51
|
Sudheesh MS, Pavithran K, M S. Revisiting the outstanding questions in cancer nanomedicine with a future outlook. NANOSCALE ADVANCES 2022; 4:634-653. [PMID: 36131837 PMCID: PMC9418065 DOI: 10.1039/d1na00810b] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/22/2021] [Indexed: 06/01/2023]
Abstract
The field of cancer nanomedicine has been fueled by the expectation of mitigating the inefficiencies and life-threatening side effects of conventional chemotherapy. Nanomedicine proposes to utilize the unique nanoscale properties of nanoparticles to address the most pressing questions in cancer treatment and diagnosis. The approval of nano-based products in the 1990s inspired scientific explorations in this direction. However, despite significant progress in the understanding of nanoscale properties, there are only very few success stories in terms of substantial increase in clinical efficacy and overall patient survival. All existing paradigms such as the concept of enhanced permeability and retention (EPR), the stealth effect and immunocompatibility of nanomedicine have been questioned in recent times. In this review we critically examine impediments posed by biological factors to the clinical success of nanomedicine. We put forth current observations on critical outstanding questions in nanomedicine. We also provide the promising side of cancer nanomedicine as we move forward in nanomedicine research. This would provide a future direction for research in nanomedicine and inspire ongoing investigations.
Collapse
Affiliation(s)
- M S Sudheesh
- Dept. of Pharmaceutics, Amrita School of Pharmacy Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara Kochi - 682041 India +91-9669372019
| | - K Pavithran
- Department of Medical Oncology, Amrita Institute of Medial Sciences and Research Centre Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara Kochi - 682041 India
| | - Sabitha M
- Dept. of Pharmaceutics, Amrita School of Pharmacy Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara Kochi - 682041 India +91-9669372019
| |
Collapse
|
52
|
Ding X, Sun X, Cai H, Wu L, Liu Y, Zhao Y, Zhou D, Yu G, Zhou X. Engineering Macrophages via Nanotechnology and Genetic Manipulation for Cancer Therapy. Front Oncol 2022; 11:786913. [PMID: 35070992 PMCID: PMC8770285 DOI: 10.3389/fonc.2021.786913] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages play critical roles in tumor progression. In the tumor microenvironment, macrophages display highly diverse phenotypes and may perform antitumorigenic or protumorigenic functions in a context-dependent manner. Recent studies have shown that macrophages can be engineered to transport drug nanoparticles (NPs) to tumor sites in a targeted manner, thereby exerting significant anticancer effects. In addition, macrophages engineered to express chimeric antigen receptors (CARs) were shown to actively migrate to tumor sites and eliminate tumor cells through phagocytosis. Importantly, after reaching tumor sites, these engineered macrophages can significantly change the otherwise immune-suppressive tumor microenvironment and thereby enhance T cell-mediated anticancer immune responses. In this review, we first introduce the multifaceted activities of macrophages and the principles of nanotechnology in cancer therapy and then elaborate on macrophage engineering via nanotechnology or genetic approaches and discuss the effects, mechanisms, and limitations of such engineered macrophages, with a focus on using live macrophages as carriers to actively deliver NP drugs to tumor sites. Several new directions in macrophage engineering are reviewed, such as transporting NP drugs through macrophage cell membranes or extracellular vesicles, reprogramming tumor-associated macrophages (TAMs) by nanotechnology, and engineering macrophages with CARs. Finally, we discuss the possibility of combining engineered macrophages and other treatments to improve outcomes in cancer therapy.
Collapse
Affiliation(s)
- Xiaoling Ding
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xinchen Sun
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, Taizhou Peoples' Hospital, Taizhou, China
| | - Huihui Cai
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, The Sixth Nantong People's Hospital, Nantong, China
| | - Lei Wu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Ying Liu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yu Zhao
- Department of Immunology, Southeast University, School of Medicine, Nanjing, China
| | - Dingjingyu Zhou
- Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| |
Collapse
|
53
|
Jiang Y, Huo Z, Qi X, Zuo T, Wu Z. Copper-induced tumor cell death mechanisms and antitumor theragnostic applications of copper complexes. Nanomedicine (Lond) 2022; 17:303-324. [PMID: 35060391 DOI: 10.2217/nnm-2021-0374] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent studies found that unbalanced copper homeostasis affect tumor growth, causing irreversible damage. Copper can induce multiple forms of cell death, including apoptosis and autophagy, through various mechanisms, including reactive oxygen species accumulation, proteasome inhibition, and antiangiogenesis. Hence, copper in vivo has attracted tremendous attention and is in the research spotlight in the field of tumor treatment. This review first highlights three typical forms of copper's antitumor mechanisms. Then, the development of diverse biomaterials and nanotechnology allowing copper to be fabricated into diverse structures to realize its theragnostic action is discussed. Novel copper complexes and their clinical applications are subsequently described.
Collapse
Affiliation(s)
- Yicheng Jiang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Zhiyi Huo
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.,Industrial Technology Innovation Platform, Zhejiang Center for Safety Study of Drug Substances, Hangzhou, 310018, China
| | - Tongmei Zuo
- Industrial Technology Innovation Platform, Zhejiang Center for Safety Study of Drug Substances, Hangzhou, 310018, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| |
Collapse
|
54
|
Galli F, Varani M, Trapasso F, Tetti S, Signore A. Radiolabeling of monocytes, NK cells and dendritic cells and quality controls. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00187-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
55
|
Zhou T, Xu Y, Gong Y, Yu M, Xu E, Aimaiti W, Ma R, Xing L, Wen H, Wang J, Jiang H. Breaking-then-curing strategy for efficient cystic echinococcosis therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.12.098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
56
|
Guo Y, Liu X. Radionanomedicine: Advanced Strategy for Precision Theranostics of Breast Cancer. J Biomed Nanotechnol 2022; 18:50-60. [PMID: 35180899 DOI: 10.1166/jbn.2022.3226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Breast carcinoma remains one of the most common and fatal cancers, and even though a series of general therapeutic approaches have been used to treat breast cancer, their outcomes are significantly affected by a variety of side effects. However, nanomedicine could offer novel strategies for dealing with breast carcinoma. In fact, an increasing number of radionanomedicine approaches have recently been used in both diagnostics and therapy. To highlight this trend, the aim of the current review is to systemically summarize the latest advances in radionanomedicine, including single-modular imaging, multiple-modular imaging, and nanomedicine-based theranostics. Barriers to clinical application, the development of next-generation radionanomedicine, and challenges associated with future design are also discussed.
Collapse
Affiliation(s)
- Yang Guo
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaoying Liu
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
57
|
Wilson CG, Aarons L, Augustijns P, Brouwers J, Darwich AS, De Waal T, Garbacz G, Hansmann S, Hoc D, Ivanova A, Koziolek M, Reppas C, Schick P, Vertzoni M, García-Horsman JA. Integration of advanced methods and models to study drug absorption and related processes: An UNGAP perspective. Eur J Pharm Sci 2021; 172:106100. [PMID: 34936937 DOI: 10.1016/j.ejps.2021.106100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.
Collapse
Affiliation(s)
- Clive G Wilson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, Glasgow, U.K.
| | | | | | | | | | | | | | | | | | | | - Mirko Koziolek
- NCE Formulation Sciences, Abbvie Deutschland GmbH & Co. KG, Germany
| | | | - Philipp Schick
- Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, Germany
| | | | | |
Collapse
|
58
|
Zhao C, Pang X, Yang Z, Wang S, Deng H, Chen X. Nanomaterials targeting tumor associated macrophages for cancer immunotherapy. J Control Release 2021; 341:272-284. [PMID: 34813877 DOI: 10.1016/j.jconrel.2021.11.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/27/2022]
Abstract
Tumor-associated macrophages (TAMs) play an important role in regulating tumor growth, invasion and metastasis, and constitute approximately 50% of tumor mass. TAMs can exist in two different subtypes, M1-polarized phenotype (pro-inflammatory and immunostimulatory) and M2-polarized phenotype (immunosuppressive myeloid cells). M2 macrophages can suppress CD8+ T cells to support tumor survival. A number of biological strategies aimed at engineering macrophages to modulate the tumor immune microenvironment remain at the forefront of cancer research. Here, we review the different therapeutic strategies that have been developed based on nanotechnology to modulate macrophage functions, such as inhibition of macrophage recruitment to tumor, depletion of M2-polarized macrophages, reprograming of M2-polarized macrophages to M1-polarized macrophages, and blocking of the CD47-signal-regulatory protein alpha (CD47-SIRPα) pathway. Furthermore, we also discuss how to image TAMs with nanoparticles to unravel novel treatment options and observe their responses to the various therapies. Overall, macrophage-mediated immune modulation based on nanotechnology can be further investigated to be effectively developed as an immunoadjuvant therapy against different cancers.
Collapse
Affiliation(s)
- Caiyan Zhao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Xiaoyu Pang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin 300072, China
| | - Zuo Yang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Sheng Wang
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin 300072, China
| | - Hongzhang Deng
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Yong Loo Lin School of Medicine, Faculty of Engineering, National University of Singapore, Singapore 119228, Singapore.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine, Faculty of Engineering, National University of Singapore, Singapore 119228, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
59
|
Liu C, Chen H, Zhou H, Yu S, Wang N, Yao W, Lu AH, Qiao W. Magnetic Resonance Imaging-Guided Multi-Stimulus-Responsive Drug Delivery Strategy for Personalized and Precise Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50716-50732. [PMID: 34668377 DOI: 10.1021/acsami.1c13853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The emergence of nano-targeted controlled release liposomal drug carriers has provided a breakthrough in cancer therapy. However, their clinical efficacy is unsatisfactory, which is related to individualized differences in targeted drugs and poor in vivo release efficiency. In this paper, we prepared a class of personalized targeted and precisely controlled-release therapeutic drug carriers (GF liposomes) by co-assembling targeting and traceable o-nitrobenzyl ester lipids to propose a magnetic resonance imaging (MRI)-guided personalized in vivo targeted drug screening strategy and a multi-stimulus superimposed controlled-release strategy. Furthermore, by following the drug release process of drug-loaded liposomes (GF-D), it was found that these liposomes could rely on energy superposition to achieve more sensitive and efficient controlled drug release. In addition, the indispensable adjustment of liposome formulation for personalized MRI-based targeted therapy was verified by differential cellular uptake and in vivo magnetic resonance imaging. In the end, the 10.22-fold tumor suppression effect in the stimulus superposition group (GF-D-UV) indicates that the multi-stimulus cumulative response strategy and MRI-guided in vivo screening strategy can more effectively treat cancer. This contribution provides a concise and clever design idea for the future development of personalized precise and efficient clinical cancer therapies.
Collapse
Affiliation(s)
- Chenyu Liu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Hailiang Chen
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Hengjun Zhou
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Simiao Yu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Ning Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Weihe Yao
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - An-Hui Lu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Weihong Qiao
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| |
Collapse
|
60
|
Soni SS, Rodell CB. Polymeric materials for immune engineering: Molecular interaction to biomaterial design. Acta Biomater 2021; 133:139-152. [PMID: 33484909 DOI: 10.1016/j.actbio.2021.01.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022]
Abstract
Biomaterials continue to evolve as complex engineered tools for interactively instructing biological systems, aiding in the understanding and treatment of various disease states through intimate biological interaction. The immune response to polymeric materials is a critical area of study, as it governs the body's response to biomaterial implants, drug delivery vehicles, and even therapeutic drug formulations. Importantly, the development of the immune response to polymeric biomaterials spans length scales - from single molecular interactions to the complex sensing of bulk biophysical properties, all of which coordinate a tissue- and systems-level response. In this review, we specifically discuss a bottom-up approach to designing biomaterials that use molecular-scale interactions to drive immune response to polymers and discuss how these interactions can be leveraged for biomaterial design. STATEMENT OF SIGNIFICANCE: The immune system is an integral controller of (patho)physiological processes, affecting nearly all aspects of human health and disease. Polymeric biomaterials, whether biologically derived or synthetically produced, can potentially alter the behavior of immune cells due to their molecular-scale interaction with individual cells, as well as their interpretation at the bulk scale. This article reviews common mechanisms by which immune cells interact with polymers at the molecular level and discusses how these interactions are being leveraged to produce the next generation of biocompatible and immunomodulatory materials.
Collapse
|
61
|
Mueller CG, Gaiddon C, Venkatasamy A. Current Clinical and Pre-Clinical Imaging Approaches to Study the Cancer-Associated Immune System. Front Immunol 2021; 12:716860. [PMID: 34539653 PMCID: PMC8446654 DOI: 10.3389/fimmu.2021.716860] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/16/2021] [Indexed: 02/01/2023] Open
Abstract
In the light of the success and the expected growth of its arsenal, immuno-therapy may become the standard neoadjuvant procedure for many cancers in the near future. However, aspects such as the identity, organization and the activation status of the peri- and intra-tumoral immune cells would represent important elements to weigh in the decision for the appropriate treatment. While important progress in non-invasive imaging of immune cells has been made over the last decades, it falls yet short of entering the clinics, let alone becoming a standard procedure. Here, we provide an overview of the different intra-vital imaging approaches in the clinics and in pre-clinical settings and discuss their benefits and drawbacks for assessing the activity of the immune system, globally and on a cellular level. Stimulated by further research, the future is likely to see many technological advances both on signal detection and emission as well as image specificity and resolution to tackle current hurdles. We anticipate that the ability to precisely determine an immune stage of cancer will capture the attention of the oncologist and will create a change in paradigm for cancer therapy.
Collapse
Affiliation(s)
- Christopher G Mueller
- CNRS UPR 3572, University of Strasbourg, Immunologie-Immunopathologie-Chimie Thérapeutique, Strasbourg, France
| | - Christian Gaiddon
- Inserm UMR_S 1113, University of Strasbourg, Interface de Recherche Fondamentale et Appliquée en Cancérologie (IRFAC), Strasbourg, France
| | - Aïna Venkatasamy
- Inserm UMR_S 1113, University of Strasbourg, Interface de Recherche Fondamentale et Appliquée en Cancérologie (IRFAC), Strasbourg, France.,IHU-Strasbourg (Institut Hospitalo-Universitaire), Strasbourg, France
| |
Collapse
|
62
|
Nanoparticles to Target and Treat Macrophages: The Ockham's Concept? Pharmaceutics 2021; 13:pharmaceutics13091340. [PMID: 34575416 PMCID: PMC8469871 DOI: 10.3390/pharmaceutics13091340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles are nanomaterials with three external nanoscale dimensions and an average size ranging from 1 to 1000 nm. Nanoparticles have gained notoriety in technological advances due to their tunable physical, chemical, and biological characteristics. However, the administration of functionalized nanoparticles to living beings is still challenging due to the rapid detection and blood and tissue clearance by the mononuclear phagocytic system. The major exponent of this system is the macrophage. Regardless the nanomaterial composition, macrophages can detect and incorporate foreign bodies by phagocytosis. Therefore, the simplest explanation is that any injected nanoparticle will be probably taken up by macrophages. This explains, in part, the natural accumulation of most nanoparticles in the spleen, lymph nodes, and liver (the main organs of the mononuclear phagocytic system). For this reason, recent investigations are devoted to design nanoparticles for specific macrophage targeting in diseased tissues. The aim of this review is to describe current strategies for the design of nanoparticles to target macrophages and to modulate their immunological function involved in different diseases with special emphasis on chronic inflammation, tissue regeneration, and cancer.
Collapse
|
63
|
Wang C, Leach BI, Lister D, Adams SR, Xu H, Hoh C, McConville P, Zhang J, Messer K, Ahrens ET. Metallofluorocarbon Nanoemulsion for Inflammatory Macrophage Detection via PET and MRI. J Nucl Med 2021; 62:1146-1153. [PMID: 33277399 PMCID: PMC8833871 DOI: 10.2967/jnumed.120.255273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is associated with a range of serious human conditions, including autoimmune and cardiovascular diseases and cancer. The ability to image active inflammatory processes greatly enhances our ability to diagnose and treat these diseases at an early stage. We describe molecular compositions enabling sensitive and precise imaging of inflammatory hotspots in vivo. Methods: A functionalized nanoemulsion with a fluorocarbon-encapsulated radiometal chelate (FERM) was developed to serve as a platform for multimodal imaging probe development. The 19F-containing FERM nanoemulsion encapsulates 89Zr in the fluorous oil via a fluorinated hydroxamic acid chelate. Simple mixing of the radiometal with the preformed aqueous nanoemulsion before use yields FERM, a stable in vivo cell tracer, enabling whole-body 89Zr PET and 19F MRI after a single intravenous injection. Results: The FERM nanoemulsion was intrinsically taken up by phagocytic immune cells, particularly macrophages, with high specificity. FERM stability was demonstrated by a high correlation between the 19F and 89Zr content in the blood (correlation coefficient > 0.99). Image sensitivity at a low dose (37 kBq) was observed in a rodent model of acute infection. The versatility of FERM was further demonstrated in models of inflammatory bowel disease and 4T1 tumor. Conclusion: Multimodal detection using FERM yields robust whole-body lesion detection and leverages the strengths of combined PET and 19F MRI. The FERM nanoemulsion has scalable production and is potentially useful for precise diagnosis, stratification, and treatment monitoring of inflammatory diseases.
Collapse
Affiliation(s)
- Chao Wang
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Benjamin I Leach
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Deanne Lister
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Stephen R Adams
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Hongyan Xu
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Carl Hoh
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Patrick McConville
- Department of Radiology, University of California San Diego, La Jolla, California
- Invicro, Inc., Boston, Massachusetts; and
| | - Jing Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Karen Messer
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Eric T Ahrens
- Department of Radiology, University of California San Diego, La Jolla, California;
| |
Collapse
|
64
|
Li R, Ng TSC, Wang SJ, Prytyskach M, Rodell CB, Mikula H, Kohler RH, Garlin MA, Lauffenburger DA, Parangi S, Dinulescu DM, Bardeesy N, Weissleder R, Miller MA. Therapeutically reprogrammed nutrient signalling enhances nanoparticulate albumin bound drug uptake and efficacy in KRAS-mutant cancer. NATURE NANOTECHNOLOGY 2021; 16:830-839. [PMID: 33958764 PMCID: PMC8491539 DOI: 10.1038/s41565-021-00897-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/11/2021] [Indexed: 05/04/2023]
Abstract
Nanoparticulate albumin bound paclitaxel (nab-paclitaxel, nab-PTX) is among the most widely prescribed nanomedicines in clinical use, yet it remains unclear how nanoformulation affects nab-PTX behaviour in the tumour microenvironment. Here, we quantified the biodistribution of the albumin carrier and its chemotherapeutic payload in optically cleared tumours of genetically engineered mouse models, and compared the behaviour of nab-PTX with other clinically relevant nanoparticles. We found that nab-PTX uptake is profoundly and distinctly affected by cancer-cell autonomous RAS signalling, and RAS/RAF/MEK/ERK inhibition blocked its selective delivery and efficacy. In contrast, a targeted screen revealed that IGF1R kinase inhibitors enhance uptake and efficacy of nab-PTX by mimicking glucose deprivation and promoting macropinocytosis via AMPK, a nutrient sensor in cells. This study thus shows how nanoparticulate albumin bound drug efficacy can be therapeutically improved by reprogramming nutrient signalling and enhancing macropinocytosis in cancer cells.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas S C Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie J Wang
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mark Prytyskach
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Christopher B Rodell
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Hannes Mikula
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Vienna University of Technology (TU Wien), Vienna, Austria
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Michelle A Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Daniela M Dinulescu
- Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nabeel Bardeesy
- MGH Cancer Center, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
65
|
Luo X, Hu D, Gao D, Wang Y, Chen X, Liu X, Zheng H, Sun M, Sheng Z. Metabolizable Near-Infrared-II Nanoprobes for Dynamic Imaging of Deep-Seated Tumor-Associated Macrophages in Pancreatic Cancer. ACS NANO 2021; 15:10010-10024. [PMID: 34060821 DOI: 10.1021/acsnano.1c01608] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tumor-associated macrophages (TAMs) play a crucial part in cancer evolution. Dynamic imaging of TAMs is of great significance for treatment outcome evaluation and precision tumor therapy. Currently, most fluorescence nanoprobes tend to accumulate in the liver and are difficult to metabolize, which leads to strong background signals and inadequate imaging quality of TAMs nearby the liver such as pancreatic cancer. Herein, we aim to develop metabolizable dextran-indocyanine green (DN-ICG) nanoprobes in the second near-infrared window (NIR-II, 1 000-1 700 nm) for dynamic imaging of TAMs in pancreatic cancer. Compared to free ICG, the NIR-II fluorescence intensity of DN-ICG nanoprobes increased by 279% with significantly improved stability. We demonstrated that DN-ICG nanoprobes could specifically target TAMs through the interaction of dextran with specific ICAM-3-grabbing nonintegrin related 1 (SIGN-R1), which were highly expressed in TAMs. Subsequently, DN-ICG nanoprobes gradually metabolized in the liver yet remained in pancreatic tumor stroma in mouse models, achieving a high signal-to-background ratio (SBR = 7) in deep tissue (∼0.5 cm) NIR-II imaging of TAMs. Moreover, DN-ICG nanoprobes could detect dynamic changes of TAMs induced by low-dose radiotherapy and zoledronic acid. Therefore, the highly biocompatible and biodegradable DN-ICG nanoprobes harbor great potential for precision therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Xinping Luo
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Dehong Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Duyang Gao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yuenan Wang
- Department of Radiaton Oncology, Peking University Shenzhen Hospital, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong Province 518036, P. R. China
| | - Xinhua Chen
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, P. R. China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Minjie Sun
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| |
Collapse
|
66
|
Starosolski Z, Courtney AN, Srivastava M, Guo L, Stupin I, Metelitsa LS, Annapragada A, Ghaghada KB. A Nanoradiomics Approach for Differentiation of Tumors Based on Tumor-Associated Macrophage Burden. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:6641384. [PMID: 34220380 PMCID: PMC8216795 DOI: 10.1155/2021/6641384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/26/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Objective Tumor-associated macrophages (TAMs) within the tumor immune microenvironment (TiME) of solid tumors play an important role in treatment resistance and disease recurrence. The purpose of this study was to investigate if nanoradiomics (radiomic analysis of nanoparticle contrast-enhanced images) can differentiate tumors based on TAM burden. Materials and Methods In vivo studies were performed in transgenic mouse models of neuroblastoma with low (N = 11) and high (N = 10) tumor-associated macrophage (TAM) burden. Animals underwent delayed nanoparticle contrast-enhanced CT (n-CECT) imaging at 4 days after intravenous administration of liposomal-iodine agent (1.1 g/kg). CT imaging-derived conventional tumor metrics (tumor volume and CT attenuation) were computed for segmented tumor CT datasets. Nanoradiomic analysis was performed using a PyRadiomics workflow implemented in the quantitative image feature pipeline (QIFP) server containing 900 radiomic features (RFs). RF selection was performed under supervised machine learning using a nonparametric neighborhood component method. A 5-fold validation was performed using a set of linear and nonlinear classifiers for group separation. Statistical analysis was performed using the Kruskal-Wallis test. Results N-CECT imaging demonstrated heterogeneous patterns of signal enhancement in low and high TAM tumors. CT imaging-derived conventional tumor metrics showed no significant differences (p > 0.05) in tumor volume between low and high TAM tumors. Tumor CT attenuation was not significantly different (p > 0.05) between low and high TAM tumors. Machine learning-augmented nanoradiomic analysis revealed two RFs that differentiated (p < 0.002) low TAM and high TAM tumors. The RFs were used to build a linear classifier that demonstrated very high accuracy and further confirmed by 5-fold cross-validation. Conclusions Imaging-derived conventional tumor metrics were unable to differentiate tumors with varying TAM burden; however, nanoradiomic analysis revealed texture differences and enabled differentiation of low and high TAM tumors.
Collapse
Affiliation(s)
- Zbigniew Starosolski
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - Amy N. Courtney
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mayank Srivastava
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
| | - Linjie Guo
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Igor Stupin
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
| | - Leonid S. Metelitsa
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Ananth Annapragada
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - Ketan B. Ghaghada
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
67
|
Chauhan DS, Dhasmana A, Laskar P, Prasad R, Jain NK, Srivastava R, Jaggi M, Chauhan SC, Yallapu MM. Nanotechnology synergized immunoengineering for cancer. Eur J Pharm Biopharm 2021; 163:72-101. [PMID: 33774162 PMCID: PMC8170847 DOI: 10.1016/j.ejpb.2021.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 12/26/2022]
Abstract
Novel strategies modulating the immune system yielded enhanced anticancer responses and improved cancer survival. Nevertheless, the success rate of immunotherapy in cancer treatment has been below expectation(s) due to unpredictable efficacy and off-target effects from systemic dosing of immunotherapeutic(s). As a result, there is an unmet clinical need for improving conventional immunotherapy. Nanotechnology offers several new strategies, multimodality, and multiplex biological targeting advantage to overcome many of these challenges. These efforts enable programming the pharmacodynamics, pharmacokinetics, and delivery of immunomodulatory agents/co-delivery of compounds to prime at the tumor sites for improved therapeutic benefits. This review provides an overview of the design and clinical principles of biomaterials driven nanotechnology and their potential use in personalized nanomedicines, vaccines, localized tumor modulation, and delivery strategies for cancer immunotherapy. In this review, we also summarize the latest highlights and recent advances in combinatorial therapies availed in the treatment of cold and complicated tumors. It also presents key steps and parameters implemented for clinical success. Finally, we analyse, discuss, and provide clinical perspectives on the integrated opportunities of nanotechnology and immunology to achieve synergistic and durable responses in cancer treatment.
Collapse
Affiliation(s)
- Deepak S Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nishant K Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA.
| |
Collapse
|
68
|
Wei Q, He J, Wang S, Hua S, Qi Y, Li F, Ling D, Zhou M. Low-dose X-ray enhanced tumor accumulation of theranostic nanoparticles for high-performance bimodal imaging-guided photothermal therapy. J Nanobiotechnology 2021; 19:155. [PMID: 34039369 PMCID: PMC8152352 DOI: 10.1186/s12951-021-00875-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/27/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Theranostic nanoparticles (NPs) have achieved rapid development owing to their capacity for personalized multimodal diagnostic imaging and antitumor therapy. However, the efficient delivery and bulk accumulation of NPs in tumors are still the decisive factors in improving therapeutic effect. It is urgent to seek other methods to alters tumor microenvironment (like vascular permeability and density) for enhancing the efficiency of nanoparticles delivery and accumulation at the tumor site. METHODS Herein, we developed a Raman-tagged hollow gold nanoparticle (termed as HAuNP@DTTC) with surface-enhanced Raman scattering (SERS) property, which could be accumulated efficiently in tumor site with the pre-irradiation of low-dose (3 Gy) X-ray and then exerted highly antitumor effect in breast cancer model. RESULTS The tumor growth inhibition (TGI) of HAuNP@DTTC-induced photothermal therapy (PTT) was increased from 60% for PTT only to 97%, and the lethal distant metastasis of 4T1 breast cancer (such as lung and liver) were effectively inhibited under the X-ray-assisted PTT treatment. Moreover, with the strong absorbance induced by localized surface plasmon resonance in near-infrared (NIR) region, the signals of Raman/photoacoustic (PA) imaging in tumor was also significantly enhanced after the administration of HAuNP@DTTC, indicating it could be used as the Raman/PA imaging and photothermal agent simultaneously under 808 nm laser irradiation. CONCLUSIONS Our studied of the as-prepared HAuNP@DTTC integrated the Raman/PA imaging and PTT functions into the single platform, and showed the good prospects for clinical applications especially with the low-dose X-ray irradiation as an adjuvant, which will be a productive strategy for enhancing drug delivery and accumulation in tumor theranostics.
Collapse
Affiliation(s)
- Qiaolin Wei
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
- Institute of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jian He
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Shuaifei Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shiyuan Hua
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Yuchen Qi
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
- State Key Laboratory of Modern Optical Instrumentations, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
69
|
Exploiting a New Approach to Destroy the Barrier of Tumor Microenvironment: Nano-Architecture Delivery Systems. Molecules 2021; 26:molecules26092703. [PMID: 34062992 PMCID: PMC8125456 DOI: 10.3390/molecules26092703] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 01/04/2023] Open
Abstract
Recent findings suggest that tumor microenvironment (TME) plays an important regulatory role in the occurrence, proliferation, and metastasis of tumors. Different from normal tissue, the condition around tumor significantly altered, including immune infiltration, compact extracellular matrix, new vasculatures, abundant enzyme, acidic pH value, and hypoxia. Increasingly, researchers focused on targeting TME to prevent tumor development and metastasis. With the development of nanotechnology and the deep research on the tumor environment, stimulation-responsive intelligent nanostructures designed based on TME have attracted much attention in the anti-tumor drug delivery system. TME-targeted nano therapeutics can regulate the distribution of drugs in the body, specifically increase the concentration of drugs in the tumor site, so as to enhance the efficacy and reduce adverse reactions, can utilize particular conditions of TME to improve the effect of tumor therapy. This paper summarizes the major components and characteristics of TME, discusses the principles and strategies of relevant nano-architectures targeting TME for the treatment and diagnosis systematically.
Collapse
|
70
|
Liu CH, Grodzinski P. Nanotechnology for Cancer Imaging: Advances, Challenges, and Clinical Opportunities. Radiol Imaging Cancer 2021; 3:e200052. [PMID: 34047667 PMCID: PMC8183257 DOI: 10.1148/rycan.2021200052] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/28/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
Nanoparticle (NP) imaging applications have the potential to improve cancer diagnostics, therapeutics, and treatment management. In biomedical research and clinical practice, NPs can serve as labels or labeled carriers for monitoring drug delivery or serve as imaging agents for enhanced imaging contrast, as well as providing improved signal sensitivity and specificity for in vivo imaging of molecular and cellular processes. These qualities offer exciting opportunities for NP-based imaging agents to address current limitations in oncologic imaging. Despite substantial advancements in NP design and development, very few NP-based imaging agents have translated into clinics within the past 5 years. This review highlights some promising NP-enabled imaging techniques and their potential to address current clinical cancer imaging limitations. Although most examples provided herein are from the preclinical space, discussed imaging solutions could offer unique in vivo tools to solve biologic questions, improve cancer treatment effectiveness, and inspire clinical translation innovation to improve patient care. Keywords: Molecular Imaging-Cancer, Molecular Imaging-Nanoparticles, Molecular Imaging-Optical Imaging, Metastases, Oncology, Surgery, Treatment Effects.
Collapse
Affiliation(s)
- Christina H. Liu
- From the Cancer Imaging Program, National Cancer Institute, National
Institutes of Health, 9609 Medical Center Dr, Room 4W216, Rockville, MD
20850
| | - Piotr Grodzinski
- From the Cancer Imaging Program, National Cancer Institute, National
Institutes of Health, 9609 Medical Center Dr, Room 4W216, Rockville, MD
20850
| |
Collapse
|
71
|
Yin Q, Pan A, Chen B, Wang Z, Tang M, Yan Y, Wang Y, Xia H, Chen W, Du H, Chen M, Fu C, Wang Y, Yuan X, Lu Z, Zhang Q, Wang Y. Quantitative imaging of intracellular nanoparticle exposure enables prediction of nanotherapeutic efficacy. Nat Commun 2021; 12:2385. [PMID: 33888701 PMCID: PMC8062465 DOI: 10.1038/s41467-021-22678-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Nanoparticle internalisation is crucial for the precise delivery of drug/genes to its intracellular targets. Conventional quantification strategies can provide the overall profiling of nanoparticle biodistribution, but fail to unambiguously differentiate the intracellularly bioavailable particles from those in tumour intravascular and extracellular microenvironment. Herein, we develop a binary ratiometric nanoreporter (BiRN) that can specifically convert subtle pH variations involved in the endocytic events into digitised signal output, enabling the accurately quantifying of cellular internalisation without introducing extracellular contributions. Using BiRN technology, we find only 10.7-28.2% of accumulated nanoparticles are internalised into intracellular compartments with high heterogeneity within and between different tumour types. We demonstrate the therapeutic responses of nanomedicines are successfully predicted based on intracellular nanoparticle exposure rather than the overall accumulation in tumour mass. This nonlinear optical nanotechnology offers a valuable imaging tool to evaluate the tumour targeting of new nanomedicines and stratify patients for personalised cancer therapy.
Collapse
Affiliation(s)
- Qingqing Yin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Anni Pan
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zenghui Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Mingmei Tang
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yue Yan
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yaoqi Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wei Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hongliang Du
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Meifang Chen
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chuanxun Fu
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yanni Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
72
|
Positron Emission Tomography Imaging of Macrophages in Cancer. Cancers (Basel) 2021; 13:cancers13081921. [PMID: 33923410 PMCID: PMC8072570 DOI: 10.3390/cancers13081921] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are large phagocytic cells that can be classified as a type of white blood cell and may be either mobile or stationary in tissues. The presence of macrophages in essentially every major disease makes them attractive candidates to serve as therapeutic targets and diagnostic biomarkers. Macrophages that are found in the microenvironment of solid tumors are referred to as tumor-associated macrophages (TAMs) and have been shown to influence chemoresistance, immune regulation, tumor initiation and tumor growth. The imaging of TAMs through Positron Emission Tomography (PET) has the potential to provide valuable information on cancer biology, tumor progression, and response to therapy. This review will highlight the versatility of macrophage imaging in cancer through the use of PET.
Collapse
|
73
|
Liberini V, Laudicella R, Capozza M, Huellner MW, Burger IA, Baldari S, Terreno E, Deandreis D. The Future of Cancer Diagnosis, Treatment and Surveillance: A Systemic Review on Immunotherapy and Immuno-PET Radiotracers. Molecules 2021; 26:2201. [PMID: 33920423 PMCID: PMC8069316 DOI: 10.3390/molecules26082201] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is an effective therapeutic option for several cancers. In the last years, the introduction of checkpoint inhibitors (ICIs) has shifted the therapeutic landscape in oncology and improved patient prognosis in a variety of neoplastic diseases. However, to date, the selection of the best patients eligible for these therapies, as well as the response assessment is still challenging. Patients are mainly stratified using an immunohistochemical analysis of the expression of antigens on biopsy specimens, such as PD-L1 and PD-1, on tumor cells, on peritumoral immune cells and/or in the tumor microenvironment (TME). Recently, the use and development of imaging biomarkers able to assess in-vivo cancer-related processes are becoming more important. Today, positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) is used routinely to evaluate tumor metabolism, and also to predict and monitor response to immunotherapy. Although highly sensitive, FDG-PET in general is rather unspecific. Novel radiopharmaceuticals (immuno-PET radiotracers), able to identify specific immune system targets, are under investigation in pre-clinical and clinical settings to better highlight all the mechanisms involved in immunotherapy. In this review, we will provide an overview of the main new immuno-PET radiotracers in development. We will also review the main players (immune cells, tumor cells and molecular targets) involved in immunotherapy. Furthermore, we report current applications and the evidence of using [18F]FDG PET in immunotherapy, including the use of artificial intelligence (AI).
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/therapeutic use
- Artificial Intelligence
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Fluorodeoxyglucose F18/administration & dosage
- Fluorodeoxyglucose F18/chemistry
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immune Checkpoint Inhibitors/chemistry
- Immune Checkpoint Inhibitors/metabolism
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Neoplasms/diagnostic imaging
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Positron-Emission Tomography/methods
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/chemical synthesis
- Signal Transduction
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Virginia Liberini
- Department of Medical Science, Division of Nuclear Medicine, University of Torino, 10126 Torino, Italy;
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (S.B.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
| | - Martina Capozza
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
- Department of Nuclear Medicine, Kantonsspital Baden, 5004 Baden, Switzerland
| | - Sergio Baldari
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (S.B.)
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Désirée Deandreis
- Department of Medical Science, Division of Nuclear Medicine, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
74
|
Soni SS, Alsasa A, Rodell CB. Applications of Macrocyclic Host Molecules in Immune Modulation and Therapeutic Delivery. Front Chem 2021; 9:658548. [PMID: 33889565 PMCID: PMC8055865 DOI: 10.3389/fchem.2021.658548] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022] Open
Abstract
The immune system plays a central role in the development and progression of human disease. Modulation of the immune response is therefore a critical therapeutic target that enables us to approach some of the most vexing problems in medicine today such as obesity, cancer, viral infection, and autoimmunity. Methods of manipulating the immune system through therapeutic delivery centralize around two common themes: the local delivery of biomaterials to affect the surrounding tissue or the systemic delivery of soluble material systems, often aided by context-specific cell or tissue targeting strategies. In either case, supramolecular interactions enable control of biomaterial composition, structure, and behavior at the molecular-scale; through rational biomaterial design, the realization of next-generation immunotherapeutics and immunotheranostics is therefore made possible. This brief review highlights methods of harnessing macromolecular interaction for immunotherapeutic applications, with an emphasis on modes of drug delivery.
Collapse
Affiliation(s)
| | | | - Christopher B. Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
75
|
Kimm MA, Klenk C, Alunni-Fabbroni M, Kästle S, Stechele M, Ricke J, Eisenblätter M, Wildgruber M. Tumor-Associated Macrophages-Implications for Molecular Oncology and Imaging. Biomedicines 2021; 9:biomedicines9040374. [PMID: 33918295 PMCID: PMC8066018 DOI: 10.3390/biomedicines9040374] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent the largest group of leukocytes within the tumor microenvironment (TME) of solid tumors and orchestrate the composition of anti- as well as pro-tumorigenic factors. This makes TAMs an excellent target for novel cancer therapies. The plasticity of TAMs resulting in varying membrane receptors and expression of intracellular proteins allow the specific characterization of different subsets of TAMs. Those markers similarly allow tracking of TAMs by different means of molecular imaging. This review aims to provides an overview of the origin of tumor-associated macrophages, their polarization in different subtypes, and how characteristic markers of the subtypes can be used as targets for molecular imaging and theranostic approaches.
Collapse
Affiliation(s)
- Melanie A. Kimm
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Christopher Klenk
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Marianna Alunni-Fabbroni
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Sophia Kästle
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Matthias Stechele
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, Freiburg University Hospital, 79106 Freiburg, Germany;
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
- Correspondence: ; Tel.: +49-0-89-4400-76640
| |
Collapse
|
76
|
Shi C, Zhou Z, Lin H, Gao J. Imaging Beyond Seeing: Early Prognosis of Cancer Treatment. SMALL METHODS 2021; 5:e2001025. [PMID: 34927817 DOI: 10.1002/smtd.202001025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Indexed: 06/14/2023]
Abstract
Assessing cancer response to therapeutic interventions has been realized as an important course to early predict curative efficacy and treatment outcomes due to tumor heterogeneity. Compared to the traditional invasive tissue biopsy method, molecular imaging techniques have fundamentally revolutionized the ability to evaluate cancer response in a spatiotemporal manner. The past few years has witnessed a paradigm shift on the efforts from manufacturing functional molecular imaging probes for seeing a tumor to a vantage stage of interpreting the tumor response during different treatments. This review is to stand by the current development of advanced imaging technologies aiming to predict the treatment response in cancer therapy. Special interest is placed on the systems that are able to provide rapid and noninvasive assessment of pharmacokinetic drug fates (e.g., drug distribution, release, and activation) and tumor microenvironment heterogeneity (e.g., tumor cells, macrophages, dendritic cells (DCs), T cells, and inflammatory cells). The current status, practical significance, and future challenges of the emerging artificial intelligence (AI) technology and machine learning in the applications of medical imaging fields is overviewed. Ultimately, the authors hope that this review is timely to spur research interest in molecular imaging and precision medicine.
Collapse
Affiliation(s)
- Changrong Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zijian Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hongyu Lin
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jinhao Gao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
77
|
Nicolson F, Kircher MF. Theranostics: Agents for Diagnosis and Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
78
|
Arlauckas S, Oh N, Li R, Weissleder R, Miller MA. Macrophage imaging and subset analysis using single-cell RNA sequencing. Nanotheranostics 2021; 5:36-56. [PMID: 33391974 PMCID: PMC7738942 DOI: 10.7150/ntno.50185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages have been associated with drug response and resistance in diverse settings, thus raising the possibility of using macrophage imaging as a companion diagnostic to inform personalized patient treatment strategies. Nanoparticle-based contrast agents are especially promising because they efficiently deliver fluorescent, magnetic, and/or radionuclide labels by leveraging the intrinsic capacity of macrophages to accumulate nanomaterials in their role as professional phagocytes. Unfortunately, current clinical imaging modalities are limited in their ability to quantify broad molecular programs that may explain (a) which particular cell subsets a given imaging agent is actually labeling, and (b) what mechanistic role those cells play in promoting drug response or resistance. Highly multiplexed single-cell approaches including single-cell RNA sequencing (scRNAseq) have emerged as resources to help answer these questions. In this review, we query recently published scRNAseq datasets to support companion macrophage imaging, with particular focus on using dextran-based nanoparticles to predict the action of anti-cancer nanotherapies and monoclonal antibodies.
Collapse
Affiliation(s)
- Sean Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Nuri Oh
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
79
|
Abstract
Lung cancer is one of the serious malignant tumors with high morbidity and mortality due to the poor diagnosis and early metastasis. The developing nanotechnology provides novel concepts and research strategies for the lung cancer diagnosis by employing nanomaterials as diagnostic reagents to enhance diagnostic efficiency. This commentary introduces recent progress using nanoparticles for lung cancer diagnosis from two aspects of in vivo and in vitro detection. The challenges and future research perspectives are proposed at the end of the paper.
Collapse
|
80
|
Abstract
Therapeutic manipulation of the immune system against cancer has revolutionized the treatment of several advanced-stage tumors. While many have benefited from these treatments, the proportion of patients responding to immunotherapies is still low. Nanomedicines have promise to revolutionize tumor treatments through spatiotemporal control of drug activity. Such control of drug function could allow enhanced therapeutic actions of immunotherapies and reduced side effects. However, only a handful of formulations have been able to reach human clinical studies so far, and even fewer systems are being used in the clinic. Among translatable formulations, self-assembled nanomedicines have shown unique and versatile features for dealing with the heterogeneity and malignancy of tumors in the clinic. Such nanomedicines can be designed to promote antitumor immune responses through a series of immunopotentiating functions after being directly injected into tumors, or achieving selective tumor accumulation upon intravenous administration. Thus, tumor-targeted nanomedicines can enhance antitumor immunity by several mechanisms, such as inducing immunogenic damage to cancer cells, altering the tumor immune microenvironment by delivering immunomodulators, or eliminating or reprogramming immunosuppressive cells, enhancing the exposure of tumor-associated antigens to antigen presenting cells, stimulating innate immunity mechanisms, and facilitating the infiltration of antitumor immune cells and their interaction with cancer cells. Moreover, nanomedicines can be engineered to sense intratumoral stimuli for activating specific immune responses or installed with ligands for increasing drug levels in tumors, granting subcellular delivery, and triggering immune signals and proliferation of immune cells. Thus, the ability of nanomedicines to exert immunomodulatory functions selectively in tumor and tumor-associated tissues, such as draining lymph nodes, increases the efficiency of the treatments, while avoiding systemic immunosuppressive toxicities and the exacerbation of adverse immune responses. Moreover, the compartmentalized structure of self-assembled nanomedicines offers the possibility to coload a variety of drugs for controlled pharmacokinetics, enhanced tumor delivery, and synergistic therapeutic output. Also, by integrating imaging functionalities into nanomedicines, it is possible to develop theranostic platforms reporting the immune settings of tumors as well as the effects of nanomedicines on the tumor immune microenvironment. Herein, we critically reviewed significant strategies for developing nanomedicines capable of potentiating antitumor immune responses by surmounting biological barriers and modulating antitumor immune signals. Moreover, the potential of these nanomedicines for developing innovative anticancer treatments by targeting particular cells is discussed. Finally, we present our perspectives on the awaiting challenges and future directions of nanomedicines in the age of immunotherapy.
Collapse
Affiliation(s)
- Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hiroaki Kinoh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
81
|
Zhao YD, Muhetaerjiang M, An HW, Fang X, Zhao Y, Wang H. Nanomedicine enables spatiotemporally regulating macrophage-based cancer immunotherapy. Biomaterials 2020; 268:120552. [PMID: 33307365 DOI: 10.1016/j.biomaterials.2020.120552] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy, leveraging the host's coordinated immune system to fight against tumor has been clinically validated. However, the modest response owing to the multiple ways of tumor immune evasion is one of the challenges in cancer immunotherapy. Tumor associated macrophages (TAMs), as a major component of the leukocytes infiltrating in all tumors, play crucial roles in driving cancer initiation, progress and metastasis via multiple mechanisms such as mediating chronic inflammation, promoting angiogenesis, taming protective immune responses, and supporting migration and intravasation. TAMs targeted therapeutics have achieved remarkable successes in clinical trials mostly through the use of small-molecule agents and antibodies. However, efforts for further application have met with challenges of limited efficacy and safety. Nanomaterials can provide versatile approaches to realize the superior spatiotemporal control over immunomodulation to amplify immune responses, ultimately enhancing the therapeutic benefits and reducing toxicity. Here, the potential drugs used in TAM-centered cancer treatment in clinic are summarized and the recent advances of TAMs targeted nanomedicines in this filed are highlighted. More importantly, we focus on how nanomedicine can exert their advantages in spatial and temporal control of immunomodulation.
Collapse
Affiliation(s)
- Yong-Dan Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China; School of Pharmacy, Shanxi Medical University, Shanxi, 030009, PR China
| | - Mamuti Muhetaerjiang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Xiaohong Fang
- University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
82
|
Luker GD. Imaging the Immune Tumor Microenvironment to Monitor and Improve Therapy. Radiology 2020; 298:133-134. [PMID: 33112715 DOI: 10.1148/radiol.2020203799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Gary D Luker
- From the Department of Radiology, University of Michigan, 109 Zina Pitcher Pl, A524 BSRB, Ann Arbor, MI 48109-2200
| |
Collapse
|
83
|
Ng TSC, Gunda V, Li R, Prytyskach M, Iwamoto Y, Kohler RH, Parangi S, Weissleder R, Miller MA. Detecting Immune Response to Therapies Targeting PDL1 and BRAF by Using Ferumoxytol MRI and Macrin in Anaplastic Thyroid Cancer. Radiology 2020; 298:123-132. [PMID: 33107799 DOI: 10.1148/radiol.2020201791] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Anaplastic thyroid cancer (ATC) is aggressive with a poor prognosis, partly because of the immunosuppressive microenvironment created by tumor-associated macrophages (TAMs). Purpose To understand the relationship between TAM infiltration, tumor vascularization, and corresponding drug delivery by using ferumoxytol-enhanced MRI and macrin in an ATC mouse model. Materials and Methods ATC tumors were generated in 6-8-week-old female B6129SF1/J mice through intrathyroid injection to model orthotopic tumors, or intravenously to model hematogenous metastasis, and prospectively enrolled randomly into treatment cohorts (n = 94 total; August 1, 2018, to January 15, 2020). Mice were treated with vehicle or combined serine/threonine-protein kinase B-Raf (BRAF) kinase inhibitor (BRAFi) and anti-PDL1 antibody (aPDL1). A subset was cotreated with therapies, including an approximately 70-nm model drug delivery nanoparticle (DDNP) to target TAM, and an antibody-neutralizing colony stimulating factor 1 receptor (CSF1R). Imaging was performed at the macroscopic level with ferumoxytol-MRI and microscopically with macrin. Genetically engineered BrafV600E/WT p53-null allografts were used and complemented by a GFP-transgenic derivative and human xenografts. Tumor-bearing organs were processed by using tissue clearing and imaged with confocal microscopy and MRI. Two-tailed Wilcoxon tests were used for comparison (≥five per group). Results TAM levels were higher in orthotopic thyroid tumors compared with pulmonary metastatic lesions by 79% ± 23 (standard deviation; P < .001). These findings were concordant with ferumoxytol MRI, which showed 136% ± 88 higher uptake in thyroid lesions (P = .02) compared with lung lesions. BRAFi and aPDL1 combination therapy resulted in higher tumor DDNP delivery by 39% ± 14 in pulmonary lesions (P = .004). Compared with the untreated group, tumors following BRAFi, aPDL1, and CSF1R-blocking antibody combination therapy did not show greater levels of TAM or DDNP (P = .82). Conclusion In a mouse model of anaplastic thyroid cancer, ferumoxytol MRI showed 136% ± 88 greater uptake in orthotopic thyroid tumors compared with pulmonary lesions, which reflected high vascularization and greater tumor-associated macrophage (TAM) levels. Serine/threonine-protein kinase B-Raf inhibitor and anti-programmed death ligand 1 antibody elicited higher local TAM levels and 43% ± 20 greater therapeutic nanoparticle delivery but not higher vascularization in pulmonary tumors. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Luker in this issue.
Collapse
Affiliation(s)
- Thomas S C Ng
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Viswanath Gunda
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Ran Li
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Mark Prytyskach
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Yoshiko Iwamoto
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Rainer H Kohler
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Sareh Parangi
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Ralph Weissleder
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Miles A Miller
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| |
Collapse
|
84
|
Poon W, Kingston BR, Ouyang B, Ngo W, Chan WCW. A framework for designing delivery systems. NATURE NANOTECHNOLOGY 2020; 15:819-829. [PMID: 32895522 DOI: 10.1038/s41565-020-0759-5] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/30/2020] [Indexed: 05/22/2023]
Abstract
The delivery of medical agents to a specific diseased tissue or cell is critical for diagnosing and treating patients. Nanomaterials are promising vehicles to transport agents that include drugs, contrast agents, immunotherapies and gene editors. They can be engineered to have different physical and chemical properties that influence their interactions with their biological environments and delivery destinations. In this Review Article, we discuss nanoparticle delivery systems and how the biology of disease should inform their design. We propose developing a framework for building optimal delivery systems that uses nanoparticle-biological interaction data and computational analyses to guide future nanomaterial designs and delivery strategies.
Collapse
Affiliation(s)
- Wilson Poon
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin R Kingston
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ben Ouyang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- MD/PhD Program, University of Toronto, Toronto, Ontario, Canada
| | - Wayne Ngo
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario, Canada.
- Department of Materials Science & Engineering, University of Toronto, Toronto, Ontaro, Canada.
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
85
|
Koch PD, Pittet MJ, Weissleder R. The chemical biology of IL-12 production via the non-canonical NFkB pathway. RSC Chem Biol 2020; 1:166-176. [PMID: 34458756 PMCID: PMC8341911 DOI: 10.1039/d0cb00022a] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
Interleukin-12 (IL-12) has emerged as an attractive cytokine for cancer therapy because it has direct anti-cancer effects and additionally plays a critical role in enhancing checkpoint inhibitors. Given these multiple modes of actions, identifying means to pharmacologically induce IL-12 production in the tumor microenvironment has become important. In this review, we highlight therapeutics that promote IL-12 induction in tumor-associated myeloid cells through the non-canonical NFkB pathway. We discuss existing clinical trials and briefly examine the additional pathway targets that warrant further exploration for drug discovery.
Collapse
Affiliation(s)
- Peter D Koch
- Center for Systems Biology, Massachusetts General Hospital 185 Cambridge St Boston MA 02114 USA
- Department of Systems Biology, Harvard Medical School 200 Longwood Ave Boston MA 02115 USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital 185 Cambridge St Boston MA 02114 USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital 185 Cambridge St Boston MA 02114 USA
- Department of Systems Biology, Harvard Medical School 200 Longwood Ave Boston MA 02115 USA
| |
Collapse
|
86
|
Nahrendorf M, Hoyer FF, Meerwaldt AE, van Leent MM, Senders ML, Calcagno C, Robson PM, Soultanidis G, Pérez-Medina C, Teunissen AJ, Toner YC, Ishikawa K, Fish K, Sakurai K, van Leeuwen EM, Klein ED, Sofias AM, Reiner T, Rohde D, Aguirre AD, Wojtkiewicz G, Schmidt S, Iwamoto Y, Izquierdo-Garcia D, Caravan P, Swirski FK, Weissleder R, Mulder WJ. Imaging Cardiovascular and Lung Macrophages With the Positron Emission Tomography Sensor 64Cu-Macrin in Mice, Rabbits, and Pigs. Circ Cardiovasc Imaging 2020; 13:e010586. [PMID: 33076700 PMCID: PMC7583675 DOI: 10.1161/circimaging.120.010586] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Macrophages, innate immune cells that reside in all organs, defend the host against infection and injury. In the heart and vasculature, inflammatory macrophages also enhance tissue damage and propel cardiovascular diseases. METHODS We here use in vivo positron emission tomography (PET) imaging, flow cytometry, and confocal microscopy to evaluate quantitative noninvasive assessment of cardiac, arterial, and pulmonary macrophages using the nanotracer 64Cu-Macrin-a 20-nm spherical dextran nanoparticle assembled from nontoxic polyglucose. RESULTS PET imaging using 64Cu-Macrin faithfully reported accumulation of macrophages in the heart and lung of mice with myocardial infarction, sepsis, or pneumonia. Flow cytometry and confocal microscopy detected the near-infrared fluorescent version of the nanoparticle (VT680Macrin) primarily in tissue macrophages. In 5-day-old mice, 64Cu-Macrin PET imaging quantified physiologically more numerous cardiac macrophages. Upon intravenous administration of 64Cu-Macrin in rabbits and pigs, we detected heightened macrophage numbers in the infarcted myocardium, inflamed lung regions, and atherosclerotic plaques using a clinical PET/magnetic resonance imaging scanner. Toxicity studies in rats and human dosimetry estimates suggest that 64Cu-Macrin is safe for use in humans. CONCLUSIONS Taken together, these results indicate 64Cu-Macrin could serve as a facile PET nanotracer to survey spatiotemporal macrophage dynamics during various physiological and pathological conditions. 64Cu-Macrin PET imaging could stage inflammatory cardiovascular disease activity, assist disease management, and serve as an imaging biomarker for emerging macrophage-targeted therapeutics.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
- These authors contributed equally: Matthias Nahrendorf, Friedrich Felix Hoyer
| | - Friedrich Felix Hoyer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- These authors contributed equally: Matthias Nahrendorf, Friedrich Felix Hoyer
| | - Anu E. Meerwaldt
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Mandy M.T. van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Max L. Senders
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia Calcagno
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip M. Robson
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Soultanidis
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlos Pérez-Medina
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Abraham J.P. Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yohana C. Toner
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth Fish
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ken Sakurai
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther M. van Leeuwen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Emma D. Klein
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Marios Sofias
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thomas Reiner
- Department of Radiology and Chemical Biology Program, Memorial Sloan-Kettering Cancer Center; Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron D. Aguirre
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephen Schmidt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Izquierdo-Garcia
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Peter Caravan
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Filip K. Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Willem J.M. Mulder
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
87
|
Xia Y, Rao L, Yao H, Wang Z, Ning P, Chen X. Engineering Macrophages for Cancer Immunotherapy and Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002054. [PMID: 32856350 DOI: 10.1002/adma.202002054] [Citation(s) in RCA: 575] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/13/2020] [Indexed: 05/23/2023]
Abstract
Macrophages play an important role in cancer development and metastasis. Proinflammatory M1 macrophages can phagocytose tumor cells, while anti-inflammatory M2 macrophages such as tumor-associated macrophages (TAMs) promote tumor growth and invasion. Modulating the tumor immune microenvironment through engineering macrophages is efficacious in tumor therapy. M1 macrophages target cancerous cells and, therefore, can be used as drug carriers for tumor therapy. Herein, the strategies to engineer macrophages for cancer immunotherapy, such as inhibition of macrophage recruitment, depletion of TAMs, reprograming of TAMs, and blocking of the CD47-SIRPα pathway, are discussed. Further, the recent advances in drug delivery using M1 macrophages, macrophage-derived exosomes, and macrophage-membrane-coated nanoparticles are elaborated. Overall, there is still significant room for development in macrophage-mediated immune modulation and macrophage-mediated drug delivery, which will further enhance current tumor therapies against various malignant solid tumors, including drug-resistant tumors and metastatic tumors.
Collapse
Affiliation(s)
- Yuqiong Xia
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Lang Rao
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Huimin Yao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Zhongliang Wang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Pengbo Ning
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| |
Collapse
|
88
|
Li R, Ng TS, Garlin MA, Weissleder R, Miller MA. Understanding the in vivo Fate of Advanced Materials by Imaging. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910369. [PMID: 38545084 PMCID: PMC10972611 DOI: 10.1002/adfm.201910369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/09/2020] [Indexed: 11/13/2024]
Abstract
Engineered materials are ubiquitous in biomedical applications ranging from systemic drug delivery systems to orthopedic implants, and their actions unfold across multiple time- and length-scales. The efficacy and safety of biologics, nanomaterials, and macroscopic implants are all dictated by the same general principles of pharmacology as apply to small molecule drugs, comprising how the body affects materials (pharmacokinetics, PK) and conversely how materials affect the body (pharmacodynamics, PD). Imaging technologies play an increasingly insightful role in monitoring both of these processes, often simultaneously: translational macroscopic imaging modalities such as MRI and PET/CT offer whole-body quantitation of biodistribution and structural or molecular response, while ex vivo approaches and optical imaging via in vivo (intravital) microscopy reveal behaviors at subcellular resolution. In this review, the authors survey developments in imaging the in situ behavior of systemically and locally administered materials, with a particular focus on using microscopy to understand transport, target engagement, and downstream host responses at a single-cell level. The themes of microenvironmental influence, controlled drug release, on-target molecular action, and immune response, especially as mediated by macrophages and other myeloid cells are examined. Finally, the future directions of how new imaging technologies may propel efficient clinical translation of next-generation therapeutics and medical devices are proposed.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Michelle A. Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
- Department of Systems Biology, Harvard Medical School
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
89
|
Siddique S, Chow JCL. Application of Nanomaterials in Biomedical Imaging and Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1700. [PMID: 32872399 PMCID: PMC7559738 DOI: 10.3390/nano10091700] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
Nanomaterials, such as nanoparticles, nanorods, nanosphere, nanoshells, and nanostars, are very commonly used in biomedical imaging and cancer therapy. They make excellent drug carriers, imaging contrast agents, photothermal agents, photoacoustic agents, and radiation dose enhancers, among other applications. Recent advances in nanotechnology have led to the use of nanomaterials in many areas of functional imaging, cancer therapy, and synergistic combinational platforms. This review will systematically explore various applications of nanomaterials in biomedical imaging and cancer therapy. The medical imaging modalities include magnetic resonance imaging, computed tomography, positron emission tomography, single photon emission computerized tomography, optical imaging, ultrasound, and photoacoustic imaging. Various cancer therapeutic methods will also be included, including photothermal therapy, photodynamic therapy, chemotherapy, and immunotherapy. This review also covers theranostics, which use the same agent in diagnosis and therapy. This includes recent advances in multimodality imaging, image-guided therapy, and combination therapy. We found that the continuous advances of synthesis and design of novel nanomaterials will enhance the future development of medical imaging and cancer therapy. However, more resources should be available to examine side effects and cell toxicity when using nanomaterials in humans.
Collapse
Affiliation(s)
- Sarkar Siddique
- Department of Physics, Ryerson University, Toronto, ON M5B 2K3, Canada;
| | - James C. L. Chow
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1X6, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
90
|
Luthria G, Li R, Wang S, Prytyskach M, Kohler RH, Lauffenburger DA, Mitchison TJ, Weissleder R, Miller MA. In vivo microscopy reveals macrophage polarization locally promotes coherent microtubule dynamics in migrating cancer cells. Nat Commun 2020; 11:3521. [PMID: 32665556 PMCID: PMC7360550 DOI: 10.1038/s41467-020-17147-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/02/2020] [Indexed: 01/07/2023] Open
Abstract
Microtubules (MTs) mediate mitosis, directional signaling, and are therapeutic targets in cancer. Yet in vivo analysis of cancer cell MT behavior within the tumor microenvironment remains challenging. Here we developed an imaging pipeline using plus-end tip tracking and intravital microscopy to quantify MT dynamics in live xenograft tumor models. Among analyzed features, cancer cells in vivo displayed higher coherent orientation of MT dynamics along their cell major axes compared with 2D in vitro cultures, and distinct from 3D collagen gel cultures. This in vivo MT phenotype was reproduced in vitro when cells were co-cultured with IL4-polarized MΦ. MΦ depletion, MT disruption, targeted kinase inhibition, and altered MΦ polarization via IL10R blockade all reduced MT coherence and/or tumor cell elongation. We show that MT coherence is a defining feature for in vivo tumor cell dynamics and migration, modulated by local signaling from pro-tumor macrophages. The regulation of microtubule (MT) dynamics in cancer cells within the tumor microenvironment is less understood. Here, the authors develop an imaging platform to examine MT dynamics in live xenograft models and show that pro-tumor macrophages modulate MT coherence and alignment to promote cancer cell migration.
Collapse
Affiliation(s)
- Gaurav Luthria
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Stephanie Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02181, USA
| | - Mark Prytyskach
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02181, USA
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA. .,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115, USA. .,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA. .,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
91
|
Wang SJ, Li R, Ng TSC, Luthria G, Oudin MJ, Prytyskach M, Kohler RH, Weissleder R, Lauffenburger DA, Miller MA. Efficient blockade of locally reciprocated tumor-macrophage signaling using a TAM-avid nanotherapy. SCIENCE ADVANCES 2020; 6:eaaz8521. [PMID: 32494745 PMCID: PMC7244320 DOI: 10.1126/sciadv.aaz8521] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/20/2020] [Indexed: 05/07/2023]
Abstract
Interpreting how multicellular interactions in the tumor affect resistance pathways to BRAF and MEK1/2 MAPK inhibitors (MAPKi) remains a challenge. To investigate this, we profiled global ligand-receptor interactions among tumor and stromal/immune cells from biopsies of MAPK-driven disease. MAPKi increased tumor-associated macrophages (TAMs) in some patients, which correlated with poor clinical response, and MAPKi coamplified bidirectional tumor-TAM signaling via receptor tyrosine kinases (RTKs) including AXL, MERTK, and their ligand GAS6. In xenograft tumors, intravital microscopy simultaneously monitored in situ single-cell activities of multiple kinases downstream of RTKs, revealing MAPKi increased TAMs and enhanced bypass signaling in TAM-proximal tumor cells. As a proof-of-principle strategy to block this signaling, we developed a multi-RTK kinase inhibitor nanoformulation that accumulated in TAMs and delayed disease progression. Thus, bypass signaling can reciprocally amplify across nearby cell types, offering new opportunities for therapeutic design.
Collapse
Affiliation(s)
- Stephanie J. Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Thomas S. C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Gaurav Luthria
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Mark Prytyskach
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
92
|
Cancer Immunoimaging with Smart Nanoparticles. Trends Biotechnol 2020; 38:388-403. [DOI: 10.1016/j.tibtech.2019.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/31/2022]
|
93
|
Wang P, Kim T, Harada M, Contag C, Huang X, Smith BR. Nano-immunoimaging. NANOSCALE HORIZONS 2020; 5:628-653. [PMID: 32226975 DOI: 10.1039/c9nh00514e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunoimaging is a rapidly growing field stoked in large part by the intriguing triumphs of immunotherapy. On the heels of immunotherapy's successes, there exists a growing need to evaluate tumor response to therapy particularly immunotherapy, stratify patients into responders vs. non-responders, identify inflammation, and better understand the fundamental roles of immune system components to improve both immunoimaging and immunotherapy. Innovative nanomaterials have begun to provide novel opportunities for immunoimaging, in part due to their sensitivity, modularity, capacity for many potentially varied ligands (high avidity), and potential for multifunctionality/multimodality imaging. This review strives to comprehensively summarize the integration of nanotechnology and immunoimaging, and the field's potential for clinical applications.
Collapse
Affiliation(s)
- Ping Wang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA
| | - Taeho Kim
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 488824, USA
| | - Xuefei Huang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Chemistry, Michigan State University, East Lansing, MI 488824, USA
| | - Bryan Ronain Smith
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Radiology, Stanford University, Stanford, CA 94306, USA
| |
Collapse
|
94
|
The expanding landscape of inflammatory cells affecting cancer therapy. Nat Biomed Eng 2020; 4:489-498. [PMID: 32203281 DOI: 10.1038/s41551-020-0524-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022]
Abstract
Tumour-infiltrating myeloid cells (TIMCs) are critical regulators of cancer growth. The different phenotypes, functions and therapeutic effects of these phagocytes have, however, been difficult to study. With the advent of single-cell-based technologies, a new 'worldview' is emerging: the classification of TIMCs into subtypes that are conserved across patients and across species. As the landscape of TIMCs is beginning to be understood, it opens up questions about the function of each TIMC subtype and its drugability. In this Perspective, we outline the current map of TIMC populations in cancer and their known and presumed functions, and discuss their therapeutic implications and the biological research questions that they give rise to. The answers should be particularly relevant for bioengineers, materials scientists and the chemical and pharmaceutical communities developing the next generation of cancer therapies.
Collapse
|
95
|
Abstract
Therapeutic targeting of the immune system in cancer is now a clinical reality and marked successes have been achieved, most notably through the use of checkpoint blockade antibodies and chimeric antigen receptor T cell therapy. However, efforts to develop new immunotherapy agents or combination treatments to increase the proportion of patients who benefit have met with challenges of limited efficacy and/or significant toxicity. Nanomedicines - therapeutics composed of or formulated in carrier materials typically smaller than 100 nm - were originally developed to increase the uptake of chemotherapy agents by tumours and to reduce their off-target toxicity. Here, we discuss how nanomedicine-based treatment strategies are well suited to immunotherapy on the basis of nanomaterials' ability to direct immunomodulators to tumours and lymphoid organs, to alter the way biologics engage with target immune cells and to accumulate in myeloid cells in tumours and systemic compartments. We also discuss early efforts towards clinical translation of nanomedicine-based immunotherapy.
Collapse
|
96
|
Koch PD, Rodell CB, Kohler RH, Pittet MJ, Weissleder R. Myeloid Cell-Targeted Nanocarriers Efficiently Inhibit Cellular Inhibitor of Apoptosis for Cancer Immunotherapy. Cell Chem Biol 2020; 27:94-104.e5. [PMID: 31902676 DOI: 10.1016/j.chembiol.2019.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/02/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023]
Abstract
Immune-checkpoint blockers can promote sustained clinical responses in a subset of cancer patients. Recent research has shown that a subpopulation of tumor-infiltrating dendritic cells functions as gatekeepers, sensitizing tumors to anti-PD-1 treatment via production of interleukin-12 (IL-12). Hypothesizing that myeloid cell-targeted nanomaterials could be used to deliver small-molecule IL-12 inducers, we performed high-content image-based screening to identify the most efficacious small-molecule compounds. Using one lead candidate, LCL161, we created a myeloid-targeted nanoformulation that induced IL-12 production in intratumoral myeloid cells in vivo, slowed tumor growth as a monotherapy, and had no significant systemic toxicity. These results pave the way for developing combination immunotherapeutics by harnessing IL-12 production for immunostimulation.
Collapse
Affiliation(s)
- Peter D Koch
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Christopher B Rodell
- 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
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
97
|
Gabizon AA, de Rosales RT, La-Beck NM. Translational considerations in nanomedicine: The oncology perspective. Adv Drug Deliv Rev 2020; 158:140-157. [PMID: 32526450 DOI: 10.1016/j.addr.2020.05.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/28/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022]
Abstract
Nanoparticles can provide effective control of the release rate and tissue distribution of their drug payload, leading to major pharmacokinetic and pharmacodynamic changes vis-à-vis the conventional administration of free drugs. In the last two decades, we have witnessed major progress in the synthesis and characterization of engineered nanoparticles for imaging and treatment of cancers, resulting in the approval for clinical use of several products and in new and promising approaches. Despite these advances, clinical applications of nanoparticle-based therapeutic and imaging agents remain limited due to biological, immunological, and translational barriers. There is a need to make high impact advances toward translation. In this review, we address biological, toxicological, immunological, and translational aspects of nanomedicine and discuss approaches to move the field forward productively. Overcoming these barriers may dramatically improve the development potential and role of nanomedicines in the oncology field and help meet the high expectations.
Collapse
|
98
|
Ng TS, Garlin MA, Weissleder R, Miller MA. Improving nanotherapy delivery and action through image-guided systems pharmacology. Theranostics 2020; 10:968-997. [PMID: 31938046 PMCID: PMC6956809 DOI: 10.7150/thno.37215] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in the translation of therapeutic nanoparticles (TNPs) into the clinic, the field continues to face challenges in predictably and selectively delivering nanomaterials for the treatment of solid cancers. The concept of enhanced permeability and retention (EPR) has been coined as a convenient but simplistic descriptor of high TNP accumulation in some tumors. However, in practice EPR represents a number of physiological variables rather than a single one (including dysfunctional vasculature, compromised lymphatics and recruited host cells, among other aspects of the tumor microenvironment) — each of which can be highly heterogenous within a given tumor, patient and across patients. Therefore, a clear need exists to dissect the specific biophysical factors underlying the EPR effect, to formulate better TNP designs, and to identify patients with high-EPR tumors who are likely to respond to TNP. The overall pharmacology of TNP is governed by an interconnected set of spatially defined and dynamic processes that benefit from a systems-level quantitative approach, and insights into the physiology have profited from the marriage between in vivo imaging and quantitative systems pharmacology (QSP) methodologies. In this article, we review recent developments pertinent to image-guided systems pharmacology of nanomedicines in oncology. We first discuss recent developments of quantitative imaging technologies that enable analysis of nanomaterial pharmacology at multiple spatiotemporal scales, and then examine reports that have adopted these imaging technologies to guide QSP approaches. In particular, we focus on studies that have integrated multi-scale imaging with computational modeling to derive insights about the EPR effect, as well as studies that have used modeling to guide the manipulation of the EPR effect and other aspects of the tumor microenvironment for improving TNP action. We anticipate that the synergistic combination of imaging with systems-level computational methods for effective clinical translation of TNPs will only grow in relevance as technologies increase in resolution, multiplexing capability, and in the ability to examine heterogeneous behaviors at the single-cell level.
Collapse
|
99
|
He W, Kapate N, Shields CW, Mitragotri S. Drug delivery to macrophages: A review of targeting drugs and drug carriers to macrophages for inflammatory diseases. Adv Drug Deliv Rev 2019; 165-166:15-40. [PMID: 31816357 DOI: 10.1016/j.addr.2019.12.001] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022]
Abstract
Macrophages play a key role in defending against foreign pathogens, healing wounds, and regulating tissue homeostasis. Driving this versatility is their phenotypic plasticity, which enables macrophages to respond to subtle cues in tightly coordinated ways. However, when this coordination is disrupted, macrophages can aid the progression of numerous diseases, including cancer, cardiovascular disease, and autoimmune disease. The central link between these disorders is aberrant macrophage polarization, which misguides their functional programs, secretory products, and regulation of the surrounding tissue microenvironment. As a result of their important and deterministic roles in both health and disease, macrophages have gained considerable attention as targets for drug delivery. Here, we discuss the role of macrophages in the initiation and progression of various inflammatory diseases, summarize the leading drugs used to regulate macrophages, and review drug delivery systems designed to target macrophages. We emphasize strategies that are approved for clinical use or are poised for clinical investigation. Finally, we provide a prospectus of the future of macrophage-targeted drug delivery systems.
Collapse
Affiliation(s)
- Wei He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Neha Kapate
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
100
|
Yang L, Gradl R, Dierolf M, Möller W, Kutschke D, Feuchtinger A, Hehn L, Donnelley M, Günther B, Achterhold K, Walch A, Stoeger T, Razansky D, Pfeiffer F, Morgan KS, Schmid O. Multimodal Precision Imaging of Pulmonary Nanoparticle Delivery in Mice: Dynamics of Application, Spatial Distribution, and Dosimetry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904112. [PMID: 31639283 DOI: 10.1002/smll.201904112] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/12/2019] [Indexed: 06/10/2023]
Abstract
Targeted delivery of nanomedicine/nanoparticles (NM/NPs) to the site of disease (e.g., the tumor or lung injury) is of vital importance for improved therapeutic efficacy. Multimodal imaging platforms provide powerful tools for monitoring delivery and tissue distribution of drugs and NM/NPs. This study introduces a preclinical imaging platform combining X-ray (two modes) and fluorescence imaging (three modes) techniques for time-resolved in vivo and spatially resolved ex vivo visualization of mouse lungs during pulmonary NP delivery. Liquid mixtures of iodine (contrast agent for X-ray) and/or (nano)particles (X-ray absorbing and/or fluorescent) are delivered to different regions of the lung via intratracheal instillation, nasal aspiration, and ventilator-assisted aerosol inhalation. It is demonstrated that in vivo propagation-based phase-contrast X-ray imaging elucidates the dynamic process of pulmonary NP delivery, while ex vivo fluorescence imaging (e.g., tissue-cleared light sheet fluorescence microscopy) reveals the quantitative 3D drug/particle distribution throughout the entire lung with cellular resolution. The novel and complementary information from this imaging platform unveils the dynamics and mechanisms of pulmonary NM/NP delivery and deposition for each of the delivery routes, which provides guidance on optimizing pulmonary delivery techniques and novel-designed NM for targeting and efficacy.
Collapse
Affiliation(s)
- Lin Yang
- Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), Munich, 81377, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Faculty of Medicine, Technical University of Munich, Munich, 80333, Germany
| | - Regine Gradl
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Munich School of BioEngineering, Technical University of Munich, Garching, 85748, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, 85748, Germany
| | - Martin Dierolf
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Munich School of BioEngineering, Technical University of Munich, Garching, 85748, Germany
| | - Winfried Möller
- Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), Munich, 81377, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - David Kutschke
- Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), Munich, 81377, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Lorenz Hehn
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, München, 81675, Germany
| | - Martin Donnelley
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, 5000, Australia
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, 5006, Australia
| | - Benedikt Günther
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Munich School of BioEngineering, Technical University of Munich, Garching, 85748, Germany
| | - Klaus Achterhold
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Munich School of BioEngineering, Technical University of Munich, Garching, 85748, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Tobias Stoeger
- Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), Munich, 81377, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Daniel Razansky
- Faculty of Medicine, Technical University of Munich, Munich, 80333, Germany
- Institute for Biological and Medical Imaging (IBMI), Helmholtz Zentrum München, Neuherberg, 85764, Germany
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, CH-8057, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Franz Pfeiffer
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Munich School of BioEngineering, Technical University of Munich, Garching, 85748, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, 85748, Germany
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, München, 81675, Germany
| | - Kaye S Morgan
- Chair of Biomedical Physics, Department of Physics, Technical University of Munich, Garching, 85748, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, 85748, Germany
- School of Physics and Astronomy, Monash University, Clayton, Victoria, 3800, Australia
| | - Otmar Schmid
- Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), Munich, 81377, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany
| |
Collapse
|