Skip to main content
Top
Published in: BMC Cancer 1/2023

Open Access 01-12-2023 | Melanoma | Research

Aspartate beta-hydroxylase domain containing 1 as a prognostic marker associated with immune infiltration in skin cutaneous melanoma

Authors: Shiquan Sun, Min Deng, Juan Wen, Xiaoyuan Chen, Jiaqi Xu, Yu Liu, Huanhuan Wan, Jin Wang, Leping Yan, Yong He, Yunsheng Xu

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Background

Skin cutaneous melanoma (SKCM) is an extremely malignant tumor and accounts for the majority of skin cancer deaths. Aspartate beta-hydroxylase domain containing 1 (ASPHD1) may participate in cancer progression through controlling α-ketoglutarate-dependent dioxygenases. However, its role in skin cutaneous melanoma (SKCM) has not been well studied.

Methods

The gene expression data of ASPDH1 and differentially expressed genes (DEGs) from TCGA and GTEx were evaluated, and verified via the GEO database. Then, we performed GO/KEGG, GSEA, PPI network analysis to analyze the functional implications of the DEGs related to ASPHD1. Then, the association between the ASPHD1 expression and clinical parameters was investigated by Cox regression analysis. Subsequently, the survival time of SKCM patients was evaluated by plotting Kaplan-Meier curves. Moreover, we investigated the correlation between the ASPHD1 expression and lymphocytic infiltration by using the data from TISIDB and TIMER 2.0. Next, we explored the association between ASPHD1 expression and drug sensitivity. Finally, we validate the expression differences by analyzing the results of qPCR, Western blot from human normal epidermal melanocytes and melanoma cells, and immunohistochemistry (IHC) from non-tumor skin as well as melanoma tissues.

Results

The ASPHD1 expression level was significantly upregulated in several cancers, including SKCM especially SKCM-metastasis tissues, and patients with an increased ASPHD1 expression had longer overall survival time than low expression ones. The functional enrichment analysis of ASPHD1-related DEGs showed an association with cell development regulation and tumorigenic pathways. Furthermore, the increased ASPHD1 expression level was associated with the level of immunostimulors, immunoinhibitors, chemokines, and TILs, such as CD4+, CD8+ T cell, mast cell, Th2 cell, and dendritic cell. More interesting, we found that ASPHD1 expression was tightly associated with CTLA4 and CD276 which are immune checkpoint markers. Moreover, the upregulated expression of ASPHD1 exhibited higher IC50 values for 24 chemotherapy drugs, including doxorubicin, and masitinib. Finally, the differential expression of ASPHD1 in SKCM was validated by the results of qPCR, Western blot, and IHC.

Conclusion

The expression of ASPHD1 in SKCM patients is closely related to patient survival. ASPHD1 may participate in the regulation of tumor immune microenvironment. Additionally, it may serve as a prognostic biomarker for SKCM and future in-depth studies are necessary to explore its value.
Appendix
Available only for authorised users
Literature
1.
go back to reference Swetter SM, Tsao H, Bichakjian CK, Curiel-Lewandrowski C, Elder DE, Gershenwald JE, Guild V, Grant-Kels JM, Halpern AC, Johnson TM, et al. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol. 2019;80(1):208–50.CrossRefPubMed Swetter SM, Tsao H, Bichakjian CK, Curiel-Lewandrowski C, Elder DE, Gershenwald JE, Guild V, Grant-Kels JM, Halpern AC, Johnson TM, et al. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol. 2019;80(1):208–50.CrossRefPubMed
2.
go back to reference Welch HG, Mazer BL, Adamson AS. The Rapid rise in cutaneous melanoma diagnoses. N Engl J Med. 2021;384(1):72–9.CrossRefPubMed Welch HG, Mazer BL, Adamson AS. The Rapid rise in cutaneous melanoma diagnoses. N Engl J Med. 2021;384(1):72–9.CrossRefPubMed
3.
go back to reference Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, McCubrey JA, Candido S, Libra M. Cutaneous melanoma: from pathogenesis to therapy (review). Int J Oncol. 2018;52(4):1071–80.PubMedPubMedCentral Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, McCubrey JA, Candido S, Libra M. Cutaneous melanoma: from pathogenesis to therapy (review). Int J Oncol. 2018;52(4):1071–80.PubMedPubMedCentral
4.
go back to reference Hartman RI, Lin JY. Cutaneous Melanoma-A review in detection, staging, and management. Hematol Oncol Clin North Am. 2019;33(1):25–38.CrossRefPubMed Hartman RI, Lin JY. Cutaneous Melanoma-A review in detection, staging, and management. Hematol Oncol Clin North Am. 2019;33(1):25–38.CrossRefPubMed
5.
go back to reference Michielin O, van Akkooi ACJ, Ascierto PA, Dummer R, Keilholz U. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019;30(12):1884–901.CrossRefPubMed Michielin O, van Akkooi ACJ, Ascierto PA, Dummer R, Keilholz U. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019;30(12):1884–901.CrossRefPubMed
6.
go back to reference Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE Jr, You MJ, DePinho RA, McMahon M, Bosenberg M. Braf(V600E) cooperates with pten loss to induce metastatic melanoma. Nat Genet. 2009;41(5):544–52.CrossRefPubMedPubMedCentral Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE Jr, You MJ, DePinho RA, McMahon M, Bosenberg M. Braf(V600E) cooperates with pten loss to induce metastatic melanoma. Nat Genet. 2009;41(5):544–52.CrossRefPubMedPubMedCentral
7.
go back to reference Nissan MH, Pratilas CA, Jones AM, Ramirez R, Won H, Liu C, Tiwari S, Kong L, Hanrahan AJ, Yao Z, et al. Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence. Cancer Res. 2014;74(8):2340–50.CrossRefPubMedPubMedCentral Nissan MH, Pratilas CA, Jones AM, Ramirez R, Won H, Liu C, Tiwari S, Kong L, Hanrahan AJ, Yao Z, et al. Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence. Cancer Res. 2014;74(8):2340–50.CrossRefPubMedPubMedCentral
8.
go back to reference Schadendorf D, Fisher DE, Garbe C, Gershenwald JE, Grob JJ, Halpern A, Herlyn M, Marchetti MA, McArthur G, Ribas A et al. Melanoma. Nat Rev Dis Primers 2015, 1:15003. Schadendorf D, Fisher DE, Garbe C, Gershenwald JE, Grob JJ, Halpern A, Herlyn M, Marchetti MA, McArthur G, Ribas A et al. Melanoma. Nat Rev Dis Primers 2015, 1:15003.
9.
go back to reference Sarkar D, Leung EY, Baguley BC, Finlay GJ, Askarian-Amiri ME. Epigenetic regulation in human melanoma: past and future. Epigenetics. 2015;10(2):103–21.CrossRefPubMedPubMedCentral Sarkar D, Leung EY, Baguley BC, Finlay GJ, Askarian-Amiri ME. Epigenetic regulation in human melanoma: past and future. Epigenetics. 2015;10(2):103–21.CrossRefPubMedPubMedCentral
10.
go back to reference Moran B, Silva R, Perry AS, Gallagher WM. Epigenetics of malignant melanoma. Semin Cancer Biol. 2018;51:80–8.CrossRefPubMed Moran B, Silva R, Perry AS, Gallagher WM. Epigenetics of malignant melanoma. Semin Cancer Biol. 2018;51:80–8.CrossRefPubMed
11.
go back to reference Scatozza F, Moschella F, D’Arcangelo D, Rossi S, Tabolacci C, Giampietri C, Proietti E, Facchiano F, Facchiano A. Nicotinamide inhibits melanoma in vitro and in vivo. J Exp Clin Cancer Res. 2020;39(1):211.CrossRefPubMedPubMedCentral Scatozza F, Moschella F, D’Arcangelo D, Rossi S, Tabolacci C, Giampietri C, Proietti E, Facchiano F, Facchiano A. Nicotinamide inhibits melanoma in vitro and in vivo. J Exp Clin Cancer Res. 2020;39(1):211.CrossRefPubMedPubMedCentral
12.
go back to reference Ross CL, Kaushik S, Valdes-Rodriguez R, Anvekar R. MicroRNAs in cutaneous melanoma: role as diagnostic and prognostic biomarkers. J Cell Physiol. 2018;233(7):5133–41.CrossRefPubMed Ross CL, Kaushik S, Valdes-Rodriguez R, Anvekar R. MicroRNAs in cutaneous melanoma: role as diagnostic and prognostic biomarkers. J Cell Physiol. 2018;233(7):5133–41.CrossRefPubMed
13.
go back to reference Guha S, Rees E, Darvasi A, Ivanov D, Ikeda M, Bergen SE, Magnusson PK, Cormican P, Morris D, Gill M, et al. Implication of a rare deletion at distal 16p11.2 in schizophrenia. JAMA Psychiatry. 2013;70(3):253–60.CrossRefPubMedPubMedCentral Guha S, Rees E, Darvasi A, Ivanov D, Ikeda M, Bergen SE, Magnusson PK, Cormican P, Morris D, Gill M, et al. Implication of a rare deletion at distal 16p11.2 in schizophrenia. JAMA Psychiatry. 2013;70(3):253–60.CrossRefPubMedPubMedCentral
14.
go back to reference Zhao S, Choi M, Overton JD, Bellone S, Roque DM, Cocco E, Guzzo F, English DP, Varughese J, Gasparrini S, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A. 2013;110(8):2916–21.CrossRefPubMedPubMedCentral Zhao S, Choi M, Overton JD, Bellone S, Roque DM, Cocco E, Guzzo F, English DP, Varughese J, Gasparrini S, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A. 2013;110(8):2916–21.CrossRefPubMedPubMedCentral
15.
16.
go back to reference Baksh SC, Finley LWS. Metabolic coordination of cell fate by α-Ketoglutarate-dependent dioxygenases. Trends Cell Biol. 2021;31(1):24–36.CrossRefPubMed Baksh SC, Finley LWS. Metabolic coordination of cell fate by α-Ketoglutarate-dependent dioxygenases. Trends Cell Biol. 2021;31(1):24–36.CrossRefPubMed
17.
go back to reference Ogawa K, Lin Q, Li L, Bai X, Chen X, Chen H, Kong R, Wang Y, Zhu H, He F, et al. Aspartate β-hydroxylase promotes pancreatic ductal adenocarcinoma metastasis through activation of SRC signaling pathway. J Hematol Oncol. 2019;12(1):144.CrossRefPubMedPubMedCentral Ogawa K, Lin Q, Li L, Bai X, Chen X, Chen H, Kong R, Wang Y, Zhu H, He F, et al. Aspartate β-hydroxylase promotes pancreatic ductal adenocarcinoma metastasis through activation of SRC signaling pathway. J Hematol Oncol. 2019;12(1):144.CrossRefPubMedPubMedCentral
18.
go back to reference Lin Q, Chen X, Meng F, Ogawa K, Li M, Song R, Zhang S, Zhang Z, Kong X, Xu Q, et al. ASPH-notch Axis guided Exosomal delivery of Prometastatic Secretome renders breast Cancer multi-organ metastasis. Mol Cancer. 2019;18(1):156.CrossRefPubMedPubMedCentral Lin Q, Chen X, Meng F, Ogawa K, Li M, Song R, Zhang S, Zhang Z, Kong X, Xu Q, et al. ASPH-notch Axis guided Exosomal delivery of Prometastatic Secretome renders breast Cancer multi-organ metastasis. Mol Cancer. 2019;18(1):156.CrossRefPubMedPubMedCentral
19.
go back to reference Benelli R, Costa D, Mastracci L, Grillo F, Olsen MJ, Barboro P, Poggi A, Ferrari N. Aspartate-β-Hydroxylase: A Promising Target to Limit the Local Invasiveness of Colorectal Cancer.Cancers (Basel)2020, 12(4). Benelli R, Costa D, Mastracci L, Grillo F, Olsen MJ, Barboro P, Poggi A, Ferrari N. Aspartate-β-Hydroxylase: A Promising Target to Limit the Local Invasiveness of Colorectal Cancer.Cancers (Basel)2020, 12(4).
20.
go back to reference Huang CK, Iwagami Y, Zou J, Casulli S, Lu S, Nagaoka K, Ji C, Ogawa K, Cao KY, Gao JS, et al. Aspartate beta-hydroxylase promotes cholangiocarcinoma progression by modulating RB1 phosphorylation. Cancer Lett. 2018;429:1–10.CrossRefPubMedPubMedCentral Huang CK, Iwagami Y, Zou J, Casulli S, Lu S, Nagaoka K, Ji C, Ogawa K, Cao KY, Gao JS, et al. Aspartate beta-hydroxylase promotes cholangiocarcinoma progression by modulating RB1 phosphorylation. Cancer Lett. 2018;429:1–10.CrossRefPubMedPubMedCentral
21.
go back to reference Aihara A, Huang CK, Olsen MJ, Lin Q, Chung W, Tang Q, Dong X, Wands JR. A cell-surface β-hydroxylase is a biomarker and therapeutic target for hepatocellular carcinoma. Hepatology. 2014;60(4):1302–13.CrossRefPubMed Aihara A, Huang CK, Olsen MJ, Lin Q, Chung W, Tang Q, Dong X, Wands JR. A cell-surface β-hydroxylase is a biomarker and therapeutic target for hepatocellular carcinoma. Hepatology. 2014;60(4):1302–13.CrossRefPubMed
22.
go back to reference Liu Y, Wan HH, Tian DM, Xu XJ, Bi CL, Zhan XY, Huang BH, Xu YS, Yan LP. Development and Characterization of High Efficacy Cell-Penetrating Peptide via Modulation of the Histidine and Arginine Ratio for Gene Therapy. Materials (Basel) 2021, 14(16). Liu Y, Wan HH, Tian DM, Xu XJ, Bi CL, Zhan XY, Huang BH, Xu YS, Yan LP. Development and Characterization of High Efficacy Cell-Penetrating Peptide via Modulation of the Histidine and Arginine Ratio for Gene Therapy. Materials (Basel) 2021, 14(16).
25.
go back to reference Kanehisa M, Furumichi M, Sato Y, Kawashima M, Ishiguro-Watanabe M. KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 2023;51(D1):D587–92.CrossRefPubMed Kanehisa M, Furumichi M, Sato Y, Kawashima M, Ishiguro-Watanabe M. KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 2023;51(D1):D587–92.CrossRefPubMed
26.
go back to reference Bedzhov I, Graham SJ, Leung CY, Zernicka-Goetz M. Developmental plasticity, cell fate specification and morphogenesis in the early mouse embryo.Philos Trans R Soc Lond B Biol Sci2014, 369(1657). Bedzhov I, Graham SJ, Leung CY, Zernicka-Goetz M. Developmental plasticity, cell fate specification and morphogenesis in the early mouse embryo.Philos Trans R Soc Lond B Biol Sci2014, 369(1657).
27.
28.
29.
go back to reference Wang YN, Lee HH, Hsu JL, Yu D, Hung MC. The impact of PD-L1 N-linked glycosylation on cancer therapy and clinical diagnosis. J Biomed Sci. 2020;27(1):77.CrossRefPubMedPubMedCentral Wang YN, Lee HH, Hsu JL, Yu D, Hung MC. The impact of PD-L1 N-linked glycosylation on cancer therapy and clinical diagnosis. J Biomed Sci. 2020;27(1):77.CrossRefPubMedPubMedCentral
30.
go back to reference Agathocleous M, Meacham CE, Burgess RJ, Piskounova E, Zhao Z, Crane GM, Cowin BL, Bruner E, Murphy MM, Chen W, et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature. 2017;549(7673):476–81.CrossRefPubMedPubMedCentral Agathocleous M, Meacham CE, Burgess RJ, Piskounova E, Zhao Z, Crane GM, Cowin BL, Bruner E, Murphy MM, Chen W, et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature. 2017;549(7673):476–81.CrossRefPubMedPubMedCentral
31.
go back to reference Ressnerova A, Raudenska M, Holubova M, Svobodova M, Polanska H, Babula P, Masarik M, Gumulec J. Zinc and copper homeostasis in Head and Neck Cancer: review and Meta-analysis. Curr Med Chem. 2016;23(13):1304–30.CrossRefPubMed Ressnerova A, Raudenska M, Holubova M, Svobodova M, Polanska H, Babula P, Masarik M, Gumulec J. Zinc and copper homeostasis in Head and Neck Cancer: review and Meta-analysis. Curr Med Chem. 2016;23(13):1304–30.CrossRefPubMed
33.
go back to reference Skrajnowska D, Bobrowska-Korczak B. Role of Zinc in Immune System and Anti-Cancer Defense Mechanisms.Nutrients2019, 11(10). Skrajnowska D, Bobrowska-Korczak B. Role of Zinc in Immune System and Anti-Cancer Defense Mechanisms.Nutrients2019, 11(10).
34.
go back to reference Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A, Knapp S, Xiao K, Campbell SL, Thiele DJ, et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature. 2014;509(7501):492–6.CrossRefPubMedPubMedCentral Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A, Knapp S, Xiao K, Campbell SL, Thiele DJ, et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature. 2014;509(7501):492–6.CrossRefPubMedPubMedCentral
35.
go back to reference Di Serio C, Doria L, Pellerito S, Prudovsky I, Micucci I, Massi D, Landriscina M, Marchionni N, Masotti G, Tarantini F. The release of fibroblast growth factor-1 from melanoma cells requires copper ions and is mediated by phosphatidylinositol 3-kinase/Akt intracellular signaling pathway. Cancer Lett. 2008;267(1):67–74.CrossRefPubMed Di Serio C, Doria L, Pellerito S, Prudovsky I, Micucci I, Massi D, Landriscina M, Marchionni N, Masotti G, Tarantini F. The release of fibroblast growth factor-1 from melanoma cells requires copper ions and is mediated by phosphatidylinositol 3-kinase/Akt intracellular signaling pathway. Cancer Lett. 2008;267(1):67–74.CrossRefPubMed
36.
go back to reference Naka K, Ochiai R, Matsubara E, Kondo C, Yang KM, Hoshii T, Araki M, Araki K, Sotomaru Y, Sasaki K, et al. The lysophospholipase D enzyme Gdpd3 is required to maintain chronic myelogenous leukaemia stem cells. Nat Commun. 2020;11(1):4681.CrossRefPubMedPubMedCentral Naka K, Ochiai R, Matsubara E, Kondo C, Yang KM, Hoshii T, Araki M, Araki K, Sotomaru Y, Sasaki K, et al. The lysophospholipase D enzyme Gdpd3 is required to maintain chronic myelogenous leukaemia stem cells. Nat Commun. 2020;11(1):4681.CrossRefPubMedPubMedCentral
37.
go back to reference Wei XL, Luo TQ, Li JN, Xue ZC, Wang Y, Zhang Y, Chen YB, Peng C. Development and validation of a Prognostic Classifier based on lipid metabolism-related genes in gastric Cancer. Front Mol Biosci. 2021;8:691143.CrossRefPubMedPubMedCentral Wei XL, Luo TQ, Li JN, Xue ZC, Wang Y, Zhang Y, Chen YB, Peng C. Development and validation of a Prognostic Classifier based on lipid metabolism-related genes in gastric Cancer. Front Mol Biosci. 2021;8:691143.CrossRefPubMedPubMedCentral
38.
go back to reference Gong Z, Cho YW, Kim JE, Ge K, Chen J. Accumulation of Pax2 transactivation domain interaction protein (PTIP) at sites of DNA breaks via RNF8-dependent pathway is required for cell survival after DNA damage. J Biol Chem. 2009;284(11):7284–93.CrossRefPubMedPubMedCentral Gong Z, Cho YW, Kim JE, Ge K, Chen J. Accumulation of Pax2 transactivation domain interaction protein (PTIP) at sites of DNA breaks via RNF8-dependent pathway is required for cell survival after DNA damage. J Biol Chem. 2009;284(11):7284–93.CrossRefPubMedPubMedCentral
39.
go back to reference Cox E, Hwang W, Uzoma I, Hu J, Guzzo CM, Jeong J, Matunis MJ, Qian J, Zhu H, Blackshaw S. Global analysis of SUMO-Binding proteins identifies SUMOylation as a Key Regulator of the INO80 chromatin remodeling complex. Mol Cell Proteomics. 2017;16(5):812–23.CrossRefPubMedPubMedCentral Cox E, Hwang W, Uzoma I, Hu J, Guzzo CM, Jeong J, Matunis MJ, Qian J, Zhu H, Blackshaw S. Global analysis of SUMO-Binding proteins identifies SUMOylation as a Key Regulator of the INO80 chromatin remodeling complex. Mol Cell Proteomics. 2017;16(5):812–23.CrossRefPubMedPubMedCentral
40.
go back to reference An N, Zhao Y, Lan H, Zhang M, Yin Y, Yi C. SEZ6L2 knockdown impairs tumour growth by promoting caspase-dependent apoptosis in colorectal cancer. J Cell Mol Med. 2020;24(7):4223–32.CrossRefPubMedPubMedCentral An N, Zhao Y, Lan H, Zhang M, Yin Y, Yi C. SEZ6L2 knockdown impairs tumour growth by promoting caspase-dependent apoptosis in colorectal cancer. J Cell Mol Med. 2020;24(7):4223–32.CrossRefPubMedPubMedCentral
41.
go back to reference Liu Y, Li RH, Ren G, Jiang J. Suppression of KIF22 Inhibits Cell Proliferation and Xenograft Tumor Growth in Tongue Squamous Cell Carcinoma. Biomed Res Int 2020, 2020:6387545. Liu Y, Li RH, Ren G, Jiang J. Suppression of KIF22 Inhibits Cell Proliferation and Xenograft Tumor Growth in Tongue Squamous Cell Carcinoma. Biomed Res Int 2020, 2020:6387545.
42.
go back to reference Wang L, He M, Fu L, Jin Y. Exosomal release of microRNA-454 by breast cancer cells sustains biological properties of cancer stem cells via the PRRT2/Wnt axis in ovarian cancer. Life Sci. 2020;257:118024.CrossRefPubMed Wang L, He M, Fu L, Jin Y. Exosomal release of microRNA-454 by breast cancer cells sustains biological properties of cancer stem cells via the PRRT2/Wnt axis in ovarian cancer. Life Sci. 2020;257:118024.CrossRefPubMed
43.
go back to reference Tomimaru Y, Mishra S, Safran H, Charpentier KP, Martin W, De Groot AS, Gregory SH, Wands JR. Aspartate-β-hydroxylase induces epitope-specific T cell responses in hepatocellular carcinoma. Vaccine. 2015;33(10):1256–66.CrossRefPubMedPubMedCentral Tomimaru Y, Mishra S, Safran H, Charpentier KP, Martin W, De Groot AS, Gregory SH, Wands JR. Aspartate-β-hydroxylase induces epitope-specific T cell responses in hepatocellular carcinoma. Vaccine. 2015;33(10):1256–66.CrossRefPubMedPubMedCentral
Metadata
Title
Aspartate beta-hydroxylase domain containing 1 as a prognostic marker associated with immune infiltration in skin cutaneous melanoma
Authors
Shiquan Sun
Min Deng
Juan Wen
Xiaoyuan Chen
Jiaqi Xu
Yu Liu
Huanhuan Wan
Jin Wang
Leping Yan
Yong He
Yunsheng Xu
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2023
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10625-8

Other articles of this Issue 1/2023

BMC Cancer 1/2023 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