Azithromycin as Potent Inhibitor of Cell Migration in Tumor Cell Line

Shumaila Usman, Kauser Ismail, Syed Saud Hasan, Mohsin Turab, Almas Jabeen, Dabeeran Zehra, Nabila Rasheed

Abstract

Continuous obstacles behind the discovery of novel drugs for cancer therapy have necessitated the development of alternative strategy of drug repurposing—the development of old drugs for new therapeutic purposes. With an improved understanding of the hallmarks of cancer, this strategy offers a cost-effective process for the treatment of human neoplastic disease, thereby facilitating rapid clinical translation. In this regard, macrolide antibiotics (MAs), which include a wide spectrum of activities against Gram-positive bacteria, have also been proposed as anticancer drugs for multiple tumor types. Over the past few years, significant progress has been achieved in anticancer therapy, but development of resistance and unavoidable side effects have weakened these attainments. Considering this severe condition, a number of drugs with novel antitumor mechanisms are under investigations including antimicrobials that have been shown to possess anti-inflammatory, immunomodulatory, and cytotoxic effects. In this regard, both conventional and novel antimicrobials are being studied to explore their anticancer potential along with underlying mechanisms that may render them as effective anticancer drugs in the future. Hence, in the latest study, we tested the role of a macrolide antibiotic drug, Azithromycin (AZM) alone, in combination with standard chemotherapeutic agent Sorafenib (Sorafenib/AZM) and its gold conjugated nanoparticles (AuAZM) as an anti angiogenic agent in hepatoma cell line hepG2 through wound healing assay. The migratory potential of HepG2 cells after being exposed to different treatments (AZM, Sorafenib, Sorafenib/AZM, and Au-AZM at IC50 concentrations) was observed at 0, 6, 24, 48, and 72 hours. The results of our study showed that AZM exhibited highly significant reduction in wound healing with p-value (< 0.001) up till 72 hours, while Sorafenib, Sorafenib/AZM, and Au-AZM inhibited wound healing up to 48 hours (p-value < 0.001). The current study revealed a comparatively higher antiangiogenic potential of AZM in cancer cells, thereby suggesting its clinical application for cancer treatment.  

 

Keywords: azithromycin, wound healing potential, tumor cell line, cell migration.

 

https://doi.org/10.55463/issn.1674-2974.49.6.5


Full Text:

PDF


References


CHA Y., EREZ T., REYNOLDS I.J., KUMAR D., ROSS J., KOYTIGER G., KUSKO R., ZESKIND B., RISSO S., KAGAN E., PAPAPETROPOULOS S., GROSSMAN I., and LAIFENFELD D. Drug repurposing from the perspective of pharmaceutical companies. British Journal of Pharmacology, 2018, 175(2): 168-180. https://doi.org/10.1111/bph.13798

PAPAPETROPOULOS A., & SZABO C. Inventing new therapies without reinventing the wheel: the power of drug repurposing. British Journal of Pharmacology, 2018, 175(2): 165-167. https://doi.org/10.1111/bph.14081

PUSHPAKOM S., IORIO F., EYERS P. A., ESCOTT K. J., HOPPER S., WELLS A., DOIG A., GUILLIAMS T., LATIMER J., MCNAMEE C., NORRIS A., SANSEAU P., CAVALLA D., and PIRMOHAMED M. Drug repurposing: progress, challenges and recommendations. Nature Reviews Drug Discovery, 2019, 18(1): 41-58. https://doi.org/10.1038/nrd.2018.168

KHANNA I. Drug discovery in pharmaceutical industry: productivity challenges and trends. Drug Discovery Today, 2012, 17(19-20): 1088-1102. https://doi.org/10.1016/j.drudis.2012.05.007

PALVE V., LIAO Y., REMSING RIX L. L., and RIX U. Turning liabilities into opportunities: Off-target based drug repurposing in cancer. Seminars in Cancer Biology, 2021, 68: 209-229. https://doi.org/10.1016/j.semcancer.2020.02.003

BOGUSKI M. S., MANDL K. D., and SUKHATME V. P. Drug discovery. Repurposing with a difference. Science, 2009, 324(5933): 1394-1395. https://doi.org/10.1126/science.1169920

AUBÉ J. Drug repurposing and the medicinal chemist. ACS Medicinal Chemistry Letters, 2012, 3(6): 442-444. https://doi.org/10.1021/ml300114c

GUPTA S. C., SUNG B., PRASAD S., WEBB L. J., and AGGARWAL B. B. Cancer drug discovery by repurposing: teaching new tricks to old dogs. Trends in Pharmacological Sciences, 2013, 34(9): 508-517. https://doi.org/10.1016/j.tips.2013.06.005

OVERINGTON J. P., AL-LAZIKANI B., and HOPKINS A. L. How many drug targets are there? Nature Reviews Drug Discovery, 2006, 5(12): 993-996. https://doi.org/10.1038/nrd2199

ENGELMAN J. A., & SETTLEMAN J. Acquired resistance to tyrosine kinase inhibitors during cancer therapy. Current Opinion in Genetics & Development, 2008, 18(1): 73-79. https://doi.org/10.1016/j.gde.2008.01.004

LOVLY C. M., & SHAW A. T. Molecular pathways: resistance to kinase inhibitors and implications for therapeutic strategies. Clinical Cancer Research, 2014, 20(9): 2249-2256. https://doi.org/10.1158/1078-0432.ccr-13-1610

SINGH A., & SETTLEMAN J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene, 2010, 29(34): 4741-4751. https://doi.org/10.1038/onc.2010.215

HOLMES M. D., & CHEN W. Y. Hiding in plain view: the potential for commonly used drugs to reduce breast cancer mortality. Breast Cancer Research, 2012, 14(6): 216. https://doi.org/10.1186/bcr3336

MESTRES J., GREGORI-PUIGJANÉ E., VALVERDE S., and SOLÉ R. V. Data completeness--the Achilles heel of drug-target networks. Nature Biotechnology, 2008, 26(9): 983-984. https://doi.org/10.1038/nbt0908-983

KATO S., MOULDER S. L., UENO N. T., WHELER J. J., MERIC-BERNSTAM F., KURZROCK R., and JANKU F. Challenges and perspective of drug repurposing strategies in early phase clinical trials. Oncoscience, 2015, 2(6): 576-580. https://doi.org/10.18632/oncoscience.173

MURRAY J. C., & LEVY B. Repurposed Drugs Trials by Cancer Type: Lung Cancer. The Cancer Journal, 2019, 25(2): 127-133. https://doi.org/10.1097/ppo.0000000000000371

LALAMI Y., PAESMANS M., AOUN M., MUNOZ-BERMEO R., REUSS K., CHERIFI S., ALEXOPOULOS C. G., and KLASTERSKY J. A prospective randomised evaluation of G-CSF or G-CSF plus oral antibiotics in chemotherapy-treated patients at high risk of developing febrile neutropenia. Supportive Care in Cancer, 2004, 12(10): 725-730. https://doi.org/10.1007/s00520-004-0658-6

LUYT C. E., BRÉCHOT N., TROUILLET J. L., and CHASTRE J. Antibiotic stewardship in the intensive care unit. Critical Care, 2014, 18(5): 480. https://doi.org/10.1186/s13054-014-0480-6

SAAD A. S., SHAHEEN S. M., ELHAMAMSY M. H., and BADARY O. A. An open-label randomized controlled phase II study of clarithromycin (CL) plus CVP in patients (pts) with previously untreated stage III/IV indolent non Hodgkin lymphoma (NHL). Journal of Clinical Oncology, 32(15_suppl). https://doi.org/10.1200/JCO.2014.32.15_SUPPL.E19510

HAMADA K., MIKASA K., YUNOU Y., KURIOKA T., MAJIMA T., NARITA N., and KITA E. Adjuvant effect of clarithromycin on chemotherapy for murine lung cancer. Chemotherapy, 2000, 46(1): 49-61. https://doi.org/10.1159/000007256

OZKAN T., HEKMATSHOAR Y., KARABAY A. Z., KOC A., ALTINOK GUNES B., KARADAG GUREL A., and SUNGUROGLU A. Assessment of azithromycin as an anticancer agent for treatment of imatinib sensitive and resistant CML cells. Leukemia Research, 2021, 102: 106523. https://doi.org/10.1016/j.leukres.2021.106523

ZHOU X., ZHANG Y., LI Y., HAO X., LIU X., and WANG Y. Azithromycin synergistically enhances anti-proliferative activity of vincristine in cervical and gastric cancer cells. Cancers, 2012, 4(4): 1318-1332. https://doi.org/10.3390/cancers4041318

QIAO X., WANG X., SHANG Y., LI Y., and CHEN S. Z. Azithromycin enhances anticancer activity of TRAIL by inhibiting autophagy and up-regulating the protein levels of DR4/5 in colon cancer cells in vitro and in vivo. Cancer Communications, 2018, 38(1): 1-13. https://doi.org/10.1186/s40880-018-0309-9

WNOROWSKA U., FIEDORUK K., PIKTEL E., PRASAD S. V., SULIK M., JANION M., DANILUK T., SAVAGE P. B., and BUCKI R. Nanoantibiotics containing membrane-active human cathelicidin LL-37 or synthetic ceragenins attached to the surface of magnetic nanoparticles as novel and innovative therapeutic tools: current status and potential future applications. Journal of Nanobiotechnology, 2020, 18(1): 3. https://doi.org/10.1186/s12951-019-0566-z

YATSUNAMI J., TSURUTA N., HARA N., and HAYASHI S. Inhibition of tumor angiogenesis by roxithromycin, a 14-membered ring macrolide antibiotic. Cancer Letters, 1998, 131(2): 137-143. https://doi.org/10.1016/S0304-3835(98)00110-4

MOOKHERJEE N., BROWN K. L., BOWDISH D. M., DORIA S., FALSAFI R., HOKAMP K., ROCHE F. M., MU R., DOHO G. H., PISTOLIC J., POWERS J. P., BRYAN J., BRINKMAN F. S., and HANCOCK R. E. Modulation of the TLR-mediated inflammatory response by the endogenous human host defense peptide LL-37. The Journal of Immunology, 2006, 176(4): 2455-2464. https://doi.org/10.4049/jimmunol.176.4.2455

MARIA-NETO S., DE ALMEIDA K. C., MACEDO M. L., and FRANCO O. L. Understanding bacterial resistance to antimicrobial peptides: From the surface to deep inside. Biochimica et Biophysica Acta (BBA) – Biomembranes, 2015, 1848(11, Part B): 3078-3088. https://doi.org/10.1016/j.bbamem.2015.02.017

BOZKURT GUZEL C., OYARDI O., and SAVAGE P. B. Comparative in vitro antimicrobial activities of CSA-142 and CSA-192, second-generation ceragenins, with CSA-13 against various microorganisms. Journal of Chemotherapy, 2018, 30(6-8): 332-337. https://doi.org/10.1080/1120009X.2018.1534567

LI F., HUANG J., JI D., MENG Q., WANG C., CHEN S., WANG X., ZHU Z., JIANG C., SHI Y., LIU S., and LI C. Azithromycin effectively inhibits tumor angiogenesis by suppressing vascular endothelial growth factor receptor 2-mediated signaling pathways in lung cancer. Oncology Letters, 2017, 14(1): 89-96. https://doi.org/10.3892/ol.2017.6103


Refbacks

  • There are currently no refbacks.