1
|
Cen J, Dai X, Zhao H, Li X, Hu X, Wu J, Duan S. Doxorubicin-Loaded Liposome with the Function of "Killing Two Birds with One Stone" against Glioma. ACS APPLIED MATERIALS & INTERFACES 2023; 15:46697-46709. [PMID: 37782688 DOI: 10.1021/acsami.3c10364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The blood-brain barrier (BBB) continues to be one of the main clinical obstacles in the treatment of glioma. Current chemotherapies always bring many different side effects, some even permanent. To date, nanomaterial-based vehicles have shown great potential in treating glioma. Herein, we developed a dual targeting liposomal delivery vector loaded with the anticancer drug doxorubicin (DOX) to treat glioma. SS31, a small peptide, has shown dual targeting effects of penetrating the BBB and specifically targeting mitochondria. In this study, a new liposomal delivery system, LS-DOX, was prepared by modifying DOX-loaded liposomes with SS31 for the treatment of in situ glioma. The liposomes demonstrated a high drug encapsulation rate and drug-loading capacity, satisfactory biocompatibility, high glioma accumulation ability, and good stability in vitro. Experimental results showed that the liposomes could effectively cross the BBB and target gliomas, and mitochondria-targeting of SS31 enhances cell uptake. In addition, the liposomes showed a good therapeutic effect on nude mice with glioma in situ with no obvious toxicity and side effects. Therefore, the present research will provide a novel alternative and reference for the effective treatment of glioma.
Collapse
Affiliation(s)
- Juan Cen
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| | - Xiaoying Dai
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| | - Han Zhao
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| | - Xiaohan Li
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| | - Xiaojiao Hu
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| | - Jing Wu
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| | - Shaofeng Duan
- School of Pharmacy, Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, PR China
| |
Collapse
|
2
|
Ghosh S, Chitgupi U, Sunar U, Lovell JF. Chemophototherapeutic Ablation of Doxorubicin-Resistant Human Ovarian Tumor Cells. Photochem Photobiol 2023; 99:844-849. [PMID: 35842741 PMCID: PMC9841062 DOI: 10.1111/php.13677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/14/2022] [Indexed: 01/18/2023]
Abstract
Porphyrin-phospholipid (PoP) liposomes loaded with Doxorubicin (Dox) have been demonstrated to be an efficient vehicle for chemophototherapy (CPT). Multidrug resistance (MDR) of cancer cells is a problematic phenomenon in which tumor cells develop resistance to chemotherapy. Herein, we report that Dox-resistant tumor cells can be ablated using our previously described formulation termed long-circulating Dox loaded in PoP liposomes (LC-Dox-PoP), which is a PEGylated formulation containing 2 mol. % of the PoP photosensitizer. In vitro studies using free Dox and LC-Dox-PoP showed that human ovarian carcinoma A2780 cells were more susceptible to Dox compared to the corresponding Dox-resistant A2780-R cells. When CPT was applied with LC-Dox-PoP liposomes, effective killing of both nonresistant and resistant A2780 cell lines was observed. An in vivo study to assess the efficiency of LC-Dox-PoP showed effective tumor shrinkage and prolonged survival of athymic nude mice bearing subcutaneous Dox-resistant A2780-R tumor xenografts when they were irradiated with a red laser. Biodistribution analysis demonstrated enhanced tumoral drug uptake in Dox-resistant tumors with CPT, suggesting that increased drug delivery was sufficient to induce ablation of resistant tumor cells.
Collapse
Affiliation(s)
- Sanjana Ghosh
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, 14260, USA
| | - Upendra Chitgupi
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, 14260, USA
- Nektar Therapeutics, Formulations Group, 455 Mission Bay Boulevard South, San Francisco, California, 94158, USA
| | - Ulas Sunar
- Department of Biomedical, Industrial & Human Factors Engineering, Wright State University, Dayton, OH 45435, USA
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, 14260, USA
| |
Collapse
|
3
|
Multimodal imaging distribution assessment of a liposomal antibiotic in an infectious disease model. J Control Release 2022; 352:199-210. [PMID: 36084816 DOI: 10.1016/j.jconrel.2022.08.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 11/08/2022]
Abstract
Liposomes are promising targeted drug delivery systems with the potential to improve the efficacy and safety profile of certain classes of drugs. Though attractive, there are unique analytical challenges associated with the development of liposomal drugs including human dose prediction given these are multi-component drug delivery systems. In this study, we developed a multimodal imaging approach to provide a comprehensive distribution assessment for an antibacterial drug, GSK2485680, delivered as a liposomal formulation (Lipo680) in a mouse thigh model of bacterial infection to support human dose prediction. Positron emission tomography (PET) imaging was used to track the in vivo biodistribution of Lipo680 over 48 h post-injection providing a clear assessment of the uptake in various tissues and, importantly, the selective accumulation at the site of infection. In addition, a pharmacokinetic model was created to evaluate the kinetics of Lipo680 in different tissues. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was then used to quantify the distribution of GSK2485680 and to qualitatively assess the distribution of a liposomal lipid throughout sections of infected and non-infected hindlimb tissues at high spatial resolution. Through the combination of both PET and MALDI IMS, we observed excellent correlation between the Lipo680-radionuclide signal detected by PET with the GSK2485680 and lipid component signals detected by MALDI IMS. This multimodal translational method can reduce drug attrition by generating comprehensive biodistribution profiles of drug delivery systems to provide mechanistic insight and elucidate safety concerns. Liposomal formulations have potential to deliver therapeutics across a broad array of different indications, and this work serves as a template to aid in delivering future liposomal drugs to the clinic.
Collapse
|
4
|
Saad MA, Hasan T. Spotlight on Photoactivatable Liposomes beyond Drug Delivery: An Enabler of Multitargeting of Molecular Pathways. Bioconjug Chem 2022; 33:2041-2064. [PMID: 36197738 DOI: 10.1021/acs.bioconjchem.2c00376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potential of photoactivating certain molecules, photosensitizers (PS), resulting in photochemical processes, has long been realized in the form of photodynamic therapy (PDT) for the management of several cancerous and noncancerous pathologies. With an improved understanding of the photoactivation process and its broader implications, efforts are being made to exploit the various facets of photoactivation, PDT, and the associated phenomenon of photodynamic priming in enhancing treatment outcomes, specifically in cancer therapeutics. The parallel emergence of nanomedicine, specifically liposome-based nanoformulations, and the convergence of the two fields of liposome-based drug delivery and PDT have led to the development of unique hybrid systems, which combine the exciting features of liposomes with adequate complementation through the photoactivation process. While initially liposomes carrying photosensitizers (PSs) were developed for enhancing the pharmacokinetics and the general applicability of PSs, more recently, PS-loaded liposomes, apart from their utility in PDT, have found several applications including enhanced targeting of drugs, coloading multiple therapeutic agents to enhance synergistic effects, imaging, priming, triggering drug release, and facilitating the escape of therapeutic agents from the endolysosomal complex. This review discusses the design strategies, potential, and unique attributes of these hybrid systems, with not only photoactivation as an attribute but also the ability to encapsulate multiple agents for imaging, biomodulation, priming, and therapy referred to as photoactivatable multiagent/inhibitor liposomes (PMILS) and their targeted versions─targeted PMILS (TPMILS). While liposomes have formed their own niche in nanotechnology and nanomedicine with several clinically approved formulations, we try to highlight how using PS-loaded liposomes could address some of the limitations and concerns usually associated with liposomes to overcome them and enhance their preclinical and clinical utility in the future.
Collapse
Affiliation(s)
- Mohammad A Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States.,Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
5
|
Gawne PJ, Pinto SMA, Nielsen KM, Keeling GP, Pereira MM, T M de Rosales R. Microwave-assisted synthesis of [ 52Mn]Mn-porphyrins: Applications in cell and liposome radiolabelling. Nucl Med Biol 2022; 114-115:6-17. [PMID: 36088876 PMCID: PMC10236072 DOI: 10.1016/j.nucmedbio.2022.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Manganese porphyrins have several therapeutic/imaging applications, including their use as radioprotectants (in clinical trials) and as paramagnetic MRI contrast agents. The affinity of porphyrins for lipid bilayers also makes them candidates for cell/liposome labelling. We hypothesised that metalation with the positron emission tomography (PET) radionuclide 52Mn (t1/2 = 5.6 d) would allow long-term in vivo biodistribution studies of Mn-porphyrins, as well as a method to label and track cells/liposomes, but methods for fast and efficient radiolabelling are lacking. RESULTS Several porphyrins were produced and radiolabelled by addition to neutralised [52Mn]MnCl2 and heating using a microwave (MW) synthesiser, and compared with non-MW heating. MW radiosynthesis allowed >95 % radiochemical yields (RCY) in just 1 h. Conversely, non-MW heating at 70 °C for 1 h resulted in low RCY (0-25 % RCY) and most porphyrins did not reach radiolabelling completion after 24 h. Formation of the 52Mn-complexes were confirmed with radio-HPLC by comparison with their non-radioactive 55Mn counterparts. Following this, several [52Mn]Mn-porphyrins were used to radiolabel liposomes resulting in 75-86 % labelling efficiency (LE). Two lead [52Mn]Mn-porphyrins were taken forward to label MDA-MB-231 cancer cells in vitro, achieving ca. 11 % LE. After 24 h, 32-45 % of the [52Mn]Mn-porphyrins was retained in cells. CONCLUSIONS In contrast to standard methods, MW heating allows the fast synthesis of [52Mn]Mn-porphyrins with >95 % radiochemical yields that avoid purification. [52Mn]Mn-porphyrins also show promising cell/liposome labelling properties. Our reported technique can potentially be exploited for the in vivo imaging of Mn-porphyrin therapeutics, as well as for the accurate in vivo quantification of Mn-porphyrin MRI agents.
Collapse
Affiliation(s)
- Peter J Gawne
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Sara M A Pinto
- Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Karin M Nielsen
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - George P Keeling
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Mariette M Pereira
- Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, UK.
| |
Collapse
|
6
|
Younis MH, Tang Z, Cai W. Multimodality imaging of nanoparticle-based vaccines: Shedding light on immunology. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1807. [PMID: 35501142 PMCID: PMC9481661 DOI: 10.1002/wnan.1807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023]
Abstract
In recent years, there have been significant innovations in the development of nanoparticle-based vaccines and vaccine delivery systems. For the purposes of both design and evaluation, these nanovaccines are imaged using the wealth of understanding established around medical imaging of nanomaterials. An important insight to the advancement of the field of nanovaccines can be given by an analysis of the design rationale of an imaging platform, as well as the significance of the information provided by imaging. Nanovaccine imaging strategies can be categorized by the imaging modality leveraged, but it is also worth understanding the superiority or convenience of a given modality over others in a given context of a particular nanovaccine. The most important imaging modalities in this endeavor are optical imaging including near-infrared fluorescence imaging (NIRF), emission tomography methods such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) with or without computed tomography (CT) or magnetic resonance (MR), the emerging magnetic particle imaging (MPI), and finally, multimodal applications of imaging which include molecular imaging with magnetic resonance imaging (MRI) and photoacoustic (PA) imaging. One finds that each of these modalities has strengths and weaknesses, but optical and PET imaging tend, in this context, to be currently the most accessible, convenient, and informative modalities. Nevertheless, an important principle is that there is not a one-size-fits-all solution, and that the specific nanovaccine in question must be compatible with a particular imaging modality. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Muhsin H. Younis
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, USA
| | - Zhongmin Tang
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, USA
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, USA
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
7
|
Ghosh S, He X, Huang WC, Lovell JF. Immune checkpoint blockade enhances chemophototherapy in a syngeneic pancreatic tumor model. APL Bioeng 2022; 6:036105. [PMID: 36164594 PMCID: PMC9509203 DOI: 10.1063/5.0099811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/29/2022] [Indexed: 11/14/2022] Open
Abstract
Pancreatic cancer (PaCa) suffers from poor treatment options for locally advanced cases. Chemophototherapy (CPT) is an emerging anti-tumor modality, and porphyrin–phospholipid liposomes have been shown to be versatile drug carriers for CPT in preclinical rodent models. Here we show that in the syngeneic subcutaneous KPC PaCa tumor model, exhausted CD8+ T cells are localized in the tumor, and that CPT is enhanced in combination with immune checkpoint blockade (ICB). Addition of ICB using anti-programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies resulted in ablation of medium-sized, established KPC tumors (∼200 mm3) without recurrence for over 100 days. Mice rejected subsequent tumor re-challenge. Flow cytometry and tumor slice analysis following injection of a fluorescently labeled anti-PD-1 antibody showed that CPT improved antibody delivery to the tumor microenvironment. Treatment of large established tumors (∼400 mm3) using with CPT and ICB induced appreciable tumor regression and delay in regrowth. Taken together, these data demonstrate the utility of combining CPT with immunotherapies.
Collapse
Affiliation(s)
- Sanjana Ghosh
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Xuedan He
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| |
Collapse
|
8
|
Radionuclide Delivery Strategies in Tumor Treatment: A Systematic Review. Curr Issues Mol Biol 2022; 44:3267-3282. [PMID: 35892711 PMCID: PMC9332578 DOI: 10.3390/cimb44080225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
The aim of this review was to assess recent progress in targeted radionuclide tumor therapy, focusing on the best delivery strategies. A literature search was conducted in PubMed, Web of Science, and Scopus using the terms "radionuclides", "liposomes", "avidin-biotin interaction", "theranostic", and "molecular docking". The 10 year filter was applied, except for the avidin-biotin interaction. Data were retrieved from both preclinical and clinical settings. Three targeting strategies were considered: pretargeting, liposomes, and ligands. Pretargeting can be achieved by exploiting the avidin-biotin interaction. This strategy seems very promising, although it has been investigated mainly in resectable tumors. Radiolabeled liposomes have attracted new interest as probes to identify the most suitable patients for treatment with liposomal formulations of common chemotherapeutics. The use of ligands for the delivery of radiotherapeutics to a specific target is still the most appealing strategy for treating tumors. The most appropriate ligand can be identified by virtually simulating its interaction with the receptor. All strategies showed great potential for use in targeted radionuclide therapy, but they also have numerous drawbacks. The most promising option is probably the one based on the use of new ligands.
Collapse
|
9
|
Vlasova KY, Ostroverkhov P, Vedenyapina D, Yakimova T, Trusova A, Lomakina GY, Vodopyanov SS, Grin M, Klyachko N, Chekhonin V, Abakumov M. Liposomal Form of 2,4-Dinitrophenol Lipophilic Derivatives as a Promising Therapeutic Agent for ATP Synthesis Inhibition. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2162. [PMID: 35808003 PMCID: PMC9268429 DOI: 10.3390/nano12132162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/07/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022]
Abstract
Mitochondrial uncoupler 2,4-dinitrophenol (2,4-DNP) is a promising antidiabetic and antiobesity agent. Its clinical use is limited by a narrow dynamic range and accumulation in non-target sensitive organs, which results in whole-body toxicity. A liposomal formulation could enable the mentioned drawbacks to be overcome and simplify the liver-targeted delivery and sustained release of 2,4-DNP. We synthesized 2,4-DNP esters with carboxylic acids of various lipophilic degrees using carboxylic acid chloride and then loaded them into liposomes. We demonstrated the effective increase in the entrapment of 2,4-DNP into liposomes when esters were used. Here, we examined the dependence of the sustained release of 2,4-DNP from liposomes on the lipid composition and LogPoct of the ester. We posit that the optimal chain length of the ester should be close to the palmitic acid and the lipid membrane should be composed of phospholipids with a certain phase transition point depending on the desired release rate. The increased effect of the ATP synthesis inhibition of the liposomal forms of caproic and palmitic acid esters compared to free molecules in liver hepatocytes was demonstrated. The liposomes' stability could well be responsible for this result. This work demonstrates promising possibilities for the liver-targeted delivery of the 2,4-DNP esters with carboxylic acids loaded into liposomes for ATP synthesis inhibition.
Collapse
Affiliation(s)
- Kseniya Yu. Vlasova
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (K.Y.V.); (V.C.)
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (G.Y.L.); (N.K.)
| | - Petr Ostroverkhov
- Department of Chemistry and Technology of Biologically Active Compounds, Medical and Organic Chemistry, Lomonosov Institute of Fine Chemical Technologies MIREA-Russian Technological University (RTU MIREA), 119571 Moscow, Russia; (P.O.); (D.V.); (M.G.)
| | - Daria Vedenyapina
- Department of Chemistry and Technology of Biologically Active Compounds, Medical and Organic Chemistry, Lomonosov Institute of Fine Chemical Technologies MIREA-Russian Technological University (RTU MIREA), 119571 Moscow, Russia; (P.O.); (D.V.); (M.G.)
| | - Tamara Yakimova
- Faculty of Materials Science, Lomonosov Moscow State University, 119991 Moscow, Russia; (T.Y.); (A.T.)
| | - Alla Trusova
- Faculty of Materials Science, Lomonosov Moscow State University, 119991 Moscow, Russia; (T.Y.); (A.T.)
| | | | - Stepan Sergeevich Vodopyanov
- College of New Materials and Nanotechnologies, National University of Science and Technology (MISIS), 119049 Moscow, Russia;
| | - Mikhail Grin
- Department of Chemistry and Technology of Biologically Active Compounds, Medical and Organic Chemistry, Lomonosov Institute of Fine Chemical Technologies MIREA-Russian Technological University (RTU MIREA), 119571 Moscow, Russia; (P.O.); (D.V.); (M.G.)
| | - Natalia Klyachko
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (G.Y.L.); (N.K.)
| | - Vladimir Chekhonin
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (K.Y.V.); (V.C.)
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia
| | - Maxim Abakumov
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (K.Y.V.); (V.C.)
- College of New Materials and Nanotechnologies, National University of Science and Technology (MISIS), 119049 Moscow, Russia;
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia
| |
Collapse
|
10
|
Liu HJ, Wang M, Shi S, Hu X, Xu P. A Therapeutic Sheep in Metastatic Wolf's Clothing: Trojan Horse Approach for Cancer Brain Metastases Treatment. NANO-MICRO LETTERS 2022; 14:114. [PMID: 35482117 PMCID: PMC9050993 DOI: 10.1007/s40820-022-00861-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/06/2022] [Indexed: 05/13/2023]
Abstract
Early-stage brain metastasis of breast cancer (BMBC), due to the existence of an intact blood-brain barrier (BBB), is one of the deadliest neurologic complications. To improve the efficacy of chemotherapy for BMBC, a Trojan horse strategy-based nanocarrier has been developed by integrating the cell membrane of a brain-homing cancer cell and a polymeric drug depot. With the camouflage of a MDA-MB-231/Br cell membrane, doxorubicin-loaded poly (D, L-lactic-co-glycolic acid) nanoparticle (DOX-PLGA@CM) shows enhanced cellular uptake and boosted killing potency for MDA-MB-231/Br cells. Furthermore, DOX-PLGA@CM is equipped with naturally selected molecules for BBB penetration, as evidenced by its boosted capacity in entering the brain of both healthy and early-stage BMBC mouse models. Consequently, DOX-PLGA@CM effectively reaches the metastatic tumor lesions in the brain, slows down cancer progression, reduces tumor burden, and extends the survival time for the BMBC animal. Furthermore, the simplicity and easy scale-up of the design opens a new window for the treatment of BMBC and other brain metastatic cancers.
Collapse
Affiliation(s)
- Hai-Jun Liu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Mingming Wang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Shanshan Shi
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Xiangxiang Hu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
11
|
Dual-Labelling Strategies for Nuclear and Fluorescence Molecular Imaging: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15040432. [PMID: 35455430 PMCID: PMC9028399 DOI: 10.3390/ph15040432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Molecular imaging offers the possibility to investigate biological and biochemical processes non-invasively and to obtain information on both anatomy and dysfunctions. Based on the data obtained, a fundamental understanding of various disease processes can be derived and treatment strategies can be planned. In this context, methods that combine several modalities in one probe are increasingly being used. Due to the comparably high sensitivity and provided complementary information, the combination of nuclear and optical probes has taken on a special significance. In this review article, dual-labelled systems for bimodal nuclear and optical imaging based on both modular ligands and nanomaterials are discussed. Particular attention is paid to radiometal-labelled molecules for single-photon emission computed tomography (SPECT) and positron emission tomography (PET) and metal complexes combined with fluorescent dyes for optical imaging. The clinical potential of such probes, especially for fluorescence-guided surgery, is assessed.
Collapse
|
12
|
Zheng F, Huang X, Ding J, Bi A, Wang S, Chen F, Zeng W. NIR-I Dye-Based Probe: A New Window for Bimodal Tumor Theranostics. Front Chem 2022; 10:859948. [PMID: 35402374 PMCID: PMC8984032 DOI: 10.3389/fchem.2022.859948] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Near-infrared (NIR, 650-1700 nm) bioimaging has emerged as a powerful strategy in tumor diagnosis. In particular, NIR-I fluorescence imaging (650-950 nm) has drawn more attention, benefiting from the high quantum yield and good biocompatibility. Since their biomedical applications are slightly limited by their relatively low penetration depth, NIR-I fluorescence imaging probes have been under extensive development in recent years. This review summarizes the particular application of the NIR-I fluorescent dye-contained bimodal probes, with emphasis on related nanoprobes. These probes have enabled us to overcome the drawbacks of individual imaging modalities as well as achieve synergistic imaging. Meanwhile, the application of these NIR-I fluorescence-based bimodal probes for cancer theranostics is highlighted.
Collapse
Affiliation(s)
- Fan Zheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Xueyan Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Jipeng Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Anyao Bi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Shifen Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Fei Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| |
Collapse
|
13
|
Chen X, Niu W, Du Z, Zhang Y, Su D, Gao X. 64Cu radiolabeled nanomaterials for positron emission tomography (PET) imaging. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.02.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
14
|
Alam Khan S, Jawaid Akhtar M. Structural modification and strategies for the enhanced doxorubicin drug delivery. Bioorg Chem 2022; 120:105599. [DOI: 10.1016/j.bioorg.2022.105599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 12/29/2022]
|
15
|
Lai X, Liu XL, Pan H, Zhu MH, Long M, Yuan Y, Zhang Z, Dong X, Lu Q, Sun P, Lovell JF, Chen HZ, Fang C. Light-Triggered Efficient Sequential Drug Delivery of Biomimetic Nanosystem for Multimodal Chemo-, Antiangiogenic, and Anti-MDSC Therapy in Melanoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106682. [PMID: 34989039 DOI: 10.1002/adma.202106682] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/14/2021] [Indexed: 06/14/2023]
Abstract
In view of the multiple pathological hallmarks of tumors, nanosystems for the sequential delivery of various drugs whose targets are separately located inside and outside tumor cells are desired for improved cancer therapy. However, current sequential delivery is mainly achieved through enzyme- or acid-dependent degradation of the nanocarrier, which would be influenced by the heterogeneous tumor microenvironment, and unloading efficiency of the drug acting on the target outside tumor cells is usually unsatisfactory. Here, a light-triggered sequential delivery strategy based on a liposomal formulation of doxorubicin (DOX)-loaded small-sized polymeric nanoparticles (DOX-NP) and free sunitinib in the aqueous cavity, is developed. The liposomal membrane is doped with photosensitizer porphyrin-phospholipid (PoP) and hybridized with red blood cell membrane to confer biomimetic features. Near-infrared light-induced membrane permeabilization triggers the "ultrafast" and "thorough" release of sunitinib (100% release in 5 min) for antiangiogenic therapy and also myeloid-derived suppressor cell (MDSC) inhibition to reverse the immunosuppressive tumor environment. Subsequently, the small-sized DOX-NP liberated from the liposomes is more easily uptaken by tumor cells for improved immunogenic chemotherapy. RNA sequencing and immune-related assay indicates therapeutic immune enhancement. This light-triggered sequential delivery strategy demonstrates the potency in cancer multimodal therapy against multiple targets in different spatial positions in tumor microenvironment.
Collapse
Affiliation(s)
- Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Xue-Liang Liu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Hong Pan
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Mei Long
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yihang Yuan
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Zhong Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Xiao Dong
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| |
Collapse
|
16
|
Qian R, Jing B, Jiang D, Gai Y, Zhu Z, Huang X, Gao Y, Lan X, An R. Multi-antitumor therapy and synchronous imaging monitoring based on exosome. Eur J Nucl Med Mol Imaging 2022; 49:2668-2681. [DOI: 10.1007/s00259-022-05696-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/19/2022] [Indexed: 02/06/2023]
|
17
|
Ariztia J, Solmont K, Moïse NP, Specklin S, Heck MP, Lamandé-Langle S, Kuhnast B. PET/Fluorescence Imaging: An Overview of the Chemical Strategies to Build Dual Imaging Tools. Bioconjug Chem 2022; 33:24-52. [PMID: 34994545 DOI: 10.1021/acs.bioconjchem.1c00503] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular imaging is a biomedical research discipline that has quickly emerged to afford the observation, characterization, monitoring, and quantification of biomarkers and biological processes in living organism. It covers a large array of imaging techniques, each of which provides anatomical, functional, or metabolic information. Multimodality, as the combination of two or more of these techniques, has proven to be one of the best options to boost their individual properties, hence offering unprecedented tools for human health. In this review, we will focus on the combination of positron emission tomography and fluorescence imaging from the specific perspective of the chemical synthesis of dual imaging agents. Based on a detailed analysis of the literature, this review aims at giving a comprehensive overview of the chemical strategies implemented to build adequate imaging tools considering radiohalogens and radiometals as positron emitters, fluorescent dyes mostly emitting in the NIR window and all types of targeting vectors.
Collapse
Affiliation(s)
- Julen Ariztia
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Kathleen Solmont
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | | | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Marie Pierre Heck
- Université Paris-Saclay, INRAE, Département Médicaments et Technologies pour la santé (DMTS), SCBM, 91191, Gif-sur-Yvette cedex, France
| | | | - Bertrand Kuhnast
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| |
Collapse
|
18
|
Ghosh S, Lovell JF. Two Laser Treatments Can Improve Tumor Ablation Efficiency of Chemophototherapy. Pharmaceutics 2021; 13:pharmaceutics13122183. [PMID: 34959464 PMCID: PMC8704214 DOI: 10.3390/pharmaceutics13122183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Chemophototherapy is an emerging tumor ablation modality that can improve local delivery of chemotherapeutic agents. Long circulating doxorubicin (Dox) in porphyrin-phospholipid (PoP) liposomes (LC-Dox-PoP) has previously been developed as an effective chemophototherapy agent. In the present study, we observed that in mice, LC-Dox-PoP showed enhanced accumulation in human pancreatic tumor xenografts even with suboptimal light doses, as assessed by fluorometric analysis of tissue homogenates and microscopic imaging of Dox and PoP in tumor slices. A second laser treatment, at a time point in which tumors had greater drug accumulation as a result of the first laser treatment, induced potent tumor ablation. Efficacy studies were carried out in two human pancreatic cancer subcutaneous mouse tumor models; MIA PaCa-2 or low-passage patient derived pancreatic cancer xenografts. A single treatment of 3 mg/kg LC-Dox-PoP and an initial 150 J/cm2 laser treatment 1 h after drug administration, followed by second laser treatment of 50 J/cm2 8 h after drug administration, was more effective than a single laser treatment of 200 J/cm2 at either of those time points. Thus, this study presents proof-of-principle and rationale for using two discrete laser treatments to enhance the efficacy of chemophototherapy.
Collapse
|
19
|
Liu C, Liu YY, Chang Q, Shu Q, Shen N, Wang H, Xie Y, Deng X. Pressure-Controlled Encapsulation of Graphene Quantum Dots into Liposomes by the Reverse-Phase Evaporation Method. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:14096-14104. [PMID: 34808057 DOI: 10.1021/acs.langmuir.1c02338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ultrasmall nanoparticles (USNPs) with sizes below 10 nm have shown great potentials in medical applications owing to their outstanding physical, chemical, optical, and biological properties. However, they suffer from a rapid renal clearance and biodegradation rate in the biological environment due to the small size. Liposomes are one of the most promising delivery nanocarriers for loading USNPs because of their excellent biocompatibility and lipid bilayer structure. Encapsulation of USNPs into liposomes in an efficient and controllable manner remains a challenge. In this study, we achieved a high loading of graphene quantum dots (GQDs, ∼4 nm), a typical USNP, into the aqueous core of liposomes (45.68 ± 1.44%), which was controllable by the pressure. The GQDs-loaded liposomes (GQDs-LPs) exhibited a very good aqueous stability for over a month. Furthermore, indocyanine green (ICG), an efficient near-infrared (NIR) photothermal agent, was introduced in the GQDs-LP system that could convert NIR laser energy into thermal energy and break down the liposomes, causing the release of GQDs in 6 min. Moreover, this NIR light-controlled release system (GQDs-ICG-LPs) also exhibited a good photothermal therapeutic performance in vitro, and 75% of cancer cells were killed at a concentration of 200 μg/mL. Overall, the successful development of the NIR light-controlled release system has laid a solid foundation for the future biomedical application of USNPs-loaded liposomes.
Collapse
Affiliation(s)
- Chenghao Liu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Qing Chang
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Qingfeng Shu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Ning Shen
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yijun Xie
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xiaoyong Deng
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
20
|
Doroudian M, Azhdari MH, Goodarzi N, O’Sullivan D, Donnelly SC. Smart Nanotherapeutics and Lung Cancer. Pharmaceutics 2021; 13:1972. [PMID: 34834387 PMCID: PMC8619749 DOI: 10.3390/pharmaceutics13111972] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is a significant health problem worldwide. Unfortunately, current therapeutic strategies lack a sufficient level of specificity and can harm adjacent healthy cells. Consequently, to address the clinical need, novel approaches to improve treatment efficiency with minimal side effects are required. Nanotechnology can substantially contribute to the generation of differentiated products and improve patient outcomes. Evidence from previous research suggests that nanotechnology-based drug delivery systems could provide a promising platform for the targeted delivery of traditional chemotherapeutic drugs and novel small molecule therapeutic agents to treat lung cancer cells more effectively. This has also been found to improve the therapeutic index and reduce the required drug dose. Nanodrug delivery systems also provide precise control over drug release, resulting in reduced toxic side effects, controlled biodistribution, and accelerated effects or responses. This review highlights the most advanced and novel nanotechnology-based strategies, including targeted nanodrug delivery systems, stimuli-responsive nanoparticles, and bio-nanocarriers, which have recently been employed in preclinical and clinical investigations to overcome the current challenges in lung cancer treatments.
Collapse
Affiliation(s)
- Mohammad Doroudian
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland; (M.D.); (D.O.)
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran; (M.H.A.); (N.G.)
| | - Mohammad H. Azhdari
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran; (M.H.A.); (N.G.)
| | - Nima Goodarzi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran; (M.H.A.); (N.G.)
| | - David O’Sullivan
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland; (M.D.); (D.O.)
| | - Seamas C. Donnelly
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland; (M.D.); (D.O.)
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Tallaght University Hospital, Tallaght, Dublin 24, Ireland
| |
Collapse
|
21
|
Yang YL, Lin K, Yang L. Progress in Nanocarriers Codelivery System to Enhance the Anticancer Effect of Photodynamic Therapy. Pharmaceutics 2021; 13:1951. [PMID: 34834367 PMCID: PMC8617654 DOI: 10.3390/pharmaceutics13111951] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising anticancer noninvasive method and has great potential for clinical applications. Unfortunately, PDT still has many limitations, such as metastatic tumor at unknown sites, inadequate light delivery and a lack of sufficient oxygen. Recent studies have demonstrated that photodynamic therapy in combination with other therapies can enhance anticancer effects. The development of new nanomaterials provides a platform for the codelivery of two or more therapeutic drugs, which is a promising cancer treatment method. The use of multifunctional nanocarriers for the codelivery of two or more drugs can improve physical and chemical properties, increase tumor site aggregation, and enhance the antitumor effect through synergistic actions, which is worthy of further study. This review focuses on the latest research progress on the synergistic enhancement of PDT by simultaneous multidrug administration using codelivery nanocarriers. We introduce the design of codelivery nanocarriers and discuss the mechanism of PDT combined with other antitumor methods. The combination of PDT and chemotherapy, gene therapy, immunotherapy, photothermal therapy, hyperthermia, radiotherapy, sonodynamic therapy and even multidrug therapy are discussed to provide a comprehensive understanding.
Collapse
Affiliation(s)
| | | | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.-L.Y.); (K.L.)
| |
Collapse
|
22
|
Huang S, Wu Y, Li C, Xu L, Huang J, Huang Y, Cheng W, Xue B, Zhang L, Liang S, Jin X, Zhu X, Xiong S, Su Y, Wang H. Tailoring morphologies of mesoporous polydopamine nanoparticles to deliver high-loading radioiodine for anaplastic thyroid carcinoma imaging and therapy. NANOSCALE 2021; 13:15021-15030. [PMID: 34533142 DOI: 10.1039/d1nr02892h] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Anaplastic thyroid carcinoma (ATC), as one of the most aggressive human malignancies, cannot be cured by 131iodine (131I) internal radiotherapy (RT) because the tumor cells cannot effectively take up 131I and are resistant to radiotherapy. In this study, a facile and simple method was proposed to synthesize mesoporous polydopamine nanoparticles (MPDA) and tailor their morphologies by component-adjusting Pluronic micelle-guided polymerization. Then, MPDA were used not only as nanocarriers to radiolabel 131I, but also as photothermal conversion agents for photothermal therapy (PTT) to promote RT. The iodine-labeling capacity and photothermal conversion efficiency of MPDA can be enhanced by optimizing their morphologies. It was found that MPDA NPs with a cerebroid pore channel structure (CPDA) showed the highest iodine-carrying capacity and a higher photothermal conversion efficiency as a result of their maximum specific surface area and unique morphology. In subsequent experiments in vitro and in vivo, our ATC animal models showed impressive therapeutic responses to CPDA-131I NPs because of the synergistic effect of PTT and RT. Additionally, CPDA-125I NPs can be utilized to obtain high-quality SPETC/CT images of tumors, which can guide clinical therapy for ATC. Considering their great biosafety, these radioiodine-labeled CPDA NPs may serve as a promising tool in combined therapy and diagnosis in ATC.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Nuclear Medicine, Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Yan Wu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Chao Li
- Department of Nuclear Medicine, Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Li Xu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Jie Huang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Yu Huang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Weiwei Cheng
- Department of Nuclear Medicine, Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Bai Xue
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Linlin Zhang
- Department of Nuclear Medicine, Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Sheng Liang
- Department of Nuclear Medicine, Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Xin Jin
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Shuqiang Xiong
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Yue Su
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | - Hui Wang
- Department of Nuclear Medicine, Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School, 1665 Kongjiang Road, Shanghai 200092, China.
| |
Collapse
|
23
|
Kilian HI, Pradhan AJ, Jahagirdar D, Ortega J, Atilla-Gokcumen GE, Lovell JF. Light-Triggered Release of Large Biomacromolecules from Porphyrin-Phospholipid Liposomes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:10859-10865. [PMID: 34450021 DOI: 10.1021/acs.langmuir.1c01848] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Liposomes containing small amounts of porphyrin-phospholipid (PoP) have been shown to encapsulate small molecular weight cargos and then release them upon exposure to red light. A putative mechanism involves transient pore formation in the bilayer induced by PoP-mediated photo-oxidation of unsaturated lipids. However, little is known about the properties of such pores. This study assesses whether large carbohydrate and protein molecules could be released from PoP liposomes upon red light exposure. A small fluorophore with ∼0.5 kDa in molecular weight, fluorescently labeled dextrans of ∼5 and ∼500 kDa, and a ∼240 kDa fluorescent protein were passively entrapped in PoP liposomes. When exposed to 665 nm irradiation, liposomes containing PoP, but not liposomes lacking it, released all these cargos in a size-dependent manner that occurred with oxidation of unsaturated lipids included in the bilayer. Thus, this study demonstrates the feasibility of light-triggered release of large biomacromolecules from liposomes.
Collapse
Affiliation(s)
- Hailey I Kilian
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - Apoorva J Pradhan
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - Dushyant Jahagirdar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Gunes Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
24
|
Feng HY, Yuan Y, Zhang Y, Liu HJ, Dong X, Yang SC, Liu XL, Lai X, Zhu MH, Wang J, Lu Q, Lin Q, Chen HZ, Lovell JF, Sun P, Fang C. Targeted Micellar Phthalocyanine for Lymph Node Metastasis Homing and Photothermal Therapy in an Orthotopic Colorectal Tumor Model. NANO-MICRO LETTERS 2021; 13:145. [PMID: 34146159 PMCID: PMC8214644 DOI: 10.1007/s40820-021-00666-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/11/2021] [Indexed: 05/04/2023]
Abstract
UNLABELLED Small-sized trastuzumab-targeted micelles (T-MP) were engineered using a surfactant-stripping approach that yielded concentrated phthalocyanines with strong near infrared absorption. T-MP accumulated more in the lymph node (LN) metastases of orthotopic colorectal cancer compared to the micelles conjugated with control IgG. Following surgical resection of the primary tumor, minimally invasive photothermal treatment of the metastatic LN with T-MP, but not the control micelles, extended mouse survival. ABSTRACT Tumor lymph node (LN) metastasis seriously affects the treatment prognosis. Studies have shown that nanoparticles with size of sub-50 nm can directly penetrate into LN metastases after intravenous administration. Here, we speculate through introducing targeting capacity, the nanoparticle accumulation in LN metastases would be further enhanced for improved local treatment such as photothermal therapy. Trastuzumab-targeted micelles (< 50 nm) were formulated using a unique surfactant-stripping approach that yielded concentrated phthalocyanines with strong near-infrared absorption. Targeted micellar phthalocyanine (T-MP) was an effective photothermal transducer and ablated HT-29 cells in vitro. A HER2-expressing colorectal cancer cell line (HT-29) was used to establish an orthotopic mouse model that developed metastatic disease in mesenteric sentinel LN. T-MP accumulated more in the LN metastases compared to the micelles conjugated with control IgG. Following surgical resection of the primary tumor, minimally invasive photothermal treatment of the metastatic LN with T-MP, but not the control micelles, extended mouse survival. Our findings demonstrate for the first time that targeted small-sized nanoparticles have potential to enable superior paradigms for dealing with LN metastases. [Image: see text] SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s40820-021-00666-8.
Collapse
Affiliation(s)
- Hai-Yi Feng
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, People's Republic of China
| | - Yihang Yuan
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Yunpeng Zhang
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, People's Republic of China
| | - Hai-Jun Liu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Xiao Dong
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Si-Cong Yang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Xue-Liang Liu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Jue Wang
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, People's Republic of China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China
| | - Quanjun Lin
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, People's Republic of China
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, People's Republic of China.
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, People's Republic of China.
| |
Collapse
|
25
|
Xu Z, Huang H, Xiong X, Wei X, Guo X, Zhao J, Zhou S. A near-infrared light-responsive extracellular vesicle as a "Trojan horse" for tumor deep penetration and imaging-guided therapy. Biomaterials 2021; 269:120647. [PMID: 33450584 DOI: 10.1016/j.biomaterials.2020.120647] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/17/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022]
Abstract
How to make the nanoparticles evade immune surveillance and deeply penetrate the tumor tissues is of great importance to maximize the therapeutic efficacy of nanomedicines. Here, a near-infrared (NIR) light-responsive extracellular vesicle as a nanoplatform is developed to realize long circulation in blood, deep penetration in tumor tissues and rapid body elimination after the treatment. Like a "Trojan horse", the nanoplatform is obtained by hiding the anti-tumor soldiers (DOX and 4.2 nm Ag2S quantum dots (QDs)) into the macrophage cell-secreted vesicle through electroporation. The natural composition and tumor targeting activity of the extracellular vesicles enable the nanoplatform to achieve a high accumulation in tumor and the in vivo biodistribution can be monitored by NIR fluorescence imaging of the Ag2S QDs. After the nanomedicines accumulate at the tumor sites, the soldiers will be released from the "Trojan horse" by utilizing the NIR photothermal effect of the Ag2S QDs. The released ultrasmall QDs and DOX can penetrate the whole tumor with a diameter of about 9 mm and effectively kill the tumor cells. Moreover, the inorganic QDs can be rapidly excreted from the body through renal clearance after the treatment to avoid the potential toxicity.
Collapse
Affiliation(s)
- Zeng Xu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Huabei Huang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Xiang Xiong
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Xiaoqing Wei
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Xing Guo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Jingya Zhao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China.
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China.
| |
Collapse
|
26
|
Faustova M, Nikolskaya E, Sokol M, Fomicheva M, Petrov R, Yabbarov N. Metalloporphyrins in Medicine: From History to Recent Trends. ACS APPLIED BIO MATERIALS 2020; 3:8146-8171. [PMID: 35019597 DOI: 10.1021/acsabm.0c00941] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The history of metalloporphyrins dates back more than 200 years ago. Metalloporphyrins are excellent catalysts, capable of forming supramolecular systems, participate in oxygen photosynthesis, transport, and used as contrast agents or superoxide dismutase mimetics. Today, metalloporphyrins represent complexes of conjugated π-electron system and metals from the entire periodic system. However, the effect of these compounds on living systems has not been fully understood, and researchers are exploring the properties of metalloporphyrins thereby extending their further application. This review provides an overview of the variety of metalloporphyrins that are currently used in different medicine fields and how metalloporphyrins became the subject of scientists' interest. Currently, metalloporphyrins utilization has expanded significantly, which gave us an opprotunuty to summarize recent progress in metalloporphyrins derivatives and prospects of their application in the treatment and diagnosis of different diseases.
Collapse
Affiliation(s)
- Mariia Faustova
- MIREA-Russian Technological University, Lomonosov Institute of Fine Chemical Technologies, 119454 Moscow, Russia.,N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena Nikolskaya
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia
| | - Maria Sokol
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia.,JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149 Moscow Russia
| | - Margarita Fomicheva
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia.,JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149 Moscow Russia
| | - Rem Petrov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Nikita Yabbarov
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia.,JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149 Moscow Russia
| |
Collapse
|
27
|
Javed I, Cui X, Wang X, Mortimer M, Andrikopoulos N, Li Y, Davis TP, Zhao Y, Ke PC, Chen C. Implications of the Human Gut-Brain and Gut-Cancer Axes for Future Nanomedicine. ACS NANO 2020; 14:14391-14416. [PMID: 33138351 DOI: 10.1021/acsnano.0c07258] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Recent clinical and pathological evidence have implicated the gut microbiota as a nexus for modulating the homeostasis of the human body, impacting conditions from cancer and dementia to obesity and social behavior. The connections between microbiota and human diseases offer numerous opportunities in medicine, most of which have limited or no therapeutic solutions available. In light of this paradigm-setting trend in science, this review aims to provide a comprehensive and timely summary of the mechanistic pathways governing the gut microbiota and their implications for nanomedicines targeting cancer and neurodegenerative diseases. Specifically, we discuss in parallel the beneficial and pathogenic relationship of the gut microbiota along the gut-brain and gut-cancer axes, elaborate on the impact of dysbiosis and the gastrointestinal corona on the efficacy of nanomedicines, and highlight a molecular mimicry that manipulates the universal cross-β backbone of bacterial amyloid to accelerate neurological disorders. This review further offers a forward-looking section on the rational design of cancer and dementia nanomedicines exploiting the gut-brain and gut-cancer axes.
Collapse
Affiliation(s)
- Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xiaoyu Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Monika Mortimer
- Institute of Environmental and Health Sciences, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Nikolaos Andrikopoulos
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
| | - Yuhuan Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai 200032, China
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai 200032, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
28
|
Wang Y, Kankala RK, Zhang J, Hao L, Zhu K, Wang S, Zhang YS, Chen A. Modeling Endothelialized Hepatic Tumor Microtissues for Drug Screening. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002002. [PMID: 33173735 PMCID: PMC7610277 DOI: 10.1002/advs.202002002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/07/2020] [Indexed: 05/03/2023]
Abstract
Compared to various traditional 2D approaches, the scaffold-based 3D tumor models have emerged as an effective strategy to investigate the complex mechanisms behind cancer progression and responses to drug treatments, by providing biomimetic extracellular matrix and stromal-like microenvironments including the vascular elements. Herein, the development of a 3D endothelialized hepatic tumor microtissue model based on the fusion of multicellular aggregates of human hepatocellular carcinoma cells and human umbilical vein endothelial cells cocultured in poly(lactic-co-glycolic acid)-based porous microspheres (PLGA PMs) is reported. In contrast to the conventional 2D culture, the cells within the PLGA PMs exhibit significantly higher half-maximal inhibitory concentration values against anticancer drugs, including doxorubicin and cisplatin. Furthermore, the feasibility of coculturing other cell types, such as fibroblasts (L929) and HepG2 cells, is investigated. Together, the findings emphasize the significance of engineered 3D hepatic tumor microtissue models using PLGA PM-based multicellular aggregates for drug screening applications.
Collapse
Affiliation(s)
- Ying Wang
- Institute of Biomaterials and Tissue EngineeringHuaqiao UniversityXiamen361021P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue EngineeringHuaqiao UniversityXiamen361021P. R. China
- Fujian Provincial Key Laboratory of Biochemical TechnologyHuaqiao UniversityXiamen361021P. R. China
| | - Jianting Zhang
- Institute of Biomaterials and Tissue EngineeringHuaqiao UniversityXiamen361021P. R. China
- Fujian Provincial Key Laboratory of Biochemical TechnologyHuaqiao UniversityXiamen361021P. R. China
| | - Liuzhi Hao
- Institute of Biomaterials and Tissue EngineeringHuaqiao UniversityXiamen361021P. R. China
| | - Kai Zhu
- Department of Cardiac SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Shibin Wang
- Institute of Biomaterials and Tissue EngineeringHuaqiao UniversityXiamen361021P. R. China
- Fujian Provincial Key Laboratory of Biochemical TechnologyHuaqiao UniversityXiamen361021P. R. China
| | - Yu Shrike Zhang
- Division of Engineering in MedicineBrigham and Women's HospitalDepartment of MedicineHarvard Medical SchoolCambridgeMA02139USA
| | - Aizheng Chen
- Institute of Biomaterials and Tissue EngineeringHuaqiao UniversityXiamen361021P. R. China
- Fujian Provincial Key Laboratory of Biochemical TechnologyHuaqiao UniversityXiamen361021P. R. China
| |
Collapse
|
29
|
Lovell JF. Thinking outside the macrocycle: Potential biomedical roles for nanostructured porphyrins and phthalocyanines — a SPP/JPP Young Investigator Award paper. J PORPHYR PHTHALOCYA 2020. [DOI: 10.1142/s1088424620300086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Porphyrins and phthalocyanines feature strong light absorption, capacity for metal chelation, and a track record of use in human therapeutic applications. Various conjugates and formulations of these macrocycles have shown potential to forge new applications in the biomedical sciences. Our lab has explored several such approaches including porphyrin polymer hydrogels, porphyrin-lipid nanovesicles, and surfactant-stripped micelles. These all feature in common a high density of tetrapyrroles, as well as unique functional properties. Porphyrin polymer hydrogels with high porphyrin density and bright fluorescence emission were demonstrated for use as a new class of implantable biosensors. Porphyrin-lipid nanovesicles hold potential for phototherapy, imaging, and also drug and vaccine delivery. Surfactant-stripped micelles have been developed for high-contrast photoacoustic imaging. In this ICPP Young Investigator Award brief perspective, we discuss our own efforts on these fronts. Taken together, the results show that tetrapyrroles enable new approaches for tackling biomedical problems and also confirm what was already well-known to members of the Society of Porphyrins and Phthalocyanines: that these molecules are remarkably versatile and enable research to flow in unexpected directions.
Collapse
Affiliation(s)
- Jonathan F. Lovell
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260 USA
| |
Collapse
|
30
|
Capriotti G, Varani M, Lauri C, Franchi G, Pizzichini P, Signore A. Copper-64 labeled nanoparticles for positron emission tomography imaging: a review of the recent literature. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:346-355. [PMID: 33073558 DOI: 10.23736/s1824-4785.20.03315-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Nuclear medicine plays a crucial role for personalized therapy, mainly in oncology. Chemotherapy and radiotherapy present some disadvantages and research is shifting toward nanotechnology with significant improvements in therapy and diagnosis of several cancers. Indeed, nanoparticles can be tagged with different radioisotopes for single photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging and for therapy. This review describes the current state of the art of 64Copper-labeled nanoparticles for PET imaging of cancer. EVIDENCE ACQUISITION We performed a systematic analysis of literature using the terms "64CuCl<inf>2</inf>," "64Cu," "Copper" AND "nanoparticle" AND "PET" in online databases: i.e. PubMed/MEDLINE and Scopus. The search was limited to English papers and original articles. We excluded articles not in English language, abstracts, case reports, review articles and meeting presentations. EVIDENCE SYNTHESIS Amongst the 116 articles retrieved, 88 were excluded because reviews, or not in English, or only in-vitro studies or meeting presentations. We considered only 28 original papers. The most used nanoparticles are liposomes and they are mainly used in breast cancer although other animal models of cancer have been also investigated. CONCLUSIONS The results showed that nanoparticles can be considered a promising radiopharmaceutical for PET imaging of different type of cancer.
Collapse
Affiliation(s)
- Gabriela Capriotti
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy - .,Unit of Nuclear Medicine, Sant'Andrea University Hospital, Rome, Italy -
| | - Michela Varani
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy
| | - Chiara Lauri
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy
| | - Gabriele Franchi
- Unit of Nuclear Medicine, Sant'Andrea University Hospital, Rome, Italy
| | | | - Alberto Signore
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy.,Unit of Nuclear Medicine, Sant'Andrea University Hospital, Rome, Italy
| |
Collapse
|
31
|
Kumar S, Jiang D, Sun B, Seeley KV, Engle JW, Sia Z, He X, Neelamegham S, Cai W, Lovell JF. Labeling of Erythrocytes by Porphyrin-Phospholipid. ADVANCED NANOBIOMED RESEARCH 2020; 1. [PMID: 34212160 DOI: 10.1002/anbr.202000013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A method is developed for membrane labeling of erythrocytes with porphyrin-phospholipid (PoP). To generate a concentrated PoP solution for labeling human red blood cells (RBCs), various surfactants and solvents are screened to identify conditions that avoid hemolysis, while minimizing non-specific PoP co-precipitation with RBCs in the pellet during centrifugation washes. Cholate, Tween 80 and Tween 40 are identified as useful surfactants for this purpose. When labeled RBCs are mixed with unlabeled ones, substantial non-specific PoP exchange is observed. Egg-yolk lecithin is included in a washing buffer to remove loosely bound PoP and reduce PoP exchange with unlabeled erythrocytes, based on flow cytometry and photodynamic hemolysis assays. Murine RBCs that are labeled with 64Cu-chelated PoP displayed altered biodistribution with longer blood circulation relative to directly administered 64Cu-chelated PoP.
Collapse
Affiliation(s)
- Sunanda Kumar
- Department of Biomedical Engineering, State University of New York at Buffalo, NY 14260, USA
| | - Dawei Jiang
- Department of Materials Science and Engineering, Department of Radiology, Department of Medical Physics, and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, Wisconsin 53705, United States
| | - Boyang Sun
- Department of Biomedical Engineering, State University of New York at Buffalo, NY 14260, USA
| | - Kaelyn V Seeley
- Department of Materials Science and Engineering, Department of Radiology, Department of Medical Physics, and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, Wisconsin 53705, United States
| | - Jonathan W Engle
- Department of Materials Science and Engineering, Department of Radiology, Department of Medical Physics, and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, Wisconsin 53705, United States
| | - Zachary Sia
- Department of Biomedical Engineering, State University of New York at Buffalo, NY 14260, USA
| | - Xuedan He
- Department of Biomedical Engineering, State University of New York at Buffalo, NY 14260, USA
| | - Sriram Neelamegham
- Department of Chemical Engineering, State University of New York at Buffalo, NY 14260, USA
| | - Weibo Cai
- Department of Materials Science and Engineering, Department of Radiology, Department of Medical Physics, and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, Wisconsin 53705, United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering, State University of New York at Buffalo, NY 14260, USA
| |
Collapse
|
32
|
Xie X, Zhan C, Wang J, Zeng F, Wu S. An Activatable Nano-Prodrug for Treating Tyrosine-Kinase-Inhibitor-Resistant Non-Small Cell Lung Cancer and for Optoacoustic and Fluorescent Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003451. [PMID: 32815304 DOI: 10.1002/smll.202003451] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/11/2020] [Indexed: 06/11/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and the cause of high rate of mortality. The epidermal growth factor receptor (EGFR)-targeted tyrosine kinase inhibitors are used to treat NSCLC, yet their curative effects are usually compromised by drug resistance. This study demonstrates a nanodrug for treating tyrosine-kinase-inhibitor-resistant NSCLC through inhibiting upstream and downstream EGFR signaling pathways. The main molecule of the nanodrug is synthesized by linking a tyrosine kinase inhibitor gefitinib and a near-infrared dye (NIR) on each side of a disulfide via carbonate bonds, and the nanodrug is then obtained through nanoparticle formation of the main molecule in aqueous medium and concomitant encapsulation of a serine threonine protein kinase (Akt) inhibitor celastrol. Upon administration, the nanodrug accumulates at the tumor region of NSCLC-bearing mice and releases the drugs for tumor inhibition, and the dye for fluorescence and optoacoustic imaging. Through suppressing the phosphorylation of upstream EGFR and downstream Akt in the EGFR pathway by gefitinib and celastrol, respectively, the nanodrug exhibits high inhibition efficacy against orthotopic NSCLC in mouse models.
Collapse
Affiliation(s)
- Xin Xie
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Chenyue Zhan
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Jie Wang
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Fang Zeng
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Shuizhu Wu
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| |
Collapse
|
33
|
Zhong D, Zhang D, Xie T, Zhou M. Biodegradable Microalgae-Based Carriers for Targeted Delivery and Imaging-Guided Therapy toward Lung Metastasis of Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000819. [PMID: 32297465 DOI: 10.1002/smll.202000819] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 06/11/2023]
Abstract
High delivery efficiency, prolonged drug release, and low systemic toxicity are effective weapons for drug delivery systems to win the battle against metastatic breast cancer. Herein, it is shown that Spirulina platensis (S. platensis) can be used as natural carriers to construct a drug-loaded system for targeted delivery and fluorescence imaging-guided chemotherapy on lung metastasis of breast cancer. The chemotherapeutic doxorubicin (DOX) is loaded into S. platensis (SP) via only one facile step to fabricate the DOX-loaded SP (SP@DOX), which exhibits ultrahigh drug loading efficiency and PH-responsive drug sustained release. The rich chlorophyll endows SP@DOX excellent fluorescence imaging capability for noninvasive tracking and real-time monitoring in vivo. Moreover, the micrometer-sized and spiral-shaped SP carriers enable the as-prepared SP@DOX to passively target the lungs and result in a significantly enhanced therapeutic efficacy on lung metastasis of 4T1 breast cancer. Finally, the undelivered carriers can be biodegraded through renal clearance without notable toxicity. The SP@DOX described here presents a novel biohybrid strategy for targeted drug delivery and effective treatment on cancer metastasis.
Collapse
Affiliation(s)
- Danni Zhong
- Eye Center and Key Laboratory of Cancer Prevention and Intervention, MOE, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Dongxiao Zhang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Tingting Xie
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Min Zhou
- Eye Center and Key Laboratory of Cancer Prevention and Intervention, MOE, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Modern Optical Instrumentations, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
34
|
Jeevarathinam AS, Lemaster JE, Chen F, Zhao E, Jokerst JV. Photoacoustic Imaging Quantifies Drug Release from Nanocarriers via Redox Chemistry of Dye-Labeled Cargo. Angew Chem Int Ed Engl 2020; 59:4678-4683. [PMID: 31840357 PMCID: PMC7101078 DOI: 10.1002/anie.201914120] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Indexed: 12/12/2022]
Abstract
We report a new approach to monitor drug release from nanocarriers via a paclitaxel-methylene blue conjugate (PTX-MB) with redox activity. This construct is in a photoacoustically silent reduced state inside poly(lactic-co-glycolic acid) (PLGA) nanoparticles (PTX-MB@PLGA NPs). During release, PTX-MB is spontaneously oxidized to produce a concentration-dependent photoacoustic signal. An in vitro drug-release study showed an initial burst release (25 %) between 0-24 h and a sustained release between 24-120 h with a cumulative release of 40.6 % and a 670-fold increase in photoacoustic signal. An in vivo murine drug release showed a photoacoustic signal enhancement of up to 649 % after 10 hours. PTX-MB@PLGA NPs showed an IC50 of 78 μg mL-1 and 44.7±4.8 % decrease of tumor burden in an orthotopic model of colon cancer via luciferase-positive CT26 cells.
Collapse
Affiliation(s)
| | - Jeanne E. Lemaster
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Fang Chen
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Department of Radiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Eric Zhao
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Jesse V. Jokerst
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Department of Radiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| |
Collapse
|
35
|
Jeevarathinam AS, Lemaster JE, Chen F, Zhao E, Jokerst JV. Photoacoustic Imaging Quantifies Drug Release from Nanocarriers via Redox Chemistry of Dye‐Labeled Cargo. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914120] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Jeanne E. Lemaster
- Department of NanoEngineering University of California San Diego 9500 Gilman Drive La Jolla CA 92093 USA
| | - Fang Chen
- Department of NanoEngineering University of California San Diego 9500 Gilman Drive La Jolla CA 92093 USA
- Department of Radiology University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
- Current address: Stanford University USA
| | - Eric Zhao
- Department of NanoEngineering University of California San Diego 9500 Gilman Drive La Jolla CA 92093 USA
| | - Jesse V. Jokerst
- Department of NanoEngineering University of California San Diego 9500 Gilman Drive La Jolla CA 92093 USA
- Materials Science and Engineering Program University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
- Department of Radiology University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
| |
Collapse
|
36
|
Liang BJ, Pigula M, Baglo Y, Najafali D, Hasan T, Huang HC. Breaking the selectivity-uptake trade-off of photoimmunoconjugates with nanoliposomal irinotecan for synergistic multi-tier cancer targeting. J Nanobiotechnology 2020; 18:1. [PMID: 31898555 PMCID: PMC6939330 DOI: 10.1186/s12951-019-0560-5] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Photoimmunotherapy involves targeted delivery of photosensitizers via an antibody conjugate (i.e., photoimmunoconjugate, PIC) followed by light activation for selective tumor killing. The trade-off between PIC selectivity and PIC uptake is a major drawback limiting the efficacy of photoimmunotherapy. Despite ample evidence showing that photoimmunotherapy is most effective when combined with chemotherapy, the design of nanocarriers to co-deliver PICs and chemotherapy drugs remains an unmet need. To overcome these challenges, we developed a novel photoimmunoconjugate-nanoliposome (PIC-Nal) comprising of three clinically used agents: anti-epidermal growth factor receptor (anti-EGFR) monoclonal antibody cetuximab (Cet), benzoporphyrin derivative (BPD) photosensitizer, and irinotecan (IRI) chemotherapy. RESULTS The BPD photosensitizers were first tethered to Cet at a molar ratio of 6:1 using carbodiimide chemistry to form PICs. Conjugation of PICs onto nanoliposome irinotecan (Nal-IRI) was facilitated by copper-free click chemistry, which resulted in monodispersed PIC-Nal-IRI with an average size of 158.8 ± 15.6 nm. PIC-Nal-IRI is highly selective against EGFR-overexpressing epithelial ovarian cancer cells with 2- to 6-fold less accumulation in low EGFR expressing cells. Successful coupling of PIC onto Nal-IRI enhanced PIC uptake and photoimmunotherapy efficacy by up to 30% in OVCAR-5 cells. Furthermore, PIC-Nal-IRI synergistically reduced cancer viability via a unique three-way mechanism (i.e., EGFR downregulation, mitochondrial depolarization, and DNA damage). CONCLUSION It is increasingly evident that the most effective therapies for cancer will involve combination treatments that target multiple non-overlapping pathways while minimizing side effects. Nanotechnology combined with photochemistry provides a unique opportunity to simultaneously deliver and activate multiple drugs that target all major regions of a cancer cell-plasma membrane, cytoplasm, and nucleus. PIC-Nal-IRI offers a promising strategy to overcome the selectivity-uptake trade-off, improve photoimmunotherapy efficacy, and enable multi-tier cancer targeting. Controllable drug compartmentalization, easy surface modification, and high clinical relevance collectively make PIC-Nal-IRI extremely valuable and merits further investigations in living animals.
Collapse
Affiliation(s)
- Barry J Liang
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA
| | - Michael Pigula
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yan Baglo
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA
| | - Daniel Najafali
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA.
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
37
|
Sun B, Chitgupi U, Li C, Federizon J, Zhang C, Ruszaj DM, Razi A, Ortega J, Neelamegham S, Zhang Y, Lovell JF. Surfactant‐Stripped Cabazitaxel Micelles Stabilized by Clotrimazole or Mifepristone. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Boyang Sun
- Department of Biomedical Engineering, University at Buffalo State University of New York (SUNY) Buffalo NY 14260 USA
- Department of Chemical and Biological Engineering, University at Buffalo SUNY Buffalo NY 14260 USA
| | - Upendra Chitgupi
- Department of Biomedical Engineering, University at Buffalo State University of New York (SUNY) Buffalo NY 14260 USA
| | - Changning Li
- Department of Chemical and Biological Engineering, University at Buffalo SUNY Buffalo NY 14260 USA
| | - Jasmin Federizon
- Department of Biomedical Engineering, University at Buffalo State University of New York (SUNY) Buffalo NY 14260 USA
| | - Changjie Zhang
- Department of Chemical and Biological Engineering, University at Buffalo SUNY Buffalo NY 14260 USA
| | - Donna M. Ruszaj
- Department of Pharmaceutical Sciences University at Buffalo SUNY Buffalo NY 14260 USA
| | - Aida Razi
- Department of Anatomy and Cell Biology Mcgill University Montreal Quebec L8S4L8 Canada
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology Mcgill University Montreal Quebec L8S4L8 Canada
| | - Sriram Neelamegham
- Department of Chemical and Biological Engineering, University at Buffalo SUNY Buffalo NY 14260 USA
| | - Yumiao Zhang
- School of Chemical Engineering and Technology Tianjin University Tianjin 301636 China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo State University of New York (SUNY) Buffalo NY 14260 USA
- Department of Chemical and Biological Engineering, University at Buffalo SUNY Buffalo NY 14260 USA
| |
Collapse
|
38
|
Chen J, Liang C, Song X, Yi X, Yang K, Feng L, Liu Z. Hybrid Protein Nano-Reactors Enable Simultaneous Increments of Tumor Oxygenation and Iodine-131 Delivery for Enhanced Radionuclide Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903628. [PMID: 31577387 DOI: 10.1002/smll.201903628] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/31/2019] [Indexed: 06/10/2023]
Abstract
It is hard for current radionuclide therapy to render solid tumors desirable therapeutic efficacy owing to insufficient tumor-targeted delivery of radionuclides and severe tumor hypoxia. In this study, a biocompatible hybrid protein nanoreactor composed of human serum albumin (HSA) and catalase (CAT) molecules is constructed via glutaraldehyde-mediated crosslinking. The obtained HSA-CAT nanoreactors (NRs) show retained and well-protected enzyme stability in catalyzing the decomposition of H2 O2 and enable efficient labeling of therapeutic radionuclide iodine-131 (131 I). Then, it is uncovered that such HSA-CAT NRs after being intravenously injected into tumor-bearing mice exhibit efficient passive tumor accumulation as vividly visualized under the fluorescence imaging system and gamma camera. As the result, such HSA-CAT NRs upon tumor accumulation would significantly attenuate tumor hypoxia by decomposing endogenous H2 O2 produced by cancer cells to molecular oxygen, and thereby remarkably improve the therapeutic efficacy of radionuclide 131 I. This study highlights the concise preparation of biocompatible protein nanoreactors with efficient tumor homing and hypoxia attenuation capacities, thus enabling greatly improved tumor radionuclide therapy with promising potential for future clinical translation.
Collapse
Affiliation(s)
- Jiawen Chen
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, P. R. China
| | - Chao Liang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, P. R. China
| | - Xuejiao Song
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, P. R. China
| | - Xuan Yi
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Kai Yang
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Liangzhu Feng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, P. R. China
| | - Zhuang Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, P. R. China
| |
Collapse
|
39
|
Ghosh S, Carter KA, Lovell JF. Liposomal formulations of photosensitizers. Biomaterials 2019; 218:119341. [PMID: 31336279 PMCID: PMC6663636 DOI: 10.1016/j.biomaterials.2019.119341] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 12/29/2022]
Abstract
Photodynamic therapy (PDT) is a clinical ablation modality to treat cancers and other diseases. PDT involves administration of a photosensitizer, followed by irradiation of target tissue with light. As many photosensitizers are small and hydrophobic, solubilization approaches and nanoscale delivery vehicles have been extensively explored. Liposomes and lipid-based formulations have been used for the past 30 years, and in some cases have been developed into well-defined commercial PDT products. This review provides an overview of common liposomal formulation strategies for photosensitizers for PDT and also photothermal therapy. Furthermore, research efforts have examined the impact of co-loading therapeutic cargo along with photosensitizers within liposomes. Additional recent approaches including imaging, overcoming hypoxia, upconversion and activatable liposomal formulations are discussed.
Collapse
Affiliation(s)
- Sanjana Ghosh
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Kevin A Carter
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA.
| |
Collapse
|
40
|
Cyclodextrin-functionalized asymmetric block copolymer films as high-capacity reservoir for drug delivery. J Memb Sci 2019. [DOI: 10.1016/j.memsci.2019.04.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
41
|
Tsolekile N, Nelana S, Oluwafemi OS. Porphyrin as Diagnostic and Therapeutic Agent. Molecules 2019; 24:E2669. [PMID: 31340553 PMCID: PMC6680575 DOI: 10.3390/molecules24142669] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 11/16/2022] Open
Abstract
The synthesis and application of porphyrins has seen a huge shift towards research in porphyrin bio-molecular based systems in the past decade. The preferential localization of porphyrins in tumors, as well as their ability to generate reactive singlet oxygen and low dark toxicities has resulted in their use in therapeutic applications such as photodynamic therapy. However, their inherent lack of bio-distribution due to water insolubility has shifted research into porphyrin-nanomaterial conjugated systems to address this challenge. This has broadened their bio-applications, viz. bio-sensors, fluorescence tracking, in vivo magnetic resonance imaging (MRI), and positron emission tomography (PET)/CT imaging to photo-immuno-therapy just to highlight a few. This paper reviews the unique theranostic role of porphyrins in disease diagnosis and therapy. The review highlights porphyrin conjugated systems and their applications. The review ends by bringing current challenges and future perspectives of porphyrin based conjugated systems and their respective applications into light.
Collapse
Affiliation(s)
- Ncediwe Tsolekile
- Department of Chemical Sciences (formerly Applied Chemistry), University of Johannesburg, P. O. Box 17011, Doornfontein 2028, Johannesburg, South Africa
- Department of Applied Chemistry, University of Johannesburg, P. O. Box 17011, Doornfontein 2028, Johannesburg, South Africa
- Department of Chemistry, Cape Peninsula University of Technology, P.O. Box 652, Cape Town 2000, South Africa
| | - Simphiwe Nelana
- Department of Chemistry, Vaal University of Technology, Private Bag X021, Vanderbijlpark 1900, South Africa.
| | - Oluwatobi Samuel Oluwafemi
- Department of Chemical Sciences (formerly Applied Chemistry), University of Johannesburg, P. O. Box 17011, Doornfontein 2028, Johannesburg, South Africa.
- Department of Applied Chemistry, University of Johannesburg, P. O. Box 17011, Doornfontein 2028, Johannesburg, South Africa.
| |
Collapse
|
42
|
Xia Y, Xu C, Zhang X, Ning P, Wang Z, Tian J, Chen X. Liposome-based probes for molecular imaging: from basic research to the bedside. NANOSCALE 2019; 11:5822-5838. [PMID: 30888379 DOI: 10.1039/c9nr00207c] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Molecular imaging is very important in disease diagnosis and prognosis. Liposomes are excellent carriers for different types of molecular imaging probes. In this work, we summarize current developments in liposome-based probes used for molecular imaging and their applications in image-guided drug delivery and tumour surgery, including computed tomography (CT), ultrasound imaging (USI), magnetic resonance imaging (MRI), positron emission tomography (PET), fluorescence imaging (FLI) and photoacoustic imaging (PAI). We also summarized liposome-based multimodal imaging probes and new targeting strategies for liposomes. This work will offer guidance for the design of liposome-based imaging probes for future clinical applications.
Collapse
Affiliation(s)
- Yuqiong Xia
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China.
| | | | | | | | | | | | | |
Collapse
|
43
|
Man F, Gawne PJ, T M de Rosales R. Nuclear imaging of liposomal drug delivery systems: A critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019; 143:134-160. [PMID: 31170428 PMCID: PMC6866902 DOI: 10.1016/j.addr.2019.05.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
The integration of nuclear imaging with nanomedicine is a powerful tool for efficient development and clinical translation of liposomal drug delivery systems. Furthermore, it may allow highly efficient imaging-guided personalised treatments. In this article, we critically review methods available for radiolabelling liposomes. We discuss the influence that the radiolabelling methods can have on their biodistribution and highlight the often-overlooked possibility of misinterpretation of results due to decomposition in vivo. We stress the need for knowing the biodistribution/pharmacokinetics of both the radiolabelled liposomal components and free radionuclides in order to confidently evaluate the images, as they often share excretion pathways with intact liposomes (e.g. phospholipids, metallic radionuclides) and even show significant tumour uptake by themselves (e.g. some radionuclides). Finally, we describe preclinical and clinical studies using radiolabelled liposomes and discuss their impact in supporting liposomal drug development and clinical translation in several diseases, including personalised nanomedicine approaches.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Peter J Gawne
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand Campus, London WC2R 2LS, United Kingdom.
| |
Collapse
|
44
|
Li X, Bottini M, Zhang L, Zhang S, Chen J, Zhang T, Liu L, Rosato N, Ma X, Shi X, Wu Y, Guo W, Liang XJ. Core-Satellite Nanomedicines for in Vivo Real-Time Monitoring of Enzyme-Activatable Drug Release by Fluorescence and Photoacoustic Dual-Modal Imaging. ACS NANO 2019; 13:176-186. [PMID: 30592401 DOI: 10.1021/acsnano.8b05136] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
It remains an unresolved challenge to achieve spatial and temporal monitoring of drug release from nanomedicines (NMs) in vivo, which is of crucial importance in disease treatment. To tackle this issue, we constructed core-satellite ICG/DOX@Gel-CuS NMs, which consist of gelatin (Gel) nanoparticles (NPs) with payloads of near-infrared fluorochrome indocyanine green (ICG) and chemo-drug doxorubicin (DOX) and surrounding CuS NPs. The fluorescence of ICG was initially shielded by satellite CuS NPs within the intact ICG/DOX@Gel-CuS NMs and increased in proportion to the amount of DOX released from NMs in response to enzyme-activated NMs degradation. For more comprehensive understanding of the drug-release profile, a theoretical model derived from computer simulation was employed to reconstruct the enzyme-activatable drug release of the ICG/DOX@Gel-CuS NMs, which demonstrated the underlying kinetics functional relationship between the released DOX amount and recovered ICG fluorescence intensity. The kinetics of drug release in vivo was assessed by administrating ICG/DOX@Gel-CuS NMs both locally and systemically into MDA-MB-231 tumor-bearing mice. Upon accumulation of ICG/DOX@Gel-CuS NMs in the tumor, overexpressed enzymes triggered the degradation of the gelatin scaffold as well as the release of DOX and ICG, which can be visually depicted with the ICG fluorescence signal increasing only in the tumor area by fluorescence imaging. Additionally, the photoacoustic signal from CuS NPs was independent from the physical status of ICG/DOX@Gel-CuS NMs and hence was utilized for real-time NMs tracking. Thus, by taking advantage of the core-satellite architecture and NMs degradability in tumor site, the DOX release profile of ICG/DOX@Gel-CuS NMs was monitored by fluorescence and photoacoustic dual-modal imaging in a real-time noninvasive manner.
Collapse
Affiliation(s)
- Xianlei Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Massimo Bottini
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Luyao Zhang
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- LNM, Institute of Mechanics, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Shuai Zhang
- CAS Key Laboratory of Molecular Imaging , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Jing Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
| | - Tingbin Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
| | - Lu Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Nicola Rosato
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Xibo Ma
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- CAS Key Laboratory of Molecular Imaging , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Xinghua Shi
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- CAS Key Laboratory for Nanosystem and Hierarchy Fabrication, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Weisheng Guo
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital , Guangzhou Medical University , Guangzhou 510260 , P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| |
Collapse
|
45
|
Asampille G, Verma BK, Swain M, Shettar A, Rosenzweig SA, Kondaiah P, Atreya HS. An ultra-stable redox-controlled self-assembling polypeptide nanotube for targeted imaging and therapy in cancer. J Nanobiotechnology 2018; 16:101. [PMID: 30526620 PMCID: PMC6286583 DOI: 10.1186/s12951-018-0427-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/24/2018] [Indexed: 01/10/2023] Open
Abstract
We introduce a self-assembling polypeptide-based nanotube system having the ability to specifically target cancer cells. The nanotubes target the cancer cell surface through integrin engagement with the help of multiple RGD units present along their surface. While the nanotubes are non-toxic towards cells in general, they can be loaded with suitable drugs to be released in a sustained manner in cancer cells. In addition, the nanotubes can be utilized for cellular imaging using any covalently tagged fluorescent dye. They are stable over a wide range of temperature due to intermolecular disulphide bonds formed during the self-assembly process. At the same time, presence of disulphide bonds provides a redox molecular switch for their degradation. Taken together this system provides a unique avenue for multimodal formulation in cancer therapy.
Collapse
Affiliation(s)
- Gitanjali Asampille
- NMR Research Centre, Indian Institute of Science, Bangalore, 560012, India.,Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Brijesh Kumar Verma
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Monalisa Swain
- NMR Research Centre, Indian Institute of Science, Bangalore, 560012, India.,Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore, 560012, India.,Basic Research Laboratory, National Cancer Institute, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Abhijith Shettar
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.,Biotechnology Engineering, Ramaiah Institute of Technology, Bangalore, Karnataka, 560054, India
| | - Steven A Rosenzweig
- Department of Cell and Molecular Pharmacology, & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Paturu Kondaiah
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| | - Hanudatta S Atreya
- NMR Research Centre, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
46
|
Ma Y, Zhou H, Hu F, Pei Z, Xu Y, Shuai Q. Multifunctional nanogel engineering with redox-responsive and AIEgen features for the targeted delivery of doxorubicin hydrochloride with enhanced antitumor efficiency and real-time intracellular imaging. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S900-S910. [DOI: 10.1080/21691401.2018.1518910] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Yuwei Ma
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi Province, PR China
| | - Huiyi Zhou
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi Province, PR China
| | - Fan Hu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi Province, PR China
| | - Zhichao Pei
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi Province, PR China
| | - Yongqian Xu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi Province, PR China
| | - Qi Shuai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi Province, PR China
| |
Collapse
|
47
|
Aguilar-Ortíz E, Jalilian AR, Ávila-Rodríguez MA. Porphyrins as ligands for 64copper: background and trends. MEDCHEMCOMM 2018; 9:1577-1588. [PMID: 30429966 PMCID: PMC6194497 DOI: 10.1039/c8md00263k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Porphyrins and 64Cu have emerged as a novel synergic option for applications in PET molecular imaging. Both the characteristics and photophysical properties of macrocyclic porphyrins and the relatively long half-life of the copper isotope, in addition to the increased tumor-specific uptake of porphyrins compared to normal cells, make this complex an attractive option not only for diagnosis but also for therapeutic applications. Herein, we present an overview of the latest results on the development of PET agents based on porphyrins and 64Cu, including methods used to improve the selectivity of these macrocycles when conjugated with biological units such as monoclonal antibodies, peptides or proteins.
Collapse
Affiliation(s)
- Edgar Aguilar-Ortíz
- Unidad Radiofarmacia-Ciclotrón , División de Investigación , Facultad de Medicina , Universidad Nacional Autónoma de México , 04510 Cd. Mx. , Mexico . ;
| | - Amir R Jalilian
- Department of Nuclear Sciences and Applications , International Atomic Energy Agency (IAEA) , Vienna , Austria
| | - Miguel A Ávila-Rodríguez
- Unidad Radiofarmacia-Ciclotrón , División de Investigación , Facultad de Medicina , Universidad Nacional Autónoma de México , 04510 Cd. Mx. , Mexico . ;
| |
Collapse
|
48
|
Cai Y, Si W, Huang W, Chen P, Shao J, Dong X. Organic Dye Based Nanoparticles for Cancer Phototheranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1704247. [PMID: 29611290 DOI: 10.1002/smll.201704247] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/03/2018] [Indexed: 06/08/2023]
Abstract
Phototheranostics, which simultaneously combines photodynamic and/or photothermal therapy with deep-tissue diagnostic imaging, is a promising strategy for the diagnosis and treatment of cancers. Organic dyes with the merits of strong near-infrared absorbance, high photo-to-radical and/or photothermal conversion efficiency, great biocompatibility, ready chemical structure fine-tuning capability, and easy metabolism, have been demonstrated as attractive candidates for clinical phototheranostics. These organic dyes can be further designed and fabricated into nanoparticles (NPs) using various strategies. Compared to free molecules, these NPs can be equipped with multiple synergistic functions and show longer lifetime in blood circulation and passive tumor-targeting property via the enhanced permeability and retention effect. In this article, the recent progress of organic dye-based NPs for cancer phototheranostic applications is summarized, which extends the anticancer arsenal and holds promise for clinical uses in the near future.
Collapse
Affiliation(s)
- Yu Cai
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Weili Si
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211800, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211800, China
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Peng Chen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Jinjun Shao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211800, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211800, China
| |
Collapse
|
49
|
Yan L, Amirshaghaghi A, Huang D, Miller J, Stein JM, Busch TM, Cheng Z, Tsourkas A. Protoporphyrin IX (PpIX)-Coated Superparamagnetic Iron Oxide Nanoparticle (SPION) Nanoclusters for Magnetic Resonance Imaging and Photodynamic Therapy. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1707030. [PMID: 29910700 PMCID: PMC5997278 DOI: 10.1002/adfm.201707030] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The ability to produce nanotherapeutics at large-scale with high drug loading efficiency, high drug loading capacity, high stability, and high potency is critical for clinical translation. However, many nanoparticle-based therapeutics under investigation suffer from complicated synthesis, poor reproducibility, low stability, and high cost. In this work, a simple method for preparing multifunctional nanoparticles is utilized that act as both a contrast agent for magnetic resonance imaging and a photosensitizer for photodynamic therapy for the treatment of cancer. In particular, the photosensitizer protoporphyrin IX (PpIX) is used to solubilize small nanoclusters of superparamagnetic iron oxide nanoparticles (SPIONs) without the use of any additional carrier materials. These nanoclusters are characterized with a high PpIX loading efficiency; a high loading capacity, stable behavior; high potency; and a synthetic approach that is amenable to large-scale production. In vivo studies of photodynamic therapy (PDT) efficacy show that the PpIX-coated SPION nanoclusters lead to a significant reduction in the growth rate of tumors in a syngeneic murine tumor model compared to both free PpIX and PpIX-loaded poly(ethylene glycol)-polycaprolactone micelles, even when injected at 1/8th the dose. These results suggest that the nanoclusters developed in this work can be a promising nanotherapeutic for clinical translation.
Collapse
Affiliation(s)
- Lesan Yan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahmad Amirshaghaghi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joann Miller
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joel M Stein
- Department of Radiology, University of Pennsylvania, PA 19104, USA
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
50
|
Luo D, Carter KA, Geng J, He X, Lovell JF. Short Drug-Light Intervals Improve Liposomal Chemophototherapy in Mice Bearing MIA PaCa-2 Xenografts. Mol Pharm 2018; 15:3682-3689. [PMID: 29608312 DOI: 10.1021/acs.molpharmaceut.8b00052] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Chemophototherapy (CPT) is an emerging tumor treatment that combines phototherapy and chemotherapy. Long-circulating (LC) liposomes can stably incorporate 2 mol % porphyrin-phospholipid (PoP) in the bilayer and load doxorubicin (Dox) to generate LC-Dox-PoP liposomes, for single-agent CPT. Following intravenous administration to mice, LC-Dox-PoP liposomes (2 mg/kg Dox) circulated with similar blood concentration ranges produced by a typical human clinical dose of DOXIL (50 mg/m2 Dox). This dosing approach aims to achieve physiologically relevant Dox and PoP concentrations as well as CPT vascular responses in mice bearing subcutaneous human pancreatic MIA PaCa-2 xenografts. Phototreatment with 2 mg/kg LC-Dox-PoP induced vascular permeabilization, leading to a 12.5-fold increase in Dox tumor influx estimated by a pharmacokinetic model, based on experimental data. Shorter drug-light intervals (0.5-3 h) led to greater tumoral drug deposition and improved treatment outcomes, compared to longer drug-light intervals. At 2 mg/kg Dox, CPT with LC-Dox-PoP liposomes induced tumor regression and growth inhibition, whereas chemotherapy using several other formulations of Dox did not. LC-Dox-PoP liposomes were well tolerated at the 2 mg/kg dose.
Collapse
Affiliation(s)
- Dandan Luo
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| | - Kevin A Carter
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| | - Jumin Geng
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| | - Xuedan He
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| |
Collapse
|