Skip to main content
Top
Published in: BMC Cancer 1/2020

Open Access 01-12-2020 | Colorectal Cancer | Research article

A novel coordination complex of platinum (PT) induces cell death in colorectal cancer by altering redox balance and modulating MAPK pathway

Authors: Khayal Al-Khayal, Mansoor-Ali Vaali-Mohammed, Mohammed Elwatidy, Thamer Bin Traiki, Omar Al-Obeed, Mohammad Azam, Zahid Khan, Maha Abdulla, Rehan Ahmad

Published in: BMC Cancer | Issue 1/2020

Login to get access

Abstract

Background

Colorectal cancer (CRC) is a heterogeneous tumor having various genetic alterations. The current treatment options had limited impact on disease free survival due to therapeutic resistance. Novel anticancer agents are needed to treat CRC specifically metastatic colorectal cancer. A novel coordination complex of platinum, (salicylaldiminato)Pt(II) complex with dimethylpropylene linkage (PT) exhibited potential anti-cancer activity. In this study, we explored the molecular mechanism of PT-induced cell death in colorectal cancer.

Methods

Colony formation was evaluated using the clonogenic assay. Apoptosis, cell cycle analysis, reactive oxygen species, mitochondrial membrane potential and caspase-3/− 7 were assessed by flow cytometry. Glutathione level was detected by colorimetric assay. PT-induced alteration in pro-apoptotic/ anti-apoptotic proteins and other signaling pathways were investigated using western blotting. P38 downregulation was performed using siRNA.

Results

In the present study, we explored the molecular mechanism of PT-mediated inhibition of cell proliferation in colorectal cancer cells. PT significantly inhibited the colony formation in human colorectal cancer cell lines (HT-29, SW480 and SW620) by inducing apoptosis and necrosis. This platinum complex was shown to significantly increase the reactive oxygen species (ROS) generation, depletion of glutathione and reduced mitochondrial membrane potential in colorectal cancer cells. Exposure to PT resulted in the downregulation of anti-apoptotic proteins (Bcl2, BclxL, XIAP) and alteration in Cyclins expression. Furthermore, PT increased cytochrome c release into cytosol and enhanced PARP cleavage leading to activation of intrinsic apoptotic pathway. Moreover, pre-treatment with ROS scavenger N-acetylcysteine (NAC) attenuated apoptosis suggesting that PT-induced apoptosis was driven by oxidative stress. Additionally, we show that PT-induced apoptosis was mediated by activating p38 MAPK and inhibiting AKT pathways. This was demonstrated by using chemical inhibitor and siRNA against p38 kinase which blocked the cytochrome c release and apoptosis in colorectal cancer cells.

Conclusion

Collectively, our data demonstrates that the platinum complex (PT) exerts its anti-proliferative effect on CRC by ROS-mediated apoptosis and activating p38 MAPK pathway. Thus, our findings reveal a novel mechanism of action for PT on colorectal cancer cells and may have therapeutic implication.
Appendix
Available only for authorised users
Literature
3.
go back to reference Fleming M, Ravula S, Tatishchev SF, Wang HL. Colorectal carcinoma: pathologic aspects. J Gastrointest Oncol. 2012;3:153–73.PubMedPubMedCentral Fleming M, Ravula S, Tatishchev SF, Wang HL. Colorectal carcinoma: pathologic aspects. J Gastrointest Oncol. 2012;3:153–73.PubMedPubMedCentral
4.
go back to reference Inamura K. Colorectal cancers: an update on their molecular pathology. Cancers (Basel). 2018;10:26. Inamura K. Colorectal cancers: an update on their molecular pathology. Cancers (Basel). 2018;10:26.
5.
go back to reference Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology. 2010;138:2044–58.PubMed Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology. 2010;138:2044–58.PubMed
6.
go back to reference Graff RE, et al. Familial risk and heritability of colorectal cancer in the nordic twin study of cancer. Clin Gastroenterol Hepatol. 2017;15:1256–64.PubMedPubMedCentral Graff RE, et al. Familial risk and heritability of colorectal cancer in the nordic twin study of cancer. Clin Gastroenterol Hepatol. 2017;15:1256–64.PubMedPubMedCentral
8.
go back to reference Folkesson J, Martling A, Kodeda K. Current considerations in colorectal cancer surgery. Colorectal Cancer. 2015;4:167–74. Folkesson J, Martling A, Kodeda K. Current considerations in colorectal cancer surgery. Colorectal Cancer. 2015;4:167–74.
9.
go back to reference Gustavsson B, Carlsson G, Machover D, Petrelli N, Roth A, Schmoll HJ, Tveit KM, Gibson F. A review of the evolution of systemic chemotherapy in the management of colorectal cancer. Clin Colorectal Cancer. 2015;14:1–10.PubMed Gustavsson B, Carlsson G, Machover D, Petrelli N, Roth A, Schmoll HJ, Tveit KM, Gibson F. A review of the evolution of systemic chemotherapy in the management of colorectal cancer. Clin Colorectal Cancer. 2015;14:1–10.PubMed
10.
go back to reference Dagoglu N, Mahadevan A, Nedea E, Poylin V, Nagle D. Stereotactic body radiotherapy (SBRT) reirradiation for pelvic recurrence from colorectal cancer. J Surg Oncol. 2015;111:478–82.PubMed Dagoglu N, Mahadevan A, Nedea E, Poylin V, Nagle D. Stereotactic body radiotherapy (SBRT) reirradiation for pelvic recurrence from colorectal cancer. J Surg Oncol. 2015;111:478–82.PubMed
11.
go back to reference Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B, Starling N. Colorectal cancer. Lancet. 2010;375:1030–47.PubMed Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B, Starling N. Colorectal cancer. Lancet. 2010;375:1030–47.PubMed
12.
go back to reference Van der Jeught K, Xu H-C, Li Y-J, et al. Drug resistance and new therapies in colorectal cancer. World J Gastroenterol. 2018;24(34):3834–48.PubMedPubMedCentral Van der Jeught K, Xu H-C, Li Y-J, et al. Drug resistance and new therapies in colorectal cancer. World J Gastroenterol. 2018;24(34):3834–48.PubMedPubMedCentral
13.
go back to reference Salonga D, Danenberg KD, Johnson M, Metzger R, Groshen S, Tsao-Wei DD, Lenz HJ, Leichman CG, Leichman L, Diasio RB, et al. Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase. Clin Cancer Res. 2000;6:1322–7.PubMed Salonga D, Danenberg KD, Johnson M, Metzger R, Groshen S, Tsao-Wei DD, Lenz HJ, Leichman CG, Leichman L, Diasio RB, et al. Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase. Clin Cancer Res. 2000;6:1322–7.PubMed
14.
go back to reference Yaffee P, Osipov A, Tan C, Tuli R, Hendifar A. Review of systemic therapies for locally advanced and metastatic rectal cancer. J Gastrointest Oncol. 2015;6:185–200.PubMedPubMedCentral Yaffee P, Osipov A, Tan C, Tuli R, Hendifar A. Review of systemic therapies for locally advanced and metastatic rectal cancer. J Gastrointest Oncol. 2015;6:185–200.PubMedPubMedCentral
15.
go back to reference Siegel R, Desantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–17.PubMed Siegel R, Desantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–17.PubMed
16.
go back to reference Milczarek M, Rosinska S, Psurski M, Maciejewska M, Kutner A, Wietrzyk J. Combined colonic cancer treatment with vitamin D analogs and irinotecan or oxaliplatin. Anticancer Res. 2013;33(2):433–44.PubMed Milczarek M, Rosinska S, Psurski M, Maciejewska M, Kutner A, Wietrzyk J. Combined colonic cancer treatment with vitamin D analogs and irinotecan or oxaliplatin. Anticancer Res. 2013;33(2):433–44.PubMed
17.
go back to reference Imajo M, Tsuchiya Y, Nishida E. Regulatory mechanisms and functions of MAP kinase signaling pathways. IUBMB Life. 2006;58:312–7.PubMed Imajo M, Tsuchiya Y, Nishida E. Regulatory mechanisms and functions of MAP kinase signaling pathways. IUBMB Life. 2006;58:312–7.PubMed
18.
go back to reference Marshall MS. Ras target proteins in eukaryotic cells. FASEB J. 1995;9:1311–8.PubMed Marshall MS. Ras target proteins in eukaryotic cells. FASEB J. 1995;9:1311–8.PubMed
19.
go back to reference Qi M, Elion EA. MAP kinase pathways. J Cell Sci. 2005;118:3569–72.PubMed Qi M, Elion EA. MAP kinase pathways. J Cell Sci. 2005;118:3569–72.PubMed
20.
go back to reference Azam M, Al-Resayes SI, Soliman SM, Trzesowska-Kruszynska A, Kruszynski R, Khan Z. A (salicylaldiminato)PT(II) complex with dimethylpropylene linkage:synthesis, structural characterization and antineoplastic activity. J Photochem Photobiol B Biol. 2017;176:150–6. Azam M, Al-Resayes SI, Soliman SM, Trzesowska-Kruszynska A, Kruszynski R, Khan Z. A (salicylaldiminato)PT(II) complex with dimethylpropylene linkage:synthesis, structural characterization and antineoplastic activity. J Photochem Photobiol B Biol. 2017;176:150–6.
22.
go back to reference Ahmad R, Vaali-Mohammed M-A, Elwatidy M, et al. Induction of ROS-mediated cell death and activation of the JNK pathway by a sulfonamide derivate. Int J Mol Medicine. 2019;44(4):1552–6. Ahmad R, Vaali-Mohammed M-A, Elwatidy M, et al. Induction of ROS-mediated cell death and activation of the JNK pathway by a sulfonamide derivate. Int J Mol Medicine. 2019;44(4):1552–6.
23.
go back to reference Al-Khayal K, Alafeefy A, Vaali-Mohammed M-A, et al. Novel derivative of aminobenzenesulfonamide (3c) induces apoptosis in colorectal cancer cells through ROS generation and inhibits cell migration. BMC Cancer. 2017;17:4.PubMedPubMedCentral Al-Khayal K, Alafeefy A, Vaali-Mohammed M-A, et al. Novel derivative of aminobenzenesulfonamide (3c) induces apoptosis in colorectal cancer cells through ROS generation and inhibits cell migration. BMC Cancer. 2017;17:4.PubMedPubMedCentral
24.
go back to reference Buttke TM, Sandstrom PA. Oxidative stress as a mediator of apoptosis. Immunol Today. 1994;15:7–10.PubMed Buttke TM, Sandstrom PA. Oxidative stress as a mediator of apoptosis. Immunol Today. 1994;15:7–10.PubMed
25.
go back to reference Fleury C, Mignotte B, Vayssiere JL. Mitochondrial reactive oxygen species in cell death signaling. Biochimie. 2002;84:131–41.PubMed Fleury C, Mignotte B, Vayssiere JL. Mitochondrial reactive oxygen species in cell death signaling. Biochimie. 2002;84:131–41.PubMed
26.
go back to reference Anderson ME. Glutathione: an overview of bio-synthesis and modulation. Chem Biol Interact. 1998;111–2:1–14. Anderson ME. Glutathione: an overview of bio-synthesis and modulation. Chem Biol Interact. 1998;111–2:1–14.
27.
go back to reference Hammond CL, Lee TK, Ballatori N. Novel roles for glutathione in gene expression, cell death, and mem-brane transport of organic solutes. J Hepatol. 2001;34:946–54.PubMed Hammond CL, Lee TK, Ballatori N. Novel roles for glutathione in gene expression, cell death, and mem-brane transport of organic solutes. J Hepatol. 2001;34:946–54.PubMed
28.
go back to reference Chen X, Carystinos GD, Batist G. Potential for selective modulation of glutathione in cancer chemotherapy. Chem Biol Interact. 1998;111–2:263–75. Chen X, Carystinos GD, Batist G. Potential for selective modulation of glutathione in cancer chemotherapy. Chem Biol Interact. 1998;111–2:263–75.
29.
go back to reference Waterhouse NJ, Goldstein JC, Ahsen OV, et al. Cytochrome c maintains mitochondrial membrane potential and ATP generation after outer mitochondrial membrane permeabilization during apoptosis process. J Cell Biol. 2001;153:319–28.PubMedPubMedCentral Waterhouse NJ, Goldstein JC, Ahsen OV, et al. Cytochrome c maintains mitochondrial membrane potential and ATP generation after outer mitochondrial membrane permeabilization during apoptosis process. J Cell Biol. 2001;153:319–28.PubMedPubMedCentral
30.
go back to reference Shamas-Din A, Kale J, Leber B, Andrews DW. Mechanism of action of Bcl-2 family proteins. Cold Spring Harb Perspect Biol. 2013;5:a008714. Shamas-Din A, Kale J, Leber B, Andrews DW. Mechanism of action of Bcl-2 family proteins. Cold Spring Harb Perspect Biol. 2013;5:a008714.
31.
go back to reference Casimiro MC, Crosariol M, Loro E, Li Z, Pestell RG. Cyclins and cell cycle control in cancer and disease. Genes Cancer. 2012;3:649–57.PubMedPubMedCentral Casimiro MC, Crosariol M, Loro E, Li Z, Pestell RG. Cyclins and cell cycle control in cancer and disease. Genes Cancer. 2012;3:649–57.PubMedPubMedCentral
32.
go back to reference Soldani C, Scovassi AI. Poly(ADP-ribose) polymerase-1 cleavage during apoptosis: an update. Apoptosis. 2002;7:321–8.PubMed Soldani C, Scovassi AI. Poly(ADP-ribose) polymerase-1 cleavage during apoptosis: an update. Apoptosis. 2002;7:321–8.PubMed
34.
go back to reference Raj, et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature. 2011;475:231–4.PubMedPubMedCentral Raj, et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature. 2011;475:231–4.PubMedPubMedCentral
35.
go back to reference Zikaki K, Aggeli IK, Gaitanaki C, Beis I. Curcumin induces theapoPTotic intrinsic pathway via upregulation od reactive oxygen species and JNKs in H9c2 cardiac myoblasts. Apoptosis. 2014;19:958–74.PubMed Zikaki K, Aggeli IK, Gaitanaki C, Beis I. Curcumin induces theapoPTotic intrinsic pathway via upregulation od reactive oxygen species and JNKs in H9c2 cardiac myoblasts. Apoptosis. 2014;19:958–74.PubMed
36.
go back to reference Chen L, Jiang K, Chen H, et al. Deguelin induces apoptosis in colorectal cancer cancer cells by activating the p38 MAPK pathway. Cancer Manag Res. 2019;11:95–105.PubMed Chen L, Jiang K, Chen H, et al. Deguelin induces apoptosis in colorectal cancer cancer cells by activating the p38 MAPK pathway. Cancer Manag Res. 2019;11:95–105.PubMed
37.
go back to reference Widmann C, Gibson S, Jarpe MB, Johnson GL. Mitogen-activated protein kinase: conservation of athree-kinase module from yeast to human. Physiol Rev. 1999;79:143–80.PubMed Widmann C, Gibson S, Jarpe MB, Johnson GL. Mitogen-activated protein kinase: conservation of athree-kinase module from yeast to human. Physiol Rev. 1999;79:143–80.PubMed
38.
go back to reference Chang L, Karin M. Mammalian MAP kinase signal-ling cascades. Nature. 2001;410:37–40.PubMed Chang L, Karin M. Mammalian MAP kinase signal-ling cascades. Nature. 2001;410:37–40.PubMed
39.
go back to reference Yuan J, Rozengurt. PKD, PKD2 and p38 MAPK mediate Hsp27 Serine-82 phosphorylation induced ny neurotensin in Pancreatic Cancer PANC-1 cells. J Cell Biochem. 2008;103:648–62.PubMed Yuan J, Rozengurt. PKD, PKD2 and p38 MAPK mediate Hsp27 Serine-82 phosphorylation induced ny neurotensin in Pancreatic Cancer PANC-1 cells. J Cell Biochem. 2008;103:648–62.PubMed
40.
go back to reference Rosenberg B, VanCamp L, Trosko JE, Mansour VH. Platinum compounds: a new class of potent antitumor agents. Nature. 1969;222:385–6.PubMed Rosenberg B, VanCamp L, Trosko JE, Mansour VH. Platinum compounds: a new class of potent antitumor agents. Nature. 1969;222:385–6.PubMed
41.
go back to reference Weiss RB, Christian MC. New cispalatin analogues in development. A review. Drugs. 1993;46:360–77.PubMed Weiss RB, Christian MC. New cispalatin analogues in development. A review. Drugs. 1993;46:360–77.PubMed
42.
go back to reference Gasser G, Ott I, Metzler-Nolte N. Organometallic anticancer compounds. J Med Chem. 2011;54:3–25.PubMed Gasser G, Ott I, Metzler-Nolte N. Organometallic anticancer compounds. J Med Chem. 2011;54:3–25.PubMed
43.
go back to reference Stoehlmacher J. Prediction of efficacy and side effects of chemotherapy in colorectal cancer. Recent Results Cancer Res. 2007;176:81–8.PubMed Stoehlmacher J. Prediction of efficacy and side effects of chemotherapy in colorectal cancer. Recent Results Cancer Res. 2007;176:81–8.PubMed
44.
go back to reference Sava G, Bergamo A, Duson PJ. Metal-based antitumor drugs in the post-genomic era: what comes next? Dalton Trans. 2011;40:9069–75.PubMed Sava G, Bergamo A, Duson PJ. Metal-based antitumor drugs in the post-genomic era: what comes next? Dalton Trans. 2011;40:9069–75.PubMed
45.
go back to reference Lazarevic T, Rilak A, Bugarcic ZD. Platinum, palladium, gold and ruthenium complexes as antitumor agents: current clinical uses, cytotoxicity studies and future perspectives. Euro J Med Chem. 2017;142:8–31. Lazarevic T, Rilak A, Bugarcic ZD. Platinum, palladium, gold and ruthenium complexes as antitumor agents: current clinical uses, cytotoxicity studies and future perspectives. Euro J Med Chem. 2017;142:8–31.
46.
go back to reference Silva TM, Andersson S, Sukumaran SK, et al. Norspermidine and novel Pd(II) and Pt(II) polynuclear complexes of norspermidine as potential antineoplastic agents against breast cancer. PLoS One. 2013;8:e55651.PubMedPubMedCentral Silva TM, Andersson S, Sukumaran SK, et al. Norspermidine and novel Pd(II) and Pt(II) polynuclear complexes of norspermidine as potential antineoplastic agents against breast cancer. PLoS One. 2013;8:e55651.PubMedPubMedCentral
47.
go back to reference Bostancioglu RB, Isik K, Genc H, et al. Studies on the cytotoxic, apoptotic and antitumoral effect of au (III) and Pt (II) complexes of 1, 10-phenanthroline on V79379A and A549 cell lines. J Enz Inhib Med Chem. 2011:1–9. Bostancioglu RB, Isik K, Genc H, et al. Studies on the cytotoxic, apoptotic and antitumoral effect of au (III) and Pt (II) complexes of 1, 10-phenanthroline on V79379A and A549 cell lines. J Enz Inhib Med Chem. 2011:1–9.
49.
go back to reference Tolan D, Gandin V, Morrison L, et al. Oxidative Stress Induced by Pt(IV) Pro-drugs Based on the Cisplatin Scaffold and Indole Carboxylic Acids in Axial Position. Sci Reports. 2016;6:29367. Tolan D, Gandin V, Morrison L, et al. Oxidative Stress Induced by Pt(IV) Pro-drugs Based on the Cisplatin Scaffold and Indole Carboxylic Acids in Axial Position. Sci Reports. 2016;6:29367.
52.
go back to reference Wilmanski T, Zhou X, Zheng W, et al. Inhibition of pyruvate carboxylase by 1α,25-dihydroxyvitamin D promotes oxidative stress in early breast cancer progression. Cancer Lett. 2017;411:171–81.PubMedPubMedCentral Wilmanski T, Zhou X, Zheng W, et al. Inhibition of pyruvate carboxylase by 1α,25-dihydroxyvitamin D promotes oxidative stress in early breast cancer progression. Cancer Lett. 2017;411:171–81.PubMedPubMedCentral
53.
go back to reference Wilmanski T, Zhou X, Zheng W et al. Inhibition of pyruvate carboxylase by 1α,25-dihydroxyvitamin D promotes oxidative stress in early breast cancer progression. Cancer Letters 2017, 411, 171-181. Wilmanski T, Zhou X, Zheng W et al. Inhibition of pyruvate carboxylase by 1α,25-dihydroxyvitamin D promotes oxidative stress in early breast cancer progression. Cancer Letters 2017, 411, 171-181.
54.
go back to reference Rahman F-U, Ali A, Duong H-Q, et al. ONS-donor ligand based Pt(II) complexes display extremely high anticancer potency through autophagic cell death pathway. Eur J Med Chem. 2019;164:546–61.PubMed Rahman F-U, Ali A, Duong H-Q, et al. ONS-donor ligand based Pt(II) complexes display extremely high anticancer potency through autophagic cell death pathway. Eur J Med Chem. 2019;164:546–61.PubMed
55.
go back to reference Kowalski S, Wyrzykowski D, Hac S, et al. New oxidovanadium(IV) coordination complex containg 2-methylnitrilotriacetate ligands induces cell cycle arrest and autophagy in human pancreatic ductal adenocarcinoma cell lines. In J Mol Sci. 2019;20:261. Kowalski S, Wyrzykowski D, Hac S, et al. New oxidovanadium(IV) coordination complex containg 2-methylnitrilotriacetate ligands induces cell cycle arrest and autophagy in human pancreatic ductal adenocarcinoma cell lines. In J Mol Sci. 2019;20:261.
56.
go back to reference Silva SLR, Bliza IRS, Dias RB, et al. Ru(II)-thymine complex causes DNA damage and apoptotic cell death in human colon carcinoma HCT116 cells mediated by JNK/p38/ERK1/2 via a p53-independent signaling. Sci Rep. 2019;9:11094.PubMedPubMedCentral Silva SLR, Bliza IRS, Dias RB, et al. Ru(II)-thymine complex causes DNA damage and apoptotic cell death in human colon carcinoma HCT116 cells mediated by JNK/p38/ERK1/2 via a p53-independent signaling. Sci Rep. 2019;9:11094.PubMedPubMedCentral
57.
go back to reference Taniguchi K, Karin M. NFkB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18:309–24.PubMed Taniguchi K, Karin M. NFkB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18:309–24.PubMed
58.
go back to reference Sethi G, Ahn KS, Sung B, Aggarwal BB. Pinistol targets nuclear factor-kappaB activation pathway leading to inhibition of gene products associated with proliferation, apoptosis, invasion and angiogenesis. Mol Cancer Ther. 2008;7:1604–14.PubMed Sethi G, Ahn KS, Sung B, Aggarwal BB. Pinistol targets nuclear factor-kappaB activation pathway leading to inhibition of gene products associated with proliferation, apoptosis, invasion and angiogenesis. Mol Cancer Ther. 2008;7:1604–14.PubMed
59.
go back to reference Wong ALA, Hirpara JL, Pervaiz S, Eu J-Q, Sethi G, Goh B-C. Do STAT3 inhibitors have potential in the future for cancer therapy. Expert Opin Investig Drugs. 2017;26:883–7.PubMed Wong ALA, Hirpara JL, Pervaiz S, Eu J-Q, Sethi G, Goh B-C. Do STAT3 inhibitors have potential in the future for cancer therapy. Expert Opin Investig Drugs. 2017;26:883–7.PubMed
60.
go back to reference Zhu Y, Zhang M, Luo L, et al. NFkB hijacking theranostic Pt(II) complex in cancer therapy. Theranostics. 2019;9:2158–66.PubMedPubMedCentral Zhu Y, Zhang M, Luo L, et al. NFkB hijacking theranostic Pt(II) complex in cancer therapy. Theranostics. 2019;9:2158–66.PubMedPubMedCentral
Metadata
Title
A novel coordination complex of platinum (PT) induces cell death in colorectal cancer by altering redox balance and modulating MAPK pathway
Authors
Khayal Al-Khayal
Mansoor-Ali Vaali-Mohammed
Mohammed Elwatidy
Thamer Bin Traiki
Omar Al-Obeed
Mohammad Azam
Zahid Khan
Maha Abdulla
Rehan Ahmad
Publication date
01-12-2020
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2020
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-07165-w

Other articles of this Issue 1/2020

BMC Cancer 1/2020 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