–311.
  16. Yokoyama, S.; Miyoshi, H. Thymic tumors and immune checkpoint inhibitors. J. Thorac. Dis. 2018, 10 (Suppl. S13), S1509–S1515.
  17. Weissferdt, A.; Fujimoto, J.; Kalhor, N.; Rodriguez, J.; Bassett, R.; Wistuba, I.; Moran, C. Expression of PD-1 and PD-L1 in thymic epithelial neoplasms. Mod. Pathol. 2017, 30, 826–833.
  18. Rajan, A.; Heery, C.R.; Thomas, A.; Mammen, A.L.; Perry, S.; O’Sullivan Coyne, G.; Guha, U.; Berman, A.; Szabo, E.; Madan, R.A.; et al. Efficacy and Tolerability of Anti-programmed Death-Ligand 1 (PD-L1) Antibody (Avelumab) Treatment in Advanced Thymoma. J. Immunother. Cancer 2019, 7, 269.
  19. Masaoutis, C.; Palamaris, K.; Kokkali, S.; Levidou, G.; Theocharis, S. Unraveling the Immune Microenvironment of Thymic Epithelial Tumors: Implications for Autoimmunity and Treatment. Int. J. Mol. Sci. 2022, 23, 7864.
  20. Fend, F.; Kirchner, T.; Marx, A.; Müller-Hermelink, H.-K. B-cells in thymic epithelial tumours. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 1993, 63, 241–247.
  21. Wang, Z.; Xu, Q.; Kaul, D.; Ismail, M.; Badakhshi, H. Significance of tumor mutation burden and immune infiltration in thymic epithelial tumors. Thorac. Cancer 2021, 12, 1995–2006.
  22. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608.
  23. Buttner, R.; Longshore, J.W.; Lopez-Rios, F.; Merkelbach-Bruse, S.; Normanno, N.; Rouleau, E.; Penault-Llorca, F. Imple-menting TMB measurement in clinical practice: Considerations on assay requirements. ESMO Open 2019, 4, e000442.
  24. Wang, X.; Chen, M.; Zhou, J.; Zhang, X. HSP27, 70 and 90, anti-apoptotic proteins, in clinical cancer therapy (Review). Int. J. Oncol. 2014, 45, 18–30.
  25. Sato, J.; Kitano, S.; Motoi, N.; Ino, Y.; Yamamoto, N.; Watanabe, S.; Ohe, Y.; Hiraoka, N. CD20 + Tumor-infiltrating Immune Cells and CD204 + M2 Macrophages Are Associated with Prognosis in Thymic Carcinoma. Cancer Sci. 2020, 111, 1921–1932.
  26. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35.
  27. Goswami, S.; Sahai, E.; Wyckoff, J.B.; Cammer, M.; Cox, D.; Pixley, F.J.; Stanley, E.R.; Segall, J.E.; Condeelis, J.S. Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res. 2005, 65, 5278–5283.
  28. Migita, T.; Sato, E.; Saito, K.; Mizoi, T.; Shiiba, K.-I.; Matsuno, S.; Nagura, H.; Ohtani, H. Differing expression of MMPs-1 and -9 and urokinase receptor between diffuse- and intestinal-type gastric carcinoma. Int. J. Cancer 1999, 84, 74–79.
  29. Souza-Moreira, L.; Soares, V.C.; Dias, S.; Bozza, P.T. Adipose-derived mesenchymal stromal cells modulate lipid metabolism and lipid droplet biogenesis via AKT/mTOR -PPARgamma Signalling in macrophages. Sci. Rep. 2019, 9, 20304.
  30. Chen, W.; Ma, T.; Shen, X.N.; Xia, X.F.; Xu, G.D.; Bai, X.L.; Liang, T.B. Macrophage-induced tumor angiogenesis is reg-ulated by the TSC2-mTOR pathway. Cancer Res. 2012, 72, 1363–1372.
  31. Lazaris, A.C.; Theodoropoulos, G.E.; Davaris, P.S.; Panoussopoulos, D.; Nakopoulou, L.; Kittas, C.; Golematis, B.C. Heat shock protein 70 and HLA-DR molecules tissue expression. Dis. Colon Rectum 1995, 38, 739–745.
  32. Glaessgen, A.; Jonmarker, S.; Lindberg, A.; Nilsson, B.; Lewensohn, R.; Ekman, P.; Valdman, A.; Egevad, L. Heat shock proteins 27, 60 and 70 as prognostic markers of prostate cancer. Apmis 2008, 116, 888–895.
  33. Joo, M.; Chi, J.G.; Lee, H. Expressions of HSP70 and HSP27 in Hepatocellular Carcinoma. J. Korean Med Sci. 2005, 20, 829–834.
  34. Małusecka, E.; Krzyzowska-Gruca, S.; Gawrychowski, J.; Fiszer-Kierzkowska, A.; Kołosza, Z.; Krawczyk, Z. Stress proteins HSP27 and HSP70i predict survival in non-small cell lung carcinoma. Anticancer Res. 2008, 28, 501–506.
  35. Mohtasham, N.; Babakoohi, S.; Montaser-Kouhsari, L.; Memar, B.; Salehinejad, J.; Rahpeyma, A.; Khageh-Ahmady, S.; Marouzi, P.; Firooz, A.; Pazoki-Toroudi, H.; et al. The expression of heat shock proteins 27 and 105 in squamous cell carci-noma of the tongue and relationship with clinicopathological index. Med. Oral. Pathol. Oral. Cir. Bucal. 2011, 16, 730–735.
  36. Janik, S.; Schiefer, A.I.; Bekos, C.; Hacker, P.; Haider, T.; Moser, J.; Klepetko, W.; Müllauer, L.; Ankersmit, H.J.; Moser, B. HSP27 and 70 expression in thymic epithelial tumors and benign thymic alterations: Diagnostic, prognostic and physiologic implications. Sci. Rep. 2016, 6, 24267.
  37. Rick, J.W.; Chandra, A.; Ore, C.D.; Nguyen, A.T.; Yagnik, G.; Aghi, M.K. Fibronectin in malignancy: Cancer-specific alterations, protumoral effects, and therapeutic implications. Semin. Oncol. 2019, 46, 284–290.
  38. Menrad, A.; Menssen, H.D. ED-B fibronectin as a target for antibody-based cancer treatments. Expert Opin. Ther. Targets 2005, 9, 491–500.
  39. Petrini, I.; Sollini, M.; Bartoli, F.; Barachini, S.; Montali, M.; Pardini, E.; Burzi, I.S.; Erba, P.A. ED-B-Containing Isoform of Fibronectin in Tumor Microenvironment of Thymomas: A Target for a Theragnostic Approach. Cancers 2022, 14, 2592.
  40. Yuan, X.; Huang, L.; Luo, W.; Zhao, Y.; Nashan, B.; Yu, F.; Liu, Y. Diagnostic and Prognostic Significances of SOX9 in Thymic Epithelial Tumor. Front. Oncol. 2021, 11.
  41. Grimm, D.; Bauer, J.; Wise, P.; Krüger, M.; Simonsen, U.; Wehland, M.; Infanger, M.; Corydon, T.J. The role of SOX family members in solid tumours and metastasis. Semin. Cancer Biol. 2019, 67, 122–153.
  42. Panda, M.; Tripathi, S.K.; Biswal, B.K. SOX9: An emerging driving factor from cancer progression to drug resistance. Biochim. Biophys. Acta Rev. Cancer 2021, 1875, 188517.
  43. Matheu, A.; Collado, M.; Wise, C.; Manterola, L.; Cekaite, L.; Tye, A.J.; Canamero, M.; Bujanda, L.; Schedl, A.; Cheah, K.S.; et al. Oncogenicity of the Developmental Transcription Factor Sox9. Cancer Res. 2012, 72, 1301–1315.
More
Video Production Service