[BACK]
BIOCELL
DOI:10.32604/biocell.2022.020209
images
Viewpoint

Role of PTX3 and complement modulation in the tumor microenvironment

GIUSEPPE STEFANO NETTI1,*, FEDERICA SPADACCINO1, VALERIA CATALANO1, GIUSEPPE CASTELLANO2, GIOVANNI STALLONE3 and ELENA RANIERI1

1Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
2Nephrology, Dialysis and Renal Transplant Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico-University of Milan, Milan, 20122, Italy
3Nephrology Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
*Address correspondence to: Giuseppe Stefano Netti, giuseppestefano.netti@unifg.it
Received: 23 December 2021; Accepted: 17 February 2022

Abstract: Pentraxin-3 (PTX3), the prototype of long pentraxins, seems to influence complement system (CS) modulation. PTX3 and CS sustain carcinogenesis, enriching tumor microenvironment (TME) with pro-inflammatory molecules promoting angiogenesis in prostate cancer (PC) and renal cell carcinoma (RCC). Furthermore, cancer cells overexpress complement regulatory proteins, such as CD46, CD55 and CD59, which negatively affect complement pathways for support cancer cells survival. This viewpoint aims to elucidate the ambivalent role of PTX3 and the CS in the context of tumor microenvironment (TME).

Keywords: Pentraxin-3; Complement system; Tumor microenvironment; Prostate cancer; Renal cell carcinoma

Introduction

Pentraxins, a superfamily of evolutionary conserved proteins, are essential components of innate immune system and play a pivotal role in vascular biology (Garlanda et al., 2005; Peri et al., 2000). Pentraxin-3 (PTX3), the prototype of long pentraxins, differs from short pentraxins for gene organization, cellular source, and ligand-binding capacities (Peri et al., 2000; Mantovani et al., 2006). Like short pentraxins, PTX3 facilitates dysregulation of mitogenic signaling pathways, sustains cellular proliferation, angiogenesis, metastasis formation, insensitivity to apoptosis, and tumor escape from immunosurveillance (Baruah et al., 2006; Rutkowski et al., 2010). Unlike short pentraxins, PTX3 is synthesized by a variety of cell types at the site of inflammation (Garlanda et al., 2005; Schenk et al., 2010), whereby it seems to regulate complement system (CS) activation (Garlanda et al., 2005; Rutkowski et al., 2010). Recent findings suggest an insidious relationship between complement and cancer in terms of cellular proliferation and regeneration as well as angiogenesis (Rutkowski et al., 2010; Diamandis et al., 2011).

This viewpoint aims to elucidate the mechanisms responsible for tumor progression focusing on the ambivalent role of PTX3 and the CS in the context of tumor microenvironment (TME).

Presentation

Recently, new investigations in the field of TME highlighted that switching from acute to chronic immunoflogosis continuously provides the tumor with a cytokines-rich milieu therefore promoting cancer survival (Korniluk et al., 2017). Of note, PTX3 has emerged as an ambivalent inflammatory mediator acting as a tumor suppressor and pro-oncogenic factor depending on the TME in which it is found (Ronca et al., 2013; Giacomini et al., 2018) (Fig. 1). PTX3 may modulate the CS by binding to C1q and MBL, thus activating the classical and the lectin pathway, and to CFH, thus affecting the alternative pathway (Ma and Garred, 2018). CS activation leads to the C3 cleavage and C5b-9 complex assembly to direct lyse the pathogens and cell debris empowering the inflammation both at the systemic and locally level.

images

Figure 1: The ambivalent role of PTX3 in cancers. The role of PTX3 in cancer pathogenesis is likely to be context and pathway dependent. PTX3 may act as a tumor suppressor factor by inducing insensitivity to apoptosis or cell-cycle arrest in Glioma, Prostate and Cervical Cancer. Of note, PTX3 may binds to members of fibroblast growth factor family (FGF), including FGF2, and FGF8b by inhibiting FGF-dependent metastasis formation in Mieloma and Melanoma. A modulation in proliferative signaling, involving the most frequently over-activated intracellular pathways PI3K/AKT was reported for lung cancer as well as a decreased tendency to sustain neoangiogenesis for breast cancer. On the contrary, PTX3 seems to act as pro-tumorigenic factor in head/neck carcinoma and cervical carcinoma by acting on epidermal growth factor (EGF) to support neoplastic proliferation. The ability to modulate the complement system leads PTX3 to induce neoangiogenesis and cancer development via the release of the anaphylatoxins in the absence of cell-lysis damage on neoplastic cells in the context of prostate and renal cancer. Of note, the lack of recruitment of FH due to PTX3 deficiency promote carcinogenesis in sarcoma. In addition, an increase of PTX3-mediated inflammation has been reported for gastric and pancreatic cancer.

In the tumour milieu complement mediators both induce an anti-tumour response mainly via their lytic activity by enriching the TME with immunosuppressive factors and promote carcinogenesis by releasing very strong pro-inflammatory molecules, such as the anaphylatoxins C3a and C5a therefore sustaining neoangiogenesis and metastasis formation (Rutkowski et al., 2010). Of note, also PTX3/CFH balance may modulate CS activation as well as CFH different expression may affect cancer development (Bonavita et al., 2015; Daugan et al., 2021b; Riihilä et al., 2019). In the attempt to avoid C5b-9-mediated lysis and to allow cell survival, cancer cells may modulate and/or remove C5b-9 complex by upregulating the membrane bound Complement Regulatory Proteins (mCRPs), including CD55, CD59, CD46 (Gigante et al., 2013; Santangelo et al., 2014; Pio et al., 2014; Castellano et al., 2019; Geller and Yan, 2019) (Fig. 2).

images

Figure 2: Involvement of PTX3 in complement system modulation in cancer. PTX3 is involved in Complement System (CS) activation and regulation by interacting with complement components, influencing inflammation and neoplastic angiogenesis. (A, B) PTX3 can binds to C1q and mannose-binding lectin (MBL) and activate classical pathway and lectin pathway, respectively or (C) can interact with complement regulators, such as Factor H, affecting the alternative pathway. As a consequence of CS activation, the cleavage of C3 and of C5 leads to the deposition of C3a and C5a anaphylotoxins, which sustain inflammation and neoangiogenesis, enhancing tumor growth. In addition, cancer cells overexpress different Membrane bound Complement Regulatory Proteins (mCRPs), including membrane cofactor protein (CD46), decay-accelerating factor (CD55) and protectin (CD59) which inhibit the formation of MAC and cell lysis, supporting cancer cells survival.

The role of CS in sustain carcinogenesis is particularly relevant in the context of genitourinary cancers. As known, patients affected by prostate cancer (PC) show increased level of prostate-specific antigen (PSA), a serine protease that among others is able to cleave C3 and C5 in vitro, acting as complement activating-protease (Manning et al., 2013). In accordance, elevated level of C3 fragments were found in the serum of PC patients (Manning et al., 2013; Karczmarski et al., 2013) and an increased expression of CD55 and CD59 were observed in patients with advanced PC and decreased overall survival (OS) (Loberg et al., 2006; Xu et al., 2005).

The ability of CS in promoting cancer progression by activating angiogenesis is amplified in renal cell carcinoma (RCC) since renal cells may produce all complement effectors mainly triggering the classical pathway (Reese et al., 2020; Tang et al., 1999, Daugan et al., 2021a; Roumenina et al., 2019). The finding of elevated C5a level in ccRCC patients resulted as an adverse prognostic feature (Zhou et al., 2001) related to C5a-mediated immunosurveillance evasion (Corrales et al., 2012; Hezmee et al., 2011). As regard mCRPs, an increased expression of CD55/CD59 and CD46 were correlated to ccRCC progression and tumor stage, respectively (Blok et al., 2000).

Noteworthy, PC and RCC patients showed an increased tissue and serum expression of PTX3, as compared to non-neoplastic patients. Looking into PC patients, elevated PTX3 serum levels were also able to discriminate PC from prostatic inflammation and benign prostatic hyperplasia (BPH) (Stallone et al., 2014; Falagario et al., 2021). Recent studies reported that PTX3 is able to modulate CS in PC and RCC TME since it co-localizes with C1q deposits, CD59 and with C3a-C5a receptors in PC biopsies and RCC tissues (Stallone et al., 2020; Netti et al., 2020). Interestingly, these studies showed no evidence of PTX3 and C5b-9 co-localization in both cancers, thus reinforcing the hypothesis of pro-tumorigenic effect of both CS and PTX3 in sustaining neoangiogenesis in the absence of cell-lysis damage on neoplastic cells.

Conclusion and Future Perspectives

Inflammation and CS have emerged as a double-edged sword acting as tumor-suppressor by generating an immunosuppressive TME or as pro-oncogenic factor promoting cancer development. PTX3 and CS are crucial and ambivalent mediator in PC and RCC. Taken together, these findings suggest a possible key role of PTX3 and CS for cancer progression in the context of TME.

Acknowledgement: The authors thank Dr. Federica Pavone, Dr. Giuseppina Prencipe and Dr. Federica Robusto, for their invaluable collaboration.

Availability of Data and Materials: No data are included within this viewpoint.

Author Contribution: GSN conceived the work and wrote the first draft. FS, VC, GC, GS and ER critically reviewed the draft. All authors contributed to drafting the work, revised the final manuscript, and approved submission.

Ethics Approval: No committees were required for this study.

Funding Statement: This work was supported by grant funding from University of Foggia (University Research Projects 2019 “PRA 2019” granted to G.S.N., 2019). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The contents are solely the responsibility of the author and do not necessarily represent the official views of the University of Foggia.

Conflicts of Interest: The authors declare that they have no conflicts of interest to report regarding the present study.

References

Baruah P, Dumitriu IE, Peri G, Russo V, Mantovani A et al. (2006). The tissue pentraxin PTX3 limits C1q-mediated complement activation and phagocytosis of apoptotic cells by dendritic cells. Journal of Leukocyte Biology 80: 87–95. DOI 10.1189/jlb.0805445. [Google Scholar] [CrossRef]

Blok VT, Daha MR, Tijsma OM, Weissglas MG, van den Broek LJ et al. (2000). A possible role of CD46 for the protection in vivo of human renal tumor cells from complement-mediated damage. Laboratory Investigation 80: 335–344. DOI 10.1038/labinvest.3780038. [Google Scholar] [CrossRef]

Bonavita E, Gentile S, Rubino M, Maina V, Papait R et al. (2015). PTX3 is an extrinsic oncosuppressor regulating complement-dependent inflammation in cancer. Cell 160: 700–714. DOI 10.1016/j.cell.2015.01.004. [Google Scholar] [CrossRef]

Castellano G, Stasi A, Franzin R, Sallustio F, Divella C et al. (2019). LPS-binding protein modulates acute renal fibrosis by inducing pericyte-to-myofibroblast trans-differentiation through TLR-4 signaling. International Journal of Molecular Sciences 20: 3682. DOI 10.3390/ijms20153682. [Google Scholar] [CrossRef]

Corrales L, Ajona D, Rafail S, Lasarte JJ, Riezu-Boj JI et al. (2012). Anaphylatoxin C5a creates a favorable microenvironment for lung cancer progression. Journal of Immunology 189: 4674–4683. [Google Scholar]

Daugan MV, Revel M, Russick J, Dragon-Durey MA, Gaboriaud C et al. (2021a). Complement C1s and C4d as prognostic biomarkers in renal cancer: Emergence of noncanonical functions of C1s. Cancer Immunology Research 9: 891–908. DOI 10.1158/2326-6066.CIR-20-0532. [Google Scholar] [CrossRef]

Daugan MV, Revel M, Thouenon R, Dragon-Durey MA, Robe-Rybkine T et al. (2021b). Intracellular factor H drives tumor progression independently of the complement cascade. Cancer Immunology Research 9: 909–925. DOI 10.1158/2326-6066.CIR-20-0787. [Google Scholar] [CrossRef]

Diamandis EP, Goodglick L, Planque C, Thornquist MD (2011). Pentraxin-3 is a novel biomarker of lung carcinoma. Clinical Cancer Research 17: 2395–2399. DOI 10.1158/1078-0432.CCR-10-3024. [Google Scholar] [CrossRef]

Falagario UG, Busetto GM, Netti GS, Sanguedolce F, Selvaggio O et al. (2021). Prospective validation of pentraxin-3 as a novel serum biomarker to predict the risk of prostate cancer in patients scheduled for prostate biopsy. Cancers 13: 1611. DOI 10.3390/cancers13071611. [Google Scholar] [CrossRef]

Garlanda C, Bottazzi B, Bastone A, Mantovani A (2005). Pentraxins at the crossroads between innate immunity, inflammation, matrix deposition, and female fertility. Annual Review of Immunology 23: 337–366. DOI 10.1146/annurev.immunol.23.021704.115756. [Google Scholar] [CrossRef]

Geller A, Yan J (2019). The role of membrane bound complement regulatory proteins in tumor development and cancer immunotherapy. Frontiers in Immunology 10: 1074. DOI 10.3389/fimmu.2019.01074. [Google Scholar] [CrossRef]

Giacomini A, Ghedini GC, Presta M, Ronca R (2018). Long pentraxin 3: A novel multifaceted player in cancer. Biochimica et Biophysica Acta–Reviews on Cancer 1869: 53–63. DOI 10.1016/j.bbcan.2017.11.004. [Google Scholar] [CrossRef]

Gigante M, d’Altilia M, Montemurno E, Diella S, Bruno F et al. (2013). Branchio-Oto-Renal Syndrome (BOR) associated with focal glomerulosclerosis in a patient with a novel EYA1 splice site mutation. BMC Nephrology 14: 60. DOI 10.1186/1471-2369-14-60. [Google Scholar] [CrossRef]

Hezmee MNM, Kyaw-Tanner M, Lee JYP, Shiels IA, Rolfe B et al. (2011). Increased expression of C5a receptor (CD88) mRNA in canine mammary tumors. Veterinary Immunology and Immunopathology 139: 50–56. DOI 10.1016/j.vetimm.2010.08.005. [Google Scholar] [CrossRef]

Karczmarski J, Rubel T, Mikula M, Wolski J, Rutkowski A et al. (2013). Pre-analytical-related variability influencing serum peptide profiles demonstrated in a mass spectrometry-based search for colorectal and prostate cancer biomarkers. Acta Biochimica Polonica 60: 417–425. DOI 10.18388/abp.2013_2002. [Google Scholar] [CrossRef]

Korniluk A, Koper O 1, Kemona H, Dymicka-Piekarska V (2017). From inflammation to cancer. Irish Journal of Medical Science 186: 57–62. DOI 10.1007/s11845-016-1464-0. [Google Scholar] [CrossRef]

Loberg RD, Day LL, Dunn R, Kalikin LM, Pienta KJ (2006). Inhibition of decay-accelerating factor (CD55) attenuates prostate cancer growth and survival in vivo. Neoplasia 8: 69–78. DOI 10.1593/neo.05679. [Google Scholar] [CrossRef]

Ma YJ, Garred P (2018). Pentraxins in complement activation and regulation. Frontiers in Immunology 9: 3046. DOI 10.3389/fimmu.2018.03046. [Google Scholar] [CrossRef]

Manning ML, Williams SA, Jelinek CA, Kostova MB, Denmeade SR (2013). Proteolysis of complement factors iC3b and C5 by the serine protease prostate-specific antigen in prostatic fluid and seminal plasma. Journal of Immunology 190: 2567–2574. DOI 10.4049/jimmunol.1200856. [Google Scholar] [CrossRef]

Mantovani A, Garlanda C, Bottazzi B, Peri G, Doni A et al. (2006). The long pentraxin PTX3 in vascular pathology. Vascular Pharmacology 45: 326–330. DOI 10.1016/j.vph.2006.08.011. [Google Scholar] [CrossRef]

Netti GS, Lucarelli G, Spadaccino F, Castellano G, Gigante M et al. (2020). PTX3 modulates the immunoflogosis in tumor microenvironment and is a prognostic factor for patients with clear cell renal cell carcinoma. Aging 12: 7585–7602. DOI 10.18632/aging.103169. [Google Scholar] [CrossRef]

Peri G, Introna M, Corradi D, Iacuitti G, Signorini S et al. (2000). PTX3, a prototypical long pentraxin, is an early indicator of acute myocardial infarction in humans. Circulation 102: 636–641. DOI 10.1161/01.CIR.102.6.636. [Google Scholar] [CrossRef]

Pio R, Corrales L, Lambris JD (2014). The role of complement in tumor growth. Advances in Experimental Medicine and Biology 772: 229–262. DOI 10.1007/978-1-4614-5915-6. [Google Scholar] [CrossRef]

Reese B, Silwal A, Daugherity E, Daugherity M, Arabi M et al. (2020). Complement as prognostic biomarker and potential therapeutic target in renal cell carcinoma. Journal of Immunology 205: 3218–3229. DOI 10.4049/jimmunol.2000511. [Google Scholar] [CrossRef]

Riihilä P, Nissinen L, Knuutila J, Rahmati Nezhad P, Viiklepp K et al. (2019). Complement system in cutaneous squamous cell carcinoma. International Journal of Molecular Sciences 20: 3550. DOI 10.3390/ijms20143550. [Google Scholar] [CrossRef]

Ronca R, Di Salle E, Giacomini A, Leali D, Alessi P et al. (2013). Long pentraxin-3 inhibits epithelial-mesenchymal transition in melanoma cells. Molecular Cancer Therapeutics 12: 2760–2771. DOI 10.1158/1535-7163.MCT-13-0487. [Google Scholar] [CrossRef]

Roumenina LT, Daugan MV, Noé R, Petitprez F, Vano YA et al. (2019). Tumor cells hijack macrophage-produced complement C1q to promote tumor growth. Cancer Immunology 7: 1091–1105. DOI 10.1158/2326-6066.CIR-18-0891. [Google Scholar] [CrossRef]

Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT (2010). Cancer and the complement cascade. Molecular Cancer Research 8: 1453–1465. DOI 10.1158/1541-7786.MCR-10-0225. [Google Scholar] [CrossRef]

Santangelo L, Gigante M, Netti GS, Diella S, Puteo F et al. (2014). A novel SMARCAL1 mutation associated with a mild phenotype of Schimke immuno-osseous dysplasia (SIOD). BMC Nephrology 15: 41. DOI 10.1186/1471-2369-15-41. [Google Scholar] [CrossRef]

Schenk JM, Kristal AR, Neuhouser ML, Tangen CM, White E et al. (2010). Biomarkers of systemic inflammation and risk of incident, symptomatic benign prostatic hyperplasia: results from the prostate cancer prevention trial. American Journal of Epidemiology 171: 571–582. DOI 10.1093/aje/kwp406. [Google Scholar] [CrossRef]

Stallone G, Cormio L, Netti GS, Infante B, Selvaggio O et al. (2014). Pentraxin 3: A novel biomarker for predicting progression from prostatic inflammation to prostate cancer. Cancer Research 74: 4230–4238. DOI 10.1158/0008-5472.CAN-14-0369. [Google Scholar] [CrossRef]

Stallone G, Netti GS, Cormio L, Castellano G, Infante B et al. (2020). Modulation of complement activation by pentraxin-3 in prostate cancer. Scientific Reports 10: 18400. DOI 10.1038/s41598-020-75376-z. [Google Scholar] [CrossRef]

Tang S, Zhou W, Sheerin NS, Vaughan RW, Sacks SH (1999). Contribution of renal secreted complement C3 to the circulating pool in humans. Journal of Immunology 162: 4336–4341. [Google Scholar]

Xu C, Jung M, Burkhardt M, Stephan C, Schnorr D et al. (2005). Increased CD59 protein expression predicts a PSA relapse in patients after radical prostatectomy. Prostate 62: 224–232. DOI 10.1002/(ISSN)1097-0045. [Google Scholar] [CrossRef]

Zhou W, Marsh JE, Sacks SH (2001). Intrarenal synthesis of complement. Kidney International 59: 1227–1235. DOI 10.1046/j.1523-1755.2001.0590041227.x. [Google Scholar] [CrossRef]

images This work is licensed under a Creative Commons Attribution 4.0 International License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.