Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Thyroid Cancer | Research

PD-1 blockade delays tumor growth by inhibiting an intrinsic SHP2/Ras/MAPK signalling in thyroid cancer cells

Authors: Federica Liotti, Narender Kumar, Nella Prevete, Maria Marotta, Daniela Sorriento, Caterina Ieranò, Andrea Ronchi, Federica Zito Marino, Sonia Moretti, Renato Colella, Efiso Puxeddu, Simona Paladino, Yoshihito Kano, Michael Ohh, Stefania Scala, Rosa Marina Melillo

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

The programmed cell death-1 (PD-1) receptor and its ligands PD-L1 and PD-L2 are immune checkpoints that suppress anti-cancer immunity. Typically, cancer cells express the PD-Ls that bind PD-1 on immune cells, inhibiting their activity. Recently, PD-1 expression has also been found in cancer cells. Here, we analysed expression and functions of PD-1 in thyroid cancer (TC).

Methods

PD-1 expression was evaluated by immunohistochemistry on human TC samples and by RT-PCR, western blot and FACS on TC cell lines. Proliferation and migration of TC cells in culture were assessed by BrdU incorporation and Boyden chamber assays. Biochemical studies were performed by western blot, immunoprecipitation, pull-down and phosphatase assays. TC cell tumorigenicity was assessed by xenotransplants in nude mice.

Results

Human TC specimens (47%), but not normal thyroids, displayed PD-1 expression in epithelial cells, which significantly correlated with tumour stage and lymph-node metastasis. PD-1 was also constitutively expressed on TC cell lines. PD-1 overexpression/stimulation promoted TC cell proliferation and migration. Accordingly, PD-1 genetic/pharmacologic inhibition caused the opposite effects. Mechanistically, PD-1 recruited the SHP2 phosphatase to the plasma membrane and potentiated its phosphatase activity. SHP2 enhanced Ras activation by dephosphorylating its inhibitory tyrosine 32, thus triggering the MAPK cascade. SHP2, BRAF and MEK were necessary for PD-1-mediated biologic functions. PD-1 inhibition decreased, while PD-1 enforced expression facilitated, TC cell xenograft growth in mice by affecting tumour cell proliferation.

Conclusions

PD-1 circuit blockade in TC, besides restoring anti-cancer immunity, could also directly impair TC cell growth by inhibiting the SHP2/Ras/MAPK signalling pathway.
Appendix
Available only for authorised users
Literature
2.
go back to reference Kruger S, Ilmer M, Kobold S, Cadilha BL, Endres S, Ormanns S, et al. Advances in cancer immunotherapy 2019 - latest trends. J Exp Clin Cancer Res. 2019;38:268.PubMedPubMedCentralCrossRef Kruger S, Ilmer M, Kobold S, Cadilha BL, Endres S, Ormanns S, et al. Advances in cancer immunotherapy 2019 - latest trends. J Exp Clin Cancer Res. 2019;38:268.PubMedPubMedCentralCrossRef
3.
5.
go back to reference Kleffel S, Posch C, Barthel SR, Mueller H, Schlapbach C, Guenova E, et al. Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth. Cell. 2015;162:1242–56.PubMedPubMedCentralCrossRef Kleffel S, Posch C, Barthel SR, Mueller H, Schlapbach C, Guenova E, et al. Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth. Cell. 2015;162:1242–56.PubMedPubMedCentralCrossRef
6.
go back to reference Li H, Li X, Liu S, Guo L, Zhang B, Zhang J, et al. Programmed cell death-1 (PD-1) checkpoint blockade in combination with a mammalian target of rapamycin inhibitor restrains hepatocellular carcinoma growth induced by hepatoma cell-intrinsic PD-1. Hepatology. 2017;66:1920–33.PubMedCrossRef Li H, Li X, Liu S, Guo L, Zhang B, Zhang J, et al. Programmed cell death-1 (PD-1) checkpoint blockade in combination with a mammalian target of rapamycin inhibitor restrains hepatocellular carcinoma growth induced by hepatoma cell-intrinsic PD-1. Hepatology. 2017;66:1920–33.PubMedCrossRef
7.
go back to reference Du S, McCall N, Park K, Guan Q, Fontina P, Ertel A, et al. Blockade of tumor-expressed PD-1 promotes lung cancer growth. Oncoimmunology. 2018;7:e1408747.PubMedPubMedCentralCrossRef Du S, McCall N, Park K, Guan Q, Fontina P, Ertel A, et al. Blockade of tumor-expressed PD-1 promotes lung cancer growth. Oncoimmunology. 2018;7:e1408747.PubMedPubMedCentralCrossRef
8.
go back to reference Zhao Y, Harrison DL, Song Y, Ji J, Huang J, Hui E. Antigen-presenting cell-intrinsic PD-1 neutralizes PD-L1 in cis to attenuate PD-1 signaling in T cells. Cell Rep. 2018;24:379–90 e6.PubMedPubMedCentralCrossRef Zhao Y, Harrison DL, Song Y, Ji J, Huang J, Hui E. Antigen-presenting cell-intrinsic PD-1 neutralizes PD-L1 in cis to attenuate PD-1 signaling in T cells. Cell Rep. 2018;24:379–90 e6.PubMedPubMedCentralCrossRef
9.
go back to reference Liotti F, Prevete N, Vecchio G, Melillo RM. Recent advances in understanding immune phenotypes of thyroid carcinomas: prognostication and emerging therapies. F1000 Faculty Rev. 2019;8:227. Liotti F, Prevete N, Vecchio G, Melillo RM. Recent advances in understanding immune phenotypes of thyroid carcinomas: prognostication and emerging therapies. F1000 Faculty Rev. 2019;8:227.
11.
go back to reference Giannini R, Moretti S, Ugolini C, Macerola E, Menicali E, Nucci N, et al. Immune profiling of thyroid carcinomas suggests the existence of two major phenotypes: an ATC-like and a PDTC-like. J Clin Endocrinol Metab. 2019;104:3557–75.PubMed Giannini R, Moretti S, Ugolini C, Macerola E, Menicali E, Nucci N, et al. Immune profiling of thyroid carcinomas suggests the existence of two major phenotypes: an ATC-like and a PDTC-like. J Clin Endocrinol Metab. 2019;104:3557–75.PubMed
12.
go back to reference Bastman JJ, Serracino HS, Zhu Y, Koenig MR, Mateescu V, Sams SB, et al. Tumor-infiltrating T cells and the PD-1 checkpoint pathway in advanced differentiated and anaplastic thyroid Cancer. J Clin Endocrinol Metab. 2016;101:2863–73.PubMedPubMedCentralCrossRef Bastman JJ, Serracino HS, Zhu Y, Koenig MR, Mateescu V, Sams SB, et al. Tumor-infiltrating T cells and the PD-1 checkpoint pathway in advanced differentiated and anaplastic thyroid Cancer. J Clin Endocrinol Metab. 2016;101:2863–73.PubMedPubMedCentralCrossRef
13.
go back to reference French JD, Kotnis GR, Said S, Raeburn CD, McIntyre RC Jr, Klopper JP, et al. Programmed death-1+ T cells and regulatory T cells are enriched in tumor-involved lymph nodes and associated with aggressive features in papillary thyroid cancer. J Clin Endocrinol Metab. 2012;97:E934–43.PubMedPubMedCentralCrossRef French JD, Kotnis GR, Said S, Raeburn CD, McIntyre RC Jr, Klopper JP, et al. Programmed death-1+ T cells and regulatory T cells are enriched in tumor-involved lymph nodes and associated with aggressive features in papillary thyroid cancer. J Clin Endocrinol Metab. 2012;97:E934–43.PubMedPubMedCentralCrossRef
14.
go back to reference Malfitano AM, Somma SD, Prevete N, Portella G. Virotherapy as a potential therapeutic approach for the treatment of aggressive thyroid Cancer. Cancers. 2019;11:1532-53. Malfitano AM, Somma SD, Prevete N, Portella G. Virotherapy as a potential therapeutic approach for the treatment of aggressive thyroid Cancer. Cancers. 2019;11:1532-53.
15.
go back to reference Saini S, Tulla K, Maker AV, Burman KD, Prabhakar BS. Therapeutic advances in anaplastic thyroid cancer: a current perspective. Mol Cancer. 2018;17:154.PubMedPubMedCentralCrossRef Saini S, Tulla K, Maker AV, Burman KD, Prabhakar BS. Therapeutic advances in anaplastic thyroid cancer: a current perspective. Mol Cancer. 2018;17:154.PubMedPubMedCentralCrossRef
16.
go back to reference Ulisse S, Tuccilli C, Sorrenti S, Antonelli A, Fallahi P, D'Armiento E, et al. PD-1 ligand expression in epithelial thyroid cancers: potential clinical implications. Int J Mol Sci. 2019;20:1405-20. Ulisse S, Tuccilli C, Sorrenti S, Antonelli A, Fallahi P, D'Armiento E, et al. PD-1 ligand expression in epithelial thyroid cancers: potential clinical implications. Int J Mol Sci. 2019;20:1405-20.
17.
go back to reference Cunha LL, Morari EC, Guihen AC, Razolli D, Gerhard R, Nonogaki S, et al. Infiltration of a mixture of immune cells may be related to good prognosis in patients with differentiated thyroid carcinoma. Clin Endocrinol. 2012;77:918–25.CrossRef Cunha LL, Morari EC, Guihen AC, Razolli D, Gerhard R, Nonogaki S, et al. Infiltration of a mixture of immune cells may be related to good prognosis in patients with differentiated thyroid carcinoma. Clin Endocrinol. 2012;77:918–25.CrossRef
18.
go back to reference Cunha LL, Marcello MA, Morari EC, Nonogaki S, Conte FF, Gerhard R, et al. Differentiated thyroid carcinomas may elude the immune system by B7H1 upregulation. Endocr Relat Cancer. 2013;20:103–10.PubMedCrossRef Cunha LL, Marcello MA, Morari EC, Nonogaki S, Conte FF, Gerhard R, et al. Differentiated thyroid carcinomas may elude the immune system by B7H1 upregulation. Endocr Relat Cancer. 2013;20:103–10.PubMedCrossRef
19.
go back to reference De Falco V, Castellone MD, De Vita G, Cirafici AM, Hershman JM, Guerrero C, et al. RET/papillary thyroid carcinoma oncogenic signaling through the Rap1 small GTPase. Cancer Res. 2007;67:381–90.PubMedCrossRef De Falco V, Castellone MD, De Vita G, Cirafici AM, Hershman JM, Guerrero C, et al. RET/papillary thyroid carcinoma oncogenic signaling through the Rap1 small GTPase. Cancer Res. 2007;67:381–90.PubMedCrossRef
20.
go back to reference Liotti F, Collina F, Pone E, La Sala L, Franco R, Prevete N, et al. Interleukin-8, but not the related chemokine CXCL1, sustains an Autocrine circuit necessary for the properties and functions of thyroid Cancer stem cells. Stem Cells. 2017;35:135–46.PubMedCrossRef Liotti F, Collina F, Pone E, La Sala L, Franco R, Prevete N, et al. Interleukin-8, but not the related chemokine CXCL1, sustains an Autocrine circuit necessary for the properties and functions of thyroid Cancer stem cells. Stem Cells. 2017;35:135–46.PubMedCrossRef
21.
go back to reference Prevete N, Liotti F, Illiano A, Amoresano A, Pucci P, de Paulis A, et al. Formyl peptide receptor 1 suppresses gastric cancer angiogenesis and growth by exploiting inflammation resolution pathways. Oncoimmunology. 2017;6:e1293213.PubMedPubMedCentralCrossRef Prevete N, Liotti F, Illiano A, Amoresano A, Pucci P, de Paulis A, et al. Formyl peptide receptor 1 suppresses gastric cancer angiogenesis and growth by exploiting inflammation resolution pathways. Oncoimmunology. 2017;6:e1293213.PubMedPubMedCentralCrossRef
22.
go back to reference Prevete N, Liotti F, Visciano C, Marone G, Melillo RM, de Paulis A. The formyl peptide receptor 1 exerts a tumor suppressor function in human gastric cancer by inhibiting angiogenesis. Oncogene. 2015;34:3826–38.PubMedCrossRef Prevete N, Liotti F, Visciano C, Marone G, Melillo RM, de Paulis A. The formyl peptide receptor 1 exerts a tumor suppressor function in human gastric cancer by inhibiting angiogenesis. Oncogene. 2015;34:3826–38.PubMedCrossRef
23.
go back to reference Paulsen EE, Kilvaer TK, Khanehkenari MR, Al-Saad S, Hald SM, Andersen S, et al. Assessing PDL-1 and PD-1 in non-small cell lung Cancer: a novel Immunoscore approach. Clin Lung Cancer. 2017;18:220–33 e8.PubMedCrossRef Paulsen EE, Kilvaer TK, Khanehkenari MR, Al-Saad S, Hald SM, Andersen S, et al. Assessing PDL-1 and PD-1 in non-small cell lung Cancer: a novel Immunoscore approach. Clin Lung Cancer. 2017;18:220–33 e8.PubMedCrossRef
24.
go back to reference Kouketsu A, Sato I, Oikawa M, Shimizu Y, Saito H, Takahashi T, et al. Expression of immunoregulatory molecules PD-L1 and PD-1 in oral cancer and precancerous lesions: a cohort study of Japanese patients. J Cranio Maxillo Facial Surg. 2019;47:33–40.CrossRef Kouketsu A, Sato I, Oikawa M, Shimizu Y, Saito H, Takahashi T, et al. Expression of immunoregulatory molecules PD-L1 and PD-1 in oral cancer and precancerous lesions: a cohort study of Japanese patients. J Cranio Maxillo Facial Surg. 2019;47:33–40.CrossRef
25.
26.
go back to reference Collina F, La Sala L, Liotti F, Prevete N, La Mantia E, Chiofalo MG, et al. AXL is a novel predictive factor and therapeutic target for radioactive iodine refractory thyroid Cancer. Cancers. 2019;11:785-805. Collina F, La Sala L, Liotti F, Prevete N, La Mantia E, Chiofalo MG, et al. AXL is a novel predictive factor and therapeutic target for radioactive iodine refractory thyroid Cancer. Cancers. 2019;11:785-805.
27.
go back to reference Avilla E, Guarino V, Visciano C, Liotti F, Svelto M, Krishnamoorthy G, et al. Activation of TYRO3/AXL tyrosine kinase receptors in thyroid cancer. Cancer Res. 2011;71:1792–804.PubMedCrossRef Avilla E, Guarino V, Visciano C, Liotti F, Svelto M, Krishnamoorthy G, et al. Activation of TYRO3/AXL tyrosine kinase receptors in thyroid cancer. Cancer Res. 2011;71:1792–804.PubMedCrossRef
28.
go back to reference Paladino S, Lebreton S, Tivodar S, Campana V, Tempre R, Zurzolo C. Different GPI-attachment signals affect the oligomerisation of GPI-anchored proteins and their apical sorting. J Cell Sci. 2008;121:4001–7.PubMedCrossRef Paladino S, Lebreton S, Tivodar S, Campana V, Tempre R, Zurzolo C. Different GPI-attachment signals affect the oligomerisation of GPI-anchored proteins and their apical sorting. J Cell Sci. 2008;121:4001–7.PubMedCrossRef
29.
go back to reference Bunda S, Burrell K, Heir P, Zeng L, Alamsahebpour A, Kano Y, et al. Inhibition of SHP2-mediated dephosphorylation of Ras suppresses oncogenesis. Nat Commun. 2015;6:8859.PubMedCrossRef Bunda S, Burrell K, Heir P, Zeng L, Alamsahebpour A, Kano Y, et al. Inhibition of SHP2-mediated dephosphorylation of Ras suppresses oncogenesis. Nat Commun. 2015;6:8859.PubMedCrossRef
30.
go back to reference Knauf JA, Fagin JA. Role of MAPK pathway oncoproteins in thyroid cancer pathogenesis and as drug targets. Curr Opin Cell Biol. 2009;21:296–303.PubMedCrossRef Knauf JA, Fagin JA. Role of MAPK pathway oncoproteins in thyroid cancer pathogenesis and as drug targets. Curr Opin Cell Biol. 2009;21:296–303.PubMedCrossRef
31.
go back to reference Rota G, Niogret C, Dang AT, Barros CR, Fonta NP, Alfei F, et al. Shp-2 is dispensable for establishing T cell exhaustion and for PD-1 signaling in vivo. Cell Rep. 2018;23:39–49.PubMedCrossRef Rota G, Niogret C, Dang AT, Barros CR, Fonta NP, Alfei F, et al. Shp-2 is dispensable for establishing T cell exhaustion and for PD-1 signaling in vivo. Cell Rep. 2018;23:39–49.PubMedCrossRef
34.
go back to reference Matozaki T, Murata Y, Saito Y, Okazawa H, Ohnishi H. Protein tyrosine phosphatase SHP-2: a proto-oncogene product that promotes Ras activation. Cancer Sci. 2009;100:1786–93.PubMedCrossRef Matozaki T, Murata Y, Saito Y, Okazawa H, Ohnishi H. Protein tyrosine phosphatase SHP-2: a proto-oncogene product that promotes Ras activation. Cancer Sci. 2009;100:1786–93.PubMedCrossRef
35.
go back to reference Kano Y, Cook JD, Lee JE, Ohh M. New structural and functional insight into the regulation of Ras. Semin Cell Dev Biol. 2016;58:70–8.PubMedCrossRef Kano Y, Cook JD, Lee JE, Ohh M. New structural and functional insight into the regulation of Ras. Semin Cell Dev Biol. 2016;58:70–8.PubMedCrossRef
36.
go back to reference Chen YN, LaMarche MJ, Chan HM, Fekkes P, Garcia-Fortanet J, Acker MG, et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535:148–52.PubMedCrossRef Chen YN, LaMarche MJ, Chan HM, Fekkes P, Garcia-Fortanet J, Acker MG, et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535:148–52.PubMedCrossRef
37.
go back to reference Xing J, Liu R, Xing M, Trink B. The BRAFT1799A mutation confers sensitivity of thyroid cancer cells to the BRAFV600E inhibitor PLX4032 (RG7204). Biochem Biophys Res Commun. 2011;404:958–62.PubMedCrossRef Xing J, Liu R, Xing M, Trink B. The BRAFT1799A mutation confers sensitivity of thyroid cancer cells to the BRAFV600E inhibitor PLX4032 (RG7204). Biochem Biophys Res Commun. 2011;404:958–62.PubMedCrossRef
38.
go back to reference Ball DW, Jin N, Rosen DM, Dackiw A, Sidransky D, Xing M, et al. Selective growth inhibition in BRAF mutant thyroid cancer by the mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244. J Clin Endocrinol Metab. 2007;92:4712–8.PubMedCrossRef Ball DW, Jin N, Rosen DM, Dackiw A, Sidransky D, Xing M, et al. Selective growth inhibition in BRAF mutant thyroid cancer by the mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244. J Clin Endocrinol Metab. 2007;92:4712–8.PubMedCrossRef
39.
go back to reference Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F, et al. Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol. 2016;13:473–86.PubMedCrossRef Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F, et al. Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol. 2016;13:473–86.PubMedCrossRef
41.
go back to reference Cunha LL, Marcello MA, Vassallo J, Ward LS. Differentiated thyroid carcinomas and their B7H1 shield. Future Oncol. 2013;9:1417–9.PubMedCrossRef Cunha LL, Marcello MA, Vassallo J, Ward LS. Differentiated thyroid carcinomas and their B7H1 shield. Future Oncol. 2013;9:1417–9.PubMedCrossRef
42.
44.
go back to reference Passariello M, Camorani S, Vetrei C, Ricci S, Cerchia L, De Lorenzo C. Ipilimumab and its derived EGFR Aptamer-based conjugate induce efficient NK cell activation against Cancer cells. Cancers. 2020;12:331-49. Passariello M, Camorani S, Vetrei C, Ricci S, Cerchia L, De Lorenzo C. Ipilimumab and its derived EGFR Aptamer-based conjugate induce efficient NK cell activation against Cancer cells. Cancers. 2020;12:331-49.
45.
go back to reference Contardi E, Palmisano GL, Tazzari PL, Martelli AM, Fala F, Fabbi M, et al. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. Int J Cancer. 2005;117:538–50.PubMedCrossRef Contardi E, Palmisano GL, Tazzari PL, Martelli AM, Fala F, Fabbi M, et al. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. Int J Cancer. 2005;117:538–50.PubMedCrossRef
47.
go back to reference Scott LM, Chen L, Daniel KG, Brooks WH, Guida WC, Lawrence HR, et al. Shp2 protein tyrosine phosphatase inhibitor activity of estramustine phosphate and its triterpenoid analogs. Bioorg Med Chem Lett. 2011;21:730–3.PubMedCrossRef Scott LM, Chen L, Daniel KG, Brooks WH, Guida WC, Lawrence HR, et al. Shp2 protein tyrosine phosphatase inhibitor activity of estramustine phosphate and its triterpenoid analogs. Bioorg Med Chem Lett. 2011;21:730–3.PubMedCrossRef
48.
go back to reference Seebacher NA, Stacy AE, Porter GM, Merlot AM. Clinical development of targeted and immune based anti-cancer therapies. J Exp Clin Cancer Res. 2019;38:156.PubMedPubMedCentralCrossRef Seebacher NA, Stacy AE, Porter GM, Merlot AM. Clinical development of targeted and immune based anti-cancer therapies. J Exp Clin Cancer Res. 2019;38:156.PubMedPubMedCentralCrossRef
49.
go back to reference Ahmed TA, Adamopoulos C, Karoulia Z, Wu X, Sachidanandam R, Aaronson SA, et al. SHP2 drives adaptive resistance to ERK signaling inhibition in molecularly defined subsets of ERK-dependent tumors. Cell Rep. 2019;26:65–78 e5.PubMedPubMedCentralCrossRef Ahmed TA, Adamopoulos C, Karoulia Z, Wu X, Sachidanandam R, Aaronson SA, et al. SHP2 drives adaptive resistance to ERK signaling inhibition in molecularly defined subsets of ERK-dependent tumors. Cell Rep. 2019;26:65–78 e5.PubMedPubMedCentralCrossRef
51.
go back to reference Hu ZQ, Ma R, Zhang CM, Li J, Li L, Hu ZT, et al. Expression and clinical significance of tyrosine phosphatase SHP2 in thyroid carcinoma. Oncol Lett. 2015;10:1507–12.PubMedPubMedCentralCrossRef Hu ZQ, Ma R, Zhang CM, Li J, Li L, Hu ZT, et al. Expression and clinical significance of tyrosine phosphatase SHP2 in thyroid carcinoma. Oncol Lett. 2015;10:1507–12.PubMedPubMedCentralCrossRef
52.
go back to reference Cabanillas ME, Ferrarotto R, Garden AS, Ahmed S, Busaidy NL, Dadu R, et al. Neoadjuvant BRAF- and immune-directed therapy for anaplastic thyroid carcinoma. Thyroid. 2018;28:945–51.PubMedPubMedCentralCrossRef Cabanillas ME, Ferrarotto R, Garden AS, Ahmed S, Busaidy NL, Dadu R, et al. Neoadjuvant BRAF- and immune-directed therapy for anaplastic thyroid carcinoma. Thyroid. 2018;28:945–51.PubMedPubMedCentralCrossRef
53.
go back to reference Iyer PC, Dadu R, Gule-Monroe M, Busaidy NL, Ferrarotto R, Habra MA, et al. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J Immunother Cancer. 2018;6:68.PubMedPubMedCentralCrossRef Iyer PC, Dadu R, Gule-Monroe M, Busaidy NL, Ferrarotto R, Habra MA, et al. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J Immunother Cancer. 2018;6:68.PubMedPubMedCentralCrossRef
Metadata
Title
PD-1 blockade delays tumor growth by inhibiting an intrinsic SHP2/Ras/MAPK signalling in thyroid cancer cells
Authors
Federica Liotti
Narender Kumar
Nella Prevete
Maria Marotta
Daniela Sorriento
Caterina Ieranò
Andrea Ronchi
Federica Zito Marino
Sonia Moretti
Renato Colella
Efiso Puxeddu
Simona Paladino
Yoshihito Kano
Michael Ohh
Stefania Scala
Rosa Marina Melillo
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01818-1

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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