1
|
Zhang H, Li S, Ma X. Transforming Healthcare with Nanomedicine: A SWOT Analysis of Drug Delivery Innovation. Drug Des Devel Ther 2024; 18:3499-3521. [PMID: 39132625 PMCID: PMC11314449 DOI: 10.2147/dddt.s470210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/11/2024] [Indexed: 08/13/2024] Open
Abstract
Objective Nanomedicine represents a transformative approach in biomedical applications. This study aims to delineate the application of nanomedicine in the biomedical field through the strengths, weaknesses, opportunities, and threats (SWOT) analysis to evaluate its efficacy and potential in clinical applications. Methods The SWOT analysis framework was employed to systematically review and assess the internal strengths and weaknesses, along with external opportunities and threats of nanomedicine. This method provides a balanced consideration of the potential benefits and challenges. Results Findings from the SWOT analysis indicate that nanomedicine presents significant potential in drug delivery, diagnostic imaging, and tissue engineering. Nonetheless, it faces substantial hurdles such as safety issues, environmental concerns, and high development costs. Critical areas for development were identified, particularly concerning its therapeutic potential and the uncertainties surrounding long-term effects. Conclusion Nanomedicine holds substantial promise in driving medical innovation. However, successful clinical translation requires addressing safety, cost, and regulatory challenges. Interdisciplinary collaboration and comprehensive strategic planning are crucial for the safe and effective application of nanomedicine.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Suping Li
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Xingming Ma
- School of Health Management, Xihua University, Chengdu, 610039, People’s Republic of China
| |
Collapse
|
2
|
Moradifar N, Moayyedkazemi A, Mohammadi HR, Ahmadi Somaghian S, Raziani Y. Investigating the potential application of organic and non-organic nanoparticles for gastric cancer treatment: An evidence-based review. ARCHIVES OF RAZI INSTITUTE 2024; 79:264-271. [PMID: 39463705 PMCID: PMC11512170 DOI: 10.32592/ari.2024.79.2.264] [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: 08/06/2023] [Accepted: 11/04/2023] [Indexed: 10/29/2024]
Abstract
Gastric cancer, which is considered a major health concern, is the sixth most frequent cancer and the second leading cause of cancer-related mortality across the globe. The present survey aimed to systematically review the anti-gastric cancer effect of all organic and inorganic nanoparticles (NPs) in in vitro, in vivo, and clinical trials. The investigation followed the PRISMA guidelines, and the findings were recorded in the CAMARADES-NC3Rs Preclinical Systematic Review and Meta-Analysis Facility database. A detailed search was conducted on various English databases, such as Scopus, Web of Science, EMBASE, PubMed, and Google Scholar, with no specified publication time frame to obtain papers regarding the anti-gastric cancer properties of nanoparticles. The search process was performed using the following terms "Nanoparticles," "Gastric cancer," "Anti-gastric cancer," "Metal nanoparticles," "Organic nanoparticles," "Inorganic nanoparticles, "in vitro," "Clinical," and "in vivo,". Out of 11,189 papers, 31 articles, including 19 (45.5%) in vitro, 3 (13.6%) in vivo, 3 (13.6%) clinical trials, and 6 (27.3%) in vitro/in vivo, up to 2023, met the inclusion criteria for discussion in this systematic review. The most widely used NPs were found to be organic nanoparticles, such as polylactic acid and poly lactic-co-glycolic acid (16, 80.0%), followed by inorganic nanoparticles, such as silver NPs (13, 41.9.0%). This review study highlighted the high anti-gastric cancer potential of a wide range of organic and non-organic NPs through their activity via some mechanisms, such as the induction of apoptosis, gene therapy, and drug delivery. Nonetheless, further studies, especially in clinical settings, are needed to confirm their anti-gastric effects and accurate mechanisms.
Collapse
Affiliation(s)
- N Moradifar
- Department of Internal Medicines, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - A Moayyedkazemi
- Department of Internal Medicines, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - H R Mohammadi
- Department of Internal Medicines, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - S Ahmadi Somaghian
- Department of Internal Medicines, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Y Raziani
- Nursing Department, Al-Mustaqbal University College, 51001 Hillah, Babylon, Iraq
| |
Collapse
|
3
|
Karimi P, Fakharzadeh S, Kalanaky S, Hafizi M, Hashemi M, Mahdavi M, Nazaran MH. Immunologic Mechanisms of BCc1 Nanomedicine Synthesized by Nanochelating Technology in Breast Tumor-bearing Mice: Immunomodulation and Tumor Suppression. Anticancer Agents Med Chem 2024; 24:1442-1456. [PMID: 39069805 DOI: 10.2174/0118715206302153240723053521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION The side effects of anti-cancer chemotherapy remain a concern for patients. So, designing alternative medications seems inevitable. In this research, the immunological mechanisms of BCc1 nanomedicine on tumor-bearing mice were investigated. METHODS BALB/c mice underwent tumor transplantation and were assigned into four groups. Group 1 was orally administered with PBS buffer, Group 2 was orally administered BCc1 10 mg/kg, and Group 3 was orally administered BCc1 40 mg/kg daily, respectively. In addition, a group of mice was administered Cyclophosphamide, 20 mg/kg daily. The weight and tumor volume of mice were evaluated bi-weekly. After 24 days of treatment, cytokines and CTL assay in the spleen cell and the tumor were assessed. Furthermore, the spleen, liver, kidney, lung, gut, and uterine tissue were stained with hematoxylin and eosin. Finally, the tumor samples were stained and analyzed for FOXP3. The survival rate of mice was recorded. RESULTS The results confirmed the histological safety of BCc1. This nanomedicine, especially BCc1 10 mg/kg, led to a strong IFN-γ response and suppressed TGF-β cytokine. The frequency of Treg in the tumor tissue of BCc1 nanomedicine groups was decreased. In addition, nanomedicine repressed tumor volume and tumor weight significantly, which was comparable to Cyclophosphamide. These immunologic events increased the survival rate of BCc1-treated groups. The results indicate that BCc1 nanomedicine can suppress tumor growth and thereby increase the survival rate of experimental mice. CONCLUSION It seems a modulation in the tumor microenvironment and polarization toward a Th1 response may be involved. So, BCc1 nanomedicine is efficient for human cancer therapy.
Collapse
Affiliation(s)
- Pegah Karimi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Advanced Therapy Medicinal Product (ATMP), Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Medical Division, Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | | |
Collapse
|
4
|
Hafizi M, Kalanaky S, Fakharzadeh S, Karimi P, Fakharian A, Lookzadeh S, Mortaz E, Mirenayat MS, Heshmatnia J, Karam MB, Zamani H, Nadji A, Toutkaboni MP, Oraee-Yazdani S, Akbari ME, Jamaati H, Nazaran MH. Beneficial effects of the combination of BCc1 and Hep-S nanochelating-based medicines on IL-6 in hospitalized moderate COVID-19 adult patients: a randomized, double-blind, placebo-controlled clinical trial. Trials 2023; 24:720. [PMID: 37951972 PMCID: PMC10638761 DOI: 10.1186/s13063-023-07624-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 09/05/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND In the severe forms of COVID-19 and many other infectious diseases, the patients develop a cytokine storm syndrome (CSS) where pro-inflammatory cytokines such as IL-6 and TNF-α play a key role in the development of this serious process. Selenium and iron are two important trace minerals, and their metabolism is tightly connected to immune system function. Numerous studies highlight the role of selenium and iron metabolism changes in the procedure of COVID-19 inflammation. The immunomodulator effect of nanomedicines that are synthesized based on nanochelating technology has been proved in previous studies. In the present study, the effects of the combination of BCc1(with iron-chelating property) and Hep-S (containing selenium) nanomedicines on mentioned cytokines levels in hospitalized moderate COVID-19 patients were evaluated. METHODS Laboratory-confirmed moderate COVID-19 patients were enrolled to participate in a randomized, double-blind, placebo-controlled study in two separate groups: combination of BCc1 and Hep-S (N = 62) (treatment) or placebo (N = 60) (placebo). The blood samples were taken before medications on day zero, at discharge, and 28 days after consumption to measure hematological and biochemical parameters and cytokine levels. The clinical symptoms of all the patients were recorded according to an assessment questionnaire before the start of the treatment and on days 3 and discharge day. RESULTS The results revealed that consumption of the nanomedicines led to a significant decrease in the mean level of IL-6 cytokine, and at the end of the study, there was a 77% downward trend in IL-6 in the nanomedicine group, while an 18% increase in the placebo group (p < 0.05). In addition, the patients in the nanomedicines group had lower TNF-α levels; accordingly, there was a 21% decrease in TNF-α level in the treatment group, while a 31% increase in this cytokine level in the placebo was observed (p > 0.05). On the other hand, in nanomedicines treated groups, clinical scores of coughing, fatigue, and need for oxygen therapy improved. CONCLUSIONS In conclusion, the combination of BCc1 and Hep-S inhibits IL-6 as a highly important and well-known cytokine in COVID-19 pathophysiology and presents a promising view for immunomodulation that can manage CSS. TRIAL REGISTRATION Iranian Registry of Clinical Trials RCT20170731035423N2 . Registered on June 12, 2020.
Collapse
Affiliation(s)
- Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Pegah Karimi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Atefeh Fakharian
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Lookzadeh
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sadat Mirenayat
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Heshmatnia
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Bakhshayesh Karam
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homa Zamani
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Nadji
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mihan Pourabdollah Toutkaboni
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Comprehensive Neurosurgical Center of Excellence, Shohada Tajrish, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hamidreza Jamaati
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
5
|
Kalanaky S, Fakharzadeh S, Karimi P, Hafizi M, Jamaati H, Hassanzadeh SM, Khorasani A, Mahdavi M, Nazaran MH. Nanoadjuvants Produced by Advanced Nanochelating Technology in the Inactivated-Severe Acute Respiratory Syndrome Coronavirus-2 Vaccine Formulation: Preliminary Results on Cytokines and IgG Responses. Viral Immunol 2023; 36:409-423. [PMID: 37506342 DOI: 10.1089/vim.2023.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023] Open
Abstract
Despite the great success of vaccines in various infectious diseases, most current vaccines are not effective enough, and on the contrary, clinically approved alum adjuvants cannot induce sufficient immune responses, including a potent cellular immune response to confer protection. In this study, we used Nanochelating Technology to develop novel nanoadjuvants to boost the potency of the alum-adjuvanted inactivated severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccine. BALB/c mice were immunized twice over 2 weeks with different doses of adjuvanted-vaccine formulations and immune responses were assessed. The analysis results of IFN-γ and IL-17 cytokines demonstrated the effectiveness of the nanoadjuvants produced by the Nanochelating Technology in shifting the alum-based vaccine toward a stronger Th1 pattern. In addition, these nanoadjuvants improved IL-2 cytokine response, which shows the efficacy of these novel formulations in inducing specific T lymphocyte proliferation. Using these nanoadjuvants increased IL-10 cytokine secretion that may be representative of a better immunoregulatory impact and may also potentially prevent immunopathology responses. Moreover, specific IgG titer analysis revealed the potency of these nanoadjuvants in improving humoral immune responses. The enzyme-linked immunosorbent assay of receptor-binding domain (RBD)-specific IgG response showed that the developed novel formulations induced strong IgG responses against this protein. This study shows that the nanostructures produced by the Advanced Nanochelating Technology have potent adjuvant effects on alum-based SARS-CoV-2 vaccines to not only compensate for alum weakness in inducing the cellular immune responses by smart regulation of the immune system but also significantly improve the humoral and cellular immune responses simultaneously.
Collapse
Affiliation(s)
- Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Pegah Karimi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mehdi Hassanzadeh
- Department of BCG Vaccine Production, Production and Research Complex, Pasteur Institute of Iran, Karaj, Iran
| | - Akbar Khorasani
- Department of FMD Vaccine Production, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Medical Division, Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Mohammad Hassan Nazaran
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
- Owner of Nanochelating Technology and Executive Manager and Chairman of Management Board of Sodour Ahrar Shargh Company, Tehran, Iran
| |
Collapse
|
6
|
Samuel JN, Booth CM, Eisenhauer E, Brundage M, Berry SR, Gyawali B. Association of Quality-of-Life Outcomes in Cancer Drug Trials With Survival Outcomes and Drug Class. JAMA Oncol 2022; 8:879-886. [PMID: 35482347 PMCID: PMC9052107 DOI: 10.1001/jamaoncol.2022.0864] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Importance Although quality of life (QOL) is an important clinical end point, cancer drugs are often approved based on overall survival (OS) or putative surrogate end points such as progression-free survival (PFS) without QOL data. Objective To ascertain whether cancer drug trials that show improvement in OS or PFS also improve global QOL of patients with cancer compared with the control treatment, as well as to assess how unchanged or detrimental QOL outcomes are reported in trial publications. Design, Setting, and Participants This retrospective cohort study included all patients with cancer in the advanced setting who were enrolled into phase 3 randomized clinical trials (RCTs) of cancer drugs reporting QOL data and published in English language in a PubMed-indexed journal in the calendar year 2019. The systematic search of PubMed was conducted in July 2020. Main Outcomes and Measures Association of QOL outcomes with OS and PFS, framing of unchanged QOL outcomes in trial publications, and the association of favorable framing with industry funding of the trials. Results A total of 45 phase 3 RCTs enrolling 24 806 participants (13 368 in the experimental arm and 11 438 in the control arm) met the inclusion criteria and were included in the study analyses. Improvement in global QOL with the experimental agent was reported in 11 (24%) RCTs. The RCTs with improved QOL were more likely to also show improved OS vs trials with unimproved QOL (7 of 11 [64%] trials vs 10 of 34 [29%] trials; χ2 = 4.13; P = .04); there was no such association observed for PFS (6 of 11 [55%] trials vs 17 of 34 [50%] trials, χ2 = 0.03; P = .87). Six trials reported worsening QOL, of which 3 (50%) were trials of targeted drugs, and 11 trials reported improvement in QOL, of which 6 (55%) were trials of immunotherapy drugs. Of the 34 trials in which QOL was not improved compared with controls, 16 (47%) reported these results in a positive frame, an observation statistically significantly associated with industry funding (χ2 = 6.35; P = .01). Conclusions and Relevance In this cohort study, a small proportion of RCTs of cancer drugs showed benefit in global QOL with the experimental agent. These results showed an association between QOL benefit and OS benefit but no such association with PFS benefit. Trials that failed to show improved QOL often reported their QOL outcomes more favorably. Non-immunotherapy-targeted drugs led to worse QOL more often than did cytotoxic agents.
Collapse
Affiliation(s)
- Joseph N Samuel
- School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Christopher M Booth
- Division of Cancer Care and Epidemiology, Cancer Research Institute, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Oncology, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Public Health Sciences, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Elizabeth Eisenhauer
- Department of Oncology, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Michael Brundage
- Division of Cancer Care and Epidemiology, Cancer Research Institute, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Oncology, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Scott R Berry
- Department of Oncology, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Bishal Gyawali
- Division of Cancer Care and Epidemiology, Cancer Research Institute, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Oncology, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Public Health Sciences, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
7
|
Zhao Z, Lu L, Li W. TAGLN2 promotes the proliferation, invasion, migration and epithelial-mesenchymal transition of colorectal cancer cells by activating STAT3 signaling through ANXA2. Oncol Lett 2021; 22:737. [PMID: 34466149 PMCID: PMC8387864 DOI: 10.3892/ol.2021.12998] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-associated mortality worldwide and currently ranks third in the USA in terms of prevalence. Transgelin-2 (TAGLN2) was previously reported to serve as a tumor promoter in various types of cancer. The present study aimed to investigate the role of TAGLN2 in the progression of CRC and to determine the potential underlying mechanism. The expression level of TAGLN2 in CRC cells (HCT116, SNU-C1, LoVo and SW480) were first detected by reverse transcription quantitative PCR and western blotting. Following TAGLN2 knockdown through transfection with short hairpin (sh)RNAs against TAGLN2, CRC cell proliferation was determined using Cell Counting Kit-8 and 5′-ethynyl-2′-deoxyuridine assays. Cell migration and invasion were evaluated using wound healing and Transwell assays, respectively. The expression levels of matrix metalloproteinase (MMP)2, MMP9 and proteins associated with epithelial-mesenchymal transition (EMT), including N-cadherin (N-cad), vimentin, zinc finger E-box binding homeobox 2 (ZEB2) and E-cadherin (E-cad), were also evaluated by western blotting. Furthermore, following TAGLN2 overexpression and the use of signal transducer and activator of transcription 3 (STAT3) inhibitors to treat CRC cells, all the aforementioned biological parameters were evaluated. The potential relationship between annexin 2 (ANXA2) and STAT3 was confirmed by western blotting analysis. The expression level of TAGLN2 was found to be particularly high in CRC cells. Following TAGLN2 knockdown, CRC cell proliferation, migration, invasion and EMT were significantly inhibited. TAGLN2 knockdown also suppressed STAT3 phosphorylation in CRC cells. In addition, the promoting effects of TAGLN2 overexpression on the progression of CRC were reversed by STAT3 inhibitor. Furthermore, ANXA2 was positively associated with STAT3. Taken together, these findings demonstrated that TAGLN2 could promote the proliferation, invasion, migration and EMT of CRC cells by activating STAT3 and regulating ANXA2 expression. This may reveal the underlying mechanism by which TAGLN2 might regulate the progression of CRC and provide potential therapeutic targets for the treatment of CRC.
Collapse
Affiliation(s)
- Zhicheng Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Weidong Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
8
|
Rupp SK, Stengel A. Influencing Factors and Effects of Treatment on Quality of Life in Patients With Gastric Cancer-A Systematic Review. Front Psychiatry 2021; 12:656929. [PMID: 34276435 PMCID: PMC8280526 DOI: 10.3389/fpsyt.2021.656929] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the leading causes of death worldwide. It is associated with several disease-related impairments contributing to the psycho-social burden of those patients, such as deterioration of well-being and overall quality of life (QOL). The aim of this study is to present the wide range of factors potentially impacting patients' overall well-being and possible preventive interventions. Methods: This systematic review was conducted in October 2020 with a search in the PubMed, MedLine, PsycInfo, and Google Scholar databases. We used the keywords "gastric cancer," "gastric neoplasm," and each of them combined with "quality of life," "depression," and "anxiety" to identify all relevant articles reporting about potential impact factors influencing the overall well-being of patients suffering from gastric cancer. Results: Finally, 125,490 articles were found, of which 125,431 were excluded in several steps of screening. Inclusion criteria were studies carried out on human ≥18 years of age, studies in English or German language, clinical trials, registry-based studies, cohort studies, population-based studies, and certain titles and abstracts. After screening for eligibility 35 potential factors influencing overall well-being in patients with GC were identified and classified into 9 important categories: genetic condition, treatment method, blood markers, nutritional status, daily living, state of health, mental state, supportive care, and alternative treatment. Conclusion: Since various factors are involved in the development of patients' overall well-being, timely treatment of psycho-social impairments by physicians and psychologists is of enormous importance. Preventing psycho-social burden by improving patients' QOL should be of high importance in the treatment regimen of patients with GC.
Collapse
Affiliation(s)
- Sophia Kristina Rupp
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Stengel
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany.,Section Psychooncology, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Tübingen, Germany.,Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
9
|
Fakharzadeh S, Kalanaky S, Argani H, Dadashzadeh S, Torbati PM, Nazaran MH, Basiri A. Ameliorative effect of a nano chromium metal-organic framework on experimental diabetic chronic kidney disease. Drug Dev Res 2020; 82:393-403. [PMID: 33230842 DOI: 10.1002/ddr.21759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/24/2020] [Accepted: 10/24/2020] [Indexed: 12/21/2022]
Abstract
Metal-Organic Frameworks (MOFs) are a new class of crystalline porous structures which can be used as a novel structure in diverse fields of medical science. Several studies have shown that chromium supplementation can be effective in amelioration of biochemical parameters of diabetes and its renal complications. Therefore, a chromium-containing MOF (DIFc) was synthetized by nanochelating technology in the present study and then its effect on biochemical indices in diabetic rats was evaluated. Diabetes was induced by high-fat diet consumption and streptozotocin (35 mg/kg) injection and then the treatment started 8 weeks after disease induction and continued for 8 weeks. The results showed that DIFc treatment decreased HOMA-IR index, blood urea nitrogen, uric acid and malondialdehyde in plasma samples. This nano MOF also reduced albumin, malondialdehyde and 8-isoprostane in urine specimen, while it increased creatinine clearance. In conclusion, DIFc MOF demonstrated promising results in the present study, indicating that it can be developed and evaluated in future investigations with the aim of designing a novel agent for management of diabetes and its renal complications.
Collapse
Affiliation(s)
- Saideh Fakharzadeh
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Hassan Argani
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Mohammadi Torbati
- Department of Pathology, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Ding Z, Sigdel K, Yang L, Liu Y, Xuan M, Wang X, Gu Z, Wu J, Xie H. Nanotechnology-based drug delivery systems for enhanced diagnosis and therapy of oral cancer. J Mater Chem B 2020; 8:8781-8793. [PMID: 33026383 DOI: 10.1039/d0tb00957a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oral cancer is a common malignant life-threatening tumor. Despite some advances in traditional therapy, mortality and mobidity rates are high due to delayed diagnosis and ineffective treatment. Additionally, some patients inevitably suffer from various fatal adverse effects during the course of therapy. Therefore, it is imperative to develop novel methods to eradicate oral cancer cells with minimal adverse effects on normal cells. Nanotechnology is a promising and novel vehicle for the diagnosis and treatment of oral cancer with encouraging recent achievements. In this review, we present state-of-the-art nanotechnology-based drug delivery systems employed in the domain of oral cancer, especially for its enhanced diagnosis and therapy. We describe in detail the types of nanotechnology used in the management of oral cancer and summarize administration routes of nanodrugs. Finally, the potential and prospects of nanotechnology-based drug delivery systems as promising modalities of diagnosis and therapy of oral cancer are highlighted.
Collapse
Affiliation(s)
- Zhangfan Ding
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Liu J, Zhang Y, Li Q, Wang Y. Transgelins: Cytoskeletal Associated Proteins Implicated in the Metastasis of Colorectal Cancer. Front Cell Dev Biol 2020; 8:573859. [PMID: 33117801 PMCID: PMC7575706 DOI: 10.3389/fcell.2020.573859] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022] Open
Abstract
Transgelins, including transgelin-1 (T-1), transgelin-2 (T-2), and transgelin-3 (T-3), are a family of actin-binding proteins (ABPs) that can alter the structure and morphology of the cytoskeleton. These proteins function by regulating migration, proliferation and apoptosis in many different cancers. Several studies have shown that in various types of tumor cells, including colorectal cancer (CRC) cells, and in the tumor microenvironment, the expression and biological effects of transgelins are diverse and may transform during tumor progression. Previous researches have demonstrated that transgelin levels are positively correlated with metastasis in CRC, and down-regulating their expression can inhibit this process. In advanced disease, T-1 is a tumor activator with increasing expression, and T-2 expression increases with the progression of CRC. Finally, T-3 is only expressed in neurons and is not associated with CRC. This evidence suggests that T-1 and T-2 are potential biomarkers and therapeutic targets for CRC metastasis.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Hafizi M, Kalanaky S, Fakharzadeh S, Janzamin E, Arjmandi T, Atashi A, Nazaran MH. GFc7 as a Smart Growth Nanofactor for ex vivo Expansion and Cryoprotection of Humans' Hematopoietic Stem Cells. Int J Nanomedicine 2020; 15:6263-6277. [PMID: 32922002 PMCID: PMC7457843 DOI: 10.2147/ijn.s256104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background Nowadays, smart synthesized nanostructures have attracted wide attention in the field of stem cell nanotechnology due to their effect on different properties of stem cells. Methods GFc7 growth nanofactor was synthesized based on nanochelating technology as an iron-containing copper chelator nanocomplex. The effect of this nanocomplex on the expansion and differentiation of hematopoietic stem cells (HSCs) as well as its performance as a cryoprotectant was evaluated in the present study. Results The results showed that the absolute count of CD34+ and CD34+CD38- cells on days 4, 7, 10 and 13; the percentage of lactate dehydrogenase enzyme on the same days and CD34+CXCR4 population on day 10 were significantly increased when they were treated with GFc7 growth nanofactor in a fetal bovine serum (FBS)-free medium. This medium also led to delayed differentiation in HSCs. One noticeable result was that CD34+CD38- cells cultured in an FBS medium were immediately differentiated into CD34+CD38+ cells, while CD34+CD38- cells treated with GFc7 growth nanofactor in FBS medium did not show such an immediate significant differentiation. De-freezing GFc7-treated CD34+ cells, which were already frozen according to cord blood bank protocols, showed a higher percentage of cell viability and a larger number of colonies according to colony-forming cell assay as compared to control. Conclusion It can be claimed that treating HSCs with GFc7 growth nanofactor leads to quality and quantity improvement of HSCs, both in terms of expansion in vitro and freezing and de-freezing processes.
Collapse
Affiliation(s)
- Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | | | - Tarlan Arjmandi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | |
Collapse
|
13
|
Fakharzadeh S, Argani H, Torbati PM, Dadashzadeh S, Kalanaky S, Nazaran MH, Basiri A. DIBc nano metal-organic framework improves biochemical and pathological parameters of experimental chronic kidney disease. J Trace Elem Med Biol 2020; 61:126547. [PMID: 32460199 DOI: 10.1016/j.jtemb.2020.126547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The growing morbidity and mortality rate of chronic kidney disease (CKD) has forced researchers to find more efficient strategies for controlling this disease. Studies have proven the important role of alteration in iron, zinc and selenium metabolism in CKD pathological process. Nanotechnology, through synthetizing nano metal-organic framework (NMOF) structures, can be employed as a valuable strategy for using these trace elements as the key for modification and improvement of CKD-related pathological events. After proving the anti-diabetic property of DIBc NMOF (which contains selenium and zinc) in the previous study, the impact of this NMOF on some important biochemical and pathological parameters of CKD was evaluated in the current study. METHODS Knowing that diabetic nephropathy (DN) is the leading cause of CKD, male wistar rats were selected and given a high fat diet for 2 weeks and then were injected with streptozotocin (35 mg/kg) to induce DN. Six weeks after streptozotocin injection, DIBc or metformin treatment started and continued for 8 weeks. RESULTS Eight weeks of DIBc treatment decreased plasma fasting blood glucose, blood urea nitrogen, uric acid, malondialdehyde (MDA) and HOMA-IR index compared to DN control and metformin groups. This NMOF significantly reduced urinary albumin excretion rate, MDA and 8-isoprostane, while it increased creatinine clearance in comparison to the above-mentioned groups. Renal histo-pathological images indicated that DIBc ameliorated glomerular basement membrane thickening and wrinkling, mesangial matrix expansion and hypercellularity and presence of intra-cytoplasmic hyaline droplets in proximal cortical tubules of kidney samples. CONCLUSION The results showed the therapeutic effect of DIBc on important biochemical and histo-pathological parameters of CKD, so this NMOF could be regarded as a promising novel anti-CKD agent.
Collapse
Affiliation(s)
- Saideh Fakharzadeh
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Hassan Argani
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Mohammadi Torbati
- Department of Pathology, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | | | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Fakharzadeh S, Argani H, Dadashzadeh S, Kalanaky S, Mohammadi Torbati P, Nazaran MH, Basiri A. BCc1 Nanomedicine Therapeutic Effects in Streptozotocin and High-Fat Diet Induced Diabetic Kidney Disease. Diabetes Metab Syndr Obes 2020; 13:1179-1188. [PMID: 32368111 PMCID: PMC7173843 DOI: 10.2147/dmso.s240757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/31/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND One common feature of chronic diseases, such as cancer, diabetes and chronic kidney disease (CKD), is the disruption of iron metabolism and increase in labile iron pool, which can result in excessive production of harmful oxidative stress. The proper management of iron metabolism in this situation can be a valuable tool to ameliorate pathological events. MATERIALS AND METHODS In the previous studies, the anti-neoplastic effects of BCc1, a nanochelating-based nanomedicine with iron-chelating property, were demonstrated in cell culture, animal models and clinical trials. In the present study, the therapeutic effects of BCc1 in animal model of diabetic kidney disease (DKD), induced by streptozotocin injection (35 mg/kg) and high-fat diet consumption, were evaluated. RESULTS The results showed that BCc1 significantly decreased HOMA-IR index, uric acid, blood urea nitrogen, malondialdehyde and 8-isoprostane. In addition, it reduced urinary albumin excretion rate and albumin-to-creatinine ratio in comparison to DKD control rats. This nanomedicine had no negative impact on liver iron content, hemoglobin level, red blood cell count, hematocrit and mean corpuscular volume, while it significantly decreased aspartate aminotransferase and alanine aminotransferase compared to DKD control group. Moreover, the histopathological assessment indicated that lesser glomerular basement membrane and wrinkling, mesangial matrix expansion and pathological changes in proximal cortical tubules were seen in the kidney samples of BCc1-treated rats. CONCLUSION In conclusion, BCc1 as an iron-chelating agent shows promising impacts in DKD animal model, which can ameliorate biochemical and pathological events of this disease.
Collapse
Affiliation(s)
- Saideh Fakharzadeh
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Hassan Argani
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Peyman Mohammadi Torbati
- Department of Pathology, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Nazaran
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
- Correspondence: Mohammad Hassan Nazaran Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran Tel/Fax +98 21 88992123 Email
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Abbas Basiri Urology and Nephrology Research Center, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran Tel/Fax +98 21 22567222 Email
| |
Collapse
|
15
|
An Investigation on the Effect of BCc1 Nanomedicine on Gastric Cancer Patients Using EORTC QLQ-STO30 Questionnaire. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2019. [DOI: 10.5812/ijcm.94190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|