101
|
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
|
102
|
Shofolawe-Bakare OT, Stokes LD, Hossain M, Smith AE, Werfel TA. Immunostimulatory biomaterials to boost tumor immunogenicity. Biomater Sci 2020; 8:5516-5537. [PMID: 33049007 PMCID: PMC7837217 DOI: 10.1039/d0bm01183e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is exhibiting great promise as a new therapeutic modality for cancer treatment. However, immunotherapies are limited by the inability of some tumors to provoke an immune response. These tumors with a 'cold' immunological phenotype are characterized by low numbers of tumor-infiltrating lymphocytes, high numbers of immunosuppressive leukocytes (e.g. regulatory T cells, tumor-associated macrophages), and high production of immune-dampening signals (e.g. IL-10, TGF-β, IDO-1). Strategies to boost the aptitude of tumors to initiate an immune response (i.e. boost tumor immunogenicity) will turn 'cold' tumors 'hot' and augment the anti-tumor efficacy of current immunotherapies. Approaches to boost tumor immunogenicity already show promise; however, multifaceted delivery and immunobiology challenges exist. For instance, systemic delivery of many immune-stimulating agents causes off-target toxicity and/or the development of autoimmunity, limiting the administrable dose below the threshold needed to achieve efficacy. Moreover, once administered in vivo, molecules such as the nucleic acid-based agonists for many pattern recognition receptors are either rapidly cleared or degraded, and don't efficiently traffic to the intracellular compartments where the receptors are located. Thus, these nucleic acid-based drugs are ineffective without a delivery system. Biomaterials-based approaches aim to enhance current strategies to boost tumor immunogenicity, enable novel strategies, and spare dose-limiting toxicities. Here, we review recent progress to improve cancer immunotherapies by boosting immunogenicity within tumors using immunostimulatory biomaterials.
Collapse
|
103
|
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
|
104
|
Mi P, Miyata K, Kataoka K, Cabral H. Clinical Translation of Self‐Assembled Cancer Nanomedicines. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000159] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center West China Hospital, Sichuan University No. 17 People's South Road Chengdu 610041 China
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| | - Kazunori Kataoka
- Institute for Future Initiatives The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐0033 Japan
- Innovation Center of NanoMedicine Kawasaki Institute of Industrial Promotion 3‐25‐14, Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| |
Collapse
|
105
|
Visualization of the distribution of nanoparticle-formulated AZD2811 in mouse tumor model using matrix-assisted laser desorption ionization mass spectrometry imaging. Sci Rep 2020; 10:15535. [PMID: 32968211 PMCID: PMC7511311 DOI: 10.1038/s41598-020-72665-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Penetration of nanoparticles into viable tumor regions is essential for an effective response. Mass spectrometry imaging (MSI) is a novel method for evaluating the intratumoral pharmacokinetics (PK) of a drug in terms of spatial distribution. The application of MSI for analysis of nanomedicine PK remains in its infancy. In this study, we evaluated the applicability of MALDI-MSI for nanoparticle-formulated drug visualization in tumors and biopsies, with an aim toward future application in clinical nanomedicine research. We established an analytic method for the free drug (AZD2811) and then applied it to visualize nanoparticle-formulated AZD2811. MSI analysis demonstrated heterogeneous intratumoral drug distribution in three xenograft tumors. The intensity of MSI signals correlated well with total drug concentration in tumors, indicating that drug distribution can be monitored quantitatively. Analysis of tumor biopsies indicated that MSI is applicable for analyzing the distribution of nanoparticle-formulated drugs in tumor biopsies, suggesting clinical applicability.
Collapse
|
106
|
Kong N, Tao W, Ling X, Wang J, Xiao Y, Shi S, Ji X, Shajii A, Gan ST, Kim NY, Duda DG, Xie T, Farokhzad OC, Shi J. Synthetic mRNA nanoparticle-mediated restoration of p53 tumor suppressor sensitizes p53-deficient cancers to mTOR inhibition. Sci Transl Med 2020; 11:11/523/eaaw1565. [PMID: 31852795 DOI: 10.1126/scitranslmed.aaw1565] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 08/23/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022]
Abstract
Loss of function in tumor suppressor genes is commonly associated with the onset/progression of cancer and treatment resistance. The p53 tumor suppressor gene, a master regulator of diverse cellular pathways, is frequently altered in various cancers, for example, in ~36% of hepatocellular carcinomas (HCCs) and ~68% of non-small cell lung cancers (NSCLCs). Current methods for restoration of p53 expression, including small molecules and DNA therapies, have yielded progressive success, but each has formidable drawbacks. Here, a redox-responsive nanoparticle (NP) platform is engineered for effective delivery of p53-encoding synthetic messenger RNA (mRNA). We demonstrate that the synthetic p53-mRNA NPs markedly delay the growth of p53-null HCC and NSCLC cells by inducing cell cycle arrest and apoptosis. We also reveal that p53 restoration markedly improves the sensitivity of these tumor cells to everolimus, a mammalian target of rapamycin (mTOR) inhibitor that failed to show clinical benefits in advanced HCC and NSCLC. Moreover, cotargeting of tumor-suppressing p53 and tumorigenic mTOR signaling pathways results in marked antitumor effects in vitro and in multiple animal models of HCC and NSCLC. Our findings indicate that restoration of tumor suppressors by the synthetic mRNA NP delivery strategy could be combined together with other therapies for potent combinatorial cancer treatment.
Collapse
Affiliation(s)
- Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Xiang Ling
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Junqing Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuling Xiao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sanjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoyuan Ji
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Cancer Pharmacology, Holistic Integrative Pharmacy Institutes, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Aram Shajii
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Silvia Tian Gan
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Na Yoon Kim
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dan G Duda
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tian Xie
- Department of Cancer Pharmacology, Holistic Integrative Pharmacy Institutes, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. .,King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
107
|
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
|
108
|
Szafraniec-Szczęsny J, Janik-Hazuka M, Odrobińska J, Zapotoczny S. Polymer Capsules with Hydrophobic Liquid Cores as Functional Nanocarriers. Polymers (Basel) 2020; 12:E1999. [PMID: 32887444 PMCID: PMC7565928 DOI: 10.3390/polym12091999] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Recent developments in the fabrication of core-shell polymer nanocapsules, as well as their current and future applications, are reported here. Special attention is paid to the newly introduced surfactant-free fabrication method of aqueous dispersions of nanocapsules with hydrophobic liquid cores stabilized by amphiphilic copolymers. Various approaches to the efficient stabilization of such vehicles, tailoring their cores and shells for the fabrication of multifunctional, navigable nanocarriers and/or nanoreactors useful in various fields, are discussed. The emphasis is placed on biomedical applications of polymer nanocapsules, including the delivery of poorly soluble active compounds and contrast agents, as well as their use as theranostic platforms. Other methods of fabrication of polymer-based nanocapsules are briefly presented and compared in the context of their biomedical applications.
Collapse
Affiliation(s)
- Joanna Szafraniec-Szczęsny
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Małgorzata Janik-Hazuka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (M.J.-H.); (J.O.)
| | - Joanna Odrobińska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (M.J.-H.); (J.O.)
| | - Szczepan Zapotoczny
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (M.J.-H.); (J.O.)
| |
Collapse
|
109
|
Joo JI, Choi M, Jang SH, Choi S, Park SM, Shin D, Cho KH. Realizing Cancer Precision Medicine by Integrating Systems Biology and Nanomaterial Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906783. [PMID: 32253807 DOI: 10.1002/adma.201906783] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/19/2019] [Indexed: 06/11/2023]
Abstract
Many clinical trials for cancer precision medicine have yielded unsatisfactory results due to challenges such as drug resistance and low efficacy. Drug resistance is often caused by the complex compensatory regulation within the biomolecular network in a cancer cell. Recently, systems biological studies have modeled and simulated such complex networks to unravel the hidden mechanisms of drug resistance and identify promising new drug targets or combinatorial or sequential treatments for overcoming resistance to anticancer drugs. However, many of the identified targets or treatments present major difficulties for drug development and clinical application. Nanocarriers represent a path forward for developing therapies with these "undruggable" targets or those that require precise combinatorial or sequential application, for which conventional drug delivery mechanisms are unsuitable. Conversely, a challenge in nanomedicine has been low efficacy due to heterogeneity of cancers in patients. This problem can also be resolved through systems biological approaches by identifying personalized targets for individual patients or promoting the drug responses. Therefore, integration of systems biology and nanomaterial engineering will enable the clinical application of cancer precision medicine to overcome both drug resistance of conventional treatments and low efficacy of nanomedicine due to patient heterogeneity.
Collapse
Affiliation(s)
- Jae Il Joo
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Minsoo Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seong-Hoon Jang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sea Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sang-Min Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongkwan Shin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
110
|
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
|
111
|
Bu Y, Hu Q, Zhang X, Li T, Xie X, Wang S. A novel cell membrane-cloaked magnetic nanogripper with enhanced stability for drug discovery. Biomater Sci 2020; 8:673-681. [PMID: 31769454 DOI: 10.1039/c9bm01411j] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cell membrane-cloaked nanotechnology has attracted increasing attention owing to its unique bionic properties, such as specific recognition and biocompatibility conferred by the integrated membrane structure and receptors. However, this technology is limited by the dissociation of the cell membrane from its carrier. Here, we report a novel type of cell membrane-cloaked modified magnetic nanoparticle with good stability in drug discovery. High α1A-adrenergic receptor (α1A-AR) expressing HEK293 cell membrane-cloaked magnetic nanogrippers (α1A/MNGs) were used as a platform for the specific targeting and binding of α1A-AR antagonists as candidate bioactive compounds from traditional Chinese medicine (TCM). Furthermore, using a dynamic covalent bonding approach, α1A/MNGs showed great stability with positive control drug recoveries of α1A/MNGs showing almost no decline after use in five adsorption-desorption cycles. Moreover, the α1A/MNGs possessed a unilamellar membrane with magnetic features and exhibited good binding capacity and selectivity. Ultimately, TCM and pharmacological studies of the bioactivity of the screened compounds confirmed the considerable targeting and binding capability of α1A/MNGs. Application of aldehyde group modification in this drug-targeting concept further improved biomaterial stability and paves the way for the development of new drug discovery strategies. More importantly, the successful application of α1A/MNGs provides new insights into methodologies to improve the integration of cell membranes with the nanoparticle platform.
Collapse
Affiliation(s)
- Yusi Bu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | | | | | | | | | | |
Collapse
|
112
|
Shi Y, van der Meel R, Chen X, Lammers T. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Am J Cancer Res 2020; 10:7921-7924. [PMID: 32685029 PMCID: PMC7359085 DOI: 10.7150/thno.49577] [Citation(s) in RCA: 481] [Impact Index Per Article: 96.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Following its discovery more than 30 years ago, the enhanced permeability and retention (EPR) effect has become the guiding principle for cancer nanomedicine development. Over the years, the tumor-targeted drug delivery field has made significant progress, as evidenced by the approval of several nanomedicinal anticancer drugs. Recently, however, the existence and the extent of the EPR effect - particularly in patients - have become the focus of intense debate. This is partially due to the disbalance between the huge number of preclinical cancer nanomedicine papers and relatively small number of cancer nanomedicine drug products reaching the market. To move the field forward, we have to improve our understanding of the EPR effect, of its cancer type-specific pathophysiology, of nanomedicine interactions with the heterogeneous tumor microenvironment, of nanomedicine behavior in the body, and of translational aspects that specifically complicate nanomedicinal drug development. In this virtual special issue, 24 research articles and reviews discussing different aspects of the EPR effect and cancer nanomedicine are collected, together providing a comprehensive and complete overview of the current state-of-the-art and future directions in tumor-targeted drug delivery.
Collapse
|
113
|
Russell LM, Liu CH, Grodzinski P. Nanomaterials innovation as an enabler for effective cancer interventions. Biomaterials 2020; 242:119926. [PMID: 32169771 DOI: 10.1016/j.biomaterials.2020.119926] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Nanomedicines have been developing very rapidly and have started to play a significant role in several cancer therapeutic modalities. Early on, the nanomedicine field focused on optimizing pharmacokinetics, toxicity, and/or biodistribution of an agent through nanoparticle formulation. In other cases, where materials science is employed more decisively, nanomedicine can include the creation of new agents that take advantage of nanoscale materials properties to enhance treatment efficacy through unique mode of action, molecular targeting, or controlled drug release. Both current and future nanomedicines will seek to contribute to the therapeutic and diagnostic landscape through creative leveraging of mechanical, electrical, optical, magnetic, and biological nanomaterial properties. In this work, we discuss how by modulating these material properties, one can design more diverse and more effective cancer interventions. We focus on six areas in cancer management, including in vitro diagnostics, clinical imaging, theranostics, combination therapy, immunotherapy, and gene therapy.
Collapse
Affiliation(s)
- Luisa M Russell
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina H Liu
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
114
|
Ullah R, Wazir J, Khan FU, Diallo MT, Ihsan AU, Mikrani R, Aquib M, Zhou X. Factors Influencing the Delivery Efficiency of Cancer Nanomedicines. AAPS PharmSciTech 2020; 21:132. [PMID: 32409932 DOI: 10.1208/s12249-020-01691-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/13/2020] [Indexed: 01/01/2023] Open
Abstract
The superiority of nanomedicine over conventional medicines in the treatment of cancer has gained immediate recognition worldwide. As traditional cancer therapies are nonspecific and detrimental to healthy cells, the ability of nanomedicine to release drugs to target tumor cells specifically instead of healthy cells has brought new hope to cancer patients. This review focuses on the effects of various factors of nanoparticles such as transport, concentration in cells, tumor microenvironment, interaction with protein, penetration, uptake by tumor cells, cancer cell mutations, and intracellular trafficking of the nanoparticle. Besides the history of nanomedicine, future perspectives of nanomedicines are also explored in this text.
Collapse
|
115
|
Ploetz E, Zimpel A, Cauda V, Bauer D, Lamb DC, Haisch C, Zahler S, Vollmar AM, Wuttke S, Engelke H. Metal-Organic Framework Nanoparticles Induce Pyroptosis in Cells Controlled by the Extracellular pH. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1907267. [PMID: 32182391 DOI: 10.1002/adma.201907267] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 05/21/2023]
Abstract
Ion homeostasis is essential for cellular survival, and elevated concentrations of specific ions are used to start distinct forms of programmed cell death. However, investigating the influence of certain ions on cells in a controlled way has been hampered due to the tight regulation of ion import by cells. Here, it is shown that lipid-coated iron-based metal-organic framework nanoparticles are able to deliver and release high amounts of iron ions into cells. While high concentrations of iron often trigger ferroptosis, here, the released iron induces pyroptosis, a form of cell death involving the immune system. The iron release occurs only in slightly acidic extracellular environments restricting cell death to cells in acidic microenvironments and allowing for external control. The release mechanism is based on endocytosis facilitated by the lipid-coating followed by degradation of the nanoparticle in the lysosome via cysteine-mediated reduction, which is enhanced in slightly acidic extracellular environment. Thus, a new functionality of hybrid nanoparticles is demonstrated, which uses their nanoarchitecture to facilitate controlled ion delivery into cells. Based on the selectivity for acidic microenvironments, the described nanoparticles may also be used for immunotherapy: the nanoparticles may directly affect the primary tumor and the induced pyroptosis activates the immune system.
Collapse
Affiliation(s)
- Evelyn Ploetz
- Department of Chemistry and Center for NanoScience (CeNS), LMU Munich, Munich, 81377, Germany
- Nanosystems Initiative Munich (NIM), LMU Munich, Munich, 81377, Germany
- Center for Integrated Protein Science Munich (CiPSM), LMU Munich, Munich, 81377, Germany
| | - Andreas Zimpel
- Department of Chemistry and Center for NanoScience (CeNS), LMU Munich, Munich, 81377, Germany
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Torino, 10129, Italy
| | - David Bauer
- Department of Chemistry, TU Munich, Munich, 81377, Germany
| | - Don C Lamb
- Department of Chemistry and Center for NanoScience (CeNS), LMU Munich, Munich, 81377, Germany
- Nanosystems Initiative Munich (NIM), LMU Munich, Munich, 81377, Germany
- Center for Integrated Protein Science Munich (CiPSM), LMU Munich, Munich, 81377, Germany
| | | | - Stefan Zahler
- Department of Pharmacy, LMU Munich, Munich, 81377, Germany
| | | | - Stefan Wuttke
- BCMaterials, Basque Center for Materials, UPV/EHU Science Park, Leioa, 48940, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain
| | - Hanna Engelke
- Department of Chemistry and Center for NanoScience (CeNS), LMU Munich, Munich, 81377, Germany
| |
Collapse
|
116
|
Ho D, Quake SR, McCabe ERB, Chng WJ, Chow EK, Ding X, Gelb BD, Ginsburg GS, Hassenstab J, Ho CM, Mobley WC, Nolan GP, Rosen ST, Tan P, Yen Y, Zarrinpar A. Enabling Technologies for Personalized and Precision Medicine. Trends Biotechnol 2020; 38:497-518. [PMID: 31980301 PMCID: PMC7924935 DOI: 10.1016/j.tibtech.2019.12.021] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Individualizing patient treatment is a core objective of the medical field. Reaching this objective has been elusive owing to the complex set of factors contributing to both disease and health; many factors, from genes to proteins, remain unknown in their role in human physiology. Accurately diagnosing, monitoring, and treating disorders requires advances in biomarker discovery, the subsequent development of accurate signatures that correspond with dynamic disease states, as well as therapeutic interventions that can be continuously optimized and modulated for dose and drug selection. This work highlights key breakthroughs in the development of enabling technologies that further the goal of personalized and precision medicine, and remaining challenges that, when addressed, may forge unprecedented capabilities in realizing truly individualized patient care.
Collapse
Affiliation(s)
- Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore; The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore; Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, CA, USA; Department of Applied Physics, Stanford University, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Wee Joo Chng
- Department of Haematology and Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Edward K Chow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xianting Ding
- Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University, NC, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis, MO, USA; Psychological & Brain Sciences, Washington University in St. Louis, MO, USA
| | - Chih-Ming Ho
- Department of Mechanical Engineering, University of California, Los Angeles, CA, USA
| | - William C Mobley
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University, CA, USA
| | - Steven T Rosen
- Comprehensive Cancer Center and Beckman Research Institute, City of Hope, CA, USA
| | - Patrick Tan
- Duke-NUS Medical School, National University of Singapore, Singapore
| | - Yun Yen
- College of Medical Technology, Center of Cancer Translational Research, Taipei Cancer Center of Taipei Medical University, Taipei, Taiwan
| | - Ali Zarrinpar
- Department of Surgery, Division of Transplantation & Hepatobiliary Surgery, University of Florida, FL, USA
| |
Collapse
|
117
|
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
|
118
|
Mi P, Cabral H, Kataoka K. Ligand-Installed Nanocarriers toward Precision Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1902604. [PMID: 31353770 DOI: 10.1002/adma.201902604] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/04/2019] [Indexed: 05/20/2023]
Abstract
Development of drug-delivery systems that selectively target neoplastic cells has been a major goal of nanomedicine. One major strategy for achieving this milestone is to install ligands on the surface of nanocarriers to enhance delivery to target tissues, as well as to enhance internalization of nanocarriers by target cells, which improves accuracy, efficacy, and ultimately enhances patient outcomes. Herein, recent advances regarding the development of ligand-installed nanocarriers are introduced and the effect of their design on biological performance is discussed. Besides academic achievements, progress on ligand-installed nanocarriers in clinical trials is presented, along with the challenges faced by these formulations. Lastly, the future perspectives of ligand-installed nanocarriers are discussed, with particular emphasis on their potential for emerging precision therapies.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), 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
|
119
|
Adir O, Poley M, Chen G, Froim S, Krinsky N, Shklover J, Shainsky-Roitman J, Lammers T, Schroeder A. Integrating Artificial Intelligence and Nanotechnology for Precision Cancer Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901989. [PMID: 31286573 PMCID: PMC7124889 DOI: 10.1002/adma.201901989] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/17/2019] [Indexed: 05/13/2023]
Abstract
Artificial intelligence (AI) and nanotechnology are two fields that are instrumental in realizing the goal of precision medicine-tailoring the best treatment for each cancer patient. Recent conversion between these two fields is enabling better patient data acquisition and improved design of nanomaterials for precision cancer medicine. Diagnostic nanomaterials are used to assemble a patient-specific disease profile, which is then leveraged, through a set of therapeutic nanotechnologies, to improve the treatment outcome. However, high intratumor and interpatient heterogeneities make the rational design of diagnostic and therapeutic platforms, and analysis of their output, extremely difficult. Integration of AI approaches can bridge this gap, using pattern analysis and classification algorithms for improved diagnostic and therapeutic accuracy. Nanomedicine design also benefits from the application of AI, by optimizing material properties according to predicted interactions with the target drug, biological fluids, immune system, vasculature, and cell membranes, all affecting therapeutic efficacy. Here, fundamental concepts in AI are described and the contributions and promise of nanotechnology coupled with AI to the future of precision cancer medicine are reviewed.
Collapse
Affiliation(s)
- Omer Adir
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Maria Poley
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Gal Chen
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Sahar Froim
- Department of Physical Electronics, School of Electrical Engineering, Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nitzan Krinsky
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Jeny Shklover
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Janna Shainsky-Roitman
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, 52074, Germany
| | - Avi Schroeder
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
120
|
Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020; 10:4557-4588. [PMID: 32292515 PMCID: PMC7150471 DOI: 10.7150/thno.38069] [Citation(s) in RCA: 358] [Impact Index Per Article: 71.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
In recent years, much progress has been motivated in stimuli-responsive nanocarriers, which could response to the intrinsic physicochemical and pathological factors in diseased regions to increase the specificity of drug delivery. Currently, numerous nanocarriers have been engineered with physicochemical changes in responding to external stimuli, such as ultrasound, thermal, light and magnetic field, as well as internal stimuli, including pH, redox potential, hypoxia and enzyme, etc. Nanocarriers could respond to stimuli in tumor microenvironments or inside cancer cells for on-demanded drug delivery and accumulation, controlled drug release, activation of bioactive compounds, probes and targeting ligands, as well as size, charge and conformation conversion, etc., leading to sensing and signaling, overcoming multidrug resistance, accurate diagnosis and precision therapy. This review has summarized the general strategies of developing stimuli-responsive nanocarriers and recent advances, presented their applications in drug delivery, tumor imaging, therapy and theranostics, illustrated the progress of clinical translation and made prospects.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, 610041, China
| |
Collapse
|
121
|
Gaspar N, Zambito G, Löwik CMWG, Mezzanotte L. Active Nano-targeting of Macrophages. Curr Pharm Des 2020; 25:1951-1961. [PMID: 31291874 DOI: 10.2174/1381612825666190710114108] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
Macrophages play a role in almost every disease such as cancer, infections, injuries, metabolic and inflammatory diseases and are becoming an attractive therapeutic target. However, understanding macrophage diversity, tissue distribution and plasticity will help in defining precise targeting strategies and effective therapies. Active targeting of macrophages using nanoparticles for therapeutic purposes is still at its infancy but holds promises since macrophages have shown high specific uptake of nanoparticles. Here, we highlight recent progress in active nanotechnology-based systems gaining pivotal roles to target diverse macrophage subsets in diseased tissues.
Collapse
Affiliation(s)
- Natasa Gaspar
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Percuros B.V., Department of Developmental BioEngineering, University of Twente, Enschede, Netherlands
| | - Giorgia Zambito
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Medres-Medical Research gmbh, Cologne, Germany
| | - Clemens M W G Löwik
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Oncology, Lausanne University Hospital (CHUV), UNIL, Switzerland
| | - Laura Mezzanotte
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
122
|
Sorrin AJ, Ruhi MK, Ferlic NA, Karimnia V, Polacheck WJ, Celli JP, Huang HC, Rizvi I. Photodynamic Therapy and the Biophysics of the Tumor Microenvironment. Photochem Photobiol 2020; 96:232-259. [PMID: 31895481 PMCID: PMC7138751 DOI: 10.1111/php.13209] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Targeting the tumor microenvironment (TME) provides opportunities to modulate tumor physiology, enhance the delivery of therapeutic agents, impact immune response and overcome resistance. Photodynamic therapy (PDT) is a photochemistry-based, nonthermal modality that produces reactive molecular species at the site of light activation and is in the clinic for nononcologic and oncologic applications. The unique mechanisms and exquisite spatiotemporal control inherent to PDT enable selective modulation or destruction of the TME and cancer cells. Mechanical stress plays an important role in tumor growth and survival, with increasing implications for therapy design and drug delivery, but remains understudied in the context of PDT and PDT-based combinations. This review describes pharmacoengineering and bioengineering approaches in PDT to target cellular and noncellular components of the TME, as well as molecular targets on tumor and tumor-associated cells. Particular emphasis is placed on the role of mechanical stress in the context of targeted PDT regimens, and combinations, for primary and metastatic tumors.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Mustafa Kemal Ruhi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
| | - Nathaniel A. Ferlic
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Vida Karimnia
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA, 02125, USA
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jonathan P. Celli
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA, 02125, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
123
|
Tan T, Wang Y, Wang J, Wang Z, Wang H, Cao H, Li J, Li Y, Zhang Z, Wang S. Targeting peptide-decorated biomimetic lipoproteins improve deep penetration and cancer cells accessibility in solid tumor. Acta Pharm Sin B 2020; 10:529-545. [PMID: 32140397 PMCID: PMC7049576 DOI: 10.1016/j.apsb.2019.05.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022] Open
Abstract
The limited penetration of nanoparticles and their poor accessibility to cancer cell fractions in tumor remain essential challenges for effective anticancer therapy. Herein, we designed a targeting peptide-decorated biomimetic lipoprotein (termed as BL-RD) to enable their deep penetration and efficient accessibility to cancer cell fractions in a tumor, thereby improving the combinational chemo-photodynamic therapy of triple negative breast cancer. BL-RD was composed of phospholipids, apolipoprotein A1 mimetic peptide (PK22), targeting peptide-conjugated cytotoxic mertansine (RM) and photodynamic agents of DiIC18(5) (DiD). The counterpart biomimetic lipoprotein system without RM (termed as BL-D) was fabricated as control. Both BL-D and BL-RD were nanometer-sized particles with a mean diameter of less than 30 nm and could be efficiently internalized by cancer cells. After intravenous injection, they can be specifically accumulated at tumor sites. When comparing to the counterpart BL-D, BL-RD displayed superior capability to permeate across the tumor mass, extravasate from tumor vasculature to distant regions and efficiently access the cancer cell fractions in a solid tumor, thus producing noticeable depression of the tumor growth. Taken together, BL-RD can be a promising delivery nanoplatform with prominent tumor-penetrating and cancer cells-accessing capability for effective tumor therapy.
Collapse
Key Words
- 4T1-GFP, 4T1 cancer cells with stable expression of green fluorescence protein
- ApoA1, apolipoprotein A1
- BL-D, biomimetic lipoprotein system without targeting peptide
- BL-RD, targeting peptide decorated biomimetic lipoprotein system
- CAF, cancer-associated fibroblasts
- CLSM, confocal laser scanning microscopy
- Cancer therapy
- DAPI, 4′,6-diamidino-2-phenylindole
- DCFH-DA, 2′,7′-dichlorodihydrofluorescein diacetate
- DOPC, 1,2-dioleoyl-sn-glycero-3-phosphocholine
- DiD, DiIC18(5)
- Drug delivery
- EC, endothelial cells
- ECM, extracellular matrix
- EE, encapsulation efficiency
- FBS, fetal bovine serum
- GSH, glutathione
- H&E staining, hematoxylin-eosin staining
- HDL, high density lipoprotein
- HPLC, high performance liquid chromatography
- IC50, half-inhibitory concentration
- Lipo-D, liposome system without targeting peptide
- Lipo-RD, targeting peptide decorated biomimetic lipoprotein system
- Lipoprotein
- MCS, multicellular spheroids
- MTT, thiazolyl blue tetrazolium bromide
- Nanoparticles
- PBS, phosphate buffered solution
- PDT, photodynamic therapy
- RM, targeting peptide-conjugated cytotoxic mertansine
- ROS, reactive oxygen species
- SOSG, singlet oxygen sensor green
- TAM, tumor-associated macrophage
- TEM, transmission electronic microscope
- TGI, tumor growth index
- Tumor penetration
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Tao Tan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuqi Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwan Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hong Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haiqiang Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| | - Siling Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| |
Collapse
|
124
|
Dasgupta A, Biancacci I, Kiessling F, Lammers T. Imaging-assisted anticancer nanotherapy. Theranostics 2020; 10:956-967. [PMID: 31938045 PMCID: PMC6956808 DOI: 10.7150/thno.38288] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/11/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer nanomedicines are submicrometer-sized formulations designed to improve the biodistribution of anticancer drugs, resulting in less off-target localization, altered toxicity profiles, improved target site accumulation and enhanced efficacy. Together, these beneficial features have resulted in the regulatory approval of about a dozen nanomedicines for the treatment of solid and hematological malignancies. In recent years, significant progress has been made in combining nanomedicines with imaging, to better understand key aspects of the tumor-targeted drug delivery process, and to address the high inter- and intra-individual heterogeneity in the Enhanced Permeability and Retention (EPR) effect. Strategies explored in this regard have included the use of traditional imaging techniques, companion diagnostics and nanotheranostics. Preclinically, integrating imaging in nanomedicine and drug delivery research has enabled the non-invasive and quantitative assessment of nanocarrier biodistribution, target site accumulation and (triggered) drug release. Clinically, imaging has been emerging as a promising tool for patient stratification, which is urgently needed to improve the translation of cancer nanomedicines. We here summarize recent progress in imaging-assisted anticancer nanotherapy and we discuss future strategies to improve the performance of cancer nanomedicines in patients.
Collapse
|
125
|
Zhao M, van Straten D, Broekman ML, Préat V, Schiffelers RM. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics 2020; 10:1355-1372. [PMID: 31938069 PMCID: PMC6956816 DOI: 10.7150/thno.38147] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
The current achievements in treating glioblastoma (GBM) patients are not sufficient because many challenges exist, such as tumor heterogeneity, the blood brain barrier, glioma stem cells, drug efflux pumps and DNA damage repair mechanisms. Drug combination therapies have shown increasing benefits against those challenges. With the help of nanocarriers, enhancement of the efficacy and safety could be gained using synergistic combinations of different therapeutic agents. In this review, we will discuss the major issues for GBM treatment, the rationales of drug combinations with or without nanocarriers and the principle of enhanced permeability and retention effect involved in nanomedicine-based tumor targeting and promising nanodiagnostics or -therapeutics. We will also summarize the recent progress and discuss the clinical perspectives of nanocarrier-based combination therapies. The goal of this article was to provide better understanding and key considerations to develop new nanomedicine combinations and nanotheranostics options to fight against GBM.
Collapse
Affiliation(s)
- Mengnan Zhao
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1 73.12, 1200 Brussels, Belgium
| | - Demian van Straten
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Marike L.D. Broekman
- Department of Neurosurgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1 73.12, 1200 Brussels, Belgium
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| |
Collapse
|
126
|
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
|
127
|
Harisinghani MG, O’Shea A, Weissleder R. Advances in clinical MRI technology. Sci Transl Med 2019; 11:11/523/eaba2591. [DOI: 10.1126/scitranslmed.aba2591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/03/2019] [Indexed: 11/02/2022]
Abstract
Advances in MRI technologies have the potential to detect, characterize, and monitor a wide variety of diseases.
Collapse
Affiliation(s)
- Mukesh G. Harisinghani
- Department of Radiology, Massachusetts General Hospital, Fruit Street, Boston, MA 02114, USA
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Aileen O’Shea
- Department of Radiology, Massachusetts General Hospital, Fruit Street, Boston, MA 02114, USA
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Ralph Weissleder
- Department of Radiology, Massachusetts General Hospital, Fruit Street, Boston, MA 02114, USA
- 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
|
128
|
Wu K, Su D, Liu J, Saha R, Wang JP. Magnetic nanoparticles in nanomedicine: a review of recent advances. NANOTECHNOLOGY 2019; 30:502003. [PMID: 31491782 DOI: 10.1088/1361-6528/ab4241] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Nanomaterials, in addition to their small size, possess unique physicochemical properties that differ from bulk materials, making them ideal for a host of novel applications. Magnetic nanoparticles (MNPs) are one important class of nanomaterials that have been widely studied for their potential applications in nanomedicine. Due to the fact that MNPs can be detected and manipulated by remote magnetic fields, it opens a wide opportunity for them to be used in vivo. Nowadays, MNPs have been used for diverse applications including magnetic biosensing (diagnostics), magnetic imaging, magnetic separation, drug and gene delivery, and hyperthermia therapy, etc. Specifically, we reviewed some emerging techniques in magnetic diagnostics such as magnetoresistive (MR) and micro-Hall (μHall) biosensors, as well as the magnetic particle spectroscopy, magnetic relaxation switching and surface enhanced Raman spectroscopy (SERS)-based bioassays. Recent advances in applying MNPs as contrast agents in magnetic resonance imaging and as tracer materials in magnetic particle imaging are reviewed. In addition, the development of high magnetic moment MNPs with proper surface functionalization has progressed exponentially over the past decade. To this end, different MNP synthesis approaches and surface coating strategies are reviewed and the biocompatibility and toxicity of surface functionalized MNP nanocomposites are also discussed. Herein, we are aiming to provide a comprehensive assessment of the state-of-the-art biological and biomedical applications of MNPs. This review is not only to provide in-depth insights into the different synthesis, biofunctionalization, biosensing, imaging, and therapy methods but also to give an overview of limitations and possibilities of each technology.
Collapse
Affiliation(s)
- Kai Wu
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | | | | | | | | |
Collapse
|
129
|
Kapitanova KS, Naumenko VA, Garanina AS, Melnikov PA, Abakumov MA, Alieva IB. Advances and Challenges of Nanoparticle-Based Macrophage Reprogramming for Cancer Immunotherapy. BIOCHEMISTRY (MOSCOW) 2019; 84:729-745. [PMID: 31509725 DOI: 10.1134/s0006297919070058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite the progress of modern medicine, oncological diseases are still among the most common causes of death of adult populations in developed countries. The current therapeutic approaches are imperfect, and the high mortality of oncological patients under treatment, the lack of personalized strategies, and severe side effects arising as a result of treatment force seeking new approaches to therapy of malignant tumors. During the last decade, cancer immunotherapy, an approach that relies on activation of the host antitumor immune response, has been actively developing. Cancer immunotherapy is the most promising trend in contemporary fundamental and practical oncology, and restoration of the pathologically altered tumor microenvironment is one of its key tasks, in particular, the reprogramming of tumor macrophages from the immunosuppressive M2-phenotype into the proinflammatory M1-phenotype is pivotal for eliciting antitumor response. This review describes the current knowledge about macrophage classification, mechanisms of their polarization, their role in formation of the tumor microenvironment, and strategies for changing the functional activity of M2-macrophages, as well as problems of targeted delivery of immunostimulatory signals to tumor macrophages using nanoparticles.
Collapse
Affiliation(s)
- K S Kapitanova
- Lomonosov Moscow State University, Department of Bioengineering and Bioinformatics, Moscow, 119991, Russia
| | - V A Naumenko
- National University of Science and Technology "MISIS", Moscow, 119049, Russia.
| | - A S Garanina
- National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - P A Melnikov
- Serbsky Federal Medical Research Center of Psychiatry and Narcology, Department of Fundamental and Applied Neurobiology, Ministry of Health of the Russian Federation, Moscow, 119034, Russia
| | - M A Abakumov
- National University of Science and Technology "MISIS", Moscow, 119049, Russia.,Russian National Research Medical University, Department of Medical Nanobiotechnology, Moscow, 117997, Russia
| | - I B Alieva
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
130
|
Rodell CB, Ahmed MS, Garris CS, Pittet MJ, Weissleder R. Development of Adamantane-Conjugated TLR7/8 Agonists for Supramolecular Delivery and Cancer Immunotherapy. Theranostics 2019; 9:8426-8436. [PMID: 31879528 PMCID: PMC6924259 DOI: 10.7150/thno.35434] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/11/2019] [Indexed: 12/27/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are often abundant in solid cancers, assuming an immunosuppressive (M2-like) phenotype which supports tumor growth and immune escape. Recent methods have focused on identification of means (e.g., drugs, nanomaterials) that polarize TAMs to a tumor suppressive (M1-like) phenotype; however, reducing the systemic side effects of these therapies and enabling their delivery to TAMs has remained a challenge. Methods: Here, we develop R848-Ad, an adamantane-modified derivative of the toll-like receptor (TLR) 7/8 agonist resiquimod (R848) through iterative drug screening against reporter cell lines. The adamantane undergoes guest-host interaction with cyclodextrin nanoparticles (CDNPs), enabling drug loading under aqueous conditions and TAM-targeted drug delivery. Therapeutic efficacy and systemic side effects were examined in a murine MC38 cancer model. Results: R848-Ad retained macrophage polarizing activity through agonization of TLR7/8, and the adamantane moiety improved drug affinity for the CDNP. In preclinical studies, nanoformulated R848-Ad resulted in a drastic reduction in measurable systemic effects (loss of body weight) relative to similarly formulated R848 alone while arresting tumor growth. Conclusions: The findings demonstrate the ability of strong nanoparticle-drug interactions to limit systemic toxicity of TLR agonists while simultaneously maintaining therapeutic efficacy.
Collapse
Affiliation(s)
| | - Maaz S. Ahmed
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Christopher S. Garris
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Graduate Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Mikael J. Pittet
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
131
|
Naumenko VA, Vlasova KY, Garanina AS, Melnikov PA, Potashnikova DM, Vishnevskiy DA, Vodopyanov SS, Chekhonin VP, Abakumov MA, Majouga AG. Extravasating Neutrophils Open Vascular Barrier and Improve Liposomes Delivery to Tumors. ACS NANO 2019; 13:12599-12612. [PMID: 31609576 DOI: 10.1021/acsnano.9b03848] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Liposomes are the most extensively used nanocarriers in cancer therapy. Despite the advantages these vehicles provide over free drugs, there are still limitations with regards to the efficiency of liposomes delivery to tumors and off-target accumulation. A better understanding of nanodrugs extravasation mechanisms in different tumor types and normal vessels is needed to improve their antitumor activity. We used intravital microscopy to track for fluorescent liposomes behavior in xenograft tumor models (murine breast cancer 4T1 and melanoma B16, human prostate cancer 22Rv1) and normal skin and identified two distinct extravasation patterns. Microleakage, a local perivascular nanoparticle deposition, was found both in malignant and healthy tissues. This type of liposomes leakage does not provide access to tumor cells and is presumably responsible for drug deposition in normal tissues. In contrast, macroleakage penetrated deep into tissues and localized predominantly on the tumor-host interface. Although neutrophils did not uptake liposomes, their extravasation appeared to initiate both micro- and macroleakages. Based on neutrophils and liposomes extravasation dynamics, we hypothesized that microleakage and macroleakage are subsequent steps of the extravasation process corresponding to liposomes transport through endothelial and subendothelial barriers. Of note, extravasation spots were detected more often in the proximity of neutrophils, and across studied tumor types, neutrophils counts correlated with leakage frequencies. Reduced liposomes accumulation in 4T1 tumors upon Ly6G depletion further corroborated neutrophils role in nanoparticles delivery. Elucidating liposomes extravasation routes has a potential to help improve existing strategies and develop effective nanodrugs for cancer therapy.
Collapse
Affiliation(s)
- Victor A Naumenko
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
| | - Kseniya Yu Vlasova
- School of Chemistry , M. V. Lomonosov Moscow State University , Moscow 119991 , Russia
| | | | - Pavel A Melnikov
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Daria M Potashnikova
- School of Biology, Department of Cell Biology and Histology , M. V. Lomonosov Moscow State University , Moscow 119234 , Russia
| | - Daniil A Vishnevskiy
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Stepan S Vodopyanov
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
| | - Vladimir P Chekhonin
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Maxim A Abakumov
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Alexander G Majouga
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
- D. Mendeleev University of Chemical Technology of Russia , Moscow 125047 , Russia
| |
Collapse
|
132
|
Samanta K, Setua S, Kumari S, Jaggi M, Yallapu MM, Chauhan SC. Gemcitabine Combination Nano Therapies for Pancreatic Cancer. Pharmaceutics 2019; 11:E574. [PMID: 31689930 PMCID: PMC6920852 DOI: 10.3390/pharmaceutics11110574] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is one of the deadliest causes of cancer-related death in the United States, with a 5-year overall survival rate of 6 to 8%. These statistics suggest that immediate medical attention is needed. Gemcitabine (GEM) is the gold standard first-line single chemotherapy agent for pancreatic cancer but, after a few months, cells develop chemoresistance. Multiple clinical and experimental investigations have demonstrated that a combination or co-administration of other drugs as chemotherapies with GEM lead to superior therapeutic benefits. However, such combination therapies often induce severe systemic toxicities. Thus, developing strategies to deliver a combination of chemotherapeutic agents more securely to patients is needed. Nanoparticle-mediated delivery can offer to load a cocktail of drugs, increase stability and availability, on-demand and tumor-specific delivery while minimizing chemotherapy-associated adverse effects. This review discusses the available drugs being co-administered with GEM and the limitations associated during the process of co-administration. This review also helps in providing knowledge of the significant number of delivery platforms being used to overcome problems related to gemcitabine-based co-delivery of other chemotherapeutic drugs, thereby focusing on how nanocarriers have been fabricated, considering the modes of action, targeting receptors, pharmacology of chemo drugs incorporated with GEM, and the differences in the physiological environment where the targeting is to be done. This review also documents the focus on novel mucin-targeted nanotechnology which is under development for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Kamalika Samanta
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sonam Kumari
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Murali M Yallapu
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| |
Collapse
|
133
|
Hartshorn CM, Russell LM, Grodzinski P. National Cancer Institute Alliance for nanotechnology in cancer-Catalyzing research and translation toward novel cancer diagnostics and therapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1570. [PMID: 31257722 PMCID: PMC6788937 DOI: 10.1002/wnan.1570] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022]
Abstract
Nanotechnology has been a burgeoning research field, which is finding compelling applications in several practical areas of everyday life. It has provided novel, paradigm shifting solutions to medical problems and particularly to cancer. In order to accelerate integration of nanotechnology into cancer research and oncology, the National Cancer Institute (NCI) of the National Institutes of Health (NIH) established the NCI Alliance for Nanotechnology in Cancer program in 2005. This effort brought together scientists representing physical sciences, chemistry, and engineering working at the nanoscale with biologists and clinicians working on cancer to form a uniquely multidisciplinary cancer nanotechnology research community. The last 14 years of the program have produced a remarkable body of scientific discovery and demonstrated its utility to the development of practical cancer interventions. This paper takes stock of how the Alliance program influenced melding of disparate research disciplines into the field of nanomedicine and cancer nanotechnology, has been highly productive in the scientific arena, and produced a mechanism of seamless transfer of novel technologies developed in academia to the clinical and commercial space. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Christopher M. Hartshorn
- Nanodelivery Systems and Devices Branch, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Luisa M. Russell
- Nanodelivery Systems and Devices Branch, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, USA
| |
Collapse
|
134
|
van der Meel R, Sulheim E, Shi Y, Kiessling F, Mulder WJM, Lammers T. Smart cancer nanomedicine. NATURE NANOTECHNOLOGY 2019; 14:1007-1017. [PMID: 31695150 PMCID: PMC7227032 DOI: 10.1038/s41565-019-0567-y] [Citation(s) in RCA: 781] [Impact Index Per Article: 130.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/30/2019] [Indexed: 05/19/2023]
Abstract
Nanomedicines are extensively employed in cancer therapy. We here propose four strategic directions to improve nanomedicine translation and exploitation. (1) Patient stratification has become common practice in oncology drug development. Accordingly, probes and protocols for patient stratification are urgently needed in cancer nanomedicine, to identify individuals suitable for inclusion in clinical trials. (2) Rational drug selection is crucial for clinical and commercial success. Opportunistic choices based on drug availability should be replaced by investments in modular (pro)drug and nanocarrier design. (3) Combination therapies are the mainstay of clinical cancer care. Nanomedicines synergize with pharmacological and physical co-treatments, and should be increasingly integrated in multimodal combination therapy regimens. (4) Immunotherapy is revolutionizing the treatment of cancer. Nanomedicines can modulate the behaviour of myeloid and lymphoid cells, thereby empowering anticancer immunity and immunotherapy efficacy. Alone and especially together, these four directions will fuel and foster the development of successful cancer nanomedicine therapies.
Collapse
Affiliation(s)
- Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
- Cancer Clinic, St. Olavs University Hospital, Trondheim, Norway
| | - Yang Shi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Willem J M Mulder
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany.
- Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands.
- Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
135
|
Characterization of Redox-Responsive LXR-Activating Nanoparticle Formulations in Primary Mouse Macrophages. Molecules 2019; 24:molecules24203751. [PMID: 31635211 PMCID: PMC6833070 DOI: 10.3390/molecules24203751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Activation of the transcription factor liver X receptor (LXR) has beneficial effects on macrophage lipid metabolism and inflammation, making it a potential candidate for therapeutic targeting in cardiometabolic disease. While small molecule delivery via nanomedicine has promising applications for a number of chronic diseases, questions remain as to how nanoparticle formulation might be tailored to suit different tissue microenvironments and aid in drug delivery. In the current study, we aimed to compare the in vitro drug delivering capability of three nanoparticle (NP) formulations encapsulating the LXR activator, GW-3965. We observed little difference in the base characteristics of standard PLGA-PEG NP when compared to two redox-active polymeric NP formulations, which we called redox-responsive (RR)1 and RR2. Moreover, we also observed similar uptake of these NP into primary mouse macrophages. We used the transcript and protein expression of the cholesterol efflux protein and LXR target ATP-binding cassette A1 (ABCA1) as a readout of GW-3956-induced LXR activation. Following an initial acute uptake period that was meant to mimic circulating exposure in vivo, we determined that although the induction of transcript expression was similar between NPs, treatment with the redox-sensitive RR1 NPs resulted in a higher level of ABCA1 protein. Our results suggest that NP formulations responsive to cellular cues may be an effective tool for targeted and disease-specific drug release.
Collapse
|
136
|
Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: Designing a new perspective in nanomedicine delivery. Theranostics 2019; 9:8091-8108. [PMID: 31754383 PMCID: PMC6857058 DOI: 10.7150/thno.37204] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
The enhanced permeability and retention (EPR) effect has underlain the predominant nanomedicine design philosophy for the past three decades. However, growing evidence suggests that it is over-represented in preclinical models, and agents designed solely using its principle of passive accumulation can only be applied to a narrow subset of clinical tumors. For this reason, strategies that can improve upon the EPR effect to facilitate nanomedicine delivery to otherwise non-responsive tumors are required for broad clinical translation. EPR-adaptive nanomedicine delivery comprises a class of chemical and physical techniques that modify tumor accessibility in an effort to increase agent delivery and therapeutic effect. In the present review, we overview the primary benefits and limitations of radiation, ultrasound, hyperthermia, and photodynamic therapy as physical strategies for EPR-adaptive delivery to EPR-insensitive tumor phenotypes, and we reflect upon changes in the preclinical research pathway that should be implemented in order to optimally validate and develop these delivery strategies.
Collapse
Affiliation(s)
- Alexander Dhaliwal
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- MD/PhD Program, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON Canada
| |
Collapse
|
137
|
Liu X, Jiang J, Meng H. Transcytosis - An effective targeting strategy that is complementary to "EPR effect" for pancreatic cancer nano drug delivery. Theranostics 2019; 9:8018-8025. [PMID: 31754378 PMCID: PMC6857052 DOI: 10.7150/thno.38587] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 08/16/2019] [Indexed: 01/10/2023] Open
Abstract
Numerous nano drug delivery systems have been developed for preclinical cancer research in the past 15 years with the hope for a fundamental change in oncology. The robust nanotherapeutic research has yielded early-stage clinical products as exemplified by the FDA-approved nano formulations (Abraxane® for paclitaxel and Onyvide® for irinotecan) for the treatment of solid tumors, including pancreatic ductal adenocarcinoma (PDAC). It is generally believed that enhanced permeability and retention (EPR) plays a key role in nanocarriers' accumulation in preclinical tumor models and is a clinically relevant phenomenon in certain cancer types. However, use of EPR effect as an across-the-board explanation for nanoparticle tumor access is likely over-simplified, particularly in the stroma rich solid tumors such as PDAC. Recently, ample evidences including our own data showed that it is possible to use transcytosis as a major mechanism for PDAC drug delivery. In this mini-review, we summarize the key studies that discuss how transcytosis can be employed to enhance EPR effect in PDAC, and potentially, other cancer malignancies. We also mentioned other vasculature engineering approaches that work beyond the classic EPR effect.
Collapse
Affiliation(s)
- Xiangsheng Liu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Huan Meng
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
138
|
Rodell CB, Koch PD, Weissleder R. Screening for new macrophage therapeutics. Theranostics 2019; 9:7714-7729. [PMID: 31695796 PMCID: PMC6831478 DOI: 10.7150/thno.34421] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid derived macrophages play a key role in many human diseases, and their therapeutic modulation via pharmacological means is receiving considerable attention. Of particular interest is the fact that these cells are i) dynamic phenotypes well suited to therapeutic manipulation and ii) phagocytic, allowing them to be efficiently targeted with nanoformulations. However, it is important to consider that macrophages represent heterogeneous populations of subtypes with often competing biological behaviors and functions. In order to develop next generation therapeutics, it is therefore essential to screen for biological effects through a combination of in vitro and in vivo assays. Here, we review the state-of-the-art techniques, including both cell based screens and in vivo imaging tools that have been developed for assessment of macrophage phenotype. We conclude with a forward-looking perspective on the growing need for noninvasive macrophage assessment and laboratory assays to be put into clinical practice and the potential broader impact of myeloid-targeted therapeutics.
Collapse
|
139
|
Lazaro-Carrillo A, Filice M, Guillén MJ, Amaro R, Viñambres M, Tabero A, Paredes KO, Villanueva A, Calvo P, Del Puerto Morales M, Marciello M. Tailor-made PEG coated iron oxide nanoparticles as contrast agents for long lasting magnetic resonance molecular imaging of solid cancers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 107:110262. [PMID: 31761230 DOI: 10.1016/j.msec.2019.110262] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/06/2019] [Accepted: 09/26/2019] [Indexed: 11/17/2022]
Abstract
Magnetic resonance imaging (MRI) is the most powerful technique for non-invasive diagnosis of human diseases and disorders. Properly designed contrast agents can be accumulated in the damaged zone and be internalized by cells, becoming interesting cellular MRI probes for disease tracking and monitoring. However, this approach is sometimes limited by the relaxation rates of contrast agents currently in clinical use, which show neither optimal pharmacokinetic parameters nor toxicity. In this work, a suitable contrast agent candidate, based on iron oxide nanoparticles (IONPs) coated with polyethyleneglycol, was finely designed, prepared and fully characterized under a physical, chemical and biological point of view. To stand out the real potential of our study, all the experiments were performed in comparison with Ferumoxytol, a FDA approved IONPs. IONPs with a core size of 15 nm and coated with polyethyleneglycol of 5 kDa (OD15-P5) resulted the best ones, being able to be uptaken by both tumoral cells and macrophages and showing no toxicity for in vitro and in vivo experiments. In vitro and in vivo MRI results for OD15-P5 showed r2 relaxivity values higher than Ferumoxitol. Furthermore, the injected OD15-P5 were completely retained at the tumor site for up to 24 h showing high potential as MRI contrast agents for real time long-lasting monitoring of the tumor evolution.
Collapse
Affiliation(s)
- Ana Lazaro-Carrillo
- Department of Biology, Universidad Autónoma de Madrid (UAM), Darwin 2, Cantoblanco, 28049, Madrid, Spain
| | - Marco Filice
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid (UCM), Plaza Ramón y Cajal, 28040, Madrid, Spain; Biomedical Research Networking Center for Respiratory Diseases (CIBERES), C/Melchor Fernandez-Almagro 3, 28029, Madrid, Spain; Fundacion Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - María José Guillén
- Research Department, PharmaMar S.A, Colmenar Viejo, 28770, Madrid, Spain
| | - Rebeca Amaro
- Department of Energy, Environment and Health, Institute of Materials Science of Madrid, ICMM-CSIC, Sor Juana Inés de La Cruz 3, Cantoblanco, 28049, Madrid, Spain
| | - Mario Viñambres
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid (UCM), Plaza Ramón y Cajal, 28040, Madrid, Spain; Department of Energy, Environment and Health, Institute of Materials Science of Madrid, ICMM-CSIC, Sor Juana Inés de La Cruz 3, Cantoblanco, 28049, Madrid, Spain
| | - Andrea Tabero
- Department of Biology, Universidad Autónoma de Madrid (UAM), Darwin 2, Cantoblanco, 28049, Madrid, Spain
| | - Karina Ovejero Paredes
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid (UCM), Plaza Ramón y Cajal, 28040, Madrid, Spain; Fundacion Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Angeles Villanueva
- Department of Biology, Universidad Autónoma de Madrid (UAM), Darwin 2, Cantoblanco, 28049, Madrid, Spain; Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, Campus Universitario de Cantoblanco, 28049, Madrid, Spain
| | - Pilar Calvo
- Research Department, PharmaMar S.A, Colmenar Viejo, 28770, Madrid, Spain
| | - Maria Del Puerto Morales
- Department of Energy, Environment and Health, Institute of Materials Science of Madrid, ICMM-CSIC, Sor Juana Inés de La Cruz 3, Cantoblanco, 28049, Madrid, Spain
| | - Marzia Marciello
- Nanobiotechnology for Life Sciences Group, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid (UCM), Plaza Ramón y Cajal, 28040, Madrid, Spain; Department of Energy, Environment and Health, Institute of Materials Science of Madrid, ICMM-CSIC, Sor Juana Inés de La Cruz 3, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
140
|
Chen L, Wu Y, Wu H, Li J, Xie J, Zang F, Ma M, Gu N, Zhang Y. Magnetic targeting combined with active targeting of dual-ligand iron oxide nanoprobes to promote the penetration depth in tumors for effective magnetic resonance imaging and hyperthermia. Acta Biomater 2019; 96:491-504. [PMID: 31302299 DOI: 10.1016/j.actbio.2019.07.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 06/28/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022]
Abstract
The combination of multi-targeting magnetic nanoprobes and multi-targeting strategies has potential to facilitate magnetic resonance imaging (MRI) and magnetic induction hyperthermia of the tumor. Although the thermo-agents based on magnetic iron oxide nanoparticles (MION) have been successfully used in the form of intratumoral injection in clinical cure of glioblastoma, the tumor-targeted thermotherapy by intravenous administration remains challenging. Herein, we constructed a c(RGDyK)- and d-glucosamine-grafted bispecific molecular nanoprobe (Fe3O4@RGD@GLU) with a magnetic iron oxide core of size 22.17 nm and a biocompatible shell of DSPE-PEG2000, which can specially target the tumor vessel and cancer cells. The selection of c(RGDyK) could make the nanoprobe enter the neovascularization endotheliocyte through αvβ3-mediated endocytosis, which drastically reduced the dependence on the enhanced permeability and retention (EPR) effect in tumor. This dual-ligand nanoprobe exhibited strong magnetic properties and favorable biocompatibility. In vitro studies confirmed the anti-phagocytosis ability against macrophages and the specific targeting capability of Fe3O4@RGD@GLU. Then, the imaging effect and anti-tumor efficacy were compared using different targeting strategies with untargeted nanoprobes, dual-targeted nanoprobes, and magnetic targeting combined with dual-targeted nanoprobes. Moreover, the combination strategy of magnetic targeting and active targeting promoted the penetration depth of nanoprobes in addition to the increased accumulation in tumor tissue. Thus, the dual-targeted magnetic nanoprobe together with the combined targeting strategy could be a promising method in tumor imaging and hyperthermia through in vivo delivery of theranostic agents. STATEMENT OF SIGNIFICANCE: Magnetic induction hyperthermia based on iron oxide nanoparticles has been used in clinic for adjuvant treatment of recurrent glioblastoma. Nonetheless, this application is limited to intratumoral injection, and tumor-targeted hyperthermia by intravenous injection remains challenging. In this study, we developed a multi-targeted strategy by combining magnetic targeting with active targeting of dual-ligand magnetic nanoprobes. This combination mode acquired optimum contrast imaging effect through MRI and tumor-suppressive effect through hyperthermia under an alternating current magnetic field. The design of the nanoprobe was suitable for targeting most tumor lesions, which enabled it to be an effective theranostic agent with extensive uses. This study showed significant enhancement of the penetration depth and accumulation of nanoprobes in the tumor tissue for efficient imaging and hyperthermia.
Collapse
|
141
|
Lee SH, Park OK, Kim J, Shin K, Pack CG, Kim K, Ko G, Lee N, Kwon SH, Hyeon T. Deep Tumor Penetration of Drug-Loaded Nanoparticles by Click Reaction-Assisted Immune Cell Targeting Strategy. J Am Chem Soc 2019; 141:13829-13840. [PMID: 31382746 DOI: 10.1021/jacs.9b04621] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nanoparticles have been extensively used to deliver therapeutic drugs to tumor tissues through the extravasation of a leaky vessel via enhanced permeation and retention effect (EPR, passive targeting) or targeted interaction of tumor-specific ligands (active targeting). However, the therapeutic efficacy of drug-loaded nanoparticles is hampered by its heterogeneous distribution owing to limited penetration in tumor tissue. Inspired by the fact that cancer cells can recruit inflammatory immune cells to support their survival, we developed a click reaction-assisted immune cell targeting (CRAIT) strategy to deliver drug-loaded nanoparticles deep into the avascular regions of the tumor. Immune cell-targeting CD11b antibodies are modified with trans-cyclooctene to enable bioorthogonal click chemistry with mesoporous silica nanoparticles functionalized with tetrazines (MSNs-Tz). Sequential injection of modified antibodies and MSNs-Tz at intervals of 24 h results in targeted conjugation of the nanoparticles onto CD11b+ myeloid cells, which serve as active vectors into tumor interiors. We show that the CRAIT strategy allows the deep tumor penetration of drug-loaded nanoparticles, resulting in enhanced therapeutic efficacy in an orthotopic 4T1 breast tumor model. The CRAIT strategy does not require ex vivo manipulation of cells and can be applied to various types of cells and nanovehicles.
Collapse
Affiliation(s)
- Soo Hong Lee
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| | - Jonghoon Kim
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| | - Kwangsoo Shin
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| | - Chan Gi Pack
- Asan Institute for Life Sciences, Asan Medical Center, Department of Convergence Medicine , University of Ulsan College of Medicine , Seoul 05505 , Republic of Korea
| | - Kang Kim
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| | - Giho Ko
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| | - Nohyun Lee
- School of Advanced Materials Engineering , Kookmin University , Seoul 02707 , Republic of Korea
| | - Seung-Hae Kwon
- Division of Bio-imaging, Korea Basic Science Institute , Seoul 02841 , Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 08826 , Republic of Korea.,School of Chemical and Biological Engineering, and Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| |
Collapse
|
142
|
Shamsi M, Mohammadi A, Manshadi MK, Sanati-Nezhad A. Mathematical and computational modeling of nano-engineered drug delivery systems. J Control Release 2019; 307:150-165. [DOI: 10.1016/j.jconrel.2019.06.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022]
|
143
|
Yong SB, Chung JY, Song Y, Kim J, Ra S, Kim YH. Non-viral nano-immunotherapeutics targeting tumor microenvironmental immune cells. Biomaterials 2019; 219:119401. [PMID: 31398571 DOI: 10.1016/j.biomaterials.2019.119401] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022]
Abstract
The tumor microenvironmental immune cells (TMICs) consists of myeloid cells (tumor-associated macrophages, dendritic cells, myeloid-derived suppressor cells, etc.) and lymphocytes (T cells and B cells), all of which could be immunologically suppressed through their interactions with cancer cells. Immunological understanding of the tumor microenvironment (TME) has led to great success in the development of clinical cancer immunotherapeutic. The most advanced cancer immunotherapies are chimeric antigen receptor-modified T cells (CAR-T cells) and checkpoint inhibiting antibodies blocking CTLA4, PD-1 and PD-L1. However, many hurdles remain that should be addressed for improved therapeutic efficacy and reduced side effects such as cytokine release syndrome and patient-death. In recent decades, nanoparticles have been demonstrated as an efficient drug delivery tool due to their ease of modification, biocompatibility and intrinsic tumor targeting effect, and also been applied for cancer immunotherapy. In this review, we briefly introduce the immunosuppressive functions of TMICs and review recent advances in the development of TMIC-targeted nanotherapeutics for cancer immunotherapy. Tumor-associated macrophage (TAM)-targeted systems have shown to deplete or repolarize macrophages to M1 state for anti-tumoral immune responses. Tumor-infiltrating T cell (TIT)-targeted strategies have provided the activation of effector T cells and suppression of regulatory T cells in tumor, overcoming the current hurdles of single regimen checkpoint inhibitors. Lastly, recent studies on dendritic cell-targeted mRNA vaccination are discussed and the future perspectives of nano-immunotherapeutic for next-generation of cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Seok-Beom Yong
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Jee Young Chung
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Yoonsung Song
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Jaehyun Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Sehee Ra
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea.
| |
Collapse
|
144
|
SoRelle ED, Yecies DW, Liba O, Bennett FC, Graef CM, Dutta R, Mitra S, Joubert LM, Cheshier S, Grant GA, de la Zerda A. Spatiotemporal Tracking of Brain-Tumor-Associated Myeloid Cells in Vivo through Optical Coherence Tomography with Plasmonic Labeling and Speckle Modulation. ACS NANO 2019; 13:7985-7995. [PMID: 31259527 PMCID: PMC8144904 DOI: 10.1021/acsnano.9b02656] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
By their nature, tumors pose a set of profound challenges to the immune system with respect to cellular recognition and response coordination. Recent research indicates that leukocyte subpopulations, especially tumor-associated macrophages (TAMs), can exert substantial influence on the efficacy of various cancer immunotherapy treatment strategies. To better study and understand the roles of TAMs in determining immunotherapeutic outcomes, significant technical challenges associated with dynamically monitoring single cells of interest in relevant live animal models of solid tumors must be overcome. However, imaging techniques with the requisite combination of spatiotemporal resolution, cell-specific contrast, and sufficient signal-to-noise at increasing depths in tissue are exceedingly limited. Here we describe a method to enable high-resolution, wide-field, longitudinal imaging of TAMs based on speckle-modulating optical coherence tomography (SM-OCT) and spectral scattering from an optimized contrast agent. The approach's improvements to OCT detection sensitivity and noise reduction enabled high-resolution OCT-based observation of individual cells of a specific host lineage in live animals. We found that large gold nanorods (LGNRs) that exhibit a narrow-band, enhanced scattering cross-section can selectively label TAMs and activate microglia in an in vivo orthotopic murine model of glioblastoma multiforme. We demonstrated near real-time tracking of the migration of cells within these myeloid subpopulations. The intrinsic spatiotemporal resolution, imaging depth, and contrast sensitivity reported herein may facilitate detailed studies of the fundamental behaviors of TAMs and other leukocytes at the single-cell level in vivo, including intratumoral distribution heterogeneity and roles in modulating cancer proliferation.
Collapse
Affiliation(s)
- Elliott Daniel SoRelle
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Biophysics Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Derek William Yecies
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Orly Liba
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | | | - Claus Moritz Graef
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Rebecca Dutta
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Siddhartha Mitra
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Lydia-Marie Joubert
- Cell Sciences Imaging Facility, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Samuel Cheshier
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Adam de la Zerda
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Biophysics Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- The Chan Zuckerberg Biohub, 499 Illinois St., San Francisco, CA 94158, USA
- To whom correspondence should be addressed:
| |
Collapse
|
145
|
Grodzinski P, Kircher M, Goldberg M, Gabizon A. Integrating Nanotechnology into Cancer Care. ACS NANO 2019; 13:7370-7376. [PMID: 31240914 DOI: 10.1021/acsnano.9b04266] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Research activity in medical and cancer nanotechnology has grown dramatically over the past 15 years. The field has become a cradle of multidisciplinary investigations bringing together physicists, chemists, and engineers working with clinicians and biologists to address paramount problems in cancer care and treatment. Some have argued that the explosion in the number of research papers has not been followed by sufficient clinical activity in nanomedicine. However, three new nanodrugs have now been approved by the U.S. Food and Drug Administration (FDA) in the past three years, confirming the validity of nanotechnology approaches in cancer. Excitingly, translational pipelines contain several additional intriguing candidates. In this Nano Focus article, we discuss potential barriers inhibiting further incorporation of nanomedicines into patient care, possible strategies to overcome these barriers, and promising new directions in cancer interventions based on nanotechnology. Insights presented herein are outcomes of discussions held at a recent strategic workshop hosted by the National Cancer Institute (NCI), which brought together research, clinical, and commercial leaders of the nanomedicine field.
Collapse
Affiliation(s)
- Piotr Grodzinski
- National Cancer Institute , National Institutes of Health , Rockville , Maryland 20814 , United States
| | - Moritz Kircher
- Dana Farber Cancer Institute , Harvard Medical School , Boston , Massachusetts 02215 , United States
| | - Michael Goldberg
- Dana Farber Cancer Institute , Harvard Medical School , Boston , Massachusetts 02215 , United States
| | - Alberto Gabizon
- Shaare Zedek Medical Center and Hebrew University-School of Medicine , Jerusalem , Israel
| |
Collapse
|
146
|
Sulaiman A, McGarry S, El-Sahli S, Li L, Chambers J, Phan A, Côté M, Cron GO, Alain T, Le Y, Lee SH, Liu S, Figeys D, Gadde S, Wang L. Co-targeting Bulk Tumor and CSCs in Clinically Translatable TNBC Patient-Derived Xenografts via Combination Nanotherapy. Mol Cancer Ther 2019; 18:1755-1764. [DOI: 10.1158/1535-7163.mct-18-0873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/18/2019] [Accepted: 07/08/2019] [Indexed: 11/16/2022]
|
147
|
Cryer AM, Chan C, Eftychidou A, Maksoudian C, Mahesh M, Tetley TD, Spivey AC, Thorley AJ. Tyrosine Kinase Inhibitor Gold Nanoconjugates for the Treatment of Non-Small Cell Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2019; 11:16336-16346. [PMID: 30986026 DOI: 10.1021/acsami.9b02986] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Gold nanoparticles (AuNPs) have emerged as promising drug delivery candidates that can be leveraged for cancer therapy. Lung cancer (LC) is a heterogeneous disease that imposes a significant burden on society, with an unmet need for new therapies. Chemotherapeutic drugs such as afatinib (Afb), which is clinically approved for the treatment of epidermal growth factor receptor positive LC, is hydrophobic and has low bioavailability leading to spread around the body, causing severe side effects. Herein, we present a novel afatinib-AuNP formulation termed Afb-AuNPs, with the aim of improving drug efficacy and biocompatibility. This was achieved by synthesis of an alkyne-bearing Afb derivative and reaction with azide-functionalized lipoic acid using copper-catalyzed click chemistry, then conjugation to AuNPs via alkylthiol-gold bond formation. The Afb-AuNPs were found to possess up to 3.7-fold increased potency when administered to LC cells in vitro and were capable of significantly inhibiting cancer cell proliferation, as assessed by MTT assay and electric cell-substrate impedance sensing, respectively. Furthermore, when exposed to Afb-AuNPs, human alveolar epithelial type I-like cells, a model of the healthy lung epithelium, maintained viability and were found to release less proinflammatory cytokines when compared to free drug, demonstrating the biocompatibility of our formulation. This study provides a new platform for the development of nontraditional AuNP conjugates which can be applied to other molecules of therapeutic or diagnostic utility, with potential to be combined with photothermal therapy in other cancers.
Collapse
|
148
|
Pal S, Ray A, Andreou C, Zhou Y, Rakshit T, Wlodarczyk M, Maeda M, Toledo-Crow R, Berisha N, Yang J, Hsu HT, Oseledchyk A, Mondal J, Zou S, Kircher MF. DNA-enabled rational design of fluorescence-Raman bimodal nanoprobes for cancer imaging and therapy. Nat Commun 2019; 10:1926. [PMID: 31028250 PMCID: PMC6486596 DOI: 10.1038/s41467-019-09173-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
Recently, surface-enhanced Raman scattering nanoprobes have shown tremendous potential in oncological imaging owing to the high sensitivity and specificity of their fingerprint-like spectra. As current Raman scanners rely on a slow, point-by-point spectrum acquisition, there is an unmet need for faster imaging to cover a clinically relevant area in real-time. Herein, we report the rational design and optimization of fluorescence-Raman bimodal nanoparticles (FRNPs) that synergistically combine the specificity of Raman spectroscopy with the versatility and speed of fluorescence imaging. DNA-enabled molecular engineering allows the rational design of FRNPs with a detection limit as low as 5 × 10−15 M. FRNPs selectively accumulate in tumor tissue mouse cancer models and enable real-time fluorescence imaging for tumor detection, resection, and subsequent Raman-based verification of clean margins. Furthermore, FRNPs enable highly efficient image-guided photothermal ablation of tumors, widening the scope of the NPs into the therapeutic realm. Currently available Raman scanners are limited in speed to acquire images of clinically relevant sizes in cancer imaging. Here, the authors developed a DNA based design principle for Raman-Fluorescence bimodal nanoparticles and demonstrate real-time, high precision image-guided tumor resections and photothermal ablation of cancer.
Collapse
Affiliation(s)
- Suchetan Pal
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Chemistry, Indian Institute of Technology Bhilai, Raipur, Chhattisgarh, 492015, India
| | - Angana Ray
- Tata Institute of Fundamental Research, Hyderabad, Telangana, 500107, India
| | - Chrysafis Andreou
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yadong Zhou
- Department of Chemistry, University of Central Florida, Orlando, FL, 32816, USA
| | - Tatini Rakshit
- Department of Bioengineering, New York University, New York, NY, 10010, USA
| | - Marek Wlodarczyk
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA
| | - Masatomo Maeda
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ricardo Toledo-Crow
- Advanced Science Research Center, City University of New York, New York, NY, 10031, USA
| | - Naxhije Berisha
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA
| | - Jiang Yang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hsiao-Ting Hsu
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anton Oseledchyk
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jagannath Mondal
- Tata Institute of Fundamental Research, Hyderabad, Telangana, 500107, India
| | - Shengli Zou
- Department of Chemistry, University of Central Florida, Orlando, FL, 32816, USA
| | - Moritz F Kircher
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA. .,Department of Radiology, Weill Cornell Medical College, New York, NY, 10021, USA. .,Department of Imaging, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
149
|
Ekladious I, Colson YL, Grinstaff MW. Polymer-drug conjugate therapeutics: advances, insights and prospects. Nat Rev Drug Discov 2019; 18:273-294. [PMID: 30542076 PMCID: PMC12032968 DOI: 10.1038/s41573-018-0005-0] [Citation(s) in RCA: 537] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Polymer-drug conjugates have long been a mainstay of the drug delivery field, with several conjugates successfully translated into clinical practice. The conjugation of therapeutic agents to polymeric carriers, such as polyethylene glycol, offers several advantages, including improved drug solubilization, prolonged circulation, reduced immunogenicity, controlled release and enhanced safety. In this Review, we discuss the rational design, physicochemical characteristics and recent advances in the development of different classes of polymer-drug conjugates, including polymer-protein and polymer-small-molecule drug conjugates, dendrimers, polymer nanoparticles and multifunctional systems. Current obstacles hampering the clinical translation of polymer-drug conjugate therapeutics and future prospects are also presented.
Collapse
Affiliation(s)
- Iriny Ekladious
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA, USA
| | - Yolonda L Colson
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.
| | - Mark W Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA, USA.
| |
Collapse
|
150
|
Caster JM, Callaghan C, Seyedin SN, Henderson K, Sun B, Wang AZ. Optimizing Advances in Nanoparticle Delivery for Cancer Immunotherapy. Adv Drug Deliv Rev 2019; 144:3-15. [PMID: 31330165 PMCID: PMC11849717 DOI: 10.1016/j.addr.2019.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/20/2022]
Abstract
Cancer immunotherapy is one of the fastest growing and most promising fields in clinical oncology. T-cell checkpoint inhibitors are revolutionizing the management of advanced cancers including non-small cell lung cancer and melanoma. Unfortunately, many common cancers are not responsive to these drugs and resistance remains problematic. A growing number of novel cancer immunotherapies have been discovered but their clinical translation has been limited by shortcomings of conventional drug delivery. Immune signaling is tightly-regulated and often requires simultaneous or near-simultaneous activation of multiple signals in specific subpopulations of immune cells. Nucleic acid therapies, which require intact intracellular delivery, are among the most promising approaches to modulate the tumor microenvironment to a pro-immunogenic phenotype. Advanced nanomedicines can be precisely engineered to overcome many of these limitations and appear well-poised to enable the clinical translation of promising cancer immunotherapies.
Collapse
Affiliation(s)
- Joseph M Caster
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Cameron Callaghan
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Steven N Seyedin
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kelly Henderson
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Bo Sun
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, Lineberger Comprehensive Cancer Center, Department of Radiation Oncology, University of North Carolina at Chapel Hill, USA
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, Lineberger Comprehensive Cancer Center, Department of Radiation Oncology, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|