1
|
Shadnoush M, Momenan M, Seidel V, Tierling S, Fatemi N, Nazemalhosseini-Mojarad E, Norooz MT, Cheraghpour M. A comprehensive update on the potential of curcumin to enhance chemosensitivity in colorectal cancer. Pharmacol Rep 2024:10.1007/s43440-024-00652-y. [PMID: 39304638 DOI: 10.1007/s43440-024-00652-y] [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: 01/15/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a major cause of cancer-related mortality worldwide. The efficacy of chemotherapy agents in CRC treatment is often limited due to toxic side effects, heterogeneity of cancer cells, and the possibility of chemoresistance which promotes cancer cell survival through several mechanisms. Combining chemotherapy agents with natural compounds like curcumin, a polyphenol compound from the Curcuma longa plant, has been reported to overcome chemoresistance and increase the sensitivity of cancer cells to chemotherapeutics. Curcumin, alone or in combination with chemotherapy agents, has been demonstrated to prevent chemoresistance by modulating various signaling pathways, reducing the expression of drug resistance-related genes. The purpose of this article is to provide a comprehensive update on studies that have investigated the ability of curcumin to enhance the efficacy of chemotherapy agents used in CRC. It is hoped that it can serve as a template for future research on the efficacy of curcumin, or other natural compounds, combined with chemotherapy agents to maximize the effectiveness of therapy and reduce the side effects that occur in CRC or other cancers.
Collapse
Affiliation(s)
- Mahdi Shadnoush
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.Box, Tehran, 16635-148, Iran
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Momenan
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Sascha Tierling
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | - Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.Box, Tehran, 16635-148, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Tayefeh Norooz
- General Surgery Department, Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Makan Cheraghpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.Box, Tehran, 16635-148, Iran.
| |
Collapse
|
2
|
Dagher D, Elmansi H, Nasr JJ, El-Enany N. Investigation of green synchronous spectrofluorimetric approach for facile sensitive estimation of two co-administered anti-cancer drugs; curcumin and doxorubicin in their laboratory-prepared mixtures, human plasma, and urine. BMC Chem 2024; 18:164. [PMID: 39252071 PMCID: PMC11385172 DOI: 10.1186/s13065-024-01272-6] [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: 05/30/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Recently, phytochemicals play an important role in cancer management. Curcumin (CUR), a natural phytochemical, has been co-administered with widespread chemotherapeutic agents such as doxorubicin (DOX) due to its excellent antitumor activity and the ability to lower the adverse reactions and drug resistance cells associated with DOX use. The present study aims to determine DOX and CUR utilizing a label-free, selective, sensitive, and precise synchronous spectrofluorimetric method. The obvious overlap between the emission spectra of DOX and CUR prevents simultaneous estimation of both analytes by conventional spectrofluorimetry. To solve such a problem, synchronous spectrofluorimetric measurements were recorded at Δλ = 20 nm, utilizing ethanol as a diluting solvent. Curcumin was recorded at 442.5 nm, whereas DOX was estimated at 571.5 nm, each at the zero-crossing point of the other one. The developed method exhibited linearity over a concentration range of 0.04-0.40 μg/mL for CUR and 0.05-0.50 μg/mL for DOX, respectively. The values of limit of detection (LOD) were 0.009 and 0.012 µg/mL, while the values of limit of quantitation (LOQ) were 0.028 and 0.037 µg/mL for CUR and DOX, respectively. The adopted approach was carefully validated according to the guidelines of ICH Q2R1. The method was utilized to estimate CUR and DOX in laboratory-prepared mixtures and human biological matrices. It showed a high percentage of recoveries with minimal RSD values. Additionally, three different tools were utilized to evaluate the greenness of the proposed approach.
Collapse
Affiliation(s)
- Diaa Dagher
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Heba Elmansi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Jenny Jeehan Nasr
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura National University, Gamasa, 7723730, Egypt
| | - Nahed El-Enany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Mansoura University, New Mansoura, 7723730, Egypt
| |
Collapse
|
3
|
Shati AA, Eid RA, El-kott AF, Alqahtani YA, Shatoor AS, Ahmed Zaki MS. Curcumin attenuates doxorubicin-induced cardiotoxicity via suppressing oxidative Stress, preventing inflammation and apoptosis: Ultrastructural and computational approaches. Heliyon 2024; 10:e27164. [PMID: 38468941 PMCID: PMC10926088 DOI: 10.1016/j.heliyon.2024.e27164] [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: 06/12/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Currently, doxorubicin (DOX) is one of the medications commonly used in chemotherapy to treat different types of tumors.Nonetheless, despite being effective in multiple tumors, yet its use is limited owing to its cytotoxic effects, the therapeutic use of DOX has been limited. This work aimed to explore whether curcumin (CMN) can prevents DOX-induced cardiotoxicity in rats. Four groups of rats were created, with the first functioning as a control, while the second group received CMN. DOX alone was administered to the third group, whereas CMN and DOX were administered to the fourth group. Lipid peroxidation assessed as Malondialdehyde (MDA), aspartate aminotransferase (AST), alanine aminotransferase (ALT), oxidative stress markers as catalase (CAT), superoxide dismutase (SOD), and inflammatory markers as tumor necrosis factor-alpha (TNF-α) in heart homogenates, each one was assessed. Heart specimens was investigated histologically and ultrastructurally. Increased, AST, and ALT serum levels, increased MDA levels, decreased SOD and CAT levels, and increased TNF-α concentrations in heart homogenates were all signs of DOX-induced myocardial injury. Histological and ultrastructural examinations revealed vacuoles and larger, swollen mitochondria in the cytoplasm. Furthermore, DOX caused significant changes in the myocardium, most notably nuclei disintegration, myofibrillar loss, and myocyte vacuolization. Using CMN with DOX reduced the harmful consequences of DOX on the myocardium by returning the increased AST and ALT levels to their original levels as compared to the control and reducing them. In cardiac tissue, CMN significantly increased the concentrations of SOD and CAT and significantly decreased the concentrations of MDA and TNF-α. Biochemical and histological studies have demonstrated that CMN has a heart-protective effect that might be related to its antioxidant and anti-inflammatory capabilities.
Collapse
Affiliation(s)
- Ayed A. Shati
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Refaat A. Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Attalla F. El-kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Youssef A. Alqahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abdullah S. Shatoor
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
4
|
Liao Y, Meng Q. Protection against cancer therapy-induced cardiovascular injury by planed-derived polyphenols and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 238:116896. [PMID: 37586453 DOI: 10.1016/j.envres.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Cancer therapy-induced heart injury is a significant concern for cancer patients undergoing chemotherapy, radiotherapy, immunotherapy, and also targeted molecular therapy. The use of these treatments can lead to oxidative stress and cardiomyocyte damage in the heart, which can result in heart failure and other cardiac complications. Experimental studies have revealed that chemotherapy drugs such as doxorubicin and cyclophosphamide can cause severe side effects such as cardiac fibrosis, electrophysiological remodeling, chronic oxidative stress and inflammation, etc., which may increase risk of cardiac disorders and attacks for patients that underwent chemotherapy. Similar consequences may also be observed for patients that undergo radiotherapy for left breast or lung malignancies. Polyphenols, a group of natural compounds with antioxidant and anti-inflammatory properties, have shown the potential in protecting against cancer therapy-induced heart injury. These compounds have been found to reduce oxidative stress, necrosis and apoptosis in the heart, thereby preserving cardiac function. In recent years, nanoparticles loaded with polyphenols have also provided for the delivery of these compounds and increasing their efficacy in different organs. These nanoparticles can improve the bioavailability and efficacy of polyphenols while minimizing their toxicity. This review article summarizes the current understanding of the protective effects of polyphenols and nanoparticles loaded with polyphenols against cancer therapy-induced heart injury. The article discusses the mechanisms by which polyphenols protect the heart, including antioxidant and anti-inflammation abilities. The article also highlights the potential benefits of using nanoparticles for the delivery of polyphenols.
Collapse
Affiliation(s)
- Yunshu Liao
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China
| | - Qinghua Meng
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
5
|
Singh M, Kadhim MM, Turki Jalil A, Oudah SK, Aminov Z, Alsaikhan F, Jawhar ZH, Ramírez-Coronel AA, Farhood B. A systematic review of the protective effects of silymarin/silibinin against doxorubicin-induced cardiotoxicity. Cancer Cell Int 2023; 23:88. [PMID: 37165384 PMCID: PMC10173635 DOI: 10.1186/s12935-023-02936-4] [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: 01/17/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Although doxorubicin chemotherapy is commonly applied for treating different malignant tumors, cardiotoxicity induced by this chemotherapeutic agent restricts its clinical use. The use of silymarin/silibinin may mitigate the doxorubicin-induced cardiac adverse effects. For this aim, the potential cardioprotective effects of silymarin/silibinin against the doxorubicin-induced cardiotoxicity were systematically reviewed. METHODS In this study, we performed a systematic search in accordance with PRISMA guideline for identifying all relevant studies on "the role of silymarin/silibinin against doxorubicin-induced cardiotoxicity" in different electronic databases up to June 2022. Sixty-one articles were obtained and screened based on the predefined inclusion and exclusion criteria. Thirteen eligible papers were finally included in this review. RESULTS According to the echocardiographic and electrocardiographic findings, the doxorubicin-treated groups presented a significant reduction in ejection fraction, tissue Doppler peak mitral annulus systolic velocity, and fractional shortening as well as bradycardia, prolongation of QT and QRS interval. However, these echocardiographic abnormalities were obviously improved in the silymarin plus doxorubicin groups. As well, the doxorubicin administration led to induce histopathological and biochemical changes in the cardiac cells/tissue; in contrast, the silymarin/silibinin co-administration could mitigate these induced alterations (for most of the cases). CONCLUSION According to the findings, it was found that the co-administration of silymarin/silibinin alleviates the doxorubicin-induced cardiac adverse effects. Silymarin/silibinin exerts its cardioprotective effects via antioxidant, anti-inflammatory, anti-apoptotic activities, and other mechanisms.
Collapse
Affiliation(s)
- Mandeep Singh
- Department of Physical Education, University of Jammu, Srinagar, Jammu, India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Zhang Y, Sultonova RD, You SH, Choi Y, Kim SY, Lee WS, Seong J, Min JJ, Hong Y. The anticancer effect of PASylated calreticulin-targeting L-ASNase in solid tumor bearing mice with immunogenic cell death-inducing chemotherapy. Biochem Pharmacol 2023; 210:115473. [PMID: 36863616 DOI: 10.1016/j.bcp.2023.115473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
L-Asparaginase (L-ASNase), a bacterial enzyme that degrades asparagine, has been commonly used in combination with several chemical drugs to treat malignant hematopoietic cancers such as acute lymphoblastic leukemia (ALL). In contrast, the enzyme was known to inhibit the growth of solid tumor cells in vitro, but not to be effective in vivo. We previously reported that two novel monobodies (CRT3 and CRT4) bound specifically with calreticulin (CRT) exposed on tumor cells and tissues during immunogenic cell death (ICD). Here, we engineered L-ASNases conjugated with monobodies at the N-termini and PAS200 tags at the C-termini (CRT3LP and CRT4LP). These proteins were expected to possess four monobody and PAS200 tag moieties, which did not disrupt the L-ASNase conformation. These proteins were expressed 3.8-fold more highly in E. coli than those without PASylation. The purified proteins were highly soluble, with much greater apparent molecular weights than expected ones. Their affinity (Kd) against CRT was about 2 nM, 4-fold higher than that of monobodies. Their enzyme activity (∼6.5 IU/nmol) was similar to that of L-ASNase (∼7.2 IU/nmol), and their thermal stability was significantly increased at 55 °C. Their half-life times were > 9 h in mouse sera, about 5-fold longer than that of L-ASNase (∼1.8 h). Moreover, CRT3LP and CRT4LP bound specifically with CRT exposed on tumor cells in vitro, and additively suppressed the tumor growth in CT-26 and MC-38 tumor-bearing mice treated with ICD-inducing drugs (doxorubicin and mitoxantrone) but not with a non-ICD-inducing drug (gemcitabine). All data indicated that PASylated CRT-targeted L-ASNases enhanced the anticancer efficacy of ICD-inducing chemotherapy. Taken together, L-ASNase would be a potential anticancer drug for treating solid tumors.
Collapse
Affiliation(s)
- Ying Zhang
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Rukhsora D Sultonova
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Sung-Hwan You
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - So-Young Kim
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Wan-Sik Lee
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Jihyoun Seong
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea.
| | - Yeongjin Hong
- Institute for Molecular Imaging and Theranostics, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun, Republic of Korea.
| |
Collapse
|
7
|
Lu Y, Feng N, Du Y, Yu R. Nanoparticle-Based Therapeutics to Overcome Obstacles in the Tumor Microenvironment of Hepatocellular Carcinoma. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162832. [PMID: 36014696 PMCID: PMC9414814 DOI: 10.3390/nano12162832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 05/09/2023]
Abstract
Hepatocellular carcinoma (HCC) is still a main health concern around the world, with a rising incidence and high mortality rate. The tumor-promoting components of the tumor microenvironment (TME) play a vital role in the development and metastasis of HCC. TME-targeted therapies have recently drawn increasing interest in the treatment of HCC. However, the short medication retention time in TME limits the efficiency of TME modulating strategies. The nanoparticles can be elaborately designed as needed to specifically target the tumor-promoting components in TME. In this regard, the use of nanomedicine to modulate TME components by delivering drugs with protection and prolonged circulation time in a spatiotemporal manner has shown promising potential. In this review, we briefly introduce the obstacles of TME and highlight the updated information on nanoparticles that modulate these obstacles. Furthermore, the present challenges and future prospects of TME modulating nanomedicines will be briefly discussed.
Collapse
Affiliation(s)
- Yuanfei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Na Feng
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
- Correspondence: (Y.D.); (R.Y.); Tel.: +86-571-88208435 (Y.D.); +86-571-87783925 (R.Y.)
| | - Risheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
- Correspondence: (Y.D.); (R.Y.); Tel.: +86-571-88208435 (Y.D.); +86-571-87783925 (R.Y.)
| |
Collapse
|
8
|
Armandeh M, Bameri B, Samadi M, Heidari S, Foroumad R, Abdollahi M. A systematic review of nonclinical studies on the effect of curcumin in chemotherapy-induced cardiotoxicity. Curr Pharm Des 2022; 28:1843-1853. [PMID: 35570565 DOI: 10.2174/1381612828666220513125312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Various anticancer drugs are effective therapeutic agents for cancer treatment; however, they cause severe toxicity in body organs. Cardiotoxicity is one of the most critical side effects of these drugs. Based on various findings, turmeric extract has positive effects on cardiac cells. OBJECTIVE This study aims to evaluate how curcumin as the main component of turmeric may affect chemotherapy-induced cardiotoxicity. METHOD Database search was performed up to April 2021 using "curcumin OR turmeric OR Curcuma longa" and "chemotherapy-induced cardiac disease," including all their equivalents and similar terms. After screening the total articles obtained from the electronic databases, 25 relevant articles were included in this systematic review. RESULTS The studies demonstrate lower body weight and increased mortality rates due to doxorubicin administration. Besides, cancer therapeutic agents induced various morphological and biochemical abnormalities compared to the non-treated groups. Based on most of the obtained results, curcumin at nontoxic doses can protect the cardiac cells mainly through modulating antioxidant capacity, regulation of cell death, and anti-inflammatory effects. Nevertheless, according to a minority of findings, curcumin increases the susceptibility of the rat cardiomyoblast cell line (H9C2) to apoptosis triggered by doxorubicin. CONCLUSION According to most nonclinical studies, curcumin can have the potential of cardioprotective effects against cardiotoxicity induced by chemotherapy. However, based on limited, contradictory findings demonstrating the function of curcumin in potentiating doxorubicin-induced cardiotoxicity, well-designed studies are needed to evaluate the safety and effectiveness of treatment with new formulations of this compound during cancer therapy.
Collapse
Affiliation(s)
- Maryam Armandeh
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Bameri
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Tehran University of Medical Sciences, Tehran, Iran
| | - Mahedeh Samadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shima Heidari
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Roham Foroumad
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Moutabian H, Ghahramani-Asl R, Mortezazadeh T, Laripour R, Narmani A, Zamani H, Ataei G, Bagheri H, Farhood B, Sathyapalan T, Sahebkar A. The cardioprotective effects of nano-curcumin against doxorubicin-induced cardiotoxicity: A systematic review. Biofactors 2022; 48:597-610. [PMID: 35080781 DOI: 10.1002/biof.1823] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/04/2022] [Indexed: 12/18/2022]
Abstract
Although the chemotherapeutic drug, doxorubicin, is commonly used to treat various malignant tumors, its clinical use is restricted because of its toxicity especially cardiotoxicity. The use of curcumin may alleviate some of the doxorubicin-induced cardiotoxic effects. Especially, using the nano-formulation of curcumin can overcome the poor bioavailability of curcumin and enhance its physicochemical properties regarding its efficacy. In this study, we systematically reviewed the potential cardioprotective effects of nano-curcumin against the doxorubicin-induced cardiotoxicity. A systematic search was accomplished based on Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines for the identification of all relevant articles on "the role of nano-curcumin on doxorubicin-induced cardiotoxicity" in the electronic databases of Scopus, PubMed, and Web of Science up to July 2021. One hundred and sixty-nine articles were screened following a predefined set of inclusion and exclusion criteria. Ten eligible scientific papers were finally included in the present systematic review. The administration of doxorubicin reduced the body and heart weights of mice/rats compared to the control groups. In contrast, the combined treatment of doxorubicin and nano-curcumin increased the body and heart weights of animals compared with the doxorubicin-treated groups alone. Furthermore, doxorubicin could significantly induce the biochemical and histological changes in the cardiac tissue; however, coadministration of nano-curcumin formulation demonstrated a pattern opposite to the doxorubicin-induced changes. The coadministration of nano-curcumin alleviates the doxorubicin-induced cardiotoxicity through various mechanisms including antioxidant, anti-inflammatory, and antiapoptotic effects. Also, the cardioprotective effect of nano-curcumin formulation against doxorubicin-induced cardiotoxicity was higher than free curcumin.
Collapse
Affiliation(s)
- Hossein Moutabian
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Ruhollah Ghahramani-Asl
- Department of Medical Physics and Radiological Sciences, Faculty of Paramedicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Tohid Mortezazadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Reza Laripour
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Hamed Zamani
- Department of Medical Physics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Gholamreza Ataei
- Department of Radiology Technology, Faculty of Paramedical Sciences, Babol University of Medical Sciences, Babol, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Trauma Research Center, Kashan University of Medical Sciences, Kashan, Iran
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School (HYMS), The University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Mashhad, Iran
| |
Collapse
|
10
|
In Vitro and In Vivo Cardioprotective Effects of Curcumin against Doxorubicin-Induced Cardiotoxicity: A Systematic Review. JOURNAL OF ONCOLOGY 2022; 2022:7277562. [PMID: 35237323 PMCID: PMC8885194 DOI: 10.1155/2022/7277562] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022]
Abstract
Objective This study aimed to review the potential chemoprotective effects of curcumin against the doxorubicin-induced cardiotoxicity. Methods According to the PRISMA guideline, a comprehensive systematic search was performed in different electronic databases (Web of Science, PubMed, and Scopus) up to July 2021. One hundred and sixty-four studies were screened in accordance with a predefined set of inclusion and exclusion criteria. Eighteen eligible articles were finally included in the current systematic review. Results According to the in vitro and in vivo findings, it was found that doxorubicin administration leads to decreased cell survival, increased mortality, decreased bodyweight, heart weight, and heart to the bodyweight ratio compared to the control groups. However, curcumin cotreatment demonstrated an opposite pattern in comparison with the doxorubicin-treated groups alone. Other findings showed that doxorubicin significantly induces biochemical changes in the cardiac cells/tissue. Furthermore, the histological changes on the cardiac tissue were observed following doxorubicin treatment. Nevertheless, for most of the cases, these biochemical and histological changes mediated by doxorubicin were reversed near to control groups following curcumin coadministration. Conclusion It can be mentioned that coadministration of curcumin alleviates the doxorubicin-induced cardiotoxicity. Curcumin exerts these cardioprotective effects through different mechanisms of antioxidant, antiapoptosis, and anti-inflammatory. Since the finding presented in this systematic review are based on in vitro and in vivo studies, suggesting the use of curcumin in cancer patients as a cardioprotector agent against cardiotoxicity mediated by doxorubicin requires further clinical studies.
Collapse
|
11
|
Zhang J, Hu K, Di L, Wang P, Liu Z, Zhang J, Yue P, Song W, Zhang J, Chen T, Wang Z, Zhang Y, Wang X, Zhan C, Cheng YC, Li X, Li Q, Fan JY, Shen Y, Han JY, Qiao H. Traditional herbal medicine and nanomedicine: Converging disciplines to improve therapeutic efficacy and human health. Adv Drug Deliv Rev 2021; 178:113964. [PMID: 34499982 DOI: 10.1016/j.addr.2021.113964] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Traditional herbal medicine (THM), an ancient science, is a gift from nature. For thousands of years, it has helped humans fight diseases and protect life, health, and reproduction. Nanomedicine, a newer discipline has evolved from exploitation of the unique nanoscale morphology and is widely used in diagnosis, imaging, drug delivery, and other biomedical fields. Although THM and nanomedicine differ greatly in time span and discipline dimensions, they are closely related and are even evolving toward integration and convergence. This review begins with the history and latest research progress of THM and nanomedicine, expounding their respective developmental trajectory. It then discusses the overlapping connectivity and relevance of the two fields, including nanoaggregates generated in herbal medicine decoctions, the application of nanotechnology in the delivery and treatment of natural active ingredients, and the influence of physiological regulatory capability of THM on the in vivo fate of nanoparticles. Finally, future development trends, challenges, and research directions are discussed.
Collapse
|
12
|
Jin M, Kong L, Han Y, Zhang S. Gut microbiota enhances the chemosensitivity of hepatocellular carcinoma to 5-fluorouracil in vivo by increasing curcumin bioavailability. Phytother Res 2021; 35:5823-5837. [PMID: 34374130 DOI: 10.1002/ptr.7240] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 01/01/2023]
Abstract
5-Fluorouracil (5-Fu) is efficient for hepatocellular carcinoma (HCC) treatment, but fast-emerging resistance limits its usage. Curcumin is being investigated for its potential chemosensitivity, but its low oral bioavailability hinders its chemosensitivity effect in vivo. Gut microbiota modulation is considered to contribute to its bioactivities in vivo. In the current study, we demonstrate that curcumin can enhance 5-Fu chemosensitivity in HCC cells in vitro, increase the apoptosis rate, arrest the cell cycle at G2/M phase, and block the PI3k/AKT/mTOR signalling pathway by inhibiting the phosphorylation of PI3K and its downstream protein kinases. Curcumin also remarkably sensitized H22 cells to 5-Fu, allowing it to inhibit tumour growth in vivo. 16S rDNA sequencing suggests that curcumin in combination with 5-Fu significantly alters the gut microbiota composition based on alpha and beta diversity analysis compared to drug treatment alone. Gut microbiota depletion abolished curcumin's chemosensitivity effect in vivo. A pharmacodynamics study suggested that the gut microbiota increased the oral bioavailability of curcumin (AUC(0-t) 15.24 ± 0.77 μM/h [wt] vs. 3.04 ± 0.18 μM/h [gut microbiota depleted]). In conclusion, curcumin can increase the chemosensitivity of HCC to 5-Fu in vitro and in vivo, and gut microbiota plays a key role in its effect in vivo.
Collapse
Affiliation(s)
- Meng Jin
- Department of Traditional Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Kong
- Department of Chinese and Western Medicine Combined with Liver Disease, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Han
- Department of Traditional Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Sen Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, China
| |
Collapse
|
13
|
Wang C, Yang H, Li J. Combination of Microwave, Ultrasonic, Enzyme Assisted Method for Curcumin Species Extraction from Turmeric (Curcuma Longa L.) and Evaluation of their Antioxidant Activity. EFOOD 2021. [DOI: 10.2991/efood.k.210329.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
14
|
Shi J, Li J, Li J, Li R, Wu X, Gao F, Zou L, Mak WWS, Fu C, Zhang J, Leung GPH. Synergistic breast cancer suppression efficacy of doxorubicin by combination with glycyrrhetinic acid as an angiogenesis inhibitor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153408. [PMID: 33234363 DOI: 10.1016/j.phymed.2020.153408] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/13/2020] [Accepted: 11/03/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Therapeutic regimens of breast cancer treatment are increasingly inclined to adopt combination strategy based on the broad spectrum antitumor effect of doxorubicin (Dox). Currently, combination therapy comprises of conventional anti-cancer drugs and angiogenesis inhibitors have been corroborated as an effective approach in cancer treatment. PURPOSE We explored the ability of a natural anti-angiogenic compound glycyrrhetinic acid (GA), derived from an edible-medicinal herb licorice, to enhance the breast cancer suppression effect of Dox. STUDY DESIGN The drug ratio of GA and Dox with synergistic anticancer effect against MCF-7 cells was optimized by combination index (CI) value in vitro, followed by evaluation of the improved anticancer effects and reduced side-effects of this combination in vitro and in vivo. METHODS Cell viability was measured by MTT assay. Analyses of mitochondrial membrane potential and cell apoptosis on MCF-7 cells were performed by JC-1 dye and Annexin V-FITC/PI assays. The cellular accumulation of Dox when combined with GA was evaluated. Levels of apoptosis-related proteins in MCF-7 cells were measured by Western blot analysis. Synergistic anti-angiogenic effects on HUVECs were evaluated. A breast cancer mouse model was established to investigate the anti-tumor effects in vivo. RESULTS Based on the optimization by CI value, Dox and GA at 1:20 molar ratio was chosen as the optimal combination drug ratio that exhibited synergistic effect against MCF-7 breast cancer cells. In addition, the combination of GA and Dox exhibited significantly enhanced cytotoxicity, apoptosis, and loss of mitochondrial membrane potential via the upregulation of a mitochondrial-dependent apoptosis pathway against MCF-7 cells. Interestingly, the addition of GA increased the intracellular accumulation of Dox in MCF-7 cells. Moreover, VEGF-induced HUVECs proliferation, migration, and tube formation were strongly inhibited by Dox when used with GA via the significant down-regulation of VEGFR2-mediated pathway, indicating that the combination of Dox and GA could exhibit ideal synergistic anti-angiogenesis effect. Expectedly, the enhanced anti-tumor efficacy of Dox and reduced Dox-induced cardiotoxicity when used in combination with GA were evident in a mouse breast tumor model. CONCLUSIONS These findings support that the combination of Dox with GA is a novel and promising therapeutic strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jinfeng Shi
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Jingjing Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region 999077, China.
| | - Jiaxin Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region 999077, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region 999077, China
| | - Fei Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Liang Zou
- School of Medicine, Chengdu University, Chengdu, Sichuan 610106, China
| | - Winston Wing Shum Mak
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region 999077, China
| | - Chaomei Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Jinming Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region 999077, China.
| |
Collapse
|
15
|
Huyut Z, Alp HH, Yaman T, Keleş ÖF, Yener Z, Türkan F, Ayengin K. Comparison of the protective effects of curcumin and caffeic acid phenethyl ester against doxorubicin-induced testicular toxicity. Andrologia 2020; 53:e13919. [PMID: 33289171 DOI: 10.1111/and.13919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/27/2020] [Accepted: 11/08/2020] [Indexed: 12/25/2022] Open
Abstract
Whether testicular toxicity is mediated by matrix metalloproteinases (MMPs) is an important question that has not been examined. This study investigated the suppressive effect of curcumin and caffeic acid phenethyl ester (CAPE) on oxidative stress, apoptosis, and whether MMPs mediate doxorubicin (DOX)-induced testicular injury. Male rats were randomly divided into eight groups (n = 8 per group). The groups were as follows: sham, dimethyl sulphoxide (100 µL), DOX (3 mg/kg), CAPE (2.68 mg/kg), curcumin (30 mg/kg), DOX+CAPE (3 mg/kg DOX and 2.68 mg/kg CAPE), DOX+curcumin (3 mg/kg DOX and 30 mg/kg curcumin) and DOX+CAPE+curcumin (3 mg/kg DOX, 2.68 mg/kg CAPE and 30 mg/kg curcumin). Injections were administered daily for 21 days. The oxidative stress, MMPs, proinflammatory cytokines and apoptotic markers in the DOX group were higher than the sham group (p < .05); these measures were lower in the groups treated with CAPE and curcumin together with DOX compared with the DOX group (p < .05). The results showed that MMPs mediated DOX-induced testicular injury, but CAPE and especially curcumin suppressed testis injury and cell apoptosis by suppressing DOX-induced increases in MMPs, oxidative stress and proinflammatory cytokines. However, curcumin exhibited more pronounced effects than CAPE in terms of all studied parameters.
Collapse
Affiliation(s)
- Zübeyir Huyut
- Medical Faculty, Department of Biochemistry, Van Yuzuncu Yıl University, Van, Turkey
| | - Hamit Hakan Alp
- Medical Faculty, Department of Biochemistry, Van Yuzuncu Yıl University, Van, Turkey
| | - Turan Yaman
- Faculty of Veterinary, Department of Pathology, Van Yuzuncu Yıl University, Van, Turkey
| | - Ömer Faruk Keleş
- Faculty of Veterinary, Department of Pathology, Van Yuzuncu Yıl University, Van, Turkey
| | - Zabit Yener
- Faculty of Veterinary, Department of Pathology, Van Yuzuncu Yıl University, Van, Turkey
| | - Fikret Türkan
- Health Services Vocational School, Igdır University, Igdır, Turkey
| | - Kemal Ayengin
- Medical Faculty, Department of Pediatric Surgery, Van Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
16
|
Polychemotherapy with Curcumin and Doxorubicin via Biological Nanoplatforms: Enhancing Antitumor Activity. Pharmaceutics 2020; 12:pharmaceutics12111084. [PMID: 33187385 PMCID: PMC7697177 DOI: 10.3390/pharmaceutics12111084] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/31/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is a well-known chemotherapeutic agent extensively applied in the field of cancer therapy. However, similar to other chemotherapeutic agents such as cisplatin, paclitaxel, docetaxel, etoposide and oxaliplatin, cancer cells are able to obtain chemoresistance that limits DOX efficacy. In respect to dose-dependent side effect of DOX, enhancing its dosage is not recommended for effective cancer chemotherapy. Therefore, different strategies have been considered for reversing DOX resistance and diminishing its side effects. Phytochemical are potential candidates in this case due to their great pharmacological activities. Curcumin is a potential antitumor phytochemical isolated from Curcuma longa with capacity of suppressing cancer metastasis and proliferation and affecting molecular pathways. Experiments have demonstrated the potential of curcumin for inhibiting chemoresistance by downregulating oncogene pathways such as MMP-2, TGF-β, EMT, PI3K/Akt, NF-κB and AP-1. Furthermore, coadministration of curcumin and DOX potentiates apoptosis induction in cancer cells. In light of this, nanoplatforms have been employed for codelivery of curcumin and DOX. This results in promoting the bioavailability and internalization of the aforementioned active compounds in cancer cells and, consequently, enhancing their antitumor activity. Noteworthy, curcumin has been applied for reducing adverse effects of DOX on normal cells and tissues via reducing inflammation, oxidative stress and apoptosis. The current review highlights the anticancer mechanism, side effects and codelivery of curcumin and DOX via nanovehicles.
Collapse
|
17
|
Sirangelo I, Sapio L, Ragone A, Naviglio S, Iannuzzi C, Barone D, Giordano A, Borriello M. Vanillin Prevents Doxorubicin-Induced Apoptosis and Oxidative Stress in Rat H9c2 Cardiomyocytes. Nutrients 2020; 12:2317. [PMID: 32752227 PMCID: PMC7468857 DOI: 10.3390/nu12082317] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (doxo) is an effective anticancer compound in several tumor types. However, as a consequence of oxidative stress induction and ROS overproduction, its high cardiotoxicity demands urgent attention. Vanillin possesses antioxidant, antiproliferative, antidepressant and anti-glycating properties. Therefore, we investigated the potential vanillin protective effects against doxo-induced cardiotoxicity in H9c2 cells. Using multiparametric approach, we demonstrated that vanillin restored both cell viability and damage in response to doxo exposure. Contextually, vanillin decreased sub-G1 appearance and caspase-3 and PARP1 activation, reducing the doxo-related apoptosis induction. From a mechanistic point of view, vanillin hindered doxo-induced ROS accumulation and impaired the ERK phosphorylation. Notably, besides the cardioprotective effects, vanillin did not counteract the doxo effectiveness in osteosarcoma cells. Taken together, our results suggest that vanillin ameliorates doxo-induced toxicity in H9c2 cells, opening new avenues for developing alternative therapeutic approaches to prevent the anthracycline-related cardiotoxicity and to improve the long-term outcome of antineoplastic treatment.
Collapse
Affiliation(s)
- Ivana Sirangelo
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.R.); (S.N.); (C.I.); (M.B.)
| | - Luigi Sapio
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.R.); (S.N.); (C.I.); (M.B.)
| | - Angela Ragone
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.R.); (S.N.); (C.I.); (M.B.)
| | - Silvio Naviglio
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.R.); (S.N.); (C.I.); (M.B.)
| | - Clara Iannuzzi
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.R.); (S.N.); (C.I.); (M.B.)
| | - Daniela Barone
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Department of Medical Biotechnology, University of Siena, 53100 Siena, Italy
| | - Margherita Borriello
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.R.); (S.N.); (C.I.); (M.B.)
| |
Collapse
|
18
|
Sesarman A, Muntean D, Abrudan B, Tefas L, Sylvester B, Licarete E, Rauca V, Luput L, Patras L, Banciu M, Vlase L, Porfire A. Improved pharmacokinetics and reduced side effects of doxorubicin therapy by liposomal co-encapsulation with curcumin. J Liposome Res 2019; 31:1-10. [PMID: 31631726 DOI: 10.1080/08982104.2019.1682604] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The goal of the current study was to investigate the pharmacokinetic profile, tissue distribution and adverse effects of long-circulating liposomes (LCL) with curcumin (CURC) and doxorubicin (DOX), in order to provide further evidence for previously demonstrated enhanced antitumor efficacy in colon cancer models. The pharmacokinetic studies were carried out in healthy rats, following the i.v. injection of a single dose of LCL-CURC-DOX (1 mg/kg DOX). For the tissue distribution study, DOX concentration in tumours, heart and liver were measured after the administration of two i.v. doses of LCL-CURC-DOX (2.5 mg/kg DOX and 5 mg/kg CURC) to Balb/c mice bearing C26 colon tumours. Markers of murine cardiac and hepatic oxidative status were determined to provide additional insights into the benefit of co-encapsulating CURC and DOX in LCL over DOX-induced adverse effects in these organs. The current study demonstrated that the liposomal association of CURC and DOX effectively improved the pharmacokinetics and biodistribution of DOX, limiting its side effects, via CURC-dependent antioxidant effects.
Collapse
Affiliation(s)
- Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dana Muntean
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bianca Abrudan
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucia Tefas
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bianca Sylvester
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Valentin Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Laurian Vlase
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
19
|
Negrette-Guzmán M. Combinations of the antioxidants sulforaphane or curcumin and the conventional antineoplastics cisplatin or doxorubicin as prospects for anticancer chemotherapy. Eur J Pharmacol 2019; 859:172513. [PMID: 31260654 DOI: 10.1016/j.ejphar.2019.172513] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
Drugs used in clinical oncology have narrow therapeutic indices with adverse toxicity often involving oxidative damage. Chemoresistance to these conventional antineoplastics is usually mediated by oxidative stress-upregulated pathways such as those of nuclear factor-kappa B (NF-κB) and hypoxia-inducible factor-1 alpha (HIF-1α). Accordingly, the use of antioxidants in combinational approaches has begun to be considered for fighting cancer because of both the protective role against adverse effects and the ability to sensitize chemoresistant cancer cells. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been identified as a mediator of the cytoprotection but it is not regularly associated with tumor chemosensitization. However, some Nrf2 inducers could be exerting cytoprotective and chemosensitizing roles through a simple integrated mechanism in which the cellular level of reactive oxygen species is controlled, thus inhibiting the oxidative damage in non-target tissues and the tumor chemoresistance mediated by NF-κB or HIF-1α. As examples to show the general idea of this antioxidant combination chemotherapy, this review explores the preclinical information available for four combinations, each composed by a paradigmatic oncological drug (cisplatin or doxorubicin) and a recognized antioxidant (sulforaphane or curcumin). The issues for translating these outcomes to clinical trials are briefly discussed.
Collapse
Affiliation(s)
- Mario Negrette-Guzmán
- Centro de Investigaciones en Enfermedades Tropicales (CINTROP), Departamento de Ciencias Básicas, Escuela de Medicina, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, 68002, Colombia.
| |
Collapse
|
20
|
Muller AG, Sarker SD, Saleem IY, Hutcheon GA. Delivery of natural phenolic compounds for the potential treatment of lung cancer. Daru 2019; 27:433-449. [PMID: 31115871 PMCID: PMC6593021 DOI: 10.1007/s40199-019-00267-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
The application of natural products to treat various diseases, such as cancer, has been an important area of research for many years. Several phytochemicals have demonstrated anticarcinogenic activity to prevent or reduce the progression of cancer by modulating various cellular mechanisms. However, poor bioavailability has hindered clinical success and the incorporation of these drugs into efficient drug delivery systems would be beneficial. For lung cancer, local delivery via the pulmonary route would also be more effective. In this article, recent in vitro scientific literature on phenolic compounds with anticancer activity towards lung cancer cell lines is reviewed and nanoparticulate delivery is mentioned as a possible solution to the problem of bioavailability. The first part of the review will explore the different classes of natural phenolic compounds and discuss recent reports on their activity on lung cancer cells. Then, the problem of the poor bioavailability of phenolic compounds will be explored, followed by a summary of recent advances in improving the efficacy of these phenolic compounds using nanoparticulate drug delivery systems. Graphical abstract The rationale for direct delivery of phenolic compounds loaded in microparticles to the lungs.
Collapse
Affiliation(s)
- Ashley G Muller
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, 3 Byrom Street, Liverpool, L3 3AF, UK.
| | - Satyajit D Sarker
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, 3 Byrom Street, Liverpool, L3 3AF, UK
| | - Imran Y Saleem
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, 3 Byrom Street, Liverpool, L3 3AF, UK
| | - Gillian A Hutcheon
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, 3 Byrom Street, Liverpool, L3 3AF, UK
| |
Collapse
|
21
|
Cao H, Duan Y, Lin Q, Yang Y, Gong Z, Zhong Y, Chen X, Shao Z. Dual-loaded, long-term sustained drug releasing and thixotropic hydrogel for localized chemotherapy of cancer. Biomater Sci 2019; 7:2975-2985. [DOI: 10.1039/c9bm00540d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A thixotropic injectable regenerated silk fibroin/hydroxypropylcellulose (RSF/HPC) hydrogel for highly sustainable dual-drug release with improved anticancer therapy and alleviated side effects.
Collapse
Affiliation(s)
- Han Cao
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Laboratory of Advanced Materials
- Fudan University
- Shanghai
| | - Yu Duan
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Laboratory of Advanced Materials
- Fudan University
- Shanghai
| | - Qinrui Lin
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Laboratory of Advanced Materials
- Fudan University
- Shanghai
| | - Yuhong Yang
- Research Center for Analysis and Measurement
- Fudan University
- Shanghai
- People's Republic of China
| | - Zuguang Gong
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Laboratory of Advanced Materials
- Fudan University
- Shanghai
| | - Yiming Zhong
- Fuels and Energy Technology Institute & Department of Chemical Engineering
- Curtin University
- Perth
- Australia
| | - Xin Chen
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Laboratory of Advanced Materials
- Fudan University
- Shanghai
| | - Zhengzhong Shao
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Laboratory of Advanced Materials
- Fudan University
- Shanghai
| |
Collapse
|
22
|
Polyethylene glycol–gum acacia-based multidrug delivery system for controlled delivery of anticancer drugs. Polym Bull (Berl) 2018. [DOI: 10.1007/s00289-018-2642-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Jain A, Rani V. Assessment of herb-drug synergy to combat doxorubicin induced cardiotoxicity. Life Sci 2018; 205:97-106. [DOI: 10.1016/j.lfs.2018.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/05/2018] [Accepted: 05/08/2018] [Indexed: 01/25/2023]
|
24
|
Cheng YY, Hsieh CH, Tsai TH. Concurrent administration of anticancer chemotherapy drug and herbal medicine on the perspective of pharmacokinetics. J Food Drug Anal 2018; 26:S88-S95. [PMID: 29703390 PMCID: PMC9326883 DOI: 10.1016/j.jfda.2018.01.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/13/2018] [Accepted: 01/20/2018] [Indexed: 02/06/2023] Open
Abstract
With an increasing number of cancer patients seeking an improved quality of life, complementary and alternative therapies are becoming more common ways to achieve such improvements. The potential risks of concurrent administration are serious and must be addressed. However, comprehensive evidence for the risks and benefits of combining anticancer drugs with traditional herbs is rare. Pharmacokinetic investigations are an efficient way to understand the influence of concomitant remedies. Therefore, this study aimed to collect the results of pharmacokinetic studies relating to the concurrent use of cancer chemotherapy and complementary and alternative therapies. According to the National Health Insurance (NHI) database in Taiwan and several publications, the three most commonly prescribed formulations for cancer patients are Xiang-Sha-Liu-Jun-Zi-Tang, Jia-Wei-Xiao-Yao-San and Bu-Zhong-Yi-Qi-Tang. The three most commonly prescribed single herbs for cancer patients are Hedyotis diffusa, Scutellaria barbata, and Astragalus membranaceus. Few studies have discussed herb-drug interactions involving these herbs from a pharmacokinetics perspective. Here, we reviewed Jia-Wei-Xiao-Yao-San, Long-Dan-Xie-Gan-Tang, Curcuma longa and milk thistle to provide information based on pharmacokinetic evidence for healthcare professionals to use in educating patients about the risks of the concomitant use of various remedies.
Collapse
Affiliation(s)
- Yung-Yi Cheng
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chen-Hsi Hsieh
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, 112, Taiwan; Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, 112, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung, 404, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Department of Chemical Engineering, National United University, Miaoli, 36063, Taiwan.
| |
Collapse
|
25
|
Fan N, He Z, Ma P, Wang X, Li C, Sun J, Sun Y, Li J. Impact of HPMC on inhibiting crystallization and improving permeability of curcumin amorphous solid dispersions. Carbohydr Polym 2018; 181:543-550. [DOI: 10.1016/j.carbpol.2017.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/22/2017] [Accepted: 12/04/2017] [Indexed: 01/30/2023]
|
26
|
Mohajeri M, Sahebkar A. Protective effects of curcumin against doxorubicin-induced toxicity and resistance: A review. Crit Rev Oncol Hematol 2017; 122:30-51. [PMID: 29458788 DOI: 10.1016/j.critrevonc.2017.12.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/28/2017] [Accepted: 12/11/2017] [Indexed: 02/08/2023] Open
Abstract
Doxorubicin (DOX)-induced toxicity and resistance are major obstacles in chemotherapeutic approaches. Despite effective in the treatment of numerous malignancies, some clinicians have voiced concern that DOX has the potential to cause debilitating consequences in organ tissues, especially the heart. The mechanisms of toxicity and resistance are respectively related to induction of reactive oxygen species (ROS) and up-regulation of ATP-binding cassette (ABC) transporter. Curcumin (CUR) with several biological and pharmacological properties is expected to restore DOX-mediated impairments to tissues. This review is intended to address the current knowledge on DOX adverse effects and CUR protective actions in the heart, kidneys, liver, brain, and reproductive organs. Coadministration of CUR and DOX is capable of ameliorating DOX toxicity pertained to antioxidant, apoptosis, autophagy, and mitochondrial permeability.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Department of Medical Biotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Chen S, Nimick M, Cridge AG, Hawkins BC, Rosengren RJ. Anticancer potential of novel curcumin analogs towards castrate-resistant prostate cancer. Int J Oncol 2017; 52:579-588. [PMID: 29207190 DOI: 10.3892/ijo.2017.4207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/14/2017] [Indexed: 11/05/2022] Open
Abstract
Prostate cancer is initially sensitive to hormone therapy; however, over time the majority of patients progress to a hormone-insensitive form classified as castration-resistant prostate cancer (CRPC). CRPC is highly metastatic and patients have a poor prognosis. Thus, new drugs for the treatment of this disease are required. In this study, we therefore examined the cytotoxic effects and anticancer mechanism(s) of action of second generation curcumin analogs towards CRPC cells. For this purpose, PC3 and DU145 cells were treated with a series of curcumin analogs at 0-10 µM for 72 h and cytotoxicity was determined by the sulforhodamine B (SRB) assay. Two compounds, 1-isopropyl-3,5-bis(pyridin-3-ylmethylene)-4-piperidone (RL118) and 1-methyl-3,5-[(6-methoxynaphthalen-2-yl)methylene]-4-piperidone (RL121), were found to have the most potent cytotoxic effect with EC50 values of 0.50 and 0.58 µM in the PC3 cells and EC50 values of 0.76 and 0.69 µM in the DU145 cells, respectively. Thus, further experiments were performed focusing on these two compounds. Flow cytometry was performed to determine their effects on the cell cycle and apoptosis. Both analogs increased the number of cells in the G2/M phase of the cell cycle and induced apoptosis. Specifically, in the PC3 cells, RL121 increased the number of cells in the G2/M phase by 86% compared to the control, while RL118 increased the number of cells in the G2/M phase by 42% compared to the control after 24 h. Moreover, both RL118 and RL121 induced the apoptosis of both cell lines. In the DU145 cells, a 38-fold increase in the number of apoptotic cells was elicited by RL118 and a 78-fold increase by RL121 compared to the control. Furthermore, the effects of both analogs on the expression of key proteins involved in cell proliferation were also determined by western blot analysis. The results revealed that both analogs inhibited the expression of nuclear factor (NF)-κB (p65/RelA), eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), p-4E-BP1, mammalian target of rapamycin (mTOR), p-mTOR, AKT and p-AKT. Thus, the findings of this study provide evidence that RL118 and RL121 have potent anticancer activity against CPRC cells, and both analogs warrant further investigation in vivo.
Collapse
Affiliation(s)
- Shuli Chen
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Mhairi Nimick
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Andrew G Cridge
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand
| | - Bill C Hawkins
- Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
28
|
Jain A, Rani V. Mode of treatment governs curcumin response on doxorubicin-induced toxicity in cardiomyoblasts. Mol Cell Biochem 2017; 442:81-96. [DOI: 10.1007/s11010-017-3195-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/09/2017] [Indexed: 01/01/2023]
|
29
|
Zhang J, Li J, Shi Z, Yang Y, Xie X, Lee SM, Wang Y, Leong KW, Chen M. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater 2017; 58:349-364. [PMID: 28455219 DOI: 10.1016/j.actbio.2017.04.029] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/22/2017] [Accepted: 04/24/2017] [Indexed: 01/24/2023]
Abstract
Co-delivery of multiple drugs with complementary anticancer mechanisms by nano-carriers offers an effective strategy to treat cancer. The combination of drugs with pro-apoptotic and anti-angiogenic activities is potentially effective in treating human hepatocellular carcinoma (HCC). Herein, we developed a co-delivery system for doxorubicin (Dox), a pro-apoptotic drug, and curcumin (Cur), a potent drug for antiangiogenesis, in pH-sensitive nanoparticles (NPs) constituted with amphiphilic poly(β-amino ester) copolymer. Dox & Cur co-loaded NPs ((D+C)/NPs) were prepared with optimized drug ratio, showing low polydispersity, high encapsulation efficiency, and enhanced release in the acidic environment of cancer cells. Furthermore, enhanced cellular internalization of cargoes delivered from (D+C)/NPs were observed in human liver cancer SMMC 7721 cells and human umbilical vein endothelial cells (HUVECs) compared to the use of free drugs. The (D+C)/NPs induced a high rate of apoptosis in SMMC 7721 cells through decreased mitochondrial membrane potential. Additionally, (D+C)/NPs exhibited stronger anti-angiogenic effects including inhibition of HUVEC proliferation, migration, invasion, and tube formation mediated VEGF pathway modulation in vitro and in vivo. Taken together, encapsulation of the pro-apoptotic drug Dox and antiangiogenic agent Cur in pH-sensitive NPs provides a promising strategy to effectively inhibit HCC progression in a synergistic manner. STATEMENT OF SIGNIFICANCE The combination of multiple drugs has been demonstrated to be more effective than single treatment. However, the different physicochemical and pharmacokinetic profiles of each drug render optimal delivery challenging. In view of the great delivery advantage of nanocarriers to unify the multiple drugs in vivo, stimulus-responsive nano-carriers are more crucial to increase efficacy and reduce toxicity from off-target exposure. Therefore, herein the pH-sensitive nanoparticles, composed by d-α-tocopheryl polyethylene glycol 1000-block-poly (β-amino ester) (TPGS-PAE) polymers, have been fabricated for doxorubicin (Dox) and curcumin (Cur) co-delivery, which exhibited diverse anticancer approaches, i.e. pro-apoptosis and antiangiogenesis. The precise intracellular target site and effective drug combination concentration result in the enhanced antitumor efficiency and the reduced systematic toxicity of Dox. The co-encapsulation of the pro-apoptotic drug and antiangiogenic agent in pH-sensitive NPs provides a promising strategy to effectively inhibit malignant neoplasm progression in a synergistic manner.
Collapse
|
30
|
Tefas LR, Sylvester B, Tomuta I, Sesarman A, Licarete E, Banciu M, Porfire A. Development of antiproliferative long-circulating liposomes co-encapsulating doxorubicin and curcumin, through the use of a quality-by-design approach. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1605-1621. [PMID: 28579758 PMCID: PMC5448697 DOI: 10.2147/dddt.s129008] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this work was to use the quality-by-design (QbD) approach in the development of long-circulating liposomes co-loaded with curcumin (CUR) and doxorubicin (DOX) and to evaluate the cytotoxic potential of these liposomes in vitro using C26 murine colon carcinoma cell line. Based on a risk assessment, six parameters, namely the phospholipid, CUR and DOX concentrations, the phospholipid:cholesterol molar ratio, the temperature during the evaporation and hydration steps and the pH of the phosphate buffer, were identified as potential risk factors for the quality of the final product. The influence of these variables on the critical quality attributes of the co-loaded liposomal CUR and DOX was investigated: particle size, zeta potential, drug loading and entrapment efficiency. For this, a 26−2 factorial design was employed to establish a proper regression model and to generate the contour plots for the responses. The obtained data served to establish the design space for which different combinations of variables yielded liposomes with characteristics within predefined specifications. The validation of the model was carried out by preparing two liposomal formulations corresponding to the robust set point from within the design space and one outside the design space and calculating the percentage bias between the predicted and actual experimental results. The in vitro antiproliferative test showed that at higher CUR concentrations, the liposomes co-encapsulating CUR and DOX had a greater cytotoxic effect than DOX-loaded liposomes. Overall, this study showed that QbD is a useful instrument for controlling and optimizing the manufacturing process of liposomes co-loaded with CUR and DOX and that this nanoparticulate system possesses a great potential for use in colon cancer therapy.
Collapse
Affiliation(s)
- Lucia Ruxandra Tefas
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Haţieganu"
| | - Bianca Sylvester
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Haţieganu"
| | - Ioan Tomuta
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Haţieganu"
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babeş-Bolyai University, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babeş-Bolyai University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babeş-Bolyai University, Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Haţieganu"
| |
Collapse
|
31
|
Peng J, Fumoto S, Miyamoto H, Chen Y, Kuroda N, Nishida K. One-step formation of lipid-polyacrylic acid-calcium carbonate nanoparticles for co-delivery of doxorubicin and curcumin. J Drug Target 2017; 25:704-714. [PMID: 28368667 DOI: 10.1080/1061186x.2017.1315687] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
A doxorubicin (Dox) and curcumin (Cur) combination treatment regimen has been widely studied in pre-clinical research. However, the nanoparticles developed for this combination therapy require a consecutive drug loading process because of the different water-solubility of these drugs. This study provides a strategy for the "one-step" formation of nanoparticles encapsulating both Dox and Cur. We took advantage of polyacrylic acid (PAA) and calcium carbonate (CaCO3) to realise a high drug entrapment efficiency (EE) and pH-sensitive drug release using a simplified preparation method. Optimisation of lipid ratios and concentrations of CaCO3 was conducted. Under optimal conditions, the mean diameter of PEGylated lipid/PAA/CaCO3 nanoparticles with encapsulated Cur and Dox (LPCCD) was less than 100 nm. An obvious pH-sensitive release of both drugs was observed, with different Dox and Cur release rates. Successful co-delivery of Cur and Dox was achieved via LPCCD on HepG2 cells. LPCCD altered the bio-distribution of Dox and Cur in vivo and decreased Dox-induced cardiotoxicity. The current investigation has developed an efficient ternary system for co-delivery of Dox and Cur to tumours, using a "one-step" formation resulting in nanoparticles possessing remarkable pH-sensitive drug release behaviour, which may be valuable for further clinical studies and eventual clinical application.
Collapse
Affiliation(s)
- Jianqing Peng
- a Graduate School of Biomedical Sciences , Nagasaki University , Nagasaki , Japan
| | - Shintaro Fumoto
- a Graduate School of Biomedical Sciences , Nagasaki University , Nagasaki , Japan
| | - Hirotaka Miyamoto
- a Graduate School of Biomedical Sciences , Nagasaki University , Nagasaki , Japan
| | - Yi Chen
- b Department of Pharmaceutics , China Pharmaceutical University , Nanjing , PR China
| | - Naotaka Kuroda
- a Graduate School of Biomedical Sciences , Nagasaki University , Nagasaki , Japan
| | - Koyo Nishida
- a Graduate School of Biomedical Sciences , Nagasaki University , Nagasaki , Japan
| |
Collapse
|
32
|
Su CL, Tseng CL, Ramesh C, Liu HS, Huang CYF, Yao CF. Using gene expression database to uncover biology functions of 1,4-disubstituted 1,2,3-triazole analogues synthesized via a copper (I)-catalyzed reaction. Eur J Med Chem 2017; 132:90-107. [PMID: 28342400 DOI: 10.1016/j.ejmech.2017.03.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 12/18/2022]
Abstract
We have synthesized bioactive 1,4-disubstituted 1,2,3-triazole analogues containing 2H-1,4-benzoxazin-3-(4H)-one derivatives via 1,3-dipolar cycloaddition in the presence of CuI. All the reactions proceeded smoothly and afforded its desired products in excellent yields. Among these analogues, 3y exhibited a better cytotoxic effect on human hepatocellular carcinoma (HCC) Hep 3B cells and displayed less cytotoxicity on normal human umbilical vein endothelial cells, compared with Sorafenib, a targeted therapy for advanced HCC. 3y also induced stronger apoptosis and autophagy. Addition of curcumin enhanced 3y-induced cytotoxicity by further induction of autophagy. Using gene expression signatures of 3y to query Connectivity Map, a glycogen synthase kinase-3 inhibitor (AR-A014418) was predicted to display similar molecular action of 3y. Experiments further demonstrate that AR-A014418 acted like 3y, and vice versa. Overall, our data suggest the chemotherapeutic potential of 3y on HCC.
Collapse
Affiliation(s)
- Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan.
| | - Chia-Ling Tseng
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan
| | - Chintakunta Ramesh
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Center of Infectious Disease and Signaling Research Center, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan; Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan.
| |
Collapse
|
33
|
Torquato HFV, Goettert MI, Justo GZ, Paredes-Gamero EJ. Anti-Cancer Phytometabolites Targeting Cancer Stem Cells. Curr Genomics 2017; 18:156-174. [PMID: 28367074 PMCID: PMC5345336 DOI: 10.2174/1389202917666160803162309] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 12/24/2015] [Accepted: 12/28/2015] [Indexed: 12/13/2022] Open
Abstract
Medicinal plants are a plentiful source of bioactive molecules with much structural diversity. In cancer treatment, molecules obtained from plants represent an attractive alternative to other treatments because several plant-derived compounds have exhibited lower toxicity and higher selectivity against cancer cells. In this review, we focus on the possible application of bioactive molecules obtained from plants against more primitive cell populations in cancers, cancer stem cells. Cancer stem cells are present in several kinds of tumors and are responsible for recurrences and metastases. Common anti-cancer drugs exhibit lower effectiveness against cancer stem cells because of their biological features. However, recently discovered natural phytometabolites exert cytotoxic effects on this rare population of cells in cancers. Therefore, this review presents the latest research on promising compounds from plants that can act as antitumor drugs and that mainly affect stem cell populations in cancers.
Collapse
Affiliation(s)
- Heron F V Torquato
- Departamento de Bioquímica, Universidade Federal de São Paulo (Campus São Paulo), São Paulo, Brazil
| | - Márcia I Goettert
- Programa de Pós-Graduação em Biotecnologia, Centro Universitário Univates, Rio Grande do Sul, Brazil
| | - Giselle Z Justo
- Departamento de Bioquímica, Universidade Federal de São Paulo (Campus São Paulo), São Paulo, Brazil;; Departamento de Ciências Biológicas (Campus Diadema), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Edgar J Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo (Campus São Paulo), São Paulo, Brazil;; Centro Interdisciplinar de Investigação Bioquímica, Universidade de Mogi das Cruzes, São Paulo, Brazil
| |
Collapse
|
34
|
Harati K, Behr B, Daigeler A, Hirsch T, Jacobsen F, Renner M, Harati A, Wallner C, Lehnhardt M, Becerikli M. Curcumin and Viscum album Extract Decrease Proliferation and Cell Viability of Soft-Tissue Sarcoma Cells: An In Vitro Analysis of Eight Cell Lines Using Real-Time Monitoring and Colorimetric Assays. Nutr Cancer 2017; 69:340-351. [PMID: 28045549 DOI: 10.1080/01635581.2017.1263349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The cytostatic effects of the polyphenol curcumin and Viscum album extract (VAE) were assessed in soft-tissue sarcoma (STS) cells. METHODS Eight human STS cell lines were used: fibrosarcoma (HT1080), liposarcoma (SW872, T778, MLS-402), synovial sarcoma (SW982, SYO1, 1273), and malignant fibrous histiocytoma (U2197). Primary human fibroblasts served as control cells. Cell proliferation, viability, and cell index (CI) were analyzed by BrdU assay, MTT assay, and real-time cell analysis (RTCA). RESULTS As indicated by BrdU and MTT, curcumin significantly decreased the cell proliferation of five cell lines (HT1080, SW872, SYO1, 1273, and U2197) and the viability of two cell lines (SW872 and SW982). VAE led to significant decreases of proliferation in eight cell lines (HT1080, SW872, T778, MLS-402, SW982, SYO1, 1293, and U2197) and reduced viability in seven STS lines (HT1080, SW872, T778, MLS-402, SW982, SYO1, and 1273). As indicated by RTCA for 160 h, curcumin decreased the CI of all synovial sarcoma cell lines as well as T778 and HT1080. VAE diminished the CI in most of the synovial sarcoma (SW982, SYO1) and liposarcoma (SW872, T778) cell lines as well as HT1080. Primary fibroblasts were not affected adversely by the two compounds in RTCA. CONCLUSION Curcumin and VAE can inhibit the proliferation and viability of STS cells.
Collapse
Affiliation(s)
- K Harati
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - B Behr
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - A Daigeler
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - T Hirsch
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - F Jacobsen
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - M Renner
- b Institute of Pathology, University of Heidelberg , Heidelberg , Germany
| | - A Harati
- c Department of Neurosurgery , Klinikum Dortmund , Dortmund , Germany
| | - C Wallner
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - M Lehnhardt
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - M Becerikli
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| |
Collapse
|
35
|
Sun X, Luo Y, Huang L, Yu BY, Tian J. A peptide-decorated and curcumin-loaded mesoporous silica nanomedicine for effectively overcoming multidrug resistance in cancer cells. RSC Adv 2017. [DOI: 10.1039/c7ra01128h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A robust peptide-functionalized mesoporous silica nanomedicine loading with curcumin and doxorubicin (DOX/CUR@MSN-Pep) has been successfully constructed to effectively overcome multidrug resistance in cancer cells.
Collapse
Affiliation(s)
- Xian Sun
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research
- Department of Complex Prescription of TCM
- China Pharmaceutical University
- Nanjing 211198
| | - Yingping Luo
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research
- Department of Complex Prescription of TCM
- China Pharmaceutical University
- Nanjing 211198
| | - Liwei Huang
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research
- Department of Complex Prescription of TCM
- China Pharmaceutical University
- Nanjing 211198
| | - Bo-Yang Yu
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research
- Department of Complex Prescription of TCM
- China Pharmaceutical University
- Nanjing 211198
| | - Jiangwei Tian
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research
- Department of Complex Prescription of TCM
- China Pharmaceutical University
- Nanjing 211198
| |
Collapse
|
36
|
Dash TK, Konkimalla VB. Comparative Study of Different Nano-Formulations of Curcumin for Reversal of Doxorubicin Resistance in K562R Cells. Pharm Res 2016; 34:279-289. [PMID: 27815791 DOI: 10.1007/s11095-016-2060-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE Curcumin is very well established as a chemo-therapeutic, chemo-preventive and chemo-sensitizing agent in diverse disease conditions. As the isolated pure form has poor solubility and pharmacokinetic problems, therefore it is encapsulated in to several nano-formulations to improve its bioavailability. Here in the current study, we aim to compare different nano-formulations of curcumin for their chemo-sensitizing activity in doxorubicin (DOX) resistant K562 cells. METHODS Four different curcumin formulations were prepared namely DMSO assisted curcumin nano-dispersion (CurD, 260 nm), liposomal curcumin (CurL, 165 nm), MPEG-PCL micellar curcumin (CurM, 18 nm) and cyclodextrin encapsulated curcumin (CurN, 37 nm). The formulations were subjected to particle characterizations (size, zeta potential, release studies), followed by biological assays such as cellular uptake, P-gp inhibitory activity and reversal of DOX resistance by co-treatment with DOX. RESULTS Curcumin uptake in K562N and K562R cells was mildly reduced when treated with CurL and CurM, while for CurD and CurN the uptake remained equivalent. However, CurL retained P-gp inhibitory activity of curcumin and with a considerable chemo-sensitizing effect but CurM showed no P-gp inhibitory activity. CurN retained above biological activities, but requires a secondary carrier under in vivo conditions. CONCLUSIONS From the results, CurM was found to be most suitable for solubilization of curcumin where as CurL can be considered as most suitable nano-formulation for reversal of DOX resistance.
Collapse
Affiliation(s)
- Tapan K Dash
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Via- Jatni, Khurda, 752050, India
| | - V Badireenath Konkimalla
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Via- Jatni, Khurda, 752050, India.
| |
Collapse
|
37
|
Klippstein R, Bansal SS, Al-Jamal KT. Doxorubicin enhances curcumin’s cytotoxicity in human prostate cancer cells in vitro by enhancing its cellular uptake. Int J Pharm 2016; 514:169-175. [DOI: 10.1016/j.ijpharm.2016.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 11/27/2022]
|
38
|
Chen S, Yang K, Tuguntaev RG, Mozhi A, Zhang J, Wang PC, Liang XJ. Targeting tumor microenvironment with PEG-based amphiphilic nanoparticles to overcome chemoresistance. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 12:269-86. [PMID: 26707818 DOI: 10.1016/j.nano.2015.10.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/24/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
UNLABELLED Multidrug resistance is one of the biggest obstacles in the treatment of cancer. Recent research studies highlight that tumor microenvironment plays a predominant role in tumor cell proliferation, metastasis, and drug resistance. Hence, targeting the tumor microenvironment provides a novel strategy for the evolution of cancer nanomedicine. The blooming knowledge about the tumor microenvironment merging with the design of PEG-based amphiphilic nanoparticles can provide an effective and promising platform to address the multidrug resistant tumor cells. This review describes the characteristic features of tumor microenvironment and their targeting mechanisms with the aid of PEG-based amphiphilic nanoparticles for the development of newer drug delivery systems to overcome multidrug resistance in cancer cells. FROM THE CLINICAL EDITOR Cancer is a leading cause of death worldwide. Many cancers develop multidrug resistance towards chemotherapeutic agents with time and strategies are urgently needed to combat against this. In this review article, the authors discuss the current capabilities of using nanomedicine to target the tumor microenvironments, which would provide new insight to the development of novel delivery systems for the future.
Collapse
Affiliation(s)
- Shizhu Chen
- Key Laboratory of Chemical Biology of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry & Environmental Science, Hebei University, Baoding, PR China
| | - Keni Yang
- CAS Key Lab of Nanomaterials Bioeffects and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, PR China
| | - Ruslan G Tuguntaev
- CAS Key Lab of Nanomaterials Bioeffects and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, PR China
| | - Anbu Mozhi
- CAS Key Lab of Nanomaterials Bioeffects and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, PR China
| | - Jinchao Zhang
- Key Laboratory of Chemical Biology of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry & Environmental Science, Hebei University, Baoding, PR China.
| | - Paul C Wang
- Fu Jen Catholic University, Taipei, Taiwan; Laboratory of Molecular Imaging, Department of Radiology, Howard University, WA, DC, USA
| | - Xing-Jie Liang
- CAS Key Lab of Nanomaterials Bioeffects and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, PR China.
| |
Collapse
|
39
|
Li H, Li M, Chen C, Fan A, Kong D, Wang Z, Zhao Y. On-demand combinational delivery of curcumin and doxorubicin via a pH-labile micellar nanocarrier. Int J Pharm 2015; 495:572-578. [DOI: 10.1016/j.ijpharm.2015.09.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/01/2015] [Accepted: 09/12/2015] [Indexed: 01/16/2023]
|
40
|
Synergistic effect of cucurbitacin B in combination with curcumin via enhancing apoptosis induction and reversing multidrug resistance in human hepatoma cells. Eur J Pharmacol 2015; 768:28-40. [PMID: 26452516 DOI: 10.1016/j.ejphar.2015.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
Cucurbitacin B is a plant-derived tetracyclic triterpenoid, which has been used for a variety of cancers, especially human hepatoma. Curcumin, isolated from a plant Curcuma longa also has found the anti-tumor property. In the present study, the synergistic effect of cucurbitacin B and curcumin was studied on BEL7402/5-Fu cells in vitro and BEL7402 tumor-bearing mice in vivo. The synergistic anticancer activity of these two compounds involves the two mechanisms. Firstly, curcumin synergistically enhanced the apoptosis of BEL7402/5-Fu cells induced by cucurbitacin B in the optimal mass ratio of 2:1 (cucurbitacin B:curcumin). The mechanism may result from the cell arresting in different phases of cell cycles and the apoptotic change of ultrastructure in BEL7402/5-Fu cells. Secondly, curcumin reversed the multidrug resistance (MDR) caused by cucurbitacin B in the optimized concentration of 67.9μM (25μg/ml). The mechanism was associated with the P-gp reduction, ΔΨm collapse and mitochondrial colocalization in BEL7402/5-Fu cells. The findings were consistent with the changes of the body weight and tumor volume, caspase3 activation and ATP down-regulation in vivo. In conclusion, cucurbitacin B in the combination with curcumin could serve as a novel, promising approach for human hepatoma.
Collapse
|
41
|
Pathak L, Kanwal A, Agrawal Y. Curcumin loaded self assembled lipid-biopolymer nanoparticles for functional food applications. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2015; 52:6143-56. [PMID: 26396362 PMCID: PMC4573133 DOI: 10.1007/s13197-015-1742-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 11/18/2014] [Accepted: 01/19/2015] [Indexed: 12/17/2022]
Abstract
The supramolecular nano-assemblies formed by electrostatic interactions of two oppositely charged lipid and polymer have been made and used as nanocarriers for curcumin to address its bioavailability and solubility issues. These curcumin encapsulated nano-supramolecular assemblies were characterized with respect to their size (dynamic light scattering), morphology (TEM, SEM), zeta potential (Laser Doppler Velocimetry), encapsulation efficiency (EE), curcumin loading (CL) etc. Stability of the nano-assemblies was assessed at different storage times as a function of varying pH and temperature. The physicochemical characterization of nano-assemblies was performed using Fourier Transform Infra Red Spectroscopy (FT-IR) and Differential Scanning Calorimetry (DSC). The in-vitro antioxidant lipid peroxidation (TBARS), radical scavenging (DPPH, NO, H2O2, reducing power) activity assays of powdered curcumin and nano-encapsulated curcumin were performed. It was found that nano-encapsulated curcumin were roughly spherical in shape, presented high positive zeta potential (>30 mV), monodisperse (polydispersity index <0.3), amorphous in nature, stable in the pH range of 2-6 and have enhanced antioxidant potency in comparison to crystalline curcumin in aqueous media. In conclusion, the curcumin encapsulated nanocarriers system has great potential as functional food ingredient of natural origin.
Collapse
Affiliation(s)
- Lokesh Pathak
- />Institute of Research and Development, Gujarat Forensics Sciences University, Gandhinagar, Gujarat 382 007 India
| | - Abhinav Kanwal
- />Medicinal Chemistry and Pharmacology Division, Indian Institute of Chemical Technology, Habsiguda, Hyderabad, 500 037 India
| | - Yadvendra Agrawal
- />Institute of Research and Development, Gujarat Forensics Sciences University, Gandhinagar, Gujarat 382 007 India
| |
Collapse
|
42
|
Cao H, Yang Y, Shao Z. Doxorubicin hydrochloride and curcumin loaded silk fibroin/hydroxypropylcellulose hydrogels for localized chemotherapy of cancer. J Control Release 2015; 213:e39. [DOI: 10.1016/j.jconrel.2015.05.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Sheu MT, Jhan HJ, Hsieh CM, Wang CJ, Ho HO. Efficacy of antioxidants as a Complementary and Alternative Medicine (CAM) in combination with the chemotherapeutic agent doxorubicin. Integr Cancer Ther 2014; 14:184-95. [PMID: 25542609 DOI: 10.1177/1534735414564425] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Although doxorubicin (Dox)-induced cardiac toxicity and pegylated liposomal doxorubicin (PLD)-induced hand-foot syndrome (HFS) were reported to be correlated with reactive oxygen species (ROS) generation, there is no effective preventive treatment at present. Therefore, the aim of this study was to investigate whether antioxidants-resveratrol (RSVL), tetrahydroxystilbene glucoside (THSG), curcumin, and the ethanolic extract of Antrodia cinnamomea (EEAC)-have the ability to reduce Dox-induced ROS and have a synergistic anticancer effect with Dox that could prevent those side effects and enhance the efficacy of cancer treatment. METHODS 3T3 normal cells were used as a model to evaluate the effects of these antioxidants in reducing ROS accumulation. Furthermore, the synergistic anticancer effect of antioxidants with Dox on the MCF-7 breast cancer model was also evaluated. RESULTS Pretreatment of cells with RSVL, curcumin, and EEAC increased the cell antioxidant ability by improving the activity of superoxide dismutase (SOD), prevented or limited intracellular damage, and ameliorated the harmful effects of ROS. Additionally, RSVL, curcumin, and EEAC had synergistic effects with Dox against MCF-7 breast cancer cells. CONCLUSION RSVL, curcumin, and EEAC have the potential to be clinically applied to prevent cardiac toxicity and HFS and enhance the anticancer efficiency of Dox.
Collapse
Affiliation(s)
| | | | | | | | - Hsiu-O Ho
- Taipei Medical University, Taipei, Taiwan, ROC
| |
Collapse
|
44
|
Curcumin is a tight-binding inhibitor of the most efficient human daunorubicin reductase--Carbonyl reductase 1. Chem Biol Interact 2014; 234:162-8. [PMID: 25541467 DOI: 10.1016/j.cbi.2014.12.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/02/2014] [Accepted: 12/14/2014] [Indexed: 11/21/2022]
Abstract
Curcumin is a major component of the plant Curcuma longa L. It is traditionally used as a spice and coloring in foods and is an important ingredient in curry. Curcuminoids have anti-oxidant and anti-inflammatory properties and gained increasing attention as potential neuroprotective and cancer preventive compounds. In the present study, we report that curcumin is a potent tight-binding inhibitor of human carbonyl reductase 1 (CBR1, Ki=223 nM). Curcumin acts as a non-competitive inhibitor with respect to the substrate 2,3-hexandione as revealed by plotting IC50-values against various substrate concentrations and most likely as a competitive inhibitor with respect to NADPH. Molecular modeling supports the finding that curcumin occupies the cofactor binding site of CBR1. Interestingly, CBR1 is one of the most effective human reductases in converting the anthracycline anti-tumor drug daunorubicin to daunorubicinol. The secondary alcohol metabolite daunorubicinol has significantly reduced anti-tumor activity and shows increased cardiotoxicity, thereby limiting the clinical use of daunorubicin. Thus, inhibition of CBR1 may increase the efficacy of daunorubicin in cancer tissue and simultaneously decrease its cardiotoxicity. Western-blots demonstrated basal expression of CBR1 in several cell lines. Significantly less daunorubicin reduction was detected after incubating A549 cell lysates with increasing concentrations of curcumin (up to 60% less with 50 μM curcumin), suggesting a beneficial effect in the co-treatment of anthracycline anti-tumor drugs together with curcumin.
Collapse
|
45
|
ZHAO FENG, GONG YUDIAN, HU YUAN, LU MINGHUI, WANG JING, DONG JIANXIN, CHEN DAQUAN, CHEN LEI, FU FENGHUA, QIU FENG. Curcumin and its major metabolites inhibit the inflammatory response induced by lipopolysaccharide: Translocation of nuclear factor-κB as potential target. Mol Med Rep 2014; 11:3087-93. [DOI: 10.3892/mmr.2014.3079] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 10/24/2014] [Indexed: 11/05/2022] Open
|
46
|
Fu Y, Gao X, Wan Y, Zhang J, Kong D, Wang Z, Zhao Y. Dendritic nanoconjugate containing optimum folic acid for targeted intracellular curcumin delivery. RSC Adv 2014. [DOI: 10.1039/c4ra08315f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
47
|
Tu Y, Sun D, Zeng X, Yao N, Huang X, Huang D, Chen Y. Piperine potentiates the hypocholesterolemic effect of curcumin in rats fed on a high fat diet. Exp Ther Med 2014; 8:260-266. [PMID: 24944632 PMCID: PMC4061201 DOI: 10.3892/etm.2014.1717] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 03/07/2014] [Indexed: 12/28/2022] Open
Abstract
It has previously been demonstrated that curcumin possesses a hypocholesterolemic effect and potentiates numerous pharmacological effects of curcumin, however, the mechanisms underlying this hypocholesterolemic effect and the interaction between curcumin and piperine remain to be elucidated. In the present study, male Sprague-Dawley rats were fed on a high-fat diet (HFD) to establish a hyperlipidemia (HLP) model. Co-administration of curcumin plus piperine was found to decrease the levels of total cholesterol (TC), triglyceride (TG) and low-density lipoprotein cholesterol in the serum and liver, as well as increase the levels of fecal TC, TG and total bile acid, compared with administration of curcumin alone. Curcumin plus piperine also markedly increased the levels of high-density lipoprotein cholesterol. Furthermore, compared with administration of curcumin alone, administration of curcumin plus piperine resulted in a significant upregulation of the activity and gene expression of apolipoprotein AI (ApoAI), lecithin cholesterol acyltransferase (LCAT), cholesterol 7α-hydroxylase (CYP7A1) and low-density lipoprotein receptor (LDLR). In conclusion, these results indicated that co-administration of curcumin plus piperine potentiates the hypocholesterolemic effects of curcumin by increasing the activity and gene expression of ApoAI, CYP7A1, LCAT and LDLR, providing a promising combination for the treatment of HLP.
Collapse
Affiliation(s)
- Yaosheng Tu
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| | - Dongmei Sun
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| | - Xiaohui Zeng
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| | - Nan Yao
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| | - Xuejun Huang
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| | - Dane Huang
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| | - Yuxing Chen
- Traditional Chinese Medicine Pharmacological Research Office, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, Guangdong 510095, P.R. China
| |
Collapse
|
48
|
|
49
|
Sun L, Deng X, Yang X, Li Z, Wang Z, Li L, Wu Q, Peng F, Liu L, Gong C. Co-delivery of doxorubicin and curcumin by polymeric micelles for improving antitumor efficacy on breast carcinoma. RSC Adv 2014. [DOI: 10.1039/c4ra07453j] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Polymeric micelles co-delivered hydrophilic doxorubicin and hydrophobic curcumin improved cytotoxicity, apoptosis, and cellular uptakein vitroand enhanced antitumor and anti-metastasis activityin vivoon breast carcinoma.
Collapse
|
50
|
Wegiel LA, Zhao Y, Mauer LJ, Edgar KJ, Taylor LS. Curcumin amorphous solid dispersions: the influence of intra and intermolecular bonding on physical stability. Pharm Dev Technol 2013; 19:976-86. [DOI: 10.3109/10837450.2013.846374] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|