151
|
Wang Y, Zhang Y, Wang J, Liang XJ. Aggregation-induced emission (AIE) fluorophores as imaging tools to trace the biological fate of nano-based drug delivery systems. Adv Drug Deliv Rev 2019; 143:161-176. [PMID: 30529308 DOI: 10.1016/j.addr.2018.12.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/18/2018] [Accepted: 12/03/2018] [Indexed: 01/10/2023]
Abstract
The vigorous development of nanotechnology has been accompanied by an equally strong interest and research efforts in nano-based drug delivery systems (NDDSs). However, only a few NDDSs have been translated into clinic thus far. One of the important hurdles is the lack of tools to comprehensively and directly trace the biological fate of NDDSs. Recently, aggregation-induced emission (AIE) fluorophores have emerged as attractive bioimaging tools due to flexible controllability, negligible toxicity and superior photostability. Herein, we recapitulate the current advances in the application of AIE fluorophores to monitor NDDSs both in vitro and in vivo. Particularly, we discuss the cellular fates of self-indicating and stimuli-responsive NDDSs with AIE fluorophores. Moreover, we highlight the in vivo application of AIE agents on the long-term tracking of therapeutics and the multi-modal monitoring of diagnostics in NDDSs. Challenges and opportunities in AIE-guided exploration of NDDSs are also discussed in detail.
Collapse
Affiliation(s)
- Yufei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuxuan Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinjin Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
152
|
Donahue ND, Acar H, Wilhelm S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv Drug Deliv Rev 2019; 143:68-96. [PMID: 31022434 DOI: 10.1016/j.addr.2019.04.008] [Citation(s) in RCA: 574] [Impact Index Per Article: 95.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/14/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
Nanoparticle-based therapeutics and diagnostics are commonly referred to as nanomedicine and may significantly impact the future of healthcare. However, the clinical translation of these technologies is challenging. One of these challenges is the efficient delivery of nanoparticles to specific cell populations and subcellular targets in the body to elicit desired biological and therapeutic responses. It is critical for researchers to understand the fundamental concepts of how nanoparticles interact with biological systems to predict and control in vivo nanoparticle transport for improved clinical benefit. In this overview article, we review and discuss cellular internalization pathways, summarize the field`s understanding of how nanoparticle physicochemical properties affect cellular interactions, and explore and discuss intracellular nanoparticle trafficking and kinetics. Our overview may provide a valuable resource for researchers and may inspire new studies to expand our current understanding of nanotechnology-biology interactions at cellular and subcellular levels with the goal to improve clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Nathan D Donahue
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Handan Acar
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States; Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States.
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States; Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States.
| |
Collapse
|
153
|
Nanotechnology in the diagnosis and treatment of lung cancer. Pharmacol Ther 2019; 198:189-205. [PMID: 30796927 DOI: 10.1016/j.pharmthera.2019.02.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
Abstract
Lung cancer is an umbrella term for a subset of heterogeneous diseases that are collectively responsible for the most cancer-related deaths worldwide. Despite the tremendous progress made in understanding lung tumour biology, advances in early diagnosis, multimodal therapy and deciphering molecular mechanisms of drug resistance, overall curative outcomes remain low, especially in metastatic disease. Nanotechnology, in particular nanoparticles (NPs), continue to progressively impact the way by which tumours are diagnosed and treated. The unique physicochemical properties of materials at the nanoscale grant access to a diverse molecular toolkit that can be manipulated for use in respiratory oncology. This realisation has resulted in several clinically approved NP formulations and many more in clinical trials. However, NPs are not a panacea and have yet to be utilised to maximal effect in lung cancer, and medicine in a wider context. This review serves to: describe the complexity of lung cancer, the current diagnostic and therapeutic environment, and highlight the recent advancements of nanotechnology based approaches in diagnosis and treatment of respiratory malignancies. Finally, a brief outlook on the future directions of nanomedicine is provided; presently the full potential of the field is yet to be realised. By gleaning lessons and integrating advancements from neighbouring disciplines, nanomedicine can be elevated to a position where the current barriers that stymie full clinical impact are lifted.
Collapse
|
154
|
Abstract
Molecular imaging is a vital tool to non-invasively measure nanoparticle delivery to solid tumors. Despite the myriad of nanoparticles studied for cancer, successful applications of nanoparticles in humans is limited by inconsistent and ineffective delivery. Successful nanoparticle delivery in preclinical models is often attributed to enhanced permeability and retention (EPR)-a set of conditions that is heterogeneous and transient in patients. Thus, researchers are evaluating therapeutic strategies to modify nanoparticle delivery, particularly treatments which have demonstrated effects on EPR conditions. Imaging nanoparticle distribution provides a means to measure the effects of therapeutic intervention on nanoparticle delivery to solid tumors. This review focuses on the utility of imaging to measure treatment-induced changes in nanoparticle delivery to tumors and provides preclinical examples studying a broad range of therapeutic interventions.
Collapse
|
155
|
Satpathy M, Wang L, Zielinski RJ, Qian W, Wang YA, Mohs AM, Kairdolf BA, Ji X, Capala J, Lipowska M, Nie S, Mao H, Yang L. Targeted Drug Delivery and Image-Guided Therapy of Heterogeneous Ovarian Cancer Using HER2-Targeted Theranostic Nanoparticles. Theranostics 2019; 9:778-795. [PMID: 30809308 PMCID: PMC6376473 DOI: 10.7150/thno.29964] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/19/2018] [Indexed: 01/30/2023] Open
Abstract
Cancer heterogeneity and drug resistance limit the efficacy of cancer therapy. To address this issue, we have developed an integrated treatment protocol for effective treatment of heterogeneous ovarian cancer. Methods: An amphiphilic polymer coated magnetic iron oxide nanoparticle was conjugated with near infrared dye labeled HER2 affibody and chemotherapy drug cisplatin. The effects of the theranostic nanoparticle on targeted drug delivery, therapeutic efficacy, non-invasive magnetic resonance image (MRI)-guided therapy, and optical imaging detection of therapy resistant tumors were examined in an orthotopic human ovarian cancer xenograft model with highly heterogeneous levels of HER2 expression. Results: We found that systemic delivery of HER2-targeted magnetic iron oxide nanoparticles carrying cisplatin significantly inhibited the growth of primary tumor and peritoneal and lung metastases in the ovarian cancer xenograft model in nude mice. Differential delivery of theranostic nanoparticles into individual tumors with heterogeneous levels of HER2 expression and various responses to therapy were detectable by MRI. We further found a stronger therapeutic response in metastatic tumors compared to primary tumors, likely due to a higher level of HER2 expression and a larger number of proliferating cells in metastatic tumor cells. Relatively long-time retention of iron oxide nanoparticles in tumor tissues allowed interrogating the relationship between nanoparticle drug delivery and the presence of resistant residual tumors by in vivo molecular imaging and histological analysis of the tumor tissues. Following therapy, most of the remaining tumors were small, primary tumors that had low levels of HER2 expression and nanoparticle drug accumulation, thereby explaining their lack of therapeutic response. However, a few residual tumors had HER2-expressing tumor cells and detectable nanoparticle drug delivery but failed to respond, suggesting additional intrinsic resistant mechanisms. Nanoparticle retention in the small residual tumors, nevertheless, produced optical signals for detection by spectroscopic imaging. Conclusion: The inability to completely excise peritoneal metastatic tumors by debulking surgery as well as resistance to chemotherapy are the major clinical challenges for ovarian cancer treatment. This targeted cancer therapy has the potential for the development of effective treatment for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Minati Satpathy
- Departments of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Rafal J. Zielinski
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiping Qian
- Departments of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | - Aaron M. Mohs
- Department of Biomedical Engineering, Emory University, Atlanta, GA, 30322, USA
| | - Brad A. Kairdolf
- Department of Biomedical Engineering, Emory University, Atlanta, GA, 30322, USA
| | - Xin Ji
- Ocean NanoTech, LLC, San Diego, CA, 92126, USA
| | - Jacek Capala
- Clinical Radiation Oncology Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| | - Malgorzata Lipowska
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shuming Nie
- Department of Biomedical Engineering, Emory University, Atlanta, GA, 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lily Yang
- Departments of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
156
|
Ahmed MS, Rodell CB, Hulsmans M, Kohler RH, Aguirre AD, Nahrendorf M, Weissleder R. A Supramolecular Nanocarrier for Delivery of Amiodarone Anti-Arrhythmic Therapy to the Heart. Bioconjug Chem 2019; 30:733-740. [PMID: 30615425 DOI: 10.1021/acs.bioconjchem.8b00882] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Amiodarone is an effective antiarrhythmic drug used to treat and prevent different types of cardiac arrhythmias. However, amiodarone can have considerable side effects resulting from accumulation in off-target tissues. Cardiac macrophages are highly prevalent tissue-resident immune cells with importance in homeostatic functions, including immune response and modulation of cardiac conduction. We hypothesized that amiodarone could be more efficiently delivered to the heart via cardiac macrophages, an important step toward reducing overall dose and off-target tissue accumulation. Toward this goal, we synthesized a nanoparticle drug carrier composed of l-lysine cross-linked succinyl-β-cyclodextrin that demonstrates amiodarone binding through supramolecular host-guest interaction as well as a high macrophage affinity. Biodistribution analyses at the organ and single-cell level demonstrate accumulation of nanoparticles in the heart resulting from rapid uptake by cardiac macrophages. Nanoparticle assisted delivery of amiodarone resulted in a 250% enhancement in the selective delivery of the drug to cardiac tissue in part due to a concomitant decrease of pulmonary accumulation, the main source of off-target toxicity.
Collapse
Affiliation(s)
- Maaz S Ahmed
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States
| | - Christopher B Rodell
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States
| | - Maarten Hulsmans
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States
| | - Rainer H Kohler
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States
| | - Aaron D Aguirre
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States.,Cardiology Division , Massachusetts General Hospital , 55 Fruit St , Boston , Massachusetts 02114 , United States
| | - Matthias Nahrendorf
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States.,Department of Radiology , Massachusetts General Hospital , 55 Fruit St , Boston , Massachusetts 02114 , United States
| | - Ralph Weissleder
- Center for Systems Biology , Massachusetts General Hospital , 185 Cambridge St , CPZN 5206, Boston , Massachusetts 02114 , United States.,Department of Radiology , Massachusetts General Hospital , 55 Fruit St , Boston , Massachusetts 02114 , United States.,Department of Systems Biology , Harvard Medical School , 200 Longwood Ave , Boston , Massachusetts 02115 , United States
| |
Collapse
|
157
|
Kalyane D, Raval N, Maheshwari R, Tambe V, Kalia K, Tekade RK. Employment of enhanced permeability and retention effect (EPR): Nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:1252-1276. [PMID: 30813007 DOI: 10.1016/j.msec.2019.01.066] [Citation(s) in RCA: 534] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/02/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
Abstract
In tumorous tissues, the absence of vasculature supportive tissues intimates the formation of leaky vessels and pores (100 nm to 2 μm in diameter) and the poor lymphatic system offers great opportunity to treat cancer and the phenomenon is known as Enhanced permeability and retention (EPR) effect. The trends in treating cancer by making use of EPR effect is increasing day by day and generate multitudes of possibility to design novel anticancer therapeutics. This review aimed to present various factors affecting the EPR effect along with important things to know about EPR effect such as tumor perfusion, lymphatic function, interstitial penetration, vascular permeability, nanoparticle retention etc. This manuscript expounds the current advances and cross-talks the developments made in the of EPR effect-based therapeutics in cancer therapy along with a transactional view of its current clinical and industrial aspects.
Collapse
Affiliation(s)
- Dnyaneshwar Kalyane
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Nidhi Raval
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Rahul Maheshwari
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Vishakha Tambe
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Kiran Kalia
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opposite Air Force Station, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
158
|
Naumenko V, Garanina A, Vodopyanov S, Nikitin A, Prelovskaya A, Demihov E, Abakumov M, Majouga A, Chekhonin V. Magnetic resonance imaging for predicting personalized antitumor nanomedicine efficacy. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2019. [DOI: 10.24075/brsmu.2018.086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Magnetic resonance imaging (MRI) is widely used to diagnose cancer and study patterns and effectiveness of nanocarrier delivery of anticancer drugs. Accumulation of nanoparticles in a tumor varies widely in a given population; it is also highly dependent on biological factors, which remain largely unstudied. In recent years, there was developed a hypothesis that suggests that MRI can be used to predict response to nanoformulations-based anticancer therapy since it provides data on accumulation of MRI contrast agents in the tumor. Pilot tests prove feasibility of the approach based on this hypothesis, however, there is a number of conceptual and technical problems and limitations that hamper its introduction into the routine clinical practice. This article discusses the advantages and disadvantages of methods to stratify tumors by level of nanoparticles accumulation. Further research in this field would facilitate development of effective algorithms of personalized treatment with anticancer drugs delivered by nanoparticles.
Collapse
Affiliation(s)
- V.A. Naumenko
- Laboratory of Biomedical Nanomaterials National University of Science and Technology MISiS, Moscow
| | - A.S. Garanina
- Research Laboratory of Tissue-Specific Ligands, Faculty of Chemistry Federal State Budget Educational Institution of Higher Education MV Lomonosov Moscow State University, Moscow
| | - S.S. Vodopyanov
- Laboratory of Biomedical Nanomaterials National University of Science and Technology MISiS, Moscow
| | - A.A. Nikitin
- Laboratory of Biomedical Nanomaterials National University of Science and Technology MISiS, Moscow; Research Laboratory of Tissue-Specific Ligands, Faculty of Chemistry Federal State Budget Educational Institution of Higher Education MV Lomonosov Moscow State University, Moscow
| | - A.O. Prelovskaya
- Laboratory of Biomedical Nanomaterials National University of Science and Technology MISiS, Moscow
| | - E.I. Demihov
- Lebedev Physical Institute, Russian Academy of Sciences, Moscow
| | - M.A. Abakumov
- Laboratory of Biomedical Nanomaterials National University of Science and Technology MISiS, Moscow; Pirogov Russian National Research Medical University, Moscow
| | - A.M. Majouga
- Laboratory of Biomedical Nanomaterials National University of Science and Technology MISiS, Moscow; Research Laboratory of Tissue-Specific Ligands, Faculty of Chemistry Federal State Budget Educational Institution of Higher Education MV Lomonosov Moscow State University, Moscow; Lebedev Physical Institute, Russian Academy of Sciences, Moscow
| | - V.P. Chekhonin
- Pirogov Russian National Research Medical University, Moscow
| |
Collapse
|
159
|
Translational Nanodiagnostics for In Vivo Cancer Detection. Bioanalysis 2019. [DOI: 10.1007/978-3-030-01775-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
160
|
|
161
|
Dadfar SM, Roemhild K, Drude NI, von Stillfried S, Knüchel R, Kiessling F, Lammers T. Iron oxide nanoparticles: Diagnostic, therapeutic and theranostic applications. Adv Drug Deliv Rev 2019. [PMID: 30639256 DOI: 10.1016/j.addr.2019.01.005.iron] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Many different iron oxide nanoparticles have been evaluated over the years, for a wide variety of biomedical applications. We here summarize the synthesis, surface functionalization and characterization of iron oxide nanoparticles, as well as their (pre-) clinical use in diagnostic, therapeutic and theranostic settings. Diagnostic applications include liver, lymph node, inflammation and vascular imaging, employing mostly magnetic resonance imaging but recently also magnetic particle imaging. Therapeutic applications encompass iron supplementation in anemia and advanced cancer treatments, such as modulation of macrophage polarization, magnetic fluid hyperthermia and magnetic drug targeting. Because of their properties, iron oxide nanoparticles are particularly useful for theranostic purposes. Examples of such setups, in which diagnosis and therapy are intimately combined and in which iron oxide nanoparticles are used, are image-guided drug delivery, image-guided and microbubble-mediated opening of the blood-brain barrier, and theranostic tissue engineering. Together, these directions highlight the versatility and the broad applicability of iron oxide nanoparticles, and indicate the integration in future medical practice of multiple iron oxide nanoparticle-based materials.
Collapse
Affiliation(s)
- Seyed Mohammadali Dadfar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Karolin Roemhild
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Institute of Pathology, Medical Faculty, RWTH Aachen University Clinic, Aachen, Germany
| | - Natascha I Drude
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Nuclear Medicine, RWTH Aachen University Clinic, Aachen, Germany; Leibniz Institute for Interactive Materials - DWI, RWTH Aachen University, Aachen, Germany
| | - Saskia von Stillfried
- Institute of Pathology, Medical Faculty, RWTH Aachen University Clinic, Aachen, Germany
| | - Ruth Knüchel
- Institute of Pathology, Medical Faculty, RWTH Aachen University Clinic, Aachen, Germany
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
162
|
Dadfar SM, Roemhild K, Drude NI, von Stillfried S, Knüchel R, Kiessling F, Lammers T. Iron oxide nanoparticles: Diagnostic, therapeutic and theranostic applications. Adv Drug Deliv Rev 2019; 138:302-325. [PMID: 30639256 PMCID: PMC7115878 DOI: 10.1016/j.addr.2019.01.005] [Citation(s) in RCA: 644] [Impact Index Per Article: 107.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/19/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022]
Abstract
Many different iron oxide nanoparticles have been evaluated over the years, for a wide variety of biomedical applications. We here summarize the synthesis, surface functionalization and characterization of iron oxide nanoparticles, as well as their (pre-) clinical use in diagnostic, therapeutic and theranostic settings. Diagnostic applications include liver, lymph node, inflammation and vascular imaging, employing mostly magnetic resonance imaging but recently also magnetic particle imaging. Therapeutic applications encompass iron supplementation in anemia and advanced cancer treatments, such as modulation of macrophage polarization, magnetic fluid hyperthermia and magnetic drug targeting. Because of their properties, iron oxide nanoparticles are particularly useful for theranostic purposes. Examples of such setups, in which diagnosis and therapy are intimately combined and in which iron oxide nanoparticles are used, are image-guided drug delivery, image-guided and microbubble-mediated opening of the blood-brain barrier, and theranostic tissue engineering. Together, these directions highlight the versatility and the broad applicability of iron oxide nanoparticles, and indicate the integration in future medical practice of multiple iron oxide nanoparticle-based materials.
Collapse
Affiliation(s)
- Seyed Mohammadali Dadfar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Karolin Roemhild
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Institute of Pathology, Medical Faculty, RWTH Aachen University Clinic, Aachen, Germany
| | - Natascha I Drude
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Nuclear Medicine, RWTH Aachen University Clinic, Aachen, Germany; Leibniz Institute for Interactive Materials - DWI, RWTH Aachen University, Aachen, Germany
| | - Saskia von Stillfried
- Institute of Pathology, Medical Faculty, RWTH Aachen University Clinic, Aachen, Germany
| | - Ruth Knüchel
- Institute of Pathology, Medical Faculty, RWTH Aachen University Clinic, Aachen, Germany
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
163
|
Miller MA, Mikula H, Luthria G, Li R, Kronister S, Prytyskach M, Kohler RH, Mitchison T, Weissleder R. Modular Nanoparticulate Prodrug Design Enables Efficient Treatment of Solid Tumors Using Bioorthogonal Activation. ACS NANO 2018; 12:12814-12826. [PMID: 30550257 PMCID: PMC6307086 DOI: 10.1021/acsnano.8b07954] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/04/2018] [Indexed: 05/18/2023]
Abstract
Prodrug strategies that facilitate localized and controlled activity of small-molecule therapeutics can reduce systemic exposure and improve pharmacokinetics, yet limitations in activation chemistry have made it difficult to assign tunable multifunctionality to prodrugs. Here, we present the design and application of a modular small-molecule caging strategy that couples bioorthogonal cleavage with a self-immolative linker and an aliphatic anchor. This strategy leverages recently discovered in vivo catalysis by a nanoencapsulated palladium compound (Pd-NP), which mediates alloxylcarbamate cleavage and triggers release of the activated drug. The aliphatic anchor enables >90% nanoencapsulation efficiency of the prodrug, while also allowing >104-fold increased cytotoxicity upon prodrug activation. We apply the strategy to a prodrug formulation of monomethyl auristatin E (MMAE), demonstrating its ability to target microtubules and kill cancer cells only after selective activation by Pd-NP. Computational pharmacokinetic modeling provides a mechanistic basis for the observation that the nanotherapeutic prodrug strategy can lead to more selective activation in the tumor, yet in a manner that is more sensitive to variable enhanced permeability and retention (EPR) effects. Combination treatment with the nanoencapsulated MMAE prodrug and Pd-NP safely blocks tumor growth, especially when combined with a local radiation therapy regimen that is known to improve EPR effects, and represents a conceptual step forward in prodrug design.
Collapse
Affiliation(s)
- Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- E-mail:
| | - Hannes Mikula
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Institute
of Applied Synthetic Chemistry, Vienna University
of Technology (TU Wien), Vienna 1060, Austria
| | - Gaurav Luthria
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Biomedical Informatics, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Ran Li
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Stefan Kronister
- Institute
of Applied Synthetic Chemistry, Vienna University
of Technology (TU Wien), Vienna 1060, Austria
| | - Mark Prytyskach
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Rainer H. Kohler
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Timothy Mitchison
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
164
|
Kim HY, Li R, Ng TS, Courties G, Rodell CB, Prytyskach M, Kohler RH, Pittet MJ, Nahrendorf M, Weissleder R, Miller MA. Quantitative Imaging of Tumor-Associated Macrophages and Their Response to Therapy Using 64Cu-Labeled Macrin. ACS NANO 2018; 12:12015-12029. [PMID: 30508377 PMCID: PMC6482841 DOI: 10.1021/acsnano.8b04338] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Tumor-associated macrophages (TAMs) are widely implicated in cancer progression, and TAM levels can influence drug responses, particularly to immunotherapy and nanomedicines. However, it has been difficult to quantify total TAM numbers and their dynamic spatiotemporal distribution in a non-invasive and translationally relevant manner. Here, we address this need by developing a pharmacokinetically optimized, 64Cu-labeled polyglucose nanoparticle (Macrin) for quantitative positron emission tomography (PET) imaging of macrophages in tumors. By combining PET with high-resolution in vivo confocal microscopy and ex vivo imaging of optically cleared tissue, we found that Macrin was taken up by macrophages with >90% selectivity. Uptake correlated with the content of macrophages in both healthy tissue and tumors ( R2 > 0.9) and showed striking heterogeneity in the TAM content of an orthotopic and immunocompetent mouse model of lung carcinoma. In a proof-of-principle application, we imaged Macrin to monitor the macrophage response to neo-adjuvant therapy, using a panel of chemotherapeutic and γ-irradiation regimens. Multiple treatments elicited 180-650% increase in TAMs. Imaging identified especially TAM-rich tumors thought to exhibit enhanced permeability and retention of nanotherapeutics. Indeed, these TAM-rich tumors accumulated >700% higher amounts of a model poly(d,l-lactic- co-glycolic acid)- b-polyethylene glycol (PLGA-PEG) therapeutic nanoparticle compared to TAM-deficient tumors, suggesting that imaging may guide patient selection into nanomedicine trials. In an orthotopic breast cancer model, chemoradiation enhanced TAM and Macrin accumulation in tumors, which corresponded to the improved delivery and efficacy of two model nanotherapies, PEGylated liposomal doxorubicin and a TAM-targeted nanoformulation of the toll-like receptor 7/8 agonist resiquimod (R848). Thus, Macrin imaging offers a selective and translational means to quantify TAMs and inform therapeutic decisions.
Collapse
Affiliation(s)
- Hye-Yeong Kim
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Christopher B. Rodell
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Mark Prytyskach
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Mikael J. Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| |
Collapse
|
165
|
Elgart V, Lin JR, Loscalzo J. Determinants of drug-target interactions at the single cell level. PLoS Comput Biol 2018; 14:e1006601. [PMID: 30571695 PMCID: PMC6319770 DOI: 10.1371/journal.pcbi.1006601] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 01/04/2019] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
The physiochemical determinants of drug-target interactions in the microenvironment of the cell are complex and generally not defined by simple diffusion and intrinsic chemical reactivity. Non-specific interactions of drugs and macromolecules in cells are rarely considered formally in assessing pharmacodynamics. Here, we demonstrate that non-specific interactions lead to very slow incorporation kinetics of DNA binding drugs. We observe a rate of drug incorporation in cell nuclei three orders of magnitude slower than in vitro due to anomalous drug diffusion within cells. This slow diffusion, however, has an advantageous consequence: it leads to virtually irreversible binding of the drug to specific DNA targets in cells. We show that non-specific interactions drive slow drug diffusion manifesting as slow reaction front propagation. We study the effect of non-specific interactions in different cellular compartments by permeabilization of plasma and nuclear membranes in order to pinpoint differential compartment effects on variability in intracellular drug kinetics. These results provide the basis for a comprehensive model of the determinants of intracellular diffusion of small-molecule drugs, their target-seeking trajectories, and the consequences of these processes on the apparent kinetics of drug-target interactions. Small-molecule drug design assumes target binding of high affinity. Most small molecules can interact with other macromolecules in the cell ‘nonspecifically,’ i.e., with significantly lower affinity. The extent to which these nonspecific interactions influence the availability and action of the drug for its specific target depends upon the relative concentrations of drug, the specific target, and nonspecific targets. The structure of the cell is quite crowded with a highly non-uniform distribution of macromolecules that can interact with the drug of interest both specifically and nonspecifically. Thus, some compartments or micro-domains within the cell may have a comparatively high concentration of nonspecific targets, sufficient to trap the drug and retard its diffusion toward the specific target. Here, using small-molecule binding to DNA and single cell monitoring, we demonstrate that this effect results in apparently anomalous small molecule-DNA binding kinetics in cells at rates that are 1000-fold slower than in a homogeneous, dilute, aqueous environment. This slow intracellular diffusion, however, has an advantageous consequence: it leads to virtually irreversible binding of the small molecule (drug) to specific DNA targets in cells. We study and quantify the effect of nonspecific interactions between small DNA-binding molecules, including known DNA-binding drugs, in different cellular compartments in order to identify factors that account for the variability in binding kinetics among individual cells.
Collapse
Affiliation(s)
- Vlad Elgart
- Laboratory for Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Jia-Ren Lin
- Laboratory for Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joseph Loscalzo
- Laboratory for Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
166
|
Kim D, Shin K, Kwon SG, Hyeon T. Synthesis and Biomedical Applications of Multifunctional Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802309. [PMID: 30133009 DOI: 10.1002/adma.201802309] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/04/2018] [Indexed: 05/20/2023]
Abstract
The accumulated knowledge of nanoparticle (NP) synthesis for the last 30 years has enabled the development of functional NPs for biomedical applications. Especially, NPs with multifunctional capabilities are gaining popularity as the demand for versatile and efficient NP agents increases. Various combinations of functional materials are integrated to form multicomponent NPs with designed size, structure, and multifunctionality. Their use as diagnostic and/or therapeutic tools is demonstrated, suggesting their application potentials in healthcare and medical practice. Here, the recent achievements in the synthesis and biomedical applications of multifunctional NPs are summarized. Starting with a brief overview regarding the advances in NP synthesis and accompanying progress in nanobiotechnology, various components to construct the multifunctional NP agents, which include polymers and mesoporous, magnetic, catalytic, and semiconducting NPs, are discussed together with their overall integration forms, such as NP assembly, hollow/porous structures, or hybrid/doped systems. Following the explanation of the features that multifunctional NP agents can offer, an outlook and a brief comment regarding the future research directions are provided.
Collapse
Affiliation(s)
- Dokyoon Kim
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Kwangsoo Shin
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Soon Gu Kwon
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
167
|
Johnston MC, Scott CJ. Antibody conjugated nanoparticles as a novel form of antibody drug conjugate chemotherapy. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:63-69. [PMID: 30553522 DOI: 10.1016/j.ddtec.2018.10.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 06/09/2023]
Abstract
Antibody conjugated nanoparticles (ACNPs) represent a novel strategy for the development of therapies exploiting antibodies to augment the delivery of chemotherapy payloads. Following in the footsteps of the success of antibody drug conjugates (ADCs), ACNPs are only now reaching clinical evaluation. In this review we discuss the success of ADCs and explore the opportunities ACNPs offer, such as broad chemotherapy payload selection, high drug to antibody ratios and the ability to finely tailor drug release in comparison to ADCs. The ability of ACNPs to elicit increased avidity due to multivalent effects and the potential to use these modular platforms in immunotherapeutic approaches is also explored. Through addressing challenges that still remain in bringing these complex formulations to the clinic, ACNPs hold obvious potential for the treatment of a wide range of cancers and other diseases where selective targeting of drug agents is essential.
Collapse
Affiliation(s)
- Michael C Johnston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, United Kingdom
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, United Kingdom.
| |
Collapse
|
168
|
Yu M, Ortega CA, Si K, Molinaro R, Schoen FJ, Leitao RFC, Xu X, Mahmoudi M, Ahn S, Liu J, Saw PE, Lee IH, Brayner MMB, Lotfi A, Shi J, Libby P, Jon S, Farokhzad OC. Nanoparticles targeting extra domain B of fibronectin-specific to the atherosclerotic lesion types III, IV, and V-enhance plaque detection and cargo delivery. Am J Cancer Res 2018; 8:6008-6024. [PMID: 30613278 PMCID: PMC6299428 DOI: 10.7150/thno.24365] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 08/22/2018] [Indexed: 01/08/2023] Open
Abstract
Extra domain B of fibronectin (FN-EDB) is upregulated in the extracellular matrix during tissue remodeling and has been postulated as a potential biomarker for atherosclerosis, yet no systematic test for FN-EDB in plaques has been reported. We hypothesized that FN-EDB expression would intensify in advanced plaques. Furthermore, engineering of FN-EDB-targeted nanoparticles (NPs) could enable imaging/diagnosis and local delivery of payloads to plaques. Methods: The amount of FN-EDB in human atherosclerotic and normal arteries (ages: 40 to 85 years) was assessed by histological staining and quantification using an FN-EDB-specific aptide (APTFN-EDB). FN-EDB-specific NPs that could serve as MRI beacons were constructed by immobilizing APTFN-EDB on the NP surface containing DTPA[Gd]. MRI visualized APTFN-EDB-[Gd]NPs administered to atherosclerotic apolipoprotein E-deficient mice in the brachiocephalic arteries. Analysis of the ascending-to-descending thoracic aortas and the aortic roots of the mice permitted quantitation of Gd, FN-EDB, and APTFN-EDB-[Gd]NPs. Cyanine, a model small molecule drug, was used to study the biodistribution and pharmacokinetics of APTFN-EDB-NPs to evaluate their utility for drug delivery. Results: Atherosclerotic tissues had significantly greater FN-EDB-positive areas than normal arteries (P < 0.001). This signal pertained particularly to Type III (P < 0.01), IV (P < 0.01), and V lesions (P < 0.001) rather than Type I and II lesions (AHA classification). FN-EDB expression was positively correlated with macrophage accumulation and neoangiogenesis. Quantitative analysis of T1-weighted images of atherosclerotic mice revealed substantial APTFN-EDB-[Gd]NPs accumulation in plaques compared to control NPs, conventional MRI contrast agent (Gd-DTPA) or accumulation in wild-type C57BL/6J mice. Additionally, the APTFN-EDB-NPs significantly prolonged the blood-circulation time (t1/2: ~ 6 h) of a model drug and increased its accumulation in plaques (6.9-fold higher accumulation vs. free drug). Conclusions: Our findings demonstrate augmented FN-EDB expression in Type III, IV, and V atheromata and that APTFN-EDB-NPs could serve as a platform for identifying and/or delivering agents locally to a subset of atherosclerotic plaques.
Collapse
|
169
|
Arlauckas SP, Garren SB, Garris CS, Kohler RH, Oh J, Pittet MJ, Weissleder R. Arg1 expression defines immunosuppressive subsets of tumor-associated macrophages. Am J Cancer Res 2018; 8:5842-5854. [PMID: 30613266 PMCID: PMC6299430 DOI: 10.7150/thno.26888] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
Tumor-associated macrophages (TAM) have attracted attention as they can modulate key cancer-related activities, yet TAM represent a heterogenous group of cells that remain incompletely characterized. In growing tumors, TAM are often referred to as M2-like macrophages, which are cells that display immunosuppressive and tumorigenic functions and express the enzyme arginase 1 (Arg1). Methods: Here we combined high resolution intravital imaging with single cell RNA seq to uncover the topography and molecular profiles of immunosuppressive macrophages in mice. We further assessed how immunotherapeutic interventions impact these cells directly in vivo. Results: We show that: i) Arg1+ macrophages are more abundant in tumors compared to other organs; ii) there exist two morphologically distinct subsets of Arg1 TAM defined by previously unknown markers (Gbp2b, Bst1, Sgk1, Pmepa1, Ms4a7); iii) anti-Programmed Cell Death-1 (aPD-1) therapy decreases the number of Arg1+ TAM while increasing Arg1- TAM; iv) accordingly, pharmacological inhibition of arginase 1 does not synergize with aPD-1 therapy. Conclusion: Overall, this research shows how powerful complementary single cell analytical approaches can be used to improve our understanding of drug action in vivo.
Collapse
|
170
|
Schoenberg SO, Attenberger UI, Solomon SB, Weissleder R. Developing a Roadmap for Interventional Oncology. Oncologist 2018; 23:1162-1170. [PMID: 29959284 PMCID: PMC6263130 DOI: 10.1634/theoncologist.2017-0654] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/05/2018] [Indexed: 01/05/2023] Open
Abstract
Interventional oncology uses image-guided procedures to enhance cancer care. Today, this specialty plays an increasingly critical role in cancer diagnosis (e.g., biopsy), cancer therapy (e.g., ablation or embolization), and cancer symptom palliation (e.g., nephrostomies or biliary drainages). Although the number of procedures and technical capabilities has improved over the last few years, challenges remain. In this article we discuss the need to advance existing procedures, develop new ones, and focus on several operational aspects that will dictate future interventional techniques to enhance cancer care, particularly by accelerating drug development and improving patient outcomes. IMPLICATIONS FOR PRACTICE Interventional oncology is vital for cancer diagnosis, therapy, and symptom palliation. This report focuses on current interventional procedures and techniques with a look toward future improvements that will improve cancer care and patient outcomes.
Collapse
Affiliation(s)
- Stefan O Schoenberg
- Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ulrike I Attenberger
- Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephen B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Ralph Weissleder
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
171
|
Biodistribution and Tumors MRI Contrast Enhancement of Magnetic Nanocubes, Nanoclusters, and Nanorods in Multiple Mice Models. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8264208. [PMID: 30344459 PMCID: PMC6174815 DOI: 10.1155/2018/8264208] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/19/2018] [Indexed: 01/26/2023]
Abstract
Magnetic resonance imaging (MRI) is a powerful technique for tumor diagnostics. Iron oxide nanoparticles (IONPs) are safe and biocompatible tools that can be used for further enhancing MR tumor contrasting. Although numerous IONPs have been proposed as MRI contrast agents, low delivery rates to tumor site limit its application. IONPs accumulation in malignancies depends on both IONPs characteristics and tumor properties. In the current paper, three differently shaped Pluronic F-127-modified IONPs (nanocubes, nanoclusters, and nanorods) were compared side by side in three murine tumor models (4T1 breast cancer, B16 melanoma, and CT26 colon cancer). Orthotopic B16 tumors demonstrated more efficient IONPs uptake than heterotopic implants. Magnetic nanocubes (MNCb) had the highest r2-relaxivity in vitro (300 mM−1·s−1) compared with magnetic nanoclusters (MNCl, 104 mM−1·s−1) and magnetic nanorods (MNRd, 51 mM−1·s−1). As measured by atomic emission spectroscopy, MNCb also demonstrated better delivery efficiency to tumors (3.79% ID) than MNCl (2.94% ID) and MNRd (1.21% ID). Nevertheless, MNCl overperformed its counterparts in tumor imaging, providing contrast enhancement in 96% of studied malignancies, whereas MNCb and MNRd were detected by MRI in 73% and 63% of tumors, respectively. Maximum MR contrasting efficiency for MNCb and MNCl was around 6-24 hours after systemic administration, whereas for MNRd maximum contrast enhancement was found within first 30 minutes upon treatment. Presumably, MNRd poor MRI performance was due to low r2-relaxivity and rapid clearance by lungs (17.3% ID) immediately after injection. MNCb and MNCl were mainly captured by the liver and spleen without significant accumulation in the lungs, kidneys, and heart. High biocompatibility and profound accumulation in tumor tissues make MNCb and MNCl the promising platforms for MRI-based tumor diagnostics and drug delivery.
Collapse
|
172
|
Nanomedicines for developing cancer nanotherapeutics: from benchtop to bedside and beyond. Appl Microbiol Biotechnol 2018; 102:9449-9470. [DOI: 10.1007/s00253-018-9352-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
|
173
|
Francia V, Aliyandi A, Salvati A. Effect of the development of a cell barrier on nanoparticle uptake in endothelial cells. NANOSCALE 2018; 10:16645-16656. [PMID: 30155550 DOI: 10.1039/c8nr03171a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
In order to improve the current success of nanomedicine, a better understanding of how nano-sized materials interact with and are processed by cells is required. Typical in vitro nanoparticle-cell interaction studies often make use of cells cultured at different cell densities. However, in vivo, for their successful delivery to the target tissue, nanomedicines need to overcome several barriers, such as endothelial and epithelial cell barriers. Unlike sub-confluent or confluent cell cultures, cell barriers are tight cell monolayers, expressing a series of specialized tight junction proteins between adjacent cells to limit paracellular transport and ensure close cell-to-cell interactions. A clear understanding on how the development of cells into a cell barrier may affect the uptake of nano-sized drug carriers is still missing. To this aim, here, human primary umbilical vein endothelial cells (HUVEC) are used as a model cell line to form endothelial cell barriers. Then, nanoparticle uptake is assessed in the developed endothelial barriers and compared to the uptake in sub-confluent or confluent HUVEC cultures. The results clearly show that the organization of cells into a cell barrier leads to a differential gene expression of endocytic markers, and - interestingly - this is accompanied by reduced nanoparticle uptake levels. Transport inhibitors are used to characterise the mechanisms involved in the uptake. However, we show that some of them can strongly compromise barrier integrity, thus impairing the interpretation of the outcomes, and overall, only a partial inhibition of nanoparticle uptake could be obtained.
Collapse
Affiliation(s)
- Valentina Francia
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | |
Collapse
|
174
|
Nomoto T, Nishiyama N. Design of drug delivery systems for physical energy-induced chemical surgery. Biomaterials 2018; 178:583-596. [DOI: 10.1016/j.biomaterials.2018.03.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/17/2018] [Accepted: 03/22/2018] [Indexed: 01/03/2023]
|
175
|
Oh N, Kim Y, Kweon HS, Oh WY, Park JH. Macrophage-Mediated Exocytosis of Elongated Nanoparticles Improves Hepatic Excretion and Cancer Phototherapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:28450-28457. [PMID: 30067899 DOI: 10.1021/acsami.8b10302] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The introduction of nanoparticle-mediated delivery and therapy has revolutionized cancer treatment approaches. However, there has been limited success in clinical trials because current approaches have not simultaneously satisfied therapeutic efficacy and biosafety criteria to an adequate degree. Here, we employ efficient macrophage-mediated exocytosis of elongated nanoparticles to facilitate their localization in tumor cells for cancer therapy and their transport to hepatocytes for hepatobiliary excretion. In vitro studies show that PEGylated high-aspect ratio gold nanoparticles exit macrophages more rapidly and remain in tumor cells longer, compared with low-aspect ratio and spherical nanoparticles. In tumors, high-aspect ratio nanoparticles tend to stay in tumor cells and escape from tumor-associated macrophages when they are taken up by those cells. In the liver, high-aspect ratio nanoparticles cleared by Kupffer cells mostly take the hepatobiliary excretion pathway through efficient Kupffer cell-hepatocyte transfer. Furthermore, we demonstrate that time-dependent localization of elongated gold nanoparticles toward tumor cells in tumor tissues enhances the overall phototherapeutic outcome. Engineering nanoparticles to modulate their exocytosis provides a new approach to improve cancer nanomedicine and pave the way toward clinical translation.
Collapse
Affiliation(s)
| | | | - Hee-Seok Kweon
- Electron Microscopy Research Center , Korea Basic Science Institute , Daejeon 34133 , Republic of Korea
| | | | | |
Collapse
|
176
|
Dai Q, Wilhelm S, Ding D, Syed AM, Sindhwani S, Zhang Y, Chen YY, MacMillan P, Chan WCW. Quantifying the Ligand-Coated Nanoparticle Delivery to Cancer Cells in Solid Tumors. ACS NANO 2018; 12:8423-8435. [PMID: 30016073 DOI: 10.1021/acsnano.8b03900] [Citation(s) in RCA: 428] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Coating the nanoparticle surface with cancer cell recognizing ligands is expected to facilitate specific delivery of nanoparticles to diseased cells in vivo. While this targeting strategy is appealing, no nanoparticle-based active targeting formulation for solid tumor treatment had made it past phase III clinical trials. Here, we quantified the cancer cell-targeting efficiencies of Trastuzumab (Herceptin) and folic acid coated gold and silica nanoparticles in multiple mouse tumor models. Surprisingly, we showed that less than 14 out of 1 million (0.0014% injected dose) intravenously administrated nanoparticles were delivered to targeted cancer cells, and that only 2 out of 100 cancer cells interacted with the nanoparticles. The majority of the intratumoral nanoparticles were either trapped in the extracellular matrix or taken up by perivascular tumor associated macrophages. The low cancer cell targeting efficiency and significant uptake by noncancer cells suggest the need to re-evaluate the active targeting process and therapeutic mechanisms using quantitative methods. This will be important for developing strategies to deliver emerging therapeutics such as genome editing, nucleic acid therapy, and immunotherapy for cancer treatment using nanocarriers.
Collapse
Affiliation(s)
- Qin Dai
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering , University of Oklahoma , 101 David L. Boren Boulevard , Norman , Oklahoma 73019 , United States
| | - Ding Ding
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Abdullah Muhammad Syed
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Shrey Sindhwani
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Yuwei Zhang
- Departments of Chemistry, Materials Science and Engineering, and Chemical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Yih Yang Chen
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Presley MacMillan
- Departments of Chemistry, Materials Science and Engineering, and Chemical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Warren C W Chan
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
- Departments of Chemistry, Materials Science and Engineering, and Chemical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
- Donnelly Center for Cellular and Biomolecular Research , University of Toronto , 160 College Street , Toronto , Ontario M5S 3E1 , Canada
| |
Collapse
|
177
|
Stapleton S, Dunne M, Milosevic M, Tran CW, Gold MJ, Vedadi A, Mckee TD, Ohashi PS, Allen C, Jaffray DA. Radiation and Heat Improve the Delivery and Efficacy of Nanotherapeutics by Modulating Intratumoral Fluid Dynamics. ACS NANO 2018; 12:7583-7600. [PMID: 30004666 DOI: 10.1021/acsnano.7b06301] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanomedicine drug delivery systems are capable of transporting significant payloads to solid tumors. However, only a modest increase in antitumor efficacy relative to the standard of care has been observed. In this study, we demonstrate that a single dose of radiation or mild hyperthermia can substantially improve tumor uptake and distribution of nanotherapeutics, resulting in improved treatment efficacy. The delivery of nanomedicine was driven by a reduction in interstitial fluid pressure (IFP) and small perturbation of steady-state fluid flow. The transient effects on fluid dynamics in tumors with high IFP was also shown to dominate over immune cell endocytic capacity, another mechanism suspected of improving drug delivery. Furthermore, we demonstrate the specificity of this mechanism by showing that delivery of nanotherapeutics to low IFP tumors with high leukocyte infiltration does not benefit from pretreatment with radiation or heat. These results demonstrate that focusing on small perturbations to steady-state fluid dynamics, rather than large sustained effects or uncertain immune cell recruitment strategies, can impart a vulnerability to tumors with high IFP and enhance nanotherapeutic drug delivery and treatment efficacy.
Collapse
Affiliation(s)
- Shawn Stapleton
- Department of Medical Biophysics , University of Toronto , Toronto , ON M5G 1L7 , Canada
| | - Michael Dunne
- Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , ON M5S 3M2 , Canada
| | - Michael Milosevic
- Department of Radiation Oncology , University of Toronto , Toronto , ON M5S 3E2 , Canada
| | - Charles W Tran
- Department of Immunology , University of Toronto , Toronto , ON M5S 1A1 , Canada
| | | | | | | | - Pamela S Ohashi
- Department of Immunology , University of Toronto , Toronto , ON M5S 1A1 , Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , ON M5S 3M2 , Canada
| | - David A Jaffray
- Department of Medical Biophysics , University of Toronto , Toronto , ON M5G 1L7 , Canada
- Department of Radiation Oncology , University of Toronto , Toronto , ON M5S 3E2 , Canada
- Techna Institute , University Health Network , Toronto , ON M5G 1L5 , Canada
| |
Collapse
|
178
|
Theek B, Opacic T, Magnuska Z, Lammers T, Kiessling F. Radiomic analysis of contrast-enhanced ultrasound data. Sci Rep 2018; 8:11359. [PMID: 30054518 PMCID: PMC6063906 DOI: 10.1038/s41598-018-29653-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/19/2018] [Indexed: 01/15/2023] Open
Abstract
Radiomics describes the use radiological data in a quantitative manner to establish correlations in between imaging biomarkers and clinical outcomes to improve disease diagnosis, treatment monitoring and prediction of therapy responses. In this study, we evaluated whether a radiomic analysis on contrast-enhanced ultrasound (CEUS) data allows to automatically differentiate three xenograft mouse tumour models. Next to conventional imaging biomarker classes, i.e. intensity-based, textural, and wavelet-based features, we included biomarkers describing morphological and functional characteristics of the tumour vasculature. In total, 235 imaging biomarkers were extracted and evaluated. Dedicated feature selection allowed us to identify user-independent and stable imaging biomarkers for each imaging biomarker class. The selected radiomic signature, composed of median image intensity, energy of grey-level co-occurrence matrix, vessel network length, and run length nonuniformity of the grey-level run length matrix from the diagonal details, was used to train a linear support vector machine (SVM) to classify tumour phenotypes. The model was trained by using a four-fold cross-validation scheme and achieved 82.1% (95% CI [0.64 0.92]) correct classifications. In conclusion, our results show that a radiomic analysis can be successfully performed on CEUS data and may help to render ultrasound-based tumour imaging more accurate, reproducible and reliable.
Collapse
Affiliation(s)
- Benjamin Theek
- Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, RWTH Aachen University Clinic, Forckenbeckstraße 55, 52074, Aachen, Germany
- Comprehensive Diagnostic Center Aachen, RWTH Aachen University Clinic, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Tatjana Opacic
- Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, RWTH Aachen University Clinic, Forckenbeckstraße 55, 52074, Aachen, Germany
| | - Zuzanna Magnuska
- Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, RWTH Aachen University Clinic, Forckenbeckstraße 55, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, RWTH Aachen University Clinic, Forckenbeckstraße 55, 52074, Aachen, Germany
- Comprehensive Diagnostic Center Aachen, RWTH Aachen University Clinic, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, RWTH Aachen University Clinic, Forckenbeckstraße 55, 52074, Aachen, Germany.
- Comprehensive Diagnostic Center Aachen, RWTH Aachen University Clinic, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
179
|
Efremova MV, Naumenko VA, Spasova M, Garanina AS, Abakumov MA, Blokhina AD, Melnikov PA, Prelovskaya AO, Heidelmann M, Li ZA, Ma Z, Shchetinin IV, Golovin YI, Kireev II, Savchenko AG, Chekhonin VP, Klyachko NL, Farle M, Majouga AG, Wiedwald U. Magnetite-Gold nanohybrids as ideal all-in-one platforms for theranostics. Sci Rep 2018; 8:11295. [PMID: 30050080 PMCID: PMC6062557 DOI: 10.1038/s41598-018-29618-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/16/2018] [Indexed: 12/21/2022] Open
Abstract
High-quality, 25 nm octahedral-shaped Fe3O4 magnetite nanocrystals are epitaxially grown on 9 nm Au seed nanoparticles using a modified wet-chemical synthesis. These Fe3O4-Au Janus nanoparticles exhibit bulk-like magnetic properties. Due to their high magnetization and octahedral shape, the hybrids show superior in vitro and in vivo T2 relaxivity for magnetic resonance imaging as compared to other types of Fe3O4-Au hybrids and commercial contrast agents. The nanoparticles provide two functional surfaces for theranostic applications. For the first time, Fe3O4-Au hybrids are conjugated with two fluorescent dyes or the combination of drug and dye allowing the simultaneous tracking of the nanoparticle vehicle and the drug cargo in vitro and in vivo. The delivery to tumors and payload release are demonstrated in real time by intravital microscopy. Replacing the dyes by cell-specific molecules and drugs makes the Fe3O4-Au hybrids a unique all-in-one platform for theranostics.
Collapse
Affiliation(s)
- Maria V Efremova
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Victor A Naumenko
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Marina Spasova
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Anastasiia S Garanina
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Maxim A Abakumov
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
- Department of Medical Nanobiotechnology, Russian National Research Medical University, Moscow, 117997, Russian Federation
| | - Anastasia D Blokhina
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Pavel A Melnikov
- Department of Fundamental and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health and Social Development of the Russian Federation, Moscow, 119034, Russian Federation
| | | | - Markus Heidelmann
- ICAN - Interdisciplinary Center for Analytics on the Nanoscale and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Zi-An Li
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Zheng Ma
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Igor V Shchetinin
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Yuri I Golovin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- Derzhavin Tambov State University, Nanocenter, Tambov, 392000, Russian Federation
| | - Igor I Kireev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Alexander G Savchenko
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Vladimir P Chekhonin
- Department of Medical Nanobiotechnology, Russian National Research Medical University, Moscow, 117997, Russian Federation
- Department of Fundamental and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health and Social Development of the Russian Federation, Moscow, 119034, Russian Federation
| | - Natalia L Klyachko
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Michael Farle
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Alexander G Majouga
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation.
- D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation.
| | - Ulf Wiedwald
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation.
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany.
| |
Collapse
|
180
|
Jiang Z, Tian Y, Shan D, Wang Y, Gerhard E, Xia J, Huang R, He Y, Li A, Tang J, Ruan H, Li Y, Li J, Yang J, Wu A. pH protective Y1 receptor ligand functionalized antiphagocytosis BPLP-WPU micelles for enhanced tumor imaging and therapy with prolonged survival time. Biomaterials 2018; 170:70-81. [DOI: 10.1016/j.biomaterials.2018.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/28/2018] [Accepted: 04/01/2018] [Indexed: 12/18/2022]
|
181
|
Sulheim E, Kim J, van Wamel A, Kim E, Snipstad S, Vidic I, Grimstad IH, Widerøe M, Torp SH, Lundgren S, Waxman DJ, de Lange Davies C. Multi-modal characterization of vasculature and nanoparticle accumulation in five tumor xenograft models. J Control Release 2018; 279:292-305. [PMID: 29684498 PMCID: PMC5972071 DOI: 10.1016/j.jconrel.2018.04.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 10/17/2022]
Abstract
Preclinical research has demonstrated that nanoparticles and macromolecules can accumulate in solid tumors due to the enhanced permeability and retention effect. However, drug loaded nanoparticles often fail to show increased efficacy in clinical trials. A better understanding of how tumor heterogeneity affects nanoparticle accumulation could help elucidate this discrepancy and help in patient selection for nanomedicine therapy. Here we studied five human tumor models with varying morphology and evaluated the accumulation of 100 nm polystyrene nanoparticles. Each tumor model was characterized in vivo using micro-computed tomography, contrast-enhanced ultrasound and diffusion-weighted and dynamic contrast-enhanced magnetic resonance imaging. Ex vivo, the tumors were sectioned for both fluorescence microscopy and histology. Nanoparticle uptake and distribution in the tumors were generally heterogeneous. Density of functional blood vessels measured by fluorescence microscopy correlated significantly (p = 0.0056) with nanoparticle accumulation and interestingly, inflow of microbubbles measured with ultrasound also showed a moderate but significant (p = 0.041) correlation with nanoparticle accumulation indicating that both amount of vessels and vessel morphology and perfusion predict nanoparticle accumulation. This indicates that blood vessel characterization using contrast-enhanced ultrasound imaging or other methods could be valuable for patient stratification for treatment with nanomedicines.
Collapse
Affiliation(s)
- Einar Sulheim
- Department of Physics, Faculty of Natural Sciences, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF, Trondheim, Norway.
| | - Jana Kim
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway; Department of Radiology and Nuclear Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Annemieke van Wamel
- Department of Physics, Faculty of Natural Sciences, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Eugene Kim
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Faculty of Natural Sciences, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Igor Vidic
- Department of Physics, Faculty of Natural Sciences, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ingeborg Hovde Grimstad
- Department of Physics, Faculty of Natural Sciences, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Marius Widerøe
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
| | - Sverre H Torp
- Department of Laboratory Medicine, Children's and Women's Health, NTNU, Trondheim, Norway; Department of Pathology, St. Olav's University Hospital, Trondheim, Norway
| | - Steinar Lundgren
- Department of Oncology, St. Olav's University Hospital, Trondheim, Norway; Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Trondheim, Norway
| | - David J Waxman
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Catharina de Lange Davies
- Department of Physics, Faculty of Natural Sciences, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
182
|
Rodell CB, Arlauckas SP, Cuccarese MF, Garris CS, Li R, Ahmed MS, Kohler RH, Pittet MJ, Weissleder R. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat Biomed Eng 2018; 2:578-588. [PMID: 31015631 PMCID: PMC6192054 DOI: 10.1038/s41551-018-0236-8] [Citation(s) in RCA: 727] [Impact Index Per Article: 103.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/13/2018] [Indexed: 12/21/2022]
Abstract
Tumour-associated macrophages (TAMs) are abundant in many cancers, and often display an immune-suppressive M2-like phenotype that fosters tumour growth and promotes resistance to therapy. Yet macrophages are highly plastic and can also acquire an anti-tumourigenic M1-like phenotype. Here, we show that R848, an agonist of the toll-like receptors (TLRs) TLR7 and TLR8 identified in a morphometric-based screen, is a potent driver of the M1 phenotype in vitro and that R848-loaded β-cyclodextrin nanoparticles (CDNPs) lead to efficient drug delivery to TAMs in vivo. As a monotherapy, the administration of CDNP-R848 in multiple tumour models in mice altered the functional orientation of the tumour immune microenvironment towards an M1 phenotype, leading to controlled tumour growth and protecting the animals against tumour rechallenge. When used in combination with the immune checkpoint inhibitor anti-PD-1, we observed improved immunotherapy response rates, also in a tumour model resistant to anti-PD-1 therapy. Our findings demonstrate the ability of rationally engineered drug–nanoparticle combinations to efficiently modulate TAMs for cancer immunotherapy.
Collapse
|
183
|
Hoogenboezem EN, Duvall CL. Harnessing albumin as a carrier for cancer therapies. Adv Drug Deliv Rev 2018; 130:73-89. [PMID: 30012492 PMCID: PMC6200408 DOI: 10.1016/j.addr.2018.07.011] [Citation(s) in RCA: 396] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Serum albumin, a natural ligand carrier that is highly concentrated and long-circulating in the blood, has shown remarkable promise as a carrier for anti-cancer agents. Albumin is able to prolong the circulation half-life of otherwise rapidly cleared drugs and, importantly, promote their accumulation within tumors. The applications for using albumin as a cancer drug carrier are broad and include both traditional cancer chemotherapeutics and new classes of biologics. Strategies for leveraging albumin for drug delivery can be classified broadly into exogenous and in situ binding formulations that utilize covalent attachment, non-covalent association, or encapsulation in albumin-based nanoparticles. These methods have shown remarkable preclinical and clinical successes that are examined in this review.
Collapse
Affiliation(s)
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN.
| |
Collapse
|
184
|
Golombek SK, May JN, Theek B, Appold L, Drude N, Kiessling F, Lammers T. Tumor targeting via EPR: Strategies to enhance patient responses. Adv Drug Deliv Rev 2018; 130:17-38. [PMID: 30009886 PMCID: PMC6130746 DOI: 10.1016/j.addr.2018.07.007] [Citation(s) in RCA: 827] [Impact Index Per Article: 118.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
The tumor accumulation of nanomedicines relies on the enhanced permeability and retention (EPR) effect. In the last 5-10 years, it has been increasingly recognized that there is a large inter- and intra-individual heterogeneity in EPR-mediated tumor targeting, explaining the heterogeneous outcomes of clinical trials in which nanomedicine formulations have been evaluated. To address this heterogeneity, as in other areas of oncology drug development, we have to move away from a one-size-fits-all tumor targeting approach, towards methods that can be employed to individualize and improve nanomedicine treatments. To this end, efforts have to be invested in better understanding the nature, the complexity and the heterogeneity of the EPR effect, and in establishing systems and strategies to enhance, combine, bypass and image EPR-based tumor targeting. In the present manuscript, we summarize key studies in which these strategies are explored, and we discuss how these approaches can be employed to enhance patient responses.
Collapse
Affiliation(s)
- Susanne K Golombek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Jan-Niklas May
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Benjamin Theek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Lia Appold
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Natascha Drude
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Nuclear Medicine, RWTH Aachen University Clinic, Aachen, Germany
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany; Department of Pharmaceutics, Utrecht University, Utrecht, the Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, the Netherlands.
| |
Collapse
|
185
|
Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun 2018; 9:1410. [PMID: 29650952 PMCID: PMC5897557 DOI: 10.1038/s41467-018-03705-y] [Citation(s) in RCA: 1391] [Impact Index Per Article: 198.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 03/05/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted delivery approaches for cancer therapeutics have shown a steep rise over the past few decades. However, compared to the plethora of successful pre-clinical studies, only 15 passively targeted nanocarriers (NCs) have been approved for clinical use and none of the actively targeted NCs have advanced past clinical trials. Herein, we review the principles behind targeted delivery approaches to determine potential reasons for their limited clinical translation and success. We propose criteria and considerations that must be taken into account for the development of novel actively targeted NCs. We also highlight the possible directions for the development of successful tumor targeting strategies. Targeted delivery strategies based on nanocarriers have immense potential to change cancer care but current strategies have been shown only limited translation in the clinic. Here, the authors survey the challenge, progress and opportunities towards targeted delivery of cancer therapeutics.
Collapse
|
186
|
Ehlerding EB, Grodzinski P, Cai W, Liu CH. Big Potential from Small Agents: Nanoparticles for Imaging-Based Companion Diagnostics. ACS NANO 2018; 12:2106-2121. [PMID: 29462554 PMCID: PMC5878691 DOI: 10.1021/acsnano.7b07252] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The importance of medical imaging in the diagnosis and monitoring of cancer cannot be overstated. As personalized cancer treatments are gaining popularity, a need for more advanced imaging techniques has grown significantly. Nanoparticles are uniquely suited to fill this void, not only as imaging contrast agents but also as companion diagnostics. This review provides an overview of many ways nanoparticle imaging agents have contributed to cancer imaging, both preclinically and in the clinic, as well as charting future directions in companion diagnostics. We conclude that, while nanoparticle-based imaging agents are not without considerable scientific and developmental challenges, they enable enhanced imaging in nearly every modality, hold potential as in vivo companion diagnostics, and offer precise cancer treatment and maximize intervention efficacy.
Collapse
Affiliation(s)
- Emily B. Ehlerding
- Office of Cancer Nanotechnology Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
- Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Piotr Grodzinski
- Office of Cancer Nanotechnology Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
- Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
- Carbone Cancer Center, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Christina H. Liu
- Office of Cancer Nanotechnology Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
187
|
Lee H, Gaddy D, Ventura M, Bernards N, de Souza R, Kirpotin D, Wickham T, Fitzgerald J, Zheng J, Hendriks BS. Companion Diagnostic 64Cu-Liposome Positron Emission Tomography Enables Characterization of Drug Delivery to Tumors and Predicts Response to Cancer Nanomedicines. Theranostics 2018; 8:2300-2312. [PMID: 29721081 PMCID: PMC5928891 DOI: 10.7150/thno.21670] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 02/25/2018] [Indexed: 11/16/2022] Open
Abstract
Deposition of liposomal drugs into solid tumors is a potentially rate-limiting step for drug delivery and has substantial variability that may influence probability of response. Tumor deposition is a shared mechanism for liposomal therapeutics such that a single companion diagnostic agent may have utility in predicting response to multiple nanomedicines. Methods: We describe the development, characterization and preclinical proof-of-concept of the positron emission tomography (PET) agent, MM-DX-929, a drug-free untargeted 100 nm PEGylated liposome stably entrapping a chelated complex of 4-DEAP-ATSC and 64Cu (copper-64). MM-DX-929 is designed to mimic the biodistribution of similarly sized therapeutic agents and enable quantification of deposition in solid tumors. Results: MM-DX-929 demonstrated sufficient in vitro and in vivo stability with PET images accurately reflecting the disposition of liposome nanoparticles over the time scale of imaging. MM-DX-929 is also representative of the tumor deposition and intratumoral distribution of three different liposomal drugs, including targeted liposomes and those with different degrees of PEGylation. Furthermore, stratification using a single pre-treatment MM-DX-929 PET assessment of tumor deposition demonstrated that tumors with high MM-DX-929 deposition predicted significantly greater anti-tumor activity after multi-cycle treatments with different liposomal drugs. In contrast, MM-DX-929 tumor deposition was not prognostic in untreated tumor-bearing xenografts, nor predictive in animals treated with small molecule chemotherapeutics. Conclusions: These data illustrate the potential of MM-DX-929 PET as a companion diagnostic strategy to prospectively select patients likely to respond to liposomal drugs or nanomedicines of similar molecular size.
Collapse
|
188
|
Miller MA, Chandra R, Cuccarese MF, Pfirschke C, Engblom C, Stapleton S, Adhikary U, Kohler RH, Mohan JF, Pittet MJ, Weissleder R. Radiation therapy primes tumors for nanotherapeutic delivery via macrophage-mediated vascular bursts. Sci Transl Med 2018; 9:9/392/eaal0225. [PMID: 28566423 DOI: 10.1126/scitranslmed.aal0225] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/23/2017] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
Abstract
Efficient delivery of therapeutic nanoparticles (TNPs) to tumors is critical in improving efficacy, yet strategies that universally maximize tumoral targeting by TNP modification have been difficult to achieve in the clinic. Instead of focusing on TNP optimization, we show that the tumor microenvironment itself can be therapeutically primed to facilitate accumulation of multiple clinically relevant TNPs. Building on the recent finding that tumor-associated macrophages (TAM) can serve as nanoparticle drug depots, we demonstrate that local tumor irradiation substantially increases TAM relative to tumor cells and, thus, TNP delivery. High-resolution intravital imaging reveals that after radiation, TAM primarily accumulate adjacent to microvasculature, elicit dynamic bursts of extravasation, and subsequently enhance drug uptake in neighboring tumor cells. TAM depletion eliminates otherwise beneficial radiation effects on TNP accumulation and efficacy, and controls with unencapsulated drug show that radiation effects are more pronounced with TNPs. Priming with combined radiation and cyclophosphamide enhances vascular bursting and tumoral TNP concentration, in some cases leading to a sixfold increase of TNP accumulation in the tumor, reaching 6% of the injected dose per gram of tissue. Radiation therapy alters tumors for enhanced TNP delivery in a TAM-dependent fashion, and these observations have implications for the design of next-generation tumor-targeted nanomaterials and clinical trials for adjuvant strategies.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Ravi Chandra
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA.,Harvard Radiation Oncology Program, 55 Fruit Street, Boston, MA 02114, USA
| | - Michael F Cuccarese
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Camilla Engblom
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Shawn Stapleton
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Utsarga Adhikary
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - James F Mohan
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA. .,Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
189
|
Mahmoudi M. Debugging Nano-Bio Interfaces: Systematic Strategies to Accelerate Clinical Translation of Nanotechnologies. Trends Biotechnol 2018; 36:755-769. [PMID: 29559165 DOI: 10.1016/j.tibtech.2018.02.014] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/23/2018] [Accepted: 02/26/2018] [Indexed: 12/21/2022]
Abstract
Despite considerable efforts in the field of nanomedicine that have been made by researchers, funding agencies, entrepreneurs, and the media, fewer nanoparticle (NP) technologies than expected have made it to clinical trials. The wide gap between the efforts and effective clinical translation is, at least in part, due to multiple overlooked factors in both in vitro and in vivo environments, a poor understanding of the nano-bio interface, and misinterpretation of the data collected in vitro, all of which reduce the accuracy of predictions regarding the NPs' fate and safety in humans. To minimize this bench-to-clinic gap, which may accelerate successful clinical translation of NPs, this opinion paper aims to introduce strategies for systematic debugging of nano-bio interfaces in the current literature.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
190
|
Kraft JC, Treuting PM, Ho RJY. Indocyanine green nanoparticles undergo selective lymphatic uptake, distribution and retention and enable detailed mapping of lymph vessels, nodes and abnormalities. J Drug Target 2018; 26:494-504. [PMID: 29388438 DOI: 10.1080/1061186x.2018.1433681] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The distributed network of lymph vessels and nodes in the body, with its complex architecture and physiology, presents a major challenge for whole-body lymphatic-targeted drug delivery. To gather physiological and pathological information of the lymphatics, near-infrared (NIR) fluorescence imaging of NIR fluorophores is used in clinical practice due to its tissue-penetrating optical radiation (700-900 nm) that safely provides real-time high-resolution in vivo images. However, indocyanine green (ICG), a common clinical NIR fluorophore, is unstable in aqueous environments and under light exposure, and its poor lymphatic distribution and retention limits its use as a NIR lymphatic tracer. To address this, we investigated in mice the distribution pathways of a novel nanoparticle formulation that stabilises ICG and is optimised for lymphatic drug delivery. From the subcutaneous space, ICG particles provided selective lymphatic uptake, lymph vessel and node retention, and extensive first-pass lymphatic distribution of ICG, enabling 0.2 mm and 5-10 cell resolution of lymph vessels, and high signal-to-background ratios for lymphatic vessel and node networks. Soluble (free) ICG readily dissipated from lymph vessels local to the injection site and absorbed into the blood. These unique characteristics of ICG particles could enable mechanistic studies of the lymphatics and diagnosis of lymphatic abnormalities.
Collapse
Affiliation(s)
- John C Kraft
- a Department of Pharmaceutics , University of Washington , Seattle , WA , USA
| | - Piper M Treuting
- b Department of Comparative Medicine , University of Washington , Seattle , WA , USA
| | - Rodney J Y Ho
- a Department of Pharmaceutics , University of Washington , Seattle , WA , USA.,c Department of Bioengineering , University of Washington , Seattle , WA , USA
| |
Collapse
|
191
|
Yi Y, Lin G, Chen S, Liu J, Zhang H, Mi P. Polyester micelles for drug delivery and cancer theranostics: Current achievements, progresses and future perspectives. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 83:218-232. [DOI: 10.1016/j.msec.2017.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/14/2022]
|
192
|
Thorat ND, Lemine OM, Bohara RA, Omri K, El Mir L, Tofail SAM. Superparamagnetic iron oxide nanocargoes for combined cancer thermotherapy and MRI applications. Phys Chem Chem Phys 2018; 18:21331-9. [PMID: 27427175 DOI: 10.1039/c6cp03430f] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nanoparticle-based cancer diagnosis-therapy integrative systems (cancer theranostics) represent an emerging approach in oncology. To address this issue in the present work iron oxide (γ-Fe2O3-maghemite) nanoparticles (IONPs) were encapsulated within the matrix of (bis(p-sulfonatophenyl)phenylphosphine)-methoxypolyethylene glycol-thiol (mPEG) polymer vesicles using a two-step process for active chemotherapeutic cargo loading in cancer theranostics. This formation method gives simple access to highly reactive surface groups present on IONPs together with good control over the vesicle size (50-100 nm). The simultaneous loading of a chemotherapeutic drug cargo (doxorubicin) and its in vitro release in cancer cells was achieved. The feasibility of controlled drug release under different pH conditions was demonstrated in the case of encapsulated doxorubicin molecules, showing the viability of the concept of stimulated drug delivery for magneto-chemotherapy. These polymer-magnetic nanocargoes (PMNCs) exhibit enhanced contrast properties that open potential applications for magnetic resonance imaging. These self-assembled magnetic polymersomes can be used as efficient multifunctional nanocarriers for combined therapy and imaging.
Collapse
Affiliation(s)
- Nanasaheb D Thorat
- Department of Physics & Energy, University of Limerick, Limerick, Ireland. and Materials & Surface Science Institute, Bernal Institute, University of Limerick, Limerick, Ireland
| | - O M Lemine
- Physics Department, College of Sciences, Al Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Raghvendra A Bohara
- Center for Interdisciplinary Research, D. Y. Patil University, Kolhapur 416006, India
| | - Karim Omri
- Laboratory of Physics of Materials and Nanomaterials Applied at Environment (LaPhyMNE), Faculty of Sciences in Gabes, Gabes, Tunisia
| | - L El Mir
- Physics Department, College of Sciences, Al Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia and Center for Interdisciplinary Research, D. Y. Patil University, Kolhapur 416006, India
| | - Syed A M Tofail
- Department of Physics & Energy, University of Limerick, Limerick, Ireland. and Materials & Surface Science Institute, Bernal Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
193
|
Chen L, Zang F, Wu H, Li J, Xie J, Ma M, Gu N, Zhang Y. Using PEGylated magnetic nanoparticles to describe the EPR effect in tumor for predicting therapeutic efficacy of micelle drugs. NANOSCALE 2018; 10:1788-1797. [PMID: 29308812 DOI: 10.1039/c7nr08319j] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Micelle drugs based on a polymeric platform offer great advantages over liposomal drugs for tumor treatment. Although nearly all of the nanomedicines approved in the clinical use can passively target to the tumor tissues on the basis of an enhanced permeability and retention (EPR) effect, the nanodrugs have shown heterogenous responses in the patients. This phenomenon may be traced back to the EPR effect of tumor, which is extremely variable in the individuals from extensive studies. Nevertheless, there is a lack of experimental data describing the EPR effect and predicting its impact on therapeutic efficacy of nanoagents. Herein, we developed 32 nm magnetic iron oxide nanoparticles (MION) as a T2-weighted contrast agent to describe the EPR effect of each tumor by in vivo magnetic resonance imaging (MRI). The MION were synthesized by a thermal decomposition method and modified with DSPE-PEG2000 for biological applications. The PEGylated MION (Fe3O4@PEG) exhibited high r2 of 571 mM-1 s-1 and saturation magnetization (Ms) of 94 emu g-1 Fe as well as long stability and favorable biocompatibility through the in vitro studies. The enhancement intensities of the tumor tissue from the MR images were quantitatively measured as TNR (Tumor/Normal tissue signal Ratio) values, which were correlated with the delay of tumor growth after intravenous administration of the PLA-PEG/PTX micelle drug. The results demonstrated that the group with the smallest TNR values (TNR < 0.5) displayed the best tumor inhibitory effect. In addition, there was a superior correlation between TNR value and relative tumor delay in individual mice. These analysis results indicated that the TNR value of the tumor region enhanced by Fe3O4@PEG (d = 32 nm) could be used to predict the therapeutic efficacy of the micelle drugs (d ≤ 32 nm) in a certain period of time. Fe3O4@PEG has a potential to serve as an ideal MRI contrast agent to visualize the EPR effect in patients for accurate medication guidance of micelle drugs in the future treatment of tumors.
Collapse
Affiliation(s)
- Ling Chen
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Hartshorn CM, Bradbury MS, Lanza GM, Nel AE, Rao J, Wang AZ, Wiesner UB, Yang L, Grodzinski P. Nanotechnology Strategies To Advance Outcomes in Clinical Cancer Care. ACS NANO 2018; 12:24-43. [PMID: 29257865 PMCID: PMC6589353 DOI: 10.1021/acsnano.7b05108] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Ongoing research into the application of nanotechnology for cancer treatment and diagnosis has demonstrated its advantages within contemporary oncology as well as its intrinsic limitations. The National Cancer Institute publishes the Cancer Nanotechnology Plan every 5 years since 2005. The most recent iteration helped codify the ongoing basic and translational efforts of the field and displayed its breadth with several evolving areas. From merely a technological perspective, this field has seen tremendous growth and success. However, an incomplete understanding of human cancer biology persists relative to the application of nanoscale materials within contemporary oncology. As such, this review presents several evolving areas in cancer nanotechnology in order to identify key clinical and biological challenges that need to be addressed to improve patient outcomes. From this clinical perspective, a sampling of the nano-enabled solutions attempting to overcome barriers faced by traditional therapeutics and diagnostics in the clinical setting are discussed. Finally, a strategic outlook of the future is discussed to highlight the need for next-generation cancer nanotechnology tools designed to address critical gaps in clinical cancer care.
Collapse
Affiliation(s)
- Christopher M Hartshorn
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| | - Michelle S Bradbury
- Department of Radiology and Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York, 10065, United States
| | - Gregory M Lanza
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63108, United States
| | - Andre E Nel
- Division of NanoMedicine, Department of Medicine, and California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jianghong Rao
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, California 94305, United States
| | - Andrew Z. Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ulrich B Wiesner
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14843, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Piotr Grodzinski
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| |
Collapse
|
195
|
Atkinson SP, Andreu Z, Vicent MJ. Polymer Therapeutics: Biomarkers and New Approaches for Personalized Cancer Treatment. J Pers Med 2018; 8:E6. [PMID: 29360800 PMCID: PMC5872080 DOI: 10.3390/jpm8010006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/11/2018] [Accepted: 01/15/2018] [Indexed: 02/06/2023] Open
Abstract
Polymer therapeutics (PTs) provides a potentially exciting approach for the treatment of many diseases by enhancing aqueous solubility and altering drug pharmacokinetics at both the whole organism and subcellular level leading to improved therapeutic outcomes. However, the failure of many polymer-drug conjugates in clinical trials suggests that we may need to stratify patients in order to match each patient to the right PT. In this concise review, we hope to assess potential PT-specific biomarkers for cancer treatment, with a focus on new studies, detection methods, new models and the opportunities this knowledge will bring for the development of novel PT-based anti-cancer strategies. We discuss the various "hurdles" that a given PT faces on its passage from the syringe to the tumor (and beyond), including the passage through the bloodstream, tumor targeting, tumor uptake and the intracellular release of the active agent. However, we also discuss other relevant concepts and new considerations in the field, which we hope will provide new insight into the possible applications of PT-related biomarkers.
Collapse
Affiliation(s)
- Stuart P Atkinson
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - Zoraida Andreu
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
196
|
Nanoparticle anchoring targets immune agonists to tumors enabling anti-cancer immunity without systemic toxicity. Nat Commun 2018; 9:6. [PMID: 29295974 PMCID: PMC5750237 DOI: 10.1038/s41467-017-02251-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/16/2017] [Indexed: 02/01/2023] Open
Abstract
Immunostimulatory agents such as agonistic anti-CD137 and interleukin (IL)−2 generate effective anti-tumor immunity but also elicit serious toxicities, hampering their clinical application. Here we show that combination therapy with anti-CD137 and an IL-2-Fc fusion achieves significant initial anti-tumor activity, but also lethal immunotoxicity deriving from stimulation of circulating leukocytes. To overcome this toxicity, we demonstrate that anchoring IL-2 and anti-CD137 on the surface of liposomes allows these immune agonists to rapidly accumulate in tumors while lowering systemic exposure. In multiple tumor models, immunoliposome delivery achieves anti-tumor activity equivalent to free IL-2/anti-CD137 but with the complete absence of systemic toxicity. Immunoliposomes stimulated tumor infiltration by cytotoxic lymphocytes, cytokine production, and granzyme expression, demonstrating equivalent immunostimulatory effects to the free drugs in the local tumor microenvironment. Thus, surface-anchored particle delivery may provide a general approach to exploit the potent stimulatory activity of immune agonists without debilitating systemic toxicities. Immunostimulatory agents used in cancer treatment often elicit serious toxicities, limiting their clinical application. Here, the authors show that the use of liposomes to intravenously deliver surface-anchored IL-2 and anti-CD137 proteins enables anti-cancer immunity and reduces the toxic side effects.
Collapse
|
197
|
Nicolas-Boluda A, Silva AK, Fournel S, Gazeau F. Physical oncology: New targets for nanomedicine. Biomaterials 2018; 150:87-99. [DOI: 10.1016/j.biomaterials.2017.10.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/28/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023]
|
198
|
Balasubramanian V, Liu Z, Hirvonen J, Santos HA. Bridging the Knowledge of Different Worlds to Understand the Big Picture of Cancer Nanomedicines. Adv Healthc Mater 2018; 7. [PMID: 28570787 DOI: 10.1002/adhm.201700432] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/27/2017] [Indexed: 12/22/2022]
Abstract
Explosive growth of nanomedicines continues to significantly impact the therapeutic strategies for effective cancer treatment. Despite the significant progress in the development of advanced nanomedicines, successful clinical translation remains challenging. As cancer nanomedicine is a multidisciplinary field, the fundamental problem is that the knowledge gaps stem from different vantage points in the understanding of cancer nanomedicines. The complexities and heterogenecity of both nanomedicines and cancer are further demanding the integration of highly diverse expertise to develop clinically translatable cancer nanomedicines. This progress report aims to discuss the current understanding of cancer nanomedicines between different research areas in terms of nanoparticle engineering, formulation, tumor patho-physiology and clinical medicine, as well as to identify the knowledge gaps lying at the interface between the different fields of research in nanomedicine. Here we also highlight for the necessity to harmonize the multidisciplinary effort in the research of nanomedicines in order to bridge the knowledge and to advance the full understanding in cancer nanomedicines. A paradigm shift is needed in the strategic development of disease specific nanomedicines in order to foster the successful translation into clinic of future cancer nanomedicines.
Collapse
Affiliation(s)
- Vimalkumar Balasubramanian
- Division of Pharmaceutical Chemistry and Technology; Drug Research Program; Faculty of Pharmacy; University of Helsinki; FI-00014 Helsinki Finland
| | - Zehua Liu
- Division of Pharmaceutical Chemistry and Technology; Drug Research Program; Faculty of Pharmacy; University of Helsinki; FI-00014 Helsinki Finland
| | - Jouni Hirvonen
- Division of Pharmaceutical Chemistry and Technology; Drug Research Program; Faculty of Pharmacy; University of Helsinki; FI-00014 Helsinki Finland
| | - Hélder A. Santos
- Helsinki Institute of Life Science; HiLIFE; University of Helsinki; FI-00014 Helsinki Finland
| |
Collapse
|
199
|
Jia Y, Sheng Z, Hu D, Yan F, Zhu M, Gao G, Wang P, Liu X, Wang X, Zheng H. Highly penetrative liposome nanomedicine generated by a biomimetic strategy for enhanced cancer chemotherapy. Biomater Sci 2018; 6:1546-1555. [DOI: 10.1039/c8bm00256h] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Biomimetic liposome nanomedicine with deep tumor penetration and specific homotypic targeting ability enhanced cancer chemotherapy.
Collapse
|
200
|
Weissleder R, Schwaiger MC, Gambhir SS, Hricak H. Imaging approaches to optimize molecular therapies. Sci Transl Med 2017; 8:355ps16. [PMID: 27605550 DOI: 10.1126/scitranslmed.aaf3936] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Imaging, including its use for innovative tissue sampling, is slowly being recognized as playing a pivotal role in drug development, clinical trial design, and more effective delivery and monitoring of molecular therapies. The challenge is that, while a considerable number of new imaging technologies and new targeted tracers have been developed for cancer imaging in recent years, the technologies are neither evenly distributed nor evenly implemented. Furthermore, many imaging innovations are not validated and are not ready for widespread use in drug development or in clinical trial designs. Inconsistent and often erroneous use of terminology related to quantitative imaging biomarkers has also played a role in slowing their development and implementation. We examine opportunities for, and challenges of, the use of imaging biomarkers to facilitate development of molecular therapies and to accelerate progress in clinical trial design. In the future, in vivo molecular imaging, image-guided tissue sampling for mutational analyses ("high-content biopsies"), and noninvasive in vitro tests ("liquid biopsies") will likely be used in various combinations to provide the best possible monitoring and individualized treatment plans for cancer patients.
Collapse
Affiliation(s)
| | | | | | - Hedvig Hricak
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|