1
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
2
|
Ghafelehbashi R, Farshbafnadi M, Aghdam NS, Amiri S, Salehi M, Razi S. Nanoimmunoengineering strategies in cancer diagnosis and therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:78-90. [PMID: 36076122 DOI: 10.1007/s12094-022-02935-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Cancer immunotherapy strategies in combination with engineered nanosystems have yielded beneficial results in the treatment of cancer and their application is increasing day by day. The pivotal role of stimuli-responsive nanosystems and nanomedicine-based cancer immunotherapy, as a subsidiary discipline in the field of immunology, cannot be ignored. Today, rapid advances in nanomedicine are used as a platform for exploring new therapeutic applications and modern smart healthcare management strategies. The progress of nanomedicine in cancer treatment has confirmed the findings of immunotherapy in the medical research phase. This study concentrates on approaches connected to the efficacy of nanoimmunoengineering strategies for cancer immunotherapies and their applications. By assessing improved approaches, different aspects of the nanoimmunoengineering strategies for cancer therapies are discussed in this study.
Collapse
Affiliation(s)
- Robabehbeygom Ghafelehbashi
- Department of Materials and Textile Engineering, College of Engineering, Razi University, Kermanshah, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Melina Farshbafnadi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Mitra Salehi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Jia M, Zhang D, Zhang C, Li C. Nanoparticle-based delivery systems modulate the tumor microenvironment in pancreatic cancer for enhanced therapy. J Nanobiotechnology 2021; 19:384. [PMID: 34809634 PMCID: PMC8607729 DOI: 10.1186/s12951-021-01134-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignant tumors with a low survival rate, partly because the tumor microenvironment (TME), which consists of extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), immune cells, and vascular systems, prevents effective drug delivery and chemoradiotherapy. Thus, modulating the microenvironment of pancreatic cancer is considered a promising therapeutic approach. Since nanoparticles are one of the most effective cancer treatment strategies, several nano-delivery platforms have been developed to regulate the TME and enhance treatment. Here, we summarize the latest advances in nano-delivery systems that alter the TME in pancreatic cancer by depleting ECM, inhibiting CAFs, reversing immunosuppression, promoting angiogenesis, or improving the hypoxic environment. We also discuss promising new targets for such systems. This review is expected to improve our understanding of how to modulate the pancreatic cancer microenvironment and guide the development of new therapies.
Collapse
Affiliation(s)
- Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China
| | - Dan Zhang
- Department of Pharmacy of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China.
| |
Collapse
|
4
|
Lôbo GCNB, Paiva KLR, Silva ALG, Simões MM, Radicchi MA, Báo SN. Nanocarriers Used in Drug Delivery to Enhance Immune System in Cancer Therapy. Pharmaceutics 2021; 13:1167. [PMID: 34452128 PMCID: PMC8399799 DOI: 10.3390/pharmaceutics13081167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer, a group of diseases responsible for the second largest cause of global death, is considered one of the main public health problems today. Despite the advances, there are still difficulties in the development of more efficient cancer therapies and fewer adverse effects for the patients. In this context, nanobiotechnology, a materials science on a nanometric scale specified for biology, has been developing and acquiring prominence for the synthesis of nanocarriers that provide a wide surface area in relation to volume, better drug delivery, and a maximization of therapeutic efficiency. Among these carriers, the ones that stand out are those focused on the activation of the immune system. The literature demonstrates the importance of this system for anticancer therapy, given that the best treatment for this disease also activates the immune system to recognize, track, and destroy all remaining tumor cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Sônia N. Báo
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (G.C.N.B.L.); (K.L.R.P.); (A.L.G.S.); (M.M.S.); (M.A.R.)
| |
Collapse
|
5
|
Zhao YD, Muhetaerjiang M, An HW, Fang X, Zhao Y, Wang H. Nanomedicine enables spatiotemporally regulating macrophage-based cancer immunotherapy. Biomaterials 2020; 268:120552. [PMID: 33307365 DOI: 10.1016/j.biomaterials.2020.120552] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy, leveraging the host's coordinated immune system to fight against tumor has been clinically validated. However, the modest response owing to the multiple ways of tumor immune evasion is one of the challenges in cancer immunotherapy. Tumor associated macrophages (TAMs), as a major component of the leukocytes infiltrating in all tumors, play crucial roles in driving cancer initiation, progress and metastasis via multiple mechanisms such as mediating chronic inflammation, promoting angiogenesis, taming protective immune responses, and supporting migration and intravasation. TAMs targeted therapeutics have achieved remarkable successes in clinical trials mostly through the use of small-molecule agents and antibodies. However, efforts for further application have met with challenges of limited efficacy and safety. Nanomaterials can provide versatile approaches to realize the superior spatiotemporal control over immunomodulation to amplify immune responses, ultimately enhancing the therapeutic benefits and reducing toxicity. Here, the potential drugs used in TAM-centered cancer treatment in clinic are summarized and the recent advances of TAMs targeted nanomedicines in this filed are highlighted. More importantly, we focus on how nanomedicine can exert their advantages in spatial and temporal control of immunomodulation.
Collapse
Affiliation(s)
- Yong-Dan Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China; School of Pharmacy, Shanxi Medical University, Shanxi, 030009, PR China
| | - Mamuti Muhetaerjiang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Xiaohong Fang
- University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
6
|
Yang Q, Guo N, Zhou Y, Chen J, Wei Q, Han M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm Sin B 2020; 10:2156-2170. [PMID: 33304783 PMCID: PMC7714989 DOI: 10.1016/j.apsb.2020.04.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages have a leading position in the tumor microenvironment (TME) which paves the way to carcinogenesis. Initially, monocytes and macrophages are recruited to the sites where the tumor develops. Under the guidance of different microenvironmental signals, macrophages would polarize into two functional phenotypes, named as classically activated macrophages (M1) and alternatively activated macrophages (M2). Contrary to the anti-tumor effect of M1, M2 exerts anti-inflammatory and tumorigenic characters. In progressive tumor, M2 tumor-associated macrophages (TAMs) are in the majority, being vital regulators reacting upon TME. This review elaborates on the role of TAMs in tumor progression. Furthermore, prospective macrophage-focused therapeutic strategies, including drugs not only in clinical trials but also at primary research stages, are summarized followed by a discussion about their clinical application values. Nanoparticulate systems with efficient drug delivery and improved antitumor effect are also summed up in this article.
Collapse
Affiliation(s)
- Qiyao Yang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ningning Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Zhou
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiejian Chen
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qichun Wei
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
7
|
Frieboes HB, Raghavan S, Godin B. Modeling of Nanotherapy Response as a Function of the Tumor Microenvironment: Focus on Liver Metastasis. Front Bioeng Biotechnol 2020; 8:1011. [PMID: 32974325 PMCID: PMC7466654 DOI: 10.3389/fbioe.2020.01011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) presents a challenging barrier for effective nanotherapy-mediated drug delivery to solid tumors. In particular for tumors less vascularized than the surrounding normal tissue, as in liver metastases, the structure of the organ itself conjures with cancer-specific behavior to impair drug transport and uptake by cancer cells. Cells and elements in the TME of hypovascularized tumors play a key role in the process of delivery and retention of anti-cancer therapeutics by nanocarriers. This brief review describes the drug transport challenges and how they are being addressed with advanced in vitro 3D tissue models as well as with in silico mathematical modeling. This modeling complements network-oriented techniques, which seek to interpret intra-cellular relevant pathways and signal transduction within cells and with their surrounding microenvironment. With a concerted effort integrating experimental observations with computational analyses spanning from the molecular- to the tissue-scale, the goal of effective nanotherapy customized to patient tumor-specific conditions may be finally realized.
Collapse
Affiliation(s)
- Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY, United States
| | - Shreya Raghavan
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, TX, United States
- Developmental Therapeutics Program, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
8
|
Etman SM, Abdallah OY, Mehanna RA, Elnaggar YSR. Lactoferrin/Hyaluronic acid double-coated lignosulfonate nanoparticles of quinacrine as a controlled release biodegradable nanomedicine targeting pancreatic cancer. Int J Pharm 2020; 578:119097. [PMID: 32032904 DOI: 10.1016/j.ijpharm.2020.119097] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
Quinacrine is an antimalarial drug that was repositioned for treatment of cancer. This is the first work to enhance quinacrine activity and minimize its associated hepatotoxicity via loading into bio-degradable, bio-renewable lignosulfonate nanoparticles. Particles were appraised for treatment of pancreatic cancer, one of the most life-threatening tumors with a five-year survival estimate. Optimum nanocomposites prepared by polyelectrolyte interaction exhibited a particle size of 138 nm, a negative surface charge (-28 mV) and a pH dependent release of the drug in an acidic environment. Ligands used for active targeting (lactoferrin and hyaluronic acid) were added to nanoparticles' surface via layer by layer coating technique. The highest anticancer activity on PANC-1 cells was demonstrated with dual active targeted particles (3-fold decrease in IC50) along with an increased ability to inhibit migration and invasion of pancreatic cancer cells. In vivo studies revealed that elaborated nanoparticles particles showed the highest tumor volume reduction with enhanced survival without any toxicity on major organs. In conclusion, the elaborated nanoparticles could be considered as a promising targeted nanotherapy for treatment of pancreatic cancer with higher efficacy& survival rate and lower organ toxicity.
Collapse
Affiliation(s)
- Samar M Etman
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| | - Radwa A Mehanna
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications CERRMA, Faculty of Medicine, Alexandria University, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt; Head of International Publication and Nanotechnology Center INCC, Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University of Alexandria, Egypt.
| |
Collapse
|
9
|
Etman SM, Abdallah OY, Mehanna RA, Elnaggar YS. Lactoferrin/Hyaluronic acid double-coated lignosulfonate nanoparticles of quinacrine as a controlled release biodegradable nanomedicine targeting pancreatic cancer. Int J Pharm 2020. [DOI: https://doi.org/10.1016/j.ijpharm.2020.119097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
10
|
Zhang R, Liu Q, Peng J, Wang M, Gao X, Liao Q, Zhao Y. Pancreatic cancer-educated macrophages protect cancer cells from complement-dependent cytotoxicity by up-regulation of CD59. Cell Death Dis 2019; 10:836. [PMID: 31685825 PMCID: PMC6828776 DOI: 10.1038/s41419-019-2065-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022]
Abstract
Tumor-associated macrophages (TAMs) are versatile immune cells that promote a variety of malignant behaviors of pancreatic cancer. CD59 is a GPI-anchored membrane protein that prevents complement activation by inhibiting the formation of the membrane attack complex, which may protect cancer cells from complement-dependent cytotoxicity (CDC). The interactions between CD59, TAMs and pancreatic cancer remain largely unknown. A tissue microarray of pancreatic cancer patients was used to evaluate the interrelationship of CD59 and TAMs and their survival impacts were analyzed. In a coculture system, THP-1 cells were used as a model to study the function of TAMs and the roles of pancreatic cancer-educated macrophages in regulating the expression of CD59 in pancreatic cancer cells were demonstrated by real-time PCR, western blot and immunofluorescence staining. The effects of macrophages on regulating CDC in pancreatic cancer cells were demonstrated by an in vitro study. To explore the potential mechanisms, RNA sequencing of pancreatic cancer cells with or without co-culture of THP-1 macrophages was performed, and the results showed that the IL-6R/STAT3 signaling pathway might participate in the regulation, which was further demonstrated by target-siRNA transfection, antibody neutralization and STAT3 inhibitors. Our data revealed that the infiltration of TAMs and the expression of CD59 of pancreatic cancer were paralleled, and higher infiltration of TAMs and higher expression of CD59 predicted worse survival of pancreatic cancer patients. Pancreatic cancer-educated macrophages could protect cancer cells from CDC by up-regulating CD59 via the IL-6R/STAT3 signaling pathway. These findings uncovered the novel mechanisms between TAMs and CD59, and contribute to providing a new promising target for the immunotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Junya Peng
- Department of Center Lab, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Mengyi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiang Gao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
11
|
Hartshorn CM, Russell LM, Grodzinski P. National Cancer Institute Alliance for nanotechnology in cancer-Catalyzing research and translation toward novel cancer diagnostics and therapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1570. [PMID: 31257722 PMCID: PMC6788937 DOI: 10.1002/wnan.1570] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022]
Abstract
Nanotechnology has been a burgeoning research field, which is finding compelling applications in several practical areas of everyday life. It has provided novel, paradigm shifting solutions to medical problems and particularly to cancer. In order to accelerate integration of nanotechnology into cancer research and oncology, the National Cancer Institute (NCI) of the National Institutes of Health (NIH) established the NCI Alliance for Nanotechnology in Cancer program in 2005. This effort brought together scientists representing physical sciences, chemistry, and engineering working at the nanoscale with biologists and clinicians working on cancer to form a uniquely multidisciplinary cancer nanotechnology research community. The last 14 years of the program have produced a remarkable body of scientific discovery and demonstrated its utility to the development of practical cancer interventions. This paper takes stock of how the Alliance program influenced melding of disparate research disciplines into the field of nanomedicine and cancer nanotechnology, has been highly productive in the scientific arena, and produced a mechanism of seamless transfer of novel technologies developed in academia to the clinical and commercial space. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Christopher M. Hartshorn
- Nanodelivery Systems and Devices Branch, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Luisa M. Russell
- Nanodelivery Systems and Devices Branch, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, USA
| |
Collapse
|
12
|
Vakili‐Ghartavol R, Mombeiny R, Salmaninejad A, Sorkhabadi SMR, Faridi‐Majidi R, Jaafari MR, Mirzaei H. Tumor‐associated macrophages and epithelial–mesenchymal transition in cancer: Nanotechnology comes into view. J Cell Physiol 2018; 233:9223-9236. [DOI: 10.1002/jcp.27027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Roghayyeh Vakili‐Ghartavol
- Department of Medical Nanotechnology School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Reza Mombeiny
- Cellular and Molecular Research Center, Iran University of Medical Sciences Tehran Iran
| | - Arash Salmaninejad
- Drug Applied Research Center, Student Research Committee, Tabriz University of Medical Science Tabriz Iran
- Department of Medical Genetics Faculty of Medicine, Student Research Committee, Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Rezayat Sorkhabadi
- Department of Medical Nanotechnology School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
- Department of Pharmacology School of Medicine, Tehran University of Medical Sciences Tehran Iran
- Department of Toxicology–Pharmacology Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS) Tehran Iran
| | - Reza Faridi‐Majidi
- Department of Medical Nanotechnology School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Nanotechnology School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Hamed Mirzaei
- Department of Biomaterials Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
13
|
Li W, Liu Z, Fontana F, Ding Y, Liu D, Hirvonen JT, Santos HA. Tailoring Porous Silicon for Biomedical Applications: From Drug Delivery to Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703740. [PMID: 29534311 DOI: 10.1002/adma.201703740] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/16/2017] [Indexed: 05/24/2023]
Abstract
In the past two decades, porous silicon (PSi) has attracted increasing attention for its potential biomedical applications. With its controllable geometry, tunable nanoporous structure, large pore volume/high specific surface area, and versatile surface chemistry, PSi shows significant advantages over conventional drug carriers. Here, an overview of recent progress in the use of PSi in drug delivery and cancer immunotherapy is presented. First, an overview of the fabrication of PSi with various geometric structures is provided, with particular focus on how the unique geometry of PSi facilitates its biomedical applications, especially for drug delivery. Second, surface chemistry and modification of PSi are discussed in relation to the strengthening of its performance in drug delivery and bioimaging. Emerging technologies for engineering PSi-based composites are then summarized. Emerging PSi advances in the context of cancer immunotherapy are also highlighted. Overall, very promising research results encourage further exploration of PSi for biomedical applications, particularly in drug delivery and cancer immunotherapy, and future translation of PSi into clinical applications.
Collapse
Affiliation(s)
- Wei Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Yaping Ding
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Dongfei Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| | - Jouni T Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
14
|
Zhang P, Xia J, Luo S. Generation of Well-Defined Micro/Nanoparticles via Advanced Manufacturing Techniques for Therapeutic Delivery. MATERIALS 2018; 11:ma11040623. [PMID: 29670013 PMCID: PMC5951507 DOI: 10.3390/ma11040623] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Micro/nanoparticles have great potentials in biomedical applications, especially for drug delivery. Existing studies identified that major micro/nanoparticle features including size, shape, surface property and component materials play vital roles in their in vitro and in vivo applications. However, a demanding challenge is that most conventional particle synthesis techniques such as emulsion can only generate micro/nanoparticles with a very limited number of shapes (i.e., spherical or rod shapes) and have very loose control in terms of particle sizes. We reviewed the advanced manufacturing techniques for producing micro/nanoparticles with precisely defined characteristics, emphasizing the use of these well-controlled micro/nanoparticles for drug delivery applications. Additionally, to illustrate the vital roles of particle features in therapeutic delivery, we also discussed how the above-mentioned micro/nanoparticle features impact in vitro and in vivo applications. Through this review, we highlighted the unique opportunities in generating controllable particles via advanced manufacturing techniques and the great potential of using these micro/nanoparticles for therapeutic delivery.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Material Processing and Controlling, School of Mechanical Engineering & Automation, Beihang University, Beijing 100191, China.
| | - Junfei Xia
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Sida Luo
- Department of Material Processing and Controlling, School of Mechanical Engineering & Automation, Beihang University, Beijing 100191, China.
| |
Collapse
|
15
|
Abstract
CD59 has been identified as a glycosylphosphatidylinositol-anchored membrane protein that acts as an inhibitor of the formation of the membrane attack complex to regulate complement activation. Recent studies have shown that CD59 is highly expressed in several cancer cell lines and tumor tissues. CD59 also regulates the function, infiltration and phenotypes of a variety of immune cells in the tumor microenvironment. Herein, we summarized recent advances related to the functions and mechanisms of CD59 in the tumor microenvironment. Therapeutic strategies that seek to modulate the functions of CD59 in the tumor microenvironment could be a promising direction for tumor immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
16
|
McInnes SJP, Santos A, Kumeria T. Porous Silicon Particles for Cancer Therapy and Bioimaging. NANOONCOLOGY 2018. [DOI: 10.1007/978-3-319-89878-0_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Leonard F, Ha NP, Sule P, Alexander JF, Volk DE, Lokesh GLR, Liu X, Cirillo JD, Gorenstein DG, Yuan J, Chatterjee S, Graviss EA, Godin B. Thioaptamer targeted discoidal microparticles increase self immunity and reduce Mycobacterium tuberculosis burden in mice. J Control Release 2017; 266:238-247. [PMID: 28987879 DOI: 10.1016/j.jconrel.2017.09.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/29/2017] [Indexed: 12/25/2022]
Abstract
Worldwide, tuberculosis (TB) remains one of the most prevalent infectious diseases causing morbidity and death in >1.5 million patients annually. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, usually resides in the alveolar macrophages. Current tuberculosis treatment methods require more than six months, and low compliance often leads to therapeutic failure and multidrug resistant strain development. Critical to improving TB-therapy is shortening treatment duration and increasing therapeutic efficacy. In this study, we sought to determine if lung hemodynamics and pathological changes in Mtb infected cells can be used for the selective targeting of microparticles to infected tissue(s). Thioaptamers (TA) with CD44 (CD44TA) targeting moiety were conjugated to discoidal silicon mesoporous microparticles (SMP) to enhance accumulation of these agents/carriers in the infected macrophages in the lungs. In vitro, CD44TA-SMP accumulated in macrophages infected with mycobacteria efficiently killing the infected cells and decreasing survival of mycobacteria. In vivo, increased accumulations of CD44TA-SMP were recorded in the lung of M. tuberculosis infected mice as compared to controls. TA-targeted carriers significantly diminished bacterial load in the lungs and caused recruitment of T lymphocytes. Proposed mechanism of action of the designed vector accounts for a combination of increased uptake of particles that leads to infected macrophage death, as well as, activation of cellular immunity by the TA, causing increased T-cell accumulation in the treated lungs. Based on our data with CD44TA-SMP, we anticipate that this drug carrier can open new avenues in TB management.
Collapse
Affiliation(s)
- Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - Ngan P Ha
- Department of Pathology and Genomic Medicine Houston, Houston Methodist Research Institute, TX 77030, United States
| | - Preeti Sule
- Texas A&M Health Science Center, Department of Microbial Pathogenesis and Immunology, Bryan, TX 77807, United States
| | - Jenolyn F Alexander
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - David E Volk
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Ganesh L R Lokesh
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - Jeffrey D Cirillo
- Texas A&M Health Science Center, Department of Microbial Pathogenesis and Immunology, Bryan, TX 77807, United States
| | - David G Gorenstein
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Jinyun Yuan
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, St. Louis, MO 63104, United States
| | - Soumya Chatterjee
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, St. Louis, MO 63104, United States
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine Houston, Houston Methodist Research Institute, TX 77030, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States.
| |
Collapse
|
18
|
Borsoi C, Leonard F, Lee Y, Zaid M, Elganainy D, Alexander JF, Kai M, Liu YT, Kang Y, Liu X, Koay EJ, Ferrari M, Godin B, Yokoi K. Gemcitabine enhances the transport of nanovector-albumin-bound paclitaxel in gemcitabine-resistant pancreatic ductal adenocarcinoma. Cancer Lett 2017; 403:296-304. [PMID: 28687352 DOI: 10.1016/j.canlet.2017.06.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/03/2017] [Accepted: 06/16/2017] [Indexed: 02/06/2023]
Abstract
The mechanism for improved therapeutic efficacy of the combination therapy with nanoparticle albumin-bound paclitaxel (nAb-PTX) and gemcitabine (gem) for pancreatic ductal adenocarcinoma (PDAC) has been ascribed to enhanced gem transport by nAb-PTX. Here, we used an orthotopic mouse model of gem-resistant human PDAC in which increasing gem transport would not improve the efficacy, thus revealing the importance of nAb-PTX transport. We aimed to evaluate therapeutic outcomes and transport of nAb-PTX to PDAC as a result of (1) encapsulating nAb-PTX in multistage nanovectors (MSV); (2) effect of gem on caveolin-1 expression. Treatment with MSV/nAb-PTX + gem was highly efficient in prolonging animal survival in comparison to other therapeutic regimens. MSV/nAb-PTX + gem also caused a substantial increase in tumor PTX accumulation, significantly reduced tumor growth and tumor cell proliferation, and increased apoptosis. Moreover, gem enhanced caveolin-1 expression in vitro and in vivo, thereby improving transport of nAb-PTX to PDAC. This data was confirmed by analysis of PDACs from patients who received gem-based neo-adjuvant chemotherapy. In conclusion, we found that nAb-PTX treatment of gem-resistant PDAC can be enhanced by (1) gem through up-regulation of caveolin-1 and (2) MSV through increasing accumulation of nAb-PTX in the tumor.
Collapse
Affiliation(s)
- Carlotta Borsoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yeonju Lee
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mohamed Zaid
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | - Megumi Kai
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yan Ting Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yaan Kang
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Eugene J Koay
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA.
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA.
| | - Kenji Yokoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA.
| |
Collapse
|
19
|
Kumeria T, McInnes SJP, Maher S, Santos A. Porous silicon for drug delivery applications and theranostics: recent advances, critical review and perspectives. Expert Opin Drug Deliv 2017; 14:1407-1422. [DOI: 10.1080/17425247.2017.1317245] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Tushar Kumeria
- School of Chemical Engineering, The University of Adelaide, Adelaide, Australia
| | - Steven J. P. McInnes
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Mawson Lakes, Australia
| | - Shaheer Maher
- School of Chemical Engineering, The University of Adelaide, Adelaide, Australia
- Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Abel Santos
- School of Chemical Engineering, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), The University of Adelaide, Adelaide, Australia
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, Australia
| |
Collapse
|
20
|
Croissant JG, Fatieiev Y, Khashab NM. Degradability and Clearance of Silicon, Organosilica, Silsesquioxane, Silica Mixed Oxide, and Mesoporous Silica Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1604634. [PMID: 28084658 DOI: 10.1002/adma.201604634] [Citation(s) in RCA: 420] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/13/2016] [Indexed: 05/27/2023]
Abstract
The biorelated degradability and clearance of siliceous nanomaterials have been questioned worldwide, since they are crucial prerequisites for the successful translation in clinics. Typically, the degradability and biocompatibility of mesoporous silica nanoparticles (MSNs) have been an ongoing discussion in research circles. The reason for such a concern is that approved pharmaceutical products must not accumulate in the human body, to prevent severe and unpredictable side-effects. Here, the biorelated degradability and clearance of silicon and silica nanoparticles (NPs) are comprehensively summarized. The influence of the size, morphology, surface area, pore size, and surface functional groups, to name a few, on the degradability of silicon and silica NPs is described. The noncovalent organic doping of silica and the covalent incorporation of either hydrolytically stable or redox- and enzymatically cleavable silsesquioxanes is then described for organosilica, bridged silsesquioxane (BS), and periodic mesoporous organosilica (PMO) NPs. Inorganically doped silica particles such as calcium-, iron-, manganese-, and zirconium-doped NPs, also have radically different hydrolytic stabilities. To conclude, the degradability and clearance timelines of various siliceous nanomaterials are compared and it is highlighted that researchers can select a specific nanomaterial in this large family according to the targeted applications and the required clearance kinetics.
Collapse
Affiliation(s)
- Jonas G Croissant
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology, Thuwal, 23955, Saudi Arabia
| | - Yevhen Fatieiev
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology, Thuwal, 23955, Saudi Arabia
| | - Niveen M Khashab
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology, Thuwal, 23955, Saudi Arabia
| |
Collapse
|
21
|
Nanoplasmonic Quantification of Tumor-derived Extracellular Vesicles in Plasma Microsamples for Diagnosis and Treatment Monitoring. Nat Biomed Eng 2017; 1. [PMID: 28791195 PMCID: PMC5543996 DOI: 10.1038/s41551-016-0021] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumour-derived extracellular vesicles (EVs) are of increasing interest as a resource of diagnostic biomarkers. However, most EV assays require large samples, are time-consuming, low-throughput and costly, and thus impractical for clinical use. Here, we describe a rapid, ultrasensitive and inexpensive nanoplasmon-enhanced scattering (nPES) assay that directly quantifies tumor-derived EVs from as little as 1 μL of plasma. The assay uses the binding of antibody-conjugated gold nanospheres and nanorods to EVs captured by EV-specific antibodies on a sensor chip to produce a local plasmon effect that enhances tumour-derived EV detection sensitivity and specificity. We identified a pancreatic cancer EV biomarker, ephrin type-A receptor 2 (EphA2), and demonstrate that an nPES assay for EphA2-EVs distinguishes pancreatic cancer patients from pancreatitis patients and healthy subjects. EphA2-EVs were also informative in staging tumour progression and in detecting early responses to neoadjuvant therapy, with better performance than a conventional enzyme-linked immunosorbent assay. The nPES assay can be easily refined for clinical use, and readily adapted for diagnosis and monitoring of other conditions with disease-specific EV biomarkers.
Collapse
|
22
|
Abstract
This review focuses on summarizing the existing work about nanomaterial-based cancer immunotherapy in detail.
Collapse
Affiliation(s)
- Lijia Luo
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Rui Shu
- University of Chinese Academy of Sciences
- Beijing 100049
- China
- Key Laboratory of Marine Materials and Related Technology
- CAS & Ningbo Institute of Materials Technology and Engineering
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| |
Collapse
|
23
|
Leonard F, Curtis LT, Yesantharao P, Tanei T, Alexander JF, Wu M, Lowengrub J, Liu X, Ferrari M, Yokoi K, Frieboes HB, Godin B. Enhanced performance of macrophage-encapsulated nanoparticle albumin-bound-paclitaxel in hypo-perfused cancer lesions. NANOSCALE 2016; 8:12544-52. [PMID: 26818212 PMCID: PMC4919151 DOI: 10.1039/c5nr07796f] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Hypovascularization in tumors such as liver metastases originating from breast and other organs correlates with poor chemotherapeutic response and higher mortality. Poor prognosis is linked to impaired transport of both low- and high-molecular weight drugs into the lesions and to high washout rate. Nanoparticle albumin-bound-paclitaxel (nAb-PTX) has demonstrated benefits in clinical trials when compared to paclitaxel and docetaxel. However, its therapeutic efficacy for breast cancer liver metastasis is disappointing. As macrophages are the most abundant cells in the liver tumor microenvironment, we design a multistage system employing macrophages to deliver drugs into hypovascularized metastatic lesions, and perform in vitro, in vivo, and in silico evaluation. The system encapsulates nAb-PTX into nanoporous biocompatible and biodegradable multistage vectors (MSV), thus promoting nAb-PTX retention in macrophages. We develop a 3D in vitro model to simulate clinically observed hypo-perfused tumor lesions surrounded by macrophages. This model enables evaluation of nAb-PTX and MSV-nab PTX efficacy as a function of transport barriers. Addition of macrophages to this system significantly increases MSV-nAb-PTX efficacy, revealing the role of macrophages in drug transport. In the in vivo model, a significant increase in macrophage number, as compared to unaffected liver, is observed in mice, confirming the in vitro findings. Further, a mathematical model linking drug release and retention from macrophages is implemented to project MSV-nAb-PTX efficacy in a clinical setting. Based on macrophage presence detected via liver tumor imaging and biopsy, the proposed experimental/computational approach could enable prediction of MSV-nab PTX performance to treat metastatic cancer in the liver.
Collapse
Affiliation(s)
- Fransisca Leonard
- Houston Methodist Research Institute, Department of Nanomedicine, R8-213, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kirui DK, Ferrari M. Intravital Microscopy Imaging Approaches for Image-Guided Drug Delivery Systems. Curr Drug Targets 2016; 16:528-41. [PMID: 25901526 DOI: 10.2174/1389450116666150330114030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/10/2014] [Accepted: 03/13/2015] [Indexed: 12/31/2022]
Abstract
Rapid technical advances in the field of non-linear microscopy have made intravital microscopy a vital pre-clinical tool for research and development of imaging-guided drug delivery systems. The ability to dynamically monitor the fate of macromolecules in live animals provides invaluable information regarding properties of drug carriers (size, charge, and surface coating), physiological, and pathological processes that exist between point-of-injection and the projected of site of delivery, all of which influence delivery and effectiveness of drug delivery systems. In this Review, we highlight how integrating intravital microscopy imaging with experimental designs (in vitro analyses and mathematical modeling) can provide unique information critical in the design of novel disease-relevant drug delivery platforms with improved diagnostic and therapeutic indexes. The Review will provide the reader an overview of the various applications for which intravital microscopy has been used to monitor the delivery of diagnostic and therapeutic agents and discuss some of their potential clinical applications.
Collapse
Affiliation(s)
| | - Mauro Ferrari
- Houston Methodist Research Institute, Department of NanoMedicine, 6670 Bertner Avenue, MS R8-460, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Tanei T, Leonard F, Liu X, Alexander JF, Saito Y, Ferrari M, Godin B, Yokoi K. Redirecting Transport of Nanoparticle Albumin-Bound Paclitaxel to Macrophages Enhances Therapeutic Efficacy against Liver Metastases. Cancer Res 2016; 76:429-39. [PMID: 26744528 DOI: 10.1158/0008-5472.can-15-1576] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022]
Abstract
Current treatments for liver metastases arising from primary breast and lung cancers are minimally effective. One reason for this unfavorable outcome is that liver metastases are poorly vascularized, limiting the ability to deliver therapeutics from the systemic circulation to lesions. Seeking to enhance transport of agents into the tumor microenvironment, we designed a system in which nanoparticle albumin-bound paclitaxel (nAb-PTX) is loaded into a nanoporous solid multistage nanovector (MSV) to enable the passage of the drug through the tumor vessel wall and enhance its interaction with liver macrophages. MSV enablement increased nAb-PTX efficacy and survival in mouse models of breast and lung liver metastasis. MSV-nAb-PTX also augmented the accumulation of paclitaxel and MSV in the liver, specifically in macrophages, whereas paclitaxel levels in the blood were unchanged after administering MSV-nAb-PTX or nAb-PTX. In vitro studies demonstrated that macrophages treated with MSV-nAb-PTX remained viable and were able to internalize, retain, and release significantly higher quantities of paclitaxel compared with treatment with nAb-PTX. The cytotoxic potency of the released paclitaxel was also confirmed in tumor cells cultured with the supernatants of macrophage treated with MSV-nAB-PTX. Collectively, our findings showed how redirecting nAb-PTX to liver macrophages within the tumor microenvironment can elicit a greater therapeutic response in patients with metastatic liver cancer, without increasing systemic side effects.
Collapse
Affiliation(s)
- Tomonori Tanei
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Jenolyn F Alexander
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Yuki Saito
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas.
| | - Kenji Yokoi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas.
| |
Collapse
|
26
|
Wolfram J, Shen H, Ferrari M. Multistage vector (MSV) therapeutics. J Control Release 2015; 219:406-415. [PMID: 26264836 PMCID: PMC4656100 DOI: 10.1016/j.jconrel.2015.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
One of the greatest challenges in the field of medicine is obtaining controlled distribution of systemically administered therapeutic agents within the body. Indeed, biological barriers such as physical compartmentalization, pressure gradients, and excretion pathways adversely affect localized delivery of drugs to pathological tissue. The diverse nature of these barriers requires the use of multifunctional drug delivery vehicles that can overcome a wide range of sequential obstacles. In this review, we explore the role of multifunctionality in nanomedicine by primarily focusing on multistage vectors (MSVs). The MSV is an example of a promising therapeutic platform that incorporates several components, including a microparticle, nanoparticles, and small molecules. In particular, these components are activated in a sequential manner in order to successively address transport barriers.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
27
|
Yokoi K, Chan D, Kojic M, Milosevic M, Engler D, Matsunami R, Tanei T, Saito Y, Ferrari M, Ziemys A. Liposomal doxorubicin extravasation controlled by phenotype-specific transport properties of tumor microenvironment and vascular barrier. J Control Release 2015; 217:293-9. [PMID: 26409121 PMCID: PMC4623952 DOI: 10.1016/j.jconrel.2015.09.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/04/2015] [Accepted: 09/22/2015] [Indexed: 11/15/2022]
Abstract
Although nanotherapeutics can be advantageous over free chemotherapy, the benefits of drug vectors can vary from patient to patient based on differences in tumor microenvironments. Although systemic pharmacokinetics (PK) of drugs is considered as the major determinant of its efficacy in clinics, recent clinical and basic research indicates that tumor-based PK can provide better representation of therapeutic efficacy. Here, we have studied the role of the tumor extravascular tissue in the extravasation kinetics of doxorubicin (DOX), delivered by pegylated liposomes (PLD), to murine lung (3LL) and breast (4T1) tumors. We found that phenotypically different 3LL and 4T1 tumors shared the similar systemic PK, but DOX extravasation in the tumor extravascular tissue was substantially different. Liquid chromatography-mass spectrometry (LC-MS) measurements showed that DOX fluorescence imaged by fluorescence microscopy could be used as a marker to study tumor microenvironment PK, providing an excellent match to DOX kinetics in tumor tissues. Our results also suggest that therapeutic responses can be closely related to the interplay of concentration levels and exposure times in extravascular tissue of tumors. Finally, the computational model of capillary drug transport showed that internalization of drug vectors was critical and could lead to 2-3 orders of magnitude more efficient drug delivery into the extravascular tissue, compared to non-internalized localization of drug vectors, and explaining the differences in therapeutic efficacy between the 3LL and 4T1 tumors. These results show that drug transport and partitioning characteristics can be phenotype- and microenvironment-dependent and are highly important in drug delivery and therapeutic efficacy.
Collapse
Affiliation(s)
- Kenji Yokoi
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Diana Chan
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Milos Kojic
- The Houston Methodist Research Institute, Houston, TX 77030, United States; Belgrade Metropolitan University, Research and Development Center for Bioengineering, 3400 Kragujevac, Serbia; Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Miljan Milosevic
- The Houston Methodist Research Institute, Houston, TX 77030, United States; Belgrade Metropolitan University, Research and Development Center for Bioengineering, 3400 Kragujevac, Serbia
| | - David Engler
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Rise Matsunami
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Tomonori Tanei
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Yuki Saito
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Mauro Ferrari
- The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Arturas Ziemys
- The Houston Methodist Research Institute, Houston, TX 77030, United States.
| |
Collapse
|
28
|
Torres Andón F, Alonso MJ. Nanomedicine and cancer immunotherapy – targeting immunosuppressive cells. J Drug Target 2015; 23:656-71. [DOI: 10.3109/1061186x.2015.1073295] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Amoozgar Z, Goldberg MS. Targeting myeloid cells using nanoparticles to improve cancer immunotherapy. Adv Drug Deliv Rev 2015; 91:38-51. [PMID: 25280471 DOI: 10.1016/j.addr.2014.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 12/23/2022]
Abstract
While nanoparticles have traditionally been used to deliver cytotoxic drugs directly to tumors to induce cancer cell death, emerging data suggest that nanoparticles are likely to generate a larger impact on oncology through the delivery of agents that can stimulate antitumor immunity. Tumor-targeted nanocarriers have generally been used to localize chemotherapeutics to tumors and thus decrease off-target toxicity while enhancing efficacy. Challengingly, tumor heterogeneity and evolution render tumor-intrinsic approaches likely to succumb to relapse. The immune system offers exquisite specificity, cytocidal potency, and long-term activity that leverage an adaptive memory response. For this reason, the ability to manipulate immune cell specificity and function would be desirable, and nanoparticles represent an exciting means by which to perform such manipulation. Dendritic cells and tumor-associated macrophages are cells of the myeloid lineage that function as natural phagocytes, so they naturally take up nanoparticles. Dendritic cells direct the specificity and potency of cellular immune responses that can be targeted for cancer vaccines. Herein, we discuss the specific criteria needed for efficient vaccine design, including but not limited to the route of administration, size, morphology, surface charge, targeting ligands, and nanoparticle composition. In contrast, tumor-associated macrophages are critical mediators of immunosuppression whose trans-migratory abilities can be exploited to localize therapeutics to the tumor core and which can be directly targeted for elimination or for repolarization to a tumor suppressive phenotype. It is likely that a combination of targeting dendritic cells to stimulate antitumor immunity and tumor-associated macrophages to reduce immune suppression will impart significant benefits and result in durable antitumor responses.
Collapse
|
30
|
Abstract
Recent developments in nanotechnology have brought new approaches to cancer diagnosis and therapy. While enhanced permeability and retention effect (EPR) promotes nanoparticle (NP) extravasation, the abnormal tumor vasculature, high interstitial pressure and dense stroma structure limit homogeneous intratumoral distribution of NP and compromise their imaging and therapeutic effect. Moreover, heterogeneous distribution of NP in nontumor-stroma cells damages the nontumor cells, and interferes with tumor-stroma crosstalk. This can lead to inhibition of tumor progression, but can also paradoxically induce acquired resistance and facilitate tumor cell proliferation and metastasis. Overall, the tumor microenvironment plays a crucial, yet controversial role in regulating NP distribution and their biological effects. In this review, we summarize recent studies on the stroma barriers for NP extravasation, and discuss the consequential effects of NP distribution in stroma cells. We also highlight design considerations to improve NP delivery and propose potential combinatory strategies to overcome acquired resistance induced by damaged stroma cells.
Collapse
Affiliation(s)
- Lei Miao
- Division of Molecular Pharmaceutics and Center of Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | |
Collapse
|
31
|
Linton SS, Sherwood SG, Drews KC, Kester M. Targeting cancer cells in the tumor microenvironment: opportunities and challenges in combinatorial nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:208-22. [PMID: 26153136 DOI: 10.1002/wnan.1358] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/01/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022]
Abstract
Cancer therapies of the future will rely on synergy between drugs delivered in combination to achieve both maximum efficacy and decreased toxicity. Nanoscale drug delivery vehicles composed of highly tunable nanomaterials ('nanocarriers') represent the most promising approach to achieve simultaneous, cell-selective delivery of synergistic ratios of combinations of drugs within solid tumors. Nanocarriers are currently being used to co-encapsulate and deliver synergistic ratios of multiple anticancer drugs to target cells within solid tumors. Investigators exploit the unique environment associated with solid tumors, termed the tumor microenvironment (TME), to make 'smart' nanocarriers. These sophisticated nanocarriers exploit the pathological conditions in the TME, thereby creating highly targeted nanocarriers that release their drug payload in a spatially and temporally controlled manner. The translational and commercial potential of nanocarrier-based combinatorial nanomedicines in cancer therapy is now a reality as several companies have initiated human clinical trials.
Collapse
Affiliation(s)
- Samuel S Linton
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, USA
| | - Samantha G Sherwood
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Kelly C Drews
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
32
|
Kovalainen M, Mönkäre J, Riikonen J, Pesonen U, Vlasova M, Salonen J, Lehto VP, Järvinen K, Herzig KH. Novel delivery systems for improving the clinical use of peptides. Pharmacol Rev 2015; 67:541-61. [PMID: 26023145 DOI: 10.1124/pr.113.008367] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Peptides have long been recognized as a promising group of therapeutic substances to treat various diseases. Delivery systems for peptides have been under development since the discovery of insulin for the treatment of diabetes. The challenge of using peptides as drugs arises from their poor bioavailability resulting from the low permeability of biological membranes and their instability. Currently, subcutaneous injection is clinically the most common administration route for peptides. This route is cost-effective and suitable for self-administration, and the development of appropriate dosing equipment has made performing the repeated injections relatively easy; however, only few clinical subcutaneous peptide delivery systems provide sustained peptide release. As a result, frequent injections are needed, which may cause discomfort and additional risks resulting from a poor administration technique. Controlled peptide delivery systems, able to provide required therapeutic plasma concentrations over an extended period, are needed to increase peptide safety and patient compliancy. In this review, we summarize the current peptidergic drugs, future developments, and parenteral peptide delivery systems. Special emphasis is given to porous silicon, a novel material in peptide delivery. Biodegradable and biocompatible porous silicon possesses some unique properties, such as the ability to carry exceptional high peptide payloads and to modify peptide release extensively. We have successfully developed porous silicon as a carrier material for improved parenteral peptide delivery. Nanotechnology, with its different delivery systems, will enable better use of peptides in several therapeutic applications in the near future.
Collapse
Affiliation(s)
- Miia Kovalainen
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Juha Mönkäre
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Joakim Riikonen
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Ullamari Pesonen
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Maria Vlasova
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Jarno Salonen
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Vesa-Pekka Lehto
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Kristiina Järvinen
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| | - Karl-Heinz Herzig
- Institute of Biomedicine and Biocenter of Oulu, Faculty of Medicine (M.K., K.-H.H.) and Medical Research Center Oulu and Oulu University Hospital (K.-H.H.), Oulu, Finland; Department of Applied Physics, Faculty of Science and Forestry (J.R.), Department of Applied Physics, Faculty of Science and Forestry (V.-P.L.), and School of Pharmacy, Faculty of Health Sciences (M.V., K.J.), University of Eastern Finland, Kuopio, Finland; Department of Pharmacology, Drug Development and Therapeutics (U.P.), and Department of Physics and Astronomy, Faculty of Mathematics and Natural Sciences (J.S.), University of Turku, Finland; and Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (J.M.)
| |
Collapse
|
33
|
Peng F, Cao Z, Ji X, Chu B, Su Y, He Y. Silicon nanostructures for cancer diagnosis and therapy. Nanomedicine (Lond) 2015; 10:2109-23. [DOI: 10.2217/nnm.15.53] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The emergence of nanotechnology suggests new and exciting opportunities for early diagnosis and therapy of cancer. During the recent years, silicon-based nanomaterials featuring unique properties have received great attention, showing high promise for myriad biological and biomedical applications. In this review, we will particularly summarize latest representative achievements on the development of silicon nanostructures as a powerful platform for cancer early diagnosis and therapy. First, we introduce the silicon nanomaterial-based biosensors for detecting cancer markers (e.g., proteins, tumor-suppressor genes and telomerase activity, among others) with high sensitivity and selectivity under molecular level. Then, we summarize in vitro and in vivo applications of silicon nanostructures as efficient nanoagents for cancer therapy. Finally, we discuss the future perspective of silicon nanostructures for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Fei Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Zhaohui Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Xiaoyuan Ji
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Binbin Chu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Yuanyuan Su
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Yao He
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| |
Collapse
|
34
|
Tzur-Balter A, Shatsberg Z, Beckerman M, Segal E, Artzi N. Mechanism of erosion of nanostructured porous silicon drug carriers in neoplastic tissues. Nat Commun 2015; 6:6208. [PMID: 25670235 PMCID: PMC4339882 DOI: 10.1038/ncomms7208] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/06/2015] [Indexed: 01/28/2023] Open
Abstract
Nanostructured porous silicon (PSi) is emerging as a promising platform for drug delivery owing to its biocompatibility, degradability and high surface area available for drug loading. The ability to control PSi structure, size and porosity enables programming its in vivo retention, providing tight control over embedded drug release kinetics. In this work, the relationship between the in vitro and in vivo degradation of PSi under (pre)clinically relevant conditions, using breast cancer mouse model, is defined. We show that PSi undergoes enhanced degradation in diseased environment compared with healthy state, owing to the upregulation of reactive oxygen species (ROS) in the tumour vicinity that oxidize the silicon scaffold and catalyse its degradation. We further show that PSi degradation in vitro and in vivo correlates in healthy and diseased states when ROS-free or ROS-containing media are used, respectively. Our work demonstrates that understanding the governing mechanisms associated with specific tissue microenvironment permits predictive material performance. The degradation of materials used in biological applications has an important bearing on their long term performance. Here, the authors show how porous silicon nanoparticle degradation can be accelerated in vivo through the influence of local tissue pathology, likely influencing drug delivery performance.
Collapse
Affiliation(s)
- Adi Tzur-Balter
- The Inter-Departmental Program of Biotechnology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Zohar Shatsberg
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Margarita Beckerman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ester Segal
- 1] Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel [2] Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Natalie Artzi
- 1] Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA [2] Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, USA
| |
Collapse
|
35
|
Montalti M, Cantelli A, Battistelli G. Nanodiamonds and silicon quantum dots: ultrastable and biocompatible luminescent nanoprobes for long-term bioimaging. Chem Soc Rev 2015; 44:4853-921. [DOI: 10.1039/c4cs00486h] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ultra-stability and low-toxicity of silicon quantum dots and fluorescent nanodiamonds for long-termin vitroandin vivobioimaging are demonstrated.
Collapse
Affiliation(s)
- M. Montalti
- Department of Chemistry “G. Ciamician”
- University of Bologna
- Bologna
- Italy
| | - A. Cantelli
- Department of Chemistry “G. Ciamician”
- University of Bologna
- Bologna
- Italy
| | - G. Battistelli
- Department of Chemistry “G. Ciamician”
- University of Bologna
- Bologna
- Italy
| |
Collapse
|
36
|
Sun J, Kim DH, Guo Y, Teng Z, Li Y, Zheng L, Zhang Z, Larson AC, Lu G. A c(RGDfE) conjugated multi-functional nanomedicine delivery system for targeted pancreatic cancer therapy. J Mater Chem B 2014; 3:1049-1058. [PMID: 32261983 DOI: 10.1039/c4tb01402b] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pancreatic cancer is one of the five most lethal malignancies and has a poor prognosis due to its abundant stromal barriers and lack of effective available therapies. Although gemcitabine has been used as a standard therapy for several decades, there has been little progress in the improvement of the 5 year survive rate due to the low targeting efficiency for pancreatic cancer cells. To achieve a targeted delivery of gemcitabine to pancreatic cancer cells, we have developed a c(RGDfE) [cyclic (Arg-Gly-Asp-d-Phe-Glu)] conjugated multi-functional nanomedicine delivery system composed of a magnetic core and mesoporous silica shell. These magnetic mesoporous nanoparticles demonstrated sufficient relaxivity properties for detection with magnetic resonance imaging (MRI). These c(RGDfE) peptide conjugated magnetic mesoporous silica nanoparticles [c(RGDfE)-pMMSNs] can target pancreatic cancer cells and increase cellular uptake in human pancreatic cancer cell lines that overexpress integrin ανβ3. Gemcitabine loaded c(RGDfE)-pMMSNs were most efficiently targeted to pancreatic cancer cells (BxPC-3). Growth inhibition of the BxPC-3 cell line was achieved in a time dependent manner consistent with observed drug release behavior. Intracellular targeted gemcitabine delivery using c(RGDfE)-pMMSNs offers a promising approach for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jing Sun
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, P.R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yokoi K, Kojic M, Milosevic M, Tanei T, Ferrari M, Ziemys A. Capillary-wall collagen as a biophysical marker of nanotherapeutic permeability into the tumor microenvironment. Cancer Res 2014; 74:4239-46. [PMID: 24853545 PMCID: PMC4134692 DOI: 10.1158/0008-5472.can-13-3494] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The capillary wall is the chief barrier to tissue entry of therapeutic nanoparticles, thereby dictating their efficacy. Collagen fibers are an important component of capillary walls, affecting leakiness in healthy or tumor vasculature. Using a computational model along with in vivo systems, we compared how collagen structure affects the diffusion flux of a 1-nm chemotherapeutic molecule [doxorubicin (DOX)] and an 80-nm chemotherapy-loaded pegylated liposome (DOX-PLD) in tumor vasculature. We found a direct correlation between the collagen content around a tumor vessel to the permeability of that vessel permeability to DOX-PLD, indicating that collagen content may offer a biophysical marker of extravasation potential of liposomal drug formulations. Our results also suggested that while pharmacokinetics determined the delivery of DOX and DOX-PLD to the same tumor phenotype, collagen content determined the extravasation of DOX-PLD to different tumor phenotypes. Transport physics may provide a deeper view into how nanotherapeutics cross biological barriers, possibly helping explain the balance between biological and physical aspects of drug delivery.
Collapse
Affiliation(s)
- Kenji Yokoi
- The Houston Methodist Research Institute; The University of Texas MD Anderson Cancer Center, Houston Texas; and
| | - Milos Kojic
- The Houston Methodist Research Institute; Belgrade Metropolitan University, Research and Development Center for Bioengineering, Kragujevac, Serbia
| | - Miljan Milosevic
- Belgrade Metropolitan University, Research and Development Center for Bioengineering, Kragujevac, Serbia
| | | | | | | |
Collapse
|
38
|
Tölli MA, Ferreira MPA, Kinnunen SM, Rysä J, Mäkilä EM, Szabó Z, Serpi RE, Ohukainen PJ, Välimäki MJ, Correia AMR, Salonen JJ, Hirvonen JT, Ruskoaho HJ, Santos HA. In vivo biocompatibility of porous silicon biomaterials for drug delivery to the heart. Biomaterials 2014; 35:8394-405. [PMID: 24985734 DOI: 10.1016/j.biomaterials.2014.05.078] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 05/28/2014] [Indexed: 11/18/2022]
Abstract
Myocardial infarction (MI), commonly known as a heart attack, is the irreversible necrosis of heart muscle secondary to prolonged ischemia, which is an increasing problem in terms of morbidity, mortality and healthcare costs worldwide. Along with the idea to develop nanocarriers that efficiently deliver therapeutic agents to target the heart, in this study, we aimed to test the in vivo biocompatibility of different sizes of thermally hydrocarbonized porous silicon (THCPSi) microparticles and thermally oxidized porous silicon (TOPSi) micro and nanoparticles in the heart tissue. Despite the absence or low cytotoxicity, both particle types showed good in vivo biocompatibility, with no influence on hematological parameters and no considerable changes in cardiac function before and after MI. The local injection of THCPSi microparticles into the myocardium led to significant higher activation of inflammatory cytokine and fibrosis promoting genes compared to TOPSi micro and nanoparticles; however, both particles showed no significant effect on myocardial fibrosis at one week post-injection. Our results suggest that THCPSi and TOPSi micro and nanoparticles could be applied for cardiac delivery of therapeutic agents in the future, and the PSi biomaterials might serve as a promising platform for the specific treatment of heart diseases.
Collapse
Affiliation(s)
- Marja A Tölli
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland
| | - Mónica P A Ferreira
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Sini M Kinnunen
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jaana Rysä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Ermei M Mäkilä
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Zoltán Szabó
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland
| | - Raisa E Serpi
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland; Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, FI-90014 Oulu, Finland
| | - Pauli J Ohukainen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland
| | - Mika J Välimäki
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland
| | - Alexandra M R Correia
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jarno J Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jouni T Hirvonen
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Heikki J Ruskoaho
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, FI-90014 Oulu, Finland; Division of Pharmacology and Pharmacotherapy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
39
|
Kirui DK, Mai J, Palange AL, Qin G, van de Ven AL, Liu X, Shen H, Ferrari M. Transient mild hyperthermia induces E-selectin mediated localization of mesoporous silicon vectors in solid tumors. PLoS One 2014; 9:e86489. [PMID: 24558362 PMCID: PMC3928046 DOI: 10.1371/journal.pone.0086489] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 12/09/2013] [Indexed: 01/03/2023] Open
Abstract
Background Hyperthermia treatment has been explored as a strategy to overcome biological barriers that hinder effective drug delivery in solid tumors. Most studies have used mild hyperthermia treatment (MHT) to target the delivery of thermo-sensitive liposomes carriers. Others have studied its application to permeabilize tumor vessels and improve tumor interstitial transport. However, the role of MHT in altering tumor vessel interfacial and adhesion properties and its relationship to improved delivery has not been established. In the present study, we evaluated effects of MHT treatment on tumor vessel flow dynamics and expression of adhesion molecules and assessed enhancement in particle localization using mesoporous silicon vectors (MSVs). We also determined the optimal time window at which maximal accumulation occur. Results In this study, using intravital microscopy analyses, we showed that temporal mild hyperthermia (∼1 W/cm2) amplified delivery and accumulation of MSVs in orthotopic breast cancer tumors. The number of discoidal MSVs (1000×400 nm) adhering to tumor vasculature increased 6-fold for SUM159 tumors and 3-fold for MCF-7 breast cancer tumors. By flow chamber experiments and Western blotting, we established that a temporal increase in E-selectin expression correlated with enhanced particle accumulation. Furthermore, MHT treatment was shown to increase tumor perfusion in a time-dependent fashion. Conclusions Our findings reveal that well-timed mild hyperthermia treatment can transiently elevate tumor transport and alter vascular adhesion properties and thereby provides a means to enhance tumor localization of non-thermally sensitive particles such as MSVs. Such enhancement in accumulation could be leveraged to increase therapeutic efficacy and reduce drug dosing in cancer therapy.
Collapse
Affiliation(s)
- Dickson K. Kirui
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Juahua Mai
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Anna-Lisa Palange
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Guoting Qin
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Anne L. van de Ven
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Xuewu Liu
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Haifa Shen
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
- Department of Cell and Development Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Mauro Ferrari
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
- Department of Internal Medicine, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
40
|
Yokoi K, Tanei T, Godin B, van de Ven AL, Hanibuchi M, Matsunoki A, Alexander J, Ferrari M. Serum biomarkers for personalization of nanotherapeutics-based therapy in different tumor and organ microenvironments. Cancer Lett 2013; 345:48-55. [PMID: 24370567 DOI: 10.1016/j.canlet.2013.11.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/14/2013] [Accepted: 11/20/2013] [Indexed: 12/11/2022]
Abstract
Enhanced permeation and retention (EPR) effect, the mechanism by which nanotherapeutics accumulate in tumors, varies in patients based on differences in the tumor and organ microenvironment. Surrogate biomarkers for the EPR effect will aid in selecting patients who will accumulate higher amounts of nanotherapeutics and show better therapeutic efficacy. Our data suggest that the differences in the vascular permeability and pegylated liposomal doxorubicin (PLD) accumulation are tumor type as well as organ-specific and significantly correlated with the relative ratio of MMP-9 to TIMP-1 in the circulation, supporting development of these molecules as biomarkers for the personalization of nanoparticle-based therapy.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/pharmacokinetics
- Biomarkers, Tumor/blood
- Brain Neoplasms/blood
- Brain Neoplasms/drug therapy
- Brain Neoplasms/metabolism
- Brain Neoplasms/secondary
- Capillary Permeability
- Doxorubicin/administration & dosage
- Doxorubicin/analogs & derivatives
- Doxorubicin/pharmacokinetics
- Drug Delivery Systems
- Female
- Liver Neoplasms, Experimental/blood
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/metabolism
- Matrix Metalloproteinase 9/blood
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Nanoparticles/administration & dosage
- Nanoparticles/metabolism
- Neoplasms, Experimental/blood
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Polyethylene Glycols/administration & dosage
- Polyethylene Glycols/pharmacokinetics
- Tissue Inhibitor of Metalloproteinase-1/blood
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kenji Yokoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Street, Houston, TX 77030, USA; Department of Cancer Biology, Cancer Metastasis Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Tomonori Tanei
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Street, Houston, TX 77030, USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Street, Houston, TX 77030, USA
| | - Anne L van de Ven
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Street, Houston, TX 77030, USA
| | - Masaki Hanibuchi
- Department of Cancer Biology, Cancer Metastasis Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Aika Matsunoki
- Department of Cancer Biology, Cancer Metastasis Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jenolyn Alexander
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Street, Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Street, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Srinivasan S, Alexander JF, Driessen WH, Leonard F, Ye H, Liu X, Arap W, Pasqualini R, Ferrari M, Godin B. Bacteriophage Associated Silicon Particles: Design and Characterization of a Novel Theranostic Vector with Improved Payload Carrying Potential. J Mater Chem B 2013; 1:10.1039/C3TB20595A. [PMID: 24409342 PMCID: PMC3881592 DOI: 10.1039/c3tb20595a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There has been extensive research on the use of nanovectors for cancer therapy. Targeted delivery of nanotherapeutics necessitates two important characteristics; the ability to accumulate at the disease locus after overcoming sequential biological barriers and the ability to carry a substantial therapeutic payload. Successful combination of the above two features is challenging, especially in solid porous materials where chemical conjugation of targeting entities on the particle surface will generally prevent successful loading of the therapeutic substance. In this study, we propose a novel strategy for decorating the surface of mesoporous silicon particles with targeting entities (bacteriophage) and gold nanoparticles (AuNP) while maintaining their payload carrying potential. The resulting Bacteriophage Associated Silicon Particles (BASP) demonstrates efficient encapsulation of macromolecules and therapeutic nanoparticles into the porous structures. In vitro targeting data show enhanced targeting efficiency with about four orders of magnitude lower concentration of bacteriophage. In vivo targeting data suggest that BASP maintain their integrity following intravenous administration in mice and display up to three fold higher accumulation in the tumor.
Collapse
Affiliation(s)
| | | | - Wouter H. Driessen
- The University of Texas MD Anderson Cancer Center, David H. Koch Center, Houston, Texas, USA
| | | | - Hu Ye
- The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Xuewu Liu
- The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Wadih Arap
- The University of Texas MD Anderson Cancer Center, David H. Koch Center, Houston, Texas, USA
| | - Renata Pasqualini
- The University of Texas MD Anderson Cancer Center, David H. Koch Center, Houston, Texas, USA
| | - Mauro Ferrari
- The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Biana Godin
- The Methodist Hospital Research Institute, Houston, Texas, USA
| |
Collapse
|