Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Research Article

Anti-Cancer Effect of Melatonin via Downregulation of Delta-like Ligand 4 in Estrogen-Responsive Breast Cancer Cells

Author(s): Ali Rajabi, Ali Saber, Mahsa Pourmahdi, Ali Emami, Reyhaneh Ravanbakhsh, Amir Khodavirdipour, Mehran Khodaei, Molood Akbarzadeh, Sepehr Abdolahi, Mohammad Ali Hosseinpourfeizi and Reza Safaralizadeh*

Volume 15, Issue 4, 2020

Page: [329 - 340] Pages: 12

DOI: 10.2174/1574892815666200929145236

Price: $65

Abstract

Background: The Notch signaling pathway has a key role in angiogenesis and Delta - Like Ligand 4 (DLL4) is one of the main ligands of Notch involved in cell proliferation in sprouting vessels.

Objective: In this study, we aimed to evaluate the expression of DLL4 in primary breast tumors and to examine the effect of melatonin on DLL4 expression in vitro.

Methods: Eighty-five breast tumor and paired adjacent non-tumor tissue samples were collected. Apoptosis assay was performed on breast cancer cells to evaluate melatonin effects. Western blot and quantitative RT-PCR were used to measure DLL4 expression. Then, we investigated the effect of melatonin on the expression of DLL4 in four breast cancer cell lines at RNA and protein levels. We also performed a probabilistic neural network analysis to study genes closely associated with DLL4 expression.

Results: Our results showed a significantly higher expression of DLL4 in tumor tissues compared to non-tumor tissues (P = 0.027). Melatonin treatment substantially attenuated DLL4 expression in BT474 and MCF-7 cells, but not in SK-BR-3 and MDA-MB-231 cells. Also, melatonin induced apoptosis in all four cell lines. Network analysis revealed a set of 15 genes that had close association and interaction with DLL4. DLL4 was overexpressed in breast cancer tissues as compared to the non-tumor tissues.

Conclusion: It can be concluded that melatonin treatment attenuated DLL4 expression only in estrogen- responsive breast cancer cells and is able to induce apoptosis in breast cancer cells.

Keywords: Angiogenesis, breast cancer, DLL4, estrogen-responsive, melatonin, tumor cell lines.

[1]
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394-424.
[http://dx.doi.org/10.3322/caac.21492] [PMID: 30207593]
[2]
Pouladi N, Abdolahi S, Farajzadeh D, Feizi MAHP. Association of the 17p13.1 region gene variants rs1042522 and rs2287499 with risk of breast cancer in Iranian-Azeri population. Meta Gene 2019; 19: 117-22.
[3]
Reigosa A, Hardisson D, Sanzi F, Caleiras E, Saldivia F, Fernández A. Subclassification of the molecular types of breast cancer based on the expression of immunohistochemical markers and evolution. Invest Clin 2016; 57(2): 187-216.
[PMID: 28429898]
[4]
Mavaddat N, Michailidou K, Dennis J, et al. ABCTB Investigators; kConFab/AOCS Investigators; NBCS Collaborators. Polygenic risk scores for prediction of breast cancer and breast cancer subtypes. Am J Hum Genet 2019; 104(1): 21-34.
[http://dx.doi.org/10.1016/j.ajhg.2018.11.002] [PMID: 30554720]
[5]
Ghanbari M, Hosseinpour-Feizi M, Safaralizadeh R, Aghazadeh A, Montazeri V. Study of KMT2B (MLL2) gene expression changes in patients with breast cancer. Breast Cancer Manag 2019; 8(2): BMT24.
[http://dx.doi.org/10.2217/bmt-2018-0016]
[6]
Dastmalchi N, Safaralizadeh R, Baradaran B, Hosseinpourfeizi M, Baghbanzadeh A. An update review of deregulated tumor suppressive microRNAs and their contribution in various molecular subtypes of breast cancer. Gene 2020; 729: 144301.
[http://dx.doi.org/10.1016/j.gene.2019.144301] [PMID: 31884105]
[7]
Morse MA, Sun W, Kim R, et al. The role of angiogenesis in hepatocellular carcinoma. Clin Cancer Res 2019; 25(3): 912-20.
[http://dx.doi.org/10.1158/1078-0432.CCR-18-1254] [PMID: 30274981]
[8]
Uzzan B, Nicolas P, Cucherat M, Perret G-Y. Microvessel density as a prognostic factor in women with breast cancer: A systematic review of the literature and meta-analysis. Cancer Res 2004; 64(9): 2941-55.
[http://dx.doi.org/10.1158/0008-5472.CAN-03-1957] [PMID: 15126324]
[9]
Nickoloff BJ, Osborne BA, Miele L. Notch signaling as a therapeutic target in cancer: A new approach to the development of cell fate modifying agents. Oncogene 2003; 22(42): 6598-608.
[http://dx.doi.org/10.1038/sj.onc.1206758] [PMID: 14528285]
[10]
Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M. The Notch target genes Hey1 and Hey2 are required for embryonic vascular development. Genes Dev 2004; 18(8): 901-11.
[http://dx.doi.org/10.1101/gad.291004] [PMID: 15107403]
[11]
Gridley T. Chapter Nine - Notch Signaling in the Vasculature. Current Topics in Developmental Biology 92. Academic Press 2010; pp. 277-309.
[12]
Hayashi H, Kume T. Foxc transcription factors directly regulate Dll4 and Hey2 expression by interacting with the VEGF-Notch signaling pathways in endothelial cells. PLoS One 2008; 3(6): e2401.
[http://dx.doi.org/10.1371/journal.pone.0002401] [PMID: 18545664]
[13]
Hayashi H, Kume T. Foxc2 transcription factor as a regulator of angiogenesis via induction of integrin β3 expression. Cell Adhes Migr 2009; 3(1): 24-6.
[http://dx.doi.org/10.4161/cam.3.1.7252] [PMID: 19372730]
[14]
Reedijk M, Odorcic S, Chang L, et al. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res 2005; 65(18): 8530-7.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-1069] [PMID: 16166334]
[15]
Kushwaha PP, Vardhan PS, Kapewangolo P, et al. Bulbine frutescens phytochemical inhibits notch signaling pathway and induces apoptosis in triple negative and luminal breast cancer cells. Life Sci 2019; 234: 116783.
[http://dx.doi.org/10.1016/j.lfs.2019.116783] [PMID: 31442552]
[16]
Das A, Narayanam MK, Paul S, et al. A novel triazole, NMK-T-057, induces autophagic cell death in breast cancer cells by inhibiting γ-secretase-mediated activation of Notch signaling. J Biol Chem 2019; 294(17): 6733-50.
[http://dx.doi.org/10.1074/jbc.RA119.007671] [PMID: 30824542]
[17]
Shutter JR, Scully S, Fan W, et al. Dll4, a novel Notch ligand expressed in arterial endothelium. Genes Dev 2000; 14(11): 1313-8.
[PMID: 10837024]
[18]
Mendonça L, Trindade A, Carvalho C, et al. Metastasis is impaired by endothelial-specific Dll4 loss-of-function through inhibition of epithelial-to-mesenchymal transition and reduction of cancer stem cells and circulating tumor cells. Clin Exp Metastasis 2019; 36(4): 365-80.
[http://dx.doi.org/10.1007/s10585-019-09973-2] [PMID: 31119445]
[19]
Pitulescu ME, Schmidt I, Giaimo BD, et al. Dll4 and Notch signalling couples sprouting angiogenesis and artery formation. Nat Cell Biol 2017; 19(8): 915-27.
[http://dx.doi.org/10.1038/ncb3555] [PMID: 28714968]
[20]
Jia X, Wang W, Xu Z, et al. A humanized anti-DLL4 antibody promotes dysfunctional angiogenesis and inhibits breast tumor growth. Sci Rep 2016; 6: 27985.
[http://dx.doi.org/10.1038/srep27985] [PMID: 27301650]
[21]
Xia S, Menden HL, Korfhagen TR, Kume T, Sampath V. Endothelial immune activation programmes cell-fate decisions and angiogenesis by inducing angiogenesis regulator DLL4 through TLR4-ERK-FOXC2 signalling. J Physiol 2018; 596(8): 1397-417.
[http://dx.doi.org/10.1113/JP275453] [PMID: 29380370]
[22]
Herman AM, Rhyner AM, Devine WP, Marrelli SP, Bruneau BG, Wythe JD. A novel reporter allele for monitoring DLL4 expression within the embryonic and adult mouse. Biol Open 2018; 7(3): bio026799.
[http://dx.doi.org/10.1242/bio.026799] [PMID: 29437553]
[23]
Ting H-A, de Almeida Nagata D, Rasky AJ, et al. Notch ligand Delta-like 4 induces epigenetic regulation of Treg cell differentiation and function in viral infection. Mucosal Immunol 2018; 11(5): 1524-36.
[http://dx.doi.org/10.1038/s41385-018-0052-1] [PMID: 30038214]
[24]
Low S, Barnes JL, Zammit PS, Beauchamp JR. Delta-like 4 activates notch 3 to regulate self-renewal in skeletal muscle stem cells. Stem Cells 2018; 36(3): 458-66.
[http://dx.doi.org/10.1002/stem.2757] [PMID: 29230914]
[25]
Choi H-J, Rho S-S, Choi D-H, Kwon Y-G. LDB2 regulates the expression of DLL4 through the formation of oligomeric complexes in endothelial cells. BMB Rep 2018; 51(1): 21-6.
[http://dx.doi.org/10.5483/BMBRep.2018.51.1.140] [PMID: 28946938]
[26]
Gu W, Monteiro R, Zuo J, et al. A novel TGFβ modulator that uncouples R-Smad/I-Smad-mediated negative feedback from R-Smad/ligand-driven positive feedback. PLoS Biol 2015; 13(2): e1002051.
[http://dx.doi.org/10.1371/journal.pbio.1002051] [PMID: 25665164]
[27]
Chen H-T, Cai Q-C, Zheng J-M, et al. High expression of delta -like ligand 4 predicts poor prognosis after curative resection for pancreatic cancer. Ann Surg Oncol 2012; 19(3)(Suppl. 3): S464-74.
[http://dx.doi.org/10.1245/s10434-011-1968-9] [PMID: 21822553]
[28]
Fernando S, Rombauts L. Melatonin: Shedding light on infertility?-A review of the recent literature. J Ovarian Res 2014; 7: 98.
[http://dx.doi.org/10.1186/s13048-014-0098-y] [PMID: 25330986]
[29]
Zamfir Chiru AA, Popescu CR, Gheorghe DC. Melatonin and cancer. J Med Life 2014; 7(3): 373-4.
[PMID: 25408757]
[30]
Lissoni P, Rovelli F, Malugani F, Bucovec R, Conti A, Maestroni GJ. Anti-angiogenic activity of melatonin in advanced cancer patients. Neuroendocrinol Lett 2001; 22(1): 45-7.
[PMID: 11335879]
[31]
Mpindi JP, Kallioniemi O, Screpanti I, Poellinger L, Sahlgren C, Lendahl U. Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKa/IKKb. Oncogene 2012; 1: 11.
[32]
Han F, Liu W, Jiang X, et al. SOX30, a novel epigenetic silenced tumor suppressor, promotes tumor cell apoptosis by transcriptional activating p53 in lung cancer. Oncogene 2015; 34(33): 4391-402.
[http://dx.doi.org/10.1038/onc.2014.370] [PMID: 25435374]
[33]
Akiyama I, Yoshino O, Osuga Y, et al. The localization and regulation of Proprotein Convertase Subtilisin/Kexin (PCSK) 6 in human ovary. Am J Reprod Immunol 2012; 68(6): 491-8.
[http://dx.doi.org/10.1111/aji.12003] [PMID: 22935039]
[34]
Ueda T, Furusawa T, Kurahashi T, Tessarollo L, Bustin M. The nucleosome binding protein HMGN3 modulates the transcription profile of pancreatic β cells and affects insulin secretion. Mol Cell Biol 2009; 29(19): 5264-76.
[http://dx.doi.org/10.1128/MCB.00526-09] [PMID: 19651901]
[35]
Harding HP, Lackey JG, Hsu H-C, et al. An intact unfolded protein response in Trpt1 knockout mice reveals phylogenic divergence in pathways for RNA ligation. RNA 2008; 14(2): 225-32.
[http://dx.doi.org/10.1261/rna.859908] [PMID: 18094117]
[36]
Gurney A, Hoey T. Anti-DLL4, a cancer therapeutic with multiple mechanisms of action. Vasc Cell 2011; 3(1): 18.
[http://dx.doi.org/10.1186/2045-824X-3-18] [PMID: 21831306]
[37]
López-Guerra M, Xargay-Torrent S, Fuentes P, Roldán J, González-Farré B, Rosich L, et al. Specific NOTCH1 antibody targets DLL4-induced proliferation, migration, and angiogenesis in NOTCH1-mutated CLL cells. Oncogene 2020; 39(6): 1185-97.
[PMID: 31616059]
[38]
Huang SF, Yang ZL, Li DQ, et al. Jagged1 and DLL4 expressions in benign and malignant pancreatic lesions and their clinicopathological significance. HBPD INT 2016; 15(6): 640-6.
[http://dx.doi.org/10.1016/S1499-3872(16)60110-8] [PMID: 27919854]
[39]
Leach A, Smyth P, Ferguson L, et al. Anti-DLL4 VNAR targeted nanoparticles for targeting of both tumour and tumour associated vasculature. Nanoscale 2020; 12(27): 14751-63.
[http://dx.doi.org/10.1039/D0NR02962A] [PMID: 32626858]
[40]
Kontomanolis E, Panteliadou M, Giatromanolaki A, et al. Delta -like ligand 4 (DLL4) in the plasma and neoplastic tissues from breast cancer patients: Correlation with metastasis. Med Oncol 2014; 31(5): 945.
[http://dx.doi.org/10.1007/s12032-014-0945-0] [PMID: 24696220]
[41]
Leon-Blanco MM, Guerrero JM, Reiter RJ, Calvo JR, Pozo D. Melatonin inhibits telomerase activity in the MCF-7 tumor cell line both in vivo and in vitro. J Pineal Res 2003; 35(3): 204-11.
[http://dx.doi.org/10.1034/j.1600-079X.2003.00077.x] [PMID: 12932205]
[42]
Li Y, Li S, Zhou Y, et al. Melatonin for the prevention and treatment of cancer. Oncotarget 2017; 8(24): 39896-921.
[http://dx.doi.org/10.18632/oncotarget.16379] [PMID: 28415828]
[43]
Ma Q, Reiter RJ, Chen Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis 2020; 23(2): 91-104.
[http://dx.doi.org/10.1007/s10456-019-09689-7] [PMID: 31650428]
[44]
Ozcan-Sezer S, Ince E, Akdemir A, Ceylan ÖÖ, Suzen S, Gurer-Orhan H. Aromatase inhibition by 2-methyl indole hydrazone derivatives evaluated via molecular docking and in vitro activity studies. Xenobiotica 2019; 49(5): 549-56.
[http://dx.doi.org/10.1080/00498254.2018.1482029] [PMID: 29804490]
[45]
Mirza-Aghazadeh-Attari M, Reiter RJ, Rikhtegar R, et al. Melatonin: An atypical hormone with major functions in the regulation of angiogenesis. IUBMB Life 2020; 72(8): 1560-84.
[http://dx.doi.org/10.1002/iub.2287] [PMID: 32329956]
[46]
Hill SM, Spriggs LL, Simon MA, Muraoka H, Blask DE. The growth inhibitory action of melatonin on human breast cancer cells is linked to the estrogen response system. Cancer Lett 1992; 64(3): 249-56.
[http://dx.doi.org/10.1016/0304-3835(92)90050-6] [PMID: 1638517]
[47]
Hasan M, Browne E, Guarinoni L, Darveau T, Hilton K, Witt-Enderby PA. Anti-cancer effects of melatonin, estrogen, and progesterone hormone therapy in MCF-7 and MDA-MB-231 breast cancer cells. FASEB J 2020; 34(S1): 1.
[PMID: 32297350]
[48]
Buzzell GR. Studies on the effects of the pineal hormone melatonin on an androgen-insensitive rat prostatic adenocarcinoma, the Dunning R 3327 HIF tumor. J Neural Transm (Vienna) 1988; 72(2): 131-40.
[http://dx.doi.org/10.1007/BF01250236] [PMID: 3385424]
[49]
Sonehara NM, Lacerda JZ, Jardim-Perassi BV, et al. Melatonin regulates tumor aggressiveness under acidosis condition in breast cancer cell lines. Oncol Lett 2019; 17(2): 1635-45.
[PMID: 30675223]
[50]
Proietti S, Cucina A, Dobrowolny G, et al. Melatonin down-regulates MDM2 gene expression and enhances p53 acetylation in MCF-7 cells. J Pineal Res 2014; 57(1): 120-9.
[http://dx.doi.org/10.1111/jpi.12150] [PMID: 24920214]
[51]
Mediavilla MD, Cos S, Sánchez-Barceló EJ. Melatonin increases p53 and p21WAF1 expression in MCF-7 human breast cancer cells in vitro. Life Sci 1999; 65(4): 415-20.
[http://dx.doi.org/10.1016/S0024-3205(99)00262-3] [PMID: 10421427]
[52]
Xiang S, Dong C, Yuan L, Dauchy RT, Blask DE, Frasch T, et al. Retinoic acid-related orphan receptor alpha 1 (RORα1) induction of AKR1C3 promotes MCF-7 breast cancer cell proliferation and tamoxifen-resistance which is suppressed by melatonin. Melatonin Research 2020; 3(1): 81-100.
[http://dx.doi.org/10.32794/mr11250050]
[53]
Han F, Jiang X, Li ZM, et al. Epigenetic inactivation of SOX30 is associated with male infertility and offers a therapy target for non-obstructive azoospermia. Mol Ther Nucleic Acids 2020; 19: 72-83.
[http://dx.doi.org/10.1016/j.omtn.2019.10.038] [PMID: 31835093]
[54]
Han F, Liu WB, Shi XY, et al. SOX30 inhibits tumor metastasis through attenuating Wnt-signaling via transcriptional and posttranslational regulation of β-catenin in lung cancer. EBioMedicine 2018; 31: 253-66.
[http://dx.doi.org/10.1016/j.ebiom.2018.04.026] [PMID: 29739711]
[55]
Hao X, Han F, Ma B, et al. SOX30 is a key regulator of desmosomal gene suppressing tumor growth and metastasis in lung adenocarcinoma. J Exp Clin Cancer Res 2018; 37(1): 111.
[http://dx.doi.org/10.1186/s13046-018-0778-3] [PMID: 29855376]
[56]
Liu Y, Wang W, Li Y, Huang Y. SOX30 confers a tumor suppressive effect in acute myeloid leukemia through inactivation of Wnt/β-catenin signaling. Mol Cell Probes 2020; 52: 101578.
[http://dx.doi.org/10.1016/j.mcp.2020.101578] [PMID: 32334007]
[57]
Fu Q, Sun Z, Yang F, Mao T, Gao Y, Wang H. SOX30, a target gene of miR-653-5p, represses the proliferation and invasion of prostate cancer cells through inhibition of Wnt/β-catenin signaling. Cell Mol Biol Lett 2019; 24(1): 71.
[http://dx.doi.org/10.1186/s11658-019-0195-4] [PMID: 31889959]
[58]
Li N, Wang Y, Forbes K, et al. Metalloproteases regulate T-cell proliferation and effector function via LAG-3. EMBO J 2007; 26(2): 494-504.
[http://dx.doi.org/10.1038/sj.emboj.7601520] [PMID: 17245433]
[59]
Angelopoulou E, Paudel YN, Villa C, Shaikh MF, Piperi C. Lymphocyte-Activation Gene 3 (LAG3) protein as a possible therapeutic Target for Parkinson’s Disease: Molecular mechanisms connecting neuroinflammation to α-synuclein spreading pathology. Biology (Basel) 2020; 9(4): 86.
[http://dx.doi.org/10.3390/biology9040086] [PMID: 32340360]
[60]
Meurette O. Shaping of the Tumor Microenvironment by Notch Signaling Tumor Microenvironment. Springer 2020; pp. 1-16.
[61]
Mok SC, Kwok TT, Berkowitz RS, Barrett AJ, Tsui FWL. Overexpression of the Protein Tyrosine Phosphatase, Nonreceptor type 6 (PTPN6), in human epithelial ovarian cancer. Gynecol Oncol 1995; 57(3): 299-303.
[http://dx.doi.org/10.1006/gyno.1995.1146] [PMID: 7774833]
[62]
Liu H, Liu Z, Wang Y, et al. Transdisciplinary Research in Cancer of the Lung (TRICL) Research Team. Functional variants in DCAF4 associated with lung cancer risk in European populations. Carcinogenesis 2017; 38(5): 541-51.
[http://dx.doi.org/10.1093/carcin/bgx033] [PMID: 28383684]
[63]
Gorbatenko A, Olesen CW, Boedtkjer E, Pedersen SF. Regulation and roles of bicarbonate transporters in cancer. Front Physiol 2014; 5: 130.
[http://dx.doi.org/10.3389/fphys.2014.00130] [PMID: 24795638]
[64]
Jansen RL, Hupperets PS, Arends JW, et al. MIB-1 labelling index is an independent prognostic marker in primary breast cancer. Br J Cancer 1998; 78(4): 460-5.
[http://dx.doi.org/10.1038/bjc.1998.515] [PMID: 9716027]
[65]
Savant S, La Porta S, Budnik A, et al. The orphan receptor Tie1 controls angiogenesis and vascular remodeling by differentially regulating Tie2 in tip and stalk cells. Cell Rep 2015; 12(11): 1761-73.
[http://dx.doi.org/10.1016/j.celrep.2015.08.024] [PMID: 26344773]
[66]
Tao J, Lin J, Nie X, Huang W, Guo J, Liu T. Thymopentin promotes ovarian angiogenesis in mice by activating N6-methyladenosine (m6A) RNA modification of key factors in the Notch/Tie1 pathway. Vascular Investigation and Therapy 2019; 2(3): 63.
[http://dx.doi.org/10.4103/VIT.VIT_17_19]
[67]
Machado MJC, Boardman R, Riu F, Emanueli C, Benest AV, Bates DO. Enhanced notch signaling modulates unproductive revascularization in response to nitric oxide-angiopoietin signaling in a mouse model of peripheral ischemia. Microcirculation 2019; 26(6): e12549.
[http://dx.doi.org/10.1111/micc.12549] [PMID: 30974486]
[68]
Witt-Enderby PA, Davis VL, Lapinsky D. Anti-cancer tamoxifen-melatonin hybrid ligand. US8785501, 2010.
[69]
Witt-Enderby PA, Davis VL. Combination Hormone Replacement Therapy (HRT) and melatonin to prevent and treat mammary cancer. US8618083, 2013.
[70]
Kruisinga RJHD. Use of melatonin in the manufacture of a medicament for treating cancer. EP1656939, 2004.
[71]
Alpert MC, Carome EF, Hansler RL, Kubulnis VE, Spayer JL. Light emitting diode lamp free of melatonin-suppressing radiation. WO2009029575, 2008.
[72]
López JL, Ruíz JC, Castelar JS, Ruíz SMJ. Melatonin analogues for treating cancer. WO2017005954, 2016.
[73]
Zeicher M. Multi-ingredient pharmaceutical composition for use in cancer therapy. WO2016059247, 2015.
[74]
Uttenthal LO. Melatonin for preventing and treating radiation vaginitis and proctitis. EP3265070, 2018.
[75]
Raje S, Barthwal R. Molecular recognition of 3+1 hybrid human telomeric G-quadruplex DNA d-[AGGG(TTAGGG)3] by anticancer drugs epirubicin and adriamycin leads to thermal stabilization. Int J Biol Macromol 2019; 139: 1272-87.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.08.109] [PMID: 3142 1170]
[76]
Lee E, Jeon H, Lee M, et al. Molecular origin of AuNPs-induced cytotoxicity and mechanistic study. Sci Rep 2019; 9(1): 2494.
[http://dx.doi.org/10.1038/s41598-019-39579-3] [PMID: 30792478]
[77]
Tewari D, Rawat P, Singh PK. Adverse drug reactions of anticancer drugs derived from natural sources. Food Chem Toxicol 2019; 123: 522-35.
[http://dx.doi.org/10.1016/j.fct.2018.11.041] [PMID: 30471312]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy