Skip to main content
Top
Published in: BMC Cancer 1/2019

Open Access 01-12-2019 | Metastasis | Research article

Blocking podoplanin suppresses growth and pulmonary metastasis of human malignant melanoma

Authors: Mengqiao Xu, Xia Wang, Yanfang Pan, Xingpeng Zhao, Bin Yan, Changgeng Ruan, Lijun Xia, Yiming Zhao

Published in: BMC Cancer | Issue 1/2019

Login to get access

Abstract

Background

Podoplanin (PDPN), a transmembrane O-glycoprotein, is up-regulated in many tumors and is involved in tumor metastasis and malignant progression. In previous studies, we generated a functional blocking monoclonal antibody (mAb, SZ168) against the extracellular domain of human PDPN. This study is aimed to investigate whether blocking PDPN by SZ168 inhibits tumor growth and metastasis.

Methods

Malignant melanoma xenograft model by inoculating subcutaneously human malignant melanoma cell line C8161 into the back of BALB/c nude mice was used. Endogenous PDPN expression in C8161 cells and nasopharyngeal cancer cell line CNE-2 was detected using western blot and flow cytometry.

Results

SZ168 significantly inhibited C8161 or CNE-2 cell-induced platelet aggregation in a dose-dependent manner with a maximal inhibition of 73.9 ± 3.0% in C8161 cells or 77.1 ± 2.7% in CNE-2 cells. Moreover, SZ168 inhibited the growth and pulmonary metastasis of C8161cells in vivo. The number of lung metastatic foci in the SZ168-treated group was significantly decreased compared with that in the control mouse IgG group (1.61 ± 0.44 vs.3.83 ± 0.60, P < 0.01). Subcutaneous tumor volume, weight, and incidence were also significantly reduced in the SZ168-treated group compared to the control group (P < 0.05). Additionally, SZ168 recognized PDPN in immunohistochemical analyses of tumor tissue sections.

Conclusions

SZ168 blocks growth and pulmonary metastasis of human malignant melanoma by inhibiting the interaction between tumor PDPN and platelet CLEC-2 and therefore is a promising antibody for therapeutic development against malignant melanoma.
Literature
1.
go back to reference Lowe KL, Navarro-Nunez L, Waston SP. Platelet CLEC-2 and podoplanin in cancer metastasis. Thromb Res. 2012;129(Suppl 1):S30–7.CrossRef Lowe KL, Navarro-Nunez L, Waston SP. Platelet CLEC-2 and podoplanin in cancer metastasis. Thromb Res. 2012;129(Suppl 1):S30–7.CrossRef
2.
go back to reference Tesfamariam B. Involvement of platelets in tumor cell metastasis. Pharmacol Ther. 2016;157:112–9.CrossRef Tesfamariam B. Involvement of platelets in tumor cell metastasis. Pharmacol Ther. 2016;157:112–9.CrossRef
3.
go back to reference Gupta GP, Massague J. Platelets and metastasis revisited: a novel fatty link. J Clin Invest. 2004;114:1691–3.CrossRef Gupta GP, Massague J. Platelets and metastasis revisited: a novel fatty link. J Clin Invest. 2004;114:1691–3.CrossRef
4.
go back to reference Stegner D, Dutting S, Nieswandt B. Mechanistic explanation for platelet contribution to cancer metastasis. Thromb Res. 2014;133(Suppl 2):S149–57.CrossRef Stegner D, Dutting S, Nieswandt B. Mechanistic explanation for platelet contribution to cancer metastasis. Thromb Res. 2014;133(Suppl 2):S149–57.CrossRef
5.
go back to reference Schacht V, Dadras SS, Johnson LA, et al. Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumors. Am J Pathol. 2005;166:913–21.CrossRef Schacht V, Dadras SS, Johnson LA, et al. Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumors. Am J Pathol. 2005;166:913–21.CrossRef
6.
go back to reference Wicki A, Lehembre F, Wick N, et al. Tumor invasion in the absence of epithelial-mesenchymal transition: podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell. 2006;9:261–72.CrossRef Wicki A, Lehembre F, Wick N, et al. Tumor invasion in the absence of epithelial-mesenchymal transition: podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell. 2006;9:261–72.CrossRef
7.
go back to reference Yuan P, Temam S, El-Naggar A, et al. Overexpression of podoplanin in oral cancer and its association with poor clinical outcome. Cancer. 2006;107:563–9.CrossRef Yuan P, Temam S, El-Naggar A, et al. Overexpression of podoplanin in oral cancer and its association with poor clinical outcome. Cancer. 2006;107:563–9.CrossRef
8.
go back to reference Kimura N, Kimura I. Podoplanin as a marker for mesothelioma. Pathol Int. 2005;55:83–6.CrossRef Kimura N, Kimura I. Podoplanin as a marker for mesothelioma. Pathol Int. 2005;55:83–6.CrossRef
9.
go back to reference Ordonez NG. D2-40 and podoplanin are highly specific and sensitive immunohistochemical markers of epithelioid malignant mesothelioma. Hum Pathol. 2005;36:372–80.CrossRef Ordonez NG. D2-40 and podoplanin are highly specific and sensitive immunohistochemical markers of epithelioid malignant mesothelioma. Hum Pathol. 2005;36:372–80.CrossRef
10.
go back to reference Weninger W, Partanen TA, Breiteneder-Geleff S, et al. Expression of vascular endothelial growth factor receptor-3 and podoplanin suggests a lymphatic endothelial cell origin of Kaposi's sarcoma tumor cells. Lab Invest. 1999;79:243–51.PubMed Weninger W, Partanen TA, Breiteneder-Geleff S, et al. Expression of vascular endothelial growth factor receptor-3 and podoplanin suggests a lymphatic endothelial cell origin of Kaposi's sarcoma tumor cells. Lab Invest. 1999;79:243–51.PubMed
11.
go back to reference Kato Y, Sasagawa I, Kaneko M, et al. Aggrus: a diagnostic marker that distinguishes seminoma from embryonal carcinoma in testicular germ cell tumors. Oncogene. 2004;23:8552–6.CrossRef Kato Y, Sasagawa I, Kaneko M, et al. Aggrus: a diagnostic marker that distinguishes seminoma from embryonal carcinoma in testicular germ cell tumors. Oncogene. 2004;23:8552–6.CrossRef
12.
go back to reference Shibahara J, Kashima T, Kikuchi Y, Kunita A, Fukayama M. Podoplanin is expressed in subsets of tumors of the central nervous system. Virchows Arch. 2006;448(4):493–9.CrossRef Shibahara J, Kashima T, Kikuchi Y, Kunita A, Fukayama M. Podoplanin is expressed in subsets of tumors of the central nervous system. Virchows Arch. 2006;448(4):493–9.CrossRef
13.
go back to reference Hoshino A, Ishii G, Ito T, et al. Podoplanin-positive fibroblasts enhance lung adenocarcinoma tumor formation: podoplanin in fibroblast functions for tumor progression. Cancer Res. 2011;71:4769–79.CrossRef Hoshino A, Ishii G, Ito T, et al. Podoplanin-positive fibroblasts enhance lung adenocarcinoma tumor formation: podoplanin in fibroblast functions for tumor progression. Cancer Res. 2011;71:4769–79.CrossRef
14.
go back to reference Kawaguchi H, El-Naggar AK, Papadimitrakopoulou V, et al. Podoplanin: a novel marker for oral cancer risk in patients with oral premalignancy. J Clin Oncol. 2008;26:354–60.CrossRef Kawaguchi H, El-Naggar AK, Papadimitrakopoulou V, et al. Podoplanin: a novel marker for oral cancer risk in patients with oral premalignancy. J Clin Oncol. 2008;26:354–60.CrossRef
15.
go back to reference Kawase A, Ishii G, Nagai K, et al. Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int J Cancer. 2008;123:1053–9.CrossRef Kawase A, Ishii G, Nagai K, et al. Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int J Cancer. 2008;123:1053–9.CrossRef
16.
go back to reference Kunita A, Kashima TG, Morishita Y, et al. The platelet aggregation-inducing factor aggrus/podoplanin promotes pulmonary metastasis. Am J Pathol. 2007;170:1337–47.CrossRef Kunita A, Kashima TG, Morishita Y, et al. The platelet aggregation-inducing factor aggrus/podoplanin promotes pulmonary metastasis. Am J Pathol. 2007;170:1337–47.CrossRef
17.
go back to reference Martin-Villar E, Megias D, Castel S, et al. Podoplanin binds ERM proteins to activate RhoA and promote epithelial-mesenchymal transition. J Cell Sci. 2006;119:4541–53.CrossRef Martin-Villar E, Megias D, Castel S, et al. Podoplanin binds ERM proteins to activate RhoA and promote epithelial-mesenchymal transition. J Cell Sci. 2006;119:4541–53.CrossRef
18.
go back to reference Scholl FG, Gamallo C, Quintanilla M. Ectopic expression of PA2.26 antigen in epidermal keratinocytes leads to destabilization of adherens junctions and malignant progression. Lab Invest. 2000;80:1749–59.CrossRef Scholl FG, Gamallo C, Quintanilla M. Ectopic expression of PA2.26 antigen in epidermal keratinocytes leads to destabilization of adherens junctions and malignant progression. Lab Invest. 2000;80:1749–59.CrossRef
19.
go back to reference Kato Y, Fujita N, Kunita A, et al. Molecular identification of Aggrus/T1alpha as a platelet aggregation-inducing factor expressed in colorectal tumors. J Biol Chem. 2003;278:51599–605.CrossRef Kato Y, Fujita N, Kunita A, et al. Molecular identification of Aggrus/T1alpha as a platelet aggregation-inducing factor expressed in colorectal tumors. J Biol Chem. 2003;278:51599–605.CrossRef
20.
go back to reference Suzuki-Inoue K, Kato Y, Inoue O, et al. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J Biol Chem. 2007;282:25993–6001.CrossRef Suzuki-Inoue K, Kato Y, Inoue O, et al. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J Biol Chem. 2007;282:25993–6001.CrossRef
21.
go back to reference Suzuki-Inoue K. Essential in vivo roles of the platelet activation receptor CLEC-2 in tumour metastasis, lymphangiogenesis and thrombus formation. J Biochem. 2011;150:127–32.CrossRef Suzuki-Inoue K. Essential in vivo roles of the platelet activation receptor CLEC-2 in tumour metastasis, lymphangiogenesis and thrombus formation. J Biochem. 2011;150:127–32.CrossRef
22.
go back to reference Suzuki-Inoue K, Fuller GL, Garcia A, et al. A novel Syk-dependent mechanism of platelet activation by the C-type lectin receptor CLEC-2. Blood. 2006;107:542–9.CrossRef Suzuki-Inoue K, Fuller GL, Garcia A, et al. A novel Syk-dependent mechanism of platelet activation by the C-type lectin receptor CLEC-2. Blood. 2006;107:542–9.CrossRef
23.
go back to reference Kato Y, Kaneko MK, Kuno A, et al. Inhibition of tumor cell-induced platelet aggregation using a novel anti-podoplanin antibody reacting with its platelet-aggregation-stimulating domain. Biochem Biophys Res Commun. 2006;349:1301–7.CrossRef Kato Y, Kaneko MK, Kuno A, et al. Inhibition of tumor cell-induced platelet aggregation using a novel anti-podoplanin antibody reacting with its platelet-aggregation-stimulating domain. Biochem Biophys Res Commun. 2006;349:1301–7.CrossRef
24.
go back to reference Zhao X, Pan Y, Ren W, et al. Plasma soluble podoplanin is a novel marker for the diagnosis of tumor occurrence and metastasis. Cancer Sci. 2018;109(2):403–11.CrossRef Zhao X, Pan Y, Ren W, et al. Plasma soluble podoplanin is a novel marker for the diagnosis of tumor occurrence and metastasis. Cancer Sci. 2018;109(2):403–11.CrossRef
25.
go back to reference Qu L, Zhao X, Fu J, et al. Stable expression of recombinant human podoplanin in Chinese hamster ovary (CHO) cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2016;32:25–8.PubMed Qu L, Zhao X, Fu J, et al. Stable expression of recombinant human podoplanin in Chinese hamster ovary (CHO) cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2016;32:25–8.PubMed
26.
go back to reference Camerer E, Qazi AA, Duong DN, et al. Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood. 2004;104:397–401.CrossRef Camerer E, Qazi AA, Duong DN, et al. Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood. 2004;104:397–401.CrossRef
27.
28.
go back to reference Philippe C, Philippe B, Fouqueray B, Perez J, Lebret M, Baud L. Protection from tumor necrosis factor-mediated cytolysis by platelets. Am J Pathol. 1993;143(6):1713–23.PubMedPubMedCentral Philippe C, Philippe B, Fouqueray B, Perez J, Lebret M, Baud L. Protection from tumor necrosis factor-mediated cytolysis by platelets. Am J Pathol. 1993;143(6):1713–23.PubMedPubMedCentral
29.
go back to reference Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer. 2011;11:123–34.CrossRef Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer. 2011;11:123–34.CrossRef
30.
go back to reference Klerk CP, Smorenburg SM, Otten HM, et al. The effect of low molecular weight heparin on survival in patients with advanced malignancy. J Clin Oncol. 2005;23:2130–5.CrossRef Klerk CP, Smorenburg SM, Otten HM, et al. The effect of low molecular weight heparin on survival in patients with advanced malignancy. J Clin Oncol. 2005;23:2130–5.CrossRef
31.
go back to reference Suzuki-Inoue K, Inoue O, Ding G, et al. Essential in vivo roles of the C-type lectin receptor CLEC-2: embryonic/neonatal lethality of CLEC-2-deficient mice by blood/lymphatic misconnections and impaired thrombus formation of CLEC-2-deficient platelets. J Biol Chem. 2010;285:24494–507.CrossRef Suzuki-Inoue K, Inoue O, Ding G, et al. Essential in vivo roles of the C-type lectin receptor CLEC-2: embryonic/neonatal lethality of CLEC-2-deficient mice by blood/lymphatic misconnections and impaired thrombus formation of CLEC-2-deficient platelets. J Biol Chem. 2010;285:24494–507.CrossRef
32.
go back to reference Eichholtz T, Jalink K, Fahrenfort I, et al. The bioactive phospholipid lysophosphatidic acid is released from activated platelets. Biochem J. 1993;291 ( Pt 3:677–80.CrossRef Eichholtz T, Jalink K, Fahrenfort I, et al. The bioactive phospholipid lysophosphatidic acid is released from activated platelets. Biochem J. 1993;291 ( Pt 3:677–80.CrossRef
33.
go back to reference Smyth SS, McEver RP, Weyrich AS, Morrell CN, Hoffman MR, Arepally GM, et al. Platelet functions beyond hemostasis. J Thromb Haemost. 2009;7(11):1759–66.CrossRef Smyth SS, McEver RP, Weyrich AS, Morrell CN, Hoffman MR, Arepally GM, et al. Platelet functions beyond hemostasis. J Thromb Haemost. 2009;7(11):1759–66.CrossRef
34.
go back to reference Takagi S, Sato S, Oh-hara T, et al. Platelets promote tumor growth and metastasis via direct interaction between Aggrus/podoplanin and CLEC-2. PLoS One. 2013;8:e73609.CrossRef Takagi S, Sato S, Oh-hara T, et al. Platelets promote tumor growth and metastasis via direct interaction between Aggrus/podoplanin and CLEC-2. PLoS One. 2013;8:e73609.CrossRef
35.
go back to reference Tajima S, Fukayama M. Possibility of D2-40 as a diagnostic and tumor differentiation-suggestive marker for some of phosphaturic mesenchymal tumors. Int J Clin Exp Pathol. 2015;8(8):9390–6.PubMedPubMedCentral Tajima S, Fukayama M. Possibility of D2-40 as a diagnostic and tumor differentiation-suggestive marker for some of phosphaturic mesenchymal tumors. Int J Clin Exp Pathol. 2015;8(8):9390–6.PubMedPubMedCentral
36.
go back to reference Afzal MZ, Mabaera R, Shirai K. Metastatic uveal melanoma showing durable response to anti-CTLA-4 and anti-PD-1 combination therapy after experiencing progression on anti-PD-1 therapy alone. J Immunother Cancer. 2018;6:13.CrossRef Afzal MZ, Mabaera R, Shirai K. Metastatic uveal melanoma showing durable response to anti-CTLA-4 and anti-PD-1 combination therapy after experiencing progression on anti-PD-1 therapy alone. J Immunother Cancer. 2018;6:13.CrossRef
37.
go back to reference Huang R, Andersen LMK, Rofstad EK. Metastatic pathway and the microvascular and physicochemical microenvironments of human melanoma xenografts. J Transl Med. 2017;15:203.CrossRef Huang R, Andersen LMK, Rofstad EK. Metastatic pathway and the microvascular and physicochemical microenvironments of human melanoma xenografts. J Transl Med. 2017;15:203.CrossRef
38.
go back to reference Lee RJ, Gremel G, Marshall A, Myers KA, Fisher N, Dunn JA, et al. Circulating tumor DNA predicts survival in patients with resected high risk stage II/III melanoma. Ann Oncol. 2018;29(2):490–6.CrossRef Lee RJ, Gremel G, Marshall A, Myers KA, Fisher N, Dunn JA, et al. Circulating tumor DNA predicts survival in patients with resected high risk stage II/III melanoma. Ann Oncol. 2018;29(2):490–6.CrossRef
Metadata
Title
Blocking podoplanin suppresses growth and pulmonary metastasis of human malignant melanoma
Authors
Mengqiao Xu
Xia Wang
Yanfang Pan
Xingpeng Zhao
Bin Yan
Changgeng Ruan
Lijun Xia
Yiming Zhao
Publication date
01-12-2019
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2019
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5808-9

Other articles of this Issue 1/2019

BMC Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine