Skip to main content
Top
Published in: Medical Oncology 5/2024

Open Access 01-05-2024 | Glioblastoma | Original Paper

Exploring the combined anti-cancer effects of sodium butyrate and celastrol in glioblastoma cell lines: a novel therapeutic approach

Authors: Bahar Kartal, Farika Nur Denizler-Ebiri, Mustafa Güven, Filiz Taşpınar, Hande Canpınar, Sedat Çetin, Tuğçe Karaduman, Serkan Küççüktürk, Javier Castresana, Mehmet Taşpınar

Published in: Medical Oncology | Issue 5/2024

Login to get access

Abstract

Glioblastoma, a highly aggressive and lethal brain cancer, lacks effective treatment options and has a poor prognosis. In our study, we explored the potential anti-cancer effects of sodium butyrate (SB) and celastrol (CEL) in two glioblastoma cell lines. SB, a histone deacetylase inhibitor, and CEL, derived from the tripterygium wilfordii plant, act as mTOR and proteasome inhibitors. Both can cross the blood–brain barrier, and they exhibit chemo- and radiosensitive properties in various cancer models. GB cell lines LN-405 and T98G were treated with SB and CEL. Cell viability was assessed by MTT assay and IC50 values were obtained. Gene expression of DNA repair, apoptosis, and autophagy-related genes was analyzed by RT-PCR. Cell cycle distribution was determined using flow cytometry. Viability assays using MTT assay revealed IC50 values of 26 mM and 22.7 mM for SB and 6.77 μM, and 9.11 μM for CEL in LN-405 and T98G cells, respectively. Furthermore, we examined the expression levels of DNA repair genes (MGMT, MLH-1, MSH-2, MSH-6), apoptosis genes (caspase-3, caspase-8, caspase-9), and an autophagy gene (ATG-6) using real-time polymerase chain reaction. Additionally, flow cytometry analysis revealed alterations in cell cycle distribution following treatment with SB, CEL and their combination. These findings indicate that SB and CEL may act through multiple mechanisms, including DNA repair inhibition, apoptosis induction, and autophagy modulation, to exert their anti-cancer effects in glioblastoma cells. This is the first study providing novel insights into the potential therapeutic effects of SB and CEL in glioblastoma.
Literature
1.
go back to reference Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015–2019. Neuro Oncol. 2022;24:v1–95.PubMedPubMedCentralCrossRef Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015–2019. Neuro Oncol. 2022;24:v1–95.PubMedPubMedCentralCrossRef
2.
go back to reference Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23:1231–51.PubMedPubMedCentralCrossRef Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23:1231–51.PubMedPubMedCentralCrossRef
3.
go back to reference Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.PubMedCrossRef Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.PubMedCrossRef
4.
go back to reference Genoud V, Migliorini D. Challenging hurdles of current targeting in glioblastoma: A focus on immunotherapeutic strategies. Int J Mol Sci. 2021;22:3493.PubMedPubMedCentralCrossRef Genoud V, Migliorini D. Challenging hurdles of current targeting in glioblastoma: A focus on immunotherapeutic strategies. Int J Mol Sci. 2021;22:3493.PubMedPubMedCentralCrossRef
5.
go back to reference Güven M, Taşpınar F, Denizler-Ebiri FN, Castresana JS, Taşpınar M. The antagonistic effects of temozolomide and trichostatin a combination on MGMT and DNA mismatch repair pathways in Glioblastoma. Med Oncol. 2023;40:223.PubMedCrossRef Güven M, Taşpınar F, Denizler-Ebiri FN, Castresana JS, Taşpınar M. The antagonistic effects of temozolomide and trichostatin a combination on MGMT and DNA mismatch repair pathways in Glioblastoma. Med Oncol. 2023;40:223.PubMedCrossRef
6.
7.
go back to reference Candido EP, Reeves R, Davie JR. Sodium butyrate inhibits histone deacetylation in cultured cells. Cell. 1978;14:105–13.PubMedCrossRef Candido EP, Reeves R, Davie JR. Sodium butyrate inhibits histone deacetylation in cultured cells. Cell. 1978;14:105–13.PubMedCrossRef
8.
go back to reference Salimi V, Shahsavari Z, Safizadeh B, Hosseini A, Khademian N, Tavakoli-Yaraki M. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis. 2017;16:208.PubMedPubMedCentralCrossRef Salimi V, Shahsavari Z, Safizadeh B, Hosseini A, Khademian N, Tavakoli-Yaraki M. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis. 2017;16:208.PubMedPubMedCentralCrossRef
9.
go back to reference Xu Z, Tao J, Chen P, Chen L, Sharma S, Wang G, et al. Sodium Butyrate inhibits colorectal cancer cell migration by downregulating Bmi-1 through enhanced miR-200c expression. Mol Nutr Food Res. 2018;62:1700844.CrossRef Xu Z, Tao J, Chen P, Chen L, Sharma S, Wang G, et al. Sodium Butyrate inhibits colorectal cancer cell migration by downregulating Bmi-1 through enhanced miR-200c expression. Mol Nutr Food Res. 2018;62:1700844.CrossRef
10.
go back to reference Taylor MA, Khathayer F, Ray SK. Quercetin and sodium butyrate synergistically increase apoptosis in rat C6 and human T98G glioblastoma cells through inhibition of autophagy. Neurochem Res. 2019;44:1715–25.PubMedCrossRef Taylor MA, Khathayer F, Ray SK. Quercetin and sodium butyrate synergistically increase apoptosis in rat C6 and human T98G glioblastoma cells through inhibition of autophagy. Neurochem Res. 2019;44:1715–25.PubMedCrossRef
12.
go back to reference Çakır T, Güven M, Taşpınar M, Denizler FN, Kartal B. The effect of sodium butyrate on radiosensitivity in glioblastoma cell. Van Med J. 2019;26:550–6.CrossRef Çakır T, Güven M, Taşpınar M, Denizler FN, Kartal B. The effect of sodium butyrate on radiosensitivity in glioblastoma cell. Van Med J. 2019;26:550–6.CrossRef
13.
go back to reference Butler R, Bates GP. Histone deacetylase inhibitors as therapeutics for polyglutamine disorders. Nat Rev Neurosci. 2006;7:784–96.PubMedCrossRef Butler R, Bates GP. Histone deacetylase inhibitors as therapeutics for polyglutamine disorders. Nat Rev Neurosci. 2006;7:784–96.PubMedCrossRef
14.
go back to reference Yang H, Chen D, Cui QC, Yuan X, Dou QP. Celastrol, a triterpene extracted from the Chinese “Thunder of God Vine”, is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice. Cancer Res. 2006;66:4758–65.PubMedCrossRef Yang H, Chen D, Cui QC, Yuan X, Dou QP. Celastrol, a triterpene extracted from the Chinese “Thunder of God Vine”, is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice. Cancer Res. 2006;66:4758–65.PubMedCrossRef
15.
go back to reference Li X, Zhu G, Yao X, Wang N, Hu R, Kong Q, et al. Celastrol induces ubiquitin-dependent degradation of mTOR in breast cancer cells. Onco Targets Ther. 2018;11:8977–85.PubMedPubMedCentralCrossRef Li X, Zhu G, Yao X, Wang N, Hu R, Kong Q, et al. Celastrol induces ubiquitin-dependent degradation of mTOR in breast cancer cells. Onco Targets Ther. 2018;11:8977–85.PubMedPubMedCentralCrossRef
17.
go back to reference Kashyap D, Sharma A, Tuli HS, Sak K, Mukherjee T, Bishayee A. Molecular targets of celastrol in cancer: Recent trends and advancements. Crit Rev Oncol Hematol. 2018;128:70–81.PubMedCrossRef Kashyap D, Sharma A, Tuli HS, Sak K, Mukherjee T, Bishayee A. Molecular targets of celastrol in cancer: Recent trends and advancements. Crit Rev Oncol Hematol. 2018;128:70–81.PubMedCrossRef
18.
go back to reference Taspinar M, Denizler F, Güven M, Yüksek V, Çetin S, Dede S. The cytotoxic effects of 5-FU and celastrol on glioblastoma. Van Heal Sci J. 2018;11:5–10. Taspinar M, Denizler F, Güven M, Yüksek V, Çetin S, Dede S. The cytotoxic effects of 5-FU and celastrol on glioblastoma. Van Heal Sci J. 2018;11:5–10.
20.
go back to reference Kazemi Sefat NA, Mohammadi MM, Hadjati J, Talebi S, Ajami M, Daneshvar H. Sodium butyrate as a histone deacetylase inhibitor affects toll-like receptor 4 expression in colorectal cancer cell lines. Immunol Invest. 2019;48:759–69.PubMedCrossRef Kazemi Sefat NA, Mohammadi MM, Hadjati J, Talebi S, Ajami M, Daneshvar H. Sodium butyrate as a histone deacetylase inhibitor affects toll-like receptor 4 expression in colorectal cancer cell lines. Immunol Invest. 2019;48:759–69.PubMedCrossRef
21.
go back to reference Joachimiak R, Kaźnica A, Drewa T. Influence of sodium butyrate on hepatocellular carcinoma (hepG2) and glioblastoma (C6) cell lines in vitro. Acta Pol Pharm. 2007;64:561–3.PubMed Joachimiak R, Kaźnica A, Drewa T. Influence of sodium butyrate on hepatocellular carcinoma (hepG2) and glioblastoma (C6) cell lines in vitro. Acta Pol Pharm. 2007;64:561–3.PubMed
22.
go back to reference Wang Z, Zhai Z, Du X. Celastrol inhibits migration and invasion through blocking the NF-κB pathway in ovarian cancer cells. Exp Ther Med. 2017;14:819–24.PubMedPubMedCentralCrossRef Wang Z, Zhai Z, Du X. Celastrol inhibits migration and invasion through blocking the NF-κB pathway in ovarian cancer cells. Exp Ther Med. 2017;14:819–24.PubMedPubMedCentralCrossRef
23.
go back to reference Huang S, Tang Y, Cai X, Peng X, Liu X, Zhang L, et al. Celastrol inhibits vasculogenesis by suppressing the VEGF-induced functional activity of bone marrow-derived endothelial progenitor cells. Biochem Biophys Res Commun. 2012;423:467–72.PubMedCrossRef Huang S, Tang Y, Cai X, Peng X, Liu X, Zhang L, et al. Celastrol inhibits vasculogenesis by suppressing the VEGF-induced functional activity of bone marrow-derived endothelial progenitor cells. Biochem Biophys Res Commun. 2012;423:467–72.PubMedCrossRef
24.
go back to reference Yan Y, Guo Y, Zhang W, Ma C, Zhang Y, Wang C, et al. Celastrol enhanced the anticancer effect of lapatinib in human hepatocellular carcinoma cells in vitro. J BUON. 2014;19:412–8.PubMed Yan Y, Guo Y, Zhang W, Ma C, Zhang Y, Wang C, et al. Celastrol enhanced the anticancer effect of lapatinib in human hepatocellular carcinoma cells in vitro. J BUON. 2014;19:412–8.PubMed
25.
go back to reference Ren B, Liu H, Gao H, Liu S, Zhang Z, Fribley AM, et al. Celastrol induces apoptosis in hepatocellular carcinoma cells via targeting ER-stress/UPR. Oncotarget. 2017;8:93039–50.PubMedPubMedCentralCrossRef Ren B, Liu H, Gao H, Liu S, Zhang Z, Fribley AM, et al. Celastrol induces apoptosis in hepatocellular carcinoma cells via targeting ER-stress/UPR. Oncotarget. 2017;8:93039–50.PubMedPubMedCentralCrossRef
26.
go back to reference Cha Z, Cheng J, Xiang H, Qin J, He Y, Peng Z, et al. Celastrol enhances TRAIL-induced apoptosis in human glioblastoma via the death receptor pathway. Cancer Chemother Pharmacol. 2019;84:719–28.PubMedCrossRef Cha Z, Cheng J, Xiang H, Qin J, He Y, Peng Z, et al. Celastrol enhances TRAIL-induced apoptosis in human glioblastoma via the death receptor pathway. Cancer Chemother Pharmacol. 2019;84:719–28.PubMedCrossRef
27.
28.
go back to reference Maysinger D, Moquin A, Choi J, Kodiha M, Stochaj U. Gold nanourchins and celastrol reorganize the nucleo- and cytoskeleton of glioblastoma cells. Nanoscale. 2018;10:1716–26.PubMedCrossRef Maysinger D, Moquin A, Choi J, Kodiha M, Stochaj U. Gold nanourchins and celastrol reorganize the nucleo- and cytoskeleton of glioblastoma cells. Nanoscale. 2018;10:1716–26.PubMedCrossRef
29.
go back to reference Tsung AJ, Guda MR, Asuthkar S, Labak CM, Purvis IJ, Lu Y, et al. Methylation regulates HEY1 expression in glioblastoma. Oncotarget. 2017;8:44398–409.PubMedPubMedCentralCrossRef Tsung AJ, Guda MR, Asuthkar S, Labak CM, Purvis IJ, Lu Y, et al. Methylation regulates HEY1 expression in glioblastoma. Oncotarget. 2017;8:44398–409.PubMedPubMedCentralCrossRef
30.
go back to reference Coxhead JM, Williams EA, Mathers JC. DNA mismatch repair status may influence anti-neoplastic effects of butyrate. Biochem Soc Trans. 2005;33:728–9.PubMedCrossRef Coxhead JM, Williams EA, Mathers JC. DNA mismatch repair status may influence anti-neoplastic effects of butyrate. Biochem Soc Trans. 2005;33:728–9.PubMedCrossRef
33.
go back to reference Cosgrove DE, Cox GS. Effects of sodium butyrate and 5-azacytidine on DNA methylation in human tumor cell lines: variable response to drug treatment and withdrawal. Biochim Biophys Acta. 1990;1087:80–6.PubMedCrossRef Cosgrove DE, Cox GS. Effects of sodium butyrate and 5-azacytidine on DNA methylation in human tumor cell lines: variable response to drug treatment and withdrawal. Biochim Biophys Acta. 1990;1087:80–6.PubMedCrossRef
34.
go back to reference Sawa H, Murakami H, Ohshima Y, Sugino T, Nakajyo T, Kisanuki T, et al. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol. 2001;18:109–14.PubMedCrossRef Sawa H, Murakami H, Ohshima Y, Sugino T, Nakajyo T, Kisanuki T, et al. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol. 2001;18:109–14.PubMedCrossRef
35.
go back to reference Kannaiyan R, Shanmugam MK, Sethi G. Molecular targets of celastrol derived from Thunder of God Vine: potential role in the treatment of inflammatory disorders and cancer. Cancer Lett. 2011;303:9–20.PubMedCrossRef Kannaiyan R, Shanmugam MK, Sethi G. Molecular targets of celastrol derived from Thunder of God Vine: potential role in the treatment of inflammatory disorders and cancer. Cancer Lett. 2011;303:9–20.PubMedCrossRef
36.
go back to reference Yadav P, Jaswal V, Sharma A, Kashyap D, Tuli HS, Garg VK, et al. Celastrol as a pentacyclic triterpenoid with chemopreventive properties. Pharm Pat Anal. 2018;7:155–67.PubMedCrossRef Yadav P, Jaswal V, Sharma A, Kashyap D, Tuli HS, Garg VK, et al. Celastrol as a pentacyclic triterpenoid with chemopreventive properties. Pharm Pat Anal. 2018;7:155–67.PubMedCrossRef
37.
go back to reference Liu X, Zhao P, Wang X, Wang L, Zhu Y, Song Y, et al. Celastrol mediates autophagy and apoptosis via the ROS/JNK and Akt/mTOR signaling pathways in glioma cells. J Exp Clin Cancer Res. 2019;38:184.PubMedPubMedCentralCrossRef Liu X, Zhao P, Wang X, Wang L, Zhu Y, Song Y, et al. Celastrol mediates autophagy and apoptosis via the ROS/JNK and Akt/mTOR signaling pathways in glioma cells. J Exp Clin Cancer Res. 2019;38:184.PubMedPubMedCentralCrossRef
38.
go back to reference Querol Cano L, Lavery DN, Bevan CL. Mini-review: Foldosome regulation of androgen receptor action in prostate cancer. Mol Cell Endocrinol. 2013;369:52–62.CrossRef Querol Cano L, Lavery DN, Bevan CL. Mini-review: Foldosome regulation of androgen receptor action in prostate cancer. Mol Cell Endocrinol. 2013;369:52–62.CrossRef
39.
go back to reference Xu S-W, Law BYK, Mok SWF, Leung ELH, Fan XX, Coghi PS, et al. Autophagic degradation of epidermal growth factor receptor in gefitinib-resistant lung cancer by celastrol. Int J Oncol. 2016;49:1576–88.PubMedCrossRef Xu S-W, Law BYK, Mok SWF, Leung ELH, Fan XX, Coghi PS, et al. Autophagic degradation of epidermal growth factor receptor in gefitinib-resistant lung cancer by celastrol. Int J Oncol. 2016;49:1576–88.PubMedCrossRef
40.
go back to reference Li H-Y, Zhang J, Sun L-L, Li B-H, Gao H-L, Xie T, et al. Celastrol induces apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells: an in vitro and in vivo study. Cell Death Dis. 2015;6:e1604.PubMedPubMedCentralCrossRef Li H-Y, Zhang J, Sun L-L, Li B-H, Gao H-L, Xie T, et al. Celastrol induces apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells: an in vitro and in vivo study. Cell Death Dis. 2015;6:e1604.PubMedPubMedCentralCrossRef
41.
go back to reference Xu S-W, Law BYK, Qu SLQ, Hamdoun S, Chen J, Zhang W, et al. SERCA and P-glycoprotein inhibition and ATP depletion are necessary for celastrol-induced autophagic cell death and collateral sensitivity in multidrug-resistant tumor cells. Pharmacol Res. 2020;153:104660.PubMedCrossRef Xu S-W, Law BYK, Qu SLQ, Hamdoun S, Chen J, Zhang W, et al. SERCA and P-glycoprotein inhibition and ATP depletion are necessary for celastrol-induced autophagic cell death and collateral sensitivity in multidrug-resistant tumor cells. Pharmacol Res. 2020;153:104660.PubMedCrossRef
42.
go back to reference Lee D, Kim IY, Saha S, Choi KS. Paraptosis in the anti-cancer arsenal of natural products. Pharmacol Ther. 2016;162:120–33.PubMedCrossRef Lee D, Kim IY, Saha S, Choi KS. Paraptosis in the anti-cancer arsenal of natural products. Pharmacol Ther. 2016;162:120–33.PubMedCrossRef
43.
go back to reference Boridy S, Le PU, Petrecca K, Maysinger D. Celastrol targets proteostasis and acts synergistically with a heat-shock protein 90 inhibitor to kill human glioblastoma cells. Cell Death Dis. 2014;5:e1216.PubMedPubMedCentralCrossRef Boridy S, Le PU, Petrecca K, Maysinger D. Celastrol targets proteostasis and acts synergistically with a heat-shock protein 90 inhibitor to kill human glioblastoma cells. Cell Death Dis. 2014;5:e1216.PubMedPubMedCentralCrossRef
44.
go back to reference Yu C, Friday BB, Yang L, Atadja P, Wigle D, Sarkaria J, et al. Mitochondrial Bax translocation partially mediates synergistic cytotoxicity between histone deacetylase inhibitors and proteasome inhibitors in glioma cells. Neuro Oncol. 2008;10:309–19.PubMedPubMedCentralCrossRef Yu C, Friday BB, Yang L, Atadja P, Wigle D, Sarkaria J, et al. Mitochondrial Bax translocation partially mediates synergistic cytotoxicity between histone deacetylase inhibitors and proteasome inhibitors in glioma cells. Neuro Oncol. 2008;10:309–19.PubMedPubMedCentralCrossRef
45.
go back to reference Asklund T, Kvarnbrink S, Holmlund C, Wibom C, Bergenheim T, Henriksson R, et al. Synergistic killing of glioblastoma stem-like cells by bortezomib and HDAC inhibitors. Anticancer Res. 2012;32:2407–13.PubMed Asklund T, Kvarnbrink S, Holmlund C, Wibom C, Bergenheim T, Henriksson R, et al. Synergistic killing of glioblastoma stem-like cells by bortezomib and HDAC inhibitors. Anticancer Res. 2012;32:2407–13.PubMed
46.
go back to reference Premkumar DR, Jane EP, Agostino NR, DiDomenico JD, Pollack IF. Bortezomib-induced sensitization of malignant human glioma cells to vorinostat-induced apoptosis depends on reactive oxygen species production, mitochondrial dysfunction, Noxa upregulation, Mcl-1 cleavage, and DNA damage. Mol Carcinog. 2013;52:118–33.PubMedCrossRef Premkumar DR, Jane EP, Agostino NR, DiDomenico JD, Pollack IF. Bortezomib-induced sensitization of malignant human glioma cells to vorinostat-induced apoptosis depends on reactive oxygen species production, mitochondrial dysfunction, Noxa upregulation, Mcl-1 cleavage, and DNA damage. Mol Carcinog. 2013;52:118–33.PubMedCrossRef
47.
go back to reference Das S, Deng X, Camphausen K, Shankavaram U. synthetic lethal drug combinations targeting proteasome and histone deacetylase inhibitors in TP53-mutated cancers. Arch cancer Biol Ther. 2020;1:42–7.PubMedPubMedCentral Das S, Deng X, Camphausen K, Shankavaram U. synthetic lethal drug combinations targeting proteasome and histone deacetylase inhibitors in TP53-mutated cancers. Arch cancer Biol Ther. 2020;1:42–7.PubMedPubMedCentral
48.
go back to reference Engelhard HH, Duncan HA, Kim S, Criswell PS, Van Eldik L. Therapeutic effects of sodium butyrate on glioma cells in vitro and in the rat C6 glioma model. Neurosurgery. 2001;48:616–25.PubMedCrossRef Engelhard HH, Duncan HA, Kim S, Criswell PS, Van Eldik L. Therapeutic effects of sodium butyrate on glioma cells in vitro and in the rat C6 glioma model. Neurosurgery. 2001;48:616–25.PubMedCrossRef
Metadata
Title
Exploring the combined anti-cancer effects of sodium butyrate and celastrol in glioblastoma cell lines: a novel therapeutic approach
Authors
Bahar Kartal
Farika Nur Denizler-Ebiri
Mustafa Güven
Filiz Taşpınar
Hande Canpınar
Sedat Çetin
Tuğçe Karaduman
Serkan Küççüktürk
Javier Castresana
Mehmet Taşpınar
Publication date
01-05-2024
Publisher
Springer US
Published in
Medical Oncology / Issue 5/2024
Print ISSN: 1357-0560
Electronic ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-024-02340-6

Other articles of this Issue 5/2024

Medical Oncology 5/2024 Go to the issue
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.