Skip to main content
Top
Published in: Journal of Translational Medicine 1/2021

Open Access 01-12-2021 | Radiotherapy | Review

GSTP1 as a novel target in radiation induced lung injury

Authors: Xiao Lei, Lehui Du, Wei Yu, Yao Wang, Na Ma, Baolin Qu

Published in: Journal of Translational Medicine | Issue 1/2021

Login to get access

Abstract

The glutathione S-transferase P1(GSTP1) is an isoenzyme in the glutathione-S transferases (GSTs) enzyme system, which is the most abundant GSTs expressed in adult lungs. Recent research shows that GSTP1 is closely related to the regulation of cell oxidative stress, inhibition of cell apoptosis and promotion of cytotoxic metabolism. Interestingly, there is evidence that GSTP1 single nucleotide polymorphisms (SNP) 105Ile/Val related to the risk of radiation induced lung injury (RILI) development, which strongly suggests that GSTP1 is closely associated with the occurrence and development of RILI. In this review, we discuss our understanding of the role of GSTP1 in RILI and its possible mechanism.
Literature
5.
go back to reference Xia C, et al. Prevention and treatment of radiation-induced lung injury. Future Med Chem. 2020;12(23):2161–73.PubMedCrossRef Xia C, et al. Prevention and treatment of radiation-induced lung injury. Future Med Chem. 2020;12(23):2161–73.PubMedCrossRef
6.
go back to reference Alizadeh E, Orlando TM, Sanche L. Biomolecular damage induced by ionizing radiation: the direct and indirect effects of low-energy electrons on DNA. Annu Rev Phys Chem. 2015;66:379–98.PubMedCrossRef Alizadeh E, Orlando TM, Sanche L. Biomolecular damage induced by ionizing radiation: the direct and indirect effects of low-energy electrons on DNA. Annu Rev Phys Chem. 2015;66:379–98.PubMedCrossRef
7.
go back to reference Pouget JP, Georgakilas AG, Ravanat JL. Targeted and off-target (bystander and abscopal) effects of radiation therapy: redox mechanisms and risk/benefit analysis. Antioxid Redox Signal. 2018;29(15):1447–87.PubMedPubMedCentralCrossRef Pouget JP, Georgakilas AG, Ravanat JL. Targeted and off-target (bystander and abscopal) effects of radiation therapy: redox mechanisms and risk/benefit analysis. Antioxid Redox Signal. 2018;29(15):1447–87.PubMedPubMedCentralCrossRef
8.
go back to reference Xue J, et al. Gene-modified mesenchymal stem cells protect against radiation-induced lung injury. Mol Ther. 2013;21(2):456–65.PubMedCrossRef Xue J, et al. Gene-modified mesenchymal stem cells protect against radiation-induced lung injury. Mol Ther. 2013;21(2):456–65.PubMedCrossRef
9.
go back to reference Zhou J, et al. Lung tissue extracellular matrix-derived hydrogels protect against radiation-induced lung injury by suppressing epithelial-mesenchymal transition. J Cell Physiol. 2020;235(3):2377–88.PubMedCrossRef Zhou J, et al. Lung tissue extracellular matrix-derived hydrogels protect against radiation-induced lung injury by suppressing epithelial-mesenchymal transition. J Cell Physiol. 2020;235(3):2377–88.PubMedCrossRef
10.
go back to reference Jin H, et al. Radiation-induced lung fibrosis: preclinical animal models and therapeutic strategies. Cancers (Basel). 2020;12(6):1561.CrossRef Jin H, et al. Radiation-induced lung fibrosis: preclinical animal models and therapeutic strategies. Cancers (Basel). 2020;12(6):1561.CrossRef
11.
go back to reference Du L, et al. GSTP1 Ile105Val polymorphism might be associated with the risk of radiation pneumonitis among lung cancer patients in Chinese population: a prospective study. J Cancer. 2018;9(4):726–35.PubMedPubMedCentralCrossRef Du L, et al. GSTP1 Ile105Val polymorphism might be associated with the risk of radiation pneumonitis among lung cancer patients in Chinese population: a prospective study. J Cancer. 2018;9(4):726–35.PubMedPubMedCentralCrossRef
12.
go back to reference Du L, et al. Association of DNA repair gene polymorphisms with the risk of radiation pneumonitis in lung cancer patients. Oncotarget. 2018;9(1):958–68.PubMedCrossRef Du L, et al. Association of DNA repair gene polymorphisms with the risk of radiation pneumonitis in lung cancer patients. Oncotarget. 2018;9(1):958–68.PubMedCrossRef
13.
go back to reference Ali-Osman F, et al. Molecular cloning, characterization, and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase Pi gene variants. Evidence for differential catalytic activity of the encoded proteins. J Biol Chem. 1997;272(15):10004–12.PubMedCrossRef Ali-Osman F, et al. Molecular cloning, characterization, and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase Pi gene variants. Evidence for differential catalytic activity of the encoded proteins. J Biol Chem. 1997;272(15):10004–12.PubMedCrossRef
14.
go back to reference Nørskov MS, Dahl M, Tybjærg-Hansen A. Genetic Variation in GSTP1, lung function, risk of lung cancer, and mortality. J Thorac Oncol. 2017;12(11):1664–72.PubMedCrossRef Nørskov MS, Dahl M, Tybjærg-Hansen A. Genetic Variation in GSTP1, lung function, risk of lung cancer, and mortality. J Thorac Oncol. 2017;12(11):1664–72.PubMedCrossRef
16.
go back to reference Chatterjee A, Gupta S. The multifaceted role of glutathione S-transferases in cancer. Cancer Lett. 2018;433:33–42.PubMedCrossRef Chatterjee A, Gupta S. The multifaceted role of glutathione S-transferases in cancer. Cancer Lett. 2018;433:33–42.PubMedCrossRef
17.
go back to reference Cui J, et al. GSTP1 and cancer: expression, methylation, polymorphisms and signaling (Review). Int J Oncol. 2020;56(4):867–78.PubMed Cui J, et al. GSTP1 and cancer: expression, methylation, polymorphisms and signaling (Review). Int J Oncol. 2020;56(4):867–78.PubMed
21.
go back to reference Mian OY, et al. GSTP1 Loss results in accumulation of oxidative DNA base damage and promotes prostate cancer cell survival following exposure to protracted oxidative stress. Prostate. 2016;76(2):199–206.PubMedCrossRef Mian OY, et al. GSTP1 Loss results in accumulation of oxidative DNA base damage and promotes prostate cancer cell survival following exposure to protracted oxidative stress. Prostate. 2016;76(2):199–206.PubMedCrossRef
22.
go back to reference Qiu K, Zheng Z, Huang Y. Long intergenic noncoding RNA 00844 promotes apoptosis and represses proliferation of prostate cancer cells through upregulating GSTP1 by recruiting EBF1. J Cell Physiol. 2020;235(11):8472–85.PubMedCrossRef Qiu K, Zheng Z, Huang Y. Long intergenic noncoding RNA 00844 promotes apoptosis and represses proliferation of prostate cancer cells through upregulating GSTP1 by recruiting EBF1. J Cell Physiol. 2020;235(11):8472–85.PubMedCrossRef
24.
go back to reference Spiteri MA, et al. Polymorphisms at the glutathione S-transferase, GSTP1 locus: a novel mechanism for susceptibility and development of atopic airway inflammation. Allergy. 2000;55(Suppl 61):15–20.PubMedCrossRef Spiteri MA, et al. Polymorphisms at the glutathione S-transferase, GSTP1 locus: a novel mechanism for susceptibility and development of atopic airway inflammation. Allergy. 2000;55(Suppl 61):15–20.PubMedCrossRef
25.
go back to reference De Nunzio C, et al. The controversial relationship between benign prostatic hyperplasia and prostate cancer: the role of inflammation. Eur Urol. 2011;60(1):106–17.PubMedCrossRef De Nunzio C, et al. The controversial relationship between benign prostatic hyperplasia and prostate cancer: the role of inflammation. Eur Urol. 2011;60(1):106–17.PubMedCrossRef
26.
go back to reference Malerba G, Pignatti PF. A review of asthma genetics: gene expression studies and recent candidates. J Appl Genet. 2005;46(1):93–104.PubMed Malerba G, Pignatti PF. A review of asthma genetics: gene expression studies and recent candidates. J Appl Genet. 2005;46(1):93–104.PubMed
27.
go back to reference Ściskalska M, Milnerowicz H. Activity of glutathione S-transferase and its π isoenzyme in the context of single nucleotide polymorphism in the GSTP1 gene (rs1695) and tobacco smoke exposure in the patients with acute pancreatitis and healthy subjects. Biomed Pharmacother. 2021;140:111589.PubMedCrossRef Ściskalska M, Milnerowicz H. Activity of glutathione S-transferase and its π isoenzyme in the context of single nucleotide polymorphism in the GSTP1 gene (rs1695) and tobacco smoke exposure in the patients with acute pancreatitis and healthy subjects. Biomed Pharmacother. 2021;140:111589.PubMedCrossRef
30.
go back to reference Wu Y, et al. Human glutathione S-transferase P1–1 interacts with TRAF2 and regulates TRAF2-ASK1 signals. Oncogene. 2006;25(42):5787–800.PubMedCrossRef Wu Y, et al. Human glutathione S-transferase P1–1 interacts with TRAF2 and regulates TRAF2-ASK1 signals. Oncogene. 2006;25(42):5787–800.PubMedCrossRef
31.
go back to reference Yang Y, et al. GSTpi regulates VE-cadherin stabilization through promoting S-glutathionylation of Src. Redox Biol. 2020;30:101416.PubMedCrossRef Yang Y, et al. GSTpi regulates VE-cadherin stabilization through promoting S-glutathionylation of Src. Redox Biol. 2020;30:101416.PubMedCrossRef
32.
go back to reference Bartolini D, et al. Glutathione S-transferase pi expression regulates the Nrf2-dependent response to hormetic diselenides. Free Radic Biol Med. 2015;88(Pt B):466–80.PubMedCrossRef Bartolini D, et al. Glutathione S-transferase pi expression regulates the Nrf2-dependent response to hormetic diselenides. Free Radic Biol Med. 2015;88(Pt B):466–80.PubMedCrossRef
33.
go back to reference Luo L, et al. Recombinant protein glutathione S-transferases P1 attenuates inflammation in mice. Mol Immunol. 2009;46(5):848–57.PubMedCrossRef Luo L, et al. Recombinant protein glutathione S-transferases P1 attenuates inflammation in mice. Mol Immunol. 2009;46(5):848–57.PubMedCrossRef
34.
go back to reference Bi X, et al. GSTP1 inhibits LPS-induced inflammatory response through regulating autophagy in THP-1 cells. Inflammation. 2020;43(3):1157–69.PubMedCrossRef Bi X, et al. GSTP1 inhibits LPS-induced inflammatory response through regulating autophagy in THP-1 cells. Inflammation. 2020;43(3):1157–69.PubMedCrossRef
35.
go back to reference Jones JT, et al. Glutathione S-transferase pi modulates NF-κB activation and pro-inflammatory responses in lung epithelial cells. Redox Biol. 2016;8:375–82.PubMedPubMedCentralCrossRef Jones JT, et al. Glutathione S-transferase pi modulates NF-κB activation and pro-inflammatory responses in lung epithelial cells. Redox Biol. 2016;8:375–82.PubMedPubMedCentralCrossRef
36.
go back to reference Rivera-Ingraham GA, et al. Salinity stress from the perspective of the energy-redox axis: lessons from a marine intertidal flatworm. Redox Biol. 2016;10:53–64.PubMedPubMedCentralCrossRef Rivera-Ingraham GA, et al. Salinity stress from the perspective of the energy-redox axis: lessons from a marine intertidal flatworm. Redox Biol. 2016;10:53–64.PubMedPubMedCentralCrossRef
37.
go back to reference Fletcher ME, et al. Influence of glutathione-S-transferase (GST) inhibition on lung epithelial cell injury: role of oxidative stress and metabolism. Am J Physiol Lung Cell Mol Physiol. 2015;308(12):L1274–85.PubMedCrossRef Fletcher ME, et al. Influence of glutathione-S-transferase (GST) inhibition on lung epithelial cell injury: role of oxidative stress and metabolism. Am J Physiol Lung Cell Mol Physiol. 2015;308(12):L1274–85.PubMedCrossRef
38.
go back to reference Giridhar P, et al. Radiation induced lung injury: prediction, assessment and management. Asian Pac J Cancer Prev. 2015;16(7):2613–7.PubMedCrossRef Giridhar P, et al. Radiation induced lung injury: prediction, assessment and management. Asian Pac J Cancer Prev. 2015;16(7):2613–7.PubMedCrossRef
39.
go back to reference Edvardsen H, et al. Germline glutathione S-transferase variants in breast cancer: relation to diagnosis and cutaneous long-term adverse effects after two fractionation patterns of radiotherapy. Int J Radiat Oncol Biol Phys. 2007;67(4):1163–71.PubMedCrossRef Edvardsen H, et al. Germline glutathione S-transferase variants in breast cancer: relation to diagnosis and cutaneous long-term adverse effects after two fractionation patterns of radiotherapy. Int J Radiat Oncol Biol Phys. 2007;67(4):1163–71.PubMedCrossRef
40.
go back to reference Terrazzino S, et al. Common variants of eNOS and XRCC1 genes may predict acute skin toxicity in breast cancer patients receiving radiotherapy after breast conserving surgery. Radiother Oncol. 2012;103(2):199–205.PubMedCrossRef Terrazzino S, et al. Common variants of eNOS and XRCC1 genes may predict acute skin toxicity in breast cancer patients receiving radiotherapy after breast conserving surgery. Radiother Oncol. 2012;103(2):199–205.PubMedCrossRef
41.
go back to reference Edvardsen H, et al. SNP in TXNRD2 associated with radiation-induced fibrosis: a study of genetic variation in reactive oxygen species metabolism and signaling. Int J Radiat Oncol Biol Phys. 2013;86(4):791–9.PubMedCrossRef Edvardsen H, et al. SNP in TXNRD2 associated with radiation-induced fibrosis: a study of genetic variation in reactive oxygen species metabolism and signaling. Int J Radiat Oncol Biol Phys. 2013;86(4):791–9.PubMedCrossRef
42.
43.
go back to reference Terrazzino S, et al. Common variants of GSTP1, GSTA1, and TGFβ1 are associated with the risk of radiation-induced fibrosis in breast cancer patients. Int J Radiat Oncol Biol Phys. 2012;83(2):504–11.PubMedCrossRef Terrazzino S, et al. Common variants of GSTP1, GSTA1, and TGFβ1 are associated with the risk of radiation-induced fibrosis in breast cancer patients. Int J Radiat Oncol Biol Phys. 2012;83(2):504–11.PubMedCrossRef
44.
go back to reference Sau A, et al. Targeting GSTP1-1 induces JNK activation and leads to apoptosis in cisplatin-sensitive and -resistant human osteosarcoma cell lines. Mol Biosyst. 2012;8(4):994–1006.PubMedCrossRef Sau A, et al. Targeting GSTP1-1 induces JNK activation and leads to apoptosis in cisplatin-sensitive and -resistant human osteosarcoma cell lines. Mol Biosyst. 2012;8(4):994–1006.PubMedCrossRef
45.
go back to reference Mohammad J, et al. JNK inhibition blocks piperlongumine-induced cell death and transcriptional activation of heme oxygenase-1 in pancreatic cancer cells. Apoptosis. 2019;24(9–10):730–44.PubMedPubMedCentralCrossRef Mohammad J, et al. JNK inhibition blocks piperlongumine-induced cell death and transcriptional activation of heme oxygenase-1 in pancreatic cancer cells. Apoptosis. 2019;24(9–10):730–44.PubMedPubMedCentralCrossRef
46.
go back to reference Yang X, et al. Novel oxadiazole analogues derived from ethacrynic acid: design, synthesis, and structure-activity relationships in inhibiting the activity of glutathione S-transferase P1–1 and cancer cell proliferation. J Med Chem. 2010;53(3):1015–22.PubMedCrossRef Yang X, et al. Novel oxadiazole analogues derived from ethacrynic acid: design, synthesis, and structure-activity relationships in inhibiting the activity of glutathione S-transferase P1–1 and cancer cell proliferation. J Med Chem. 2010;53(3):1015–22.PubMedCrossRef
47.
go back to reference Wang T, et al. Glutathione S-transferase P1–1 (GSTP1-1) inhibits c-Jun N-terminal kinase (JNK1) signaling through interaction with the C terminus. J Biol Chem. 2001;276(24):20999–1003.PubMedCrossRef Wang T, et al. Glutathione S-transferase P1–1 (GSTP1-1) inhibits c-Jun N-terminal kinase (JNK1) signaling through interaction with the C terminus. J Biol Chem. 2001;276(24):20999–1003.PubMedCrossRef
48.
go back to reference Ruscoe JE, et al. Pharmacologic or genetic manipulation of glutathione S-transferase P1–1 (GSTpi) influences cell proliferation pathways. J Pharmacol Exp Ther. 2001;298(1):339–45.PubMed Ruscoe JE, et al. Pharmacologic or genetic manipulation of glutathione S-transferase P1–1 (GSTpi) influences cell proliferation pathways. J Pharmacol Exp Ther. 2001;298(1):339–45.PubMed
49.
go back to reference Liu G, et al. Ethacrynic acid oxadiazole analogs induce apoptosis in malignant hematologic cells through downregulation of Mcl-1 and c-FLIP, which was attenuated by GSTP1-1. Mol Cancer Ther. 2013;12(9):1837–47.PubMedCrossRef Liu G, et al. Ethacrynic acid oxadiazole analogs induce apoptosis in malignant hematologic cells through downregulation of Mcl-1 and c-FLIP, which was attenuated by GSTP1-1. Mol Cancer Ther. 2013;12(9):1837–47.PubMedCrossRef
50.
go back to reference Mcmillan D, et al. Attenuation of GSTP1 decreases Fas SGlutathionylation-mediated lung epithelial cell apoptosis and fibrotic remodeling. Free Radical Biol Med. 2014;76:S164.CrossRef Mcmillan D, et al. Attenuation of GSTP1 decreases Fas SGlutathionylation-mediated lung epithelial cell apoptosis and fibrotic remodeling. Free Radical Biol Med. 2014;76:S164.CrossRef
52.
go back to reference Pollom EL, et al. Normal tissue constraints for abdominal and thoracic stereotactic body radiotherapy. Semin Radiat Oncol. 2017;27(3):197–208.PubMedCrossRef Pollom EL, et al. Normal tissue constraints for abdominal and thoracic stereotactic body radiotherapy. Semin Radiat Oncol. 2017;27(3):197–208.PubMedCrossRef
53.
go back to reference Duijm M, et al. Dose and volume of the irradiated main bronchi and related side effects in the treatment of central lung tumors with stereotactic radiotherapy. Semin Radiat Oncol. 2016;26(2):140–8.PubMedCrossRef Duijm M, et al. Dose and volume of the irradiated main bronchi and related side effects in the treatment of central lung tumors with stereotactic radiotherapy. Semin Radiat Oncol. 2016;26(2):140–8.PubMedCrossRef
55.
56.
go back to reference Xu S, Liu C, Ji HL. Concise review: therapeutic potential of the mesenchymal stem cell derived secretome and extracellular vesicles for radiation-induced lung injury: progress and hypotheses. Stem Cells Transl Med. 2019;8(4):344–54.PubMedPubMedCentralCrossRef Xu S, Liu C, Ji HL. Concise review: therapeutic potential of the mesenchymal stem cell derived secretome and extracellular vesicles for radiation-induced lung injury: progress and hypotheses. Stem Cells Transl Med. 2019;8(4):344–54.PubMedPubMedCentralCrossRef
57.
go back to reference Guan D, et al. Lung endothelial cell-targeted peptide-guided bFGF promotes the regeneration after radiation induced lung injury. Biomaterials. 2018;184:10–9.PubMedCrossRef Guan D, et al. Lung endothelial cell-targeted peptide-guided bFGF promotes the regeneration after radiation induced lung injury. Biomaterials. 2018;184:10–9.PubMedCrossRef
58.
go back to reference Meziani L, et al. CSF1R inhibition prevents radiation pulmonary fibrosis by depletion of interstitial macrophages. Eur Respir J. 2018;51(3):1702120.PubMedCrossRef Meziani L, et al. CSF1R inhibition prevents radiation pulmonary fibrosis by depletion of interstitial macrophages. Eur Respir J. 2018;51(3):1702120.PubMedCrossRef
59.
go back to reference Townsend DM, Tew KD. Cancer drugs, genetic variation and the glutathione-S-transferase gene family. Am J Pharmaco Genomics. 2003;3(3):157–72.CrossRef Townsend DM, Tew KD. Cancer drugs, genetic variation and the glutathione-S-transferase gene family. Am J Pharmaco Genomics. 2003;3(3):157–72.CrossRef
60.
go back to reference Gurioli G, et al. GSTP1 methylation in cancer: a liquid biopsy biomarker? Clin Chem Lab Med. 2018;56(5):702–17.PubMedCrossRef Gurioli G, et al. GSTP1 methylation in cancer: a liquid biopsy biomarker? Clin Chem Lab Med. 2018;56(5):702–17.PubMedCrossRef
61.
go back to reference Okamura T, et al. Phosphorylation of glutathione S-transferase P1 (GSTP1) by epidermal growth factor receptor (EGFR) promotes formation of the GSTP1-c-Jun N-terminal kinase (JNK) complex and suppresses JNK downstream signaling and apoptosis in brain tumor cells. J Biol Chem. 2015;290(52):30866–78.PubMedPubMedCentralCrossRef Okamura T, et al. Phosphorylation of glutathione S-transferase P1 (GSTP1) by epidermal growth factor receptor (EGFR) promotes formation of the GSTP1-c-Jun N-terminal kinase (JNK) complex and suppresses JNK downstream signaling and apoptosis in brain tumor cells. J Biol Chem. 2015;290(52):30866–78.PubMedPubMedCentralCrossRef
63.
go back to reference Lei K, et al. C/EBPβ mediates NQO1 and GSTP1 anti-oxidative reductases expression in glioblastoma, promoting brain tumor proliferation. Redox Biol. 2020;34:101578.PubMedPubMedCentralCrossRef Lei K, et al. C/EBPβ mediates NQO1 and GSTP1 anti-oxidative reductases expression in glioblastoma, promoting brain tumor proliferation. Redox Biol. 2020;34:101578.PubMedPubMedCentralCrossRef
64.
go back to reference Li Q, et al. Association of GSTP1 and P16 promoter methylation with the risk of HBV-related hepatocellular carcinoma: a meta-analysis. Onco Targets Ther. 2018;11:5789–96.PubMedPubMedCentralCrossRef Li Q, et al. Association of GSTP1 and P16 promoter methylation with the risk of HBV-related hepatocellular carcinoma: a meta-analysis. Onco Targets Ther. 2018;11:5789–96.PubMedPubMedCentralCrossRef
65.
go back to reference Patel PG, et al. A three-gene DNA methylation biomarker accurately classifies early stage prostate cancer. Prostate. 2019;79(14):1705–14.PubMedCrossRef Patel PG, et al. A three-gene DNA methylation biomarker accurately classifies early stage prostate cancer. Prostate. 2019;79(14):1705–14.PubMedCrossRef
66.
go back to reference Yang SJ, et al. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene. 2017;623:5–14.PubMedCrossRef Yang SJ, et al. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene. 2017;623:5–14.PubMedCrossRef
67.
go back to reference Li J, et al. Transcriptional activation of Gstp1 by MEK/ERK signaling confers chemo-resistance to cisplatin in lung cancer stem cells. Front Oncol. 2019;9:476.PubMedPubMedCentralCrossRef Li J, et al. Transcriptional activation of Gstp1 by MEK/ERK signaling confers chemo-resistance to cisplatin in lung cancer stem cells. Front Oncol. 2019;9:476.PubMedPubMedCentralCrossRef
68.
go back to reference Zhang P, et al. DLJ14, a novel chemo-sensitization agent, enhances therapeutic effects of adriamycin against MCF-7/A cells both in vitro and in vivo. J Pharm Pharmacol. 2014;66(3):398–407.PubMedCrossRef Zhang P, et al. DLJ14, a novel chemo-sensitization agent, enhances therapeutic effects of adriamycin against MCF-7/A cells both in vitro and in vivo. J Pharm Pharmacol. 2014;66(3):398–407.PubMedCrossRef
Metadata
Title
GSTP1 as a novel target in radiation induced lung injury
Authors
Xiao Lei
Lehui Du
Wei Yu
Yao Wang
Na Ma
Baolin Qu
Publication date
01-12-2021
Publisher
BioMed Central
Keyword
Radiotherapy
Published in
Journal of Translational Medicine / Issue 1/2021
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-021-02978-0

Other articles of this Issue 1/2021

Journal of Translational Medicine 1/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.