1
|
Escalera-Anzola S, Rosado M, Yang Y, Parra-Sanchez D, Pedro-Liberal CS, Acedo P. Breakthroughs in nanoparticle-based strategies for pancreatic cancer therapy. Biochem Pharmacol 2024; 232:116685. [PMID: 39613113 DOI: 10.1016/j.bcp.2024.116685] [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: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, mainly due to its high heterogeneity, resistance to therapy and late diagnosis, with a 5-year survival rate of less than 10%. This dismal prognosis has promoted strategies to develop more effective treatments. Nanoparticle-based strategies have emerged, in the last decades, as a great opportunity because they can enhance drug delivery and promote controlled release, presenting lower side effects than conventional therapeutic regimens. Moreover, nanoparticles can often be modified to target specific cells or to achieve a sustained release of the drugs into the tumor. However, very few nanoparticle-based therapies are clinically approved. Concretely for pancreatic cancer treatment only two nanoformulations have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) so far. Clinical translation of nanoparticles remains a challenge for modern medicine, and in particular for pancreatic cancer therapy, because of the complexity of the disease, and a lack of studies been performed in clinically relevant in vitro and in vivo models. In this review, we have summarized the most recent clinical trials using nanoparticle-based formulations in PDAC, giving a small context of the diverse types of nanoparticles employed and the most recent advancements in the field.
Collapse
Affiliation(s)
- Sara Escalera-Anzola
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom; Smart Devices for Nano Medicine Group, Unidad Excelencia Instituto de BioMedicina y Genética Molecular (IBGM) de Valladolid, University of Valladolid and CSIC, Valladolid, Spain
| | - Maria Rosado
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Yuchen Yang
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Daniel Parra-Sanchez
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Carolina San Pedro-Liberal
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom.
| |
Collapse
|
2
|
Angelocci LV, Sgrignoli SS, de Souza CD, Antunes PCG, Rostelato MECM, Zeituni CA. In silicodosimetry for a prostate cancer treatment using 198Au nanoparticles. Biomed Phys Eng Express 2024; 11:015002. [PMID: 39447593 DOI: 10.1088/2057-1976/ad8acc] [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: 08/22/2024] [Accepted: 10/24/2024] [Indexed: 10/26/2024]
Abstract
Objective. To estimate dose rates delivered by using radioactive198Au nanoparticles for prostate cancer nanobrachytherapy, identifying contribution by photons and electrons emmited from the source.Approach. Utilizingin silicomodels, two different anatomical representations were compared: a mathematical model and a unstructured mesh model based on the International Commission on Radiological Protection (ICRP) Publication 145 phantom. Dose rates by activity were calculated to the tumor and nearby healthy tissues, including healthy prostate tissue, urinary bladder wall and rectum, using Monte Carlo code MCNP6.2.Main results. Results indicate that both models provide dose rate estimates within the same order of magnitude, with the mathematical model overestimating doses to the prostate and bladder by approximately 20% compared to the unstructured mesh model. The discrepancies for the tumor and rectum were below 4%. Photons emmited from the source were defined as the primary contributors to dose to other organs, while 97.9% of the dose to the tumor was due to electrons emmited from the source.Significance. Our findings emphasize the importance of model selection in dosimetry, particularly the advantages of using realistic anatomical phantoms for accurate dose calculations. The study demonstrates the feasibility and effectiveness of198Au nanoparticles in achieving high dose concentrations in tumor regions while minimizing exposure to surrounding healthy tissues. Beta emissions were found to be predominantly responsible for tumor dose delivery, reinforcing the potential of198Au nanoparticles in localized radiation therapy. We advocate for using realistic body phantoms in further research to enhance reliability in dosimetry for nanobrachytherapy, as the field still lacks dedicated protocols.
Collapse
Affiliation(s)
- Lucas Verdi Angelocci
- Centro de Tecnologia das Radiações, Instituto de Pesquisas Energéticas e Nucleareas, São Paulo, SP, Brazil
| | | | - Carla Daruich de Souza
- Centro de Tecnologia das Radiações, Instituto de Pesquisas Energéticas e Nucleareas, São Paulo, SP, Brazil
- Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Carlos Alberto Zeituni
- Centro de Tecnologia das Radiações, Instituto de Pesquisas Energéticas e Nucleareas, São Paulo, SP, Brazil
| |
Collapse
|
3
|
Qin X, Liu J. Nanoformulations for the diagnosis and treatment of metabolic dysfunction-associated steatohepatitis. Acta Biomater 2024; 184:37-53. [PMID: 38879104 DOI: 10.1016/j.actbio.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive phase of metabolic dysfunction-associated steatotic liver disease (MASLD) that develops into irreversible liver cirrhosis and hepatocellular carcinoma, ultimately necessitating liver transplantation as the sole life-saving option. However, given the drawbacks of liver transplantation, including invasiveness, chronic immunosuppression, and a lack of donor livers, prompt diagnosis and effective treatment are indispensable. Due to the limitations of liver biopsy and conventional imaging modalities in diagnosing MASH, as well as the potential hazards associated with liver-protecting medicines, numerous nanoformulations have been created for MASH theranostics. Particularly, there has been significant study interest in artificial nanoparticles, natural biomaterials, and bionic nanoparticles that exhibit exceptional biocompatibility and bioavailability. In this review, we summarized extracellular vesicles (EVs)-based omics analysis and Fe3O4-based functional magnetic nanoparticles as magnetic resonance imaging (MRI) contrast agents for MASH diagnosis. Additionally, artificial nanoparticles such as organic and inorganic nanoparticles, as well as natural biomaterials such as cells and cell-derived EVs and bionic nanoparticles including cell membrane-coated nanoparticles, have also been reported for MASH treatment owing to their specific targeting and superior therapeutic effect. This review has the potential to stimulate advancements in nanoformulation fabrication techniques. By exploring their compatibility with cell biology, it could lead to the creation of innovative material systems for efficient theragnostic uses for MASH. STATEMENT OF SIGNIFICANCE: People with metabolic dysfunction-associated steatohepatitis (MASH) will progress to fibrosis, cirrhosis, or even liver cancer. It is imperative to establish effective theragnostic techniques to stop MASH from progressing into a lethal condition. In our review, we summarize the advancement of artificial, natural, and bionic nanoparticles applied in MASH theragnosis. Furthermore, the issues that need to be resolved for these cutting-edge techniques are summarized to realize a more significant clinical impact. We forecast the key fields that will advance further as nanotechnology and MASH research progress. Generally, our discovery has significant implications for the advancement of nanoformulation fabrication techniques, and their potential to be compatible with cell biology could lead to the creation of innovative materials systems for effective MASH theragnostic.
Collapse
Affiliation(s)
- Xueying Qin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Jingjing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China.
| |
Collapse
|
4
|
Zhang Y, Watson S, Ramaswamy Y, Singh G. Intravitreal therapeutic nanoparticles for age-related macular degeneration: Design principles, progress and opportunities. Adv Colloid Interface Sci 2024; 329:103200. [PMID: 38788306 DOI: 10.1016/j.cis.2024.103200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 05/11/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss in the elderly. The current standard treatment for AMD involves frequent intravitreal administrations of therapeutic agents. While effective, this approach presents challenges, including patient discomfort, inconvenience, and the risk of adverse complications. Nanoparticle-based intravitreal drug delivery platforms offer a promising solution to overcome these limitations. These platforms are engineered to target the retina specifically and control drug release, which enhances drug retention, improves drug concentration and bioavailability at the retinal site, and reduces the frequency of injections. This review aims to uncover the design principles guiding the development of highly effective nanoparticle-based intravitreal drug delivery platforms for AMD treatment. By gaining a deeper understanding of the physiology of ocular barriers and the physicochemical properties of nanoparticles, we establish a basis for designing intravitreal nanoparticles to optimize drug delivery and drug retention in the retina. Furthermore, we review recent nanoparticle-based intravitreal therapeutic strategies to highlight their potential in improving AMD treatment efficiency. Lastly, we address the challenges and opportunities in this field, providing insights into the future of nanoparticle-based drug delivery to improve therapeutic outcomes for AMD patients.
Collapse
Affiliation(s)
- Yuhang Zhang
- The School of Biomedical Engineering, Faculty of IT and Engineering, Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2008, Australia
| | - Stephanie Watson
- Faculty of Medicine and Health, Clinical Ophthalmology and Eye Health, Save Sight Institute, The University of Sydney, Camperdown, NSW 2008, Australia
| | - Yogambha Ramaswamy
- The School of Biomedical Engineering, Faculty of IT and Engineering, Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2008, Australia
| | - Gurvinder Singh
- The School of Biomedical Engineering, Faculty of IT and Engineering, Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2008, Australia.
| |
Collapse
|
5
|
Li H, Wang S, Yang Z, Meng X, Niu M. Nanomaterials modulate tumor-associated macrophages for the treatment of digestive system tumors. Bioact Mater 2024; 36:376-412. [PMID: 38544737 PMCID: PMC10965438 DOI: 10.1016/j.bioactmat.2024.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 11/25/2024] Open
Abstract
The treatment of digestive system tumors presents challenges, particularly in immunotherapy, owing to the advanced immune tolerance of the digestive system. Nanomaterials have emerged as a promising approach for addressing these challenges. They provide targeted drug delivery, enhanced permeability, high bioavailability, and low toxicity. Additionally, nanomaterials target immunosuppressive cells and reshape the tumor immune microenvironment (TIME). Among the various cells in the TIME, tumor-associated macrophages (TAMs) are the most abundant and play a crucial role in tumor progression. Therefore, investigating the modulation of TAMs by nanomaterials for the treatment of digestive system tumors is of great significance. Here, we present a comprehensive review of the utilization of nanomaterials to modulate TAMs for the treatment of gastric cancer, colorectal cancer, hepatocellular carcinoma, and pancreatic cancer. We also investigated the underlying mechanisms by which nanomaterials modulate TAMs to treat tumors in the digestive system. Furthermore, this review summarizes the role of macrophage-derived nanomaterials in the treatment of digestive system tumors. Overall, this research offers valuable insights into the development of nanomaterials tailored for the treatment of digestive system tumors.
Collapse
Affiliation(s)
- Hao Li
- Department of Interventional Radiology, First Hospital of China Medical University, Shenyang, China
| | - Shuai Wang
- Department of Interventional Radiology, First Hospital of China Medical University, Shenyang, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Meng Niu
- China Medical University, Shenyang, China
| |
Collapse
|
6
|
Fatima T, Abrar H, Jahan N, Shamim S, Ahmed N, Ali AB, Begum I, Ahmed W. Molecular marker identification, antioxidant, antinociceptive, and anti-inflammatory responsiveness of malonic acid capped silver nanoparticle. Front Pharmacol 2024; 14:1319613. [PMID: 38357362 PMCID: PMC10864560 DOI: 10.3389/fphar.2023.1319613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Nano-sized silver has drawn a great deal of attention in the field of health sciences owing to its remarkable therapeutic applications. Interestingly, the method applied to synthesize nanoparticles and the choice of reagents considerably influence their therapeutic potential and toxicities. Current research has explored the toxicity, anti-inflammatory, antinociceptive, and antioxidant responses of the malonic acid-capped silver nanoparticles (MA-AgNPs (C) by using sodium borohydride as a reducing agent at low temperatures by employing both in vitro and in vivo approaches. Furthermore, it has highlighted the synergistic effect of these novel compounds with conventional anti-inflammatory therapeutic agents. Acute and sub-acute toxicity analysis performed following OECD guidelines showed that the studied MA-AgNPs (C) are safer, and prominent toxic signs have not been detected at the highest studied dose of 2,000 mg/kg. Cytotoxicity evaluation through brine shrimp lethality revealed 20% lethality at the highest concentration of 169.8 μg/mL. Significantly, positive anti-inflammatory and analgesic responses alone as well as synergism with the standard were identified through in vitro as well as in vivo methods which were more potent at a lower dose (200 mg/kg). Notably synergistic outcomes were more pronounced than individual ones, indicating their prominent effect as a feasible drug delivery system. IL-6 and TNF-α assessment in excised paw tissue through RTPCR technique further supported their anti-inflammatory potential. DPPH assay revealed eminent in vitro antioxidant activity which was further corroborated by in vivo antioxidant assessment through evaluation of SOD in excised paw tissue.
Collapse
Affiliation(s)
- Tehrim Fatima
- Department of Pharmacology, Dow College of Pharmacy, Faculty of Pharmaceutical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Hina Abrar
- Department of Pharmacology, Dow College of Pharmacy, Faculty of Pharmaceutical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Noor Jahan
- Department of Pharmacology, Dow College of Pharmacy, Faculty of Pharmaceutical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Sana Shamim
- Department of Pharmaceutical Chemistry, Dow College of Pharmacy, Faculty of Pharmaceutical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Nazia Ahmed
- Dow Research Institute of Biotechnology and Biosciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Asma Basharat Ali
- Department of Anatomy, Jinnah Medical and Dental College, Karachi, Pakistan
| | - Irshad Begum
- Department of Chemistry, University of Karachi, Karachi, Pakistan
| | - Waqas Ahmed
- School of Public Health, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
7
|
Mekapogu AR, Xu Z, Pothula S, Perera C, Pang T, Hosen SMZ, Damalanka V, Janetka J, Goldstein D, Pirola R, Wilson J, Apte M. HGF/c-Met pathway inhibition combined with chemotherapy increases cytotoxic T-cell infiltration and inhibits pancreatic tumour growth and metastasis. Cancer Lett 2023:216286. [PMID: 37354984 DOI: 10.1016/j.canlet.2023.216286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Pancreatic cancer (PC) is a deadly cancer with a high mortality rate. The unique characteristics of PC, including desmoplasia and immunosuppression, have made it difficult to develop effective treatment strategies. Pancreatic stellate cells (PSCs) play a crucial role in the progression of the disease by interacting with cancer cells. One of the key mediators of PSC - cancer cell interactions is the hepatocyte growth factor (HGF)/c-MET pathway. Using an immunocompetent in vivo model of PC as well as in vitro experiments, this study has shown that a combined approach using HGF/c-MET inhibitors to target stromal-tumour interactions and chemotherapy (gemcitabine) to target cancer cells effectively decreases tumour volume, EMT, and stemness, and importantly, eliminates metastasis. Notably, HGF/c-MET inhibition decreases TGF-β secretion by cancer cells, resulting in an increase in cytotoxic T-cell infiltration, thus contributing to cancer cell death in tumours. HGF/c-MET inhibition + chemotherapy was also found to normalise the gut microbiome and improve gut microbial diversity. These findings provide a strong platform for assessment of this triple therapy (HGF/c-MET inhibition + chemotherapy) approach in the clinical setting.
Collapse
Affiliation(s)
- Alpha Raj Mekapogu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Srinivasa Pothula
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia; AbCellera, Beaconsfield, New South Wales, United Kingdom
| | - Chamini Perera
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Tony Pang
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia; Surgical Innovations Unit, Westmead Hospital, Sydney, Australia; Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - S M Zahid Hosen
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Vishnu Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - James Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - David Goldstein
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Romano Pirola
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Jeremy Wilson
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Minoti Apte
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia.
| |
Collapse
|
8
|
Soni A, Bhandari MP, Tripathi GK, Bundela P, Khiriya PK, Khare PS, Kashyap MK, Dey A, Vellingiri B, Sundaramurthy S, Suresh A, Pérez de la Lastra JM. Nano-biotechnology in tumour and cancerous disease: A perspective review. J Cell Mol Med 2023; 27:737-762. [PMID: 36840363 PMCID: PMC10002932 DOI: 10.1111/jcmm.17677] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/07/2022] [Accepted: 11/18/2022] [Indexed: 02/26/2023] Open
Abstract
In recent years, drug manufacturers and researchers have begun to consider the nanobiotechnology approach to improve the drug delivery system for tumour and cancer diseases. In this article, we review current strategies to improve tumour and cancer drug delivery, which mainly focuses on sustaining biocompatibility, biodistribution, and active targeting. The conventional therapy using cornerstone drugs such as fludarabine, cisplatin etoposide, and paclitaxel has its own challenges especially not being able to discriminate between tumour versus normal cells which eventually led to toxicity and side effects in the patients. In contrast to the conventional approach, nanoparticle-based drug delivery provides target-specific delivery and controlled release of the drug, which provides a better therapeutic window for treatment options by focusing on the eradication of diseased cells via active targeting and sparing normal cells via passive targeting. Additionally, treatment of tumours associated with the brain is hampered by the impermeability of the blood-brain barriers to the drugs, which eventually led to poor survival in the patients. Nanoparticle-based therapy offers superior delivery of drugs to the target by breaching the blood-brain barriers. Herein, we provide an overview of the properties of nanoparticles that are crucial for nanotechnology applications. We address the potential future applications of nanobiotechnology targeting specific or desired areas. In particular, the use of nanomaterials, biostructures, and drug delivery methods for the targeted treatment of tumours and cancer are explored.
Collapse
Affiliation(s)
- Ambikesh Soni
- School of NanotechnologyRajiv Gandhi Proudyogiki VishwavidyalayaBhopalIndia
| | | | | | - Priyavand Bundela
- School of NanotechnologyRajiv Gandhi Proudyogiki VishwavidyalayaBhopalIndia
| | | | | | - Manoj Kumar Kashyap
- Amity Stem Cell Institute, Amity Medical SchoolAmity University HaryanaHaryanaIndia
| | - Abhijit Dey
- Department of Life SciencesPresidency UniversityWest BengalKolkataIndia
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational ResearchDepartment of ZoologySchool of Basic Sciences, Central University of PunjabMaulana Azad National Institute of TechnologyBathindaIndia
| | - Suresh Sundaramurthy
- Department of Chemical EngineeringMaulana Azad National Institute of TechnologyMadhya PradeshBhopalIndia
| | - Arisutha Suresh
- Department of EnergyMaulana Azad National Institute of Technology & M/s Eco Science & TechnologyMadhya PradeshBhopalIndia
| | - José M. Pérez de la Lastra
- Biotecnología de macromoléculasInstituto de Productos Naturales y Agrobiología, (IPNA‐CSIC)San Cristóbal de la LagunaSpain
| |
Collapse
|
9
|
Oladipo A, Ejeromedoghene O, Adebayo A, Ogunyemi O, Egejuru G. A mini review on the prospects of Fagara zanthoxyloides extract based composites: a remedy for COVID-19 and associated replica? PHYSICAL SCIENCES REVIEWS 2023. [DOI: 10.1515/psr-2022-0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Abstract
Studies are still being conducted to find a sustainable and long-lasting solution to the lethal consequences of the feared virus characterized as coronavirus disease (Covid-19) and its accompanying pathogenic replication, which pose a serious threat to human survival in the wake of its broad distribution. Since its emergence, researchers have investigated synthetic approaches in search of a dependable vaccine or treatment and curtail the spread of the virus and also enhance the health of a patient who has been affected. Unfortunately, the infection is yet to be entirely eradicated in many parts of the world. Despite the introduction of synthetic pharmaceuticals like remdesivir and derivatives of chloroquine, plant extracts may be an alternative reliable strategy that could successfully combat the operation of the virus. Herein, we investigated the prospects of fagara zanthoxyloides lam. (rutaceae) (syn. zanthoxylum zanthoxyloides), a well-known medicinal tree whose extracts have demonstrated success in treating many microbiological and viral-related infections. The distinctive plant extracts contain several bioactive phytochemicals with promising biological activity with minimal or no side effects and are being researched for a variety of applications, particularly in the pharmaceutical and medicinal industries. Consequently, in this review, we examined the crude extracts from the Fagara species and suggested that careful consideration should be given to its independent use or combination with other bioactive molecules, such as biopolymers and nano-metallic composites, to combat the terrifying Covid-19 virus and its associates.
Collapse
Affiliation(s)
- Abiodun Oladipo
- Co-Innovation Center for Sustainable Forestry in Southern China , College of Forestry , Nanjing Forestry University , 210037 Nanjing , Jiangsu , P. R. China
| | - Onome Ejeromedoghene
- School of Chemistry and Chemical Engineering , Southeast University , Jiangning District , Nanjing , Jiangsu Province , 211189 , P. R. China
| | - Ademola Adebayo
- Department of Forest and Conservation Sciences , Faculty of Forestry , University of British Columbia , V6T1Z4 B.C , Vancouver , Canada
| | - Olakunle Ogunyemi
- Department of Forestry and Wildlife Management , Federal University of Agriculture , PMB 2240 Abeokuta , Ogun State , Nigeria
| | - George Egejuru
- School of Public Health , Southeast University , Jiangning District , Nanjing , Jiangsu Province , 211189 , P. R. China
| |
Collapse
|
10
|
Liao C, Wu Z, Lin C, Chen X, Zou Y, Zhao W, Li X, Huang G, Xu B, Briganti GE, Qi Y, Wang X, Zeng T, Wuethrich A, Zou H. Nurturing the marriages of urinary liquid biopsies and nano-diagnostics for precision urinalysis of prostate cancer. SMART MEDICINE 2023; 2:e20220020. [PMID: 39188554 PMCID: PMC11236013 DOI: 10.1002/smmd.20220020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/04/2022] [Indexed: 08/28/2024]
Abstract
Prostate cancer remains the second-most common cancer diagnosed in men, despite the increasingly widespread use of serum prostate-specific antigen (PSA) screening. The controversial clinical implications and cost benefits of PSA screening have been highlighted due to its poor specificity, resulting in a high rate of overdiagnosis and underdiagnosis. Thus, the development of novel biomarkers for prostate cancer detection remains an intriguing challenge. Urine is emerging as a source for prostate cancer biomarker discovery. Currently, new urine biomarkers already outperform serum PSA in clinical diagnosis. Meanwhile, the advances in nanotechnology have provided a suite of diagnostic tools to study prostate cancer in more detail, sparking a new era of biomarker discoveries. In this review, we envision that future prostate cancer diagnosis will probably integrate multiplex nano-diagnostic approaches to detect novel urinary biomarkers. However, challenges remain in differentiating indolent from aggressive cancers to better inform treatment decisions, and clinical translation still needs to be overcome.
Collapse
Affiliation(s)
- Caizhi Liao
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Zhihao Wu
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Chan Lin
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Xiaofeng Chen
- School of Environmental and Geographical SciencesShanghai Normal UniversityShanghaiChina
- School of ChemistryNorthwestern UniversityChicagoIllinoisUSA
| | - Yaqun Zou
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Wan Zhao
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Xin Li
- Department of UrologySir Run Run Shaw HospitalZhejiang UniversityHangzhouChina
| | | | - Baisheng Xu
- Department of UrologyThe First People's Hospital of XiushuiJiujiangChina
| | | | - Yan Qi
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Xianshu Wang
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
| | - Tao Zeng
- Department of Urologythe Second Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Alain Wuethrich
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of QueenslandBrisbaneQueenslandAustralia
| | - Hongzhi Zou
- Creative Biosciences (Guangzhou) Co., LtdGuangzhouChina
- The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
11
|
Zinc Oxide Nanoparticles as Diagnostic Tool for Cancer Cells. Int J Biomater 2022; 2022:2807644. [PMID: 36387955 PMCID: PMC9646305 DOI: 10.1155/2022/2807644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/21/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
ZnO nanoparticles have various characteristics that make them attractive to be used in many medical applications like a cancer diagnosis. It can be used as a nanoprobe for targeting different types of cancer cells in vitro as a cancer cell recognition system. The present study aims to investigate the permeability of ZnO NPs through both normal and cancerous cell lines in humans. In vitro experiments for ZnO NPs inside the environment of living cells have been described, which would contribute to the visualization of nanoparticles as cancer diagnostic and scanning techniques. MCF7, AMJ13, and RD cancer cells, and also the normal breast cell line HBL, were used in in vitro imaging experiments. The findings revealed that ZnO NPs specifically incorporated within tumor cells while accumulating less inside normal cells. Our findings show that ZnO NPs may be identified inside cancer cells after 1 h of exposure and can endure up to 3 h, providing them appropriate for tumor cell imaging. The findings showed that ZnO NPs might be employed as an alternate fluorophore for diagnostic imaging in the early identification of solid cancers. Therefore, here we studied in vitro applications of ZnO NPs and their beneficial use as a diagnostic tool for cancer cell lines rather than normal cells. Taken together, ZnO NPs can be used as good targeting NPs for the development of imaging agents for early diagnosis of cancers.
Collapse
|
12
|
Lefler JE, MarElia-Bennett CB, Thies KA, Hildreth BE, Sharma SM, Pitarresi JR, Han L, Everett C, Koivisto C, Cuitino MC, Timmers CD, O'Quinn E, Parrish M, Romeo MJ, Linke AJ, Hobbs GA, Leone G, Guttridge DC, Zimmers TA, Lesinski GB, Ostrowski MC. STAT3 in tumor fibroblasts promotes an immunosuppressive microenvironment in pancreatic cancer. Life Sci Alliance 2022; 5:e202201460. [PMID: 35803738 PMCID: PMC9270499 DOI: 10.26508/lsa.202201460] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 01/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with an incredibly dense stroma, which contributes to its recalcitrance to therapy. Cancer-associated fibroblasts (CAFs) are one of the most abundant cell types within the PDAC stroma and have context-dependent regulation of tumor progression in the tumor microenvironment (TME). Therefore, understanding tumor-promoting pathways in CAFs is essential for developing better stromal targeting therapies. Here, we show that disruption of the STAT3 signaling axis via genetic ablation of Stat3 in stromal fibroblasts in a Kras G12D PDAC mouse model not only slows tumor progression and increases survival, but re-shapes the characteristic immune-suppressive TME by decreasing M2 macrophages (F480+CD206+) and increasing CD8+ T cells. Mechanistically, we show that loss of the tumor suppressor PTEN in pancreatic CAFs leads to an increase in STAT3 phosphorylation. In addition, increased STAT3 phosphorylation in pancreatic CAFs promotes secretion of CXCL1. Inhibition of CXCL1 signaling inhibits M2 polarization in vitro. The results provide a potential mechanism by which CAFs promote an immune-suppressive TME and promote tumor progression in a spontaneous model of PDAC.
Collapse
Affiliation(s)
- Julia E Lefler
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Catherine B MarElia-Bennett
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Katie A Thies
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Blake E Hildreth
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Sudarshana M Sharma
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Jason R Pitarresi
- Division of Gastroenterology, Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Lu Han
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Caroline Everett
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Christopher Koivisto
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Maria C Cuitino
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Cynthia D Timmers
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Elizabeth O'Quinn
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Melodie Parrish
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Martin J Romeo
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda J Linke
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - G Aaron Hobbs
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Gustavo Leone
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Denis C Guttridge
- Department of Pediatrics and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Teresa A Zimmers
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Michael C Ostrowski
- Hollings Cancer Center and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
13
|
Tong S, Yu Z, Yin F, Yang Q, Chu J, Huang L, Gao W, Qian M. Manganese-based Prussian blue nanoparticles inhibit tumor proliferation and migration via the MAPK pathway in pancreatic cancer. Front Chem 2022; 10:1026924. [PMID: 36353142 PMCID: PMC9638070 DOI: 10.3389/fchem.2022.1026924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest gastrointestinal malignancies. Advances in molecular biology and surgery have significantly improved survival rates for other tumors in recent decades, but clinical outcomes for PC remained relatively unchanged. Chemodynamic therapy (CDT) and Photothermal therapy (PTT) represent an efficient and relatively safe cancer treatment modality. Here, we synthesized Mn-doped Prussian blue nanoparticles (MnPB NPs) through a simple and mild method, which have a high loading capacity for drugs and excellent CDT/PTT effect. Cell line experiments in vitro and animal experiments in vivo proved the safety of MnPB NPs. We stimulated the PC cells with MnPB NPs and performed transwell migration assays. The migration of PC cells was reduced company with the decrease of two classical proteins: matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9). Moreover, MnPB NPs induced ferroptosis, which mediated the MAPK pathway and achieved tumor elimination in nude mice. This effective and safe strategy controlled by irradiation represents a promising strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Shanshi Tong
- Department of General Surgery, Shanghai tenth People’s Hospital, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Zhilong Yu
- Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing, China
| | - Fang Yin
- Shanghai Engineering Technology Research Center for the Functional Development of Human Intestinal Flora, Shanghai tenth People’s Hospital, Shanghai, China
| | - Qilin Yang
- Department of General Surgery, Shanghai tenth People’s Hospital, Shanghai, China
| | - Juhang Chu
- School of Medicine, Tongji University, Shanghai, China
| | - Luyao Huang
- Department of General Surgery, Shanghai tenth People’s Hospital, Shanghai, China
| | - Wenxue Gao
- Clinical Research Management Office, Shanghai tenth People’s Hospital, Shanghai, China
- *Correspondence: Wenxue Gao, ; Mingping Qian,
| | - Mingping Qian
- Department of General Surgery, Shanghai tenth People’s Hospital, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Wenxue Gao, ; Mingping Qian,
| |
Collapse
|
14
|
Improvement of the reactivity of triethyl phosphate and structural behavior of hydroxyapatite versus the synthesis conditions by sol–gel route. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-021-01938-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Morás AM, Henn JG, Steffens Reinhardt L, Lenz G, Moura DJ. Recent developments in drug delivery strategies for targeting DNA damage response in glioblastoma. Life Sci 2021; 287:120128. [PMID: 34774874 DOI: 10.1016/j.lfs.2021.120128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma is the most frequent and malignant brain tumor. The median survival for this disease is approximately 15 months, and despite all the available treatment strategies employed, it remains an incurable disease. Preclinical and clinical research have shown that the resistance process related to DNA damage repair pathways, glioma stem cells, blood-brain barrier selectivity, and dose-limiting toxicity of systemic treatment leads to poor clinical outcomes. In this context, the advent of drug delivery systems associated with localized treatment seems to be a promising and versatile alternative to overcome the failure of the current treatment approaches. In order to bypass therapeutic tumor resistance mechanisms, more effective combinatorial therapies should be identified, such as the use of cytotoxic drugs combined with the inhibition of DNA damage response (DDR)-related targets. Additionally, critical reasoning about the delivery approach and administration route in brain tumors treatment innovation is essential. The outcomes of future experimental studies regarding the association of delivery systems, alternative treatment routes, and DDR targets are expected to lead to the development of refined therapeutic interventions. Novel therapeutic approaches could improve the life's quality of glioblastoma patients and increase their survival rate.
Collapse
Affiliation(s)
- A M Morás
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| | - J G Henn
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| | - L Steffens Reinhardt
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| | - G Lenz
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| | - D J Moura
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| |
Collapse
|
16
|
Ali N, Srivastava N. Recent Advancements for the Management of Pancreatic Cancer: Current Insights. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394717666210625153256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the most fatal forms of cancer includes cancer of the pancreas And the most
rapid malignancy is observed in PDAC (pancreatic ductal adenocarcinoma). The high lethality rate
is generally due to very late diagnosis and resistance to traditional chemotherapeutic agents. Desmoplastic
stromal barrier results in resistance to immunotherapy. Other reasons for the high lethality
rate include the absence of effective treatment and standard screening tests. Hence, there is a
need for effective novel carrier systems. “A formulation, method, or device that allows the desired
therapeutic substance to reach its site of action in such a manner that nontarget cells experience
minimum effect is referred to as a drug delivery system”. The delivery system is responsible for introducing
the active component into the body. They are also liable for boosting the efficacy and desirable
targeted action on the tumorous tissues. Several studies, researches, and developments have
yielded various advanced drug delivery systems, which include liposomes, nanoparticles, carbon
nanotubules, renovoCath, etc. These systems control rate and location of the release. They are designed
while taking into consideration characteristic properties of the tumor and tumor stroma. These
delivery systems overcome the barriers in drug deliverance in pancreatic cancer. Alongside providing
palliative benefits, these delivery systems also aim to correct the underlying reason for the
defect. The following review article aims and focuses to bring out a brief idea about systems, methods,
and technologies for futuristic drug deliverance in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Naureen Ali
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Lucknow Campus, Lucknow,India
| | - Nimisha Srivastava
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Lucknow Campus, Lucknow,India
| |
Collapse
|
17
|
Palzer J, Mues B, Goerg R, Aberle M, Rensen SS, Olde Damink SWM, Vaes RDW, Cramer T, Schmitz-Rode T, Neumann UP, Slabu I, Roeth AA. Magnetic Fluid Hyperthermia as Treatment Option for Pancreatic Cancer Cells and Pancreatic Cancer Organoids. Int J Nanomedicine 2021; 16:2965-2981. [PMID: 33935496 PMCID: PMC8079353 DOI: 10.2147/ijn.s288379] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/16/2021] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a cancer with a meager prognosis due to its chemotherapy resistance. A new treatment method may be magnetic fluid hyperthermia (MFH). Magnetoliposomes (ML), consisting of superparamagnetic iron oxide nanoparticles (SPION) stabilized with a phospholipid-bilayer, are exposed to an alternating magnetic field (AMF) to generate heat. To optimize this therapy, we investigated the effects of MFH on human PDAC cell lines and 3D organoid cultures. MATERIAL AND METHODS ML cytotoxicity was tested on Mia PaCa-2 and PANC-1 cells and on PDAC 3D organoid cultures, generated from resected tissue of patients. The MFH was achieved by AMF application with an amplitude of 40-47 kA/m and a frequency of 270 kHz. The MFH effect on the cell viability of the cell lines and the organoid cultures was investigated at two different time points. Clonogenic assays evaluated the impairment of colony formation. Altering ML set-ups addressed differences arising from intra- vs extracellular ML locations. RESULTS Mia PaCa-2 and PANC-1 cells showed no cytotoxic effects at ML concentrations up to 300 µg(Fe)/mL and 225 µg(Fe)/mL, respectively. ML at a concentration of 225 µg(Fe)/mL were also non-toxic for PDAC organoid cultures. MFH treatment using exclusively extracellular ML presented the highest impact on cell viability. Clonogenic assays demonstrated remarkable impairment as long-term outcome in MFH-treated PDAC cell lines. Additionally, we successfully treated PDAC organoids with extracellular ML-derived MFH, resulting in notably reduced cell viabilities 2h and 24 h post treatment. Still, PDAC organoids seem to partly recover from MFH after 24 h as opposed to conventional 2D-cultures. CONCLUSION Treatment with MFH strongly diminished pancreatic cancer cell viability in vitro, making it a promising treatment strategy. As organoids resemble the more advanced in vivo conditions better than conventional 2D cell lines, our organoid model holds great potential for further investigations.
Collapse
Affiliation(s)
- Julian Palzer
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Institute of Applied Medical Engineering, Helmholtz Institute Aachen, RWTH Aachen University Hospital, Aachen, Germany
| | - Benedikt Mues
- Institute of Applied Medical Engineering, Helmholtz Institute Aachen, RWTH Aachen University Hospital, Aachen, Germany
| | - Richard Goerg
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Institute of Applied Medical Engineering, Helmholtz Institute Aachen, RWTH Aachen University Hospital, Aachen, Germany
| | - Merel Aberle
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Sander S Rensen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Steven W M Olde Damink
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Rianne D W Vaes
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Thorsten Cramer
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Thomas Schmitz-Rode
- Institute of Applied Medical Engineering, Helmholtz Institute Aachen, RWTH Aachen University Hospital, Aachen, Germany
| | - Ulf P Neumann
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Ioana Slabu
- Institute of Applied Medical Engineering, Helmholtz Institute Aachen, RWTH Aachen University Hospital, Aachen, Germany
| | - Anjali A Roeth
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
18
|
Hu X, Xia F, Lee J, Li F, Lu X, Zhuo X, Nie G, Ling D. Tailor-Made Nanomaterials for Diagnosis and Therapy of Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002545. [PMID: 33854877 PMCID: PMC8025024 DOI: 10.1002/advs.202002545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/25/2020] [Indexed: 05/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide due to its aggressiveness and the challenge to early diagnosis and treatment. In recent decades, nanomaterials have received increasing attention for diagnosis and therapy of PDAC. However, these designs are mainly focused on the macroscopic tumor therapeutic effect, while the crucial nano-bio interactions in the heterogeneous microenvironment of PDAC remain poorly understood. As a result, the majority of potent nanomedicines show limited performance in ameliorating PDAC in clinical translation. Therefore, exploiting the unique nature of the PDAC by detecting potential biomarkers together with a deep understanding of nano-bio interactions that occur in the tumor microenvironment is pivotal to the design of PDAC-tailored effective nanomedicine. This review will introduce tailor-made nanomaterials-enabled laboratory tests and advanced noninvasive imaging technologies for early and accurate diagnosis of PDAC. Moreover, the fabrication of a myriad of tailor-made nanomaterials for various PDAC therapeutic modalities will be reviewed. Furthermore, much preferred theranostic multifunctional nanomaterials for imaging-guided therapies of PDAC will be elaborated. Lastly, the prospects of these nanomaterials in terms of clinical translation and potential breakthroughs will be briefly discussed.
Collapse
Affiliation(s)
- Xi Hu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Fan Xia
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Jiyoung Lee
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Fangyuan Li
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| | - Xiaoyang Lu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Xiaozhen Zhuo
- Department of Cardiologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyNo.11 Zhongguancun BeiyitiaoBeijing100190China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510700China
| | - Daishun Ling
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| |
Collapse
|
19
|
Yuan T, Sun J, Tian J, Hu J, Yin H, Yin J. Involvement of ABC transporters in the detoxification of non-substrate nanoparticles in lung and cervical cancer cells. Toxicology 2021; 455:152762. [PMID: 33766574 DOI: 10.1016/j.tox.2021.152762] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
This paper aimed to systemically investigate the role of adenosine triphosphate-binding cassette (ABC transporters) in the detoxification of non-substrate nanoparticles including titanium dioxide (n-TiO2, 5-10 nm) and gold (AuNPs, 3 nm, 15 nm, and 80 nm, named as Au-3, Au-15 and Au-80) in human lung cancer (A549) and human cervical cancer (HeLa) cells. All these nanoparticles were of larger hydrophilic diameters than the channel sizes of ABC transporters, thus should not be the substrates of membrane proteins. After 24-h treatment, they induced significant cytotoxicity as reflected by the reduction in cell viability and glutathione (GSH) contents, as well as the increase in reactive oxygen species (ROS) level. At median-lethal concentrations (10 mg/L n-TiO2, 2 mg/L Au-3, 5 mg/L Au-15, and 10 mg/L Au-80 for A549 cells; 20 mg/L n-TiO2, 2 mg/L Au-3, 5 mg/L Au-15, and 10 mg/L Au-80 for Hela cells), all the nanoparticles significantly induced the gene expressions and activities of ABC transporters including P-glycoprotein (PGP) and multidrug resistance associated protein 1 (MRP1). Addition of transporter inhibitors enhanced the ROS levels produced by nanoparticles, but didn't alter their death-inducing effects and intracellular accumulations. With specific suppressors, transcription factors like nuclear factor-erythroid 2-related factor-2 (NRF2) and pregnane X receptor (PXR) were proved to be important in the induction of ABC transporters by nanoparticles. After all, this paper revealed a damage-dependent modulation of ABC transporters by non-substrate nanoparticles. The up-regulated ABC transporters could help in reducing the oxidative stress produced by nanoparticles. Such information should be useful in assessing the environmental risk of nanoparticles, as well as their interactions with other chemical toxicants or drugs.
Collapse
Affiliation(s)
- Tongkuo Yuan
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Jinan Guo Ke Medical Technology Development Co., Ltd, PR China
| | - Jiaojiao Sun
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China
| | - Jingjing Tian
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Academy for Engineering & Technology, Fudan University, Shanghai 200433, PR China
| | - Jia Hu
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Huancai Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Jinan Guo Ke Medical Technology Development Co., Ltd, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| | - Jian Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Jinan Guo Ke Medical Technology Development Co., Ltd, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| |
Collapse
|
20
|
|
21
|
Radi I, Samaha R, El Hajj J, Samaha H, Kourie HR. Perioperative chemotherapy with modified FOLFIRINOX for nonmetastatic pancreatic cancer: a new standard of care? Future Oncol 2020; 17:229-233. [PMID: 33305614 DOI: 10.2217/fon-2020-0836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Imad Radi
- Hotel-Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut, 166830, Lebanon
| | - Ramy Samaha
- Hotel-Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut, 166830, Lebanon
| | - Joanna El Hajj
- Hotel-Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut, 166830, Lebanon
| | - Hady Samaha
- Hotel-Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut, 166830, Lebanon
| | - Hampig Raphael Kourie
- Hotel-Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut, 166830, Lebanon
| |
Collapse
|
22
|
Ferreira BL, Martel F, Silva C, Santos T, Daniel-da-Silva A. Nanostructured functionalized magnetic platforms for the sustained delivery of cisplatin: Synthesis, characterization and in vitro cytotoxicity evaluation. J Inorg Biochem 2020; 213:111258. [DOI: 10.1016/j.jinorgbio.2020.111258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 01/09/2023]
|
23
|
Jiang B, Zhou L, Lu J, Wang Y, Liu C, You L, Guo J. Stroma-Targeting Therapy in Pancreatic Cancer: One Coin With Two Sides? Front Oncol 2020; 10:576399. [PMID: 33178608 PMCID: PMC7593693 DOI: 10.3389/fonc.2020.576399] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignancy with one of the worst prognoses worldwide and has an overall 5-year survival rate of only 9%. Although chemotherapy is the recommended treatment for patients with advanced PDAC, its efficacy is not satisfactory. The dense dysplastic stroma of PDAC is a major obstacle to the delivery of chemotherapy drugs and plays an important role in the progression of PDAC. Therefore, stroma-targeting therapy is considered a potential treatment strategy to improve the efficacy of chemotherapy and patient survival. While several preclinical studies have shown encouraging results, the anti-tumor potential of the PDAC stroma has also been revealed, and the extreme depletion might promote tumor progression and undermine patient survival. Therefore, achieving a balance between stromal abundance and depletion might be the further of stroma-targeting therapy. This review summarized the current progress of stroma-targeting therapy in PDAC and discussed the double-edged sword of its therapeutic effects.
Collapse
Affiliation(s)
- Bolun Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yizhi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengxi Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Zhu X, Kong Q, Niu X, Chen L, Ge C. Mapping Intellectual Structure and Research Performance for the Nanoparticles in Pancreatic Cancer Field. Int J Nanomedicine 2020; 15:5503-5516. [PMID: 32801702 PMCID: PMC7415461 DOI: 10.2147/ijn.s253599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/13/2020] [Indexed: 01/15/2023] Open
Abstract
Objective To comprehensively analyze the global scientific outputs of nanoparticles in pancreatic cancer research. Methods Publications regarding the nanoparticles in pancreatic cancer research published from 1986 to 2019 were retrieved from the Web of Science Core Collection (WoSCC). Highly frequent keywords, publication years, journals, cited papers, cited journals and cited authors were identified using BICOMB software, and then a binary matrix and a co-word matrix were constructed. gCLUTO was used for double clustering of highly frequent journals. Co-citation analysis was performed using CiteSpace V software, including keywords, references, journals author or institution cooperation network. Results A total of 1171 publications were included in this study. Publications mainly came from 10 countries, led by the US (n=470) and China (n=349). Among the top 20 journals ranked by the number of citations, nanoscience nanotechnology was the leader with 300. Cluster analysis of citation network identified 12 co-citation clusters, headed by “stromal barrier” and “emerging inorganic nanomaterial”. Conclusion Our findings reveal the research performance and intellectual structure of the nanoparticles in pancreatic cancer research, which may help researchers understand the research trends and hotspots in this field.
Collapse
Affiliation(s)
- Xuan Zhu
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China.,Institute of Translational Medicine, China Medical University, Shenyang, Liaoning 110122, People's Republic of China.,Anshan Hospital, The First Affiliated Hospital of China Medical University, Anshan, Liaoning 114011, People's Republic of China
| | - Qingquan Kong
- Institute of Translational Medicine, China Medical University, Shenyang, Liaoning 110122, People's Republic of China
| | - Xing Niu
- Department of Second Clinical College, Shengjing Hospital Affiliated to China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Lijie Chen
- Department of Second Clinical College, Shengjing Hospital Affiliated to China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Chunlin Ge
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| |
Collapse
|
25
|
Brachi G, Bussolino F, Ciardelli G, Mattu C. Nanomedicine for Imaging and Therapy of Pancreatic Adenocarcinoma. Front Bioeng Biotechnol 2019; 7:307. [PMID: 31824928 PMCID: PMC6880757 DOI: 10.3389/fbioe.2019.00307] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/17/2019] [Indexed: 12/20/2022] Open
Abstract
Pancreatic adenocarcinoma has the worst outcome among all cancer types, with a 5-year survival rate as low as 10%. The lethal nature of this cancer is a result of its silent onset, resistance to therapies, and rapid spreading. As a result, most patients remain asymptomatic and present at diagnosis with an already infiltrating and incurable disease. The tumor microenvironment, composed of a dense stroma and of disorganized blood vessels, coupled with the dysfunctional signal pathways in tumor cells, creates a set of physical and biological barriers that make this tumor extremely hard-to-treat with traditional chemotherapy. Nanomedicine has great potential in pancreatic adenocarcinoma, because of the ability of nano-formulated drugs to overcome biological barriers and to enhance drug accumulation at the target site. Moreover, monitoring of disease progression can be achieved by combining drug delivery with imaging probes, resulting in early detection of metastatic patterns. This review describes the latest development of theranostic formulations designed to concomitantly treat and image pancreatic cancer, with a specific focus on their interaction with physical and biological barriers.
Collapse
Affiliation(s)
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute -IRCCS-FPO, Candiolo, Italy
| | | | | |
Collapse
|
26
|
Han Z, Shang W, Liang X, Yan H, Hu M, Peng L, Jiang H, Fang C, Wang K, Tian J. An Innovation for Treating Orthotopic Pancreatic Cancer by Preoperative Screening and Imaging-Guided Surgery. Mol Imaging Biol 2019; 21:67-77. [PMID: 29858735 DOI: 10.1007/s11307-018-1209-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Pancreatic cancer is still associated with a poor outcome and low patient quality of life, which are mainly attributed to the late detection and requirement of distal pancreatectomy with extended resection of pancreatic tumors. Therefore, novel strategies for early screening and precise tumor resection are urgently needed. In this study, we evaluated the feasibility of a low-density lipoprotein receptor (LDLR)-targeted small-molecule contrast agent (peptide-22-Cy7) for early screening with photoacoustic tomography and near-infrared (NIR) imaging as guided surgical navigation to achieve precise resection. PROCEDURE Normal pancreatic cells (HPDE6-C7) and cancer cells (PANC-1) were respectively used in the in vitro targeting evaluations. The ability of peptide-22-Cy7 for preoperative in vivo pancreatic tumor detection was investigated in a mouse orthotopic pancreatic cancer model (n = 10) using photoacoustic tomography; 18 tumor-bearing mice were further divided into three groups for different treatments. After intravenous injection of peptide-22-Cy7, surgical navigation was conducted through laparotomy. Histopathological analysis was used to further confirm the tumor area and the state of surgical margins. RESULTS Flow cytometry demonstrated that peptide-22 is highly specific to pancreatic cancer cells, with a fluorescence intensity of approximately 87.3 %. Orthotopic pancreatic tumors with a size of 4 mm could be accurately detected by photoacoustic tomography. Surgical navigation effectively achieved R0 resection and minimized the range of resection, which led to increased body weight of the mice following surgery. CONCLUSION Overall, our newly developed targeted contrast agent facilitated the accurate positioning and resection of pancreatic tumors. Photoacoustic tomography and optical imaging-guided surgical navigation may be a novel direction for improving the survival, quality of life, and disease management of pancreatic cancer patients.
Collapse
Affiliation(s)
- Ziyu Han
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.,CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, 100080, China.,Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Wenting Shang
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaoyuan Liang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.,Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Hao Yan
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Min Hu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.,Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Li Peng
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hongmei Jiang
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China. .,Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China.
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,University of Chinese Academy of Sciences, Beijing, 100080, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing, China. .,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Zhongguancun East Road #95, Haidian Dist, Beijing, 100190, China.
| |
Collapse
|
27
|
Yang F, Jin C, Fu DL, Warshaw AL. Modified FOLFIRINOX for resected pancreatic cancer: Opportunities and challenges. World J Gastroenterol 2019; 25:2839-2845. [PMID: 31249443 PMCID: PMC6589737 DOI: 10.3748/wjg.v25.i23.2839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer death worldwide. Adjuvant chemotherapy has been developed based on the experiences made with palliative chemotherapy, and advocated to improve long-term survival of patients with this disease. However, the optimal chemotherapeutic regimen remains controversial. Recently, Conroy et al demonstrated the impressive benefits of modified FOLFIRINOX over gemcitabine alone in the multicenter Partenariat de Recherche en Oncologie Digestive 24 (PRODIGE-24) trial. The remarkable results mark a new milestone in treating resectable pancreatic cancer and have now changed the standard of care for this patient population. In this commentary, we discuss an issue of difference of tumor grade between the PRODIGE-24 trial and previous phase III trials. We also discuss potential biomarkers predicting therapeutic response to modified FOLFIRINOX. Finally, we summarize several ongoing clinical trials of replacing part of the FOLFIRINOX regimen with Xeloda/S-1/nanoliposomal irinotecan for pancreatic cancer.
Collapse
Affiliation(s)
- Feng Yang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
28
|
Asghari A, Hosseini M, Khordad E, Alipour F, Marefati N, Ebrahimzadeh Bideskan A. Hippocampal apoptosis of the neonates born from TiO2 nanoparticles-exposed rats is mediated by inducible nitric oxide synthase. TOXIN REV 2019. [DOI: 10.1080/15569543.2019.1570269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Amir Asghari
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Khordad
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Alipour
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Marefati
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
29
|
Di Carlo C, Brandi J, Cecconi D. Pancreatic cancer stem cells: Perspectives on potential therapeutic approaches of pancreatic ductal adenocarcinoma. World J Stem Cells 2018; 10:172-182. [PMID: 30631392 PMCID: PMC6325076 DOI: 10.4252/wjsc.v10.i11.172] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/10/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive solid tumours of the pancreas, characterised by a five-year survival rate less than 8%. Recent reports that pancreatic cancer stem cells (PCSCs) contribute to the tumorigenesis, progression, and chemoresistance of pancreatic cancer have prompted the investigation of new therapeutic approaches able to directly target PCSCs. In the present paper the non-cancer related drugs that have been proposed to target CSCs that could potentially combat pancreatic cancer are reviewed and evaluated. The role of some pathways and deregulated proteins in PCSCs as new therapeutic targets are also discussed with a focus on selected specific inhibitors. Finally, advances in the development of nanoparticles for targeting PCSCs and site-specific drug delivery are highlighted, and their limitations considered.
Collapse
Affiliation(s)
- Claudia Di Carlo
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy
| | - Jessica Brandi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy.
| | - Daniela Cecconi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy
| |
Collapse
|
30
|
Aliebrahimi S, Kouhsari SM, Arab SS, Shadboorestan A, Ostad SN. Phytochemicals, withaferin A and carnosol, overcome pancreatic cancer stem cells as c-Met inhibitors. Biomed Pharmacother 2018; 106:1527-1536. [PMID: 30119228 DOI: 10.1016/j.biopha.2018.07.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 01/04/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) are pharmaceutically attractive targets due to their fundamental role in tumor formation. The hallmark of pancreatic cancer is its high mortality rate attributed to the existence of cancer stem cell (CSC) subpopulations which result in therapy resistance and recurrence. c-Met is a known pancreatic CSC marker that belongs to the family of RTKs. To surmount the hurdles related to ligand-independent c-Met activation, we aimed to elucidate the inhibitory mechanisms of withaferin A (WA) and carnosol (CA) as two hit phytochemicals against c-Met kinase domain. Both tested compounds attenuated HGF-mediated proliferation across various established c-Met+ cancer cell lines and altered cell cycle distribution accompanied by apoptosis induction. Scratch assay confirmed the anti-migratory activity of WA and CA in AsPC-1 cells. The blockade of HGF-driven cellular growth and motility was reflected by the suppression of c-Met phosphorylation and its downstream pro-survival pathway Akt. Further studies showed that the administration of WA and CA diminished the sphere-formation and clonogenic potential which was validated by down-regulation of pluripotency maintaining genes (oct-4 and nanog), demonstrating their potentiality to target pancreatic CSCs. As more than 60% of anti-cancer drugs are composed of natural product-derived inhibitors known as fourth generation inhibitors, our present data suggest that WA and CA may hold promise to eradicate CSCs in c-Met-dependent cancers.
Collapse
Affiliation(s)
- Shima Aliebrahimi
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Shideh Montasser Kouhsari
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Seyed Shahriar Arab
- Department of Biophysics, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Asghari A, Hosseini M, Beheshti F, Shafei MN, Mehri S. Inducible nitric oxide inhibitor aminoguanidine, ameliorated oxidative stress, interleukin-6 concentration and improved brain-derived neurotrophic factor in the brain tissues of neonates born from titanium dioxide nanoparticles exposed rats. J Matern Fetal Neonatal Med 2018; 32:3962-3973. [PMID: 29788817 DOI: 10.1080/14767058.2018.1480602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Introduction: An interaction between oxidative stress, neuroinflammation, and nitric oxide (NO) has been suggested to have a role neurotoxicity. The aim of current research was to investigate the effect of aminoguanidine (AG) as an inducible NO synthase (iNOS) inhibitor, on brain-derived neurotrophic factor (BDNF), oxidative stress, and interleukin-6 (IL-6) concentrations in the brain tissues of neonates born from the rats exposed to titanium dioxide nanoparticles (TiO2 NPs) during gestation. Methods: The pregnant rats were grouped into three and received: (1) saline, (2) TiO2 (200 mg/kg, gavage), and (3) TiO2-AG [200 mg/kg intraperitoneal (IP)]. The treatment was started since the second gestation day up to the delivery time. The neonates born from the rats were deeply anesthetized, sacrificed, and the brains were collected for biochemical evaluations. Results: The neonates born from the rats exposed to TiO2 showed a lower BDNF (p < .001) but a higher IL-6 (p < .01) concentrations in their hippocampal tissue. TiO2 exposure also increased malondialdehyde (MDA) (p < .001) and NO metabolites (p < .001), while diminished thiol (p < .001), superoxide (SOD) (p < .001), and catalase (CAT) (p < .001) in all hippocampal, cortical, and cerebellar tissues. Administration of AG improved BDNF (p < .01) but attenuated IL-6 (p < .01) concentrations in the hippocampal tissue. AG also decreased MDA (p < .001) and NO metabolites (p < .01-p < .001), while increased thiol (p < .01-p < .001), SOD (p < .001), and CAT (p < .05-p < .001) in all cerebellar, hippocampal, cortical, and tissues. Conclusion: The results of the current research revealed that iNOS inhibitor AG, ameliorated oxidative stress, IL-6 concentration, and improved BDNF in the brain tissues of neonates born from TiO2 NPs exposed rats.
Collapse
Affiliation(s)
- Amir Asghari
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Farimah Beheshti
- Department of Basic Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences , Torbat Heydariyeh , Iran
| | - Mohammad Naser Shafei
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
32
|
Zhang L, Sanagapalli S, Stoita A. Challenges in diagnosis of pancreatic cancer. World J Gastroenterol 2018; 24:2047-2060. [PMID: 29785074 PMCID: PMC5960811 DOI: 10.3748/wjg.v24.i19.2047] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/28/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a growing source of cancer related death, yet has poor survival rates which have not improved in the last few decades. Its high mortality rate is attributed to pancreatic cancer biology, difficulty in early diagnosis and the lack of standardised international guidelines in assessing suspicious pancreatic masses. This review aims to provide an update in the current state of play in pancreatic cancer diagnosis and to evaluate the benefits and limitations of available diagnostic technology. The main modalities discussed are imaging with computed tomography, magnetic resonance imaging, endoscopic ultrasound and positron emission tomography and tissue acquisition with fine needle aspiration. We also review the improvements in the techniques used for tissue acquisition and the opportunity for personalised cancer medicine. Screening of high risk individuals, promising biomarkers and common mimickers of pancreatic cancer are also explored, as well as suggestions for future research directions to allow for earlier detection of pancreatic cancer. Timely and accurate diagnosis of pancreatic cancer can lead to improvements in the current poor outcome of this disease.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Gastroenterology, St. Vincent’s Hospital Sydney, Darlinghurst 2010, NSW, Australia
| | - Santosh Sanagapalli
- Department of Gastroenterology, St. Vincent’s Hospital Sydney, Darlinghurst 2010, NSW, Australia
| | - Alina Stoita
- Department of Gastroenterology, St. Vincent’s Hospital Sydney, Darlinghurst 2010, NSW, Australia
| |
Collapse
|
33
|
Sielaff CM, Mousa SA. Status and future directions in the management of pancreatic cancer: potential impact of nanotechnology. J Cancer Res Clin Oncol 2018; 144:1205-1217. [PMID: 29721665 DOI: 10.1007/s00432-018-2651-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at a late stage, has limited treatments, and patients have poor survival rates. It currently ranks as the seventh leading cause of cancer deaths globally and has increasing rates of diagnosis. Improved PDAC treatment requires the development of innovative, effective, and economical therapeutic drugs. The late stage diagnosis limits options for surgical resection, and traditional PDAC chemotherapeutics correlate with increased organ and hematologic toxicity. In addition, PDAC tumor tissue is dense and highly resistant to many traditional chemotherapeutic applications, making the disease difficult to treat and impeding options for palliative care. New developments in nanotechnology may offer innovative options for targeted PDAC therapeutic drug delivery. Nanotechnology can be implemented using multimodality methods that offer increased opportunities for earlier diagnosis, precision enhanced imaging, targeted long-term tumor surveillance, and controlled drug delivery, as well as improved palliative care and patient comfort. Nanoscale delivery methods have demonstrated the capacity to infiltrate the dense, fibrous tumor tissue associated with PDAC, increasing delivery and effectiveness of chemotherapeutic agents and reducing toxicity through the loading of multiple drug therapies on a single nano delivery vehicle. This review presents an overview of nanoscale drug delivery systems and multimodality carriers at the forefront of new PDAC treatments.
Collapse
Affiliation(s)
- Catherine M Sielaff
- Department of Toxicology, School of Pharmacy, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA.
| |
Collapse
|
34
|
Karthik CS, Manukumar HM, Sandeep S, Sudarshan BL, Nagashree S, Mallesha L, Rakesh KP, Sanjay KR, Mallu P, Qin HL. Development of piperazine-1-carbothioamide chitosan silver nanoparticles (P1C-Tit*CAgNPs) as a promising anti-inflammatory candidate: a molecular docking validation. MEDCHEMCOMM 2018; 9:713-724. [PMID: 30108962 DOI: 10.1039/c7md00628d] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
Natural products are important leads in drug discovery. The search for effective plant-derived agents or their synthetic analogues has continued to be of interest to biologists and chemists for a long time. Herein, we have synthesized a novel compound, P1C, and P1C-Tit*CAgNPs from chitosan; P1C is a precursor and an anti-inflammatory candidate, which has been validated by molecular docking studies. The synthesized P1C-Tit*CAgNPs showed monodisperse, spherical, and cationic nature and antioxidant properties, protecting destabilization of the erythrocyte membrane by the azo compound 2,2'-azobis(2-amidinopropane)dihydrochloride (AAPH); the involvement of NPs as a protective agent for biomolecules, such as DNA and protein, followed by the treatment of NPs with AAPH was confirmed. The inhibition of cellular damage and leakage of cellular inflammatory agents was confirmed by AFM, SEM, TEM, SDS-PAGE, LDH, and PLA2 enzyme inhibition via in vitro studies. The anti-inflammatory property of P1C was further validated by in silico molecular docking studies and showed that, the P1C best pose aligned to PLA2 compared to standard drug. The significant anticancer property of P1C-Tit*CAgNPs was confirmed against MCF7, U373, and C6 cancer cell lines. Thus, the present study highlights the synthesized P1C in P1C-Tit*CAgNPs as a target PLA2-specific anti-inflammatory candidate, and further tuning of small and development-functionalized nanoparticles has a great future in medicine; hence, their clinical applications are warranted.
Collapse
Affiliation(s)
- C S Karthik
- Department of Chemistry , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India .
| | - H M Manukumar
- Department of Chemistry , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India . .,Department of Studies in Biotechnology , University of Mysore , Manasagangotri , Mysuru-570006 , Karnataka , India
| | - S Sandeep
- Department of Chemistry , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India .
| | - B L Sudarshan
- Department of Biotechnology , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India
| | - S Nagashree
- Department of Chemistry , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India .
| | - L Mallesha
- PG Department of Chemistry , JSS College of Arts, Commerce and Science , Mysuru-570 025 , Karnataka , India
| | - K P Rakesh
- Department of Pharmaceutical Engineering , School of Chemistry, Chemical Engineering and Life Science , Wuhan University of Technology , 205 Luoshi Road , Wuhan , 430073 , PR China . ; ; ; Fax: +86 27 87749300
| | - K R Sanjay
- Department of Biotechnology , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India
| | - P Mallu
- Department of Chemistry , Sri Jayachamarajendra College of Engineering , Mysuru-570 006 , Karnataka , India .
| | - Hua-Li Qin
- Department of Pharmaceutical Engineering , School of Chemistry, Chemical Engineering and Life Science , Wuhan University of Technology , 205 Luoshi Road , Wuhan , 430073 , PR China . ; ; ; Fax: +86 27 87749300
| |
Collapse
|
35
|
Pandita D, Munjal A, Godara S, Lather V. Nanocarriers in Drug and Gene Delivery. ADVANCES IN ANIMAL BIOTECHNOLOGY AND ITS APPLICATIONS 2018:71-102. [DOI: 10.1007/978-981-10-4702-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
36
|
Lv Y, Cao Y, Li P, Liu J, Chen H, Hu W, Zhang L. Ultrasound-Triggered Destruction of Folate-Functionalized Mesoporous Silica Nanoparticle-Loaded Microbubble for Targeted Tumor Therapy. Adv Healthc Mater 2017; 6. [PMID: 28671341 DOI: 10.1002/adhm.201700354] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 04/21/2017] [Indexed: 12/19/2022]
Abstract
A multifunctional drug delivery vehicle, which combines the active targeted mesoporous silica nanoparticle (MSN) and microbubble (MB) drug delivery system, is proposed and fabricated. The resulting delivery vehicle integrates the merits of high drug loading capacity, multitargeting, and ultrasound-guided releasing. Folate (FA), which serves as an active ligand, is modified to the surface of MSN (MSN-FA) to enhance cell membrane translocation. MSN-FA is loaded with tanshinone IIA (TAN), then encapsulated in a microbubble (denoted as MSN-FA-TAN-MB) for more precise tumor targeting. The conjunction between FA and MSN is confirmed by fourier transform infrared spectroscopy (FTIR). The characteristics and morphology of MSN-FA-TAN-MB are investigated by confocal microscopy and transmission electron microscopy. In vitro cytotoxicity and cellular uptake studies of MSN-FA-TAN-MB are conducted on A549 and HeLa tumor cells. FA-facilitated MSN-FA-TAN uptake is shown by HeLa cells that overexpress FA receptors via a FA-receptor-mediated endocytosis mechanism. The ultrasound response property of MSN-FA-TAN-MB is also verified. MSN-FA-TAN-MB shows significant antitumor efficacy in vivo with the assistance of FA, MB, and an external ultrasound irradiation. Thus, this multifunctional vehicle may provide a novel strategy for tumor targeting and imaging in tumor therapy.
Collapse
Affiliation(s)
- Yongjiu Lv
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology Chongqing Research Center for Pharmaceutical Engineering School of Pharmacy Chongqing Medical University Chongqing 400016 P. R. China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging Institute of Ultrasound Imaging Chongqing Medical University Chongqing 400016 P. R. China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging Institute of Ultrasound Imaging Chongqing Medical University Chongqing 400016 P. R. China
| | - Jianxin Liu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging Institute of Ultrasound Imaging Chongqing Medical University Chongqing 400016 P. R. China
| | - Huali Chen
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology Chongqing Research Center for Pharmaceutical Engineering School of Pharmacy Chongqing Medical University Chongqing 400016 P. R. China
| | - Wenjing Hu
- Chongqingshi Shapingba District People's Hospital Chongqing 400030 P. R. China
| | - Liangke Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology Chongqing Research Center for Pharmaceutical Engineering School of Pharmacy Chongqing Medical University Chongqing 400016 P. R. China
| |
Collapse
|
37
|
Zhao R, Han X, Li Y, Wang H, Ji T, Zhao Y, Nie G. Photothermal Effect Enhanced Cascade-Targeting Strategy for Improved Pancreatic Cancer Therapy by Gold Nanoshell@Mesoporous Silica Nanorod. ACS NANO 2017; 11:8103-8113. [PMID: 28738680 DOI: 10.1021/acsnano.7b02918] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Pancreatic cancer, one of the leading causes of cancer-related mortality, is characterized by desmoplasia and hypovascular cancerous tissue, with a 5 year survival rate of <8%. To overcome the severe resistance of pancreatic cancer to conventional therapies, we synthesized gold nanoshell-coated rod-like mesoporous silica (GNRS) nanoparticles which integrated cascade tumor targeting (mediated by photothermal effect and molecular receptor binding) and photothermal treatment-enhanced gemcitabine chemotherapy, under mild near-infrared laser irradiation condition. GNRS significantly improved gemcitabine penetration and accumulation in tumor tissues, thus destroying the dense stroma barrier of pancreatic cancer and reinforcing chemosensitivity in mice. Our current findings strongly support the notion that further development of this integrated plasmonic photothermal strategy may represent a promising translational nanoformulation for effective treatment of pancreatic cancer with integral cascade tumor targeting strategy and enhanced drug delivery efficacy.
Collapse
Affiliation(s)
- Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Xuexiang Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Tianjiao Ji
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School , 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, P. R. China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| |
Collapse
|
38
|
Ahmed S, Fujita S, Matsumura K. A Freeze-Concentration and Polyampholyte-Modified Liposome-Based Antigen-Delivery System for Effective Immunotherapy. Adv Healthc Mater 2017; 6. [PMID: 28493521 DOI: 10.1002/adhm.201700207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/21/2017] [Indexed: 12/13/2022]
Abstract
Immunotherapy is an exciting new approach to cancer treatment. The development of a novel freeze-concentration method is described that could be applicable in immunotherapy. The method involves freezing cells in the presence of pH-sensitive, polyampholyte-modified liposomes with encapsulated ovalbumin (OVA) as the antigen. In RAW 264.7 cells, compared to unfrozen, freeze-concentration of polyampholyte-modified liposomes encapsulating OVA resulted in efficient OVA uptake and also allowed its delivery to the cytosol. Efficient delivery of OVA to the cytosol was shown to be partly due to the pH-dependence of the polyampholyte-modified liposomes. Cytosolic OVA delivery also resulted in significant up-regulation of the major histocompatibility complex class I pathway through cross-stimulation, as well as an increase in the release of IL-1β, IL-6, and TNF-α. The results demonstrate that the combination of a simple freeze-concentration method and polyampholyte-modified liposomes might be useful in future immunotherapy applications.
Collapse
Affiliation(s)
- Sana Ahmed
- School of Materials Science; Japan Advanced Institute of Science and Technology; 1-1 Asahidai Nomi Ishikawa 923-1292 Japan
| | - Satoshi Fujita
- Department of Fiber Technology and Science; Graduate School of Engineering; University of Fukui; Fukui 910-8507 Japan
| | - Kazuaki Matsumura
- School of Materials Science; Japan Advanced Institute of Science and Technology; 1-1 Asahidai Nomi Ishikawa 923-1292 Japan
| |
Collapse
|
39
|
The relationship between SPARC expression in primary tumor and metastatic lymph node of resected pancreatic cancer patients and patients' survival. Hepatobiliary Pancreat Dis Int 2017; 16:104-109. [PMID: 28119265 DOI: 10.1016/s1499-3872(16)60168-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Previous researches in pancreatic cancer demonstrated a negative correlation between secreted protein acidic and rich in cysteine (SPARC) expression in primary tumor and survival, but not for SPARC expression in lymph node. In the present study, we aimed to evaluate the SPARC expression in various types of tissues and its impact on patients' prognosis. METHODS The expression of SPARC was examined by immunohistochemistry in resected pancreatic cancer specimens. Kaplan-Meier analyses and Cox proportional hazards regression were applied to assess the mortality risk. RESULTS A total of 222 tissue samples from 73 patients were collected to evaluate the SPARC expression, which included 73 paired primary tumor and adjacent normal tissues, 38 paired metastatic and normal lymph nodes. The proportion of positive SPARC expression in metastatic lymph node was high (32/38), whereas in normal lymph node it was negative (0/38). Positive SPARC expression in primary tumor cells was associated with a significantly decreased overall survival (P=0.007) and disease-free survival (P=0.003), whereas in other types of tissues it did not show a predictive role for prognosis. Univariate and multivariate analyses both confirmed this significance. CONCLUSION SPARC can serve a dual function role as both predictor for prognosis and potentially biomarker for lymph node metastasis in resected pancreatic cancer patients.
Collapse
|
40
|
Synthesis of Superparamagnetic Hydroxyapatite Core-Shell Nanostructure by a Rapid Sol-Gel Route. E-JOURNAL OF SURFACE SCIENCE AND NANOTECHNOLOGY 2017. [DOI: 10.1380/ejssnt.2017.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
41
|
Bayir E, Bilgi E, Urkmez AS. Implementation of Nanoparticles in Cancer Therapy. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Cancer is a wide group of diseases and generally characterized by uncontrolled proliferation of cells whose metabolic activities are disrupted. Conventionally, chemotherapy, radiotherapy, and surgery are used in the treatment of cancer. However, in theory, even a single cancer cell may trigger recurrence. Therefore, these treatments cannot provide high survival rate for deadly types. Identification of alternative methods in treatment of cancers is inevitable because of adverse effects of conventional methods. In the last few decades, nanotechnology developed by scientists working in different disciplines—physics, chemistry, and biology—offers great opportunities. It is providing elimination of both circulating tumor cells and solid cancer cells by targeting cancer cells. In this chapter, inadequate parts of conventional treatment methods, nanoparticle types used in new treatment methods of cancer, and targeting methods of nanoparticles are summarized; furthermore, recommendations of future are provided.
Collapse
|
42
|
Liu Y, Yang X, Huang Z, Huang P, Zhang Y, Deng L, Wang Z, Zhou Z, Liu Y, Kalish H, Khachab NM, Chen X, Nie Z. Magneto-Plasmonic Janus Vesicles for Magnetic Field-Enhanced Photoacoustic and Magnetic Resonance Imaging of Tumors. Angew Chem Int Ed Engl 2016; 55:15297-15300. [PMID: 27862808 PMCID: PMC5131874 DOI: 10.1002/anie.201608338] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/28/2016] [Indexed: 12/31/2022]
Abstract
Magneto-plasmonic Janus vesicles (JVs) integrated with gold nanoparticles (AuNPs) and magnetic NPs (MNPs) were prepared asymmetrically in the membrane for in vivo cancer imaging. The hybrid JVs were produced by coassembling a mixture of hydrophobic MNPs, free amphiphilic block copolymers (BCPs), and AuNPs tethered with amphiphilic BCPs. Depending on the size and content of NPs, the JVs acquired spherical or hemispherical shapes. Among them, hemispherical JVs containing 50 nm AuNPs and 15 nm MNPs showed a strong absorption in the near-infrared (NIR) window and enhanced the transverse relaxation (T2 ) contrast effect, as a result of the ordering and dense packing of AuNPs and MNPs in the membrane. The magneto-plasmonic JVs were used as drug delivery vehicles, from which the release of a payload can be triggered by NIR light and the release rate can be modulated by a magnetic field. Moreover, the JVs were applied as imaging agents for in vivo bimodal photoacoustic (PA) and magnetic resonance (MR) imaging of tumors by intravenous injection. With an external magnetic field, the accumulation of the JVs in tumors was significantly increased, leading to a signal enhancement of approximately 2-3 times in the PA and MR imaging, compared with control groups without a magnetic field.
Collapse
Affiliation(s)
- Yijing Liu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742 (USA). Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA) and Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA)
| | - Xiangyu Yang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA) and Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA)
| | - Zhiqi Huang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742 (USA)
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060 (P.R. China)
| | - Yang Zhang
- Smart Hybrid Materials (SHMs) Lab Department of Chemical Sciences and Engineering, Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900 (Kingdom of Saudi Arabia)
| | - Lin Deng
- Smart Hybrid Materials (SHMs) Lab Department of Chemical Sciences and Engineering, Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900 (Kingdom of Saudi Arabia)
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA) and Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA)
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA) and Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA)
| | - Yi Liu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742 (USA)
| | - Heather Kalish
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA) and Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA)
| | - Niveen M. Khachab
- Smart Hybrid Materials (SHMs) Lab Department of Chemical Sciences and Engineering, Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900 (Kingdom of Saudi Arabia)
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA) and Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (USA)
| | - Zhihong Nie
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742 (USA)
| |
Collapse
|
43
|
Liu Y, Yang X, Huang Z, Huang P, Zhang Y, Deng L, Wang Z, Zhou Z, Liu Y, Kalish H, Khachab NM, Chen X, Nie Z. Magneto-Plasmonic Janus Vesicles for Magnetic Field-Enhanced Photoacoustic and Magnetic Resonance Imaging of Tumors. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201608338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yijing Liu
- Department of Chemistry and Biochemistry; University of Maryland; College Park MD 20742 USA
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
| | - Xiangyu Yang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
| | - Zhiqi Huang
- Department of Chemistry and Biochemistry; University of Maryland; College Park MD 20742 USA
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering; Shenzhen University; Shenzhen 518060 P.R. China
| | - Yang Zhang
- Smart Hybrid Materials (SHMs) Lab, Department of Chemical Sciences and Engineering, Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology (KAUST); Thuwal 23955-6900 Kingdom of Saudi Arabia
| | - Lin Deng
- Smart Hybrid Materials (SHMs) Lab, Department of Chemical Sciences and Engineering, Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology (KAUST); Thuwal 23955-6900 Kingdom of Saudi Arabia
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
| | - Yi Liu
- Department of Chemistry and Biochemistry; University of Maryland; College Park MD 20742 USA
| | - Heather Kalish
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
| | - Niveen M. Khachab
- Smart Hybrid Materials (SHMs) Lab, Department of Chemical Sciences and Engineering, Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology (KAUST); Thuwal 23955-6900 Kingdom of Saudi Arabia
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB); National Institutes of Health; USA
| | - Zhihong Nie
- Department of Chemistry and Biochemistry; University of Maryland; College Park MD 20742 USA
| |
Collapse
|
44
|
He CF, Wang SH, Yu YJ, Shen HY, Zhao Y, Gao HL, Wang H, Li LL, Liu HY. Advances in biodegradable nanomaterials for photothermal therapy of cancer. Cancer Biol Med 2016; 13:299-312. [PMID: 27807498 PMCID: PMC5069834 DOI: 10.20892/j.issn.2095-3941.2016.0052] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/30/2016] [Indexed: 12/25/2022] Open
Abstract
Photothermal cancer therapy is an alternative to chemotherapy, radiotherapy, and surgery. With the development of nanophotothermal agents, this therapy holds immense potential in clinical translation. However, the toxicity issues derived from the fact that nanomaterials are trapped and retained in the reticuloendothelial systems limit their biomedical application. Developing biodegradable photothermal agents is the most practical route to address these concerns. In addition to the physicochemical properties of nanomaterials, various internal and external stimuli play key roles on nanomaterials uptake, transport, and clearance. In this review, we summarized novel nanoplatforms for photothermal therapy; these nanoplatforms can elicit stimuli-triggered degradation. We focused on the recent innovative designs endowed with biodegradable photothermal agents under different stimuli, including enzyme, pH, and near-infrared (NIR) laser.
Collapse
Affiliation(s)
- Chao-Feng He
- School of Material Science and Engineering, Henan Polytechnic University, Jiaozuo 454000, China
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shun-Hao Wang
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ying-Jie Yu
- Department of Materials Science and Engineering, State University of New York at Stony Brook, Stony Brook, NY 11790, USA
| | - He-Yun Shen
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yan Zhao
- Department of Emergency, Shandong Heze Municipal Hospital, Heze 274031, China
| | - Hui-Ling Gao
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Lin-Lin Li
- Beijing Institute of Nanoenergy and Nanosystems, National Center for Nanoscience and Technology (NCNST), Chinese Academy of Sciences, Beijing 100083, China
| | - Hui-Yu Liu
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
45
|
Dubey RD, Saneja A, Gupta PK, Gupta PN. Recent advances in drug delivery strategies for improved therapeutic efficacy of gemcitabine. Eur J Pharm Sci 2016; 93:147-62. [PMID: 27531553 DOI: 10.1016/j.ejps.2016.08.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 02/07/2023]
Abstract
Gemcitabine (2',2'-difluoro-2'-deoxycytidine; dFdC) is an efficacious anticancer agent acting against a wide range of solid tumors, including pancreatic, non-small cell lung, bladder, breast, ovarian, thyroid and multiple myelomas. However, short plasma half-life due to metabolism by cytidine deaminase necessitates administration of high dose, which limits its medical applicability. Further, due to its hydrophilic nature, it cannot traverse cell membranes by passive diffusion and, therefore, enters via nucleoside transporters that may lead to drug resistance. To circumvent these limitations, macromolecular prodrugs and nanocarrier-based formulations of Gemcitabine are gaining wide recognition. The nanoformulations based approaches by virtue of their controlled release and targeted delivery have proved to improve bioavailability, increase therapeutic efficacy and reduce adverse effects of the drug. Furthermore, the combination of Gemcitabine with other anticancer agents as well as siRNAs using nanocarriers has also been investigated in order to enhance its therapeutic potential. This review deals with challenges and recent advances in the delivery of Gemcitabine with particular emphasis on macromolecular prodrugs and nanomedicines.
Collapse
Affiliation(s)
- Ravindra Dhar Dubey
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India
| | - Ankit Saneja
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India
| | - Prasoon K Gupta
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India.
| | - Prem N Gupta
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India.
| |
Collapse
|
46
|
Hong F, Wu N, Zhao X, Tian Y, Zhou Y, Chen T, Zhai Y, Ji L. Titanium dioxide nanoparticle-induced dysfunction of cardiac hemodynamics is involved in cardiac inflammation in mice. J Biomed Mater Res A 2016; 104:2917-2927. [DOI: 10.1002/jbm.a.35831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/24/2016] [Accepted: 07/07/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Nan Wu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Xiangyu Zhao
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Yusheng Tian
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Yingjun Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Ting Chen
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Yanyu Zhai
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| | - Li Ji
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection; Huaiyin Normal University; Huaian 223300 China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake; Huaiyin Normal University; Huaian 223300 China
- School of Life Sciences; Huaiyin Normal University; Huaian 223300 China
| |
Collapse
|
47
|
Coelho SC, Almeida GM, Santos-Silva F, Pereira MC, Coelho MAN. Enhancing the efficiency of bortezomib conjugated to pegylated gold nanoparticles: an in vitro study on human pancreatic cancer cells and adenocarcinoma human lung alveolar basal epithelial cells. Expert Opin Drug Deliv 2016; 13:1075-81. [PMID: 27087021 DOI: 10.1080/17425247.2016.1178234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Gold nanoparticles have become promising vectors for cancer diagnosis and treatment. The present study investigates the effect of bortezomib (BTZ), a proteasome inhibitor, conjugated with pegylated gold nanoparticles (PEGAuNPs) in pancreatic and lung cancer cells. METHODS Synthesized gold nanoparticles (PEGAuNPs) were conjugated with bortezomib antitumor drug. We investigated the cytotoxicity induced by BTZ conjugated with functionalized gold nanoparticles in vitro, in the human pancreatic (S2-013) and lung (A549) cancer cell lines. RESULTS We found an efficient of conjugation of BTZ with PEGAuNPs. In vitro assays showed that after 72 h' incubation with PEGAuNPs-BTZ cancer cells revealed alterations in morphology; also for S2-013 and A549 cancer cells, the IC50 value of free BTZ is respectively 1.5 and 4.3 times higher than the IC50 value of PEGAuNPs-BTZ. Furthermore, for TERT-HPNE, the IC50 value is around 63 times lower for free BTZ than the conjugated nanovehicle. Cell growth inhibition results showed a remarkable enhancement in the effect of BTZ when conjugated with AuNPs. CONCLUSIONS Our findings showed that conjugation with PEGAuNPs enhance the BTZ growth-inhibition effect on human cancer cells (S2-013 and A549) and decreases its toxicity against normal cells (TERT-HPNE).
Collapse
Affiliation(s)
- Sílvia Castro Coelho
- a LEPABE, Department of Chemical Engineering, Faculty of Engineering , University of Porto , Porto , Portugal
| | - Gabriela M Almeida
- b Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c Expression Regulation in Cancer Group , IPATIMUP , Porto , Portugal
| | - Filipe Santos-Silva
- b Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,d Public Awareness of Cancer Unit , IPATIMUP , Porto , Portugal.,e Faculty of Medicine , University of Porto , Porto , Portugal.,f Department of Biochemistry and Molecular Biology, Eppley Institute , University of Nebraska Medical Center , Omaha , NE , USA
| | - Maria Carmo Pereira
- a LEPABE, Department of Chemical Engineering, Faculty of Engineering , University of Porto , Porto , Portugal
| | - Manuel A N Coelho
- a LEPABE, Department of Chemical Engineering, Faculty of Engineering , University of Porto , Porto , Portugal
| |
Collapse
|
48
|
Zhou Y, Xu M, Liu Y, Bai Y, Deng Y, Liu J, Chen L. Green synthesis of Se/Ru alloy nanoparticles using gallic acid and evaluation of theiranti-invasive effects in HeLa cells. Colloids Surf B Biointerfaces 2016; 144:118-124. [PMID: 27085043 DOI: 10.1016/j.colsurfb.2016.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/02/2016] [Accepted: 04/04/2016] [Indexed: 12/15/2022]
Abstract
Methods for the synthesis of nanoparticles (NPs) for biomedical applications ideally involve the use of nontoxic reducing and capping agents, and more importantly, enable control over the shape and size of the particles. As such, we used gallic acid (GA) as both a reducing and a capping agent in a simple and "green" synthesis of stable Se/Rualloy NPs (GA-Se/RuNPs). The diameter and morphology of the Se/Ru alloy NPs were regulated by GA concentration, and the presence of Ru was found to be a key factor in regulating and controlling the size of GA-Se/RuNPs. Moreover, GA-Se/RuNPs suppressed HeLa cell proliferation through the induction of apoptosis at concentrations that were nontoxic in normal cells. Furthermore, GA-Se/RuNPs effectively inhibited migration and invasion in HeLa cells via the inhibition of MMP-2 and MMP-9 proteins. Our findings confirm that bimetallic (Se/Ru) NPs prepared via GA-mediated synthesis exhibit enhanced anticancer effects.
Collapse
Affiliation(s)
- Yanhui Zhou
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Meng Xu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yanan Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yan Bai
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yuqian Deng
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Jie Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Lanmei Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China; School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China.
| |
Collapse
|
49
|
Deb S, Ghosh K, Shetty SD. Nanoimaging in cardiovascular diseases: Current state of the art. Indian J Med Res 2016; 141:285-98. [PMID: 25963489 PMCID: PMC4442326 DOI: 10.4103/0971-5916.156557] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nanotechnology has been integrated into healthcare system in terms of diagnosis as well as therapy. The massive impact of imaging nanotechnology has a deeper intervention in cardiology i.e. as contrast agents, to target vulnerable plaques with site specificity and in a theranostic approach to treat these plaques, stem cell delivery in necrotic myocardium, etc. Thus cardiovascular nanoimaging is not limited to simple diagnosis but also can help real time tracking during therapy as well as surgery. The present review provides a comprehensive description of the molecular imaging techniques for cardiovascular diseases with the help of nanotechnology and the potential clinical implications of nanotechnology for future applications.
Collapse
Affiliation(s)
- Suryyani Deb
- Department of Hemostasis & Thrombosis, National Institute of Immunohaematology (ICMR), Mumbai, India
| | | | | |
Collapse
|
50
|
Paul B, Bhuyan B, Purkayastha DD, Dhar SS. Photocatalytic and antibacterial activities of gold and silver nanoparticles synthesized using biomass of Parkia roxburghii leaf. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 154:1-7. [PMID: 26590801 DOI: 10.1016/j.jphotobiol.2015.11.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 12/26/2022]
Abstract
The present study reports a green approach for synthesis of gold (Au) and silver (Ag) nanoparticles (NPs) using dried biomass of Parkia roxburghii leaf. The biomass of the leaf acts as both reductant as well as stabilizer. The as-synthesized nanoparticles were characterized by time-dependent UV-visible, Fourier transform infrared (FT-IR), powder X-ray diffraction (XRD), and transmission electron microscopy (TEM) analyses. The UV-visible spectra of synthesized Au and Ag NPs showed surface plasmon resonance (SPR) at 555 and 440 nm after 12h. Powder XRD studies revealed formation of face-centered cubic structure for both Au and Ag NPs with average crystallite size of 8.4 and 14.74 nm, respectively. The TEM image showed the Au NPs to be monodispersed, spherical in shape with sizes in the range of 5-25 nm. On the other hand, Ag NPs were polydispersed, quasi-spherical in shape with sizes in the range of 5-25 nm. Investigation of photocatalytic activities of Au and Ag NPs under solar light illumination reveals that both these particles have pronounced effect on degradation of dyes viz., methylene blue (MB) and rhodamine b (RhB). Antibacterial activity of the synthesized NPs was studied on Gram positive bacteria Staphylococcus aureus and Gram negative bacteria Escherichia coli. Both Au and Ag NPs showed slightly higher activity on S. aureus than on E. coli.
Collapse
Affiliation(s)
- Bappi Paul
- Department of Chemistry, National Institute of Technology Silchar, Silchar 788010, Assam, India
| | - Bishal Bhuyan
- Department of Chemistry, National Institute of Technology Silchar, Silchar 788010, Assam, India
| | - Debraj Dhar Purkayastha
- Department of Chemistry, National Institute of Technology Silchar, Silchar 788010, Assam, India.
| | - Siddhartha Sankar Dhar
- Department of Chemistry, National Institute of Technology Silchar, Silchar 788010, Assam, India.
| |
Collapse
|