Cytokines Drive Angiogenesis: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Cytokines are important molecular players in cancer development, progression, and potential targets for treatment. Despite being small and overlooked, research has revealed that cytokines influence cancer biology in multiple ways. Cytokines are often found to contribute to immune function, cell damage, inflammation, angiogenesis, metastasis, and several other cellular processes important to tumor survival. Cytokines have also proven to have powerful effects on complex tumor microenvironment molecular biology and microbiology.  Angiogenesis, also termed neovascularization, is blood vessel development from pre-existing vasculature. It is regulated by a careful balance of pro- and anti-angiogenic factors. In this paper, we will discuss the role of several cytokines in angiogenesis.

  • angiogenesis
  • cytokines
  • inflammation
  • tumor microenvironment

1. Introduction

In 2020, an estimated 19.3 million new cancer cases and nearly 10 million cancer deaths occurred worldwide [1]. Due to cancer’s increasing global impact, understanding factors that drive cancer development has never been more critical. Cancer is characterized by improper regulation of cell differentiation, proliferation, apoptosis avoidance, growth suppressor evasion, increased vasculature, invasion, metastasis, reprogrammed cellular metabolism, and immune evasion [2]. Many of these cancer-promoting processes are highly regulated by cytokines, small protein molecular messengers produced by both normal cells and cancer cells. Cytokines facilitate various interactions between cancer cells, immune cells, and non-immune cells [3,4]. In normal immune activation, cytokines regulate T-cell activation, priming, and CD4+ differentiation and, therefore, play an important role in anticancer immunity [5]. In addition to T cells, cytokines are secreted by other immune cells. For example, tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and myeloid-derived suppressor cells (MDSCs) secrete chemokines C–C motif chemokine ligand 2 (CCL2), CCL4, and CCL5 and inflammatory cytokines tumor necrosis factor α (TNF-α), transforming growth factor β (TGF-β), interleukin-1β (IL-1β), IL-6, and IL-23 to activate T helper 17 (Th17) cell expansion [6]. Generally, pro-inflammatory cytokines mediate key immune interactions to promote antitumor activity. In contrast, cytokines secreted by cells in the tumor microenvironment (TME) and some normal cells promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, tumor progression, and maintain cancer stem-cell-like (CSCs) cells [7]. There are several cytokines involved in these processes. Some cytokines even have complex dual roles and may be involved in both immune activation and cancer development, while others are undergoing investigation to become novel therapeutic targets to treat cancer by disrupting cancer processes. Overall, cytokines are heavily involved in multiple aspects of cancer development and may drive carcinogenesis or promote antitumorigenic effects.

2. Cytokines Drive Angiogenesis

Angiogenesis, also termed neovascularization, is blood vessel development from pre-existing vasculature. It is regulated by a careful balance of pro- and anti-angiogenic factors. Normal wound healing, tissue regeneration, and physiological growth are associated with angiogenesis; however, unbalanced angiogenesis is a prerequisite for solid tumor development and, therefore, an important research area [8,9]. Tumor vasculature is characterized by an abnormal basement membrane, discontinuous and irregularly shaped endothelium, and tortuous hyperpermeable blood vessels that leak nutrients to support cancer growth, development, and resistance [10]. Tumor blood vessels are disorganized and difficult to identify as arterioles, venules, or capillaries [11]. A diverse group of cells, including endothelial cells, cancer cells, cancer-associated adipocytes, fibroblasts, neutrophils, and macrophages influence tumor vasculature morphology and functionality (Figure 1) [12,13,14,15]. For example, TAMs promote angiogenesis by secreting various pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), metalloproteinases (MMPs), TGF-β, platelet-derived growth factor (PDGF), and adrenomedullin (ADM) [16]. Overall, angiogenesis is a complicated process that involves a diverse group of cells that secrete various angiogenic factors and cytokines. Here, we discuss some major cytokines that are specifically involved in angiogenesis.
Figure 1. The TME is composed of a diverse group of cells, including cancer cells, CSCs, adipocytes, neutrophils, TAMs, CAFs, and lymphocytes. Various cytokines secreted by these cells enhance angiogenesis and overall tumor progression.

2.1. Angiogenesis and the VEGF Family

The VEGF family includes VEGF-A, -B, -C, -D, and placental growth factor (PIGF). VEGF-A is known to bind VEGF receptors 1 and 2, while VEGF-B and PIGF bind VEGF receptor 1 only [17]. Under oxygen-deprived conditions, hypoxia-inducible factors (HIF-1α and HIF-2α) induce TAMs to increase VEGF-A secretion and, thus, promote angiogenesis [18]. CAFs are major stromal resident cells that secrete several aggressive angiogenic factors, including FGF2, FGF7, IL-6, and VEGF-A [19]. Several studies indicate that VEGF regulates angiogenesis in several cancer types. For example, VEGF serum levels are correlated with vascular invasion, metastases, tumor stage, and tumor grade of bladder cancer [20]. One study indicated that TAMs overexpressing VEGF-C promoted lymphovascularization in Merkel cell carcinoma [21,22]. As mentioned previously, MMPs also play a major role in driving angiogenesis and may be related to VEGF levels. Early chicken chorioallantoic membrane models and mouse models demonstrated that MMP-2 downregulation and deficiencies are associated with tumor angiogenesis [23,24]. Interestingly, MMP-9- and MT1-MMP-deficient mice present decreased angiogenesis, compared with wild type [25,26,27]. One study proposed that MMP-2 may modify VEGF expression after observing reduced VEGF expression in A549 lung cancer xenograft tissue samples from mice treated with MMP-2 siRNA [28]. Several studies demonstrate that MMPs enhance VEGF production; however, the exact mechanism is unknown [29]. In addition to angiogenesis in solid tumors, VEGF may be associated with hematological malignancies [30]. Recently, Filipiak et al. used an enzyme-linked immunosorbent assay (ELISA) to measure VEGF-A levels in 42 Hodgkin’s lymphoma patients and found that VEGF-A concentration was significantly elevated compared to healthy patient levels [31]. Another study showed that treating Raji cells with oxacetaxine and curcumin, which have antiproliferative effects on lymphoma cells, results in suppressed VEGF-A levels in exosomes derived from Raji cells, and decreases phosphorylated VEGF receptor 2 (p-VEGFR2) levels [32]. VEGF may also be used to predict the prognosis of diffuse large B cell lymphoma (DLBCL) patients. Sang et al. performed a retrospective study and discovered that upregulated VEGF was related to poor therapeutic response and survival of DLBCL patients [33]. In summary, extensive research has identified VEGF as an angiogenesis biomarker and potential therapeutic target.
A recent preclinical study showed that propofol, a common intravenous anesthetic, could inhibit VEGF/VEGFR2- and mTOR/eIF4E-mediated signaling pathways to induce anti-angiogenic activity [34]. However, the relationship between propofol and angiogenesis will require further investigation because both anti-angiogenic and pro-angiogenic effects are reported [35,36]. Anti-VEGF antibodies may target VEGF and inhibit angiogenesis. For example, humanized mAb bevacizumab targets VEGF-A isoforms and was approved in the US in 2004 to be used in combination with chemotherapy for colorectal cancer treatment. Today, it is also approved to treat ovarian, cervical, glioblastoma, and renal cancers [20]. Bevacizumab is also being tested as a potential treatment option for other cancers (Table 1). In one case study, a 21-year-old female with central nervous system (CNS) acute myeloid leukemia (AML) relapse was treated with bevacizumab and intrathecal (IT) chemotherapy. As a result, she remained in complete remission for nearly 1 year. Results suggest that bevacizumab may prove to be a good combination treatment for AML patients [37].
Table 1. Representative clinical trials: cytokine targeted immunotherapy strategies in monotherapy or combination (as of April 2022) may prove promising for the treatment of various cancers.
PIGF is a potential prognostic cancer biomarker, like other VEGF family members, and is involved in endothelial stimulation, bone marrow-derived cell activation, and angiogenesis [38]. In one study, the serum levels of PIGF from 49 clear cell ccRCC patients were tested before surgery and 3 months post-surgery. Before surgery, patients with primary metastatic ccRCC had significantly elevated PIGF levels, compared with localized, without-relapse ccRCC patients [39]. Phage-display technology identified PIGF-specific nanobodies in the nM range that demonstrate anti-angiogenic activity [40,41]. Humanized anti-PIGF antibody TB-403 was tested in patients with advanced solid tumors in phase I clinical trial and pediatric patients with relapsed or refractory medulloblastoma (NCT02748135) [42]. Results are expected to be released in 2022 [43].

2.2. Angiogenesis and Other Major Cytokines

Hepatocyte growth factor (HGF) is a scatter factor that promotes EMT, by activating metalloproteinases, and potently binds to heparin-binding angiogenic factor [44]. HGF is a stromal-cell-derived cytokine and the natural endogenous ligand for mesenchymal–epithelial transition (MET) receptor tyrosine kinase (RTK), which is encoded by the MET proto-oncogene on human chromosome 7 [45]. HGF/MET signaling activation drives angiogenesis, cell proliferation, and tumor aggressiveness [46]. Elevated levels are associated with poor colorectal and lung cancer survival [47,48]. Recently, Katayama et al. revealed that high HGF serum levels in muscle-invasive bladder cancer (MIBC) patients are associated with worse cancer-specific survival, recurrence-free survival, and overall survival [49]. Another study found that patients with gastric cancer have elevated plasma HGF levels, compared with patients with normal gastric mucosa or gastric ulcers [50]. Developing novel treatments targeting both HGF-dependent and HGF-independent MET activation will be important for future treatments targeting the oncogenic-driving MET pathway [51].
Interleukins are involved in multiple cancer processes, including angiogenesis. The IL-1 family comprises IL-1α, IL-1β, and IL-1 receptor antagonist (IL-1RA). IL-1α and IL-1β are cancer-promoting and drive angiogenesis, tumor progression, and tumor aggressiveness [52]. CAFs and adipocytes are known to promote these processes through IL-1β secretion [53]. Early studies of B16 melanoma mouse models showed that IL-1β is required for angiogenesis via lymphotoxin and VEGF-A induction, while IL-1α promoted a similar but weaker phenotype [54,55]. Endothelial cells act as both direct and indirect targets of IL-1 signaling and produce VEGF upon activation (Figure 2). IL-1 and VEGF may synergize to enhance angiogenesis [53]. IL-1β/IL-1R1 signaling induces additional IL-1β transcription in macrophages, which promotes fibroblasts and endothelial cells to also contribute to angiogenesis [56]. Recently, Machelke et al. found that epidermal growth factor receptor (EGFR) inhibition in A549 lung cancer cells leads to reduced IL-1β-induced tissue factor (TF) expression, which normally promotes tumor progression and angiogenesis [57]. The Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) trial evaluated anti-IL-1β to treat atherosclerosis, and results indicated that treatment significantly reduced lung cancer incidence [52,58].
Figure 2. Several cytokines, cell types, and signaling pathways drive angiogenesis. Increased vasculature supports tumor growth and aggressiveness. Bevacizumab may be used to block VEGF, while TB-403 may potentially be used to inhibit PIGF to further prevent angiogenesis in TMEs.
Other interleukins, such as IL-6 and IL-8, may also mediate angiogenesis. Early studies showed that serum IL-6 and VEGF levels are related. IL-6 may induce VEGF expression to increase vasculature [59]. IL-6 is known to transmit signals through several signaling pathways, including JAK/STAT, RAS/MAPK, PI3K/Akt, and NF-κB, to drive tumor progression [60]. IL-6 binds to the IL-6 receptor (IL-6R), which results in the release of its associated Janus kinase (JAK) to phosphorylate transcription 3 (STAT3) and initiates downstream signals to promote angiogenesis, proliferation, and prevent apoptosis [61]. Li et al. demonstrated that IL-6 increases angiogenesis via the STAT5/P-STAT5 signaling pathway and that 6-phosphofrutcto-2-kinase/fructose-2, 6-bisphosphatase 4 (PFKFB4) expression elicits IL-6 upregulation via NF-κB signaling to increase breast cancer angiogenesis [62]. Several potential treatment strategies to target IL-6 are currently undergoing investigation. Examples include (1) small molecule Madindoline A to inhibit dimerization of IL-6/IL-6R/gp130 complexes, (2) siltuximab and CNTO-136 to inhibit IL-6 activity, and (3) mAb tocilizumab to block IL-6R [61]. IL-8 is a chemokine that binds C–X–C motif chemokine receptor 1 (CXCR1) and CXCR2, which are G-protein coupled receptors displayed on granulocytes, monocytes, and endothelial cells to increase angiogenesis, recruit immunosuppressive cells to the tumor site, and worsen prognosis [63]. IL-8 was demonstrated to be the primary cytokine involved in increasing endothelial cell permeability and cell junction disruption in glioblastoma [64]. In human colorectal cancer mouse models, IL-8 induces significant increases in CD31+ peritumoral vasculature, while CXCR2 knockout results in significantly reduced tumor growth, potentially due to lack of IL-8 signaling [65]. Interestingly, gastric cancer cells treated with nicotine, an alkaloid found in tobacco, show enhanced angiogenesis and proliferation in the TME by stimulating IL-8 expression via ROS/NF-κB and ROS/MAPK (Erk1/2, p38)/AP-1 pathways [66]. Like other interleukins, IL-8 may also be targeted for cancer treatment. Recently, escin, a pentacyclic triterpenoid derived from horse chestnut, demonstrated antitumor activity against pancreatic cancer cells by influencing IL-8 expression. Results indicated that escin-treated pancreatic cancer cells had significantly reduced NF-κB activity and IL-8 and VEGF secretion, resulting in inhibited angiogenesis [67].
The angiopoietin family consists of four members that bind to Tie-2 receptors on endothelial cells [44]. Generally, Ang-1 is described as a strong Tie-2 agonist, while Ang-2 is a Tie-2 antagonist that competes with Ang-1. However, recent studies characterize Ang-2 as capable of acting as a Tie-2 agonist or antagonist [68]. In colorectal cancer, Ang-2 expression is negatively associated with patient overall survival [69]. One study demonstrated that glucocorticoid-treated colon cancer-derived myofibroblasts reduce Ang-2 levels and inhibit endothelial cell angiogenesis and cell migration [70]. Ang-4 and Ang-3, a mouse orthologue, are both Tie-2 agonists, but their effect on angiogenesis is not well characterized [71]. A recent study showed that Ang-3 was elevated in cervical cancer cells, compared with normal cervical cells, and Ang-3 silencing inhibited human umbilical vein endothelial cell angiogenesis and integrin alpha v beta 3 (αvβ3). Results also showed that upregulated αvβ3 expression increases VEGF and VEGFR2 secretion and blood vessel formation, suggesting Ang-3 as a potential novel therapeutic target for treating cervical cancer [72]. In another study, Ang-4 was discovered to be overexpressed in ovarian cancer cells. Immunoprecipitation results suggested that Ang-4 suppression leads to VEGFR2/VE-cadherin/Src complex dissociation and phosphorylation of VEGFR2 in A2780 and CAOV3 ovarian cancer cell lines. Researchers concluded that Ang-4 silencing significantly inhibits tumor angiogenesis and progression [73].

3. Conclusion

Due to cancer’s global impact, understanding how cancer develops and progresses is critical. Cytokines are heavily involved in regulating several cancer developmental processes. Cytokines have profound effects on cells and may lead to pro- or anti-tumor activity, depending on environmental conditions and the presence of other cytokines. Overall, studies have shown that cytokines are powerful molecular players that often control angiogenesis. The impact and therapeutic potential of cytokines are beginning to be uncovered through already existing and developing cytokine-targeted therapies. In this review, we highlighted only a few cytokines involved in angiogenesis. Further research focusing on the intricate relationship between angiogenesis and cytokines will be required to increase our understanding of how cancer develops. Cytokine research will continue to play a key role in revealing the molecular mechanisms behind cancer development and angiogenesis.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA. Cancer J. Clin. 2021, 71, 209–249.
  2. Hanahan, D. Hallmarks of Cancer: New DimensionsHallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46.
  3. Mascaux, C.; Angelova, M.; Vasaturo, A.; Beane, J.; Hijazi, K.; Anthoine, G.; Buttard, B.; Rothe, F.; Willard-Gallo, K.; Haller, A.; et al. Immune evasion before tumour invasion in early lung squamous carcinogenesis. Nature. 2019, 571, 570–575.
  4. Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in cancer: from biology to therapy. Nat. Rev. Cancer. 2021, 21, 481–499.
  5. Qiu, Y.; Su, M.; Liu, L.; Tang, Y.; Pan, Y.; Sun, J. Clinical Application of Cytokines in Cancer Immunotherapy. Drug Des. Devel. Ther. 2021, 15, 2269.
  6. Chen, C.; Gao, F.H. Th17 Cells Paradoxical Roles in Melanoma and Potential Application in Immunotherapy. Front. Immunol. 2019, 10, 187.
  7. Pradhan, R.; Chatterjee, S.; Hembram, K.C.; Sethy, C.; Mandal, M.; Kundu, C.N. Nano formulated Resveratrol inhibits metastasis and angiogenesis by reducing inflammatory cytokines in oral cancer cells by targeting tumor associated macrophages. J. Nutr. Biochem. 2021, 92, 108624.
  8. Bhat, S.M.; Badiger, V.A.; Vasishta, S.; Chakraborty, J.; Prasad, S.; Ghosh, S.; Joshi, M.B. 3D tumor angiogenesis models: recent advances and challenges. J. Cancer Res. Clin. Oncol. 2021, 147, 3477.
  9. Otrock, Z.K.; Mahfouz, R.A.R.; Makarem, J.A.; Shamseddine, A.I. Understanding the biology of angiogenesis: Review of the most important molecular mechanisms. Blood Cells, Mol. Dis. 2007, 39, 212–220.
  10. Siemann, D.W. The Unique Characteristics of Tumor Vasculature and Preclinical Evidence for its Selective Disruption by Tumor-Vascular Disrupting Agents. Cancer Treat. Rev. 2011, 37, 63.
  11. Konerding, M.A.; Fait, E.; Gaumann, A. 3D microvascular architecture of pre-cancerous lesions and invasive carcinomas of the colon. Br. J. Cancer. 2001, 84, 1354.
  12. 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.
  13. Wang, F.T.; Sun, W.E.I.; Zhang, J.T.; Fan, Y.Z. Cancer-associated fibroblast regulation of tumor neo-angiogenesis as a therapeutic target in cancer. Oncol. Lett. 2019, 17, 3055.
  14. Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor Associated Neutrophils. Their Role in Tumorigenesis, Metastasis, Prognosis and Therapy. Front. Oncol. 2019, 9, 1146.
  15. Hida, K.; Maishi, N.; Torii, C.; Hida, Y. Tumor angiogenesis—characteristics of tumor endothelial cells. Int. J. Clin. Oncol. 2016, 21, 206–212.
  16. Aras, S.; Raza Zaidi, M. TAMeless traitors: macrophages in cancer progression and metastasis. Br. J. Cancer 2017, 117, 1583.
  17. Uemura, A.; Fruttiger, M.; D’Amore, P.A.; De Falco, S.; Joussen, A.M.; Sennlaub, F.; Brunck, L.R.; Johnson, K.T.; Lambrou, G.N.; Rittenhouse, K.D.; et al. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog. Retin. Eye Res. 2021, 84, 100954.
  18. Joshi, S.; Singh, A.R.; Zulcic, M.; Durden, D.L. A Macrophage-Dominant PI3K Isoform Controls Hypoxia-Induced HIF1α and HIF2α Stability and Tumor Growth, Angiogenesis, and Metastasis. Mol. Cancer Res. 2014, 12, 1520–1531.
  19. Fu, R.; Han, C.F.; Ni, T.; Di, L.; Liu, L.J.; Lv, W.C.; Bi, Y.R.; Jiang, N.; He, Y.; Li, H.M.; et al. A ZEB1/p53 signaling axis in stromal fibroblasts promotes mammary epithelial tumours. Nat. Commun. 2019, 10, 3210.
  20. Ghafouri, S.; Burkenroad, A.; Pantuck, M.; Almomani, B.; Stefanoudakis, D.; Shen, J.; Drakaki, A. VEGF inhibition in urothelial cancer: the past, present and future. World J. Urol. 2021, 39, 741–749.
  21. Matsumoto-Okazaki, Y.; Yamane, J.; Kajiya, K. Real-time imaging of interaction between macrophages and lymphatic vessels in an in vitro model to study inflammatory resolution. J. Dermatol. Sci. 2015, 77, 76–79.
  22. Werchau, S.; Toberer, F.; Enk, A.; Dammann, R.; Helmbold, P. Merkel cell carcinoma induces lymphatic microvessel formation. J. Am. Acad. Dermatol. 2012, 67, 215–225.
  23. Itoh, T.; Tanioka, M.; Yoshida, H.; Yoshioka, T.; Nishimoto, H.; Itohara, S. Reduced Angiogenesis and Tumor Progression in Gelatinase A-deficient Mice. Cancer Res. 1998, 58, 1048-1051.
  24. Fang, J.; Shing, Y.; Wiederschain, D.; Yan, L.; Butterfield, C.; Jackson, G.; Harper, J.; Tamvakopoulos, G.; Moses, M.A. Matrix metalloproteinase-2 is required for the switch to the angiogenic phenotype in a tumor model. Proc. Natl. Acad. Sci. U. S. A. 2000, 97, 3884.
  25. Vu, T.H.; Shipley, J.M.; Bergers, G.; Berger, J.E.; Helms, J.A.; Hanahan, D.; Shapiro, S.D.; Senior, R.M.; Werb, Z. MMP-9/Gelatinase B Is a Key Regulator of Growth Plate Angiogenesis and Apoptosis of Hypertrophic Chondrocytes. Cell. 1998, 93, 411.
  26. Zhou, Z.; Apte, S.S.; Soininen, R.; Cao, R.; Baaklini, G.Y.; Rauser, R.W.; Wang, J.; Cao, Y.; Tryggvason, K. Impaired endochondral ossification and angiogenesis in mice deficient in membrane-type matrix metalloproteinase I. Proc. Natl. Acad. Sci. U. S. A. 2000, 97, 4052.
  27. Huang, S.; Van Arsdall, M.; Tedjarati, S.; McCarty, M.; Wu, W.; Langley, R.; Fidler, I.J. Contributions of Stromal Metalloproteinase-9 to Angiogenesis and Growth of Human Ovarian Carcinoma in Mice. JNCI J. Natl. Cancer Inst. 2002, 94, 1134–1142.
  28. Chetty, C.; Lakka, S.S.; Bhoopathi, P.; Rao, J.S. MMP-2 Alters VEGF Expression via αVβ3 Integrin-Mediated PI3K/AKT Signaling in A549 Lung Cancer Cells. Int. J. Cancer 2010, 127, 1081.
  29. Carey, P.; Low, E.; Harper, E.; Stack, M.S. Metalloproteinases in Ovarian Cancer. Int. J. Mol. Sci. 2021, 22, 3403.
  30. Das, S.; Amin, S.A.; Jha, T. Inhibitors of gelatinases (MMP-2 and MMP-9) for the management of hematological malignancies. Eur. J. Med. Chem. 2021, 223, 113623.
  31. Filipiak, J.; Boinska, J.; Ziołkowska, K.; Zduńska, M.; Zarychta, E.; Rość, D. Assessment of endothelial progenitor cells, VEGF-A and SDF-1α in Hodgkin’s lymphoma. Blood Coagul. Fibrinolysis. 2021, 32, 266–272.
  32. Zhang, Y.; Xiang, J.; Zhu, N.; Ge, H.; Sheng, X.; Deng, S.; Chen, J.; Yu, L.; Zhou, Y.; Shen, J. Curcumin in Combination With Omacetaxine Suppress Lymphoma Cell Growth, Migration, Invasion, and Angiogenesis via Inhibition of VEGF/Akt Signaling Pathway. Front. Oncol. 2021, 11, 656045.
  33. Sang, W.; Zhou, H.; Qin, Y.; Shen, Z.; Yan, D.; Sun, C.; Song, X.; Ma, Y.; Tu, D.; Bian, Z.; et al. Risk stratification model based on VEGF and International Prognostic Index accurately identifies low-risk diffuse large B-cell lymphoma patients in the rituximab era. Int. J. Hematol. 2021, 114, 189–198.
  34. Wang, Z.; Cao, B.; Ji, P.; Yao, F. Propofol inhibits tumor angiogenesis through targeting VEGF/VEGFR and mTOR/eIF4E signaling. Biochem. Biophys. Res. Commun. 2021, 555, 13–18.
  35. Chang, C.Y.; Chen, P.H.; Lu, S.C.; Hsieh, M.C.; Lin, C.W.; Lee, H.M.; Jawan, B.; Kao, Y.H. Propofol-enhanced autophagy increases motility and angiogenic capacity of cultured human umbilical vascular endothelial cells. Life Sci. 2015, 142, 49–59.
  36. Gao, Y.; Yu, X.; Zhang, F.; Dai, J. Propofol inhibits pancreatic cancer progress under hypoxia via ADAM8. J. Hepatobiliary. Pancreat. Sci. 2019, 26, 219–226.
  37. Cao, H.-Y.; Tao, T.; Shen, X.-D.; Bai, L.; Wan, C.-L.; Wu, D.-P.; Li, J.-L.; Xue, S.-L. Efficiency of anti-VEGF therapy in central nervous system AML relapse: A case report and literature review. Clin. Case Reports 2022, 10, e05367.
  38. Ruggiero, D.; Nutile, T.; Nappo, S.; Tirozzi, A.; Bellenguez, C.; Leutenegger, A.L.; Ciullo, M. Genetics of PlGF plasma levels highlights a role of its receptors and supports the link between angiogenesis and immunity. Sci. Rep. 2021, 11, 16821.
  39. Cechova, M.; Chocholaty, M.; Babjuk, M.; Zima, T.; Havlova, K.; Koldova, M.; Schmidt, M.; Kalousova, M. Diagnostic and prognostic value of placental growth factor serum concentration in clear cell renal cell carcinoma. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc. Czech. Repub. 2021, 165, 375–379.
  40. Arezumand, R.; Mahdian, R.; Zeinali, S.; Hassanzadeh-Ghassabeh, G.; Mansouri, K.; Khanahmad, H.; Namvar-asl, N.; Rahimi, H.; Behdani, M.; Cohan, R.A.; et al. Identification and characterization of a novel nanobody against human placental growth factor to modulate angiogenesis. Mol. Immunol. 2016, 78, 183–192.
  41. Nikooharf, A.; Arezumand, R.; Mansouri, K.; Khoshi, A.H.; Namdar Ahmadabad, H. Development of a Recombinant Monospecific Anti-PLGF Bivalent Nanobody and Evaluation of it in Angiogenesis Modulation. Mol. Biotechnol. 2020, 62, 580–588.
  42. Lassen, U.; Nielsen, D.L.; Sørensen, M.; Winstedt, L.; Niskanen, T.; Stenberg, Y.; Pakola, S.; Stassen, J.M.; Glazer, S. A phase I, dose-escalation study of TB-403, a monoclonal antibody directed against PlGF, in patients with advanced solid tumours. Br. J. Cancer. 2012, 106, 678.
  43. Ye, X.; Gaucher, J.F.; Vidal, M.; Broussy, S. A Structural Overview of Vascular Endothelial Growth Factors Pharmacological Ligands: From Macromolecules to Designed Peptidomimetics. Molecules. 2021, 26, 6759.
  44. Neufeld, G.; Kessler, O. Pro-angiogenic cytokines and their role in tumor angiogenesis. Cancer Metastasis Rev. 2006, 62, 580-588.
  45. Moosavi, F.; Giovannetti, E.; Saso, L.; Firuzi, O. HGF/MET pathway aberrations as diagnostic, prognostic, and predictive biomarkers in human cancers. Crit. Rev. Clin. Lab. Sci. 2019, 56, 533–566.
  46. Mukai, S.; Yamasaki, K.; Fujii, M.; Nagai, T.; Terada, N.; Kataoka, H.; Kamoto, T. Dysregulation of Type II Transmembrane Serine Proteases and Ligand-Dependent Activation of MET in Urological Cancers. Int. J. Mol. Sci. 2020, 21, 2663.
  47. Tsuji, T.; Sakamori, Y.; Ozasa, H.; Yagi, Y.; Ajimizu, H.; Yasuda, Y.; Funazo, T.; Nomizo, T.; Yoshida, H.; Nagai, H.; et al. Clinical impact of high serum hepatocyte growth factor in advanced non-small cell lung cancer. Oncotarget. 2017, 8, 71805–71816.
  48. Toiyama, Y.; Miki, C.; Inoue, Y.; Okugawa, Y.; Tanaka, K.; Kusunoki, M. Serum hepatocyte growth factor as a prognostic marker for stage II or III colorectal cancer patients. Int. J. Cancer. 2009, 125, 1657–1662.
  49. Katayama, S.; Schuettfort, V.M.; Pradere, B.; Mori, K.; Mostafaei, H.; Quhal, F.; Sari Motlagh, R.; Laukhtina, E.; Grossmann, N.C.; Aydh, A.; et al. Prognostic value of hepatocyte growth factor for muscle-invasive bladder cancer. J. Cancer Res. Clin. Oncol. 2022, 21, 1–12.
  50. Gao, L.-M.; Wang, F.; Zheng, Y.; Fu, Z.-Z.; Zheng, L.; Chen, L.-L. Roles of Fibroblast Activation Protein and Hepatocyte Growth Factor Expressions in Angiogenesis and Metastasis of Gastric Cancer. Pathol. Oncol. Res. 2019, 25, 369–376.
  51. Faiella, A.; Riccardi, F.; Cartenì, G.; Chiurazzi, M.; Onofrio, L. The Emerging Role of c-Met in Carcinogenesis and Clinical Implications as a Possible Therapeutic Target. J. Oncol. 2022, 2022, 1–12.
  52. Zhang, J.; Veeramachaneni, N. Targeting interleukin-1β and inflammation in lung cancer. Biomark. Res. 2022, 10, 5.
  53. Zhang, W.; Borcherding, N.; Kolb, R. IL-1 Signaling in Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1240, 1–23.
  54. Carmi, Y.; Dotan, S.; Rider, P.; Kaplanov, I.; White, M.R.; Baron, R.; Abutbul, S.; Huszar, M.; Dinarello, C.A.; Apte, R.N.; et al. The Role of IL-1β in the Early Tumor Cell–Induced Angiogenic Response. J. Immunol. 2013, 190, 3500–3509.
  55. Voronov, E.; Shouval, D.S.; Krelin, Y.; Cagnano, E.; Benharroch, D.; Iwakura, Y.; Dinarello, C.A.; Apte, R.N. IL-1 is required for tumor invasiveness and angiogenesis. Proc. Natl. Acad. Sci. U. S. A. 2003, 100, 2645.
  56. Young, H.L.; Rowling, E.J.; Bugatti, M.; Giurisato, E.; Luheshi, N.; Arozarena, I.; Acosta, J.C.; Kamarashev, J.; Frederick, D.T.; Cooper, Z.A.; et al. An adaptive signaling network in melanoma inflammatory niches confers tolerance to MAPK signaling inhibition. J. Exp. Med. 2017, 214, 1691.
  57. Mechelke, T.; Wittig, F.; Ramer, R.; Hinz, B. Interleukin-1β Induces Tissue Factor Expression in A549 Cells via EGFR-Dependent and -Independent Mechanisms. Int. J. Mol. Sci. 2021, 22, 6606.
  58. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131.
  59. Salgado, R.; Vermeulen, P.B.; Benoy, I.; Weytjens, R.; Huget, P.; Van Marck, E.; Dirix, L.Y. Platelet number and interleukin-6 correlate with VEGF but not with bFGF serum levels of advanced cancer patients. Br. J. Cancer. 1999, 80, 892–897.
  60. Xia, T.; Li, J.; Ren, X.; Liu, C.; Sun, C. Research progress of phenolic compounds regulating IL-6 to exert antitumor effects. Phyther. Res. 2021, 35, 6720–6734.
  61. Xu, J.; Lin, H.; Wu, G.; Zhu, M.; Li, M. IL-6/STAT3 Is a Promising Therapeutic Target for Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 760971.
  62. Li, D.; Tang, J.; Gao, R.; Lan, J.; Shen, W.; Liu, Y.; Chen, Y.; Sun, H.; Yan, J.; Nie, Y.; et al. PFKFB4 promotes angiogenesis via IL-6/STAT5A/P-STAT5 signaling in breast cancer. J. Cancer. 2022, 13, 212.
  63. Fousek, K.; Horn, L.A.; Palena, C. Interleukin-8: A chemokine at the intersection of cancer plasticity, angiogenesis, and immune suppression. Pharmacol. Ther. 2021, 219, 107692.
  64. Dwyer, J.; Hebda, J.K.; Le Guelte, A.; Galan-Moya, E.M.; Smith, S.S.; Azzi, S.; Bidere, N.; Gavard, J. Glioblastoma Cell-Secreted Interleukin-8 Induces Brain Endothelial Cell Permeability via CXCR2. PLoS One. 2012, 7, 45562.
  65. Lee, Y.S.; Choi, I.; Ning, Y.; Kim, N.Y.; Khatchadourian, V.; Yang, D.; Chung, H.K.; Choi, D.; Labonte, M.J.; Ladner, R.D.; et al. Interleukin-8 and its receptor CXCR2 in the tumour microenvironment promote colon cancer growth, progression and metastasis. Br. J. Cancer. 2012, 106, 1833–1841.
  66. Lian, S.; Li, S.; Zhu, J.; Xia, Y.; Do Jung, Y. Nicotine stimulates IL-8 expression via ROS/NF-κB and ROS/MAPK/AP-1 axis in human gastric cancer cells. Toxicology. 2022, 466, 153062.
  67. Omi, K.; Matsuo, Y.; Ueda, G.; Aoyama, Y.; Kato, T.; Hayashi, Y.; Imafuji, H.; Saito, K.; Tsuboi, K.; Morimoto, M.; et al. Escin inhibits angiogenesis by suppressing interleukin-8 and vascular endothelial growth factor production by blocking nuclear factor-κB activation in pancreatic cancer cell lines. Oncol. Rep. 2021, 45, 55.
  68. Tsakogiannis, D.; Nikolakopoulou, A.; Zagouri, F.; Stratakos, G.; Syrigos, K.; Zografos, E.; Koulouris, N.; Bletsa, G. Update Overview of the Role of Angiopoietins in Lung Cancer. Medicina.. 2021, 57, 1191.
  69. Kim, H.K.; Yang, Y.; Byeon, S.; Jeong, Y.; Kwon, J.; Lee, K.H.; Son, S.M.; Han, H.S. E-Cadherin and Angiopoietin-2 as Potential Biomarkers for Colorectal Cancer With Peritoneal Carcinomatosis. Anticancer Res. 2021, 41, 4497–4504.
  70. Drebert, Z.; MacAskill, M.; Doughty-Shenton, D.; De Bosscher, K.; Bracke, M.; Hadoke, P.W.F.; Beck, I.M. Colon cancer-derived myofibroblasts increase endothelial cell migration by glucocorticoid-sensitive secretion of a pro-migratory factor. Vascul. Pharmacol. 2017, 89, 19.
  71. Lee, H.J.; Cho, C.-H.; Hwang, S.-J.; Choi, H.-H.; Kim, K.-T.; Ahn, S.Y.; Kim, J.-H.; Oh, J.-L.; Lee, G.M.; Koh, G.Y. Biological characterization of angiopoietin-3 and angiopoietin-4. FASEB J. 2004, 18, 1200–1208.
  72. Zhong, L.; Tang, L.; He, X. Angiopoietin-like 3 (ANGPTL3) drives cell proliferation, migration and angiogenesis in cervical cancer via binding to integrin alpha v beta 3. Bioengineered. 2022, 13, 2971–2980.
  73. Wu, Y.; Gao, J.; Liu, X. Deregulation of angiopoietin-like 4 slows ovarian cancer progression through vascular endothelial growth factor receptor 2 phosphorylation. Cancer Cell Int. 2021, 21, 171.

 

This entry is adapted from the peer-reviewed paper 10.3390/cancers14092178

This entry is offline, you can click here to edit this entry!
Video Production Service