1
|
Khayat S, Choudhary K, Claude Nshimiyimana J, Gurav J, Hneini A, Nazir A, Chaito H, Wojtara M, Uwishema O. Pancreatic cancer: from early detection to personalized treatment approaches. Ann Med Surg (Lond) 2024; 86:2866-2872. [PMID: 38694319 PMCID: PMC11060269 DOI: 10.1097/ms9.0000000000002011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/19/2024] [Indexed: 05/04/2024] Open
Abstract
Pancreatic cancer is notorious for its persistently poor prognosis and health outcomes, so some of the questions that may be begged are "Why is it mostly diagnosed at end stage?", "What could we possibly do with the advancing technology in today's world to detect early pancreatic cancer and intervene?", and "Are there any implementation of the existing novel imaging technologies?". Well, to start with, this is in part because the majority of patients presented would already have reached a locally advanced or metastatic stage at the time of diagnosis due to its highly aggressive characteristics and lack of symptoms. Due to this striking disparity in survival, advancements in early detection and intervention are likely to significantly increase patients' survival. Presently, screening is frequently used in high-risk individuals in order to obtain an early pancreatic cancer diagnosis. Having a thorough understanding of the pathogenesis and risk factors of pancreatic cancer may enable us to identify individuals at high risk, diagnose the disease early, and begin treatment promptly. In this review, the authors outline the clinical hurdles to early pancreatic cancer detection, describe high-risk populations, and discuss current screening initiatives for high-risk individuals. The ultimate goal of this current review is to study the roles of both traditional and novel imaging modalities for early pancreatic cancer detection. A lot of the novel imaging techniques mentioned seem promising, but they need to be put to the test on a large scale and may need to be combined with other non-invasive biomarkers before they can be widely used.
Collapse
Affiliation(s)
| | | | | | | | - Asmaa Hneini
- Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Hassan Chaito
- Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Magda Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
2
|
Viegas C, Patrício AB, Prata J, Fonseca L, Macedo AS, Duarte SOD, Fonte P. Advances in Pancreatic Cancer Treatment by Nano-Based Drug Delivery Systems. Pharmaceutics 2023; 15:2363. [PMID: 37765331 PMCID: PMC10536303 DOI: 10.3390/pharmaceutics15092363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Pancreatic cancer represents one of the most lethal cancer types worldwide, with a 5-year survival rate of less than 5%. Due to the inability to diagnose it promptly and the lack of efficacy of existing treatments, research and development of innovative therapies and new diagnostics are crucial to increase the survival rate and decrease mortality. Nanomedicine has been gaining importance as an innovative approach for drug delivery and diagnosis, opening new horizons through the implementation of smart nanocarrier systems, which can deliver drugs to the specific tissue or organ at an optimal concentration, enhancing treatment efficacy and reducing systemic toxicity. Varied materials such as lipids, polymers, and inorganic materials have been used to obtain nanoparticles and develop innovative drug delivery systems for pancreatic cancer treatment. In this review, it is discussed the main scientific advances in pancreatic cancer treatment by nano-based drug delivery systems. The advantages and disadvantages of such delivery systems in pancreatic cancer treatment are also addressed. More importantly, the different types of nanocarriers and therapeutic strategies developed so far are scrutinized.
Collapse
Affiliation(s)
- Cláudia Viegas
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal;
- Center for Marine Sciences (CCMar), University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ana B. Patrício
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - João Prata
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Leonor Fonseca
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ana S. Macedo
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- LAQV, REQUIMTE, Applied Chemistry Lab—Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sofia O. D. Duarte
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Pedro Fonte
- Center for Marine Sciences (CCMar), University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (A.B.P.); (S.O.D.D.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
| |
Collapse
|
3
|
Jiang P, Liang B, Zhang Z, Fan B, Zeng L, Zhou Z, Mao Z, Xu Q, Yao W, Shen Q. New insights into nanosystems for non-small-cell lung cancer: diagnosis and treatment. RSC Adv 2023; 13:19540-19564. [PMID: 37388143 PMCID: PMC10300523 DOI: 10.1039/d3ra03099g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023] Open
Abstract
Lung cancer is caused by a malignant tumor that shows the fastest growth in both incidence and mortality and is also the greatest threat to human health and life. At present, both in terms of incidence and mortality, lung cancer is the first in male malignant tumors, and the second in female malignant tumors. In the past two decades, research and development of antitumor drugs worldwide have been booming, and a large number of innovative drugs have entered clinical trials and practice. In the era of precision medicine, the concept and strategy of cancer from diagnosis to treatment are experiencing unprecedented changes. The ability of tumor diagnosis and treatment has rapidly improved, the discovery rate and cure rate of early tumors have greatly improved, and the overall survival of patients has benefited significantly, with a tendency to transform to a chronic disease with tumor. The emergence of nanotechnology brings new horizons for tumor diagnosis and treatment. Nanomaterials with good biocompatibility have played an important role in tumor imaging, diagnosis, drug delivery, controlled drug release, etc. This article mainly reviews the advancements in lipid-based nanosystems, polymer-based nanosystems, and inorganic nanosystems in the diagnosis and treatment of non-small-cell lung cancer (NSCLC).
Collapse
Affiliation(s)
- Piao Jiang
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
- The First Clinical Medical College, Nanchang University Nanchang China
| | - Bin Liang
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
| | - Zhen Zhang
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College Nanchang China
| | - Bing Fan
- Department of Radiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College Nanchang China
| | - Lin Zeng
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
| | - Zhiyong Zhou
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
| | - Zhifang Mao
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
| | - Quan Xu
- Department of Thoracic Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College Nanchang China
| | - Weirong Yao
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
| | - Qinglin Shen
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College No. 152 Aiguo Road, Donghu District Nanchang 330006 China
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College Nanchang China
| |
Collapse
|
4
|
Bao J, Tu H, Li J, Dong Y, Dang L, Yurievna KE, Zhang F, Xu L. Interfacial engineered iron oxide nanoring for T2-weighted magnetic resonance imaging-guided magnetothermal-chemotherapy. Front Bioeng Biotechnol 2022; 10:1005719. [PMID: 36277375 PMCID: PMC9582775 DOI: 10.3389/fbioe.2022.1005719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Due to no penetration depth limitation, low cost, and easy control, magnetic nanoparticles mediated magnetic hyperthermia therapy (MHT) has shown great potential in experimental and clinal treatments of various diseases. However, the low heating conversion efficiencies and short circulation times are major drawback for most existing magnetic-thermal materials. Additionally, single MHT treatment always leads to resistance and recurrence. Herein, a highly efficient magnetic-thermal conversion, ferrimagnetic vortex nanoring Fe3O4 coated with hyaluronic acid (HA) nanoparticles (Fe3O4@HA, FVNH NPs) was firstly constructed. Additionally, the doxorubicin (DOX) was successfully enclosed inside the FVNH and released remotely for synergetic magnetic–thermal/chemo cancer therapy. Due to the ferrimagnetic vortex-domain state, the ring shape Fe3O4 displays a high specific absorption rate (SAR) under an external alternating magnetic field (AMF). Additionally, antitumor drug (DOX) can be encapsulated inside the single large hole of FVNH by the hyaluronic acid (HA) shell and quickly released in response the tumor acidic microenvironments and AMF. What’s more, the non-loaded FVNH NPs show good biocompatibility but high cytotoxicity after loading DOX under AMF. Furthermore, the synthesized FVNH can efficiently reduce the transverse relaxation time and enhance negative magnetic resonance imaging (MRI). The impressive in vivo systemic therapeutic efficacy of FVNH was also proved in this work. Taken together, the results of this study demonstrate that the synthesized FVNH NPs offer the promise of serving as multifunctional theranostic nanoplatforms for medical imaging-guided tumor therapies.
Collapse
Affiliation(s)
- Jianfeng Bao
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Hui Tu
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jing Li
- Office of Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yanbo Dong
- School of Education, Pingdingshan University, Pingdingshan, China
| | - Le Dang
- School of Education, Pingdingshan University, Pingdingshan, China
| | - Korjova Elena Yurievna
- Institute of Psychology, The Herzen State Pedagogical University of Russia, Saint Petersburg, Russia
| | - Fengshou Zhang
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
- *Correspondence: Fengshou Zhang, ; Lei Xu,
| | - Lei Xu
- Department of Clinical Laboratory, Huai’an Second People’s Hospital, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
- *Correspondence: Fengshou Zhang, ; Lei Xu,
| |
Collapse
|
5
|
Renal cell carcinoma management: A step to nano-chemoprevention. Life Sci 2022; 308:120922. [PMID: 36058262 DOI: 10.1016/j.lfs.2022.120922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022]
Abstract
Renal cell carcinoma (RCC) is one of the most common kidney cancers, responsible for nearly 90 % of all renal malignancies. Despite the availability of many treatment strategies, RCC still remains to be an incurable disease due to its resistivity towards conventional therapies. Nanotechnology is an emerging field of science that offers newer possibilities in therapeutics including cancer medicine, specifically by targeted delivery of anticancer drugs. Several phytochemicals are known for their anti-cancer properties and have been regarded as chemopreventive agents. However, the hydrophobic nature of many phytochemicals decreases its bioavailability and distribution, thus showing limited therapeutic effect. Application of nanotechnology to enhance chemoprevention is an effective strategy to increase the bioavailability of phytochemicals and thereby its therapeutic efficacy. The present review focuses on the utility of nanotechnology in RCC treatment and chemopreventive agents of RCC. We have also visualized the future prospects of nanomolecules in the prevention and cure of RCC.
Collapse
|
6
|
Ahmadian E, Janas D, Eftekhari A, Zare N. Application of carbon nanotubes in sensing/monitoring of pancreas and liver cancer. CHEMOSPHERE 2022; 302:134826. [PMID: 35525455 DOI: 10.1016/j.chemosphere.2022.134826] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 06/14/2023]
Abstract
Liver and pancreatic tumors are among the third leading causes of cancer-associated death worldwide. In addition to poor prognosis, both cancer types are diagnosed at advanced and metastatic stages without typical prior symptoms. Unfortunately, the existing theranostic approaches are inefficient in cancer diagnosis and treatment. Carbon nanotubes (CNTs) have attracted increasing attention in this context due to their distinct properties, including variable functionalization capability, biocompatibility, and excellent thermodynamic and optical features. As a consequence, they are now regarded as one of the most promising materials for this application. The current review aims to summarize and discuss the role of CNT in pancreatic and liver cancer theranostics. Accordingly, the breakthroughs achieved so far are classified based on the cancer type and analyzed in detail. The most feasible tactics utilizing CNT-based solutions for both cancer diagnosis and treatment are presented from the biomedical point of view. Finally, a future outlook is provided, which anticipates how the R&D community can build on the already developed methodologies and the subsequent biological responses of the pancreatic and liver cancer cells to the directed procedures.
Collapse
Affiliation(s)
- Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dawid Janas
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100, Gliwice, Poland.
| | - Aziz Eftekhari
- Department of Pharmacology & Toxicology, Tabriz University of Medical Sciences, Tabriz, Iran; Health Innovation & Acceleration Centre, Tabriz University of Medical Sciences, Tabriz, 51664, Iran; Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych, Ukraine, Baku, Azerbaijan.
| | - Najme Zare
- Department of Chemical Engineering, Quchan University of Technology, Quchan, Iran.
| |
Collapse
|
7
|
Alshememry AK, Alsaleh NB, Alkhudair N, Alzhrani R, Alshamsan A. Recent nanotechnology advancements to treat multidrug-resistance pancreatic cancer: Pre-clinical and clinical overview. Front Pharmacol 2022; 13:933457. [PMID: 36091785 PMCID: PMC9449524 DOI: 10.3389/fphar.2022.933457] [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: 04/30/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal and incurable forms of cancer and has a poor prognosis. One of the significant therapeutic challenges in PC is multidrug resistance (MDR), a phenomenon in which cancer cells develop resistance toward administered therapy. Development of novel therapeutic platforms that could overcome MDR in PC is crucial for improving therapeutic outcomes. Nanotechnology is emerging as a promising tool to enhance drug efficacy and minimize off-target responses via passive and/or active targeting mechanisms. Over the past decade, tremendous efforts have been made to utilize nanocarriers capable of targeting PC cells while minimizing off-target effects. In this review article, we first give an overview of PC and the major molecular mechanisms of MDR, and then we discuss recent advancements in the development of nanocarriers used to overcome PC drug resistance. In doing so, we explore the developmental stages of this research in both pre-clinical and clinical settings. Lastly, we discuss current challenges and gaps in the literature as well as potential future directions in the field.
Collapse
Affiliation(s)
- Abdullah K. Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nasser B. Alsaleh
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nora Alkhudair
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rami Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Aws Alshamsan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- *Correspondence: Aws Alshamsan,
| |
Collapse
|
8
|
Iyengar D, Tatiparti K, Gavande NS, Sau S, Iyer AK. Nanomedicine for overcoming therapeutic and diagnostic challenges associated with pancreatic cancer. Drug Discov Today 2022; 27:1554-1559. [PMID: 35247592 DOI: 10.1016/j.drudis.2022.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 11/29/2022]
Abstract
Pancreatic cancer is the second leading cause of cancer-related death in the USA. The 5-year survival rate for pancreatic cancer is as low as 10%, making it one of the most deadly cancers. This dismal prognosis is caused, in part, by the lack of early detection and screening options, leading to late-stage detection of the disease, at a point at which chemotherapy is no longer effective. However, nanoparticle (NP) drug delivery systems have increased the efficacy of chemotherapeutics by improving the targeting ability of drugs to the tumor site, while also decreasing the risk of local and systemic toxicity. Such efforts can contribute to the development of early diagnosis and routine screening tests, which will drastically improve the survival rates and prognosis of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Disha Iyengar
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Katyayani Tatiparti
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
9
|
Farr KP, Moses D, Haghighi KS, Phillips PA, Hillenbrand CM, Chua BH. Imaging Modalities for Early Detection of Pancreatic Cancer: Current State and Future Research Opportunities. Cancers (Basel) 2022; 14:cancers14102539. [PMID: 35626142 PMCID: PMC9139708 DOI: 10.3390/cancers14102539] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary While survival rates for many cancers have improved dramatically over the last 20 years, patients with pancreatic cancer have persistently poor outcomes. The majority of patients with pancreatic cancer are not suitable for potentially curative surgery due to locally advanced or metastatic disease stage at diagnosis. Therefore, early detection would potentially improve survival of pancreatic cancer patients through earlier intervention. Here, we present clinical challenges in the early detection of pancreatic cancer, characterise high risk groups for pancreatic cancer and current screening programs in high-risk individuals. The aim of this scoping review is to investigate the role of both established and novel imaging modalities for early detection of pancreatic cancer. Furthermore, we investigate innovative imaging techniques for early detection of pancreatic cancer, but its widespread application requires further investigation and potentially a combination with other non-invasive biomarkers. Abstract Pancreatic cancer, one of the most lethal malignancies, is increasing in incidence. While survival rates for many cancers have improved dramatically over the last 20 years, people with pancreatic cancer have persistently poor outcomes. Potential cure for pancreatic cancer involves surgical resection and adjuvant therapy. However, approximately 85% of patients diagnosed with pancreatic cancer are not suitable for potentially curative therapy due to locally advanced or metastatic disease stage. Because of this stark survival contrast, any improvement in early detection would likely significantly improve survival of patients with pancreatic cancer through earlier intervention. This comprehensive scoping review describes the current evidence on groups at high risk for developing pancreatic cancer, including individuals with inherited predisposition, pancreatic cystic lesions, diabetes, and pancreatitis. We review the current roles of imaging modalities focusing on early detection of pancreatic cancer. Additionally, we propose the use of advanced imaging modalities to identify early, potentially curable pancreatic cancer in high-risk cohorts. We discuss innovative imaging techniques for early detection of pancreatic cancer, but its widespread application requires further investigation and potentially a combination with other non-invasive biomarkers.
Collapse
Affiliation(s)
- Katherina P. Farr
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia; (K.S.H.); (B.H.C.)
- Correspondence:
| | - Daniel Moses
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia;
| | - Koroush S. Haghighi
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia; (K.S.H.); (B.H.C.)
- Department of General Surgery, Prince of Wales Hospital, Sydney, NSW 2052, Australia
| | - Phoebe A. Phillips
- Pancreatic Cancer Translational Research Group, School of Clinical Medicine, Lowy Cancer Research Centre, UNSW, Sydney, NSW 2052, Australia;
| | - Claudia M. Hillenbrand
- Research Imaging NSW, Division of Research & Enterprise, UNSW, Sydney, NSW 2052, Australia;
| | - Boon H. Chua
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia; (K.S.H.); (B.H.C.)
- Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Sydney, NSW 2052, Australia
| |
Collapse
|
10
|
Tarannum M, Vivero-Escoto JL. Nanoparticle-based therapeutic strategies targeting major clinical challenges in pancreatic cancer treatment. Adv Drug Deliv Rev 2022; 187:114357. [PMID: 35605679 DOI: 10.1016/j.addr.2022.114357] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers due to its aggressiveness and the challenges for early diagnosis and treatment. Recently, nanotechnology has demonstrated relevant strategies to overcome some of the major clinical issues in the treatment of PDAC. This review is focused on the pathological hallmarks of PDAC and the impact of nanotechnology to find solutions. It describes the use of nanoparticle-based systems designed for the delivery of chemotherapeutic agents and combinatorial alternatives that address the chemoresistance associated with PDAC, the development of combination therapies targeting the molecular heterogeneity in PDAC, the investigation of novel therapies dealing with the improvement of immunotherapy and handling the desmoplastic stroma in PDAC by remodeling the tumor microenvironment. A special section is dedicated to the design of nanoparticles for unique non-traditional modalities that could be promising in the future for the improvement in the dismal prognosis of PDAC.
Collapse
|
11
|
Wu K, Liu Y, Liu L, Peng Y, Pang H, Sun X, Xia D. Emerging Trends and Research Foci in Tumor Microenvironment of Pancreatic Cancer: A Bibliometric and Visualized Study. Front Oncol 2022; 12:810774. [PMID: 35515122 PMCID: PMC9063039 DOI: 10.3389/fonc.2022.810774] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/21/2022] [Indexed: 01/07/2023] Open
Abstract
Background Pancreatic cancer (PC) is a serious disease with high mortality. The tumor microenvironment plays a key role in the occurrence and development of PC. The purpose of this study is to analyze trends by year, country, institution, journal, reference and keyword in publications on the PC microenvironment and to predict future research hotspots. Methods The Web of Science Core Collection was used to search for publications. We analyzed the contributions of various countries/regions, institutes, and authors and identified research hotspots and promising future trends using the CiteSpace and VOSviewer programs. We also summarized relevant completed clinical trials. Results A total of 2,155 papers on the PC microenvironment published between 2011 and 2021 were included in the study. The number of publications has increased every year. The average number of citations per article was 32.69. The USA had the most publications, followed by China, and a total of 50 influential articles were identified through co-citation analysis. Clustering analysis revealed two clusters of keywords: basic research and clinical application. The co-occurrence cluster analysis showed glutamine metabolism, carcinoma-associated fibroblasts, oxidative phosphorylation as the highly concerned research topics of basic research in recently. The three latest hot topics in clinical application are liposomes, endoscopic ultrasound and photodynamic therapy. Conclusion The number of publications and research interest have generally increased, and the USA has made prominent contributions to the study of the tumor microenvironment of PC. The current research hotspots mainly focus on energy metabolism in the hypoxic tumor microenvironment, cancer associated fibroblasts in regulating the tumor microenvironment, accurate diagnosis, drug delivery and new treatments.
Collapse
Affiliation(s)
- Kaiwen Wu
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China.,Southwest Jiaotong University College of Medicine, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Ye Liu
- Naval Medical University, Shanghai, China
| | - Lei Liu
- Medical Research Center, Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yunlan Peng
- Southwest Jiaotong University College of Medicine, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Honglin Pang
- Southwest Jiaotong University College of Medicine, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Xiaobin Sun
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Demeng Xia
- Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| |
Collapse
|
12
|
Nanomedicine in Pancreatic Cancer: Current Status and Future Opportunities for Overcoming Therapy Resistance. Cancers (Basel) 2021; 13:cancers13246175. [PMID: 34944794 PMCID: PMC8699181 DOI: 10.3390/cancers13246175] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Despite access to a vast arsenal of anticancer agents, many fail to realise their full therapeutic potential in clinical practice. One key determinant of this is the evolution of multifaceted resistance mechanisms within the tumour that may either pre-exist or develop during the course of therapy. This is particularly evident in pancreatic cancer, where limited responses to treatment underlie dismal survival rates, highlighting the urgent need for new therapeutic approaches. Here, we discuss the major features of pancreatic tumours that contribute to therapy resistance, and how they may be alleviated through exploitation of the mounting and exciting promise of nanomedicines; a unique collection of nanoscale platforms with tunable and multifunctional capabilities that have already elicited a widespread impact on cancer management. Abstract The development of drug resistance remains one of the greatest clinical oncology challenges that can radically dampen the prospect of achieving complete and durable tumour control. Efforts to mitigate drug resistance are therefore of utmost importance, and nanotechnology is rapidly emerging for its potential to overcome such issues. Studies have showcased the ability of nanomedicines to bypass drug efflux pumps, counteract immune suppression, serve as radioenhancers, correct metabolic disturbances and elicit numerous other effects that collectively alleviate various mechanisms of tumour resistance. Much of this progress can be attributed to the remarkable benefits that nanoparticles offer as drug delivery vehicles, such as improvements in pharmacokinetics, protection against degradation and spatiotemporally controlled release kinetics. These attributes provide scope for precision targeting of drugs to tumours that can enhance sensitivity to treatment and have formed the basis for the successful clinical translation of multiple nanoformulations to date. In this review, we focus on the longstanding reputation of pancreatic cancer as one of the most difficult-to-treat malignancies where resistance plays a dominant role in therapy failure. We outline the mechanisms that contribute to the treatment-refractory nature of these tumours, and how they may be effectively addressed by harnessing the unique capabilities of nanomedicines. Moreover, we include a brief perspective on the likely future direction of nanotechnology in pancreatic cancer, discussing how efforts to develop multidrug formulations will guide the field further towards a therapeutic solution for these highly intractable tumours.
Collapse
|
13
|
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
|
14
|
Biomarkers in Pancreatic Cancer as Analytic Targets for Nanomediated Imaging and Therapy. MATERIALS 2021; 14:ma14113083. [PMID: 34199998 PMCID: PMC8200189 DOI: 10.3390/ma14113083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022]
Abstract
As the increase in therapeutic and imaging technologies is swiftly improving survival chances for cancer patients, pancreatic cancer (PC) still has a grim prognosis and a rising incidence. Practically everything distinguishing for this type of malignancy makes it challenging to treat: no approved method for early detection, extended asymptomatic state, limited treatment options, poor chemotherapy response and dense tumor stroma that impedes drug delivery. We provide a narrative review of our main findings in the field of nanoparticle directed treatment for PC, with a focus on biomarker targeted delivery. By reducing drug toxicity, increasing their tumor accumulation, ability to modulate tumor microenvironment and even improve imaging contrast, it seems that nanotechnology may one day give hope for better outcome in pancreatic cancer. Further conjugating nanoparticles with biomarkers that are overexpressed amplifies the benefits mentioned, with potential increase in survival and treatment response.
Collapse
|
15
|
Regulation of tumor microenvironment for pancreatic cancer therapy. Biomaterials 2021; 270:120680. [PMID: 33588140 DOI: 10.1016/j.biomaterials.2021.120680] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 02/05/2023]
Abstract
Pancreatic cancer (PC) is one kind of the most lethal malignancies worldwide, owing to its insidious symptoms, early metastases, and negative responses to current therapies. With an increasing understanding of pathology, the tumor microenvironment (TME) plays a significant role in ineffective treatment and poor prognosis of PC. Thus, a growing number of studies have focused on whether components of the TME could be effective targets for PC therapy. Biomaterials have been widely applied in cancer therapy, and numerous organic or inorganic biomaterials for TME regulation have been developed to inhibit the growth and metastasis of PC, as well as reverse therapeutic resistance. In this review, we discuss various biomaterials utilized to treat PC based on different components of the TME, including, but not limited to, extracellular matrix (ECM), abnormal tumor vascularization, and tumor-associated immune cells, as well as other unconventional therapeutic strategies. Besides, the perspectives on the underlying future of theranostic nanomedicines for PC therapy are also presented.
Collapse
|
16
|
99mTc-citrate-gold nanoparticles as a tumor tracer: synthesis, characterization, radiolabeling and in-vivo studies. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2019-3208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Targeted drug delivery system can reduce the side effects of high drug concentration by improving drug pharmacokinetics at lower doses. Citrate-gold nanoparticles (AuNPs) as a drug delivery system were synthesized via green nanotechnology technique to be used as a new imaging platform for tumor targeting. Citrate-AuNPs were synthesized with core size of 10 nm. Citrate-AuNPs were labeled with technetium-99m (99mTc) with radiochemical yield of 95.20 ± 2.70% with good in-vitro stability in both saline and human serum and well in-vivo studied in both normal and solid tumor bearing mice. The in-vivo biodistribution study of [99mTc]Tc-citrate-AuNPs in solid tumor bearing mice (as preliminary study) showed a high accumulation in tumor site with tumor/muscle of 4.35 ± 0.22 after 30 min post injection. The direct intratumoral (I.T) injection of [99mTc]Tc-citrate-AuNPs showed that this complex was retained in the tumor up to 77.86 ± 1.90 % at 5 min and still around 50.00 ± 1.42 % after 30 min post injection (p.i.). The newly presented nano-platform could be presented as a new potential radiopharmaceutical tumor imaging probe.
Collapse
|
17
|
Kumar A, Chaudhary RK, Singh R, Singh SP, Wang SY, Hoe ZY, Pan CT, Shiue YL, Wei DQ, Kaushik AC, Dai X. Nanotheranostic Applications for Detection and Targeting Neurodegenerative Diseases. Front Neurosci 2020; 14:305. [PMID: 32425743 PMCID: PMC7203731 DOI: 10.3389/fnins.2020.00305] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 03/16/2020] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology utilizes engineered materials and devices which function with biological systems at the molecular level and could transform the management of neurodegenerative diseases (NDs) by provoking, reacting to, and intermingling with target sites to stimulate physiological responses while minimizing side effects. Blood-brain barrier (BBB) protects the brain from harmful agents, and transporting drugs across the BBB is a major challenge for diagnosis, targeting, and treatment of NDs. The BBB provides severe limitations for diagnosis and treatment of Alzheimer's disease (AD), Parkinson's disease (PD), and various other neurological diseases. Conventional drug delivery systems generally fail to cross the BBB, thus are inefficient in treatment. Although gradual development through research is ensuring the progress of nanotheranostic approaches from animal to human modeling, aspects of translational applicability and safety are a key concern. This demands a deep understanding of the interaction of body systems with nanomaterials. There are various plant-based nanobioactive compounds which are reported to have applicability in the diagnosis and treatment of these NDs. This review article provides an overview of applications of nanotheranostics in AD and PD. The review also discusses nano-enabled drug delivery systems and their current and potential applications for the treatment of various NDs.
Collapse
Affiliation(s)
- Ajay Kumar
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ravi Kumar Chaudhary
- Department of Biotechnology, Institute of Applied Medicines & Research, Ghaziabad, India
| | - Rachita Singh
- Department of Electrical and Electronics Engineering, IIMT Engineering College, Uttar Pradesh Technical University, Meerut, India
| | - Satya P. Singh
- School of Computer Science & Engineering, Nanyang Technological University, Singapore, Singapore
| | - Shao-Yu Wang
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Zheng-Yu Hoe
- Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Tang Pan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Dong-Qing Wei
- Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aman Chandra Kaushik
- Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
18
|
Kokkinos J, Ignacio RMC, Sharbeen G, Boyer C, Gonzales-Aloy E, Goldstein D, Australian Pancreatic Cancer Genome Initiative Apgi, McCarroll JA, Phillips PA. Targeting the undruggable in pancreatic cancer using nano-based gene silencing drugs. Biomaterials 2020; 240:119742. [PMID: 32088410 DOI: 10.1016/j.biomaterials.2019.119742] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/03/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer is predicted to be the second leading cause of cancer-related death by 2025. The best chemotherapy only extends survival by an average of 18 weeks. The extensive fibrotic stroma surrounding the tumor curbs therapeutic options as chemotherapy drugs cannot freely penetrate the tumor. RNA interference (RNAi) has emerged as a promising approach to revolutionize cancer treatment. Small interfering RNA (siRNA) can be designed to inhibit the expression of any gene which is important given the high degree of genetic heterogeneity present in pancreatic tumors. Despite the potential of siRNA therapies, there are hurdles limiting their clinical application such as poor transport across biological barriers, limited cellular uptake, degradation, and rapid clearance. Nanotechnology can address these challenges. In fact, the past few decades have seen the conceptualization, design, pre-clinical testing and recent clinical approval of a RNAi nanodrug to treat disease. In this review, we comment on the current state of play of clinical trials evaluating siRNA nanodrugs and review pre-clinical studies investigating the efficacy of siRNA therapeutics in pancreatic cancer. We assess the physiological barriers unique to pancreatic cancer that need to be considered when designing and testing new nanomedicines for this disease.
Collapse
Affiliation(s)
- John Kokkinos
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia; Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia
| | - Rosa Mistica C Ignacio
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - Cyrille Boyer
- Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia; Centre for Advanced Macromolecular Design, School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Estrella Gonzales-Aloy
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia; Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, NSW, 2052, Australia
| | | | - Joshua A McCarroll
- Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia; Tumour Biology & Targeting Program, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia, 2031; School of Women's and Children's Health, Faculty of Medicine, UNSW, Sydney, NSW, 2052, Australia.
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia; Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia.
| |
Collapse
|
19
|
Santos-Rebelo A, Kumar P, Pillay V, Choonara YE, Eleutério C, Figueira M, Viana AS, Ascensão L, Molpeceres J, Rijo P, Correia I, Amaral J, Solá S, Rodrigues CMP, Gaspar MM, Reis CP. Development and Mechanistic Insight into the Enhanced Cytotoxic Potential of Parvifloron D Albumin Nanoparticles in EGFR-Overexpressing Pancreatic Cancer Cells. Cancers (Basel) 2019; 11:cancers11111733. [PMID: 31694306 PMCID: PMC6895893 DOI: 10.3390/cancers11111733] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 01/04/2023] Open
Abstract
Pancreatic cancer is one of the most lethal cancers, with an extremely poor prognosis. The development of more effective therapies is thus imperative. Natural origin compounds isolated from Plectranthus genus, such as parvifloron D (PvD), have cytotoxic and antiproliferative activity against human tumour cells. However, PvD is a very low water-soluble compound, being nanotechnology a promising alternative strategy to solve this problem. Therefore, the aim of this study was to optimize a nanosystem for preferential delivery of PvD to pancreatic tumour cells. Albumin nanoparticles (BSA NPs) were produced through a desolvation method. Glucose cross-linking and bioactive functionalization profiles of BSA platform were elucidated and analysed using static lattice atomistic simulations in vacuum. Using the optimized methodology, PvD was encapsulated (yield higher than 80%) while NPs were characterized in terms of size (100–400 nm) and morphology. Importantly, to achieve a preferential targeting to pancreatic cancer cells, erlotinib and cetuximab were attached to the PvD-loaded nanoparticle surface, and their antiproliferative effects were evaluated in BxPC3 and Panc-1 cell lines. Erlotinib conjugated NPs presented the highest antiproliferative effect toward pancreatic tumour cells. Accordingly, cell cycle analysis of the BxPC3 cell line showed marked accumulation of tumour cells in G1-phase and cell cycle arrest promoted by NPs. As a result, erlotinib conjugated PvD-loaded BSA NPs must be considered a suitable and promising carrier to deliver PvD at the tumour site, improving the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ana Santos-Rebelo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal; (A.S.-R.); (P.R.)
- Department of Biomedical Sciences, Faculty of Pharmacy, University of Alcalá, Ctra. A2 km 33,600 Campus Universitario, 28871 Alcalá de Henares, Spain;
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, School of Therapeutics Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; (P.K.); (V.P.); (Y.E.C.)
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, School of Therapeutics Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; (P.K.); (V.P.); (Y.E.C.)
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, School of Therapeutics Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; (P.K.); (V.P.); (Y.E.C.)
| | - Carla Eleutério
- Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (C.E.); (M.F.)
| | - Mariana Figueira
- Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (C.E.); (M.F.)
| | - Ana S. Viana
- CQB, CQE, Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016 Lisboa, Portugal;
| | - Lia Ascensão
- CESAM, Universidade de Lisboa, Faculdade de Ciências, Campo Grande 1749-016 Lisboa, Portugal;
| | - Jesús Molpeceres
- Department of Biomedical Sciences, Faculty of Pharmacy, University of Alcalá, Ctra. A2 km 33,600 Campus Universitario, 28871 Alcalá de Henares, Spain;
| | - Patrícia Rijo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal; (A.S.-R.); (P.R.)
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.A.); (S.S.); (C.M.P.R.); (M.M.G.)
| | - Isabel Correia
- Centro de Química Estrutural, Instituto Superior Técnico, Departamento de Engenharia Química, Universidade de Lisboa,1049-001 Lisboa, Portugal;
| | - Joana Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.A.); (S.S.); (C.M.P.R.); (M.M.G.)
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.A.); (S.S.); (C.M.P.R.); (M.M.G.)
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.A.); (S.S.); (C.M.P.R.); (M.M.G.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.A.); (S.S.); (C.M.P.R.); (M.M.G.)
| | - Catarina Pinto Reis
- Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (C.E.); (M.F.)
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.A.); (S.S.); (C.M.P.R.); (M.M.G.)
- IBEB, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Correspondence: ; Tel.: +351-217-946-400; Fax: +351-217-946-470
| |
Collapse
|
20
|
Bidve P, Prajapati N, Kalia K, Tekade R, Tiwari V. Emerging role of nanomedicine in the treatment of neuropathic pain. J Drug Target 2019; 28:11-22. [PMID: 30798636 DOI: 10.1080/1061186x.2019.1587444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neuropathic pain (NeP) is a complex chronic pain condition associated with nerve injury. Approximately, 7-10% of the general population across the globe is suffering from this traumatic condition, but the existing treatment strategies are inadequate to deliver pain relief and are associated with severe adverse effects. To overcome these limitations, lot of research is focussed on developing new molecules with high potency and fewer side effects, novel cell and gene-based therapies and modification of the previously approved drugs by different formulation aspects. Nanomedicine has attracted a lot of attention in the treatment of many diverse pathological conditions because of their unique physiochemical and biological properties. In this manuscript, we highlighted the emerging role of nanomedicine in different therapies (drug, cell and gene), also we emphasised on the challenges associated with nanomedicine such as development of well-characterised nanoformulation, scaling of batches with reproducible results and toxicity along with this we discussed about the future of nanomedicine in the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Pankaj Bidve
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Namrata Prajapati
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Rakesh Tekade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Vinod Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| |
Collapse
|
21
|
McCarroll JA, Sharbeen G, Kavallaris M, Phillips PA. The Use of Star Polymer Nanoparticles for the Delivery of siRNA to Mouse Orthotopic Pancreatic Tumor Models. Methods Mol Biol 2019; 1974:329-353. [PMID: 31099013 DOI: 10.1007/978-1-4939-9220-1_23] [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: 12/23/2022]
Abstract
Pancreatic cancer is a lethal malignancy which is refractory to most chemotherapy drugs. Recent landmark studies have shed new light on the complex genetic heterogeneity of pancreatic cancer and provide an opportunity to utilize "precision-based medicines" to target genes based on the genetic profile of an individual's tumor to increase the efficiency of chemotherapy and decrease tumor growth and metastases. Gene-silencing drugs in the form of short-interfering RNA (siRNA) have the potential to play an important role in precision medicine for pancreatic cancer by silencing the expression of genes including those considered difficult to inhibit (undruggable) using chemical agents. However, before siRNA can reach its clinical potential a delivery vehicle is needed to carry siRNA across the cell membrane and into the cytoplasm of the cell. Herein, we detail the methods required to use star polymer nanoparticles to deliver siRNA to pancreatic tumors in an orthotopic pancreatic cancer mouse model to silence the expression of an "undruggable" gene (βIII-tubulin) that regulates pancreatic cancer growth and chemosensitivity.
Collapse
Affiliation(s)
- Joshua A McCarroll
- Tumour Biology and Targeting Program, Lowy Cancer Research Centre, Children's Cancer Institute, UNSW Sydney, Sydney, NSW, Australia.,Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Maria Kavallaris
- Tumour Biology and Targeting Program, Lowy Cancer Research Centre, Children's Cancer Institute, UNSW Sydney, Sydney, NSW, Australia.,Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia
| | - Phoebe A Phillips
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia. .,Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
22
|
Li BY, He LJ, Zhang XL, Liu H, Liu B. High expression of RAB38 promotes malignant progression of pancreatic cancer. Mol Med Rep 2018; 19:909-918. [PMID: 30569114 PMCID: PMC6323198 DOI: 10.3892/mmr.2018.9732] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
Ras-Related Protein Rab-38 (RAB38), which belongs to the RAB family, is involved in the biogenesis of lysosome-related organelles and defense against certain microbial infections. However, the clinical significance and potential function of RAB38 in pancreatic adenocarcinoma remain unclear. In the present study, an immunohistochemical assay was performed to analyze the expression of RAB38 in pancreatic adenocarcinoma tumor specimens from 82 patients, and the clinicopathological characteristics and survival rate of these patients were further examined. To validate the role of RAB38 in tumors, the effect of RAB38 on tumor cell proliferation, migration and invasion was assessed by establishing RAB38 knockdown cell lines. Reverse transcription-quantitative polymerase chain reaction and western blotting were used to examine the expression levels of proteins associated with the cancer cell behavior. In addition, the inhibitory effect of RAB38 silencing on pancreatic cancer was examined in mice. The immunohistochemistry results revealed that RAB38 was upregulated and positively correlated with the grade of progression in pancreatic adenocarcinoma patients. Further investigation indicated that RAB38 downregulation significantly suppressed the proliferation, migration and invasive capacity of pancreatic cancer cells, as well as decreased the expression levels of Ki67, proliferating cell nuclear antigen, and matrix metalloproteinases 2 and 9. RAB38 silencing also inhibited the development of pancreatic cancer in vivo. Taken together, a high level of RAB38 was significantly associated with the malignant phenotypes of pancreatic cancer, suggesting that RAB38 may serve as a novel biomarker and a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Bao-Yu Li
- Department of General Surgery, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Li-Jie He
- Department of General Surgery, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xiang-Lian Zhang
- Department of General Surgery, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Hui Liu
- Department of General Surgery, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Bin Liu
- Department of General Surgery, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
23
|
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
|
24
|
Vittorio O, Le Grand M, Makharza SA, Curcio M, Tucci P, Iemma F, Nicoletta FP, Hampel S, Cirillo G. Doxorubicin synergism and resistance reversal in human neuroblastoma BE(2)C cell lines: An in vitro study with dextran-catechin nanohybrids. Eur J Pharm Biopharm 2017; 122:176-185. [PMID: 29129733 DOI: 10.1016/j.ejpb.2017.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 11/28/2022]
Abstract
Hybrid nanocarrier consisting in nanographene oxide coated by a dextran-catechin conjugate was proposed in the efforts to find more efficient Neuroblastoma treatment with Doxorubicin chemotherapy. The dextran-catechin conjugate was prepared by immobilized laccase catalysis and its peculiar reducing ability exploited for the synthesis of the hybrid carrier. Raman spectra and DSC thermograms were recorded to check the physicochemical properties of the nanohybrid, while DLS measurements, SEM, TEM, and AFM microscopy allowed the determination of its morphological and dimensional features. A pH dependent Doxorubicin release was observed, with 30 and 75% doxorubicin released at pH 7.4 and 5.0, respectively. Viability assays on parental BE(2)C and resistant BE(2)C/ADR cell lines proved that the high anticancer activity of dextran-catechin conjugate (IC50 19.9 ± 0.6 and 18.4 ± 0.7 µg mL-1) was retained upon formation of the nanohybrids (IC50 24.8 ± 0.7 and 22.9 ± 1 µg mL-1). Combination therapy showed a synergistic activity between doxorubicin and either bioconjugate or nanocarrier on BE(2)C. More interestingly, on BE(2)C/ADR we recorded both the reversion of doxorubicin resistance mechanism as a consequence of decreased P-gp expression (Western Blot analysis) and a synergistic effect on cell viability, confirming the proposed nanohybrid as a very promising starting point for further research in neuroblastoma treatment.
Collapse
Affiliation(s)
- Orazio Vittorio
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia; Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, NSW, Sydney, Australia
| | - Marion Le Grand
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia; Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, NSW, Sydney, Australia
| | - Sami A Makharza
- College of Pharmacy and Medical Sciences, Hebron University, Hebron, Palestine
| | - Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy
| | - Paola Tucci
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy
| | - Silke Hampel
- Leibniz Institute of Solid State and Material Research Dresden, 01171 Dresden, Germany
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy.
| |
Collapse
|
25
|
Matta-Domjan B, King A, Totti S, Matta C, Dover G, Martinez P, Zakhidov A, La Ragione R, Macedo H, Jurewicz I, Dalton A, Velliou EG. Biophysical interactions between pancreatic cancer cells and pristine carbon nanotube substrates: Potential application for pancreatic cancer tissue engineering. J Biomed Mater Res B Appl Biomater 2017; 106:1637-1644. [PMID: 28976640 DOI: 10.1002/jbm.b.34012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/23/2017] [Accepted: 09/17/2017] [Indexed: 12/16/2022]
Abstract
Novel synthetic biomaterials able to support direct tissue growth and retain cellular phenotypical properties are promising building blocks for the development of tissue engineering platforms for accurate and fast therapy screening for cancer. The aim of this study is to validate an aligned, pristine multi-walled carbon nanotube (CNT) platform for in vitro studies of pancreatic cancer as a systematic understanding of interactions between cells and these CNT substrates is lacking. Our results demonstrate that our CNT scaffolds-which are easily tuneable to form sheets/fibers-support growth, proliferation, and spatial organization of pancreatic cancer cells, indicating their great potential in cancer tissue engineering. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1637-1644, 2018.
Collapse
Affiliation(s)
- Brigitta Matta-Domjan
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, Faculty of Engineering & Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Alice King
- Department of Physics and Astronomy, School of Mathematical and Physical Sciences, University of Sussex, Brighton, BN1 9QH, UK
| | - Stella Totti
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, Faculty of Engineering & Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Csaba Matta
- Department of Veterinary Preclinical Sciences, School of Veterinary Science and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, UK.,Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - George Dover
- Department of Physics, Faculty of Engineering & Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Patricia Martinez
- NanoTech Institute, University of Texas at Dallas, Richardson, Texas, 75083-068875080
| | - Anvar Zakhidov
- NanoTech Institute, University of Texas at Dallas, Richardson, Texas, 75083-068875080.,National University of Science and Technology, MISIS, Moscow, 119049, Russia.,Laboratory of Hybrid Nanophotonics and Optoelectronics, Department of Physics and Technology, ITMO University, St. Petersburg, 197101, Russia
| | - Roberto La Ragione
- Department of Pathology and Infectious Diseases, School of Veterinary Science and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, UK
| | | | - Izabela Jurewicz
- Department of Physics, Faculty of Engineering & Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Alan Dalton
- Department of Physics and Astronomy, School of Mathematical and Physical Sciences, University of Sussex, Brighton, BN1 9QH, UK
| | - Eirini G Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, Faculty of Engineering & Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| |
Collapse
|
26
|
Montiel Schneider MG, Lassalle VL. Magnetic iron oxide nanoparticles as novel and efficient tools for atherosclerosis diagnosis. Biomed Pharmacother 2017; 93:1098-1115. [PMID: 28738519 DOI: 10.1016/j.biopha.2017.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/14/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular complications derivate from atherosclerosis are the main cause of death in western world. An early detection of vulnerable atherosclerotic plaques is primordial for a better care of patients suffering the pathology. In this context nanotechnology has emerged as a promising tool to achieve this goal. Nanoparticles based on magnetic iron oxide (MNPs) have been extensively studied in cardiovascular diseases diagnosis, as well as in the treatment and diagnostic of other pathologies. The present review aims to describe and analyze the most current literature regarding to this topic, offering the level of detail required to reproduce the experimental tasks providing a critical input of the latest available reports. The current diagnostic features are presented and compared, highlighting their advantages and disadvantages. Information on novel technology intended to this purpose is also recompiled and in deep analyzed. Special emphasis is placed in magnetic nanotechnology, remarking the possibility to assess selective and multifunctional systems to the early detection of artherosclerotic pathologies. Finally, in view of the state of the art, the future perspectives about the trends on MNPs in artherosclerorsis diagnostic and treatment have also been addressed.
Collapse
Affiliation(s)
| | - Verónica Leticia Lassalle
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS)-CONICET, Av. Alem 1253, 8000 Bahía Blanca, Argentina.
| |
Collapse
|
27
|
Khan S, Ansari AA, Rolfo C, Coelho A, Abdulla M, Al-Khayal K, Ahmad R. Evaluation of in vitro cytotoxicity, biocompatibility, and changes in the expression of apoptosis regulatory proteins induced by cerium oxide nanocrystals. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2017; 18:364-373. [PMID: 28634498 PMCID: PMC5468938 DOI: 10.1080/14686996.2017.1319731] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 04/12/2017] [Accepted: 04/12/2017] [Indexed: 06/09/2023]
Abstract
Cerium oxide nanocrystals (CeO2-NCs) exhibit superoxide dismutase and catalase mimetic activities. Based on these catalytic activities, CeO2-NCs have been suggested to have the potential to treat various diseases. The crystalline size of these materials is an important factor that influences the performance of CeO2-NCs. Previous reports have shown that several metal-based nanocrystals, including CeO2-NCs, can induce cytotoxicity in cancer cells. However, the underlying mechanisms have remained unclear. To characterize the anticancer activities of CeO2-NCs, several assays related to the mechanism of cytotoxicity and induction of apoptosis has been performed. Here, we have carried out a systematic study to characterize CeO2-NCs phase purity (X-ray diffraction), morphology (electron microscopy), and optical features (optical absorption, Raman scattering, and photoluminescence) to better establish their potential as anticancer drugs. Our study revealed anticancer effects of CeO2-NCs in HT29 and SW620 colorectal cancer cell lines with half-maximal inhibitory concentration (IC50) values of 2.26 and 121.18 μg ml-1, respectively. Reductions in cell viability indicated the cytotoxic potential of CeO2-NCs in HT29 cells based on inverted and florescence microscopy assessments. The mechanism of cytotoxicity confirmed by estimating possible changes in the expression levels of Bcl2, BclxL, Bax, PARP, cytochrome c, and β-actin (control) proteins in HT29 cells. Down-regulation of Bcl2 and BclxL and up-regulation of Bax, PARP, and cytochrome c proteins suggested the significant involvement of CeO2-NCs exposure in the induction of apoptosis. Furthermore, biocompatibility assay showed minimum effect of CeO2-NCs on human red blood cells.
Collapse
Affiliation(s)
- Shahanavaj Khan
- Nanomedicine & Biotechnology Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Bioscience, Shri Ram Group of College (SRGC), Muzaffarnagar, India
| | - Anees A. Ansari
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Christian Rolfo
- Phase I- Early Clinical Trials Unit, Oncology Department and Multidisciplinary Oncology Center Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Andreia Coelho
- Phase I- Early Clinical Trials Unit, Oncology Department and Multidisciplinary Oncology Center Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Maha Abdulla
- Colorectal Research Center, College of Medicine King Saud University, Riyadh, Saudi Arabia
| | - Khayal Al-Khayal
- Colorectal Research Center, College of Medicine King Saud University, Riyadh, Saudi Arabia
| | - Rehan Ahmad
- Colorectal Research Center, College of Medicine King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Sharbeen G, Youkhana J, Mawson A, McCarroll J, Nunez A, Biankin A, Johns A, Goldstein D, Phillips P. MutY-Homolog (MYH) inhibition reduces pancreatic cancer cell growth and increases chemosensitivity. Oncotarget 2017; 8:9216-9229. [PMID: 27999205 PMCID: PMC5354726 DOI: 10.18632/oncotarget.13985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 12/12/2016] [Indexed: 12/30/2022] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PC) have a poor prognosis due to metastases and chemoresistance. PC is characterized by extensive fibrosis, which creates a hypoxic microenvironment, and leads to increased chemoresistance and intracellular oxidative stress. Thus, proteins that protect against oxidative stress are potential therapeutic targets for PC. A key protein that maintains genomic integrity against oxidative damage is MutY-Homolog (MYH). No prior studies have investigated the function of MYH in PC cells. Using siRNA, we showed that knockdown of MYH in PC cells 1) reduced PC cell proliferation and increased apoptosis; 2) further decreased PC cell growth in the presence of oxidative stress and chemotherapy agents (gemcitabine, paclitaxel and vincristine); 3) reduced PC cell metastatic potential; and 4) decreased PC tumor growth in a subcutaneous mouse model in vivo. The results from this study suggest MYH may be a novel therapeutic target for PC that could potentially improve patient outcome by reducing PC cell survival, increasing the efficacy of existing drugs and reducing metastatic spread.
Collapse
Affiliation(s)
- George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Amanda Mawson
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, Australia
| | - Andrea Nunez
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Andrew Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, Scotland, United Kingdom
- The Kinghorn Cancer Centre, Cancer Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Amber Johns
- The Kinghorn Cancer Centre, Cancer Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Phoebe Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
29
|
Dobiasch S, Szanyi S, Kjaev A, Werner J, Strauss A, Weis C, Grenacher L, Kapilov-Buchman K, Israel LL, Lellouche JP, Locatelli E, Franchini MC, Vandooren J, Opdenakker G, Felix K. Synthesis and functionalization of protease-activated nanoparticles with tissue plasminogen activator peptides as targeting moiety and diagnostic tool for pancreatic cancer. J Nanobiotechnology 2016; 14:81. [PMID: 27993133 PMCID: PMC5168863 DOI: 10.1186/s12951-016-0236-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/03/2016] [Indexed: 11/24/2022] Open
Abstract
Background Functionalized nanoparticles (NPs) are one promising tool for detecting specific molecular targets and combine molecular biology and nanotechnology aiming at modern imaging. We aimed at ligand-directed delivery with a suitable target-biomarker to detect early pancreatic ductal adenocarcinoma (PDAC). Promising targets are galectins (Gal), due to their strong expression in and on PDAC-cells and occurrence at early stages in cancer precursor lesions, but not in adjacent normal tissues. Results Molecular probes (10-29 AA long peptides) derived from human tissue plasminogen activator (t-PA) were selected as binding partners to galectins. Affinity constants between the synthesized t-PA peptides and Gal were determined by microscale thermophoresis. The 29 AA-long t-PA-peptide-1 with a lactose-functionalized serine revealed the strongest binding properties to Gal-1 which was 25-fold higher in comparison with the native t-PA protein and showed additional strong binding to Gal-3 and Gal-4, both also over-expressed in PDAC. t-PA-peptide-1 was selected as vector moiety and linked covalently onto the surface of biodegradable iron oxide nanoparticles (NPs). In particular, CAN-doped maghemite NPs (CAN-Mag), promising as contrast agent for magnetic resonance imaging (MRI), were selected as magnetic core and coated with different biocompatible polymers, such as chitosan (CAN-Mag-Chitosan NPs) or polylactic co glycolic acid (PLGA) obtaining polymeric nanoparticles (CAN-Mag@PNPs), already approved for drug delivery applications. The binding efficacy of t-PA-vectorized NPs determined by exposure to different pancreatic cell lines was up to 90%, as assessed by flow cytometry. The in vivo targeting and imaging efficacy of the vectorized NPs were evaluated by applying murine pancreatic tumor models and assessed by 1.5 T magnetic resonance imaging (MRI). The t-PA-vectorized NPs as well as the protease-activated NPs with outer shell decoration (CAN-Mag@PNPs-PEG-REGAcp-PEG/tPA-pep1Lac) showed clearly detectable drop of subcutaneous and orthotopic tumor staining-intensity indicating a considerable uptake of the injected NPs. Post mortem NP deposition in tumors and organs was confirmed by Fe staining of histopathology tissue sections. Conclusions The targeted NPs indicate a fast and enhanced deposition of NPs in the murine tumor models. The CAN-Mag@PNPs-PEG-REGAcp-PEG/tPA-pep1Lac interlocking steps strategy of NPs delivery and deposition in pancreatic tumor is promising.
Collapse
Affiliation(s)
- Sophie Dobiasch
- Department of Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Technische Universität München, Munich, Germany
| | - Szilard Szanyi
- Department of Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Aleko Kjaev
- Department of Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Jens Werner
- Department of Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany.,Department of General-, Visceral-, Transplantations-, Vascular- and Thorax-Surgery LMU Munich, München, Germany
| | - Albert Strauss
- Department of Radiology, University of Heidelberg, Heidelberg, Germany
| | - Christian Weis
- Department of Radiology, University of Heidelberg, Heidelberg, Germany
| | - Lars Grenacher
- Department of Radiology, University of Heidelberg, Heidelberg, Germany.,Diagnostik München, Diagnostic Imaging and Prevention Center, Munich, Germany
| | - Katya Kapilov-Buchman
- Nanomaterials Research Center, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel
| | - Liron-Limor Israel
- Nanomaterials Research Center, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel
| | - Jean-Paul Lellouche
- Nanomaterials Research Center, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel
| | - Erica Locatelli
- Department of Industrial Chemistry Toso Montanari, University of Bologna, Bologna, Italy
| | - Mauro Comes Franchini
- Department of Industrial Chemistry Toso Montanari, University of Bologna, Bologna, Italy
| | - Jennifer Vandooren
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Klaus Felix
- Department of Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany.
| |
Collapse
|
30
|
Thombre RS, Shinde V, Thaiparambil E, Zende S, Mehta S. Antimicrobial Activity and Mechanism of Inhibition of Silver Nanoparticles against Extreme Halophilic Archaea. Front Microbiol 2016; 7:1424. [PMID: 27679615 PMCID: PMC5020055 DOI: 10.3389/fmicb.2016.01424] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 08/29/2016] [Indexed: 01/31/2023] Open
Abstract
Haloarchaea are salt-loving halophilic microorganisms that inhabit marine environments, sea water, salterns, and lakes. The resistance of haloarchaea to physical extremities that challenge organismic survival is ubiquitous. Metal and antibiotic resistance of haloarchaea has been on an upsurge due to the exposure of these organisms to metal sinks and drug resistance genes augmented in their natural habitats due to anthropogenic activities and environmental pollution. The efficacy of silver nanoparticles (SNPs) as a potent and broad spectrum inhibitory agent is known, however, there are no reports on the inhibitory activity of SNPs against haloarchaea. In the present study, we have investigated the antimicrobial potentials of SNPs synthesized using aqueous leaf extract of Cinnamomum tamala against antibiotic resistant haloarchaeal isolates Haloferax prahovense RR8, Haloferax lucentense RR15, Haloarcula argentinensis RR10 and Haloarcula tradensis RR13. The synthesized SNPs were characterized by UV-Vis spectroscopy, scanning electron microscopy, energy dispersive X-ray spectroscopy, dynamic light scattering, X-ray diffraction and Fourier transform infrared spectroscopy. The SNPs demonstrated potent antimicrobial activity against the haloarchaea with a minimum inhibitory concentration of 300-400 μg/ml. Growth kinetics of haloarchaea in the presence of SNPs was studied by employing the Baranyi mathematical model for microbial growth using the DMFit curve fitting program. The C. tamala SNPs also demonstrated cytotoxic activity against human lung adenocarcinoma epithelial cell line (A540) and human breast adenocarcinoma cell line (MCF-7). The mechanism of inhibition of haloarchaea by the SNPs was investigated. The plausible mechanism proposed is the alterations and disruption of haloarchaeal membrane permeability by turbulence, inhibition of respiratory dehydrogenases and lipid peroxidation causing cellular and DNA damage resulting in cell death.
Collapse
Affiliation(s)
- Rebecca S. Thombre
- Department of Biotechnology, Modern College of Arts, Science and CommercePune, India
| | - Vinaya Shinde
- Department of Biotechnology, Modern College of Arts, Science and CommercePune, India
| | - Elvina Thaiparambil
- Department of Biotechnology, Modern College of Arts, Science and CommercePune, India
| | - Samruddhi Zende
- Department of Biotechnology, Modern College of Arts, Science and CommercePune, India
| | - Sourabh Mehta
- National Center for Nanosciences and Nanotechnology, University of MumbaiMumbai, India
| |
Collapse
|
31
|
Au M, Emeto TI, Power J, Vangaveti VN, Lai HC. Emerging Therapeutic Potential of Nanoparticles in Pancreatic Cancer: A Systematic Review of Clinical Trials. Biomedicines 2016; 4:E20. [PMID: 28536387 PMCID: PMC5344258 DOI: 10.3390/biomedicines4030020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is an aggressive disease with a five year survival rate of less than 5%, which is associated with late presentation. In recent years, research into nanomedicine and the use of nanoparticles as therapeutic agents for cancers has increased. This article describes the latest developments in the use of nanoparticles, and evaluates the risks and benefits of nanoparticles as an emerging therapy for pancreatic cancer. The Preferred Reporting Items of Systematic Reviews and Meta-Analyses checklist was used. Studies were extracted by searching the Embase, MEDLINE, SCOPUS, Web of Science, and Cochrane Library databases from inception to 18 March 2016 with no language restrictions. Clinical trials involving the use of nanoparticles as a therapeutic or prognostic option in patients with pancreatic cancer were considered. Selected studies were evaluated using the Jadad score for randomised control trials and the Therapy CA Worksheet for intervention studies. Of the 210 articles found, 10 clinical trials including one randomised control trial and nine phase I/II clinical trials met the inclusion criteria and were analysed. These studies demonstrated that nanoparticles can be used in conjunction with chemotherapeutic agents increasing their efficacy whilst reducing their toxicity. Increased efficacy of treatment with nanoparticles may improve the clinical outcomes and quality of life in patients with pancreatic cancer, although the long-term side effects are yet to be defined. The study registration number is CRD42015020009.
Collapse
Affiliation(s)
- Minnie Au
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, James Cook Drive, Douglas, Townsville QLD 4811, Australia.
- Townsville Cancer Centre, The Townsville Hospital, Townsville QLD 4814, Australia.
| | - Theophilus I Emeto
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, James Cook Drive, Douglas, Townsville QLD 4811, Australia.
| | - Jacinta Power
- Townsville Cancer Centre, The Townsville Hospital, Townsville QLD 4814, Australia.
| | - Venkat N Vangaveti
- College of Medicine and Dentistry, James Cook University, James Cook Drive, Douglas, Townsville QLD 4811, Australia.
| | - Hock C Lai
- Townsville Cancer Centre, The Townsville Hospital, Townsville QLD 4814, Australia.
| |
Collapse
|
32
|
Teo J, McCarroll JA, Boyer C, Youkhana J, Sagnella SM, Duong HTT, Liu J, Sharbeen G, Goldstein D, Davis TP, Kavallaris M, Phillips PA. A Rationally Optimized Nanoparticle System for the Delivery of RNA Interference Therapeutics into Pancreatic Tumors in Vivo. Biomacromolecules 2016; 17:2337-51. [DOI: 10.1021/acs.biomac.6b00185] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Joann Teo
- Tumour
Biology and Targeting Program, Children’s Cancer Institute,
Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales 2052, Australia
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Joshua A. McCarroll
- Tumour
Biology and Targeting Program, Children’s Cancer Institute,
Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales 2052, Australia
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Cyrille Boyer
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
- Centre
for Advanced Macromolecular Design, School of Chemical Engineering, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Janet Youkhana
- Pancreatic
Cancer Translational Research Group, Lowy Cancer Research Centre,
Prince of Wales Clinical School, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Sharon M. Sagnella
- Tumour
Biology and Targeting Program, Children’s Cancer Institute,
Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales 2052, Australia
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Hien T. T. Duong
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
- Centre
for Advanced Macromolecular Design, School of Chemical Engineering, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Jie Liu
- Pancreatic
Cancer Translational Research Group, Lowy Cancer Research Centre,
Prince of Wales Clinical School, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - George Sharbeen
- Pancreatic
Cancer Translational Research Group, Lowy Cancer Research Centre,
Prince of Wales Clinical School, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - David Goldstein
- Pancreatic
Cancer Translational Research Group, Lowy Cancer Research Centre,
Prince of Wales Clinical School, UNSW Australia, Sydney, New South Wales 2052, Australia
- Prince
of Wales Hospital, Prince of Wales Clinical School, Sydney, New South Wales 2052, Australia
| | - Thomas P. Davis
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology
Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, Victoria 3800, Australia
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Maria Kavallaris
- Tumour
Biology and Targeting Program, Children’s Cancer Institute,
Lowy Cancer Research Centre, UNSW Australia, Sydney, New South Wales 2052, Australia
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
- ARC Centre
of Excellence in Convergent Bio-Nano Science and Technology UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Phoebe A. Phillips
- Australian
Centre for NanoMedicine, UNSW Australia, Sydney, New South Wales 2052, Australia
- Pancreatic
Cancer Translational Research Group, Lowy Cancer Research Centre,
Prince of Wales Clinical School, UNSW Australia, Sydney, New South Wales 2052, Australia
| |
Collapse
|
33
|
Sápi J, Kovács L, Drexler DA, Kocsis P, Gajári D, Sápi Z. Tumor Volume Estimation and Quasi-Continuous Administration for Most Effective Bevacizumab Therapy. PLoS One 2015; 10:e0142190. [PMID: 26540189 PMCID: PMC4635016 DOI: 10.1371/journal.pone.0142190] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 10/19/2015] [Indexed: 02/04/2023] Open
Abstract
Background Bevacizumab is an exogenous inhibitor which inhibits the biological activity of human VEGF. Several studies have investigated the effectiveness of bevacizumab therapy according to different cancer types but these days there is an intense debate on its utility. We have investigated different methods to find the best tumor volume estimation since it creates the possibility for precise and effective drug administration with a much lower dose than in the protocol. Materials and Methods We have examined C38 mouse colon adenocarcinoma and HT-29 human colorectal adenocarcinoma. In both cases, three groups were compared in the experiments. The first group did not receive therapy, the second group received one 200 μg bevacizumab dose for a treatment period (protocol-based therapy), and the third group received 1.1 μg bevacizumab every day (quasi-continuous therapy). Tumor volume measurement was performed by digital caliper and small animal MRI. The mathematical relationship between MRI-measured tumor volume and mass was investigated to estimate accurate tumor volume using caliper-measured data. A two-dimensional mathematical model was applied for tumor volume evaluation, and tumor- and therapy-specific constants were calculated for the three different groups. The effectiveness of bevacizumab administration was examined by statistical analysis. Results In the case of C38 adenocarcinoma, protocol-based treatment did not result in significantly smaller tumor volume compared to the no treatment group; however, there was a significant difference between untreated mice and mice who received quasi-continuous therapy (p = 0.002). In the case of HT-29 adenocarcinoma, the daily treatment with one-twelfth total dose resulted in significantly smaller tumors than the protocol-based treatment (p = 0.038). When the tumor has a symmetrical, solid closed shape (typically without treatment), volume can be evaluated accurately from caliper-measured data with the applied two-dimensional mathematical model. Conclusion Our results provide a theoretical background for a much more effective bevacizumab treatment using optimized administration.
Collapse
Affiliation(s)
- Johanna Sápi
- Research and Innovation Center of Obuda University, Physiological Controls Group, Obuda University, Budapest, Hungary
| | - Levente Kovács
- Research and Innovation Center of Obuda University, Physiological Controls Group, Obuda University, Budapest, Hungary
| | - Dániel András Drexler
- Department of Control Engineering and Information Technology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Pál Kocsis
- Preclinical Imaging Center, Gedeon Richter Plc., Budapest, Hungary
| | - Dávid Gajári
- Preclinical Imaging Center, Gedeon Richter Plc., Budapest, Hungary
| | - Zoltán Sápi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
34
|
Chen C, Wu CQ, Chen TW, Tang MY, Zhang XM. Molecular Imaging with MRI: Potential Application in Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:624074. [PMID: 26579537 PMCID: PMC4633535 DOI: 10.1155/2015/624074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/02/2015] [Accepted: 10/04/2015] [Indexed: 12/11/2022]
Abstract
Despite the variety of approaches that have been improved to achieve a good understanding of pancreatic cancer (PC), the prognosis of PC remains poor, and the survival rates are dismal. The lack of early detection and effective interventions is the main reason. Therefore, considerable ongoing efforts aimed at identifying early PC are currently being pursued using a variety of methods. In recent years, the development of molecular imaging has made the specific targeting of PC in the early stage possible. Molecular imaging seeks to directly visualize, characterize, and measure biological processes at the molecular and cellular levels. Among different imaging technologies, the magnetic resonance (MR) molecular imaging has potential in this regard because it facilitates noninvasive, target-specific imaging of PC. This topic is reviewed in terms of the contrast agents for MR molecular imaging, the biomarkers related to PC, targeted molecular probes for MRI, and the application of MRI in the diagnosis of PC.
Collapse
Affiliation(s)
- Chen Chen
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, Sichuan 637000, China
| | - Chang Qiang Wu
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, Sichuan 637000, China
| | - Tian Wu Chen
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, Sichuan 637000, China
| | - Meng Yue Tang
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, Sichuan 637000, China
| | - Xiao Ming Zhang
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, Sichuan 637000, China
| |
Collapse
|
35
|
Zheng K, Dou Y, He L, Li H, Zhang Z, Chen Y, Ye A, Liu W, Kong L. Improved sensitivity and specificity for prostate cancer diagnosis based on the urine PCA3/PSA ratio acquired by sequence‑specific RNA capture. Oncol Rep 2015; 34:2439-44. [PMID: 26351770 DOI: 10.3892/or.2015.4266] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 07/17/2015] [Indexed: 11/05/2022] Open
Abstract
Prostate cancer antigen 3 (PCA3) is a non-coding RNA fragment that is overexpressed in prostate cancer cells. However, the clinical applications of PCA3 are highly limited due to the instability of RNA and the lack of reliable and efficient RNA extraction and purification methods. Thus, in the present study, we compared three different methods of RNA extraction to further confirm the higher yield of commercial magnetic beads with poly-T functionalization and a capturer strand. The current protocols for RNA extraction of i) the phenol-chloroform method, ii) the affinity column method and iii) magnetic beads with poly-T functionalization and a capturer strand were applied separately for RNA extraction in urine samples. Reverse transcription‑quantitative polymerase chain reaction was performed to evaluate the yield of the three methods of RNA extraction. Furthermore, 52 urine samples after prostate massage from patients suspected of a diagnosis of prostate cancer were collected. The Mag-Cap method and RT-PCR were applied to obtain the PCA3 score. The clinical value of the PCA3 score was investigated by comparison with the pathology of the prostate biopsy. The yield of the Mag-Cap method was higher than that of the phenol‑chloroform method and commercial kits. Thirty‑four patients were pathologically diagnosed with prostate cancer and 18 with benign prostatic hyperplasia (BPH). It was confirmed that the median PCA3 score was higher among the prostate cancer patients than those with benign disease (53.5 vs. 17, p=0.000). A sensitivity of 82.4% and a specificity of 77.8% were obtained when the cut-off value for the PCA3 score was 28.5. The Mag-Cap method was found to be more efficient for RNA extraction. The urinary PCA3 score is a promising method for prostate cancer screening, detection and diagnosis, and has the potential to reduce unnecessary prostate biopsies.
Collapse
Affiliation(s)
- Kewen Zheng
- Urology Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yaling Dou
- Laboratory Medicine Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Linfu He
- Institute of Bioengineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hanzhong Li
- Urology Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Zhicai Zhang
- Institute of Bioengineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yu Chen
- Laboratory Medicine Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Ali Ye
- Laboratory Medicine Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Wenjing Liu
- Laboratory Medicine Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Lingjun Kong
- Laboratory Medicine Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
36
|
McCarroll JA, Dwarte T, Baigude H, Dang J, Yang L, Erlich RB, Kimpton K, Teo J, Sagnella SM, Akerfeldt MC, Liu J, Phillips PA, Rana TM, Kavallaris M. Therapeutic targeting of polo-like kinase 1 using RNA-interfering nanoparticles (iNOPs) for the treatment of non-small cell lung cancer. Oncotarget 2015; 6:12020-34. [PMID: 25557168 PMCID: PMC4494920 DOI: 10.18632/oncotarget.2664] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/27/2014] [Indexed: 01/29/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the most common cause of cancer death worldwide due its resistance to chemotherapy and aggressive tumor growth. Polo-like kinase 1 (PLK1) is a serine-threonine protein kinase which is overexpressed in cancer cells, and plays a major role in regulating tumor growth. A number of PLK1 inhibitors are in clinical trial; however, poor tumor bioavailability and off-target effects limit their efficacy. Short-interfering-RNA (siRNA) holds promise as a class of therapeutics, which can selectively silence disease-causing genes. However, siRNA cannot enter cells without a delivery vehicle. Herein, we investigated whether RNAi-interfering nanoparticles could deliver siRNA to NSCLC cells and silence PLK1 expression in vitro and in vivo. iNOP-7 was non-toxic, and delivered siRNA with high efficiency to NSCLC cells. iNOP-7-PLK1 siRNA silenced PLK1 expression and reduced NSCLC growth in vitro. Notably, iNOP-7 delivered siRNA to orthotopic lung tumors in mice, and administration of iNOP-7-PLK1 siRNA reduced lung tumor burden. These novel data show that iNOP-7 can deliver siRNA against PLK1 to NSCLC cells, and decrease cell proliferation both in vitro and in vivo. iNOP-7-PLK1 siRNA may provide a novel therapeutic strategy for the treatment of NSCLC as well as other cancers which aberrantly express this gene.
Collapse
Affiliation(s)
- Joshua A. McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, NSW, Australia
| | - Tanya Dwarte
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
| | - Huricha Baigude
- Program for RNA Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Jason Dang
- Program for RNA Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Lu Yang
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
| | - Rafael B. Erlich
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, NSW, Australia
| | - Kathleen Kimpton
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
| | - Joann Teo
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, NSW, Australia
| | - Sharon M. Sagnella
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, NSW, Australia
| | - Mia C. Akerfeldt
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
| | - Jie Liu
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW, NSW, Australia
| | - Phoebe A. Phillips
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, NSW, Australia
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW, NSW, Australia
| | - Tariq M. Rana
- Program for RNA Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, UNSW Australia (UNSW), NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, NSW, Australia
| |
Collapse
|
37
|
Zhao X, Huang Q, Jin Y. Gold nanorod delivery of LSD1 siRNA induces human mesenchymal stem cell differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 54:142-9. [PMID: 26046277 DOI: 10.1016/j.msec.2015.05.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/28/2015] [Accepted: 05/07/2015] [Indexed: 10/23/2022]
Abstract
Over the past decade, theranostic nanoparticles with microsize and multifunctional ability have emerged as a new platform in biomedical field, such as cancer therapy, optical imaging and gene therapy. Gene therapy has been recently shown as a promising tool for tissue engineering as safe and effective nanotechnology-based delivery methods are developed. Controlling adhesion and differentiation of stem cells is critical for tissue regeneration. In this study, we have developed poly-sodium 4-styrenesulfonate (PSS) and poly-allylamine hydrochloride (PAH) coated AuNR-based nanocarriers, which are capable of delivering small interfering RNA (siRNA) against LSD1 to induce the differentiation of human mesenchymal stem cells. To further study the mechanism, we tested the stemness and differentiation genes and found that they have been changed with LSD1 down-regulation. In addition, with the hepatocyte growth factor (HGF), LSD1 siRNA delivery by AuNRs could promote the differentiation of the human mesenchymal stem cells (human MSCs) into a hepatocyte lineage in vitro. Our results suggest for the first time use of AuNRs as nanocarriers of delivery LSD1 siRNA to induce the differentiation of human MSCs into a hepatocyte lineage, and envision the potential application of nanotechnology in tissue remodeling (such as liver and bone) in vivo, eventually translating to clinical applications.
Collapse
Affiliation(s)
- Xiongfei Zhao
- R&D center, Shanghai Angecon Biotechnology Co., Ltd, 588 Qiangdai Road, Pudong District, Shanghai 201318, China
| | - Qianying Huang
- R&D center, Shanghai Angecon Biotechnology Co., Ltd, 588 Qiangdai Road, Pudong District, Shanghai 201318, China
| | - Yiqiang Jin
- R&D center, Shanghai Angecon Biotechnology Co., Ltd, 588 Qiangdai Road, Pudong District, Shanghai 201318, China.
| |
Collapse
|
38
|
Yin F, Yang C, Wang Q, Zeng S, Hu R, Lin G, Tian J, Hu S, Lan RF, Yoon HS, Lu F, Wang K, Yong KT. A Light-Driven Therapy of Pancreatic Adenocarcinoma Using Gold Nanorods-Based Nanocarriers for Co-Delivery of Doxorubicin and siRNA. Theranostics 2015; 5:818-33. [PMID: 26000055 PMCID: PMC4440440 DOI: 10.7150/thno.11335] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/14/2015] [Indexed: 01/05/2023] Open
Abstract
In this work, we report the engineering of polyelectrolyte polymers coated Gold nanorods (AuNRs)-based nanocarriers that are capable of co-delivering small interfering RNA (siRNA) and an anticancer drug doxorubicin (DOX) to Panc-1 cancer cells for combination of both chemo- and siRNA-mediated mutant K-Ras gene silencing therapy. Superior anticancer efficacy was observed through synergistic combination of promoted siRNA and DOX release upon irradiating the nanoplex formulation with 665 nm light. Our antitumor study shows that the synergistic effect of AuNRs nanoplex formulation with 665 nm light treatment is able to inhibit the in vivo tumor volume growth rate by 90%. The antitumor effect is contributed from the inactivation of K-Ras gene and thereby causing a profound synthesis (S) phase arrest in treated Panc-1 cells. Our study shows that the percentage of Panc-1 cells treated by nanoplex formulation with S phase is determined to be 35% and it is 17% much higher than that of Panc-1 cells without any treatments. The developed nanotherapy formulation here, that combines chemotherapy, RNA silencing and NIR window light-mediated therapy, will be seen to be the next natural step to be taken in the clinical research for improving the therapeutic outcomes of the pancreatic adenocarcinoma treatment.
Collapse
Affiliation(s)
- Feng Yin
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Chengbin Yang
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Qianqian Wang
- 3. Laboratory of Chemical Genetics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Shuwen Zeng
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
- 6. CINTRA CNRS/NTU/THALES, UMI 3288, Research Techno Plaza, 50 Nanyang Drive, Border X Block, Singapore, 637553
| | - Rui Hu
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Guimiao Lin
- 5. The key lab of Biomedical Engineering and Research Institute of Uropoiesis and Reproduction, School of Medical Sciences, Shenzhen University, Shenzhen, 518060, China
| | - Jinglin Tian
- 5. The key lab of Biomedical Engineering and Research Institute of Uropoiesis and Reproduction, School of Medical Sciences, Shenzhen University, Shenzhen, 518060, China
| | - Siyi Hu
- 7. School of Science, Changchun University of Science and Technology, Changchun, 130022, China
| | - Rong Feng Lan
- 8. Institute of Research and Continuing Education, Hong Kong Baptist University (Shenzhen), Shenzhen 518057, China
| | - Ho Sup Yoon
- 2. Division of Structural Biology & Biochemistry, School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
- 9. Department of Genetic Engineering, College of Life Sciences, Kyung Hee University, Yongin-si Gyeonggi-do, 446-701, Republic of Korea
| | - Fei Lu
- 3. Laboratory of Chemical Genetics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Kuan Wang
- 4. Nanomedicine Program and Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Ken-Tye Yong
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
39
|
Sharbeen G, McCarroll J, Goldstein D, Phillips PA. Exploiting base excision repair to improve therapeutic approaches for pancreatic cancer. Front Nutr 2015; 2:10. [PMID: 25988138 PMCID: PMC4428371 DOI: 10.3389/fnut.2015.00010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/10/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly chemoresistant and metastatic disease with a dismal 5-year survival rate of 6%. More effective therapeutic targets and approaches are urgently needed to tackle this devastating disease. The base excision repair (BER) pathway has been identified as a predictor of therapeutic response, prognostic factor, and therapeutic target in a variety of cancers. This review will discuss our current understanding of BER in PDA and its potential to improve PDA treatment.
Collapse
Affiliation(s)
- George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Australia , Sydney, NSW , Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Australia , Sydney, NSW , Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Australia , Sydney, NSW , Australia
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Australia , Sydney, NSW , Australia
| |
Collapse
|
40
|
Frasco MF, Almeida GM, Santos-Silva F, Pereira MDC, Coelho MAN. Transferrin surface-modified PLGA nanoparticles-mediated delivery of a proteasome inhibitor to human pancreatic cancer cells. J Biomed Mater Res A 2014; 103:1476-84. [PMID: 25046528 DOI: 10.1002/jbm.a.35286] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/23/2014] [Accepted: 07/18/2014] [Indexed: 12/17/2022]
Abstract
The aim of this study was to develop a drug delivery system based on poly(lactic-co-glycolic acid) (PLGA) nanoparticles for an efficient and targeted action of the proteasome inhibitor bortezomib against pancreatic cancer cells. The PLGA nanoparticles were formulated with a poloxamer, and further surface-modified with transferrin for tumor targeting. The nanoparticles were characterized as polymer carriers of bortezomib, and the cellular uptake and growth inhibitory effects were evaluated in pancreatic cells. Cellular internalization of nanoparticles was observed in normal and cancer cells, but with higher uptake by cancer cells. The sustained release of the loaded bortezomib from PLGA nanoparticles showed cytotoxic effects against pancreatic normal and cancer cells. Noteworthy differential cytotoxicity was attained by transferrin surface-modified PLGA nanoparticles since significant cell growth inhibition by delivered bortezomib was only observed in cancer cells. These findings demonstrate that the ligand transferrin enhanced the targeted delivery of bortezomib-loaded PLGA nanoparticles to pancreatic cancer cells. These in vitro results highlight the transferrin surface-modified PLGA nanoparticles as a promising system for targeted delivery of anticancer drugs.
Collapse
Affiliation(s)
- Manuela F Frasco
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | | | | | | | | |
Collapse
|
41
|
Targeting the C-terminal focal adhesion kinase scaffold in pancreatic cancer. Cancer Lett 2014; 353:281-9. [PMID: 25067788 DOI: 10.1016/j.canlet.2014.07.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/16/2014] [Accepted: 07/19/2014] [Indexed: 12/20/2022]
Abstract
Preliminary studies in our laboratory have demonstrated the importance of both the NH2 and COOH terminus scaffolding functions of focal adhesion kinase (FAK). Here, we describe a new small molecule inhibitor, C10, that targets the FAK C-terminus scaffold. C10 showed marked selectivity for cells overexpressing VEGFR3 when tested in isogenic cell lines, MCF7 and MCF7-VEGFR3. C10 preferentially inhibited pancreatic tumor growth in vivo in cells with high FAK-Y925 and VEGFR3 expression. Treatment with C10 led to a significant inhibition in endothelial cell proliferation and tumor endothelial and lymphatic vessel density and decrease in interstitial fluid pressure. These results highlight the underlying importance of targeting the FAK scaffold to treat human cancers.
Collapse
|