851
|
Tang J, Zhu G, Li P, Zhang P, Peng F, Meng F. Novel recognition mechanism based on oxidative addition of Pt(II) complex-based luminescent probes for hypochlorite ion detection. Analyst 2021; 146:5691-5703. [PMID: 34515701 DOI: 10.1039/d1an01048d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Platinum(II) complexes are the most commonly used anticancer drugs and potential optical materials, but the detectability of Pt(II) complex-based probes is seldom reported. In our previous work, a tetradentate Pt(II) complex Pt-CHO was utilised as a 'turn-off' probe to detect ClO- and image cancer cells. However, the recognition mechanism has not been completely clarified and there are still doubts. In this work, three Pt(II) complexes, Pt-H, Pt-CHO and Pt-COOH, were developed to elucidate the mechanism of this class of complexes and refine their property studies. As a result, the UV-visible absorption and luminescence emission experiments, as well as the mass spectrum, proved that the oxidation of Pt(II) to Pt(IV) was the real reason for luminescence quenching, which has nothing to do with aldehyde groups. This first reported mechanism introduces a new type of ClO- probe based on Pt(II) complexes, thereby expanding the application fields of platinum complexes. Moreover, the quantum yield measurements, the effect of biomolecules and reversibility were studied to improve the properties of the probes. Theoretical calculations were used to gain an in-depth understanding of optical characteristics and related mechanisms. The cell imaging of RAW264.7 cells under endogenous ClO- proved the potential of the probes in bioimaging.
Collapse
Affiliation(s)
- Jingjie Tang
- Institute of Bioengineering, Guangdong Academy of Sciences, Guangzhou 510316, China. .,Guangdong Province Engineering Research Center for Green Technology of Sugar Industry, Guangzhou 510316, China
| | - Guoxun Zhu
- School of Chemical Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Peng Li
- Institute of Bioengineering, Guangdong Academy of Sciences, Guangzhou 510316, China. .,Guangdong Province Engineering Research Center for Green Technology of Sugar Industry, Guangzhou 510316, China
| | - Pingjun Zhang
- Institute of Bioengineering, Guangdong Academy of Sciences, Guangzhou 510316, China. .,Guangdong Province Engineering Research Center for Green Technology of Sugar Industry, Guangzhou 510316, China
| | - Fang Peng
- Guangdong Second Provincial General Hospital, Guangzhou 510316, China.
| | - Fei Meng
- Institute of Bioengineering, Guangdong Academy of Sciences, Guangzhou 510316, China. .,Guangdong Province Engineering Research Center for Green Technology of Sugar Industry, Guangzhou 510316, China
| |
Collapse
|
852
|
Lenis-Rojas OA, Cordeiro S, Horta-Meireles M, Fernández JAA, Fernández Vila S, Rubiolo JA, Cabezas-Sainz P, Sanchez L, Fernandes AR, Royo B. N-Heterocyclic Carbene Iron Complexes as Anticancer Agents: In Vitro and In Vivo Biological Studies. Molecules 2021; 26:molecules26185535. [PMID: 34577006 PMCID: PMC8470334 DOI: 10.3390/molecules26185535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/03/2021] [Accepted: 09/09/2021] [Indexed: 12/04/2022] Open
Abstract
Cisplatin and its derivatives are commonly used in chemotherapeutic treatments of cancer, even though they suffer from many toxic side effects. The problems that emerge from the use of these metal compounds led to the search for new complexes capable to overcome the toxic side effects. Here, we report the evaluation of the antiproliferative activity of Fe(II) cyclopentadienyl complexes bearing n-heterocyclic carbene ligands in tumour cells and their in vivo toxicological profile. The in vitro antiproliferative assays demonstrated that complex Fe1 displays the highest cytotoxic activity both in human colorectal carcinoma cells (HCT116) and ovarian carcinoma cells (A2780) with IC50 values in the low micromolar range. The antiproliferative effect of Fe1 was even higher than cisplatin. Interestingly, Fe1 showed low in vivo toxicity, and in vivo analyses of Fe1 and Fe2 compounds using colorectal HCT116 zebrafish xenograft showed that both reduce the proliferation of human HCT116 colorectal cancer cells in vivo.
Collapse
Affiliation(s)
- Oscar A. Lenis-Rojas
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Av. da República, 2780-157 Oeiras, Portugal;
- Correspondence: (O.A.L.-R.); (A.R.F.); (B.R.)
| | - Sandra Cordeiro
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, NOVA University, Campus de Caparica, 2829-516 Caparica, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University, 2819-516 Caparica, Portugal
| | - Marta Horta-Meireles
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Av. da República, 2780-157 Oeiras, Portugal;
| | - Jhonathan Angel Araujo Fernández
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (J.A.A.F.); (S.F.V.); (J.A.R.); (P.C.-S.); (L.S.)
- Laboratory of Zebrafish, Department of Medical Genetics and Genomic Medicine—School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, SP, Brazil
| | - Sabela Fernández Vila
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (J.A.A.F.); (S.F.V.); (J.A.R.); (P.C.-S.); (L.S.)
| | - Juan Andrés Rubiolo
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (J.A.A.F.); (S.F.V.); (J.A.R.); (P.C.-S.); (L.S.)
- Facultad de Ciencias Bioquímicas y Farmacéuticas-Centro Científico y Tecnológico Acuario del Río Paraná, Universidad Nacional de Rosario, Rosario 2000, Argentina
| | - Pablo Cabezas-Sainz
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (J.A.A.F.); (S.F.V.); (J.A.R.); (P.C.-S.); (L.S.)
| | - Laura Sanchez
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (J.A.A.F.); (S.F.V.); (J.A.R.); (P.C.-S.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 5706 Santiago de Compostela, Spain
| | - Alexandra R. Fernandes
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, NOVA University, Campus de Caparica, 2829-516 Caparica, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University, 2819-516 Caparica, Portugal
- Correspondence: (O.A.L.-R.); (A.R.F.); (B.R.)
| | - Beatriz Royo
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Av. da República, 2780-157 Oeiras, Portugal;
- Correspondence: (O.A.L.-R.); (A.R.F.); (B.R.)
| |
Collapse
|
853
|
Patel H, Wu ZX, Chen Y, Bo L, Chen ZS. Drug resistance: from bacteria to cancer. MOLECULAR BIOMEDICINE 2021; 2:27. [PMID: 35006446 PMCID: PMC8607383 DOI: 10.1186/s43556-021-00041-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
The phenomenon of drug resistance has been a hindrance to therapeutic medicine since the late 1940s. There is a plethora of factors and mechanisms contributing to progression of drug resistance. From prokaryotes to complex cancers, drug resistance is a prevailing issue in clinical medicine. Although there are numerous factors causing and influencing the phenomenon of drug resistance, cellular transporters contribute to a noticeable majority. Efflux transporters form a huge family of proteins and are found in a vast number of species spanning from prokaryotes to complex organisms such as humans. During the last couple of decades, various approaches in analyses of biochemistry and pharmacology of transporters have led us to understand much more about drug resistance. In this review, we have discussed the structure, function, potential causes, and mechanisms of multidrug resistance in bacteria as well as cancers.
Collapse
Affiliation(s)
- Harsh Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Yanglu Chen
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA.
| |
Collapse
|
854
|
Mu Q, Lv Y, Luo C, Liu X, Huang C, Xiu Y, Tang L. Research Progress on the Functions and Mechanism of circRNA in Cisplatin Resistance in Tumors. Front Pharmacol 2021; 12:709324. [PMID: 34566636 PMCID: PMC8458655 DOI: 10.3389/fphar.2021.709324] [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] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Cisplatin is a common chemotherapeutic drug that has been used to treat of numerous tumors, including testicular, lung, bladder, ovarian, liver and head and neck cancers. Although clinical chemotherapy based on cisplatin has shown a remarkable therapeutic effect, the resistance to cisplatin becomes increasingly obvious as a patient uses it for a prolonged period. It not only affects the prognosis of these tumors, but also causes the recurrence of cancer and decreases the overall survival rate. The development of cisplatin resistance involves several mechanisms, including DNA damage repair, ATP-binding cassette (ABC) transporter, autophagy, cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), and other related signaling pathways. Interestingly, these mechanisms have been found to be influenced by circular RNAs (circRNAs) to regulate tumor proliferation, invasion, chemosensitivity, and other biological behaviors in the tumor microenvironment (TME). In recent years, circRNAs in cisplatin resistance in tumors, especially lung cancer and gastric cancer, have gradually drawn peoples' attention. This review summarizes recent studies on the functions and mechanisms of circRNAs in cisplatin resistance. We emphasize that circRNA can be used as a promising target gene to improve drug resistance and therapeutic efficacy.
Collapse
Affiliation(s)
- Qingchun Mu
- The People’s Hospital of Gaozhou, Gaozhou, China
| | - Yue Lv
- Department of Urology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chunmei Luo
- The People’s Hospital of Gaozhou, Gaozhou, China
| | - Xiaojing Liu
- The People’s Hospital of Gaozhou, Gaozhou, China
| | | | - Youcheng Xiu
- Department of Urology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | | |
Collapse
|
855
|
Nishiumi F, Kawai Y, Nakura Y, Yoshimura M, Wu HN, Hamaguchi M, Kakizawa S, Suzuki Y, Glass JI, Yanagihara I. Blockade of endoplasmic reticulum stress-induced cell death by Ureaplasma parvum vacuolating factor. Cell Microbiol 2021; 23:e13392. [PMID: 34490709 DOI: 10.1111/cmi.13392] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/03/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022]
Abstract
Previously, we found that Ureaplasma parvum internalised into HeLa cells and cytosolic accumulation of galectin-3. U. parvum induced the host cellular membrane damage and survived there. Here, we conducted vesicular trafficking inhibitory screening in yeast to identify U. parvum vacuolating factor (UpVF). U. parvum triggered endoplasmic reticulum (ER) stress and upregulated the unfolded protein response-related factors, including BiP, P-eIF2 and IRE1 in the host cells, but it blocked the induction of the downstream apoptotic factors. MicroRNA library screening of U. parvum-infected cells and UpVF-transfected cells identified miR-211 and miR-214 as the negative regulators of the apoptotic cascade under ER stress. Transient expression of UpVF induced HeLa cell death with intracellular vacuolization; however, some stable UpVF transformant survived. U. parvum-infected cervical cell lines showed resistance to actinomycin D, and UpVF stable transformant cell lines exhibited resistance to X-ray irradiation, as well as cisplatin and paclitaxel. UpVF expressing cervical cancer xenografts in nude mice also acquired resistance to cisplatin and paclitaxel. A mycoplasma expression vector based on Mycoplasma mycoides, Syn-MBA (multiple banded antigen)-UpVF, reduced HeLa cell survival compared with that of Syn-MBA after 72 hr of infection. These findings together suggest novel mechanisms for Ureaplasma infection and the possible implications for cervical cancer malignancy. TAKE AWAYS: • Ureaplasmal novel virulence factor, UpVF, was identified. • UpVF triggered ER stress but suppressed apoptotic cascade via miR-211 and -214. • UpVF conferred resistance to anticancer treatments both in vivo and in vitro. • Dual expression of MBA and UpVF in JCVI-syn3B showed host cell damage.
Collapse
Affiliation(s)
- Fumiko Nishiumi
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| | - Yasuhiro Kawai
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan.,Health Evaluation Center, Kanazawa Medical University Himi Municipal Hospital, Toyama, Japan
| | - Yukiko Nakura
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| | - Michinobu Yoshimura
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan.,Department of Microbiology and Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Heng Ning Wu
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| | - Mitsuhide Hamaguchi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Emergency and Critical Care Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Shigeyuki Kakizawa
- Bioproduction Research Institute,, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Yo Suzuki
- Synthetic Biology Group, J. Craig Venter Institute, La Jolla, California, USA
| | - John I Glass
- Synthetic Biology Group, J. Craig Venter Institute, La Jolla, California, USA
| | - Itaru Yanagihara
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| |
Collapse
|
856
|
Fotopoulou E, Titilas I, Ronconi L. Metallodrugs as Anticancer Chemotherapeutics and Diagnostic Agents: A Critical Patent Review (2010-2020). Recent Pat Anticancer Drug Discov 2021; 17:42-54. [PMID: 34493191 DOI: 10.2174/1574892816666210907101146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The development of metallodrugs with potential applications in cancer treatment and diagnosis has been a hot topic since the approval and subsequent marketing of the anticancer drug cisplatin in 1978. Since then, thousands of metal-based derivatives have been reported and evaluated for their chemotherapeutic or tumor imaging properties, but only a very limited number gained clinical status. Nonetheless, research in the field has been increasing exponentially over the years, especially in a view to exploiting novel drug designing approaches and strategies aimed at improving pharmacological outcomes and, at the same time, reducing side-effects. OBJECTIVE This review article reports on the patents filed during the last decade and strictly focusing on the development of metal-based anticancer and diagnostic agents. The goal is to identify the latest trends and designing strategies in the field, which would represent a valuable starting point to researchers interested in the development of metallodrugs. METHODS The most relevant patents filed in the 2010-2020 timeframe have been retrieved from various databases using dedicated search engines (such as SciFinder, Google Patents, PatentPak, Espacenet, Global Dossier, PatentScope), sorted by type of metallodrug and screened to include those reporting a substantial amount of biological data. RESULTS The majority of patents here reviewed are concerned with metallodrugs (mostly platinum-based) showing interesting pharmacological properties but no specific tumor-targeting features. Nonetheless, some promising trends in the development of novel drug delivery strategies and/or metallodrugs with potential applications in targeted chemotherapy are envisaged. CONCLUSION In this review, the latest trends in the development of metallodrugs from recent patents are summarized and critically discussed. Such trends would be of interest not only to the scientific community but also to lay audiences aiming to broaden their knowledge of the field and industrial stakeholders potentially interested in the exploitation and commercialization of this class of pharmaceuticals.
Collapse
Affiliation(s)
- Eirini Fotopoulou
- School of Chemistry, College of Science and Engineering, National University of Ireland Galway, University Road, Galway H91 TK33. Ireland
| | - Ioannis Titilas
- School of Chemistry, College of Science and Engineering, National University of Ireland Galway, University Road, Galway H91 TK33. Ireland
| | - Luca Ronconi
- School of Chemistry, College of Science and Engineering, National University of Ireland Galway, University Road, Galway H91 TK33. Ireland
| |
Collapse
|
857
|
Xin C, Huang F, Wang J, Li J, Chen Q. Roles of circRNAs in cancer chemoresistance (Review). Oncol Rep 2021; 46:225. [PMID: 34468007 DOI: 10.3892/or.2021.8176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 03/11/2021] [Indexed: 02/05/2023] Open
Abstract
Circular RNA (circRNA) is a type of endogenous, high‑stability, noncoding RNA. circRNAs exhibit various biological functions, and are involved in physiological and pathological processes occurring in various diseases, including cancers. They can not only act as microRNA and protein sponges, but also interact with proteins, translated peptides, and transcriptional and translational regulators, and compete with pre‑mRNA splicing. Chemotherapy is one of the most important types of cancer treatment. However, the resistance of cancer cells to chemotherapy is a leading reason for the failure of chemotherapy. It has been reported that circRNAs play important roles in cancer resistance via a number of mechanisms. The functions of the circRNAs provide insight into their roles in chemoresistance pathways. In addition, some circRNAs may serve as novel biomarkers for the diagnosis and prognosis of cancer resistance. Obtaining improved understanding of the molecular regulatory networks featuring circRNAs in tumors and searching for markers for the diagnosis and treatment of cancer resistance are leading issues in circRNA research. The present review introduced the functions of circRNAs, illustrated the mechanisms underlying drug resistance in cancer, described the contributions of circRNAs to this resistance and discussed the potential application of circRNAs in the treatment of drug‑resistant cancer. In particular, the review aimed to reveal the main mechanisms of circRNAs in cancer drug resistance, including mechanisms involving drug transport and metabolism, alterations of drug targets, DNA damage repair, downstream resistance mechanisms, adaptive responses and the tumor microenvironment. The findings may provide novel therapeutic targets for clinical treatment of cancer chemoresistance.
Collapse
Affiliation(s)
- Chuan Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fei Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
858
|
Amini Chermahini F, Raeisi E, Aazami MH, Mirzaei A, Heidarian E, Lemoigne Y. Does Bromelain-Cisplatin Combination Afford In-Vitro Synergistic Anticancer Effects on Human Prostatic Carcinoma Cell Line, PC3? Galen Med J 2021; 9:e1749. [PMID: 34466585 PMCID: PMC8343875 DOI: 10.31661/gmj.v9i0.1749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/07/2019] [Accepted: 01/23/2020] [Indexed: 12/28/2022] Open
Abstract
Background:
Bromelain enhances anticancer impacts to chemotherapeutic agents. The question as to whether bromelain does promote in-vitro cytotoxic and proapoptotic effects of cisplatin on human prostatic carcinoma PC3 cell line was investigated.
Materials and Methods:
PC3 (human prostatic carcinoma) cells were treated either single or in combination with bromelain and/or cisplatin. MTT, clonogenic assay, flow cytometry and real-time quantitative polymerase chain reaction were used to investigate cell viability, colony formation, proapoptotic potential and p53 gene expression, respectively.
Results:
Cisplatin (IC10) combined with bromelain (IC40) significantly affected PC3 cell viability, inhibited colony formation, as well increased p53 proapoptotic gene expression compared to cisplatin single treatment. Nevertheless, bromelain-cisplatin chemoherbal combination did not display any additive proapoptotic effect compared to single treatments.
Conclusion:
Bromelain-cisplatin chemoherbal combination demonstrated synergistic in-vitro anticancer effect on human prostatic carcinoma cell line, PC3, that drastically reduced required cisplatin dose.
Collapse
Affiliation(s)
- Fatemeh Amini Chermahini
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Raeisi
- Department of Medical Physics and Radiology, School of Allied Medical Sciences, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Correspondence to: Elham Raeisi, Shahrekord University of Medical Sciences, School of Allied Medical Sciences, Rahmatiyeh, Shahrekord, Iran Telephone Number: +983833346692 Email Address:
| | - Mathias Hossain Aazami
- Department of Cardiology and Cardiac Surgery, Kashani and Hajar University Hospitals, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Abbas Mirzaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Esfandiar Heidarian
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Yves Lemoigne
- Department of Medical Physics, Institute for Medical Physics, Ambilly, France
| |
Collapse
|
859
|
Hou L, Jia X, Wu Y, Li J, Yao D, Gou Y, Huang G. Aroylhydrazone Cu(Ⅱ) complexes: Syntheses, crystal structures, and anticancer properties. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
860
|
Zheng X, Wu H, Shen Z, Wang C, Ma Y. COOH-Terminated Silicon Quantum Dots as a Highly Efficient Drug Nanocarrier for Targeted Tumor Cell Imaging. J Biomed Nanotechnol 2021; 17:1830-1839. [PMID: 34688328 DOI: 10.1166/jbn.2021.3151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Fluorescent silicon quantum dots (SiQDs) characterized by exceptional photostability and colloidal robustness as well as beneficial biocompatibility are fast becoming new pharmaceutical nanocarriers. With a view to efficiently loading cisplatin (CDDP) onto SiQDs, carboxylate group (COOH) terminated SiQDs were imperative because of chelate formation with CDDP. In this work, we employed a facial microwave irradiation route for rapidly synthesizing high-quality COOH-SiQDs through the use of 3-aminopropyl trimethoxy silane (APTMS) molecules to fulfil the role of silicon precursor and maleic acid (MA) as the agent for facilitating reduction. The SiQDs showed blue fluorescence with an associated photoluminescence quantum yield (PLQY) of 40.2%, the size of which was small at 3.2 ±0.6 nm, and long-lasting stability (an extensive range in pH (4-12) and concentrations of electrolytes reaching 3 Molarity of a solution of sodium chloride). As nanocarriers, carboxylic acids chelation generated a high loading of CDDP onto SiQDs (drug loading capacity, DLC up to 32.2% at pH = 9) and a drug release of CDDP up to 57.6% at pH = 5. Furthermore, the MTT assays demonstrated the non or low cytotoxicity of SiQDs and the role of the controlled release of SiQD-CDDP Finally, the prepared SiQD-CDDP were used for cell imaging, and further targeted labeling of some tumors after folic acid (FA) conjugation. These characteristics allow for the deployment of SiQDs as a highly efficient nanocarrier that facilitate the delivery of clinical drugs for the future.
Collapse
Affiliation(s)
- Xiaohui Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongyu Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhe Shen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Caihong Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yunfei Ma
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
861
|
Gu S, Wu Q, Li Y, Jia W, Zhang D, Jiao L, Zhang Y. Investigation of differentially expressed gene profile for cisplatin-treated lung cancer patients. Anticancer Drugs 2021; 32:875-881. [PMID: 33967201 DOI: 10.1097/cad.0000000000001075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The purpose of the study was to establish a comprehensive differential gene profile for lung cancer patients treated with cisplatin compared with control patients without any chemotherapy drug treatment. The RNA sequencing data and miRNA sequencing data of 108 lung cancer patients treated with cisplatin only and 232 lung cancer patients treated without any chemotherapeutic drugs, were analyzed using differential expression, protein-protein interaction, and immune cell infiltration ratio analysis. Compared with control patients, the cisplatin-treated patients demonstrated 336 differentially expressed genes, which included 48 upregulated genes and 288 downregulated genes. Meanwhile, 12 differentially expressed miRNAs (DEMs), including 7 upregulated miRNAs and 5 downregulated miRNAs showed a differentially expressed pattern. With further instigation, five miRNAs (hsa-miR-548ah, hsa-miR-466, hsa-miR-552, hsa-miR-371a, and hsa-miR-4445) were suggested to be the key targets in the cisplatin-treated patients. At the same time, we also found a significant correlation between the cisplatin treatment and six immune checkpoints including programmed cell death ligand. This study helped us better understand the potential targets and underline molecular mechanisms for cisplatin treatment and provided references to eliminate existing side effects in the future.
Collapse
Affiliation(s)
- Songtao Gu
- Tianjin Medical University
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Qi Wu
- Tianjin Medical University General Hospital
| | - Yuechuan Li
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Wei Jia
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Dongrui Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Lina Jiao
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Yating Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
862
|
Xu Z, Wang Z, Deng Z, Zhu G. Recent advances in the synthesis, stability, and activation of platinum(IV) anticancer prodrugs. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213991] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
863
|
Oishi T, Sasaki Y, Tong Y, Chen L, Onodera T, Iwasa S, Udo E, Furusato B, Fujimori H, Imamichi S, Honda T, Bessho T, Fukuoka J, Ashizawa K, Yanagihara K, Nakao K, Yamada Y, Hiraoka N, Masutani M. A newly established monoclonal antibody against ERCC1 detects major isoforms of ERCC1 in gastric cancer. Glob Health Med 2021; 3:226-235. [PMID: 34532603 DOI: 10.35772/ghm.2021.01001] [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: 01/10/2021] [Revised: 04/25/2021] [Accepted: 05/10/2021] [Indexed: 11/08/2022]
Abstract
Identifying patients resistant to cisplatin treatment is expected to improve cisplatin-based chemotherapy for various types of cancers. Excision repair cross-complementing group 1 (ERCC1) is involved in several repair processes of cisplatin-induced DNA crosslinks. ERCC1 overexpression is reported as a candidate prognostic factor and considered to cause cisplatin resistance in major solid cancers. However, anti-ERCC1 antibodies capable of evaluating expression levels of ERCC1 in clinical specimens were not fully optimized. A mouse monoclonal antibody against human ERCC1 was generated in this study. The developed antibody 9D11 specifically detected isoforms of 201, 202, 203 but not 204, which lacks the exon 3 coding region. To evaluate the diagnostic usefulness of this antibody, we have focused on gastric cancer because it is one of the major cancers in Japan. When ERCC1 expression was analyzed in seventeen kinds of human gastric cancer cell lines, all the cell lines were found to express either 201, 202, and/or 203 as major isoforms of ERCC1, but not 204 by Western blotting analysis. Immunohistochemical staining showed that ERCC1 protein was exclusively detected in nuclei of the cells and a moderate level of constant positivity was observed in nuclei of vascular endothelial cells. It showed a clear staining pattern in clinical specimens of gastric cancers. Antibody 9D11 may thus be useful for estimating expression levels of ERCC1 in clinical specimens.
Collapse
Affiliation(s)
- Takayuki Oishi
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuka Sasaki
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ying Tong
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Lichao Chen
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takae Onodera
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Emiko Udo
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Bungo Furusato
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroaki Fujimori
- Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shoji Imamichi
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Clinical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Junya Fukuoka
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuto Ashizawa
- Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Nagasaki, Japan.,Department of Clinical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yasuhide Yamada
- Department of Medical Oncology, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Comprehensive Cancer Center, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Division of Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Mitsuko Masutani
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
864
|
Dai G, Zheng D, Liu G, Song Q. Synergistic Anticancer Effects of Cisplatin Combined with Combretastatin A4 Phosphate on Human Osteosarcoma-Xenografted Mice. Cells Tissues Organs 2021; 210:293-300. [PMID: 34433168 DOI: 10.1159/000517446] [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: 02/16/2021] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
This study aimed to investigate the effectiveness of anticancer therapy combining a cytotoxic chemotherapeutic agent, cisplatin (DDP), and a vascular disruptive drug, combretastatin A4 phosphate (CA4P), in osteosarcoma. First, a human osteosarcoma-xenografted mice model was established. Second, the transplanted tumor models were treated with DDP and CA4P in combination or as monotherapy. Third, the therapeutic effects and the mechanism of the drug combination in the inhibition of transplanted tumors was studied. Finally, the toxic effects of the drugs were observed and recorded. The results showed that DDP combined with CA4P significantly inhibited the growth and lung metastasis of transplanted tumors compared with the monotherapy drug group and vehicle control group. Histopathological analysis revealed that apoptotic and necrotic cell death significantly increased in the combination group, and combined treatment significantly inhibited the proliferation of osteosarcoma cells compared with either agent alone or the vehicle control. Additionally, no obvious toxic effects were observed in the combination group. These results indicate that the combined effects of DDP and CA4P on the progression of human osteosarcoma in vivo were superior to that of either agent alone. DDP combined with CA4P exerted synergistic effects at lower concentrations and promoted apoptosis and necrosis, as well as inhibited proliferation of osteosarcoma cells, but it did not increase the systemic toxic effects of chemotherapy. Our findings highlight CA4P as an effective anticancer agent candidate for combination with DDP in clinical applications to treat osteosarcoma.
Collapse
Affiliation(s)
- Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaiwei Liu
- Department of Orthopedics, Jingzhou Central Hospital, Jingzhou, China
| | - Qi Song
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, China
| |
Collapse
|
865
|
Wu J, Yang T, Wang X, Li W, Pang M, Sun H, Liang H, Yang F. Development of a multi-target anticancer Sn(ii) pyridine-2-carboxaldehyde thiosemicarbazone complex. Dalton Trans 2021; 50:10909-10921. [PMID: 34313274 DOI: 10.1039/d1dt01272j] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study, we proposed to design effective multi-target anticancer agents based on the chelation of nontoxic metals with ligands that possess anticancer activity. In total, five Sn(ii) pyridine-2-carboxaldehyde thiosemicarbazone complexes are synthesized and their activities are tested. Among these complexes, C5 is found to show the highest cytotoxicity on investigating their structure-activity relationships. In addition, C5 not only exhibits an effective inhibitory effect against tumor growth in vivo, but also suppresses angiogenesis and restricts the metastasis of cancer cells in vitro. Multiple mechanisms underlie the antitumor effect of C5, and they include acting against DNA, inducing apoptosis, and inhibiting the activities of anti-apoptotic Bcl-xL protein, metalloproteinase MMP2 and topoisomerase II.
Collapse
Affiliation(s)
- Junmiao Wu
- 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, 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, China.
| | - Xiaojun Wang
- 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, China.
| | - Wenjuan Li
- 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, China.
| | - Min Pang
- 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, China.
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu, 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, 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, China.
| |
Collapse
|
866
|
Maliyakkal N, Appadath Beeran A, Udupa N. Nanoparticles of cisplatin augment drug accumulations and inhibit multidrug resistance transporters in human glioblastoma cells. Saudi Pharm J 2021; 29:857-873. [PMID: 34408546 PMCID: PMC8363105 DOI: 10.1016/j.jsps.2021.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/04/2021] [Indexed: 11/16/2022] Open
Abstract
Background Cisplatin (CSP) is a potent anticancer drug widely used in treating glioblastoma multiforme (GBM). However, CSP's clinical efficacy in GBM contrasted with low therapeutic ratio, toxicity, and multidrug resistance (MDR). Therefore, we have developed a system for the active targeting of cisplatin in GBM via cisplatin loaded polymeric nanoplatforms (CSP-NPs). Methods CSP-NPs were prepared by modified double emulsion and nanoprecipitation techniques. The physiochemical characterizations of CSP-NPs were performed using zeta sizer, scanning electron microscopy (SEM), drug release kinetics, and drug content analysis. Cytotoxicity, induction of apoptosis, and cell cycle-specific activity of CSP-NPs in human GBM cell lines were evaluated by MTT assay, fluorescent microscopy, and flow cytometry. Intracellular drug uptake was gauged by fluorescent imaging and flow cytometry. The potential of CSP-NPs to inhibit MDR transporters were assessed by flow cytometry-based drug efflux assays. Results CSP-NPs have smooth surface properties with discrete particle size with required zeta potential, polydispersity index, drug entrapment efficiency, and drug content. CSP-NPs has demonstrated an ‘initial burst effect’ followed by sustained drug release properties. CSP-NPs imparted dose and time-dependent cytotoxicity and triggered apoptosis in human GBM cells. Interestingly, CSP-NPs significantly increased uptake, internalization, and accumulations of anticancer drugs. Moreover, CSP-NPs significantly reversed the MDR transporters (ABCB1 and ABCG2) in human GBM cells. Conclusion The nanoparticulate system of cisplatin seems to has a promising potential for active targeting of cisplatin as an effective and specific therapeutic for human GBM, thus eliminating current chemotherapy's limitations.
Collapse
Key Words
- ABC, ATP-binding cassette
- ANOVA, Analysis of variance
- Active drug targeting
- BBB, Blood brain barrier
- BCRP, Breast cancer resistance protein
- CSP, Cisplatin
- CSP-NPs, Cisplatin nanoparticles DMEM, Dulbecco’s modified eagle medium
- Cisplatin nanoparticles
- DMSO, Dimethyl sulfoxide
- DNR, Daunorubicin
- DOX, Doxorubicin
- Drug uptake and accumulations
- EDTA, Ethylenediaminetetraacetic acid
- EPR, Enhanced permeability retention
- FACS, Fluorescence activated cell sorting
- FBS, Fetal bovine serum
- FTC, Fumitremorgin C
- GBM, Glioblastoma multiforme
- HBSS, Hank’s balanced salt solution
- HPLC, High Performance Liquid Chromatography
- Induction of Apoptosis
- MDR, Multidrug resistance
- MTT, Methyl tetrazolium
- MX, Mitoxantrone
- NPs, Nanoparticles
- O.D., Optical density
- PBS, Phosphate buffer saline
- PI, Propidium iodide
- PLGA, Poly (lactic-co-glycolic) acid
- RT, Room temperature
- Rho-123, Rhodamine 123
- SDS, Sodium dodecyl sulfate
- SEM, Scanning electron microscopy
- Targeting multidrug resistance (MDR) transporters
- nm, Nanometer
Collapse
Affiliation(s)
- Naseer Maliyakkal
- Department of Basic Medical Sciences, College of Applied Medical Sciences in Khamis Mushait, King Khalid University, Abha, Saudi Arabia.,Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Cancer Research Unit, King Khalid University, Abha, Saudi Arabia
| | - Asmy Appadath Beeran
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nayanabhirama Udupa
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
867
|
Li T, Liu T, Zhu W, Xie S, Zhao Z, Feng B, Guo H, Yang R. Targeting MDSC for Immune-Checkpoint Blockade in Cancer Immunotherapy: Current Progress and New Prospects. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2021; 15:11795549211035540. [PMID: 34408525 PMCID: PMC8365012 DOI: 10.1177/11795549211035540] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 07/07/2021] [Indexed: 01/06/2023]
Abstract
Immune-checkpoint blockade (ICB) demonstrated inspiring effect and great promise in anti-cancer therapy. However, many obstacles, such as drug resistance and difficulty in patient selection, limited the efficacy of ICB therapy and awaited to be overcome. By timely identification and intervention of the key immune-suppressive promotors in the tumor microenvironment (TME), we may better understand the mechanisms of cancer immune-escape and use novel strategies to enhance the therapeutic effect of ICB. Myeloid-derived suppressor cell (MDSC) is recognized as a major immune suppressor in the TME. In this review, we summarized the roles MDSC played in the cancer context, focusing on its negative biologic functions in ICB therapy, discussed the strategies targeted on MDSC to optimize the diagnosis and therapy process of ICB and improve the efficacy of ICB therapy against malignancies.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Tianyao Liu
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Wenjie Zhu
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Shangxun Xie
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Zihan Zhao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Baofu Feng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| |
Collapse
|
868
|
Ciaffaglione V, Modica MN, Pittalà V, Romeo G, Salerno L, Intagliata S. Mutual Prodrugs of 5-Fluorouracil: From a Classic Chemotherapeutic Agent to Novel Potential Anticancer Drugs. ChemMedChem 2021; 16:3496-3512. [PMID: 34415107 PMCID: PMC9290623 DOI: 10.1002/cmdc.202100473] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/18/2021] [Indexed: 12/18/2022]
Abstract
The development of potent antitumor agents with a low toxicological profile against healthy cells is still one of the greatest challenges facing medicinal chemistry. In this context, the “mutual prodrug” approach has emerged as a potential tool to overcome undesirable physicochemical features and mitigate the side effects of approved drugs. Among broad‐spectrum chemotherapeutics available for clinical use today, 5‐fluorouracil (5‐FU) is one of the most representative, also included in the World Health Organization model list of essential medicines. Unfortunately, severe side effects and drug resistance phenomena are still the primary limits and drawbacks in its clinical use. This review describes the progress made over the last ten years in developing 5‐FU‐based mutual prodrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues.
Collapse
Affiliation(s)
- Valeria Ciaffaglione
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Maria N Modica
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
869
|
Singh R, Adhya P, Sharma SS. Redox-sensitive TRP channels: a promising pharmacological target in chemotherapy-induced peripheral neuropathy. Expert Opin Ther Targets 2021; 25:529-545. [PMID: 34289785 DOI: 10.1080/14728222.2021.1956464] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chemotherapy-induced peripheral neuropathy (CIPN) and its related pain is a major side effect of certain chemotherapeutic agents used in cancer treatment. Available analgesics are mostly symptomatic, and on prolonged treatment, patients become refractive to them. Hence, the development of improved therapeutics that act on novel therapeutic targets is necessary. Potential targets include the redox-sensitive TRP channels [e.g. TRPA1, TRPC5, TRPC6, TRPM2, TRPM8, TRPV1, TRPV2, and TRPV4] which are activated under oxidative stress associated with CIPN. AREAS COVERED We have examined numerous neuropathy-inducing cancer chemotherapeutics and their pathophysiological mechanisms. Oxidative stress and its downstream targets, the redox-sensitive TRP channels, together with their potential pharmacological modulators, are discussed. Finally, we reflect upon the barriers to getting new therapeutic approaches into the clinic. The literature search was conducted in PubMed upto and including April 2021. EXPERT OPINION Redox-sensitive TRP channels are a promising target in CIPN. Pharmacological modulators of these channels have reduced pain in preclinical models and in clinical studies. Clinical scrutiny suggests that TRPA1, TRPM8, and TRPV1 are the most promising targets because of their pain-relieving potential. In addition to the analgesic effect, TRPV1 agonist-Capsaicin possesses a disease-modifying effect in CIPN through its restorative property in damaged sensory nerves.
Collapse
Affiliation(s)
- Ramandeep Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Pratik Adhya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| |
Collapse
|
870
|
Cai Z, Teng Y, Chen Y. The Effect of Shenyi Capsule on Non-Small-Cell Lung Cancer Combined with Chemotherapy from the Yin-Yang Perspective. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:1653750. [PMID: 34512772 PMCID: PMC8426066 DOI: 10.1155/2021/1653750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/26/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
As an example of Shenyi capsule on non-small-cell lung cancer combined with chemotherapy, this review discusses the synergistic effect and mechanism of natural drugs in oncotherapy from the yin-yang perspective in ancient Chinese philosophy, so as to reflect the therapeutic principle of natural drugs for tumor more comprehensively. The major focuses of this review are on the philosophical thinking of yin-yang as a tool which can not only explain the effect of Shenyi capsule in NSCLC combined with chemotherapy but also explore the mechanism of Shenyi capsule at the cellular and molecular level. Learning from the "yin-yang" thinking of ancient Chinese philosophy will bring more enlightenment to the research and development of traditional Chinese drugs in the future.
Collapse
Affiliation(s)
- Zhixing Cai
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Yue Teng
- Outpatient Department of Clinic Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai 200437, China
| | - Yue Chen
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| |
Collapse
|
871
|
Annunziata A, Liberti D, Bedini E, Cucciolito ME, Loreto D, Monti DM, Merlino A, Ruffo F. Square-Planar vs. Trigonal Bipyramidal Geometry in Pt(II) Complexes Containing Triazole-Based Glucose Ligands as Potential Anticancer Agents. Int J Mol Sci 2021; 22:ijms22168704. [PMID: 34445409 PMCID: PMC8395886 DOI: 10.3390/ijms22168704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022] Open
Abstract
This article describes the synthesis, characterization, and biological activity of novel square-planar cationic platinum(II) complexes containing glucoconjugated triazole ligands and a comparison with the results obtained from the corresponding five-coordinate complexes bearing the same triazole ligands. Stability in solution, reactivity with DNA and small molecules of the new compounds were evaluated by NMR, fluorescence, and UV–vis absorption spectroscopy, together with their cytotoxic action against pairs of immortalized and tumorigenic cell lines. The results show that the square-planar species exhibit greater stability than the corresponding five-coordinate ones. Furthermore, although the square-planar complexes are less cytotoxic than the latter ones, they exhibit a certain selectivity. These results simultaneously demonstrate that overall stability is a fundamental prerequisite for preserving the performance of the agents and that coordinative saturation constitutes a point in favor of their biological action.
Collapse
|
872
|
Fan H, Ou Q, Su Q, Li G, Deng Z, Huang X, Bi J. ZIPK activates the IL-6/STAT3 signaling pathway and promotes cisplatin resistance in gastric cancer cells. FEBS Open Bio 2021; 11:2655-2667. [PMID: 34375503 PMCID: PMC8409285 DOI: 10.1002/2211-5463.13270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/06/2021] [Accepted: 08/09/2021] [Indexed: 11/24/2022] Open
Abstract
Gastric cancer is one of the most common malignant cancers globally. Chemotherapy resistance remains a major obstacle in the treatment of gastric cancer, and the molecular mechanisms underlying drug resistance are still not well understood. We previously reported that Zipper interacting protein kinase (ZIPK), also known as death‐associated protein kinase3, exerts an oncogenic effect on gastric cancer via activation of Akt/NF‐κB signaling and promotion of stemness. Here, we explored the roles of ZIPK in cisplatin resistance. We report that ZIPK enhances cell proliferation and invasion and reduces the antitumor activity of cisplatin in gastric cancer. In addition, our western blot data suggest that ZIPK activated the IL‐6/STAT3 signaling pathway. Furthermore, ZIPK increased the expression of IL‐6 and multidrug‐resistance genes. Using the STAT3 inhibitor stattic to block the IL‐6/STAT3 signaling pathway strongly increased the sensitivity of ZIPK‐expressed cells to cisplatin. In conclusion, ZIPK may play a role in cisplatin resistance through activation of the IL‐6/ STAT3 signaling pathway. Inhibition of STAT3 in gastric cancer overexpressing ZIPK might have potential to improve the efficacy of cisplatin.
Collapse
Affiliation(s)
- Haonan Fan
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qifeng Ou
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiao Su
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanman Li
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,School of Medicine (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Zhijuan Deng
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Ultrasound Medical Center, the First people's Hospital of Chenzhou, Chenzhou, China
| | - Xiaohui Huang
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiong Bi
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
873
|
Guo T, Gu C, Li B, Xu C. Dual inhibition of FGFR4 and BCL-xL inhibits multi-resistant ovarian cancer with BCL2L1 gain. Aging (Albany NY) 2021; 13:19750-19759. [PMID: 34351305 PMCID: PMC8386571 DOI: 10.18632/aging.203386] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022]
Abstract
Aim: Overexpression of BCL2L1 (BCL-xL) was associated with platinum resistance in ovarian cancer (OvCa). However, role of copy number (CN) gain of BCL2L1 in OvCa remains elusive. Methods: In silico analyses of multiple public datasets were perform. Validation was carried out in our tissue microarray (TMA) of OvCa cases. In vitro and in vivo assays was performed to explore potential targeted compound against BCL2L1-gained OvCa. Results: BCL2L1 was gained in ~60% of OvCa. BCL2L1 was differentially expressed between healthy and cancerous ovarian cases. BCL2L1 gain was not prognostic either in overall or in progression-free survival but higher BCL2L1 expression was associated with worsened survival, indicating biological distinction between CN gain and overexpression of the gene. BCL2L1 gain was associated with multi-resistance to various drug with no significant sensitivity to any single agent. Only CRISPR-mediated BCL2L1 knockout, but not shRNA could be inhibitive. Combined genetic silencing of FGFR4/NCAM and BCL2L1 with shRNA induced potent inhibition of BCL2L1-gained OvCa with durable effect. Combined inhibition of FGFR/BCL-xL was required for inhibiting BCL2L1-gained OvCa in vitro and in vivo. Only dual inhibition of FGFR/BCL-xL without platinum was tolerable in vivo. Conclusion: Gain of BCL2L1 is associated with resistance to multiple anti-cancer agents in OvCa. Dual inhibition of FGFR4 and BCL-xL showed potent effect and tolerable toxicity, holding promise to further translation.
Collapse
Affiliation(s)
- Ting Guo
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P.R. China
| | - Chao Gu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P.R. China
| | - Bin Li
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P.R. China
| | - Congjian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P.R. China
| |
Collapse
|
874
|
Zuccolo M, Arrighetti N, Perego P, Colombo D. Recent Progresses in Conjugation with Bioactive Ligands to Improve the Anticancer Activity of Platinum Compounds. Curr Med Chem 2021; 29:2566-2601. [PMID: 34365939 DOI: 10.2174/0929867328666210806110857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Platinum (Pt) drugs, including cisplatin, are widely used for the treatment of solid tumors. Despite the clinical success, side effects and occurrence of resistance represent major limitations to the use of clinically available Pt drugs. To overcome these problems, a variety of derivatives have been designed and synthetized. Here, we summarize the recent progress in the development of Pt(II) and Pt(IV) complexes with bioactive ligands. The development of Pt(II) and Pt(IV) complexes with targeting molecules, clinically available agents, and other bioactive molecules is an active field of research. Even if none of the reported Pt derivatives has been yet approved for clinical use, many of these compounds exhibit promising anticancer activities with an improved pharmacological profile. Thus, planning hybrid compounds can be considered as a promising approach to improve the available Pt-based anticancer agents and to obtain new molecular tools to deepen the knowledge of cancer progression and drug resistance mechanisms.
Collapse
Affiliation(s)
- Marco Zuccolo
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan. Italy
| | - Noemi Arrighetti
- Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan. Italy
| | - Paola Perego
- Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan. Italy
| | - Diego Colombo
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan. Italy
| |
Collapse
|
875
|
Hussain Y, Islam L, Khan H, Filosa R, Aschner M, Javed S. Curcumin-cisplatin chemotherapy: A novel strategy in promoting chemotherapy efficacy and reducing side effects. Phytother Res 2021; 35:6514-6529. [PMID: 34347326 DOI: 10.1002/ptr.7225] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
The efficacy of chemotherapy in cancer therapy is limited due to resistance, treatment selectivity, and severe adverse effects. Immunotherapy, chemotherapy, targeted therapy, radiation, and surgery are the most common therapeutic strategies for treatment, with chemotherapy being the most successful. Nonetheless, these treatments exhibit poor effectiveness due to toxicity and resistance. Therefore, combination therapies of natural products may be used as an effective and novel strategy to overcome such barriers. Cisplatin is a platinum-based chemotherapy agent, and when administered alone, it can lead to severe adverse effects and resistance mechanism resulting in therapeutic failure. Curcumin is a polyphenolic compound extracted from turmeric (Curcuma longa) exhibiting anticancer potential with minimal adverse effects. The combination therapy of curcumin and cisplatin is a novel strategy to mitigate/attenuate cisplatin-related adverse effects and improve the barrier of resistance reducing unwanted effects. However, there are uncertainties on the efficacy of curcumin, and more in depth and high-quality studies are needed. This review aims to explain the adverse effects related to individual cisplatin delivery, the positive outcome of individual curcumin delivery, and the combination therapy of curcumin and cisplatin from nano platform as a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Controlled Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Lubna Islam
- Department of Pharmacy, University of Malakand, Dir Lower Chakdara, KPK, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Rosanna Filosa
- Department of Experimental Medicine, University of Campania, "L. Vanvitelli", Naples, Italy
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Saba Javed
- Department of Zoology, Hazara University, Mansehra, Pakistan
| |
Collapse
|
876
|
Yang K, Yu G, Yang Z, Yue L, Zhang X, Sun C, Wei J, Rao L, Chen X, Wang R. Supramolecular Polymerization-Induced Nanoassemblies for Self-Augmented Cascade Chemotherapy and Chemodynamic Therapy of Tumor. Angew Chem Int Ed Engl 2021; 60:17570-17578. [PMID: 34041833 DOI: 10.1002/anie.202103721] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/05/2021] [Indexed: 01/19/2023]
Abstract
The clinical application of chemodynamic therapy is impeded by the insufficient intracellular H2 O2 level in tumor tissues. Herein, we developed a supramolecular nanoparticle via a simple one-step supramolecular polymerization-induced self-assembly process using platinum (IV) complex-modified β-cyclodextrin-ferrocene conjugates as supramolecular monomers. The supramolecular nanoparticles could dissociate rapidly upon exposure to endogenous H2 O2 in the tumor and release hydroxyl radicals as well as platinum (IV) prodrugs in situ, which is reduced into cisplatin to significantly promote the generation of H2 O2 in the tumor tissue. Thus, the supramolecular nanomedicine overcomes the limitation of conventional chemodynamic therapy via the self-augmented cascade radical generation and drug release. In addition, dissociated supramolecular nanoparticles could be readily excreted from the body via renal clearance to effectively avoid systemic toxicity and ensure long term biocompatibility of the nanomedicine. This work may provide new insights on the design and development of novel supramolecular nanoassemblies for cascade chemo/chemodynamic therapy.
Collapse
Affiliation(s)
- Kuikun Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Guocan Yu
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, China
| | - Zhiqing Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Ludan Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Xiangjun Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Chen Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Jianwen Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macau, China
| |
Collapse
|
877
|
Mei H, Li J, Cai S, Zhang X, Shi W, Zhu H, Cao J, He B. Mitochondria-acting carrier-free nanoplatform self-assembled by α-tocopheryl succinate carrying cisplatin for combinational tumor therapy. Regen Biomater 2021; 8:rbab029. [PMID: 34221448 PMCID: PMC8242230 DOI: 10.1093/rb/rbab029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022] Open
Abstract
Unsatisfactory drug loading capability, potential toxicity of the inert carrier and the limited therapeutic effect of a single chemotherapy drug are all vital inhibitory factors of carrier-assisted drug delivery systems for chemotherapy. To address the above obstacles, a series of carrier-free nanoplatforms self-assembled by dual-drug conjugates was constructed to reinforce chemotherapy against tumors by simultaneously disrupting intratumoral DNA activity and inhibiting mitochondria function. In this nanoplatform, the mitochondria-targeting small-molecular drug, α-tocopheryl succinate (TOS), firstly self-assembled into nanoparticles, which then were used as the carrier to conjugate cisplatin (CDDP). Systematic characterization results showed that this nanoplatform exhibited suitable particle size and a negative surface charge with good stability in physicochemical environments, as well as pH-sensitive drug release and efficient cellular uptake. Due to the combined effects of reactive oxygen species (ROS) generation by TOS and DNA damage by CDDP, the developed nanoplatform could induce mitochondrial dysfunction and elevated cell apoptosis, resulting in highly efficient anti-tumor outcomes in vitro. Collectively, the combined design principles adopted for carrier-free nanodrugs construction in this study aimed at targeting different intracellular organelles for facilitating ROS production and DNA disruption can be extended to other carrier-free nanodrugs-dependent therapeutic systems.
Collapse
Affiliation(s)
- Heng Mei
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Jing Li
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Shengsheng Cai
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Xuequan Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Wenqiang Shi
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Hai Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| |
Collapse
|
878
|
Niu L, Ren G, Hou T, Shen X, Zhu D. Synthesis, structure and anticancer activity of three platinum(II) complexes with 2-phenylpyridine derivatives. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.108737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
879
|
Diorganotin(IV) complexes derived from thiazole Schiff bases: synthesis, characterization, antimicrobial and cytotoxic studies. RESEARCH ON CHEMICAL INTERMEDIATES 2021. [DOI: 10.1007/s11164-021-04557-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
880
|
Chen KB, Yang W, Xuan Y, Lin AJ. miR-526b-3p inhibits lung cancer cisplatin-resistance and metastasis by inhibiting STAT3-promoted PD-L1. Cell Death Dis 2021; 12:748. [PMID: 34321456 PMCID: PMC8319181 DOI: 10.1038/s41419-021-04033-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
Chemotherapy remains the primary treatment of advanced solid cancer, including lung cancer. However, as first-line treatment, cisplatin-based therapy is restricted by the frequent development of drug resistance. Increasing data showed that the programmed cell death protein ligand 1 (PD-L1) plays a vital role in regulating cisplatin resistance. However, the underlying mechanisms are not fully understood. We found that miR-526b-3p expression declined while PD-L1 was elevated in cisplatin-resistant lung cancer compared to that in cisplatin-sensitive lung cancer by analyzing clinical samples. Significantly, miR-526b-3p was associated with response to cisplatin negatively. We further demonstrated that miR-526b-3p reversed cisplatin resistance, suppressed metastasis, and activated CD8+ T cells in a STAT3/PD-L1-dependent manner. Thus, our findings extended the knowledge of PD-L1-mediated cisplatin resistance of lung cancer. In addition, the introduction of miR-526b-3p provided a new clue to improve the anti-tumor effects of the combination of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Kuan-Bing Chen
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Yang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Ying Xuan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ai-Jun Lin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
881
|
Lin Y, Betts H, Keller S, Cariou K, Gasser G. Recent developments of metal-based compounds against fungal pathogens. Chem Soc Rev 2021; 50:10346-10402. [PMID: 34313264 DOI: 10.1039/d0cs00945h] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This review provides insight into the rapidly expanding field of metal-based antifungal agents. In recent decades, the antibacterial resistance crisis has caused reflection on many aspects of public health where weaknesses in our medicinal arsenal may potentially be present - including in the treatment of fungal infections, particularly in the immunocompromised and those with underlying health conditions where mortality rates can exceed 50%. Combination of organic moieties with known antifungal properties and metal ions can lead to increased bioavailability, uptake and efficacy. Development of such organometallic drugs may alleviate pressure on existing antifungal medications. Prodigious antimicrobial moieties such as azoles, Schiff bases, thiosemicarbazones and others reported herein lend themselves easily to the coordination of a host of metal ions, which can vastly improve the biocidal activity of the parent ligand, thereby extending the library of antifungal drugs available to medical professionals for treatment of an increasing incidence of fungal infections. Overall, this review shows the impressive but somewhat unexploited potential of metal-based compounds to treat fungal infections.
Collapse
Affiliation(s)
- Yan Lin
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Harley Betts
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Sarah Keller
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| |
Collapse
|
882
|
Chen TY, Tai YY, Chang LC, Wu PC. Fabrication, optimisation and evaluation of cisplatin-loaded nanostructured carriers for improved urothelium permeability for intravesical administration. J Microencapsul 2021; 38:405-413. [PMID: 34275419 DOI: 10.1080/02652048.2021.1957037] [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: 10/20/2022]
Abstract
AIM To design microemulsions as carriers to improve cisplatin permeation capability for intravesical administration. METHOD The response surface methodology with factorial design was used to investigate and optimise the influence of the compositions e.g. capryol 90 and 5-pentanediol/transcutol mixture on the permeation accumulation amount and tissue deposition amount of cisplatin-loaded microemulsions. The in vitro permeation study and in vivo intravesical test were conducted to prove the effect of microemulsions. RESULTS The droplet size and the viscosity of all drug-loaded formulations ranged 235.8-309.3 nm and 550.8-861.7 cps, respectively. The permeation accumulation amounts significantly increased about 26-fold, by used microemulsion as carriers. In vivo study, the cisplatin deposition amount in bladder tissue significantly increased 4.1-fold (p < 0.05) and the penetration depth increased from 60 μm up 120 μm. The nanocarrier showed considerable thermodynamic stability. CONCLUSION The designed nanocarrier was considered to be a promising delivery system for cisplatin intravesical administration.
Collapse
Affiliation(s)
- Ting-Yu Chen
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung City, Taiwan, ROC
| | - Yu-Yao Tai
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung City, Taiwan, ROC
| | - Li-Ching Chang
- School of Medicine for International Students, I-Shou University, Kaohsiung City, Taiwan, ROC
| | - Pao-Chu Wu
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung City, Taiwan, ROC.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan, ROC
| |
Collapse
|
883
|
Ruthenium Complexes as Promising Candidates against Lung Cancer. Molecules 2021; 26:molecules26154389. [PMID: 34361543 PMCID: PMC8348655 DOI: 10.3390/molecules26154389] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Lung cancer is one of the most common malignancies with the highest mortality rate and the second-highest incidence rate after breast cancer, posing a serious threat to human health. The accidental discovery of the antitumor properties of cisplatin in the early 1960s aroused a growing interest in metal-based compounds for cancer treatment. However, the clinical application of cisplatin is limited by serious side effects and drug resistance. Therefore, other transition metal complexes have been developed for the treatment of different malignant cancers. Among them, Ru(II/III)-based complexes have emerged as promising anticancer drug candidates due to their potential anticancer properties and selective cytotoxic activity. In this review, we summarized the latest developments of Ru(II/III) complexes against lung cancer, focusing mainly on the mechanisms of their biological activities, including induction of apoptosis, necroptosis, autophagy, cell cycle arrest, inhibition of cell proliferation, and invasion and metastasis of lung cancer cells.
Collapse
|
884
|
Li HT, Zhu X. Quinoline-based Compounds with Potential Activity against Drugresistant Cancers. Curr Top Med Chem 2021; 21:426-437. [PMID: 32552650 DOI: 10.2174/1568026620666200618113957] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/06/2020] [Accepted: 05/12/2020] [Indexed: 11/22/2022]
Abstract
Drug resistance is the major cause of the failure of cancer chemotherapy, so one of the most important features in developing effective cancer therapeutic strategies is to overcome drug resistance. Quinoline moiety has become one of the most privileged structural motifs in anticancer agent discovery since its derivatives possess potent activity against various cancers including drug-resistant cancers. Several quinoline-based compounds which are represented by Anlotinib, Bosutinib, Lenvatinib, and Neratinib have already been applied in clinical practice to fight against cancers, so quinoline-based compounds are potential anticancer agents. The present short review article provides an overview of the recent advances of quinoline-based compounds with potential activity against drug-resistant cancers. The structure-activity relationship and mechanisms of action are also discussed.
Collapse
Affiliation(s)
- Huan-Ting Li
- Department of Pharmacy, Baotou Medical College, Baotou, Inner Mongolia, 014040, China
| | - Xiaoyong Zhu
- Department of Oncology, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province 311800, China
| |
Collapse
|
885
|
Sun B, Hu N, Cong D, Chen K, Li J. MicroRNA-25-3p promotes cisplatin resistance in Non-small-cell lung carcinoma (NSCLC) through adjusting PTEN/PI3K/AKT route. Bioengineered 2021; 12:3219-3228. [PMID: 34266345 PMCID: PMC8806525 DOI: 10.1080/21655979.2021.1939577] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs exert crucial effects in the drug resistance. The purpose of this research was to investigate the miR-25-3p effects on DDP resistance in NSCLC. We used RT-qPCR to evaluate the expression of miR-25-3p. Cell growth was determined using MTS assay. Cellular bio-activity was analyzed via Colony formation, Annexin V/PI, and Transwell assay. Luciferase reporter assay was used to determine miR-25-3p and PTEN binding. Western blot was used to determine PTEN, PI3K, p-AKT/AKT expression. In-vivo study was used to determine the effects of miR-25-3p on the tumor growth. Expression of miR-25-3p is increased in NSCLC cisplatin resistant A549 and H1299 cells. Furthermore, miR-25-3p mimic enhanced drug resistance, and accelerated cell invasion and metastasis. Moreover, miR-25-3p mimic resulted in the activation of PTEN/PI3K/AKT pathway. However, miR-25-3p inhibitors exhibited the opposite trend. We further identified PTEN as a potential target of miR-25-3p. PTEN knockout promoted cisplatin resistance, while PTEN mimic displayed opposite effects. Interestingly, miR-25-3p further boosted cisplatin resistance cells in vivo, and miR-25-3p inhibitors reduced the in-vivo tumor volume. MiR-25-3p/PTEN/PI3K/AKT axis might accelerate DDP tolerance in NSCLC, which may serve as a potential target for chemotherapy resistance in NSCLC.
Collapse
Affiliation(s)
- Butong Sun
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun City, China
| | - Nanjun Hu
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun City, China
| | - Dan Cong
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun City, China
| | - Kang Chen
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun City, China
| | - Jun Li
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun City, China
| |
Collapse
|
886
|
Du J, Wang X, Li Y, Ren X, Zhou Y, Hu W, Zhou C, Jing Q, Yang C, Wang L, Li H, Fang L, Zhou Y, Tong X, Wang Y. DHA exhibits synergistic therapeutic efficacy with cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via modulation of iron metabolism. Cell Death Dis 2021; 12:705. [PMID: 34262021 PMCID: PMC8280115 DOI: 10.1038/s41419-021-03996-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal cancer with limited treatment options. Cisplatin (DDP) is used as a mainstay of chemotherapeutic agents in combination with other drugs or radiotherapy for PDAC therapy. However, DDP exhibits severe side-effects that can lead to discontinuation of therapy, and the acquired drug resistance of tumor cells presents serious clinical obstacles. Therefore, it is imperative to develop a more effective and less toxic therapeutic strategy. We and others have previously discovered that dihydroartemisinin (DHA) represents a safe and promising therapeutic agent to preferentially induce cancer cell ferroptosis. In the present study, we find that DHA could intensively strengthen the cytotoxicity of DDP and significantly reduce its effective concentrations both in vitro and in vivo. Combination of DHA and DDP synergistically inhibits the proliferation and induces DNA damage of PDAC cells. Mechanically, the combinative treatment impairs mitochondrial homeostasis, characterized by destroyed mitochondrial morphology, decreased respiratory capacity, reduced ATP production, and accumulated mitochondria-derived ROS. Further studies show that ferroptosis contributes to the cytotoxic effects in PDAC cells under the challenge of DHA and DDP, together with catastrophic accumulation of free iron and unrestricted lipid peroxidation. Moreover, pharmacologic depleting of the free iron reservoir or reconstituted expression of FTH contributes to the tolerance of DHA/DDP-induced ferroptosis, while iron addition accelerates the ferroptotic cell death. In summary, these results provide experimental evidence that DHA acts synergistically with DDP and renders PDAC cells vulnerable to ferroptosis, which may act as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Jing Du
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xu Wang
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Xueying Ren
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yi Zhou
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wanye Hu
- Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Chaoting Zhou
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qiangan Jing
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Chen Yang
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Luyang Wang
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huanjuan Li
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lijuan Fang
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Yonglie Zhou
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Xiangmin Tong
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Bengbu Medical College, Bengbu, Anhui, 233000, China.
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China.
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Ying Wang
- Bengbu Medical College, Bengbu, Anhui, 233000, China.
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China.
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
887
|
Ruthenium(II) and Platinum(II) Complexes with Biologically Active Aminoflavone Ligands Exhibit In Vitro Anticancer Activity. Int J Mol Sci 2021; 22:ijms22147568. [PMID: 34299199 PMCID: PMC8306828 DOI: 10.3390/ijms22147568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022] Open
Abstract
Continuing our studies on the mechanisms underlying the cytotoxicity of potential drugs, we have described several aspects of the in vitro anticancer activity of ruthenium(II) and platinum(II) complexes with bioactive, synthetic aminoflavone ligands. We examined the mechanism of proapoptotic activity of cis-dichlorobis(3-imino-2-methoxyflavanone)ruthenium(II), cis-dichlorobis(3-imino-2-ethoxyflavanone)ruthenium(II), and trans-dichlorobis(3-aminoflavone)platinum(II). Cisplatin was used as a reference compound. The cytotoxicity was investigated by MTT assay. The mechanism of proapoptotic activity of the tested compounds was investigated by evaluation of caspase-8 activity, cytometric analysis of annexin-V positive cells, and mitochondrial potential loss measurement. The results showed that ruthenium compounds break partially or completely the cisplatin resistance by activating the caspase 8-dependent apoptosis pathway and loss of mitochondrial membrane potential. Platinum compounds also have a cytostatic effect, but their action requires more exposure time. Potential mechanisms underlying drug resistance in the two pairs of cancer cell lines were investigated: total glutathione content, P-glycoprotein activity, and differences in the activity of DNA repair induced by nucleotide excision. Results showed that cisplatin-resistant cells have elevated glutathione levels relative to sensitive cells. Moreover, they indicated the mechanisms enabling cells to avoid apoptosis caused by DNA damage. Pg-P activity has no effect on the development of cisplatin resistance in the cell lines described.
Collapse
|
888
|
Byun EB, Song HY, Kim WS, Han JM, Seo HS, Park SH, Kim K, Byun EH. Protective Effect of Polysaccharides Extracted from Cudrania tricuspidata Fruit against Cisplatin-Induced Cytotoxicity in Macrophages and a Mouse Model. Int J Mol Sci 2021; 22:ijms22147512. [PMID: 34299130 PMCID: PMC8304288 DOI: 10.3390/ijms22147512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022] Open
Abstract
Although cisplatin is one of most effective chemotherapeutic drugs that is widely used to treat various types of cancer, it can cause undesirable damage in immune cells and normal tissue because of its strong cytotoxicity and non-selectivity. This study was conducted to investigate the cytoprotective effects of Cudrania tricuspidata fruit-derived polysaccharides (CTPS) against cisplatin-induced cytotoxicity in macrophages, lung cancer cell lines, and a mouse model, and to explore the possibility of application of CTPS as a supplement for anticancer therapy. Both cisplatin alone and cisplatin with CTPS induced a significant cytotoxicity in A549 and H460 lung cancer cells, whereas cytotoxicity was suppressed by CTPS in cisplatin-treated RAW264.7 cells. CTPS significantly attenuated the apoptotic and necrotic population, as well as cell penetration in cisplatin-treated RAW264.7 cells, which ultimately inhibited the upregulation of Bcl-2-associated X protein (Bax), cytosolic cytochrome c, poly (adenosine diphosphateribose) polymerase (PARP) cleavage, and caspases-3, -8, and -9, and the downregulation of B cell lymphoma-2 (Bcl-2). The CTPS-induced cytoprotective action was mediated with a reduction in reactive oxygen species production and mitochondrial transmembrane potential loss in cisplatin-treated RAW264.7 cells. In agreement with the results obtained above, CTPS induced the attenuation of cell damage in cisplatin-treated bone marrow-derived macrophages (primary cells). In in vivo studies, CTPS significantly inhibited metastatic colonies and bodyweight loss as well as immunotoxicity in splenic T cells compared to the cisplatin-treated group in lung metastasis-induced mice. Furthermore, CTPS decreased the level of CRE and BUN in serum. In summation, these results suggest that CTPS-induced cytoprotective action may play a role in alleviating the side effects induced by chemotherapeutic drugs.
Collapse
Affiliation(s)
- Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Ha-Yeon Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Korea;
| | - Jeong Moo Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Ho Seong Seo
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Sang-Hyun Park
- Department of Food Science and Technology, Kongju National University, Yesan 32439, Korea; (S.-H.P.); (K.K.)
| | - Kwangwook Kim
- Department of Food Science and Technology, Kongju National University, Yesan 32439, Korea; (S.-H.P.); (K.K.)
| | - Eui-Hong Byun
- Department of Food Science and Technology, Kongju National University, Yesan 32439, Korea; (S.-H.P.); (K.K.)
- Correspondence: ; Tel.: +82-413-301-481; Fax: +82-413-301-489
| |
Collapse
|
889
|
Li B, Zhou M, Wang J, Xu H, Yang M. Suppressing ERK Pathway Impairs Glycochenodeoxycholate-Mediated Survival and Drug-Resistance in Hepatocellular Carcinoma Cells. Front Oncol 2021; 11:663944. [PMID: 34327135 PMCID: PMC8313996 DOI: 10.3389/fonc.2021.663944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/22/2021] [Indexed: 12/02/2022] Open
Abstract
Glycochenodeoxycholate (GCDA), a toxic component in bile salts, is involved in carcinogenesis of gastrointestinal tumors. The objective of this research was to study the function of ERK1/2 in the GCDA-mediated survival and drug-resistance in hepatocellular carcinoma cells (HCCs). Firstly, extracellular signal-regulated kinase 1/2 (ERK1/2) was detected extensively expressed in liver cancer cells, and silencing ERK1/2 by RNA interference could suppress GCDA-stimulated survival and promote apoptosis. Furthermore, phosphorylation of endogenous ERK1/2 could be potently stimulated by GCDA in combination with enhanced chemoresistance in QGY-7703 hepatocellular carcinoma cells. The GCDA-mediated proliferation and chemoresistance could be impaired by PD98059, which acted as an inhibitor to block the phosphorylation of ERK1/2. Mechanistically, PD98059 was able to potently suppress GCDA-stimulated nuclear aggregation of ERK1/2 and p-ERK1/2, upregulate pro-survival protein Mcl-1 and downregulate pro-apoptotic protein Bim. The results of this study indicated that disruption of ERK1/2 by blocking phosphorylation or nuclear translocation may put forward new methods for solving the problem of GCDA-related proliferation and drug-resistance in liver cancer treatment.
Collapse
Affiliation(s)
- Bingxin Li
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| | - Maojun Zhou
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, National Center for Geriatrics Clinical Research, State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Jue Wang
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongjuan Xu
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| | - Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
890
|
Cisplatin-functionalized nanodiamonds: preparation and characterization, with potential antineoplastic application. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-01955-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
891
|
Programmed cell death, redox imbalance, and cancer therapeutics. Apoptosis 2021; 26:385-414. [PMID: 34236569 DOI: 10.1007/s10495-021-01682-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/06/2023]
Abstract
Cancer cells are disordered by nature and thus featured by higher internal redox level than healthy cells. Redox imbalance could trigger programmed cell death if exceeded a certain threshold, rendering therapeutic strategies relying on redox control a possible cancer management solution. Yet, various programmed cell death events have been consecutively discovered, complicating our understandings on their associations with redox imbalance and clinical implications especially therapeutic design. Thus, it is imperative to understand differences and similarities among programmed cell death events regarding their associations with redox imbalance for improved control over these events in malignant cells as well as appropriate design on therapeutic approaches relying on redox control. This review addresses these issues and concludes by bringing affront cold atmospheric plasma as an emerging redox controller with translational potential in clinics.
Collapse
|
892
|
Shueng PW, Yu LY, Chiu HC, Chang HC, Chiu YL, Kuo TY, Yen YW, Lo CL. Early phago-/endosomal escape of platinum drugs via ROS-responsive micelles for dual cancer chemo/immunotherapy. Biomaterials 2021; 276:121012. [PMID: 34252800 DOI: 10.1016/j.biomaterials.2021.121012] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/01/2021] [Accepted: 07/04/2021] [Indexed: 02/06/2023]
Abstract
Recent studies have indicated that cancer treatment based on immunotherapy alone is not viable. Combined treatment with other strategies is required to achieve the expected therapeutic effect. Reactive oxygen species (ROS) play an important role in regulating cancer cells and the tumor microenvironment, even in immune cells. However, rigorous regulation of the ROS level within the entire tumor tissue is difficult, limiting the application of ROS in cancer therapy. Therefore, we design an early phago-/endosome-escaping micelle that can release platinum-based drugs into the cytoplasm of macrophages and cancer cells, thereby enhancing the ROS levels of the entire tumor tissue; inducing apoptosis of cancer cells, down-regulation of CD47 expression of cancer cells, polarization of M1 macrophages, and phagocytosis of cancer cells by M1 macrophages; and achieving the dual effect of chemotherapy and macrophage-mediated immunotherapy.
Collapse
Affiliation(s)
- Pei-Wei Shueng
- Division of Radiation Oncology, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan, ROC; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan, ROC; Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 112, Taiwan, ROC
| | - Lu-Yi Yu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC; Department of Biomedical Engineering, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Hsin-Cheng Chiu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing-Hua University, Hsinchu, 300, Taiwan, ROC
| | - Hui-Ching Chang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC; Department of Biomedical Engineering, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Yen-Ling Chiu
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan City, 320, Taiwan, ROC; Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, 100, Taiwan, ROC; Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan, ROC
| | - Tzu-Yu Kuo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC; Department of Biomedical Engineering, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Yu-Wei Yen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC; Department of Biomedical Engineering, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Chun-Liang Lo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC; Department of Biomedical Engineering, National Yang-Ming University, Taipei, 112, Taiwan, ROC; Center for Advanced Pharmaceutics and Drug Delivery Research, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, ROC.
| |
Collapse
|
893
|
Zhao X, Liu Y, Chen X, Mi Z, Li W, Wang P, Shan X, Lu X. Detection and Characterization of Single Cisplatin Adducts on DNA by Nanopore Sequencing. ACS OMEGA 2021; 6:17027-17034. [PMID: 34250360 PMCID: PMC8264939 DOI: 10.1021/acsomega.1c02106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/07/2021] [Indexed: 05/13/2023]
Abstract
Detection and characterization of an individual cisplatin adduct on a single DNA molecule is a demanding task. We explore the characteristic features of cisplatin adducts in the nanopore sequencing signal in aspects of dwell time, genome anchored current trace, and basecalling accuracy. The offset between the motor protein and the nanopore constriction region is revealed by dwell time analysis to be about 14 bases in the nanopore device as we examined. Characteristic distortions due to cisplatin adducts are illustrated in genome anchored current trace analysis, constituting the fingerprint for identification of cisplatin adduct. The sharp increase in odds ratio at the location of adducting sites provides additional feature in the detection of the adduct. By these combined methods, single cisplatin adducts can be detected with high fidelity on a single read of the DNA sequence. The study demonstrates an effective method in the detection and characterization of single cisplatin adducts on DNA at the single-molecule level and with single nucleotide spatial resolution.
Collapse
Affiliation(s)
- Xinjia Zhao
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
- School
of Physical Sciences, University of Chinese
Academy of Sciences, Beijing 100190, China
| | - Yuru Liu
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoyu Chen
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
- School
of Physical Sciences, University of Chinese
Academy of Sciences, Beijing 100190, China
| | - Zhuang Mi
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
- School
of Physical Sciences, University of Chinese
Academy of Sciences, Beijing 100190, China
| | - Wei Li
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Pengye Wang
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
- School
of Physical Sciences, University of Chinese
Academy of Sciences, Beijing 100190, China
- Songshan
Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| | - Xinyan Shan
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Xinghua Lu
- Beijing
National Laboratory for Condensed-Matter Physics and Institute of
Physics, Chinese Academy of Sciences, Beijing 100190, China
- School
of Physical Sciences, University of Chinese
Academy of Sciences, Beijing 100190, China
- Center
for Excellence in Topological Quantum Computation, Chinese Academy of Sciences, Beijing 100190, China
- Songshan
Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| |
Collapse
|
894
|
Erfani Majd N, Tabandeh MR, Hosseinifar SH, Sadeghi M. Protective Effect of Aloe vera Gel against Cisplatin-Induced Testicular Damage, Sperm Alteration and Oxidative stress in Rats. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2021; 15:210-218. [PMID: 34155868 PMCID: PMC8233928 DOI: 10.22074/ijfs.2020.134691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/02/2020] [Indexed: 11/12/2022]
Abstract
BACKGROUND Cisplatin (CIS) is an effective antineoplas tic drug that is used to treat various types of cancers. However, it causes side effects on the male reproductive sys tem. The present s tudy aimed to inves tigate the possible protective effects of Aloe vera (AL) gel (known as an antioxidant plant) on CIS-induced changes in rat sperm parameters, tes ticular s tructure, and oxidative s tress markers. MATERIALS AND METHODS In this experimental study, forty-eight adult male rats were divided into 6 groups including: control, CIS, AL, metformin (MET), CIS+AL, and CIS+MET. CIS was used intraperitoneally at a dose of 5 mg/kg on days 7, 14, 21, and 28 of the experiment. AL gel (400 mg/kg per day) and MET (200 mg/kg per day) were administered orally for 35 days (started one week before the beginning of the experiment). Testes weight and dimensions, and morphometrical and histological alterations, activities of antioxidant enzymes including superoxide dismutase (SOD) and glutathione peroxidase (GPx), serum testosterone concentration, lipid peroxidation level, and sperm parameters were examined. RESULTS CIS caused a significant decrease (P<0.05) in relative weight and dimension of the testis, germinal epithelium thickness and diameter of seminiferoustubules, the numbers of testicular cells, and spermatogenesis indexes. The malondialdehyde (MDA) levels increased and antioxidant enzymes activities decreased in the CIS group compared to the control group (P<0.05). Additionally, sperm parameters (concentration, viability, motility, and normal morphology), and testosterone levels reduced significantly in CIS-treated rats (P<0.05). Also, CIS induced histopathological damages including disorganization, desquamation, atrophy, and vacuolation in the testis. However, administration of AL gel to CIS-treated rats attenuated the CIS-induced alterations, mitigated testicular oxidative stress and increased testosterone concentration. CONCLUSION The results suggest that AL as a potential antioxidant plant and due to free radicals scavenging activities, has a protective effect against CIS-induced testicular alterations.
Collapse
Affiliation(s)
- Naeem Erfani Majd
- Department of Basic Sciences, Histology Section, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Stem Cell and Transgenic Technology Research Center, Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Reza Tabandeh
- Stem Cell and Transgenic Technology Research Center, Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - S Hima Hosseinifar
- Department of Basic Sciences, Histology Section, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mahin Sadeghi
- Department of Basic Sciences, Histology Section, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
895
|
Zhou S, Zhou Y, Yu J, Jiang L, Xiang Y, Wang J, Du Y, Cui X, Ge F. A neutral polysaccharide from Ophiocordyceps lanpingensis restrains cisplatin-induced nephrotoxicity. Food Sci Nutr 2021; 9:3602-3616. [PMID: 34262721 PMCID: PMC8269674 DOI: 10.1002/fsn3.2317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 04/18/2021] [Indexed: 12/11/2022] Open
Abstract
Ophiocordyceps lanpingensis is an edible mushroom distributed over the south-eastern part of the Tibet Plateau, which is also recognized as an effective ethnomedicine to alleviate diseases. This study explored the effects of a kind of Ophiocordyceps lanpingensis neutral polysaccharide (ONP) on RAW264.7 macrophages and cisplatin-induced nephrotoxicity. The results showed that ONP relieved the inflammatory response of RAW264.7 macrophages by increasing the expression level of anti-inflammatory factor IL-10. Furthermore, ONP treatment significantly prolonged the survival of the mice treated by cisplatin through decelerating pathological progress and alleviating damaged functions of the kidneys. Compared with the cisplatin group, ONP reduced the oxidative stress of the renal cells and the expression levels of pro-inflammatory factors. Apoptosis of renal cells was also weakened in the ONP treatment group. These findings indicated that ONP alleviated cisplatin nephrotoxicity mainly by inhibiting oxidative stress, inflammation, and apoptosis in the kidneys, underscoring the potential of ONP supplementation to alleviate the side effects of cisplatin chemotherapy.
Collapse
Affiliation(s)
- Shubo Zhou
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Yongchun Zhou
- Yunnan Cancer Center Molecular Diagnostics CenterYunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Jiaji Yu
- Department of Microbiology, Immunology & Molecular GeneticsUniversity of CaliforniaLos AngelesCAUSA
| | - Li Jiang
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Yingying Xiang
- Department of StomatologyYan’an Hospital Affiliated to Kunming Medical UniversityKunmingChina
| | - Juan Wang
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Yaxi Du
- Yunnan Cancer Center Molecular Diagnostics CenterYunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Xiuming Cui
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Feng Ge
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| |
Collapse
|
896
|
Yu P, Li X, Cheng G, Zhang X, Wu D, Chang J, Wang S. Hydrogen peroxide-generating nanomedicine for enhanced chemodynamic therapy. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
897
|
Marzo T, La Mendola D. The Effects on Angiogenesis of Relevant Inorganic Chemotherapeutics. Curr Top Med Chem 2021; 21:73-86. [PMID: 33243124 DOI: 10.2174/1568026620666201126163436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiogenesis is a key process allowing the formation of blood vessels. It is crucial for all the tissues and organs, ensuring their function and growth. Angiogenesis is finely controlled by several mechanisms involving complex interactions between pro- or antiangiogenic factors, and an imbalance in this control chain may result in pathological conditions. Metals as copper, zinc and iron cover an essential role in regulating angiogenesis, thus therapies having physiological metals as target have been proposed. In addition, some complexes of heavier metal ions (e.g., Pt, Au, Ru) are currently used as established or experimental anticancer agents targeting genomic or non-genomic targets. These molecules may affect the angiogenic mechanisms determining different effects that have been only poorly and non-systematically investigated so far. Accordingly, in this review article, we aim to recapitulate the impact on the angiogenic process of some reference anticancer drugs, and how it is connected to the overall pharmacological effects. In addition, we highlight how the activity of these drugs can be related to the role of biological essential metal ions. Overall, this may allow a deeper description and understanding of the antineoplastic activity of both approved or experimental metal complexes, providing important insights for the synthesis of new inorganic drugs able to overcome resistance and recurrence phenomena.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
898
|
Pettenuzzo A, Vezzù K, Di Paolo ML, Fotopoulou E, Marchiò L, Via LD, Ronconi L. Design, physico-chemical characterization and in vitro biological activity of organogold(III) glycoconjugates. Dalton Trans 2021; 50:8963-8979. [PMID: 34110336 DOI: 10.1039/d1dt01100f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To develop new metal-based glycoconjugates as potential anticancer agents, four organometallic gold(iii)-dithiocarbamato glycoconjugates of the type [AuIII(2-Bnpy)(SSC-Inp-GlcN)](PF6) (2-Bnpy: 2-benzylpyridine; Inp: isonipecotic moiety; GlcN: amino-glucose scaffold; Au3-Au6) and the corresponding model non-glycosylated counterparts [AuIII(2-Bnpy)(SSC-Inp-R)](PF6) (R: OEt (Au1), NH2 (Au2)) have been generated and characterized by means of several analytical techniques (elemental analysis, FT-IR, 1H-/13C-NMR, ESI-MS, UV-Vis, X-ray crystallography). Their stability under physiologically-relevant conditions (PBS solution) and n-octanol/PBS distribution coefficient (D7.4) have also been evaluated. Gold(iii) glycoconjugates showed an antiproliferative effect against ovarian carcinoma A2780 cells, with GI50 values in the low micromolar range. Remarkably, their cell growth inhibitory effect increases upon the addition of a glucose transporter 1 (GLUT1) inhibitor, thus ruling out the involvement of GLUT1 in their transport inside the cell. Additional mechanistic studies have been carried out in A2780 cells, supporting the hypothesis of a facilitated diffusion mechanism (possibly mediated by glucose transporters other than GLUT1), and revealing their capability to act as topoisomerase I and II inhibitors and to disrupt mitochondrial membrane integrity, leading to the generation of ROS, thus resulting in the promotion of oxidative stress and, eventually, cell death.
Collapse
Affiliation(s)
- Andrea Pettenuzzo
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| | - Keti Vezzù
- University of Padova, Department of Industrial Engineering, Via F. Marzolo 8, 35131 Padova, Italy
| | - Maria Luisa Di Paolo
- University of Padova, Department of Molecular Medicine, Via G. Colombo 3, 35131 Padova, Italy
| | - Eirini Fotopoulou
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| | - Luciano Marchiò
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parco Area delle Scienze 11/a, 43124 Parma, Italy
| | - Lisa Dalla Via
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy.
| | - Luca Ronconi
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| |
Collapse
|
899
|
Yang K, Yu G, Yang Z, Yue L, Zhang X, Sun C, Wei J, Rao L, Chen X, Wang R. Supramolecular Polymerization‐Induced Nanoassemblies for Self‐Augmented Cascade Chemotherapy and Chemodynamic Therapy of Tumor. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Kuikun Yang
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| | - Guocan Yu
- Key Lab of Organic Optoelectronics & Molecular Engineering Department of Chemistry Tsinghua University Beijing China
| | - Zhiqing Yang
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| | - Ludan Yue
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| | - Xiangjun Zhang
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| | - Chen Sun
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| | - Jianwen Wei
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering Shenzhen Bay Laboratory Shenzhen China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering Yong Loo Lin School of Medicine and Faculty of Engineering National University of Singapore Singapore Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Nanomedicine Translational Research Program NUS Center for Nanomedicine Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Science University of Macau Avenida da Universidade Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology University of Macau Avenida da Universidade Taipa Macau China
| |
Collapse
|
900
|
Combined Action of Anti-MUC1 Monoclonal Antibody and Pyrazole-Platinum(II) Complexes Reveals Higher Effectiveness towards Apoptotic Response in Comparison with Monotherapy in AGS Gastric Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13070968. [PMID: 34206951 PMCID: PMC8309157 DOI: 10.3390/pharmaceutics13070968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
MUC1 mucin is a transmembrane glycoprotein aberrantly overexpressed and underglycosylated in most epithelium origin cancers. Combining chemotherapeutics with monoclonal antibodies toward cancer-related antigens is one of the new strategies in cancer therapies. In this study, we assessed the effectiveness of 10 μM cisplatin (cisPt), two pyrazole-platinum(II) complexes (PtPz4 and PtPz6), and 5 μg/mL anti-MUC1 used as monotherapy, as well as cisplatin and its derivatives combined with mAb on apoptotic response and specific cancer-related sugar antigens in AGS gastric cancer cells. Flow cytometry, RT-PCR, Western blotting, and ELISA tests were applied to determine the influence of examined compounds on analyzed factors. PtPz6 combined with anti-MUC1 revealed the strongest apoptotic response compared to control and monotherapy. The combined action of both cisPt derivatives and anti-MUC1 was more effective than monotherapy in relation to Bad, Bcl-xL, Bcl-2, caspase-9, caspase-3, as well as pro- and cleaved caspase-3 protein, and T, sialyl Tn sugar antigens in cell lysates, and Tn, T, sialyl Tn, sialyl T antigens in culture medium. Additionally, PtPz4 administrated with mAb was revealed to be more potent than used alone with regard to Bax protein and Bid expression, and PtPz6 used in complex with anti-MUC1 revealed more efficient action towards Akt and sialyl T antigen expression. These data indicate the rationality of the potential application of combined treatment of anti-MUC1 and cisPt derivatives in gastric cancer therapy.
Collapse
|