1
|
Karamanolis NN, Kounatidis D, Vallianou NG, Dimitriou K, Tsaroucha E, Tsioulos G, Anastasiou IA, Mavrothalassitis E, Karampela I, Dalamaga M. Unraveling the Anti-Cancer Mechanisms of Antibiotics: Current Insights, Controversies, and Future Perspectives. Antibiotics (Basel) 2024; 14:9. [PMID: 39858295 PMCID: PMC11762948 DOI: 10.3390/antibiotics14010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Cancer persists as a significant global health challenge, claiming millions of lives annually despite remarkable strides in therapeutic innovation. Challenges such as drug resistance, toxicity, and suboptimal efficacy underscore the need for novel treatment paradigms. In this context, the repurposing of antibiotics as anti-cancer agents has emerged as an attractive prospect for investigation. Diverse classes of antibiotics have exhibited promising anti-cancer properties in both in vitro and in vivo studies. These mechanisms include the induction of apoptosis and cell cycle arrest, generation of reactive oxygen species, and inhibition of key regulators of cell proliferation and migration. Additional effects involve the disruption of angiogenesis and modulation of pivotal processes such as inflammation, immune response, mitochondrial dynamics, ferroptosis, and autophagy. Furthermore, antibiotics have demonstrated the potential to enhance the efficacy of conventional modalities like chemotherapy and radiotherapy, while alleviating treatment-induced toxicities. Nevertheless, the integration of antibiotics into oncological applications remains contentious, with concerns centered on their disruption of gut microbiota, interference with immunotherapeutic strategies, contribution to microbial resistance, and potential association with tumorigenesis. This narrative review explores the mechanisms of antibiotics' anti-cancer activity, addresses controversies about their dual role in cancer biology, and envisions future perspectives that include the development of novel derivatives and innovative frameworks for their incorporation into cancer treatment paradigms.
Collapse
Affiliation(s)
- Nikolaos Nektarios Karamanolis
- Second Department of Internal Medicine, Hippokratio General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.N.K.); (K.D.)
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (I.A.A.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Krystalia Dimitriou
- Second Department of Internal Medicine, Hippokratio General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.N.K.); (K.D.)
| | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Georgios Tsioulos
- Fourth Department of Internal Medicine, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Ioanna A. Anastasiou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (I.A.A.)
| | - Evangelos Mavrothalassitis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12461 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Mali KK, Gavhane YN, Chakole RD. Natural Polymer-Based Nanogel for pH-Responsive Delivery of Sorafenib Tosylate in Hemangiosarcoma. AAPS PharmSciTech 2024; 25:83. [PMID: 38605211 DOI: 10.1208/s12249-024-02797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Smart nanomedicinal treatment for cancer manifests a solubility challenge with inherent nanoscale size and nonspecific release with stimuli-responsive potential. This is the limelight in novel chemotherapy to pursue physiochemical differences between the tumor microenvironment (TME) and normal cells, which introduces active groups of nanocarriers responding to various stimuli, endowing them with concise responses to various tumor-related signals. The nanogels were successfully prepared by a modified solvent evaporation technique. Nine batches were formulated by changing the chitosan concentration (12, 14, 16 mg/ml) and sonication time (5, 10, 15 min). The formulations were optimized for particle size and zeta potential with high percent entrapment efficiency (%EE) through Central Composite Design software. The optimized batch F7 had a 182-nm size and high zeta potential (64.5 mV) with 98% EE. The drug release of F7 was higher at pH 6 (97.556%) than at pH 7.4 (45.113%). The pharmacokinetic study shows that the release follows the Hixon plot model (R2 = 0.9334) that shifts to zero order (R2 = 0.9149). The nanogel F7 was observed for stability and showed an absence of color change, phase separation, and opacity for 6 months. In the present study, the pH difference between cancer cells and normal cells is the key point of the smart nanogel. This study is promising but challenging depending on the in vivo study. The nanogel was successfully prepared and evaluated for pH-responsive release. As hemangiosarcoma commonly occurs in dogs, this formulation helps to limit the difficulties with administration.
Collapse
Affiliation(s)
- Kiran K Mali
- Department of Pharmaceutics, Government College of Pharmacy, Karad, Shivaji University, Kolhapur, Maharashtra, 415124, India.
- Department of Pharmaceutics, Krishna Foundation's, Jaywant Institute of Pharmacy, Wathar, DBATU, Lonere, Karad, Maharashtra, 415139, India.
| | - Yogeshkumar N Gavhane
- Department of Pharmaceutics, Government College of Pharmacy, Karad, Shivaji University, Kolhapur, Maharashtra, 415124, India
| | - Rita D Chakole
- Department of Pharmacy, Government College of Pharmacy, Karad, Shivaji University, Kolhapur, Maharashtra, 415124, India
| |
Collapse
|
3
|
Banti CN, Kalousi FD, Psarra AMG, Moushi EE, Leonidas DD, Hadjikakou SK. Silver ciprofloxacin (CIPAG): a multitargeted metallodrug in the development of breast cancer therapy. J Biol Inorg Chem 2024; 29:177-186. [PMID: 38581541 PMCID: PMC11098868 DOI: 10.1007/s00775-024-02048-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/06/2024] [Indexed: 04/08/2024]
Abstract
The anti-proliferative activity of the known metalloantibiotic {[Ag(CIPH)2]NO3∙0.75MeOH∙1.2H2O} (CIPAG) (CIPH = ciprofloxacin) against the human breast adenocarcinoma cancer cells MCF-7 (hormone dependent (HD)) and MDA-MB-231 (hormone independent (HI)) is evaluated. The in vitro toxicity and genotoxicity of the metalloantibiotic were estimated toward fetal lung fibroblast (MRC-5) cells. The molecular mechanism of the CIPAG activity against MCF-7 cells was clarified by the (i) cell morphology, (ii) cell cycle arrest, (iii) mitochondrial membrane permeabilization, and (iv) by the assessment of the possible differential effect of CIPAG on estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) transcriptional activation, applying luciferase reporter gene assay. Moreover, the ex vivo mechanism of CIPAG was clarified by its binding affinity toward calf thymus (CT-DNA).
Collapse
Affiliation(s)
- Christina N Banti
- Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece.
| | - Foteini D Kalousi
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Anna-Maria G Psarra
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Eleni E Moushi
- Department of Life Sciences, The School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Demetres D Leonidas
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Sotiris K Hadjikakou
- Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece.
- Institute of Materials Science and Computing, University Research Centre of Ioannina (URCI), Ioannina, Greece.
| |
Collapse
|
4
|
Liu H, Shen W, Liu W, Yang Z, Yin D, Xiao C. From oncolytic peptides to oncolytic polymers: A new paradigm for oncotherapy. Bioact Mater 2024; 31:206-230. [PMID: 37637082 PMCID: PMC10450358 DOI: 10.1016/j.bioactmat.2023.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/18/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
Traditional cancer therapy methods, especially those directed against specific intracellular targets or signaling pathways, are not powerful enough to overcome tumor heterogeneity and therapeutic resistance. Oncolytic peptides that can induce membrane lysis-mediated cancer cell death and subsequent anticancer immune responses, has provided a new paradigm for cancer therapy. However, the clinical application of oncolytic peptides is always limited by some factors such as unsatisfactory bio-distribution, poor stability, and off-target toxicity. To overcome these limitations, oncolytic polymers stand out as prospective therapeutic materials owing to their high stability, chemical versatility, and scalable production capacity, which has the potential to drive a revolution in cancer treatment. This review provides an overview of the mechanism and structure-activity relationship of oncolytic peptides. Then the oncolytic peptides-mediated combination therapy and the nano-delivery strategies for oncolytic peptides are summarized. Emphatically, the current research progress of oncolytic polymers has been highlighted. Lastly, the challenges and prospects in the development of oncolytic polymers are discussed.
Collapse
Affiliation(s)
- Hanmeng Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, 230012, China
| | - Wanguo Liu
- Department of Orthopaedic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, 230012, China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
5
|
Yang S, Leong J, Wang Y, Sim R, Tan KH, Chua YH, Tan N, Lee ALZ, Tay J, Yang YY. Drug-free neutrally charged polypeptide nanoparticles as anticancer agents. J Control Release 2022; 345:464-474. [PMID: 35331785 DOI: 10.1016/j.jconrel.2022.03.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 01/11/2023]
Abstract
Cationic synthetic anticancer polymers and peptides have attracted increasing attention for advancing cancer treatment without causing drug resistance development. To circumvent in vivo instability and toxicity caused by cationic charges of the anticancer polymers/peptides, we report, for the first time, a nanoparticulate delivery system self-assembled from a negatively charged pH-sensitive polypeptide poly(ethylene glycol)-b-poly(ʟ-lysine)-graft-cyclohexene-1,2-dicarboxylic anhydride and a cationic anticancer polypeptide guanidinium-functionalized poly(ʟ-lysine) (PLL-Gua) via electrostatic interaction. The formation of nanoparticles (Gua-NPs) neutralized the positive charges of PLL-Gua. Both PLL-Gua and Gua-NPs killed cancer cells in a dose- and time-dependent manner, and induced cell death via apoptosis. Confocal microscopic studies demonstrated that PLL-Gua and Gua-NPs readily entered cancer cells, and Gua-NPs were taken up by the cells via endocytosis. Notably, Gua-NPs and PLL-Gua exhibited similar in vitro anticancer efficacy against MCF-7 and resistant MCF-7/ADR. PLL-Gua and Gua-NPs also induced similar morphological changes in MCF-7/ADR cells compared to MCF-7 cells, further indicating their ability to bypass drug resistance mechanisms in the MCF-7/ADR cells. More importantly, Gua-NPs with higher LD50 and enhanced tumor accumulation significantly inhibited tumor growth with negligible side effects in vivo. Our findings shed light on the in vivo delivery of anticancer peptides and opened a new avenue for cancer treatment.
Collapse
Affiliation(s)
- Shengcai Yang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Jiayu Leong
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yanming Wang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Rachel Sim
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Ko Hui Tan
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yau Hong Chua
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Nathanael Tan
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore; School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Ashlynn L Z Lee
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Joyce Tay
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore.
| |
Collapse
|