1
|
Chen F, Wen X, Li S, Wu J, Luo Y, Gao Y, Yu X, Chen L. Targeting hypoxia-mediated chemo-immuno resistance by a hybrid NBDHEX-Pt(IV) prodrug via declining nuclear STING1-promoted AhR-CIN in human lung squamous cell carcinoma. Transl Oncol 2025; 55:102350. [PMID: 40138855 PMCID: PMC11985067 DOI: 10.1016/j.tranon.2025.102350] [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: 08/12/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
As found in human lung squamous cell carcinoma (LUSC), STING1 involved in ER-Golgi intermediate compartment (ERGIC) could coordinate immune responses to ectopic DNA triggered by DNA-targeted chemotherapy. ERGIC STING1 is considered to compete with nuclear STING1 to decline aryl hydrocarbon receptor (AhR)-chromosomal instability (CIN)-triggered chronic STING activation which could cause therapeutic resistance. Moreover, GSTP1 was proved to inhibit ERGIC-STING1 via promoting S-glutathione modification of STING1. Hence, a potent GSTP1-targeted Pt(IV) hybrid NBDHEX-DN604, was designed via conjugating a GSTP1 inhibitor NBDHEX to the axial position of Pt(IV) prodrug. As mentioned, hypoxia is mainly observed in malignant tumors and develops acquired drug resistance. In vitro bio-properties of hypoxic SK-MES-1/cDDP cells demonstrated that NBDHEX-DN604 could reverse chemo-immuno resistance via intercepting GSTP1 to activate ERGIC STING1, leading to the decrease of nuclear STING1. The mechanistic data indicated that NBDHEX-DN604 could elevate ERGIC STING1 to mitigate nuclear STING1-mediated AhR-TLS-CIN-chronic activation. Meanwhile, NBDHEX-DN604 was found to decline STING1-AhR-CIN to circumvent chemo-immuno resistance, resulting in predominant in vivo antitumor effect in HY-KLN-205/cDDP-inoculated BALB/c mice. The data provide a novel rationale for the mixed chemo-immunotherapy of NBDHEX-DN604 as a potent Pt(IV) therapeutic method for patients with resistant LUSC.
Collapse
Affiliation(s)
- Feihong Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| | - Xin Wen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Shan Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Jiani Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yaxuan Luo
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuan Gao
- Senior Department of Obstetrics & Gynecology, the Seventh Medical Center of PLA General Hospital, Beijing 100700, China.
| | - Xiaoxuan Yu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China.
| | - Li Chen
- Suzhou Institute for Drug Control, Suzhou 215104, PR China.
| |
Collapse
|
2
|
Fernández-González M, Armisen R, Fernández MI. Predator-Prey Model for Simulating the Genetic Carcinogenicity of Aggressive Toxicant-Related Cancer. J Appl Toxicol 2025. [PMID: 40234059 DOI: 10.1002/jat.4790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/17/2025]
Abstract
The mechanism of how toxicant exposure leads to aggressive tumors remains unresolved. A genetic-based hypothesis predicts that under stress, the transcription of growth-related genes will be inhibited by the activation of mitogenic pathways, redirecting energy toward stress response and increasing survival. This hypothesis fails to explain why epidemiological data suggest that growth and stress response are activated, as patients exposed to toxicants exhibit more aggressive growth than nonexposed individuals. This co-occurrence requires increased energy availability to prevent the activation of mitogenic pathways, as seen in the Warburg effect. We hypothesize that if pollutant effects cease, it might drive aggressive cancer, as excess energy that is no longer used for stress response can fuel rapid growth. We model this allocation between growth and stress response as a trophic competition using the Lotka-Volterra equations and using as input RNA-Seq data from growth- and stress-related genes obtained from cancer cells exposed to copper, cadmium, and carboplatin. Our findings suggest that the energy allocation to growth and its rate of allocation is higher in exposed than nonexposed tumors and results in overgrowth in unexposed cells. This study helps to understand how certain scenarios, such as partial or total cessation of exposure, in toxicant-related cancer can drive cancer aggressiveness.
Collapse
Affiliation(s)
- Mauricio Fernández-González
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Ricardo Armisen
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Mario I Fernández
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Departamento de Urología, Clinica Alemana, Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
3
|
Gomaa S, Nassef M, Tabl G, Gabry SE. Immunoenhancing of the anti-cancer therapy and anti-oxidative stress by co-administration of granulocyte-colony stimulating factor-mobilized stem cells or cells derived from bone marrow and/or spleen plus vaccination with chemotherapeutic cyclophosphamide. Immunol Res 2025; 73:62. [PMID: 40091102 DOI: 10.1007/s12026-025-09610-z] [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: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The combination of immunotherapy and chemotherapy, referred to as chemo-immunotherapy, represents a promising regimen for developing new cancer treatments that target the local tumor microenvironment and target tumors in their early stages. However, this approach carries potential risks, including myelo- and immunosuppression, as well as the emergence of chemo-resistant tumor cells. The purpose of this study was to investigate how well mobilizing hematopoietic stem cells (HSCs) work when used alongside chemotherapy and immunotherapy to enhance and modulate the immune response, thereby overcoming immunosuppression and eliminating distant cancer cells. Ehrlich ascetic carcinoma (EAC) tumor-bearing mice were intraperitoneal (i.p.) preconditioned with CTX (4 mg/mouse). EAC-bearing mice that were preconditioned with CTX were intravenous (i.v.) administered with adoptive transferred naive mice-derived bone marrow cells (nBMCs) at 5 × 106 through lateral tail vein (nBMCs group), adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) at 5 × 106 cell/mouse (tBMCs group), a combination of adoptive transferred naïve mice-derived bone marrow cells (nBMCs) and naïve mice-derived splenocytes (nSPs) at 5 × 106 (nBMCs/nSPs group), a combination of adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) and tumor-bearing mice derived-splenocytes (tSPs) at 5 × 106 cell/mouse (tBMCs/tSPs group), or G-CSF administrated subcutaneously (s.c.) at 5 µg/mouse (G-CSF group). Subsequently, all mice groups were vaccinated with tumor lysate at a dosage of 100 µg/mouse. Treating EAC tumor-bearing mice with G-CSF, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, adoptive transferred tBMCs/tSPs, resulted in a significantly enhanced anti-tumor effect that was evidenced by increased anti-proliferative activity and growth inhibition against EAC tumor cells, increased necrosis and apoptosis rates among EAC tumor cells, restricted tumor growth in EAC tumor-bearing mice, and reduced levels of carcinoembryonic antigen (CEA) tumor marker. Furthermore, there was an improvement in serum levels of antioxidant enzyme superoxide dismutase (SOD) and malondialdehyde (MDA) in EAC tumor-bearing mice receiving G-CSF, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, and adoptive transferred tBMCs/tSPs. Notably, this treatment regimen ameliorates liver and kidney damage associated with CTX administration in EA tumor-bearing mice. The integration of G-CSF-mobilized HSCs, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs combination, and adoptive transferred tBMCs/tSPs combination may yield powerful anti-cancer therapy, thereby facilitating more effective anti-tumor immunotherapy strategies when align with anti-tumor responses. This research may propose a novel therapeutic approach that combines chemotherapy and immunotherapy for addressing early-stage cancer. Further research is necessary to connect the biomedical application and heterogeneity of human tumors and immune systems of this regimen to both diagnostic and therapeutic methodologies.
Collapse
Affiliation(s)
- Soha Gomaa
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt.
| | - Mohamed Nassef
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Ghada Tabl
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Shaimaa El Gabry
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| |
Collapse
|
4
|
Kumari S, Thakur M, Chauhan C, Kumari M. Synthesis, characterization, biological activity and computation-based efficacy of cobalt(II) complexes of biphenyl-2-ol against SARS-CoV-2 virus. J Biomol Struct Dyn 2025; 43:483-497. [PMID: 37990487 DOI: 10.1080/07391102.2023.2283144] [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: 07/13/2023] [Accepted: 11/06/2023] [Indexed: 11/23/2023]
Abstract
Cobalt(II) complexes of biphenyl-2-ol of composition, CoCl2-n(OC6H4C6H5-2)n(H2O)4 (where n = 1 or 2), were prepared by reacting cobaltous(II) chloride with equi- and bimolar ratios of sodium salt of biphenyl-2-ol. The structural characterization of the synthesized complexes was accomplished by NMR, FTIR, thermogravimetry (TGA), high resolution mass spectroscopy (HRMS), electronic spectroscopic techniques coupled with density functional theory (DFT). The stability of the complexes in different pH media of solvent was studied. Chemical reactivity parameters of the newly synthesized complexes, computed using DFT, indicated greater reactivity of complex 2 over complex 1 and free ligand as indicated by its low HOMO-LUMO energy gap corresponding to 1.71 eV. Molecular docking (MD) studies were carried out in order to study the binding affinities between amino acid residues of DNA duplex (PDB ID: 1BNA) and SARS-CoV-2 (PDB ID: 7T9K) with newly synthesized complexes. Complex 2 has shown promising antivirus behaviour with an inhibition constant value of 0.0423 µmol-1 with amino acid residues of SARS-CoV-2 virus. Toxicity of the complexes was predicted using ProTox-II online server. Antibacterial studies have indicated the complexes to exhibit greater efficacy than the free ligand, while the antioxidant activities have suggested them to display enhanced antioxidant behaviour as compared to reference compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shalima Kumari
- Department of Chemistry, Himachal Pradesh University, Shimla, India
| | - Maridula Thakur
- Department of Chemistry, Himachal Pradesh University, Shimla, India
| | - Chetan Chauhan
- Department of Chemistry, Himachal Pradesh University, Shimla, India
| | - Meena Kumari
- Department of Chemistry, Himachal Pradesh University, Shimla, India
| |
Collapse
|
5
|
Man X, Li W, Zhu M, Li S, Xu G, Zhang Z, Liang H, Yang F. Anticancer Tetranuclear Cu(I) Complex Catalyzes a Click Reaction to Synthesize a Chemotherapeutic Agent in situ to Achieve Targeted Dual-Agent Combination Therapy for Cancer. Angew Chem Int Ed Engl 2024; 63:e202411846. [PMID: 39295439 DOI: 10.1002/anie.202411846] [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: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 09/21/2024]
Abstract
To develop next-generation metal-based drugs and dual-drug combination therapy for cancer, we proposed to develop a copper (Cu) complex that exerts anticancer function by integrating chemotherapy, immunotherapy and catalyzes a click reaction for the in situ synthesis of a chemotherapeutic agent, thereby achieving targeted dual-agent combination therapy. We designed and synthesized a tetranuclear Cu(I) complex (Cu4) with remarkable cytotoxicity and notable catalytic ability for the in situ synthesis of a chemotherapeutic agent via Cu(I)-catalyzed azide-alkyne 1,3-cycloaddition (CuAAC). We also constructed an apoferritin (AFt)-Cu4 nanoparticles (NPs) delivery system. Aft-Cu4 NPs not only showed an enhanced performance of tumor growth inhibition, but also improved the targeting ability and reduced the systemic toxicity of Cu4 in vivo. Importantly, the anticancer effect was enhanced by combining the Aft-Cu4 NPs with the resveratrol analogue obtained from the CuAAC reaction in situ. Finally, we revealed the anticancer mechanism of the Cu4/Aft-Cu4 NPs, which involves both cuproptosis and cuproptosis-induced systemic immune response.
Collapse
Affiliation(s)
- Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
- School of Pharmaceutical Sciences, Jiangxi Normal University, Nanchang, Jiangxi, 330022, China
| |
Collapse
|
6
|
Sahoo D, Deb P, Basu T, Bardhan S, Patra S, Sukul PK. Advancements in platinum-based anticancer drug development: A comprehensive review of strategies, discoveries, and future perspectives. Bioorg Med Chem 2024; 112:117894. [PMID: 39214013 DOI: 10.1016/j.bmc.2024.117894] [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: 06/18/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Platinum-based anticancer drugs have been at the forefront of cancer chemotherapy, with cisplatin emerging as a pioneer in the treatment of various malignancies. This review article provides a comprehensive overview of the evolution of platinum-based anticancer therapeutics, focusing on the development of cisplatin, platinum(IV) prodrugs, and the integration of photodynamic therapy (PDT) for enhanced cancer treatment results. The first section of the review delves into the historical context and molecular mechanisms underlying the success of cisplatin, highlighting its DNA binding properties and subsequent interference with cellular processes. Despite its clinical efficacy, the inherent limitations, including dose-dependent toxicities and acquired resistance, accelerated the exploration of novel platinum derivatives. This led to the emergence of platinum(IV) prodrugs, designed to overcome resistance mechanisms and enhance selectivity through targeted drug delivery. The subsequent section provides an in-depth analysis of the principles of design and structural modifications employed in the development of platinum(IV) prodrugs. The transitions to the incorporation of photodynamic therapy (PDT) stands out as a synergistic approach to platinum-based anticancer treatment. The photophysical properties of platinum complexes are discussed in the context of their potential application in PDT, emphasizing on combined cytotoxic effects of platinum-based drugs and light-induced reactive oxygen species generation. This dual-action approach holds great promise for overcoming the limitations of traditional chemotherapy as well as producing superior therapeutic outcomes. Overall, the present report explores the latest developments in the development and use of platinum complexes, highlighting novel strategies such combination treatments, targeted delivery methods, and the generation of multifunctional complexes. It also provides a comprehensive overview of the current landscape while proposing future directions for the development of next-generation platinum-based anticancer therapeutics.
Collapse
Affiliation(s)
- Debsankar Sahoo
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Action Area-II, Kadampukur, New Town, Rajarhat, Kolkata 700135, India
| | - Priya Deb
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Action Area-II, Kadampukur, New Town, Rajarhat, Kolkata 700135, India
| | - Tamal Basu
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Action Area-II, Kadampukur, New Town, Rajarhat, Kolkata 700135, India
| | - Srishti Bardhan
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Action Area-II, Kadampukur, New Town, Rajarhat, Kolkata 700135, India
| | - Sayan Patra
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Action Area-II, Kadampukur, New Town, Rajarhat, Kolkata 700135, India
| | - Pradip K Sukul
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Action Area-II, Kadampukur, New Town, Rajarhat, Kolkata 700135, India; Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| |
Collapse
|
7
|
Son B, Lee W, Kim H, Shin H, Park HH. Targeted therapy of cancer stem cells: inhibition of mTOR in pre-clinical and clinical research. Cell Death Dis 2024; 15:696. [PMID: 39349424 PMCID: PMC11442590 DOI: 10.1038/s41419-024-07077-8] [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: 12/29/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Cancer stem cells (CSCs) are a type of stem cell that possesses not only the intrinsic abilities of stem cells but also the properties of cancer cells. Therefore, CSCs are known to have self-renewal and outstanding proliferation capacity, along with the potential to differentiate into specific types of tumor cells. Cancers typically originate from CSCs, making them a significant target for tumor treatment. Among the related cascades of the CSCs, mammalian target of rapamycin (mTOR) pathway is regarded as one of the most important signaling pathways because of its association with significant upstream signaling: phosphatidylinositol 3‑kinase/protein kinase B (PI3K/AKT) pathway and mitogen‑activated protein kinase (MAPK) cascade, which influence various activities of stem cells, including CSCs. Recent studies have shown that the mTOR pathway not only affects generation of CSCs but also the maintenance of their pluripotency. Furthermore, the maintenance of pluripotency or differentiation into specific types of cancer cells depends on the regulation of the mTOR signal in CSCs. Consequently, the clinical potential and importance of mTOR in effective cancer therapy are increasing. In this review, we demonstrate the association between the mTOR pathway and cancer, including CSCs. Additionally, we discuss a new concept for anti-cancer drug development aimed at overcoming existing drawbacks, such as drug resistance, by targeting CSCs through mTOR inhibition.
Collapse
Affiliation(s)
- Boram Son
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Wonhwa Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyeonjeong Kim
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
8
|
Wang C, Li Q, Song K, Wang W, Zhang N, Dai L, Di W. Nanoparticle co-delivery of carboplatin and PF543 restores platinum sensitivity in ovarian cancer models through inhibiting platinum-induced pro-survival pathway activation. NANOSCALE ADVANCES 2024; 6:4082-4093. [PMID: 39114142 PMCID: PMC11302180 DOI: 10.1039/d4na00227j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/29/2024] [Indexed: 08/10/2024]
Abstract
Resistance to platinum-based chemotherapy is the major cause of poor prognosis and cancer-associated mortality in ovarian cancer patients, so novel therapeutic strategies to restore platinum sensitivity are needed to improve patient outcomes. Sphingosine Kinase (SphK) 1 is involved in regulating multiple pro-survival pathways, key mediators in the sensitivity of tumor cells toward platinum. By encapsulating CBP and the SphK1 inhibitor PF543 in PLGA (poly lactic-co-glycolic acid) nanoparticles, a dual-drug delivery system (C/PNPs) was formed to simultaneously deliver CBP and PF543. The physicochemical characteristics, cell uptake rate and biodistribution behavior of C/PNPs were evaluated. Then the anti-tumor ability of C/PNPs in vitro and in vivo was further investigated. The C/PNPs could deliver CBP and PF543 simultaneously to a platinum-insensitive cell line (SKOV3) both in vitro and in vivo. Furthermore, benefiting from the enhanced permeability and retention (EPR) effect of PLGA NPs, C/PNPs exhibited an improved tumor region accumulation. As a result, a synergistic anti-tumor effect was found in the SKOV3 tumor-bearing mice, with tumor volume inhibiting rates of 84.64% and no side effects in major organs. The mechanistic studies confirmed that the inhibition of SphK1 by PF543 sensitized SKOV3 cells to CBP chemotherapy, partly by inhibiting the CBP-induced activation of pro-survival pathways, including ERK, AKT and STAT3 signaling. Our study reveals that C/PNPs can serve as an efficient dual-drug delivery system to restore platinum sensitivity in ovarian cancer models partly through inhibiting platinum-induced pro-survival pathway activation.
Collapse
Affiliation(s)
- Chen Wang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Qing Li
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Keqi Song
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Wenjing Wang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Ning Zhang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Lan Dai
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| |
Collapse
|
9
|
Luković D, Franich AA, Živković MD, Rajković S, Stojanović B, Gajović N, Jurišević M, Pavlović S, Simović Marković B, Jovanović M, Stojanović BS, Pavlović R, Jovanović I. Biological Evaluation of Dinuclear Platinum(II) Complexes with Aromatic N-Heterocycles as Bridging Ligands. Int J Mol Sci 2024; 25:8525. [PMID: 39126093 PMCID: PMC11312983 DOI: 10.3390/ijms25158525] [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: 07/13/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
The history of effective anti-cancer medications begins with the discovery of cisplatin's anti-cancer properties. Second-generation analogue, carboplatin, with a similar range of effectiveness, made progress in improving these drugs with fewer side effects and better solubility. Renewed interest in platinum-based drugs has been increasing in the past several years. These developments highlight a revitalized enthusiasm and ongoing exploration in platinum chemotherapy based on the series of dinuclear platinum(II) complexes, [{Pt(L)Cl}2(μ-bridging ligand)]2+, which have been synthesized and evaluated for their biological activities. These complexes are designed to target various cancerous conditions, exhibiting promising antitumor, antiproliferative, and apoptosis-inducing activities. The current work aims to shed light on the potential of these complexes as next-generation platinum-based therapies, highlighting their enhanced efficacy and reduced side effects, which could revolutionize the approach to chemotherapy.
Collapse
Affiliation(s)
- Desimir Luković
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Andjela A. Franich
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia; (A.A.F.); (S.R.)
| | - Marija D. Živković
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia
| | - Snežana Rajković
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia; (A.A.F.); (S.R.)
| | - Bojan Stojanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia
| | - Nevena Gajović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Milena Jurišević
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
- Department of Clinical Pharmacy, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia;
| | - Slađana Pavlović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Bojana Simović Marković
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Marina Jovanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Bojana S. Stojanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia
| | - Radiša Pavlović
- Department of Clinical Pharmacy, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia;
| | - Ivan Jovanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| |
Collapse
|
10
|
Roufosse B, Serbu C, Marschner C, Prince S, Blom B. Homo and heteromultimetallic complexes containing a group 8 transition metal and μ-diphosphine bridging ligands involved in anticancer research: A review. Eur J Med Chem 2024; 274:116528. [PMID: 38805938 DOI: 10.1016/j.ejmech.2024.116528] [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: 04/04/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Herein, we present a comprehensive review focusing on synthetic strategies, detailed structural analysis, and anticancer activity investigations of complexes following the general formula [LnM(μ-diphosphine)M'Lm] where M = group 8 metal; M' = any transition metal; μ-diphosphine = bridging ligand; Ln and Lm = ligand spheres). Both homo- and heteromultimetallic complexes will be discussed in detail. We review in vitro, in vivo and in silico anticancer activity investigations, in an attempt to draw comparisons between the various complexes and derive structure-activity relationships (SAR). This review solely focuses on complexes falling under the general formula stated above that have been studied for their anticancer activities, other complexes falling into that scheme but which have not undergone anticancer testing are not included in this review. We compare the anticancer activities of these complexes to their mononuclear counterparts, and a positive control (cisplatin) when possible and present a summary of all existing data to date and attempt to draw some conclusions on the future development of these complexes.
Collapse
Affiliation(s)
- Basile Roufosse
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, the Netherlands
| | - Christi Serbu
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, the Netherlands
| | - Christoph Marschner
- Institut für Anorganische Chemie, Technische Universität Graz, Stremayrgasse 9, A-8010, Graz, Austria
| | - Sharon Prince
- Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa
| | - Burgert Blom
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, the Netherlands.
| |
Collapse
|
11
|
Nandhini S, Thiruppathi G, Ranjani M, Puschmann H, Ravi M, Sundararaj P, Prabhakaran R. Effect of ruthenium(II) complexes on MDA-MB-231 cells and lifespan/tumor growth in gld-1mutant, Daf-16 TF and stress productive genes: A perspective study. J Inorg Biochem 2024; 257:112580. [PMID: 38701694 DOI: 10.1016/j.jinorgbio.2024.112580] [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: 02/19/2024] [Revised: 04/17/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
Pincer type coumarin based N-substituted semicarbazone ligands HL1-4 and their corresponding ruthenium(II) complexes (1-4) were synthesized, analyzed and confirmed by various spectro analytical techniques. The molecular structure of the ligand HL3 and complex 3 was confirmed by single crystal X-ray diffraction analysis. The stoichiometry of complexes 1, 2 and 4 was confirmed by high resolution mass spectroscopy (HRMS). The binding affinity of the compounds with CT-DNA (Calf Thymus DNA) and BSA (Bovine Serum Albumin) was established by absorption and emission titration methods. The results of In vitro cytotoxicity showed the significant cytotoxic potential of the complexes against MDA-MB-231 cells (TNBC- Triple-negative breast cancer). Among the complexes, 1 and 4 have shown appreciable results. Further, antimigratory activity against the MDA-MB-231 cells was studied for the complexes 1 and 4. The percentage cell cycle arrest, apoptosis and necrosis were explored by flow cytometry. The in vivo anti-tumor activity of the complexes 1 and 4 using C. elegans as model organism was established by using the tumoral C. elegans strain JK1466 (gld-1(q485)), which bears a mutation in the gld-1 tumor suppressor gene. We have determined the effect of our complexes on tumor gonad reduction and found to be non toxic to the JK1466 worms and they have prolonged their mean lifespan with potential antioxidant ability by overcoming stress responses. Overall, our study reported herein demonstrated that the complexes 1 and 4 could be established as potential metallo-drugs substantiating further exploration.
Collapse
Affiliation(s)
- S Nandhini
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India
| | - G Thiruppathi
- Department of Zoology, Bharathiar University, Coimbatore 641 046, India
| | - M Ranjani
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India
| | - Horst Puschmann
- Department of Chemistry, Durham University, Durham DH1 3LE, UK
| | - M Ravi
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600025, India
| | - P Sundararaj
- Department of Zoology, Bharathiar University, Coimbatore 641 046, India
| | - R Prabhakaran
- Department of Chemistry, Bharathiar University, Coimbatore 641 046, India.
| |
Collapse
|
12
|
Hobsteter AW, Irazoqui AP, Gonzalez A, Picco AS, Rubert AA, Buitrago CG, Lo Fiego MJ, Silbestri GF. Acetylated galactopyranosyl N-heterocyclic monocarbene complexes of Silver(I) as novel anti-proliferative agents in a rhabdomyosarcoma cell line. Bioorg Med Chem 2024; 107:117756. [PMID: 38759255 DOI: 10.1016/j.bmc.2024.117756] [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: 04/15/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Herein, four silver(I) complexes bearing acetylated d-galactopyranoside-based N-heterocyclic carbene ligands were synthesized and fully characterized by elemental analysis, NMR, and X-ray photoelectron spectroscopy. All complexes were obtained with an anomeric β-configuration and as monocarbene species. In this study, we investigated the biological effects of the silver(I) complexes 2a-d on the human rhabdomyosarcoma cell line, RD. Our results show concentration-dependent effects on cell density, growth inhibition, and activation of key signaling pathways such as Akt 1/2, ERK 1/2, and p38-MAPK, indicating their potential as anticancer agents. Notably, at 35.5 µM, the complexes induced mitochondrial network disruption, as observed with 2b and 2c, whereas with 2a, this disruption was accompanied by nuclear content release. These results provide insight into the utility of carbohydrate incorporated NHC complexes of silver(I) as new agents in cancer therapy.
Collapse
Affiliation(s)
- Ariana W Hobsteter
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina
| | - Ana P Irazoqui
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina; Comisión de Investigaciones Científicas de la provincia de Buenos Aires (CIC PBA), Argentina.
| | - Agustina Gonzalez
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Agustín S Picco
- INIFTA, Fac. de Cs. Exactas, Universidad Nacional de La Plata-CONICET, 1900 La Plata, Argentina
| | - Aldo A Rubert
- INIFTA, Fac. de Cs. Exactas, Universidad Nacional de La Plata-CONICET, 1900 La Plata, Argentina
| | - Claudia G Buitrago
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Marcos J Lo Fiego
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina.
| | - Gustavo F Silbestri
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina
| |
Collapse
|
13
|
Chen Q, Fang C, Xia F, Wang Q, Li F, Ling D. Metal nanoparticles for cancer therapy: Precision targeting of DNA damage. Acta Pharm Sin B 2024; 14:1132-1149. [PMID: 38486992 PMCID: PMC10934341 DOI: 10.1016/j.apsb.2023.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/30/2023] [Accepted: 08/15/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer, a complex and heterogeneous disease, arises from genomic instability. Currently, DNA damage-based cancer treatments, including radiotherapy and chemotherapy, are employed in clinical practice. However, the efficacy and safety of these therapies are constrained by various factors, limiting their ability to meet current clinical demands. Metal nanoparticles present promising avenues for enhancing each critical aspect of DNA damage-based cancer therapy. Their customizable physicochemical properties enable the development of targeted and personalized treatment platforms. In this review, we delve into the design principles and optimization strategies of metal nanoparticles. We shed light on the limitations of DNA damage-based therapy while highlighting the diverse strategies made possible by metal nanoparticles. These encompass targeted drug delivery, inhibition of DNA repair mechanisms, induction of cell death, and the cascading immune response. Moreover, we explore the pivotal role of physicochemical factors such as nanoparticle size, stimuli-responsiveness, and surface modification in shaping metal nanoparticle platforms. Finally, we present insights into the challenges and future directions of metal nanoparticles in advancing DNA damage-based cancer therapy, paving the way for novel treatment paradigms.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiyue Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| |
Collapse
|
14
|
Pei L, Zhao F, Zhang Y. USP43 impairs cisplatin sensitivity in epithelial ovarian cancer through HDAC2-dependent regulation of Wnt/β-catenin signaling pathway. Apoptosis 2024; 29:210-228. [PMID: 38087046 PMCID: PMC10830728 DOI: 10.1007/s10495-023-01873-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2023] [Indexed: 02/01/2024]
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of cancer death all over the world. USP43 functions as a tumor promoter in various malignant cancers. Nevertheless, the biological roles and mechanisms of USP43 in EOC remain unknown. In this study, USP43 was highly expressed in EOC tissues and cells, and high expression of USP43 were associated with a poor prognosis of EOC. USP43 overexpression promoted EOC cell proliferation, enhanced the ability of migration and invasion, decreased cisplatin sensitivity and inhibited apoptosis. Knockdown of USP43 in vitro effectively retarded above malignant progression of EOC. In vivo xenograft tumors, silencing USP43 slowed tumor growth and enhanced cisplatin sensitivity. Mechanistically, USP43 inhibited HDAC2 degradation and enhanced HDAC2 protein stability through its deubiquitylation function. USP43 diminished the sensitivity of EOC cells to cisplatin through activation of the Wnt/β-catenin signaling pathway mediated by HDAC2. Taken together, the data in this study revealed the functions of USP43 in proliferation, migration, invasion, chemoresistance of EOC cells, and the mechanism of HDAC2-mediated Wnt/β-catenin signaling pathway. Thus, USP43 might serve as a potential target for the control of ovarian cancer progression.
Collapse
Affiliation(s)
- Lipeng Pei
- Department of Obstetrics and Gynecology, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Yi Zhang
- Department of Gynecology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang, People's Republic of China.
| |
Collapse
|
15
|
Gomaa S, Nassef M, Tabl G, Zaki S, Abdel-Ghany A. Doxorubicin and folic acid-loaded zinc oxide nanoparticles-based combined anti-tumor and anti-inflammatory approach for enhanced anti-cancer therapy. BMC Cancer 2024; 24:34. [PMID: 38178054 PMCID: PMC10768430 DOI: 10.1186/s12885-023-11714-4] [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: 06/20/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Zinc oxide nanoparticles (ZnONPs) have impressively shown their efficacy in targeting and therapy of cancer. The present research was designated to investigate the potential of ZnONP nanocomposites as a cancer chemotherapeutic-based drug delivery system and to assess the anti-tumor and anti-inflammatory effectiveness of ZnONP nanocomposites combination with systemic chemotherapeutic drugs doxorubicin (DOX) and folic acid (FA) in Ehrlich ascites carcinoma (EAC) tumor cell line both in vitro and in vivo. METHODS Anti-tumor potential of ZnONP nanocomposites: ZnONPs, ZnONPs/FA, ZnONPs/DOX and ZnONPs/DOX/FA against EAC tumor cell line was evaluated in vitro by MTT assay. Anti-tumor and anti-inflammatory efficacy of ZnONP nanocomposites were analyzed in vivo by examination of the proliferation rate and apoptosis rate of EAC tumor cells by flow cytometry, splenocytes count, level of inflammatory markers interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α), as well as liver and kidney function in EAC-challenged mice. RESULTS In vitro results showed that ZnONP nanocomposites showed a high anti-proliferative potency against EAC tumor cells. Furthermore, the in vivo study revealed that the treatment EAC-challenged mice with ZnONPs, ZnONPs/DOX, ZnONPs/FA and ZnONPs/DOX/FA hindered the proliferation rate of implanted EAC tumor cells through lowering their number and increasing their apoptosis rate. Moreover, the treatment of EAC-challenged mice with ZnONPs/DOX/FA markedly decreased the level of IL-6 and TNF-α and remarkably ameliorated the liver and kidney damages that were elevated by implantation of EAC tumor cells, restoring the liver and kidney functions to be close to the naïve mice control. CONCLUSION ZnONP nanocomposites may be useful as a cancer chemotherapeutic-based drug delivery system. ZnONP nanocomposites: ZnONPs/DOX, ZnONPs/FA and ZnONPs/DOX/FA regimen may have anti-inflammatory approaches and a great potential to increase anti-tumor effect of conventional chemotherapy, overcoming resistance to cancer systemic chemotherapeutics and reducing their side effects, offering a promising regimen for cancer therapy.
Collapse
Affiliation(s)
- Soha Gomaa
- Zoology department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed Nassef
- Zoology department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Ghada Tabl
- Zoology department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Somia Zaki
- Zoology department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Asmaa Abdel-Ghany
- Zoology department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
16
|
Qu B, Liu J, Peng Z, Xiao Z, Li S, Wu J, Li S, Luo J. CircSOD2 polarizes macrophages towards the M1 phenotype to alleviate cisplatin resistance in gastric cancer cells by targeting the miR-1296/STAT1 axis. Gene 2023; 887:147733. [PMID: 37625563 DOI: 10.1016/j.gene.2023.147733] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Cisplatin is the first-line drug for gastric cancer (GC). Cisplatin resistance is the most important cause of poor prognosis for GC. Increasing evidence has identified the important role of macrophage polarization in chemoresistance. CircRNAs are newly discovered non-coding RNAs, characterized by covalently closed loops with high stability. Previous studies have reported a significant difference between circRNA profiles expressed in classically activated M1 macrophages, and those expressed in alternatively activated M2 macrophages. However, the underlying mechanism behind the regulation of GC cisplatin resistance by macrophages remains unclear. In our study, we observed the aberrant high expression of circSOD2 in M1 macrophages derived from THP-1. These expression patterns were confirmed in macrophages from patients with GC. Detection of the M1 and M2 markers confirmed that overexpression of circSOD2 enhances M1 polarization. The viability of cisplatin-treated GC cells was significantly reduced in the presence of macrophages overexpressing circSOD2, and cisplatin-induced apoptosis increased dramatically. In vivo experiments showed that macrophages expressing circSOD2 enhanced the effect of cisplatin. Moreover, we demonstrated that circSOD2 acts as a microRNA sponge for miR-1296 and regulates the expression of its target gene STAT1 (signal transducer and activator of transcription 1). CircSOD2 exerts its function through the miR-1296/STAT1 axis. Inhibition of circSOD2/miR-1296/STAT1 may therefore reduce M1 polarization. Overexpression of circSOD2 promotes the polarization of M1 macrophages and enhances the effect of cisplatin in GC. CircSOD2 is a novel positive regulator of M1 macrophages and may serve as a potential target for GC chemotherapy.
Collapse
Affiliation(s)
- Bing Qu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiasheng Liu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhiyang Peng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhe Xiao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shijun Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jianguo Wu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shengbo Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jianfei Luo
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
17
|
Kumari P, Ghosh S, Acharya S, Mitra P, Roy S, Ghosh S, Maji M, Singh S, Mukherjee A. Cytotoxic Imidazolyl-Mesalazine Ester-Based Ru(II) Complexes Reduce Expression of Stemness Genes and Induce Differentiation of Oral Squamous Cell Carcinoma. J Med Chem 2023; 66:14061-14079. [PMID: 37831489 DOI: 10.1021/acs.jmedchem.3c01092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The aggressiveness and recurrence of cancer is linked to cancer stem cells (CSCs), but drugs targeting CSCs may not succeed in the clinic due to the lack of a distinct CSC subpopulation. Clinical Pt(II) drugs can increase stemness. We screened 15 RuII or IrIII complexes with mesalazine or 3-aminobenzoate Schiff bases of the general formulas [Ru(p-cym)L]+, [Ru(p-cym)L], and [Ir(Cp*)L]+ (L = L1-L9) and found three complexes (2, 12, and 13) that are active against oral squamous cell carcinoma (OSCC) CSCs. There is a putative oncogenic role of transcription factors (viz. NOTCH1, SOX2, c-MYC) to enhance the stemness. Our work shows that imidazolyl-mesalazine ester-based RuII complexes inhibit growth of CSC-enriched OSCC 3D spheroids at low micromolar doses (2 μM). Complexes 2, 12, and 13 reduce stemness gene expression and induce differentiation markers (Involucrin, CK10) in OSCC 3D cultures. The imidazolyl-mesalazine ester-based RuII complex 13 shows the strongest effect. Downregulating c-MYC suggests that RuII complexes may target c-MYC-driven cancers.
Collapse
Affiliation(s)
- Pragya Kumari
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Subhashis Ghosh
- National Institute of Biomedical Genomics, Kalyani-741251, West Bengal, India
| | - Sourav Acharya
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Paromita Mitra
- National Institute of Biomedical Genomics, Kalyani-741251, West Bengal, India
| | - Souryadip Roy
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Shilpendu Ghosh
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Moumita Maji
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Sandeep Singh
- National Institute of Biomedical Genomics, Kalyani-741251, West Bengal, India
| | - Arindam Mukherjee
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| |
Collapse
|
18
|
Oh SJ, Lim JY, Son MK, Ahn JH, Song KH, Lee HJ, Kim S, Cho EH, Chung JY, Cho H, Kim H, Kim JH, Park J, Choi J, Hwang SW, Kim TW. TRPV1 inhibition overcomes cisplatin resistance by blocking autophagy-mediated hyperactivation of EGFR signaling pathway. Nat Commun 2023; 14:2691. [PMID: 37165076 PMCID: PMC10172196 DOI: 10.1038/s41467-023-38318-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/25/2023] [Indexed: 05/12/2023] Open
Abstract
Cisplatin resistance along with chemotherapy-induced neuropathic pain is an important cause of treatment failure for many cancer types and represents an unmet clinical need. Therefore, future studies should provide evidence regarding the mechanisms of potential targets that can overcome the resistance as well as alleviate pain. Here, we show that the emergence of cisplatin resistance is highly associated with EGFR hyperactivation, and that EGFR hyperactivation is arisen by a transcriptional increase in the pain-generating channel, TRPV1, via NANOG. Furthermore, TRPV1 promotes autophagy-mediated EGF secretion via Ca2+ influx, which activates the EGFR-AKT signaling and, consequentially, the acquisition of cisplatin resistance. Importantly, TRPV1 inhibition renders tumors susceptible to cisplatin. Thus, our findings indicate a link among cisplatin resistance, EGFR hyperactivation, and TRPV1-mediated autophagic secretion, and implicate that TRPV1 could be a crucial drug target that could not only overcome cisplatin resistance but also alleviate pain in NANOG+ cisplatin-resistant cancer.
Collapse
Affiliation(s)
- Se Jin Oh
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Ji Yeon Lim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Physiology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Min Kyu Son
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jun Hyeok Ahn
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Kwon-Ho Song
- Department of Cell biology, Daegu Catholic University School of Medicine, Daegu, 42472, Republic of Korea
| | - Hyo-Jung Lee
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Suyeon Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Eun Ho Cho
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Hyosun Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Jooyoung Park
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sun Wook Hwang
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Physiology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Tae Woo Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- NEX-I Inc., Seoul, 05854, Republic of Korea.
| |
Collapse
|
19
|
Odachowski M, Neven R, Perversi G, Romano D, Slabber CA, Hadiji M, Honing M, Zhao Y, Munro OQ, Blom B. Ionic mononuclear [Fe] and heterodinuclear [Fe,Ru] bis(diphenylphosphino)alkane complexes: Synthesis, spectroscopy, DFT structures, cytotoxicity, and biomolecular interactions. J Inorg Biochem 2023; 242:112156. [PMID: 36801621 DOI: 10.1016/j.jinorgbio.2023.112156] [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: 11/11/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/06/2023]
Abstract
Iron(II) and Ru(II) half-sandwich compounds encompass some promising pre-clinical anticancer agents whose efficacy may be tuned by structural modification of the coordinated ligands. Here, we combine two such bioactive metal centres in cationic bis(diphenylphosphino)alkane-bridged heterodinuclear [Fe2+, Ru2+] complexes to delineate how ligand structural variations modulate compound cytotoxicity. Specifically, Fe(II) complexes of the type [(η5-C5H5)Fe(CO)2(κ1-PPh2(CH2)nPPh2)]{PF6} (n = 1-5), compounds 1-5, and heterodinuclear [Fe2+, Ru2+] complexes, [(η5-C5H5)Fe(CO)2(μ-PPh2(CH2)nPPh2))(η6-p-cymene)RuCl2]{PF6} (n = 2-5) (compounds 7-10), were synthesized and characterised. The mononuclear complexes were moderately cytotoxic against two ovarian cancer cell lines (A2780 and cisplatin resistant A2780cis) with IC50 values ranging from 2.3 ± 0.5 μM to 9.0 ± 1.4 μM. For 7-10, the cytotoxicity increased with increasing Fe⋅⋅⋅Ru distance, consistent with their DNA affinity. UV-visible spectroscopy suggested the chloride ligands in heterodinuclear 8-10 undergo stepwise substitution by water on the timescale of the DNA interaction experiments, probably affording the species [RuCl(OH2)(η6-p-cymene)(PRPh2)]2+ and [Ru(OH)(OH2)(η6-p-cymene)(PRPh2)]2+ (where PRPh2 has R = [-(CH2)5PPh2-Fe(C5H5)(CO)2]+). One interpretation of the combined DNA-interaction and kinetic data is that the mono(aqua) complex may interact with dsDNA through nucleobase coordination. Heterodinuclear 10 reacts with glutathione (GSH) to form stable mono- and bis(thiolate) adducts, 10-SG and 10-SG2, with no evidence of metal ion reduction (k1 = 1.07 ± 0.17 × 10-1 min-1 and k2 = 6.04 ± 0.59 × 10-3 min-1 at 37 °C). This work highlights the synergistic effect of the Fe2+/Ru2+ centres on both the cytotoxicity and biomolecular interactions of the present heterodinuclear complexes.
Collapse
Affiliation(s)
- Matylda Odachowski
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul Henri Spaaklaan 1, 6229 EN Maastricht, The Netherlands
| | - Robin Neven
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul Henri Spaaklaan 1, 6229 EN Maastricht, The Netherlands
| | - Giuditta Perversi
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul Henri Spaaklaan 1, 6229 EN Maastricht, The Netherlands
| | - Dario Romano
- King Abdullah University of Science and Technology, Department of Chemical Sciences, Division of Physical Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Cathryn A Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, PO WITS 2050 Johannesburg, South Africa
| | - Mouna Hadiji
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH1015 Lausanne, Switzerland
| | - Maarten Honing
- Maastricht Multimodal Molecular Imaging (M4i) Institute, Division of Imaging Mass Spectrometry Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Yuandi Zhao
- Maastricht Multimodal Molecular Imaging (M4i) Institute, Division of Imaging Mass Spectrometry Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, PO WITS 2050 Johannesburg, South Africa.
| | - Burgert Blom
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul Henri Spaaklaan 1, 6229 EN Maastricht, The Netherlands.
| |
Collapse
|
20
|
Aputen AD, Elias MG, Gilbert J, Sakoff JA, Gordon CP, Scott KF, Aldrich-Wright JR. Versatile Platinum(IV) Prodrugs of Naproxen and Acemetacin as Chemo-Anti-Inflammatory Agents. Cancers (Basel) 2023; 15:cancers15092460. [PMID: 37173934 PMCID: PMC10177380 DOI: 10.3390/cancers15092460] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/22/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Developing new and versatile platinum(IV) complexes that incorporate bioactive moieties is a rapidly evolving research strategy for cancer drug discovery. In this study, six platinum(IV) complexes (1-6) that are mono-substituted in the axial position with a non-steroidal anti-inflammatory molecule, naproxen or acemetacin, were synthesised. A combination of spectroscopic and spectrometric techniques confirmed the composition and homogeneity of 1-6. The antitumour potential of the resultant complexes was assessed on multiple cell lines and proved to be significantly improved compared with cisplatin, oxaliplatin and carboplatin. The platinum(IV) derivatives conjugated with acemetacin (5 and 6) were determined to be the most biologically potent, demonstrating GI50 values ranging between 0.22 and 250 nM. Remarkably, in the Du145 prostate cell line, 6 elicited a GI50 value of 0.22 nM, which is 5450-fold more potent than cisplatin. A progressive decrease in reactive oxygen species and mitochondrial activity was observed for 1-6 in the HT29 colon cell line, up to 72 h. The inhibition of the cyclooxygenase-2 enzyme was also demonstrated by the complexes, confirming that these platinum(IV) complexes may reduce COX-2-dependent inflammation and cancer cell resistance to chemotherapy.
Collapse
Affiliation(s)
- Angelico D Aputen
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South, Sydney, NSW 2751, Australia
| | - Maria George Elias
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South, Sydney, NSW 2751, Australia
- Ingham Institute, Liverpool, Sydney, NSW 2170, Australia
| | - Jayne Gilbert
- Calvary Mater Newcastle Hospital, Waratah, Newcastle, NSW 2298, Australia
| | - Jennette A Sakoff
- Calvary Mater Newcastle Hospital, Waratah, Newcastle, NSW 2298, Australia
| | - Christopher P Gordon
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South, Sydney, NSW 2751, Australia
| | - Kieran F Scott
- Ingham Institute, Liverpool, Sydney, NSW 2170, Australia
- School of Medicine, Western Sydney University, Locked Bag 1797, Penrith South, Sydney, NSW 2751, Australia
| | - Janice R Aldrich-Wright
- School of Science, Western Sydney University, Locked Bag 1797, Penrith South, Sydney, NSW 2751, Australia
| |
Collapse
|
21
|
Shi C, Huang K, Soto J, Sankaran R, Kalia V, Onwumere O, Young M, Einbond L, Redenti S. Piperlongumine inhibits proliferation and oncogenic MYCN expression in chemoresistant metastatic retinoblastoma cells directly and through extracellular vesicles. Biomed Pharmacother 2023; 161:114554. [PMID: 36940616 PMCID: PMC10157982 DOI: 10.1016/j.biopha.2023.114554] [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: 11/06/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
Ocular retinoblastoma malignancies, which develop into metastatic phenotypes, result in poor prognosis and survival for infant and child patients. To improve the prognosis of metastatic retinoblastoma, it is important to identify novel compounds with less toxic side effects and higher therapeutic efficacy compared to existing chemotherapeutics. Piperlongumine (PL), a neuroprotective, plant-derived compound has been explored for its anticancer activities both in vitro and in vivo. Here, we analyze the potential efficacy of PL for metastatic retinoblastoma cell treatment. Our data reveal that PL treatment significantly inhibits cell proliferation in metastatic retinoblastoma Y79 cells compared to the commonly used retinoblastoma chemotherapeutic drugs carboplatin, etoposide, and vincristine. PL treatment also significantly increases cell death compared to treatment with other chemotherapeutic drugs. PL-induced cell-death signaling was associated with significantly higher caspase 3/7 activities and greater loss of mitochondrial membrane potential. PL was also internalized into Y79 cells with an estimated concentration of 0.310pM and expression analysis revealed reduced MYCN oncogene levels. We next examined extracellular vesicles derived from PL-treated Y79 cells. Extracellular vesicles in other cancers are pro-oncogenic, mediating systemic toxicities via the encapsulation of chemotherapeutic drugs. Within metastatic Y79 EV samples, an estimated PL concentration of 0.026pM was detected. PL treatment significantly downregulated Y79 EV cargo of the oncogene MYCN transcript. Interestingly, non-PL-treated Y79 cells incubated with EVs from PL-treated cells exhibited significantly reduced cell growth. These findings indicate that in metastatic Y79 cells, PL exhibits potent anti-proliferation effects and oncogene downregulation. Importantly, PL is also incorporated into extracellular vesicles released from treated metastatic cells with measurable anti-cancer effects on target cells at a distance from the site of primary treatment. The use of PL in the treatment of metastatic retinoblastoma may reduce primary tumor proliferation and inhibit metastatic cancer activity systemically via extracellular vesicle circulation.
Collapse
Affiliation(s)
- Cui Shi
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; Biochemistry Doctoral Program, The Graduate School, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Kunhui Huang
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; Biochemistry Doctoral Program, The Graduate School, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - John Soto
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA
| | - Renuka Sankaran
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; Biochemistry Doctoral Program, The Graduate School, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Onyekwere Onwumere
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; Biology Doctoral Program, The Graduate School of the City University of New York, 365 5th Avenue, New York, NY 10016, USA
| | - Michael Young
- The Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | - Linda Einbond
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA
| | - Stephen Redenti
- Lehman College, the City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; Biochemistry Doctoral Program, The Graduate School, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; Biology Doctoral Program, The Graduate School of the City University of New York, 365 5th Avenue, New York, NY 10016, USA.
| |
Collapse
|
22
|
Liu XM, Li Z, Xie XR, Wang JQ, Qiao X, Qiao X, Xie CZ, Xu JY. Combination of DNA Damage, Autophagy, and ERK Inhibition: Novel Evodiamine-Inspired Multi-Action Pt(IV) Prodrugs with High-Efficiency and Low-Toxicity Antitumor Activity. J Med Chem 2023; 66:1852-1872. [PMID: 36715603 DOI: 10.1021/acs.jmedchem.2c01660] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Exploring multi-targeting chemotherapeutants with advantages over single-targeting agents and drug combinations is of great significance in drug discovery. Herein, we employed phytogenic evodiamine (EVO) and conventional Pt(II) drugs to design and synthesize multi-target EVO-Pt(IV) anticancer prodrugs (4-14). Among them, compound 10 exhibited a 118-fold enhancement in the IC50 value compared to cisplatin and low toxicity to normal cells. Further studies proved that 10 significantly enhanced intracellular Pt accumulation and DNA damage, perturbed mitochondrial membrane potential, inhibited cell migration and invasion, upregulated reactive oxygen species levels, and induced apoptosis and autophagic cell death. Molecular docking assay revealed that 10 fits perfectly into the extracellular signal-regulated protein kinase (ERK)-1 pocket, which was verified to produce profound ERK suppression. Most strikingly, compound 10 exhibited superior in vivo antitumor efficiency and effectively attenuated systemic toxicity. Our results emphasize that functionalizing platinum drugs with the multi-target EVO could generate synergistically excellent anticancer activity with low toxicity and decreased resistance, which may represent a brand-new cancer therapy modality.
Collapse
Affiliation(s)
- Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin-Ru Xie
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jia-Qian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Cheng-Zhi Xie
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
23
|
Szefler B, Czeleń P. Will the Interactions of Some Platinum (II)-Based Drugs with B-Vitamins Reduce Their Therapeutic Effect in Cancer Patients? Comparison of Chemotherapeutic Agents such as Cisplatin, Carboplatin and Oxaliplatin-A Review. Int J Mol Sci 2023; 24:ijms24021548. [PMID: 36675064 PMCID: PMC9862491 DOI: 10.3390/ijms24021548] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Pt (II) derivatives show anti-cancer activity by interacting with nucleobases of DNA, thus causing some spontaneous and non-spontaneous reactions. As a result, mono- and diaqua products are formed which further undergo complexation with guanine or adenine. Consequently, many processes are triggered, which lead to the death of the cancer cell. The theoretical and experimental studies confirm that such types of interactions can also occur with other chemical compounds. The vitamins from B group have a similar structure to the nucleobases of DNA and have aromatic rings with single-pair orbitals. Theoretical and experimental studies were performed to describe the interactions of B vitamins with Pt (II) derivatives such as cisplatin, oxaliplatin and carboplatin. The obtained results were compared with the values for guanine. Two levels of simulations were implemented at the theoretical level, namely, B3LYP/6-31G(d,p) with LANL2DZ bases set for platinum atoms and MN15/def2-TZVP. The polarizable continuum model (IEF-PCM preparation) and water as a solvent were used. UV-Vis spectroscopy was used to describe the drug-nucleobase and drug-B vitamin interactions. Values of the free energy (ΔGr) show spontaneous reactions with mono- and diaqua derivatives of cisplatin and oxaliplatin; however, interactions with diaqua derivatives are more preferable. The strength of these interactions was also compared. Carboplatin products have the weakest interaction with the studied structures. The presence of non-covalent interactions was demonstrated in the tested complexes. A good agreement between theory and experiment was also demonstrated.
Collapse
|
24
|
Zheng Y, Li Z, Wang Y, Chen W, Lin Y, Guo J, Ye G. CircRNA: A new class of targets for gastric cancer drug resistance therapy. Pathol Oncol Res 2023; 29:1611033. [PMID: 37065861 PMCID: PMC10097900 DOI: 10.3389/pore.2023.1611033] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/22/2023] [Indexed: 04/18/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. Patients with advanced GC need palliative care to ensure survival. This includes the use of chemotherapy agents, such as cisplatin, 5-fluorouracil, oxaliplatin, paclitaxel, and pemetrexed, as well as targeted agents. However, the emergence of drug resistance evidence in poor patient outcomes and poor prognosis is a motivation to determine the specific mechanism of drug resistance. Interestingly, circular RNAs (circRNAs) play an important part in the carcinogenesis and progression of GC and are involved in GC drug resistance. This review systematically summarizes the functions and mechanisms of circRNAs underlying GC drug resistance, especially chemoresistance. It also emphasizes that circRNAs can serve as promising targets for improving drug resistance and therapeutic efficacy.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Institute of Digestive Diseases of Ningbo University, Ningbo, China
| | - Zhe Li
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Institute of Digestive Diseases of Ningbo University, Ningbo, China
| | - Yao Wang
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Institute of Digestive Diseases of Ningbo University, Ningbo, China
| | - Wanjiao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yifan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Institute of Digestive Diseases of Ningbo University, Ningbo, China
| | - Junming Guo
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Institute of Digestive Diseases of Ningbo University, Ningbo, China
- *Correspondence: Junming Guo, ; Guoliang Ye,
| | - Guoliang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Institute of Digestive Diseases of Ningbo University, Ningbo, China
- *Correspondence: Junming Guo, ; Guoliang Ye,
| |
Collapse
|
25
|
Al-Khdour MS, Khabour OF, Al-Eitan LN, Alzoubi KH. Genotoxicity of nedaplatin in cultured lymphocytes: modulation by vitamin E. Drug Chem Toxicol 2023; 46:176-180. [PMID: 34965829 DOI: 10.1080/01480545.2021.2015369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nedaplatin is a chemotherapeutic agent used widely in cancer therapy. Nedaplatin has been shown to cause DNA damage to cells via the induction of oxidative stress. Vitamin E (Vit E) has an anti-mutagenic activity that can protect cells from DNA damaging agents. The objective of this study is to examine the genotoxic and cytotoxic effects of nedaplatin in human cultured lymphocytes. In addition, modulation of such effects by Vit E was also examined. The frequencies of sister chromatid exchange (SCE) and chromosomal aberrations (CAs) were used as an indicator for genotoxicity. The mitotic and proliferative indices were used to examine the cytotoxic effects of nedaplatin. The results showed that nedaplatin significantly elevated SCE and CA frequencies in human lymphocytes (p ˂ 0.01). The increases in the frequencies of SCE and CA caused by nedaplatin were lowered by pretreatment treatment with Vit E (p < 0.05). Nedaplatin significantly lowered mitotic index but Vit E pretreatment did not modulate this effect. These results suggest that Vit E has the potential to ameliorate the genotoxicity of nedaplatin in cultured lymphocytes.
Collapse
Affiliation(s)
- Muntaha S Al-Khdour
- Department of Applied Biological Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Laith N Al-Eitan
- Department of Applied Biological Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
26
|
Papo TR, Jaganyi D, Mambanda A. Substitution reactions of cis-platinum(II) complexes containing bidentate N,N-donor pyridinecarboxamide ligands with different substituents. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2149327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tshephiso R. Papo
- School of Chemistry and Physics, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Deogratius Jaganyi
- School of Pure and Applied Sciences, Mount Kenya University, Thika, Kenya
- Department of Chemistry, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Allen Mambanda
- School of Chemistry and Physics, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| |
Collapse
|
27
|
Xie FL, Wang Y, Zhu JW, Xu HH, Guo QF, Wu Y, Liu SH. Anticancer mechanism studies of iridium(III) complexes inhibiting osteosarcoma HOS cells proliferation. J Inorg Biochem 2022; 237:112011. [PMID: 36252336 DOI: 10.1016/j.jinorgbio.2022.112011] [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: 03/25/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 01/18/2023]
Abstract
Three iridium (III) polypyridine complexes [Ir(bzq)2(maip)](PF6) (Ir1,bzq = benzo[h]quinoline, maip = 3-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline), [Ir(bzq)2(apip)](PF6) (Ir2, apip = 2-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ir(bzq)2(paip)](PF6) (Ir3, paip = 4-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline) were synthesized and characterized. The cytotoxic activities of the three complexes against human osteosarcoma HOS, U2OS, MG63 and normal LO2 cells were evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) method. The results showed that Ir1-3 exhibited moderate antitumor activity against HOS with IC50 of 21.8 ± 0. 4 μM,10.5 ± 1.8 μM and 7.4 ± 0.4 μM, respectively. We found that Ir1-3 can effectively inhibit HOS cells growth and blocked the cell cycle at the G0/G1 phase. Further studies revealed that complexes can increase intracellular reactive oxygen species (ROS) and Ca2+, which accompanied by mitochondria-mediated intrinsic apoptosis pathway. In addition, autophagy was also investigated. Taken together, the complexes induce HOS apoptosis through a ROS-mediated mitochondrial dysfunction pathway and inhibition of the PI3K (phosphatidylinositol 3-kinase)/AKT (protein kinase B)/mTOR (mammalian target of rapamycin) signaling pathway. This study provides useful help for understanding the anticancer mechanism of iridium (III) complexes toward osteosarcoma treatment.
Collapse
Affiliation(s)
- Fu-Li Xie
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China
| | - Yan Wang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China
| | - Jian-Wei Zhu
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China
| | - Hui-Hua Xu
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China
| | - Qi-Feng Guo
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China.
| | - Yong Wu
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China.
| | - Si-Hong Liu
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China; Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, PR China.
| |
Collapse
|
28
|
Kim JH, Ofori S, Tagmount A, Vulpe CD, Awuah SG. Genome-wide CRISPR Screen Reveal Targets of Chiral Gold(I) Anticancer Compound in Mammalian Cells. ACS OMEGA 2022; 7:39197-39205. [PMID: 36340096 PMCID: PMC9631916 DOI: 10.1021/acsomega.2c05166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/06/2022] [Indexed: 06/09/2023]
Abstract
Metal-based drugs, such as cisplatin and auranofin, are used for the treatment of cancer and rheumatoid arthritis, respectively. Auranofin and other gold-derived compounds have been shown to possess anticancer, anti-inflammatory, antimicrobial, and antiparasitic activity in preclinical and clinical trials. Unlike platinum agents which are known to target DNA, the target of gold is not well elucidated. To better understand the targets and effects of gold agents in mammalian cells, we used a targeted CRISPR (ToxCRISPR) screen in K562 cancer cells to identify genes that modulate cellular sensitivity to gold. We synthesized a novel chiral gold(I) compound, JHK-21, with potent anticancer activity. Among the most sensitizing hits were proteins involved in mitochondrial carriers, mitochondrial metabolism, and oxidative phosphorylation. Further analysis revealed that JHK-21 induced inner mitochondria membrane dysfunction and modulated ATP-binding cassette subfamily member C (ABCC1) function in a manner distinct from auranofin. Characterizing the therapeutic effects and toxicities of metallodrugs in mammalian cells is of growing interest to guide future drug discovery, and cellular and preclinical/clinical studies.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel Ofori
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Abderrahmane Tagmount
- Department
of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Chris D. Vulpe
- Department
of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Samuel G. Awuah
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- Markey
Cancer Center, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
29
|
El-Sheekh MM, Nassef M, Bases E, Shafay SE, El-Shenody R. Antitumor immunity and therapeutic properties of marine seaweeds-derived extracts in the treatment of cancer. Cancer Cell Int 2022; 22:267. [PMID: 35999584 PMCID: PMC9396856 DOI: 10.1186/s12935-022-02683-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022] Open
Abstract
Marine seaweeds are important sources of drugs with several pharmacological characteristics. The present study aims to evaluate the antitumor and antitumor immunological potentials of the extracts from the brown alga Padina pavonica and the red alga Jania rubens, inhibiting the Egyptian marine coasts. Hep-G2 cell lines were used for assessment of the antitumor efficacy of Padina pavonica and Jania rubens extracts in vitro, while Ehrlich ascites carcinoma (EAC) cells were applied to gain more antitumor immunity and antitumor insights of P. pavonica and J. rubens extracts in vivo. In vitro antitumor potentials of P. pavonica and J. rubens extracts were analyzed against human liver cancer Hep-G2 cells by MTT and trypan blue exclusion assays. In vivo antitumor immunological potentials of P. pavonica and J. rubens extracts at low, high, and prophylactic doses were analyzed by blood counting and flow cytometry in mice challenged with Ehrlich ascites carcinoma (EAC) cells. In vitro results revealed that P. pavonica and J. rubens extracts caused significant decreases in the number and viability of Hep-G2 cells in a dose-dependent manner as compared to untreated Hep-G2 cells or Cisplatin®-treated Hep-G2 cells. In vivo findings showed that P. pavonica and J. rubens extracts at low, high, and prophylactic doses significantly reduced the number and viability of EAC tumor cells accompanied by increases in EAC apoptosis compared to naïve EAC mouse. Additionally, P. pavonica and J. rubens extracts at low and prophylactic doses remarkably increased both the total WBC count and the relative numbers of lymphocytes and decreased the relative numbers of neutrophils and monocytes. Flow cytometric analysis showed that P. pavonica and J. rubens extracts at the treatment and the prophylactic doses resulted in a significant increase in the phenotypic expressions of CD4+ T, CD8+ T, and CD335 cells compared to naïve EAC mouse. Overall, both extracts P. pavonica and J. rubens possess potential antitumor and antitumor immunological effects with less toxicity, opening new approaches for further studies of the chemical and biological mechanisms behind these effects.
Collapse
Affiliation(s)
- Mostafa M El-Sheekh
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed Nassef
- Zoology Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Eman Bases
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Shimaa El Shafay
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Rania El-Shenody
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
30
|
Identification of Malignant Cell Populations Associated with Poor Prognosis in High-Grade Serous Ovarian Cancer Using Single-Cell RNA Sequencing. Cancers (Basel) 2022; 14:cancers14153580. [PMID: 35892844 PMCID: PMC9331511 DOI: 10.3390/cancers14153580] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Ovarian cancer has a high recurrence rate (~75%), and tumor heterogeneity is associated with such tumor recurrence. However, it is still poorly understood in ovarian cancer. To reveal tumor heterogeneity, we performed single-cell RNA sequencing (RNA-seq) of serous ovarian cancer cells from four different patients: two with primary carcinoma, one with recurrent carcinoma, and one with carcinoma treated with interval debulking surgery. As a result, we found two malignant tumor cell subtypes associated with poor prognosis. One malignant population included the earliest cancer cells and cancer stem-like cells. SLC3A1 and PEG10 were identified as the marker genes of cancer-initiating cells. The other malignant population expressing CA125 (MUC16), the most common biomarker for ovarian cancer, is associated with a decrease in the number of tumor-infiltrating cytotoxic T lymphocytes (CTLs). Our findings will offer new markers for diagnosis and choosing treatments targeting the malignant populations in ovarian cancer. Abstract To reveal tumor heterogeneity in ovarian cancer, we performed single-cell RNA sequencing (RNA-seq). We obtained The Cancer Genome Atlas (TCGA) survival data and TCGA gene expression data for a Kaplan–Meier plot showing the association of each tumor population with poor prognosis. As a result, we found two malignant tumor cell subtypes associated with poor prognosis. Next, we performed trajectory analysis using scVelo and Monocle3 and cell–cell interaction analysis using CellphoneDB. We found that one malignant population included the earliest cancer cells and cancer stem-like cells. Furthermore, we identified SLC3A1 and PEG10 as the marker genes of cancer-initiating cells. The other malignant population expressing CA125 (MUC16) is associated with a decrease in the number of tumor-infiltrating cytotoxic T lymphocytes (CTLs). Moreover, cell–cell interaction analysis implied that interactions mediated by LGALS9 and GAS6, expressed by this malignant population, caused the CTL suppression. The results of this study suggest that two tumor cell populations, including a cancer-initiating cell population and a population expressing CA125, survive the initial treatment and suppress antitumor immunity, respectively, and are associated with poor prognosis. Our findings offer a new understanding of ovarian cancer heterogeneity and will aid in the development of diagnostic tools and treatments.
Collapse
|
31
|
Improvement of Kiteplatin Efficacy by a Benzoato Pt(IV) Prodrug Suitable for Oral Administration. Int J Mol Sci 2022; 23:ijms23137081. [PMID: 35806087 PMCID: PMC9266928 DOI: 10.3390/ijms23137081] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022] Open
Abstract
Kiteplatin, [PtCl2(cis-1,4-DACH)] (DACH = diaminocyclohexane), contains an isomeric form of the oxaliplatin diamine ligand trans-1R,2R-DACH and has been proposed as a valuable drug candidate against cisplatin- and oxaliplatin-resistant tumors, in particular, colorectal cancer. To further improve the activity of kiteplatin, it has been transformed into a Pt(IV) prodrug by the addition of two benzoato groups in the axial positions. The new compound, cis,trans,cis-[PtCl2(OBz)2(cis-1,4-DACH)] (1; OBz = benzoate), showed cytotoxic activity at nanomolar concentration against a wide panel of human cancer cell lines. Based on these very promising results, the investigation has been extended to the in vivo activity of compound 1 in a Lewis Lung Carcinoma (LLC) model and its suitability for oral administration. Compound 1 resulted to be remarkably stable in acidic conditions (pH 1.5 to mimic the stomach environment) undergoing a drop of the initial concentration to ~60% of the initial one only after 72 h incubation at 37 °C; thus resulting amenable for oral administration. Interestingly, in a murine model (2·106 LLC cells implanted i.m. into the right hind leg of 8-week old male and female C57BL mice), a comparable reduction of tumor mass (~75%) was observed by administering compound 1 by oral gavage and the standard drug cisplatin by intraperitoneal injection, thus indicating that, indeed, there is the possibility of oral administration for this dibenzoato prodrug of kiteplatin. Moreover, since the mechanism of action of Pt(IV) prodrugs involves an initial activation by chemical reduction to cytotoxic Pt(II) species, the reduction of 1 by two bioreductants (ascorbic acid/sodium ascorbate and glutathione) was investigated resulting to be rather slow (not complete after 120 h incubation at 37 °C). Finally, the neurotoxicity of 1 was evaluated using an in vitro assay.
Collapse
|
32
|
Karumban KS, Raut R, Gupta P, Muley A, Giri B, Kumbhakar S, Misra A, Maji S. Mononuclear cobalt(II) complexes with polypyridyl ligands: Synthesis, characterization, DNA interactions and in vitro cytotoxicity towards human cancer cells. J Inorg Biochem 2022; 233:111866. [DOI: 10.1016/j.jinorgbio.2022.111866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/28/2022] [Accepted: 05/17/2022] [Indexed: 02/02/2023]
|
33
|
Gou Y, Jia X, Hou LX, Deng JG, Huang GJ, Jiang HW, Yang F. Dithiocarbazate-Fe III, -Co III, -Ni II, and -Zn II Complexes: Design, Synthesis, Structure, and Anticancer Evaluation. J Med Chem 2022; 65:6677-6689. [PMID: 35446587 DOI: 10.1021/acs.jmedchem.1c02186] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-platinum-metal complexes show great potential as anticancer agents. Herein, a series of dithiocarbazate non-Pt-metal complexes, including [FeIII(L)2]·Cl·2H2O 1, [CoIII(L)2]·NO3·2.5H2O 2, [NiII(L)2] 3, and [ZnII(L)2] 4, have been designed and evaluated for their efficacy as antineoplastic agents. Among them, complex 2 exhibited higher anticancer efficacy than complexes 1, 3, 4, and cisplatin against several cancer cell lines. Hemolysis assays revealed that complex 2 showed comparable hemolysis with cisplatin. In vivo anticancer evaluations showed that complex 2 could retard tumor xenograft growth effectively with low systemic toxicity. Further studies revealed that complex 2 suppressed cancer cells by triggering multiple mechanisms involving the simultaneous inhibition of mitochondria and glycolytic bioenergetics. Overall, our study provides new insights into the anticancer mechanism of Co complexes, which can be used as a good strategy to overcome the flexibility of cancer cells to chemotherapy adaptation.
Collapse
Affiliation(s)
- Yi Gou
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Xiaoying Jia
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Norma University, Guilin 541004, Guangxi, China
| | - Li Xia Hou
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Jun Gang Deng
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Guo Jin Huang
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Hao Wen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
| | - Feng Yang
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China.,State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Norma University, Guilin 541004, Guangxi, China
| |
Collapse
|
34
|
Jiang M, Yang T, Chu Y, Zhang Z, Sun H, Liang H, Yang F. Design of a novel Pt(II) complex to reverse cisplatin-induced resistance in lung cancer via a multi-mechanism. Dalton Trans 2022; 51:5257-5270. [PMID: 35285843 DOI: 10.1039/d1dt03964d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In order to develop a novel platinum (Pt) complex aiming to overcome cisplatin resistance, we synthesised a series of novel Pt complexes (C1-C6). These Pt complexes displayed potent cytotoxicity activity against resistant lung cancer cells (A549cisR) in vitro and efficiently inhibited tumour growth in vivo. The Pt complexes can target DNA, lead to DNA platination and cause cell cycle arrest in the S phase, thus impeding precise DNA synthesis. C6, in particular, induced not only apoptosis but also lethal autophagy in A549cisR cells. In addition, these novel Pt complexes reversed cisplatin-induced resistance via inhibiting the expression of P-glycoprotein and decreasing the level of glutathione in A549cisR cells. Moreover, the ERK pathway was involved in C6-induced overcoming cisplatin resistance.
Collapse
Affiliation(s)
- Ming Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China. .,School of food and biochemical engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Tongfu Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Yong Chu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| |
Collapse
|
35
|
Wu H, Zhao X, Wang J, Jiang X, Cheng Y, He Y, Sun L, Zhang G. Circular RNA CDR1as Alleviates Cisplatin-Based Chemoresistance by Suppressing MiR-1299 in Ovarian Cancer. Front Genet 2022; 12:815448. [PMID: 35154259 PMCID: PMC8826532 DOI: 10.3389/fgene.2021.815448] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/17/2021] [Indexed: 11/30/2022] Open
Abstract
Cisplatin (CDDP) chemoresistance seriously affects the prognosis and survival of patients with ovarian cancer (OC). Previous research has shown that circular RNA CDR1as is biologically associated with a large number of cancers. However, the molecular mechanism underlying the role of CDR1as in CDDP chemoresistance in OC remains unclear. Here, we investigated the mechanism of CDR1as in CDDP-resistant OC. First, we employed bioinformatics analysis and quantitative real-time PCR (qRT-PCR) to determine the expression of CDR1as and related RNAs in CDDP-sensitive and -resistant OC tissues and cells. Then, functional experiments were used to determine cell proliferation, invasion, migration, and apoptosis in CDDP chemoresistance and parent OC cells in vitro. The effect of CDR1as in CDDP chemoresistance OC progression was tested in nude mice in vivo. Moreover, dual-luciferase assays and RNA immunoprecipitation (RIP) were performed to confirm the interactions of CDR1as and related RNAs. Finally, we used Western blotting to determine protein expression levels. Our findings interpret the underlying mechanisms of the CDR1as/miR-1299/PPP1R12B axis and shed light on the clinical applications for CDDP-chemoresistant OC.
Collapse
Affiliation(s)
- Han Wu
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xibo Zhao
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jing Wang
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xinyan Jiang
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yan Cheng
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yanan He
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Liyuan Sun
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guangmei Zhang
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
36
|
Jana A, Aher A, Brandao P, Bera P, Sharda S, Phadikar U, Manna SK, Mahapatra AK, Bera P. Evaluation of the anticancer activities with various ligand substituents in Co(II/III)-picolyl phenolate derivatives: synthesis, characterization, DFT, DNA cleavage, and molecular docking studies. Dalton Trans 2022; 51:2346-2363. [PMID: 35043134 DOI: 10.1039/d1dt02825a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The reactions between 2-(pyridine-2-ylmethoxy)-benzaldehyde (L) and various primary amines furnish tridentate (L1 to L3) and tetradentate (L4) chelating ligands. The choice of different primary amines in the condensation reaction incorporates the chiral carbon atom in L2 and L3. A series of mononuclear cobalt(II) complexes, [CoII(L1)(Cl)2] (1), [CoII(L2)(Cl)2]·CH3CN (2), [CoII(L3)(Cl)2] (3), and [CoIII(L4)(N3)2] (4) are synthesized in the pure crystalline state from the resulting solution of cobalt(II) chloride and/or azide and respective ligand. The new ligands and cobalt complexes are characterized using spectral (UV-Vis, 1H-NMR, IR, and HRMS), cyclovoltammetric, and DFT studies. The structure of L1, L2, and all four cobalt complexes are determined by single X-ray crystallography. Cytotoxic activity of the compounds is evaluated using three different tissues of origin e.g., U-937 (histiocytic lymphoma), HEK293T (embryonic kidney), and A549 (lung carcinoma). The cobalt complexes are more active than the corresponding ligands against U-937 and HEK293T. The MTT assay demonstrates that the cobalt compounds are more effective anticancer agents against U-937 cancer cells than HEK293T and A549. The toxicity order, 1 (7.2 ± 0.3 μM) > 3 (11.4 ± 0.6 μM) > 2 (12 ± 0.1 μM) > 4 (29 ± 1 μM) is observed against U-937 cancer cells. All the compounds induce cell death through an apoptosis mechanism and all are ineffective against PBMCs. The mechanism of activity against U937 cancer cells involves caspase-3 activation and two different mitogen-activated protein kinases attesting the programmed cell death. Among the compounds, complexes 1, 2, and 3 show DNA cleavage activity both in oxidizing (H2O2) and reducing (GSH) environments. The mechanistic study reveals that singlet oxygen (1O2) is the major species involved in DNA cleavage. The absolute chemical hardness values of the ligands and 4 are relatively higher than 1, 2, and 3, which tacitly support the DNA cleavage experiment. The docking result indicates that the compounds under investigation strongly interact with DNA base pairs through a variety of interactions which attests successfully to the experimental results. A structure-activity relationship has been drawn to correlate the variation of antitumor activity with ligand conformations.
Collapse
Affiliation(s)
- Abhimanyu Jana
- Post Graduate Department of Chemistry, Panskura Banamali College (Autonomous) (Vidyasagar University), Panskura R. S, Midnapore (East), West Bengal, 721152, India.
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, West Bengal, 711103, India
| | - Abhishek Aher
- Centre for DNA Fingerprinting & Diagnostics (CDFD), Hyderabad, 500 039, Telangana, India
- Graduate Studies, Regional Centre for Biotechnology, Faridabad, Haryana-121001, India
| | - Paula Brandao
- Department of Chemistry, CICECO, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pradip Bera
- Post Graduate Department of Chemistry, Panskura Banamali College (Autonomous) (Vidyasagar University), Panskura R. S, Midnapore (East), West Bengal, 721152, India.
- Department of Chemistry, Kandi Raj College, Murshidabad, West Bengal, 742137, India
| | - Saphy Sharda
- Centre for DNA Fingerprinting & Diagnostics (CDFD), Hyderabad, 500 039, Telangana, India
- Graduate Studies, Regional Centre for Biotechnology, Faridabad, Haryana-121001, India
| | - Ujjwal Phadikar
- Post Graduate Department of Chemistry, Panskura Banamali College (Autonomous) (Vidyasagar University), Panskura R. S, Midnapore (East), West Bengal, 721152, India.
| | - Sunil Kumar Manna
- Centre for DNA Fingerprinting & Diagnostics (CDFD), Hyderabad, 500 039, Telangana, India
- Adjunct Faculty, Regional Centre for Biotechnology, Faridabad, Haryana, 121001, India
| | - Ajit Kumar Mahapatra
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, West Bengal, 711103, India
| | - Pulakesh Bera
- Post Graduate Department of Chemistry, Panskura Banamali College (Autonomous) (Vidyasagar University), Panskura R. S, Midnapore (East), West Bengal, 721152, India.
| |
Collapse
|
37
|
Margiotta N, Pacifico C, Saltarella T, Natile G, Intini FP. Synthesis and characterization of new platinum(II) complexes with cyclic iminoether-type ligands having the azomethine group out of cycle. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2021.120655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
38
|
Hou Y, Gan T, Fang T, Zhao Y, Luo Q, Liu X, Qi L, Zhang Y, Jia F, Han J, Li S, Wang S, Wang F. OUP accepted manuscript. Nucleic Acids Res 2022; 50:3070-3082. [PMID: 35258624 PMCID: PMC8989551 DOI: 10.1093/nar/gkac151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 11/23/2022] Open
Abstract
Pyridostatin (PDS) is a well-known G-quadruplex (G4) inducer and stabilizer, yet its target genes have remained unclear. Herein, applying MS proteomics strategy, we revealed PDS significantly downregulated 22 proteins but upregulated 16 proteins in HeLa cancer cells, of which the genes both contain a number of G4 potential sequences, implying that PDS regulation on gene expression is far more complicated than inducing/stabilizing G4 structures. The PDS-downregulated proteins consequently upregulated 6 proteins to activate cyclin and cell cycle regulation, suggesting that PDS itself is not a potential anticancer agent, at least toward HeLa cancer cells. Importantly, SUB1, which encodes human positive cofactor and DNA lesion sensor PC4, was downregulated by 4.76-fold. Further studies demonstrated that the downregulation of PC4 dramatically promoted the cytotoxicity of trans-[PtCl2(NH3)(thiazole)] (trans-PtTz) toward HeLa cells to a similar level of cisplatin, contributable to retarding the repair of 1,3-trans-PtTz crosslinked DNA lesion mediated by PC4. These findings not only provide new insights into better understanding on the biological functions of PDS but also implicate a strategy for the rational design of novel multi-targeting platinum anticancer drugs via conjugation of PDS as a ligand to the coordination scaffold of transplatin for battling drug resistance to cisplatin.
Collapse
Affiliation(s)
- Yinzhu Hou
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
- College of Chemical Science, University of Chinese Academy of Sciences, Yuquan Road, Shijingshan District, 100049 Beijing, P.R. China
| | - Tieliang Gan
- College of Chemical Science, University of Chinese Academy of Sciences, Yuquan Road, Shijingshan District, 100049 Beijing, P.R. China
| | - Tiantian Fang
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
| | - Yao Zhao
- Correspondence may also be addressed to Yao Zhao. Tel: +86 010 62529069;
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
- College of Chemical Science, University of Chinese Academy of Sciences, Yuquan Road, Shijingshan District, 100049 Beijing, P.R. China
| | - Xingkai Liu
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
- College of Chemical Science, University of Chinese Academy of Sciences, Yuquan Road, Shijingshan District, 100049 Beijing, P.R. China
| | - Luyu Qi
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
- College of Chemical Science, University of Chinese Academy of Sciences, Yuquan Road, Shijingshan District, 100049 Beijing, P.R. China
| | - Yanyan Zhang
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
| | - Feifei Jia
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
| | - Juanjuan Han
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
| | - Shumu Li
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; National Centre for Mass Spectrometry in Beijing, Institute of Chemistry, Chinese Academy of Sciences, No. 2 Zhongguancun North First Street, Haidian District, 100190 Beijing, P.R. China
| | - Shijun Wang
- Correspondence may also be addressed to Shijun Wang. Tel: +86 0531 89628750;
| | - Fuyi Wang
- To whom correspondence should be addressed. Tel: +86 010 62529069;
| |
Collapse
|
39
|
Gao Y, Kabotyanski EB, Shepherd JH, Villegas E, Acosta D, Hamor C, Sun T, Montmeyor-Garcia C, He X, Dobrolecki LE, Westbrook TF, Lewis MT, Hilsenbeck SG, Zhang XHF, Perou CM, Rosen JM. Tumor suppressor PLK2 may serve as a biomarker in triple-negative breast cancer for improved response to PLK1 therapeutics. CANCER RESEARCH COMMUNICATIONS 2021; 1:178-193. [PMID: 35156101 PMCID: PMC8827906 DOI: 10.1158/2767-9764.crc-21-0106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Polo-like kinase (PLK) family members play important roles in cell cycle regulation. The founding member PLK1 is oncogenic and preclinically validated as a cancer therapeutic target. Paradoxically, frequent loss of chromosome 5q11-35 which includes PLK2 is observed in basal-like breast cancer. In this study, we found that PLK2 was tumor suppressive in breast cancer, preferentially in basal-like and triple-negative breast cancer (TNBC) subtypes. Knockdown of PLK1 rescued phenotypes induced by PLK2-loss both in vitro and in vivo. We also demonstrated that PLK2 directly interacted with PLK1 at prometaphase through the kinase but not the polo-box domains of PLK2, suggesting PLK2 functioned at least partially through the interaction with PLK1. Furthermore, an improved treatment response was seen in both Plk2-deleted/low mouse preclinical and PDX TNBC models using the PLK1 inhibitor volasertib alone or in combination with carboplatin. Re-expression of PLK2 in an inducible PLK2-null mouse model reduced the therapeutic efficacy of volasertib. In summary, this study delineates the effects of chromosome 5q loss in TNBC that includes PLK2, the relationship between PLK2 and PLK1, and how this may render PLK2-deleted/low tumors more sensitive to PLK1 inhibition in combination with chemotherapy.
Collapse
Affiliation(s)
- Yang Gao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Elena B. Kabotyanski
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | | | - Deanna Acosta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Clark Hamor
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Tingting Sun
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | | | - Xiaping He
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lacey E. Dobrolecki
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Thomas F. Westbrook
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Michael T. Lewis
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Susan G. Hilsenbeck
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xiang H.-F. Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- McNair Medical Institute, Baylor College of Medicine, Houston, Texas
| | - Charles M. Perou
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey M. Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Corresponding Author: Jeffrey M. Rosen, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030. Phone: 832-215-9503; E-mail:
| |
Collapse
|
40
|
Hain BA, Xu H, Waning DL. Loss of REDD1 prevents chemotherapy-induced muscle atrophy and weakness in mice. J Cachexia Sarcopenia Muscle 2021; 12:1597-1612. [PMID: 34664403 PMCID: PMC8718092 DOI: 10.1002/jcsm.12795] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chemotherapy is an essential treatment to combat solid tumours and mitigate metastasis. Chemotherapy causes side effects including muscle wasting and weakness. Regulated in Development and DNA Damage Response 1 (REDD1) is a stress-response protein that represses the mechanistic target of rapamycin (mTOR) in complex 1 (mTORC1), and its expression is increased in models of muscle wasting. The aim of this study was to determine if deletion of REDD1 is sufficient to attenuate chemotherapy-induced muscle wasting and weakness in mice. METHODS C2C12 myotubes were treated with carboplatin, and changes in myotube diameter were measured. Protein synthesis was measured by puromycin incorporation, and REDD1 mRNA and protein expression were analysed in myotubes treated with carboplatin. Markers of mTORC1 signalling were measured by western blot. REDD1 global knockout mice and wild-type mice were treated with a single dose of carboplatin and euthanized 7 days later. Body weight, hindlimb muscle weights, forelimb grip strength, and extensor digitorum longus whole muscle contractility were measured in all groups. Thirty minutes prior to euthanasia, mice were injected with puromycin to measure puromycin incorporation in skeletal muscle. RESULTS C2C12 myotube diameter was decreased at 24 (P = 0.0002) and 48 h (P < 0.0001) after carboplatin treatment. Puromycin incorporation was decreased in myotubes treated with carboplatin for 24 (P = 0.0068) and 48 h (P = 0.0008). REDD1 mRNA and protein expression were increased with carboplatin treatment (P = 0.0267 and P = 0.0015, respectively), and this was accompanied by decreased phosphorylation of Akt T308 (P < 0.0001) and S473 (P = 0.0006), p70S6K T389 (P = 0.0002), and 4E-binding protein 1 S65 (P = 0.0341), all markers of mTORC1 activity. REDD1 mRNA expression was increased in muscles from mice treated with carboplatin (P = 0.0295). Loss of REDD1 reduced carboplatin-induced body weight loss (P = 0.0013) and prevented muscle atrophy in mice. REDD1 deletion prevented carboplatin-induced decrease of protein synthesis (P = 0.7626) and prevented muscle weakness. CONCLUSIONS Carboplatin caused loss of body weight, muscle atrophy, muscle weakness, and inhibition of protein synthesis. Loss of REDD1 attenuates muscle atrophy and weakness in mice treated with carboplatin. Our study illustrates the importance of REDD1 in the regulation of muscle mass with chemotherapy treatment and may be an attractive therapeutic target to combat cachexia.
Collapse
Affiliation(s)
- Brian A Hain
- Dept. of Cellular and Molecular Physiology, The Penn State College of Medicine, Hershey, PA, USA
| | - Haifang Xu
- Dept. of Cellular and Molecular Physiology, The Penn State College of Medicine, Hershey, PA, USA
| | - David L Waning
- Dept. of Cellular and Molecular Physiology, The Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
41
|
Boonmee A, Benjaskulluecha S, Kueanjinda P, Wongprom B, Pattarakankul T, Palaga T. The chemotherapeutic drug carboplatin affects macrophage responses to LPS and LPS tolerance via epigenetic modifications. Sci Rep 2021; 11:21574. [PMID: 34732786 PMCID: PMC8566489 DOI: 10.1038/s41598-021-00955-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/18/2021] [Indexed: 12/22/2022] Open
Abstract
Following re-exposure to lipopolysaccharide (LPS), macrophages exhibit an immunosuppressive state known as LPS tolerance, which is characterized by repressed proinflammatory cytokine production. LPS-induced tolerance in macrophages is mediated in part by epigenetic changes. Carboplatin, an anticancer chemotherapeutic drug, exerts its effect by inhibiting DNA replication and transcription, as well as through epigenetic modifications. Through an unbiased screen, we found that carboplatin rescued TNF-α and IL-6 production in LPS-tolerant macrophages. Transcriptomic analysis and gene set enrichment analyses revealed that p53 was one of the most significantly upregulated hallmarks in both LPS-primed and LPS-tolerant macrophages in the presence of carboplatin, while E2F and G2/M were the most negatively regulated hallmarks. Heterochromatin protein 1 (HP1-α), which is associated with gene silencing, was significantly reduced in carboplatin-treated LPS-tolerant macrophages at the mRNA and protein levels. Dynamic changes in the mRNA level of genes encoding H3K9me3 methyltransferases, setdb2, kdm4d, and suv39h1 were induced in the presence of carboplatin in LPS-tolerant macrophages. Taken together, we provide evidence that carboplatin treatment interferes with proinflammatory cytokine production during the acute LPS response and LPS tolerance in macrophages, possibly via H3K9me3 modification.
Collapse
Affiliation(s)
- Atsadang Boonmee
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Salisa Benjaskulluecha
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
- Inter-Disciplinary Graduate Program in Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Patipark Kueanjinda
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Benjawan Wongprom
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Thitiporn Pattarakankul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
42
|
Chen F, Xu G, Tian W, Gou S. Breakdown of chemo-immune resistance by a TDO2-targeted Pt(IV) prodrug via attenuating endogenous Kyn-AhR-AQP4 metabolic circuity and TLS-promoted genomic instability. Biochem Pharmacol 2021; 193:114785. [PMID: 34562469 DOI: 10.1016/j.bcp.2021.114785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022]
Abstract
A tryptophan-2,3-dioxygenase 2 (TDO2)-targeted Pt(IV) prodrug, DN604-TDOi, was designed to prove that the multi-action compound could overcome drug resistance and relieve immunosuppression via introducing a TDO2 inhibitor to the axial position of a six-coordinate Pt(IV) hybrid. Several in vitro biological studies on cisplatin-resistant NSCLC cancer cells suggested that TDO2-targeted Pt(IV) prodrug could combat cisplatin resistance via influencing TDO2-kynurenine (Kyn)-aryl hydrocarbon receptor (AhR)-Aquaporin-4 (AQP4) metabolic circuity and AhR-human DNA polymerase (hpol) κ-induced translesion DNA synthesis (TLS) genomic instability, which are positive in drug-resistant human tumors associated with malignant progression and poor survival. Remarkably, we observed that DN604-TDOi could inhibit TDO2-mediated constitutive Kyn-AhR-AQP4 signaling pathway and suppress hpol κ expression, leading to potential decrease of cell motility and genomic instability in A549/cDDP cells. It was confirmed that TDO2-targeted Pt(IV) prodrug could harness Kyn-AhR-AQP4 metabolic circuitry and TLS genomic instability, exerting antitumor effects in C57BL6 but not TDO2-/- mice. Moreover, the Pt(IV) prodrug improved the intratumoral infiltration of Teff cells and reduced the recruitment of Treg cells. The results provided compelling preclinical evidence that TDO2-targeted Pt(IV) prodrug could abrogate immune chemotherapeutic resistance via decaying TDO2-mediated Kyn-AhR-AQP4 immunosuppression and AhR-hpol κ-induced TLS genomic instability, underscoring the development of a novel Pt(IV)-based candidate as a potent immunotherapeutic agent for chemo-immune resistance prevention.
Collapse
Affiliation(s)
- Feihong Chen
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Gang Xu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Wenyuan Tian
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
43
|
Weiner F, Schille JT, Hein JI, Wu XF, Beller M, Junghanß C, Murua Escobar H, Nolte I. Evaluation of combination protocols of the chemotherapeutic agent FX-9 with azacitidine, dichloroacetic acid, doxorubicin or carboplatin on prostate carcinoma cell lines. PLoS One 2021; 16:e0256468. [PMID: 34432846 PMCID: PMC8386839 DOI: 10.1371/journal.pone.0256468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/06/2021] [Indexed: 11/25/2022] Open
Abstract
The isoquinolinamine FX-9 is a novel potential chemotherapeutic agent showing antiproliferative effects against hematologic and prostate cancer cell lines such as B- and T-acute lymphoblastic leukemia and prostate cancer (PC) of different species. Interestingly, FX-9 shows no hemolytic activity and low toxicity in benign adherent cells. The detailed FX-9 molecular mode of action is currently not fully understood. But application on neoplastic cells induces pro-apoptotic and antimitotic effects. Canine prostate cancer (cPC) represents a unique spontaneous occurring animal model for human androgen-independent PC. Human androgen-independent PC as well as cPC are currently not satisfactorily treatable with chemotherapeutic protocols. Accordingly, the evaluation of novel agent combinations bears significant potential for identifying novel treatment strategies. In this study, we combined FX-9 with the currently approved therapeutic agents doxorubicin, carboplatin, the demethylating substance azacitidine as well as further potentially antitumorigenic agents such as dichloroacetic acid (DCA) in order to evaluate the respective synergistic potential. The combinations with 1–5 μM FX-9 were evaluated regarding the effect after 72 hours on cell viability, cell count and apoptotic/necrotic cells in two human prostate cancer cell lines (LNCaP, PC-3) and a canine prostate cancer cell line (Adcarc1258) representing androgen-dependent and -independent PC/cPC forms. FX-9 in combination with azacitidine decreases cell viability and increases cell death with positive Bliss values. Furthermore, this decreases the cell count with neutral Bliss values on PC-3. Carboplatin in combination with FX-9 reduces cell viability with a neutral Bliss value and increases cell death on LNCaP with calculated positive Bliss values. DCA or doxorubicin in combination with FX-9 do not show synergistic or additive effects on the cell viability. Based on these results, azacitidine or carboplatin in combination with FX-9 offers synergistic/additive efficacy against prostate adenocarcinoma cell lines in vitro. The beneficial effects of both combinations are worth further investigation.
Collapse
Affiliation(s)
- Franziska Weiner
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, University of Rostock, Rostock, Germany
| | - Jan Torben Schille
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, University of Rostock, Rostock, Germany
| | - Jens Ingo Hein
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Xiao-Feng Wu
- Leibniz Institute for Catalysis, Rostock, Germany
| | | | - Christian Junghanß
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, University of Rostock, Rostock, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, University of Rostock, Rostock, Germany
- * E-mail:
| | - Ingo Nolte
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
44
|
Valente A, Podolski-Renić A, Poetsch I, Filipović N, López Ó, Turel I, Heffeter P. Metal- and metalloid-based compounds to target and reverse cancer multidrug resistance. Drug Resist Updat 2021; 58:100778. [PMID: 34403910 DOI: 10.1016/j.drup.2021.100778] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Drug resistance remains the major cause of cancer treatment failure especially at the late stage of the disease. However, based on their versatile chemistry, metal and metalloid compounds offer the possibility to design fine-tuned drugs to circumvent and even specifically target drug-resistant cancer cells. Based on the paramount importance of platinum drugs in the clinics, two main areas of drug resistance reversal strategies exist: overcoming resistance to platinum drugs as well as multidrug resistance based on ABC efflux pumps. The current review provides an overview of both aspects of drug design and discusses the open questions in the field. The areas of drug resistance covered in this article involve: 1) Altered expression of proteins involved in metal uptake, efflux or intracellular distribution, 2) Enhanced drug efflux via ABC transporters, 3) Altered metabolism in drug-resistant cancer cells, 4) Altered thiol or redox homeostasis, 5) Altered DNA damage recognition and enhanced DNA damage repair, 6) Impaired induction of apoptosis and 7) Altered interaction with the immune system. This review represents the first collection of metal (including platinum, ruthenium, iridium, gold, and copper) and metalloid drugs (e.g. arsenic and selenium) which demonstrated drug resistance reversal activity. A special focus is on compounds characterized by collateral sensitivity of ABC transporter-overexpressing cancer cells. Through this approach, we wish to draw the attention to open research questions in the field. Future investigations are warranted to obtain more insights into the mechanisms of action of the most potent compounds which target specific modalities of drug resistance.
Collapse
Affiliation(s)
- Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Isabella Poetsch
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nenad Filipović
- Department of Chemistry and Biochemistry, Faculty of Agriculture, University of Belgrade, Belgrade, Serbia
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
45
|
Nabiyeva T, Roufosse B, Odachowski M, Baumgartner J, Marschner C, Verma AK, Blom B. Osmium Arene Germyl, Stannyl, Germanate, and Stannate Complexes as Anticancer Agents. ACS OMEGA 2021; 6:19252-19268. [PMID: 34337263 PMCID: PMC8320079 DOI: 10.1021/acsomega.1c02665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
Herein, we describe the synthesis, full spectroscopic characterization, DFT (density functional theory) calculations, and single-crystal X-ray diffraction analyses of a series of osmium arene σ-germyl, germanate, σ-stannyl, and stannate complexes, along with their cytotoxic (anticancer) investigations. The known dimer complexes [OsCl2(η6-C6H6)]2 (1) and [OsCl2(η6-p-cymene)]2 (2) were reacted with PPh3 to form the known mononuclear complex [OsCl2(η6-p-cymene)(PPh3)] (3) and the new complex [OsCl2(η6-C6H6)(PPh3)] (6); complex 3 was reacted with GeCl2·(dioxane) and SnCl2 to afford, by insertion into the Os-Cl bond, the neutral σ-germyl and stannyl complexes [OsCl(η6-p-cymene)(PPh3)(GeCl3)] (7) and [OsCl(η6-p-cymene)(PPh3)(SnCl3)] (11), respectively, as a mixture of enantiomers. Similarly, the reaction of complex 6 with GeCl2·(dioxane) afforded [OsCl(η6-C6H6)(PPh3)(GeCl3)] (9). Complex 2, upon reaction with 1,1-bis(diphenylphosphino)methane (dppm), formed a mixture of [OsCl2(η6-p-cymene)(κ1-dppm)] (4) and [Os(η6-p-cymene)(κ2-dppm)Cl]+Cl- (5) when prepared in acetonitrile and a mixture of 4 and the dinuclear complex [[OsCl2(η6-p-cymene)]2(μ-dppm)] (0) when prepared in dichloromethane. By utilizing either isolated 4 or a mixture of 4 and 5, the synthesis of κ2-dppm germanate and stannate salts, [OsCl(η6-p-cymene)(κ2-dppm)]+GeCl3 - (8) and [OsCl(η6-p-cymene)(κ2-dppm)]+SnCl3 - (10), were accomplished via halide-abstracting reactions with GeCl2·(dioxane) or SnCl2, respectively. All resulting complexes were characterized by means of multinuclear NMR, FT-IR, ESI-MS, and UV/Vis spectroscopy. X-ray diffraction analyses of 4, 8, 9, 10, and 11 were performed and are reported. DFT studies (B3LYP, basis set LANL2DZ for Os, and def2-TZVPP for Sn, Ge, Cl, P, C, and H) were performed on complex 9 and the benzene analogue of complex 11, 11-benzene, to evaluate the structural changes and the effects on the frontier molecular orbitals arising from the substitution of Ge for Sn. Finally, complexes 3 and 7-11 were investigated for potential anticancer activities considering cell cytotoxicity and apoptosis assays against Dalton's lymphoma (DL) and Ehrlich ascites carcinoma (EAC) malignant cancer cell lines. The complexes were also tested against healthy peripheral blood mononuclear cells (PBMCs). All cell lines were also treated with the reference drug cisplatin to draw a comparison with the results obtained from the reported complexes. The study was further corroborated with in silico molecular interaction simulations and a pharmacokinetic study.
Collapse
Affiliation(s)
- Tomiris Nabiyeva
- Maastricht
Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan, 1, P.O.
Box 616, 6200 MD Maastricht, The Netherlands
| | - Basile Roufosse
- Maastricht
Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan, 1, P.O.
Box 616, 6200 MD Maastricht, The Netherlands
| | - Matylda Odachowski
- Maastricht
Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan, 1, P.O.
Box 616, 6200 MD Maastricht, The Netherlands
| | - Judith Baumgartner
- Institut
für Anorganische Chemie, Technische
Universität Graz, Stremayrgasse 9, A-8010 Graz, Austria
| | - Christoph Marschner
- Institut
für Anorganische Chemie, Technische
Universität Graz, Stremayrgasse 9, A-8010 Graz, Austria
| | - Akalesh Kumar Verma
- Department
of Zoology, Cell & Biochemical Technology Laboratory, Cotton University, Guwahati 781001, India
| | - Burgert Blom
- Maastricht
Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan, 1, P.O.
Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
46
|
Kuijpers T, Blom B. Homo and heterobimetallic palladium and platinum complexes bearing μ-diphosphane bridges involved in biological studies. Eur J Med Chem 2021; 223:113651. [PMID: 34214843 DOI: 10.1016/j.ejmech.2021.113651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 02/03/2023]
Abstract
Given the increasing reports of well-defined bimetallic molecular complexes as potential anticancer agents in the last decades, along with the prevalence of platinum in anticancer therapy, we report here a detailed survey of bimetallic platinum and palladium complexes investigated as potential anticancer agents. Specifically, we will concentrate on the synthesis, characterisation and biological (anticancer) studies of a sub-class of these agents, namely homo and heterobimetallic complexes bearing a bridging phosphane ligand of the type: [LnM1(μ-R2P(CH2)nPR2)M2Lm] (where M1 is platinum or palladium, M2 is any other transition metal, R = alkyl or aryl substituents, Ln or Lm are co-ligands, n = 1-6). We will review the in vitro and in vivo activities and any mechanistic anticancer studies of these complexes with a view of trying to delineate patterns in biological activity and structure-activity relationships (SAR). We do not include the review of bimetallic complexes in this class that have not undergone any anticancer testing, nor those that have been involved in other biological investigations unrelated to cancer studies.
Collapse
Affiliation(s)
- Talita Kuijpers
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan, 1, PO Box 616, 6200 MD, Maastricht, the Netherlands
| | - Burgert Blom
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan, 1, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| |
Collapse
|
47
|
Interference between copper transport systems and platinum drugs. Semin Cancer Biol 2021; 76:173-188. [PMID: 34058339 DOI: 10.1016/j.semcancer.2021.05.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/17/2021] [Indexed: 01/06/2023]
Abstract
Cisplatin, or cis-diamminedichloridoplatinum(II) cis-[PtCl2(NH3)2], is a platinum-based anticancer drug largely used for the treatment of various types of cancers, including testicular, ovarian and colorectal carcinomas, sarcomas, and lymphomas. Together with other platinum-based drugs, cisplatin triggers malignant cell death by binding to nuclear DNA, which appears to be the ultimate target. In addition to passive diffusion across the cell membrane, other transport systems, including endocytosis and some active or facilitated transport mechanisms, are currently proposed to play a pivotal role in the uptake of platinum-based drugs. In this review, an updated view of the current literature regarding the intracellular transport and processing of cisplatin will be presented, with special emphasis on the plasma membrane copper permease CTR1, the Cu-transporting ATPases, ATP7A and ATP7B, located in the trans-Golgi network, and the soluble copper chaperone ATOX1. Their role in eliciting cisplatin efficacy and their exploitation as pharmacological targets will be addressed.
Collapse
|
48
|
Hou L, Li H, Wang H, Ma D, Liu J, Ma L, Wang Z, Yang Z, Wang F, Xia H. The circadian clock gene PER2 enhances chemotherapeutic efficacy in nasopharyngeal carcinoma when combined with a targeted nanosystem. J Mater Chem B 2021; 8:5336-5350. [PMID: 32458942 DOI: 10.1039/d0tb00595a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Treatment failure occurs in more than 40% of advanced nasopharyngeal carcinoma (NPC) patients including local recurrence and distant metastasis due to chemoradioresistance. Circadian clock genes were identified as regulating cancer progression and chemoradiosensitivity in a time-dependent manner. A novel nanosystem can ensure the accumulation and controllable release of chemotherapeutic agents at the tumour site at a set time. In this study, we investigated the expression of circadian clock genes and identified that period circadian regulator 2 (PER2) as a tumour suppressor plays a key role in NPC progression. A label-free proteomic approach showed that PER2 overexpression can inhibit the ERK/MAPK pathway. The chemotherapeutic effect of PER2 overexpression was assessed in NPC together with the nanosystem comprising folic acid (FA), upconverting nanoparticles covalently coupled with Rose Bengal (UCNPs-RB), 10-hydroxycamptothecin (HCPT) and lipid-perfluorohexane (PFH) (FURH-PFH-NPs). PER2 overexpression combined with the targeted and controlled release of nanoagents elevated chemotherapeutic efficacy in NPC, which has potential application value for the chronotherapy of tumours.
Collapse
Affiliation(s)
- Li Hou
- Department of Otolaryngology, Head and Neck Surgery, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China and Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China.
| | - Hailiang Li
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China. and Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Haiyan Wang
- Department of Gynaecology, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Dede Ma
- Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Jing Liu
- Department of Otolaryngology, Head and Neck Surgery, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Liqiong Ma
- Department of Pathology, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Zhihua Wang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Zhihua Yang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| | - Faxuan Wang
- School of Public Health, Ningxia Medical University, Yin Chuan, 750004 Ningxia, P. R. China
| | - Hechun Xia
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China. and Department of Neurosurgery, General Hospital of Ningxia Medical University, Yin Chuan, 750004, Ningxia, P. R. China
| |
Collapse
|
49
|
Jeon J, Lee S, Kim H, Kang H, Youn H, Jo S, Youn B, Kim HY. Revisiting Platinum-Based Anticancer Drugs to Overcome Gliomas. Int J Mol Sci 2021; 22:ijms22105111. [PMID: 34065991 PMCID: PMC8151298 DOI: 10.3390/ijms22105111] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Although there are many patients with brain tumors worldwide, there are numerous difficulties in overcoming brain tumors. Among brain tumors, glioblastoma, with a 5-year survival rate of 5.1%, is the most malignant. In addition to surgical operations, chemotherapy and radiotherapy are generally performed, but the patients have very limited options. Temozolomide is the most commonly prescribed drug for patients with glioblastoma. However, it is difficult to completely remove the tumor with this drug alone. Therefore, it is necessary to discuss the potential of anticancer drugs, other than temozolomide, against glioblastomas. Since the discovery of cisplatin, platinum-based drugs have become one of the leading chemotherapeutic drugs. Although many studies have reported the efficacy of platinum-based anticancer drugs against various carcinomas, studies on their effectiveness against brain tumors are insufficient. In this review, we elucidated the anticancer effects and advantages of platinum-based drugs used in brain tumors. In addition, the cases and limitations of the clinical application of platinum-based drugs are summarized. As a solution to overcome these obstacles, we emphasized the potential of a novel approach to increase the effectiveness of platinum-based drugs.
Collapse
Affiliation(s)
- Jaewan Jeon
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea; (J.J.); (S.J.)
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea;
| | - Sunmi Jo
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea; (J.J.); (S.J.)
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: (B.Y.); (H.Y.K.); Tel.: +82-51-510-2264 (B.Y.); +82-51-797-3923 (H.Y.K.)
| | - Hae Yu Kim
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea
- Correspondence: (B.Y.); (H.Y.K.); Tel.: +82-51-510-2264 (B.Y.); +82-51-797-3923 (H.Y.K.)
| |
Collapse
|
50
|
Kim JH, Ofori S, Parkin S, Vekaria H, Sullivan PG, Awuah SG. Anticancer gold(iii)-bisphosphine complex alters the mitochondrial electron transport chain to induce in vivo tumor inhibition. Chem Sci 2021; 12:7467-7479. [PMID: 34163837 PMCID: PMC8171344 DOI: 10.1039/d1sc01418h] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 01/10/2023] Open
Abstract
Expanding the chemical diversity of metal complexes provides a robust platform to generate functional bioactive reagents. To access an excellent repository of metal-based compounds for probe/drug discovery, we capitalized on the rich chemistry of gold to create organometallic gold(iii) compounds by ligand tuning. We obtained novel organogold(iii) compounds bearing a 1,2-bis(diphenylphosphino)benzene ligand, providing structural diversity with optimal physiological stability. Biological evaluation of the lead compound AuPhos-89 demonstrates mitochondrial complex I-mediated alteration of the mitochondrial electron transport chain (ETC) to drive respiration and diminish cellular energy in the form of adenosine triphosphate (ATP). Mechanism-of-action efforts, RNA-Seq, quantitative proteomics, and NCI-60 screening reveal a highly potent anticancer agent that modulates mitochondrial ETC. AuPhos-89 inhibits the tumor growth of metastatic triple negative breast cancer and represents a new strategy to study the modulation of mitochondrial respiration for the treatment of aggressive cancer and other disease states where mitochondria play a pivotal role in the pathobiology.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Hemendra Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky USA
- Department of Neuroscience, University of Kentucky USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky USA
- Department of Neuroscience, University of Kentucky USA
- Lexington Veterans' Affairs Healthcare System USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky Lexington Kentucky 40536 USA
| |
Collapse
|