1
|
Al-Taie A, Özcan Bülbül E. A paradigm use of monoclonal antibodies-conjugated nanoparticles in breast cancer treatment: current status and potential approaches. J Drug Target 2024; 32:45-56. [PMID: 38096045 DOI: 10.1080/1061186x.2023.2295803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/12/2023] [Indexed: 12/30/2023]
Abstract
Monoclonal antibodies (mAbs) are integral to cancer treatment over conventional non-specific therapy methods. This study provides a scoping review of the clinically approved mAbs, focusing on the current application of different nanocarrier technologies as drug delivery targets for mAb-conjugated nanoparticles (NPs) as potential features for breast cancer (BC) treatment. An extensive literature search was conducted between the years 2000 and 2023 using various sources of databases. The first part covered mAb classification, types, and mechanisms of action, pharmacokinetics and clinical applications in BC. The second part covered polymeric, lipid and inorganic-based NPs, which are a variety of mAb-conjugated NPs targeting BC. A total of 20 relevant studies were enrolled indicating there are three different types of nanoparticular systems (polymeric NPs, inorganic NPs and lipid-based NPs) that can be used for BC treatment by being loaded with various active substances and conjugated with these antibodies. While mAbs have altered the way in cancer treatment due to targeting cancer cells specifically, the delivery of mAbs with nanoparticulate systems is important in the treatment of BC, as NPs are still being investigated as distinctive and promising drug delivery methods that can be employed for effective treatment of BC.
Collapse
Affiliation(s)
- Anmar Al-Taie
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| | - Ece Özcan Bülbül
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| |
Collapse
|
2
|
Prasad R, Kumari R, Chaudhari R, Kumar R, Kundu GC, Kumari S, Roy G, Gorain M, Chandra P. Emissive Lipid Nanoparticles as Biophotonic Contrast Agent for Site-Selective Solid Tumor Imaging in Pre-Clinical Models. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53393-53404. [PMID: 39324588 DOI: 10.1021/acsami.4c08273] [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: 09/27/2024]
Abstract
Small organic dye-based fluorescent agents are highly potent in solid tumor imaging but face challenges such as poor photostability, nonspecific distribution, low circulation, and weak tumor binding. Nanocarriers overcome these issues with better physicochemical and biological performance, particularly in cancer imaging. Among the various nanosized carriers, lipid formulations are clinically approved but yet to be designed as bright nanocontrast agents for solid tumor diagnosis without affecting surrounding tissues. Herein, indocyanine green (ICG) encapsulated targetable lipid nanoparticles (698 ICG/LNPs) as safe contrast agents (∼200 nm) have been developed and tested for solid tumor imaging and biodistribution. Our findings reveal that nanoprecipitation produces ICG-LNPs with a unique assembly, which contributes to their high brightness with improved quantum yield (3.5%) in aqueous media. The bright, optically stable (30 days) biophotonic agents demonstrate rapid accumulation (within 1 h) and prolonged retention (for up to 168 h) at the primary tumor site, with better signal intensity following a one-time dose administration (17.7 × 109 LNP per dose). Incorporated folic acid (735 folic acid/LNPs) helps in selective tumor binding and the specific biodistribution of intravenously injected nanoparticles without affecting healthy tissues. Designed targetable ICG-LNP (634 MESF) demonstrates high-contrast fluorescence and resolution from the tumor area as compared to the targetable ICG-liposomal nanoparticles (532 MESF). Various in vitro and in vivo findings reveal that the cancer diagnostic efficacy elicited by designed bright lipid nanoparticles are comparable to reported clinically accepted imaging agents. Thus, such LNPs hold translational potential for cancer diagnosis at an early stage.
Collapse
Affiliation(s)
- Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rohini Kumari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Ruchita Chaudhari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rahul Kumar
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Gopal Chandra Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411008, India
- School of Biotechnology and Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Simpy Kumari
- Sahu Bio-Tech Services, Utsav Society, Nandoshi Road, Kirkatwadi, Pune, Maharashtra 411024, India
| | - Gaurab Roy
- Sahu Bio-Tech Services, Utsav Society, Nandoshi Road, Kirkatwadi, Pune, Maharashtra 411024, India
| | - Mahadeo Gorain
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411008, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
3
|
Giordano A, Provenza AC, Reverchon G, Baldino L, Reverchon E. Lipid-Based Nanocarriers: Bridging Diagnosis and Cancer Therapy. Pharmaceutics 2024; 16:1158. [PMID: 39339195 PMCID: PMC11434863 DOI: 10.3390/pharmaceutics16091158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Theranostics is a growing field that matches diagnostics and therapeutics. In this approach, drugs and techniques are uniquely coupled to diagnose and treat medical conditions synergically or sequentially. By integrating diagnostic and treatment functions in a single platform, the aim of theranostics is to improve precision medicine by tailoring treatments based on real-time information. In this context, lipid-based nanocarriers have attracted great scientific attention due to their biodegradability, biocompatibility, and targeting capabilities. The present review highlights the latest research advances in the field of lipid-based nanocarriers for cancer theranostics, exploring several ways of improving in vivo performance and addressing associated challenges. These nanocarriers have significant potential to create new perspectives in the field of nanomedicine and offer promise for a significant step towards more personalized and precise medicine, reducing side effects and improving clinical outcomes for patients. This review also presents the actual barriers to and the possible challenges in the use of nanoparticles in the theranostic field, such as regulatory hurdles, high costs, and technological integration. Addressing these issues through a multidisciplinary and collaborative approach among institutions could be essential for advancing lipid nanocarriers in the theranostic field. Such collaborations can leverage diverse expertise and resources, fostering innovation and overcoming the complex challenges associated with clinical translation. This approach will be crucial for realizing the full potential of lipid-based nanocarriers in precision medicine.
Collapse
Affiliation(s)
- Alessandra Giordano
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.G.); (A.C.P.); (E.R.)
| | - Anna Chiara Provenza
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.G.); (A.C.P.); (E.R.)
| | - Giorgio Reverchon
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Via G.C. Pupilli, 1, 40136 Bologna, Italy;
| | - Lucia Baldino
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.G.); (A.C.P.); (E.R.)
| | - Ernesto Reverchon
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.G.); (A.C.P.); (E.R.)
| |
Collapse
|
4
|
Lin Q, Jing Y, Yan C, Chen X, Zhang Q, Lin X, Xu Y, Chen B. Design and Application of pH-Responsive Liposomes for Site-Specific Delivery of Cytotoxin from Cobra Venom. Int J Nanomedicine 2024; 19:5381-5395. [PMID: 38859950 PMCID: PMC11164093 DOI: 10.2147/ijn.s461728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/25/2024] [Indexed: 06/12/2024] Open
Abstract
Background Current immunotherapies with unexpected severe side effects and treatment resistance have not resulted in the desired outcomes for patients with melanoma, and there is a need to discover more effective medications. Cytotoxin (CTX) from Cobra Venom has been established to have favorable cytolytic activity and antitumor efficacy and is regarded as a promising novel anticancer agent. However, amphiphilic CTX with excellent anionic phosphatidylserine lipid-binding ability may also damage normal cells. Methods We developed pH-responsive liposomes with a high CTX load (CTX@PSL) for targeted acidic-stimuli release of drugs in the tumor microenvironment. The morphology, size, zeta potential, drug-release kinetics, and preservation stability were characterized. Cell uptake, apoptosis-promoting effects, and cytotoxicity were assessed using MTT assay and flow cytometry. Finally, the tissue distribution and antitumor effects of CTX@PSL were systematically assessed using an in vivo imaging system. Results CTX@PSL exhibited high drug entrapment efficiency, drug loading, stability, and a rapid release profile under acidic conditions. These nanoparticles, irregularly spherical in shape and small in size, can effectively accumulate at tumor sites (six times higher than free CTX) and are rapidly internalized into cancer cells (2.5-fold higher cell uptake efficiency). CTX@PSL displayed significantly stronger cytotoxicity (IC50 0.25 μg/mL) and increased apoptosis in than the other formulations (apoptosis rate 71.78±1.70%). CTX@PSL showed considerably better tumor inhibition efficacy than free CTX or conventional liposomes (tumor inhibition rate 79.78±5.93%). Conclusion Our results suggest that CTX@PSL improves tumor-site accumulation and intracellular uptake for sustained and targeted CTX release. By combining the advantages of CTX and stimuli-responsive nanotechnology, the novel CTX@PSL nanoformulation is a promising therapeutic candidate for cancer treatment.
Collapse
Affiliation(s)
- Qing Lin
- Department of Pharmacy, Affiliated Fuzhou First Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
| | - Yafei Jing
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Cailing Yan
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
| | - Xinyi Chen
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
| | - Qiong Zhang
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
| | - Xinhua Lin
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Yunlu Xu
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
- Center of Translational Hematology, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Bing Chen
- School of Pharmacy, Fujian Medical University, Fujian, People’s Republic of China
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| |
Collapse
|
5
|
Myat YY, Sahatsapan N, Rojanarata T, Ngawhirunpat T, Opanasopit P, Pornpitchanarong C, Patrojanasophon P. Antibody-decorated chitosan-iodoacetamide-coated nanocarriers for the potential delivery of doxorubicin to breast cancer cells. Int J Biol Macromol 2024; 258:128797. [PMID: 38104687 DOI: 10.1016/j.ijbiomac.2023.128797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Using an active targeting approach of chemotherapeutics-loaded nanocarriers (NCs) with monoclonal antibodies is a potential strategy to improve the specificity of the delivery systems and reduce adverse reactions of chemotherapeutic drugs. Specific targeting of the human epidermal growth factor receptor-2 (HER-2), expressed excessively in HER-2-positive breast cancer cells, can be achieved by conjugating NCs with an anti-HER-2 monoclonal antibody. We constructed trastuzumab-conjugated chitosan iodoacetamide-coated NCs containing doxorubicin (Tras-Dox-CHI-IA-NCs) as a tumor-targeted drug delivery system, during the study. Chitosan-iodoacetamide (CHI-IA) was synthesized and utilized to prepare trastuzumab-conjugated NCs (Tras-NCs). The morphology, physicochemical properties, drug loading, drug release, and biological activities of the NCs were elucidated. The Tras-NCs were spherical, with a particle size of approximately 76 nm, and had a positive zeta potential; after incorporating the drug, the size of the Tras-NC increased. A prolonged, 24-h drug release from the NCs was achieved. The Tras-NCs exhibited high cellular accumulation and significantly higher antitumor activity against HER-2-positive breast cancer cells than the unconjugated NCs and the drug solution. Therefore, Tras-Dox-CHI-IA-NCs could be a promising nanocarrier for HER-2-positive breast cancer.
Collapse
Affiliation(s)
- Yin Yin Myat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Nitjawan Sahatsapan
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 160 00, Czech Republic
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Chaiyakarn Pornpitchanarong
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
6
|
Nasri N, Saharkhiz S, Dini G, Yousefnia S. Thermo- and pH-responsive targeted lipid-coated mesoporous nano silica platform for dual delivery of paclitaxel and gemcitabine to overcome HER2-positive breast cancer. Int J Pharm 2023; 648:123606. [PMID: 37972671 DOI: 10.1016/j.ijpharm.2023.123606] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/09/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023]
Abstract
In the current study, a new monoclonal antibody conjugated dual stimuli lipid-coated mesoporous silica nanoparticles (L-MSNs) platform was developed and investigated for specific co-delivery of the paclitaxel (PTX) and gemcitabine (Gem) to cancer cells and preventing their side effects during the treatment process. First, MSNs were synthesized and then coated with as-prepared pH-, and thermo-sensitive niosomes to produce L-MSNs. For this aim, Dipalmitoylphosphatidylcholine (DPPC) was used to create thermo-sensitivity, and 1, 2-Distearoyl-sn-glycerol-3-phosphoethanolamine -Citraconic Anhydride-Polyethylene Glycol (DSPE-CA-PEG) polymers were prepared and incorporated to the lipid layer for creation of pH-sensitivity. In the next step, trastuzumab as a monoclonal antibody (mAb) was conjugated to the maleimide groups of the 1, 2-Distearoyl-sn-glycerol-3-phosphoethanolamine DSPE-polyethylene glycol (PEG)-maleimide agents in the lipid bilayer via a disulfide bond. Dynamic light scattering (DLS) and zeta potential measurements, Fourier-transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), Brunauer-Emmett-Teller (BET), and scanning electron microscopy (SEM) analyses were utilized to characterize the synthesized particles before and after surface modification. The encapsulation efficiency (EE%) and loading efficiency (LE%) of the particles were also evaluated. Additionally, the drug release study and MTT assay were done to evaluate the bioactivity potential of the fabricated platforms. The results of DLS and zeta potential measurements revealed an average size of 200 nm and a neutral zeta potential of about -1 mV for mAb-L-MSNs. Also, the FTIR spectra confirmed the formation of mAb-L-MSNs. Moreover, SEM analysis showed spherical-shaped MSNs with amorphous structure confirmed by XRD analysis, and BET test revealed ∼ 820 m2/g specific surface area and pore about 5 nm in size. The values of EE% and LE% of PTX were 90.3 % and 26.7 %, while these values for GEM were 89.5 % and 38.8 % in the co-loaded form, respectively. The thermo-pH-sensitivity examination showed approximately 500 nm of size increase after the change of pH and temperature from 7.4 and 37˚C to 5 and 42˚C. The release profile showed a pH-, and thermo-dependence manner, which led to about 89 % and 95 % of PTX and GEM released from the co-loaded platform at a pH of 5 and 42 °C while these values were 31.1 % and 32.2 % at pH of 7.4 and 37˚C, respectively. MTT assay data presented that when the mAb-L-co-loaded-MSNs platform containing 250 µg/mL drug was used, about 92 % of cells died in human epidermal receptors (HER2)-positive breast cancer cells (SKBR3), while just about 4 % of HER2-negative normal cells were killed. However, the growth inhibition rate of SKBR3 cells was caused by empty-mAb-L-MSNs, pure PTX and GEM combination were 9 % and 87 %, respectively. Moreover, the half inhibitory concentration (IC50) of the pure PTX, pure GEM, and mAb-coloaded-L-MSNs were 33, 17.6, and 6.5 µg/mL. The synergic effect of co-encapsulation of PTX and GEM in addition to trastuzumab conjugated L-MSNs was confirmed by a combinational index (CI) of 0.34. Therefore, this strategy leads to specific targeted drug delivery to cancer cells using a key-lock interaction between the trastuzumab and HER-2 receptors on the cancer cell membrane which stimuli the endocytosis of the particles to the cells followed by the destruction of the lipid layer in the acidic pH and the temperature of the lysosome, leading to enhanced release of PTX and GEM (pH of 5 and 42˚C). So, this platform can be considered a suitable carrier for cancer treatment.
Collapse
Affiliation(s)
- Negar Nasri
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Shaghayegh Saharkhiz
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Ghasem Dini
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran.
| | - Saghar Yousefnia
- Department of Cell and Molecular Biology, Semnan University, Semnan, Iran
| |
Collapse
|
7
|
Kommineni N, Chaudhari R, Conde J, Tamburaci S, Cecen B, Chandra P, Prasad R. Engineered Liposomes in Interventional Theranostics of Solid Tumors. ACS Biomater Sci Eng 2023; 9:4527-4557. [PMID: 37450683 DOI: 10.1021/acsbiomaterials.3c00510] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Engineered liposomal nanoparticles have unique characteristics as cargo carriers in cancer care and therapeutics. Liposomal theranostics have shown significant progress in preclinical and clinical cancer models in the past few years. Liposomal hybrid systems have not only been approved by the FDA but have also reached the market level. Nanosized liposomes are clinically proven systems for delivering multiple therapeutic as well as imaging agents to the target sites in (i) cancer theranostics of solid tumors, (ii) image-guided therapeutics, and (iii) combination therapeutic applications. The choice of diagnostics and therapeutics can intervene in the theranostics property of the engineered system. However, integrating imaging and therapeutics probes within lipid self-assembly "liposome" may compromise their overall theranostics performance. On the other hand, liposomal systems suffer from their fragile nature, site-selective tumor targeting, specific biodistribution and premature leakage of loaded cargo molecules before reaching the target site. Various engineering approaches, viz., grafting, conjugation, encapsulations, etc., have been investigated to overcome the aforementioned issues. It has been studied that surface-engineered liposomes demonstrate better tumor selectivity and improved therapeutic activity and retention in cells/or solid tumors. It should be noted that several other parameters like reproducibility, stability, smooth circulation, toxicity of vital organs, patient compliance, etc. must be addressed before using liposomal theranostics agents in solid tumors or clinical models. Herein, we have reviewed the importance and challenges of liposomal medicines in targeted cancer theranostics with their preclinical and clinical progress and a translational overview.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Center for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ruchita Chaudhari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa; Lisboa 1169-056, Portugal
| | - Sedef Tamburaci
- Department of Chemical Engineering, Izmir Institute of Technology, Gulbahce Campus, Izmir 35430, Turkey
| | - Berivan Cecen
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
8
|
Oloo SO, Smith KM, Vicente MDGH. Multi-Functional Boron-Delivery Agents for Boron Neutron Capture Therapy of Cancers. Cancers (Basel) 2023; 15:3277. [PMID: 37444386 DOI: 10.3390/cancers15133277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/03/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Boron neutron capture therapy (BNCT) is a binary cancer treatment that involves the irradiation of 10B-containing tumors with low-energy neutrons (thermal or epithermal). The alpha particles and recoiling Li nuclei that are produced in the 10B-capture nuclear reaction are high-linear-energy transfer particles that destroy boron-loaded tumor cells; therefore, BNCT has the potential to be a localized therapeutic modality. Two boron-delivery agents have been used in clinical trials of BNCT in patients with malignant brain tumors, cutaneous melanoma, or recurrent tumors of the head and neck region, demonstrating the potential of BNCT in the treatment of difficult cancers. A variety of potentially highly effective boron-delivery agents have been synthesized in the past four decades and tested in cells and animal models. These include boron-containing nucleosides, peptides, proteins, polyamines, porphyrins, liposomes, monoclonal antibodies, and nanoparticles of various types. The most promising agents are multi-functional boronated molecules and nanoparticles functionalized with tumor cell-targeting moieties that increase their tumor selectivity and contain a radiolabel or fluorophore to allow quantification of 10B-biodistribution and treatment planning. This review discusses multi-functional boron agents reported in the last decade, but their full potential can only be ascertained after their evaluation in BNCT clinical trials.
Collapse
Affiliation(s)
- Sebastian O Oloo
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Kevin M Smith
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | | |
Collapse
|
9
|
Nel J, Elkhoury K, Velot É, Bianchi A, Acherar S, Francius G, Tamayol A, Grandemange S, Arab-Tehrany E. Functionalized liposomes for targeted breast cancer drug delivery. Bioact Mater 2023; 24:401-437. [PMID: 36632508 PMCID: PMC9812688 DOI: 10.1016/j.bioactmat.2022.12.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/05/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Despite the exceptional progress in breast cancer pathogenesis, prognosis, diagnosis, and treatment strategies, it remains a prominent cause of female mortality worldwide. Additionally, although chemotherapies are effective, they are associated with critical limitations, most notably their lack of specificity resulting in systemic toxicity and the eventual development of multi-drug resistance (MDR) cancer cells. Liposomes have proven to be an invaluable drug delivery system but of the multitudes of liposomal systems developed every year only a few have been approved for clinical use, none of which employ active targeting. In this review, we summarize the most recent strategies in development for actively targeted liposomal drug delivery systems for surface, transmembrane and internal cell receptors, enzymes, direct cell targeting and dual-targeting of breast cancer and breast cancer-associated cells, e.g., cancer stem cells, cells associated with the tumor microenvironment, etc.
Collapse
Affiliation(s)
- Janske Nel
- Université de Lorraine, LIBio, F-54000, Nancy, France
| | | | - Émilie Velot
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Arnaud Bianchi
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Samir Acherar
- Université de Lorraine, CNRS, LCPM, F-54000, Nancy, France
| | | | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | | | | |
Collapse
|
10
|
Chang R, Fu R, Huang Y, Zhang J, Feng C, Wang R, Yan H, Li G, Chu X, Yuan F, Jia D, Li J. Codelivery of TRAIL and Mitomycin C via Liposomes Shows Improved Antitumor Effect on TRAIL-Resistant Tumors. Mol Pharm 2023. [PMID: 37134184 DOI: 10.1021/acs.molpharmaceut.2c01013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Although tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) constitutes a promising antitumor drug, tumor resistance to TRAIL has become a major obstacle in its clinical application. Mitomycin C (MMC) is an effective TRAIL-resistant tumor sensitizer, which indicates a potential utility of combination therapy. However, the efficacy of this combination therapy is limited owing to its short half-life and the cumulative toxicity of MMC. To address these issues, we successfully developed a multifunctional liposome (MTLPs) with human TRAIL protein on the surface and MMC encapsulated in the internal aqueous phase to codeliver TRAIL and MMC. MTLPs are uniform spherical particles that exhibit efficient cellular uptake by HT-29 TRAIL-resistant tumor cells, thereby inducing a stronger killing effect compared with control groups. In vivo assays revealed that MTLPs efficiently accumulated in tumors and safely achieved 97.8% tumor suppression via the synergistic effect of TRAIL and MMC in an HT-29 tumor xenograft model while ensuring biosafety. These results suggest that the liposomal codelivery of TRAIL and MMC provides a novel approach to overcome TRAIL-resistant tumors.
Collapse
Affiliation(s)
- Rui Chang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Rongrong Fu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Yujiao Huang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jibing Zhang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Changshun Feng
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Rui Wang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Hui Yan
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Guangyong Li
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Xiaohong Chu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng 252000, China
- School of Clinical Medicine, Shandong University, Jinan 250012, China
| | - Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| | - Jun Li
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
11
|
Matoori S. Vesicular Diagnostics: A Spotlight on Lactate- and Ammonia-Sensing Systems. ACS APPLIED BIO MATERIALS 2023; 6:1315-1322. [PMID: 36917016 DOI: 10.1021/acsabm.3c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Liposomes are a highly successful drug delivery system with over 15 FDA-approved formulations. Beyond delivering drugs, lipid and polymer vesicles have successfully been used for diagnostic applications. These applications range from more traditional uses, such as releasing diagnostic agents in a controlled manner, to leveraging the unique membrane properties to separate analytes and provide isolated reaction compartments in complex biological matrices. In this Spotlight on Applications, I highlight the complexities in the development and translation of diagnostic vesicles with two case studies, a liposomal reaction compartment for lactate sensing and a transmembrane pH-gradient polymersome for ammonia sensing.
Collapse
Affiliation(s)
- Simon Matoori
- Faculté de Pharmacie, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
12
|
Shan H, Sun X, Liu X, Sun Q, He Y, Chen Z, Lin Q, Jiang Z, Chen X, Chen Z, Zhao S. One-Step Formation of Targeted Liposomes in a Versatile Microfluidic Mixing Device. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205498. [PMID: 36449632 DOI: 10.1002/smll.202205498] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/28/2022] [Indexed: 06/17/2023]
Abstract
Targeted liposomes, as a promising carrier, have received tremendous attention in COVID-19 vaccines, molecular imaging, and cancer treatment, due to their enhanced cellular uptake and payload accumulation at target sites. However, the conventional methods for preparing targeted liposomes still suffer from limitations, including complex operation, time-consuming, and poor reproducibility. Herein, a facile and scalable strategy is developed for one-step construction of targeted liposomes using a versatile microfluidic mixing device (MMD). The engineered MMD provides an advanced synthesis platform for multifunctional liposome with high production rate and controllability. To validate the method, a programmed death-ligand 1 (PD-L1)-targeting aptamer modified indocyanine green (ICG)-liposome (Apt-ICG@Lip) is successfully constructed via the MMD. ICG and the PD-L1-targeting aptamer are used as model drug and targeting moiety, respectively. The Apt-ICG@Lip has high encapsulation efficiency (89.9 ± 1.4%) and small mean diameter (129.16 ± 5.48 nm). In vivo studies (PD-L1-expressing tumor models) show that Apt-ICG@Lip can realize PD-L1 targeted photoacoustic imaging, fluorescence imaging, and photothermal therapy. To verify the versatility of this approach, various targeted liposomes with different functions are further prepared and investigated. These experimental results demonstrate that this method is concise, efficient, and scalable to prepare multifunctional targeted liposomal nanoplatforms for molecular imaging and disease theranostics.
Collapse
Affiliation(s)
- Han Shan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Xin Sun
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Xin Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qi Sun
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yao He
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Ziyan Chen
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Qibo Lin
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Zixi Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
| | - Zeyu Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| |
Collapse
|
13
|
Zhou S, Li C, Yuan Y, Jiang L, Chen W, Jiang X. Dendritic lipopeptide liposomes decorated with dual-targeted proteins. Biomater Sci 2022; 10:7032-7041. [PMID: 36318065 DOI: 10.1039/d2bm00952h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to their homing effects, cell and cell membrane-derived nanocarriers have been widely used to enhance drug target delivery. Inspired by the protein-anchored cell membrane architecture, we here report a tumor-targeted liposome, dtDLP, which was constructed through the electrostatic interaction between dendritic lipopeptide liposomes and a dual-targeted recombinant protein, achieving superior tumor homing, cellular endocytotic and penetration abilities. The dual-targeted recombinant protein consists of an anti-epidermal growth factor receptor single domain antibody and a peptide ligand for the integrin αvβ3. dtDLPs substantially reduced macrophage phagocytosis and increased drug internalization in both 4T1 cells and HeLa cells by providing more endocytic pathways. In addition, the dtDLPs showed great penetration ability in both multicellular spheroids and tumor tissues. Due to the improved cancer cellular uptake and tumor penetration, the dtDLPs exhibited a superior anticancer effect in both HeLa and 4T1 tumor-bearing mice. This work will be helpful for the design of cell-specific liposomes with admirable tumor targeting, endocytotic and penetration abilities.
Collapse
Affiliation(s)
- Sensen Zhou
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Cheng Li
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Yang Yuan
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Lei Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China. .,State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| |
Collapse
|
14
|
Ying N, Lin X, Xie M, Zeng D. Effect of surface ligand modification on the properties of anti-tumor nanocarrier. Colloids Surf B Biointerfaces 2022; 220:112944. [DOI: 10.1016/j.colsurfb.2022.112944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/31/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022]
|
15
|
Kurihara Y, Yokota H, Takahashi M. Water-Dispersible Carboxymethyl Dextran-Coated Melamine Nanoparticles for Biosensing Applications. ACS OMEGA 2022; 7:41641-41650. [PMID: 36406549 PMCID: PMC9670359 DOI: 10.1021/acsomega.2c05653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
In this study, we developed a simple method for preparing highly dispersed, stable, and streptavidin (SA)-functionalized carboxymethyl dextran (CMD)-coated melamine nanoparticles (MNPs) in an aqueous buffer at neutral pH. Dynamic light scattering (DLS) revealed the agglomeration of MNPs in an aqueous buffer at neutral pH. When CMD, N-hydroxysuccinimide (NHS), and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) were simultaneously mixed with the MNPs, CMD was bound to the MNPs, promoting their dispersibility. Preparation of SA-CMD-MNPs was accomplished simply by adding SA solution to the CMD-MNPs. The amount of SA bound to the CMD-MNPs was quantified by the bicinchoninic assay, and the amount of SA molecules bound to each CMD-MNP was 417 ± 4. SA-CMD-MNPs exhibited high dispersity (polydispersity index = 0.058) in a neutral phosphate buffer and maintained it for 182 days with dispersion using a probe sonicator (5 s) before DLS characterization. The performance of the SA-CMD-MNPs in biosensing was evaluated by immunohistochemistry, which revealed that the nanoparticles could specifically stain MCF-7 cells derived from breast cancer cells with low HER2 expression. This study provides an effective method for synthesizing highly dispersible nanoparticles for biosensing.
Collapse
|
16
|
Cepero A, Luque C, Cabeza L, Perazzoli G, Quiñonero F, Mesas C, Melguizo C, Prados J. Antibody-Functionalized Nanoformulations for Targeted Therapy of Colorectal Cancer: A Systematic Review. Int J Nanomedicine 2022; 17:5065-5080. [PMID: 36345508 PMCID: PMC9635983 DOI: 10.2147/ijn.s368814] [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: 03/31/2022] [Accepted: 08/21/2022] [Indexed: 11/06/2022] Open
Abstract
The failure of chemotherapeutic treatment in colorectal cancer (CRC), the second most mortal cancer worldwide, is associated with several drug limitations, such as non-selective distribution, short half-life, and development of multiple resistances. One of the most promising strategies in CRC therapy is the development of delivery systems based on nanomaterials that can transport antitumor agents to the tumor site more efficiently, increasing accumulation within the tumor and thus the antitumor effect. In addition to taking advantage of the increased permeability and retention effect (EPR) of solid tumors, these nanoformulations can be conjugated with monoclonal antibodies that recognize molecular markers that are specifically over-expressed on CRC cells. Active targeting of nanoformulations reduces the adverse effects associated with the cytotoxic activity of drugs in healthy tissues, which will be of interest for improving the quality of life of cancer patients in the future. This review focuses on in vitro and in vivo studies of drug delivery nanoformulations functionalized with monoclonal antibodies for targeted therapy of CRC.
Collapse
Affiliation(s)
- Ana Cepero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Cristina Luque
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain,Correspondence: Consolación Melguizo, Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain, Tel +34-958-249833, Email
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| |
Collapse
|
17
|
Sarwar U, Naeem M, Nurjis F, Karim S, Raza A. Ultrasound-mediated in vivo biodistribution of coumarin-labeled sorafenib-loaded liposome-based nanotheranostic system. Nanomedicine (Lond) 2022; 17:1909-1927. [PMID: 36695214 DOI: 10.2217/nnm-2022-0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aim: This study aimed to synthesize folate-conjugated sorafenib-loaded (FCSL) liposomes for theranostic application using ultrasound (US). Materials & methods: US parameter optimization, in vitro release, anticancer effect, in vivo biodistribution, optical imaging and biocompatibility of liposomes were studied. Results: With 84% in vitro release after 4 min of US exposure at 3 MHz (1.2 mechanical index), FCSL liposomes showed lower IC50 (8.70 μM) versus sorafenib (9.34 μM) against HepG2 cells. In vivo biodistribution of FCSL liposomes versus sorafenib after 9 mg/kg injection in the liver (8.63 vs 0.55) > intestine (8.45 vs 1.07) > stomach (5.62 vs 0.57) > kidney (5.46 vs 0.91) showed longer circulation time in plasma and can be tracked in mice. Conclusion: A threefold higher drug concentration in the liver in US-exposed mice makes this a successful nanotheranostic approach.
Collapse
Affiliation(s)
- Usama Sarwar
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers & Optronics College (NILOP-C), Pakistan Institute of Engineering & Applied Sciences, Nilore, Islamabad, 45650, Pakistan.,Department of Biotechnology, Medical Genetics Research Laboratory, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Naeem
- Department of Biotechnology, Medical Genetics Research Laboratory, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Farwa Nurjis
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers & Optronics College (NILOP-C), Pakistan Institute of Engineering & Applied Sciences, Nilore, Islamabad, 45650, Pakistan
| | - Shafqat Karim
- Nano Materials Research Group, Pakistan Institute of Nuclear Science & Technology (PINSTECH), Nilore, Islamabad, 45650, Pakistan
| | - Abida Raza
- National Center of Industrial Biotechnology, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, 46000, Pakistan
| |
Collapse
|
18
|
Zhao Z, Swartchick CB, Chan J. Targeted contrast agents and activatable probes for photoacoustic imaging of cancer. Chem Soc Rev 2022; 51:829-868. [PMID: 35094040 PMCID: PMC9549347 DOI: 10.1039/d0cs00771d] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Photoacoustic (PA) imaging has emerged as a powerful technique for the high resolution visualization of biological processes within deep tissue. Through the development and application of exogenous targeted contrast agents and activatable probes that can respond to a given cancer biomarker, researchers can image molecular events in vivo during cancer progression. This information can provide valuable details that can facilitate cancer diagnosis and therapy monitoring. In this tutorial review, we provide a step-by-step guide to select a cancer biomarker and subsequent approaches to design imaging agents for in vivo use. We envision this information will be a useful summary to those in the field, new members to the community, and graduate students taking advanced imaging coursework. We also highlight notable examples from the recent literature, with emphasis on the molecular designs and their in vivo PA imaging performance. To conclude, we provide our outlook and future perspective in this exciting field.
Collapse
Affiliation(s)
- Zhenxiang Zhao
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, USA.
| | - Chelsea B Swartchick
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, USA.
| | - Jefferson Chan
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, USA.
| |
Collapse
|
19
|
Thakur R, Suri CR, Kaur IP, Rishi P. Review. Crit Rev Ther Drug Carrier Syst 2022; 40:49-100. [DOI: 10.1615/critrevtherdrugcarriersyst.2022040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Khurana N, Yathavan B, Jedrzkiewicz J, Gill AS, Pulsipher A, Alt JA, Ghandehari H. Vascular permeability in chronic rhinosinusitis enhances accumulation and retention of nanoscale pegylated liposomes. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 38:102453. [PMID: 34363985 PMCID: PMC10499165 DOI: 10.1016/j.nano.2021.102453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 11/17/2022]
Abstract
Chronic rhinosinusitis (CRS) is a debilitating inflammatory disorder of the sinonasal mucosa that substantially diminishes patient quality of life. Progress surrounding management of this disease has been crippled by a lack of therapeutic innovation. It has been posited that increased vascularity within the diseased sinuses of patients with CRS may allow for improved systemic drug delivery via nanoscale liposomal carriers. Such a system could enhance drug distribution, accumulation, and retention within the sinuses, ultimately leading to improved patient outcomes. PEGylated liposomes loaded with indocyanine green (ICG) were synthesized, characterized and systemically administered in a mouse model of CRS. Accumulation and retention of ICG in sinonasal tissue were evaluated. Compared to healthy controls, CRS mice showed significant sinonasal tissue accumulation and retention of PEGylated liposomal ICG for up to 21 days (P < 0.001). Conversely, free ICG was eliminated from the body after 24 h in both groups.
Collapse
Affiliation(s)
- Nitish Khurana
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA
| | - Bhuvanesh Yathavan
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA
| | | | - Amarbir S Gill
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Abigail Pulsipher
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jeremiah A Alt
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| | - Hamidreza Ghandehari
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
21
|
Nitheesh Y, Pradhan R, Hejmady S, Taliyan R, Singhvi G, Alexander A, Kesharwani P, Dubey SK. Surface engineered nanocarriers for the management of breast cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112441. [PMID: 34702526 DOI: 10.1016/j.msec.2021.112441] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022]
Abstract
Breast cancer is commonly known life-threatening malignancy in women after lung cancer. The standard of care (SOC) treatment for breast cancer primarily includes surgery, radiotherapy, hormonal therapy, and chemotherapy. However, the effectiveness of conventional chemotherapy is restricted by several limitations such as poor targeting, drug resistance, poor drug delivery, and high toxicity. Nanoparticulate drug delivery systems have gained a lot of interest in the scientific community because of its unique features and promising potential in breast cancer diagnosis and treatment. The unique physicochemical and biological properties of the nanoparticulate drug delivery systems promotes the drug accumulation, Pharmacokinetic profile towards the tumor site and thereby, reduces the cytotoxicity towards healthy cells. In addition, to improve tumor-specific drug delivery, researchers have focused on surface engineered nanocarrier system with targeting molecules/ligands that are specific to overexpressed receptors present on cancer cells. In this review, we have summarized the different biological ligands and surface-engineered nanoparticles, enlightening the physicochemical characteristics, toxic effects, and regulatory considerations of nanoparticles involved in treatment of breast cancer.
Collapse
Affiliation(s)
- Yanamandala Nitheesh
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Rajesh Pradhan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Siddhant Hejmady
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Amit Alexander
- National Institute of Pharmaceutical Education and Research (NIPER-G), Ministry of Chemicals & Fertilizers, Govt. of India NH 37, NITS Mirza, Kamrup-781125, Guwahati, Assam, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Sunil Kumar Dubey
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia 700056, Kolkata, India.
| |
Collapse
|
22
|
Clinically translatable quantitative molecular photoacoustic imaging with liposome-encapsulated ICG J-aggregates. Nat Commun 2021; 12:5410. [PMID: 34518530 PMCID: PMC8438038 DOI: 10.1038/s41467-021-25452-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/11/2021] [Indexed: 02/08/2023] Open
Abstract
Photoacoustic (PA) imaging is a functional and molecular imaging technique capable of high sensitivity and spatiotemporal resolution at depth. Widespread use of PA imaging, however, is limited by currently available contrast agents, which either lack PA-signal-generation ability for deep imaging or their absorbance spectra overlap with hemoglobin, reducing sensitivity. Here we report on a PA contrast agent based on targeted liposomes loaded with J-aggregated indocyanine green (ICG) dye (i.e., PAtrace) that we synthesized, bioconjugated, and characterized to addresses these limitations. We then validated PAtrace in phantom, in vitro, and in vivo PA imaging environments for both spectral unmixing accuracy and targeting efficacy in a folate receptor alpha-positive ovarian cancer model. These study results show that PAtrace concurrently provides significantly improved contrast-agent quantification/sensitivity and SO2 estimation accuracy compared to monomeric ICG. PAtrace's performance attributes and composition of FDA-approved components make it a promising agent for future clinical molecular PA imaging.
Collapse
|
23
|
Abbasi H, Rahbar N, Kouchak M, Khalil Dezfuli P, Handali S. Functionalized liposomes as drug nanocarriers for active targeted cancer therapy: a systematic review. J Liposome Res 2021; 32:195-210. [PMID: 33729077 DOI: 10.1080/08982104.2021.1903035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a broad term used to describe a group of diseases that have more than 270 types. Today, due to the suffering of patients from the side effects of existing methods in the treatment of cancer such as chemotherapy and radiotherapy, the employment of targeted methods in the treatment of this disease has been received much consideration. In recent years, nanoparticles have revolutionized in the treatment of many diseases such as cancer. Among these nanoparticles, liposomes are more considerable. Active targeted liposomes show an important role in the selective action of the drug on cancer cells. Until now, a variety of anti-cancer agents have been reported for targeted delivery to cancer cells using liposomes. The results of in vitro and studies in vivo have been shown that selective action of the targeted liposomes is increased with reduced side effects and toxicity compared with free drugs or non-targeted liposomes. This systematic review expresses the reports of this type of drug delivery system. Search terms were searched through several online databases including PubMed, Scopus, and Science Direct from 1990 to 2019 and the quality evaluation was performed. Out of 11,676 published articles, 196 articles met the inclusion criteria. The current report reviews developments in the liposomes targeted with aptamer, transferrin, folate, and monoclonal antibodies.
Collapse
Affiliation(s)
- Hanieh Abbasi
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parna Khalil Dezfuli
- School of Pharmacy Library, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Handali
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Oladipo AO, Unuofin JO, Iku SII, Nkambule TTI, Mamba BB, Msagati TAM. Bimetallic Au@Pd nanodendrite system incorporating multimodal intracellular imaging for improved doxorubicin antitumor efficiency. Int J Pharm 2021; 602:120661. [PMID: 33933638 DOI: 10.1016/j.ijpharm.2021.120661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/19/2022]
Abstract
The sufficient accumulation of drugs is crucial for efficient treatment in a complex tumor microenvironment. Drug delivery systems (DDS) with high surface area and selective cytotoxicity present a novel approach to mitigate insufficient drug loading for improved therapeutic response. Herein, a doxorubicin-conjugated bimetallic gold-core palladium-shell nanocarrier with multiple dense arrays of branches (Au@PdNDs.PEG/DOX) was characterized and its efficacy against breast adenocarcinoma (MCF-7) and lung adenocarcinoma (A549) cells were evaluated. Enhanced darkfield and hyperspectral imaging (HSI) microscopy were used to study the intracellular uptake and accumulation of the DOX-loaded nanodendrites A fascinating data from a 3D-CytoViva fluorescence imaging technique provided information about the dynamics of localization and distribution of the nanocarrier. In vitro cytotoxicity assays indicated that Au@PdNDs.PEG/DOX inhibited the proliferative effects of MCF-7 cells at equivalent IC50 dosage compared to DOX alone. The nanocarrier triggered higher induction of apoptosis proved by a time-dependent phosphatidylserine V release, cell cycle arrest, and flow cytometry analysis. Moreover, the cell cycle phase proportion increase suggests that the enhanced apoptotic effect induced by Au@PdNDs.PEG/DOX was via a G2/M phase arrest. Thus, this study demonstrated the potential of dendritic nanoparticles to improve DOX therapeutic efficiency and plasmonic-mediated intracellular imaging as a suitable theranostic platform for deployment in nanomedicine.
Collapse
Affiliation(s)
- Adewale O Oladipo
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa.
| | - Jeremiah O Unuofin
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Solange I I Iku
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Thabo T I Nkambule
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Bhekie B Mamba
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Titus A M Msagati
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa.
| |
Collapse
|
25
|
Zhuo S, Zhang F, Yu J, Zhang X, Yang G, Liu X. pH-Sensitive Biomaterials for Drug Delivery. Molecules 2020; 25:E5649. [PMID: 33266162 PMCID: PMC7730929 DOI: 10.3390/molecules25235649] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
The development of precise and personalized medicine requires novel formulation strategies to deliver the therapeutic payloads to the pathological tissues, producing enhanced therapeutic outcome and reduced side effects. As many diseased tissues are feathered with acidic characteristics microenvironment, pH-sensitive biomaterials for drug delivery present great promise for the purpose, which could protect the therapeutic payloads from metabolism and degradation during in vivo circulation and exhibit responsive release of the therapeutics triggered by the acidic pathological tissues, especially for cancer treatment. In the past decades, many methodologies, such as acidic cleavage linkage, have been applied for fabrication of pH-responsive materials for both in vitro and in vivo applications. In this review, we will summarize some pH-sensitive drug delivery system for medical application, mainly focusing on the pH-sensitive linkage bonds and pH-sensitive biomaterials.
Collapse
Affiliation(s)
- Shijie Zhuo
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.Z.); (F.Z.); (J.Y.)
| | - Feng Zhang
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.Z.); (F.Z.); (J.Y.)
| | - Junyu Yu
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.Z.); (F.Z.); (J.Y.)
| | - Xican Zhang
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.Z.); (F.Z.); (J.Y.)
| | - Guangbao Yang
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China;
| | - Xiaowen Liu
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.Z.); (F.Z.); (J.Y.)
| |
Collapse
|
26
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
27
|
Heshmati Aghda N, Abdulsahib SM, Severson C, Lara EJ, Torres Hurtado S, Yildiz T, Castillo JA, Tunnell JW, Betancourt T. Induction of immunogenic cell death of cancer cells through nanoparticle-mediated dual chemotherapy and photothermal therapy. Int J Pharm 2020; 589:119787. [PMID: 32898630 DOI: 10.1016/j.ijpharm.2020.119787] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/11/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
The use of nanomedicines to induce immunogenic cell death is a new strategy that aims to increase tumor immunogenicity and thereby prime tumors for further immunotherapies. In this study, we developed a nanoparticle formulation for combinatory chemotherapy and photothermal therapy based only on materials previously used in FDA-approved products and investigated the effect of the combinatory therapy on the growth inhibition and induction of immunogenic cell death in human MDA-MB-231 breast cancer cells. The formulation consists of ~108-nm nanoparticles made of poly(lactic acid)-b-methoxy poly(ethylene glycol) which carry doxorubicin for chemotherapy and indocyanine green for photothermal therapy. A 0.3 mg/mL suspension of NPs increased the medium temperature up to 10 °C upon irradiation with an 808-nm diode laser. In vitro studies showed that combination of laser assisted indocyanine green-mediated photothermal therapy and doxorubicin-mediated chemotherapy effectively eradicated cancer cells and resulted in the highest level of damage-associated molecular pattern presentation (calreticulin, high mobility group box 1, and adenosine triphosphate) compared to the individual treatments alone. These results demonstrate that our nanoparticle-mediated combinatory approach led to the most intense immunogenic cell death when compared to individual chemotherapy or photothermal therapy, making it a potent option for future in vivo studies in combination with cancer immunotherapies.
Collapse
Affiliation(s)
- Niloofar Heshmati Aghda
- Materials Science, Engineering and Commercialization Program, Texas State University, San Marcos, TX, USA
| | - Shahad M Abdulsahib
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Carli Severson
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Emilio J Lara
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Susana Torres Hurtado
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Tugba Yildiz
- Materials Science, Engineering and Commercialization Program, Texas State University, San Marcos, TX, USA
| | - Juan A Castillo
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - James W Tunnell
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Tania Betancourt
- Materials Science, Engineering and Commercialization Program, Texas State University, San Marcos, TX, USA; Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA.
| |
Collapse
|
28
|
Cheng YA, Wu TH, Wang YM, Cheng TL, Chen IJ, Lu YC, Chuang KH, Wang CK, Chen CY, Lin RA, Chen HJ, Liao TY, Liu ES, Chen FM. Humanized bispecific antibody (mPEG × HER2) rapidly confers PEGylated nanoparticles tumor specificity for multimodality imaging in breast cancer. J Nanobiotechnology 2020; 18:118. [PMID: 32854720 PMCID: PMC7457265 DOI: 10.1186/s12951-020-00680-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Developing a universal strategy to improve the specificity and sensitivity of PEGylated nanoaparticles (PEG-NPs) for assisting in the diagnosis of tumors is important in multimodality imaging. Here, we developed the anti-methoxypolyethylene glycol (mPEG) bispecific antibody (BsAb; mPEG × HER2), which has dual specificity for mPEG and human epidermal growth factor receptor 2 (HER2), with a diverse array of PEG-NPs to confer nanoparticles with HER2 specificity and stronger intensity. Result We used a one-step formulation to rapidly modify the nanoprobes with mPEG × HER2 and optimized the modified ratio of BsAbs on several PEG-NPs (Lipo-DiR, SPIO, Qdot and AuNP). The αHER2/PEG-NPs could specifically target MCF7/HER2 cells (HER2++) but not MCF7/neo1 cells (HER2+/−). The αHER2/Lipo-DiR and αHER2/SPIO could enhance the sensitivity of untargeted PEG-NPs on MCF7/HER2 (HER2++). In in vivo imaging, αHER2/Lipo-DiR and αHER2/SPIO increased the specific targeting and enhanced PEG-NPs accumulation at 175% and 187% on 24 h, respectively, in HER2-overexpressing tumors. Conclusion mPEG × HER2, therefore, provided a simple one-step formulation to confer HER2-specific targeting and enhanced sensitivity and contrast intensity on HER2 positive tumors for multimodality imaging. ![]()
Collapse
Affiliation(s)
- Yi-An Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Tung-Ho Wu
- Cardiovascular Division of Surgical Department, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd, Zuoying Dist, Kaohsiung, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, No. 1001, University Road, Hsinchu, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - I-Ju Chen
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Yun-Chi Lu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, No.172-1, Sec. 2, Keelung Rd, Taipei, Taiwan
| | - Chih-Kuang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Chiao-Yun Chen
- Department of Radiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan
| | - Rui-An Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Huei-Jen Chen
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Tzu-Yi Liao
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Fang-Ming Chen
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan. .,Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, No.68, Jhonghua 3rd Rd, Cianjin District, Kaohsiung, Taiwan. .,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan. .,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.
| |
Collapse
|
29
|
Near-infrared phototherapy for patient-derived orthotopic xenograft model of hepatocellular carcinoma in combination with indocyanine green. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 209:111938. [PMID: 32590285 DOI: 10.1016/j.jphotobiol.2020.111938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Hepatocellular carcinoma notably takes up and retains indocyanine green (ICG). Here, we investigated whether patient-derived orthotopic xenograft of hepatocellular carcinoma could accumulate ICG and show full remission via phototherapy. METHODS NIR light and ICG were tested for cytotoxicity in cancerous cell lines (Huh-7, Hep3B). Patient-derived orthotopic xenograft (PDoX) mice were subjected to phototherapy comprising of daily NIR exposure (0.5-1.75 W/cm2) and intravenous injection of ICG (5-20 mg/kg2). Moreover, NIR laser was flashed on individual mouse until hepatocellular carcinoma completely loss the fluorescence, as determined by NIR camera. RESULTS Cytotoxicity increased in response to the input energy, but insufficient energy (< 150 joule/cm2) was irresponsive at all irradiances. NIR irradiance in the range of 0.5-1.75 W/cm2 took 5-7 days to elicit complete remission from PDoX mice in combination with 20 mg/kg ICG. In contrast, phototherapy could completely ablate hepatocellular carcinoma at 5-15 mg/kg ICG. CONCLUSIONS ICG could potentiate the tumoricidal ability of NIR light in a dose-dependent manner, and vice versa. Regardless of ICG dosage, however, phototherapy treated group showed a relatively high survival rate compared to the non-treated group. Notably, real-time phototherapy could halve the effective ICG dosage for full remission of deep-seated tumor.
Collapse
|
30
|
Xu D, Li L, Chu C, Zhang X, Liu G. Advances and perspectives in near-infrared fluorescent organic probes for surgical oncology. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1635. [PMID: 32297455 DOI: 10.1002/wnan.1635] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/25/2020] [Accepted: 03/19/2020] [Indexed: 12/11/2022]
Abstract
Surgical resection of solid tumors is currently the most efficient and preferred therapeutic strategy for treating cancer. Despite significant medical, technical, and scientific advances, the complete treatment of this lethal disease is still a challenging task. New imaging techniques and contrast agents are urgently needed to improve cytoreductive surgery and patient outcomes. Tumor-targeted probes are valuable for guiding a surgical resection of tumor from subjective judgments to visual inspection. Near-infrared (NIR) fluorescent imaging is a promising technology in preclinical and clinical tumor diagnosis and therapy. The rapid development in NIR fluorophores with improved optical properties, targeting strategies, and imaging devices has brought about prospective study of novel NIR nanomaterials for intraoperative tumor detection. In this review, we summarize the recent development in NIR-emitting organic fluorophores and cancer-targeting strategies that specifically target and accumulate in tumors for the molecular imaging of cancerous cells. We believe this technique utilizing new fluorescent probes with an intraoperative optical imaging capacity could provide a more sensitive and accurate method for cancer resection guidance, thereby resulting in better surgical outcomes. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Dazhuang Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China.,Department of Chemistry, Nanchang University, Nanchang, China
| | - Lei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Chengchao Chu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xiaoyong Zhang
- Department of Chemistry, Nanchang University, Nanchang, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
31
|
Abstract
This review highlights the pharmacokinetic features and tumor imaging preponderance of renal clearable AuNCs for in vivo tumor imaging.
Collapse
Affiliation(s)
- Huili Li
- Engineering Research Center of Cell and Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiaotong University
- Shanghai 200240
| | - Hongle Li
- Department of Molecular Pathology
- The Affiliated Cancer Hospital
- Zhengzhou University
- Zhengzhou
- China
| | - Ajun Wan
- National Engineering Research Center of Protected Agriculture
- School of Medicine
- Tongji University
- Shanghai 200092
- China
| |
Collapse
|
32
|
Jaymand M. Chemically Modified Natural Polymer-Based Theranostic Nanomedicines: Are They the Golden Gate toward a de Novo Clinical Approach against Cancer? ACS Biomater Sci Eng 2019; 6:134-166. [DOI: 10.1021/acsbiomaterials.9b00802] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| |
Collapse
|
33
|
Al-Ahmady ZS, Donno R, Gennari A, Prestat E, Marotta R, Mironov A, Newman L, Lawrence MJ, Tirelli N, Ashford M, Kostarelos K. Enhanced Intraliposomal Metallic Nanoparticle Payload Capacity Using Microfluidic-Assisted Self-Assembly. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:13318-13331. [PMID: 31478662 DOI: 10.1021/acs.langmuir.9b00579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Hybrids composed of liposomes (L) and metallic nanoparticles (NPs) hold great potential for imaging and drug delivery purposes. However, the efficient incorporation of metallic NPs into liposomes using conventional methodologies has so far proved to be challenging. In this study, we report the fabrication of hybrids of liposomes and hydrophobic gold NPs of size 2-4 nm (Au) using a microfluidic-assisted self-assembly process. The incorporation of increasing amounts of AuNPs into liposomes was examined using microfluidics and compared to L-AuNP hybrids prepared by the reverse-phase evaporation method. Our microfluidics strategy produced L-AuNP hybrids with a homogeneous size distribution, a smaller polydispersity index, and a threefold increase in loading efficiency when compared to those hybrids prepared using the reverse-phase method of production. Quantification of the loading efficiency was determined by ultraviolet spectroscopy, inductively coupled plasma mass spectroscopy, and centrifugal field flow fractionation, and qualitative validation was confirmed by transmission electron microscopy. The higher loading of gold NPs into the liposomes achieved using microfluidics produced a slightly thicker and more rigid bilayer as determined with small-angle neutron scattering. These observations were confirmed using fluorescent anisotropy and atomic force microscopy. Structural characterization of the liposomal-NP hybrids with cryo-electron microscopy revealed the coexistence of membrane-embedded and interdigitated NP-rich domains, suggesting AuNP incorporation through hydrophobic interactions. The microfluidic technique that we describe in this study allows for the automated production of monodisperse liposomal-NP hybrids with high loading capacity, highlighting the utility of microfluidics to improve the payload of metallic NPs within liposomes, thereby enhancing their application for imaging and drug delivery.
Collapse
Affiliation(s)
- Zahraa S Al-Ahmady
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health , University of Manchester , Av Hill Building , Manchester M13 9PT , U.K
- Pharmacology Department, School of Science and Technology , Nottingham Trent University , Nottingham NG11 8NS , U.K
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
| | - Roberto Donno
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
- Laboratory of Polymers and Biomaterials , Fondazione Istituto Italiano di Tecnologia , 16163 , Genova , Italy
| | - Arianna Gennari
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
- Laboratory of Polymers and Biomaterials , Fondazione Istituto Italiano di Tecnologia , 16163 , Genova , Italy
| | - Eric Prestat
- SuperSTEM Laboratory , SciTech Daresbury Campus , Keckwick Lane, Warrington WA4 4AD , U.K
| | - Roberto Marotta
- Electron Microscopy Laboratory , Fondazione Istituto Italiano di Tecnologia , 16163 Genova , Italy
| | | | - Leon Newman
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health , University of Manchester , Av Hill Building , Manchester M13 9PT , U.K
| | - M Jayne Lawrence
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
| | - Nicola Tirelli
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
- Laboratory of Polymers and Biomaterials , Fondazione Istituto Italiano di Tecnologia , 16163 , Genova , Italy
| | - Marianne Ashford
- Advanced Drug Delivery Pharmaceutical Sciences, IMED Biotech Unit , AstraZeneca , Macclesfield SK10 2NA , U.K
| | - Kostas Kostarelos
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health , University of Manchester , Av Hill Building , Manchester M13 9PT , U.K
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
| |
Collapse
|
34
|
Miranda D, Wan C, Kilian HI, Mabrouk MT, Zhou Y, Jin H, Lovell JF. Indocyanine green binds to DOTAP liposomes for enhanced optical properties and tumor photoablation. Biomater Sci 2019; 7:3158-3164. [PMID: 31232421 PMCID: PMC6650340 DOI: 10.1039/c9bm00551j] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Indocyanine green (ICG) is a clinically-approved near infrared (NIR) dye used for optical imaging. The dye is only slightly soluble in water and is prone to aggregation in saline solutions, so that alternative formulations can improve photophysical performance. Numerous nanoscale formulations of ICG have been described in the literature, but we sought to develop an approach that does not require additional purification steps. Pre-formed liposomes incorporating 45 mol% of the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) rapidly bind ICG, resulting in enhanced NIR optical properties. ICG binding is dependent on the amount of DOTAP incorporated in the liposomes. A dye-to-lipid mass ratio of [0.5 : 25] is sufficient for full complexation, without additional purification steps following mixing. NIR absorption, fluorescence intensity, and photoacoustic signals are increased for the liposome-bound dye. Not only is the optical character enhanced by simple mixing of ICG with liposomes, but retention in 4T1 mammary tumors is observed following intratumor injection, as assessed by fluorescence and photoacoustic imaging. Subsequent photothermal therapy with 808 nm laser irradiation is effective and results in tumor ablation without regrowth for at least 30 days. Thus, ICG optical properties and photothermal ablation outcomes can be improved by mixing the dye with pre-formed DOTAP liposomes in conditions that result in full dye-binding to the liposomes.
Collapse
Affiliation(s)
- Dyego Miranda
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Hailey I Kilian
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Moustafa T Mabrouk
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Yuhan Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
35
|
Abstract
Fuelled by innovation, optical microscopy plays a critical role in the life sciences and medicine, from basic discovery to clinical diagnostics. However, optical microscopy is limited by typical penetration depths of a few hundred micrometres for in vivo interrogations in the visible spectrum. Optoacoustic microscopy complements optical microscopy by imaging the absorption of light, but it is similarly limited by penetration depth. In this Review, we summarize progress in the development and applicability of optoacoustic mesoscopy (OPAM); that is, optoacoustic imaging with acoustic resolution and wide-bandwidth ultrasound detection. OPAM extends the capabilities of optical imaging beyond the depths accessible to optical and optoacoustic microscopy, and thus enables new applications. We explain the operational principles of OPAM, its placement as a bridge between optoacoustic microscopy and optoacoustic macroscopy, and its performance in the label-free visualization of tissue pathophysiology, such as inflammation, oxygenation, vascularization and angiogenesis. We also review emerging applications of OPAM in clinical and biological imaging.
Collapse
|
36
|
Targeting Ovarian Cancer Cells Overexpressing CD44 with Immunoliposomes Encapsulating Glycosylated Paclitaxel. Int J Mol Sci 2019; 20:ijms20051042. [PMID: 30818864 PMCID: PMC6429518 DOI: 10.3390/ijms20051042] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 01/06/2023] Open
Abstract
Paclitaxel (PTX) is one of the front-line drugs approved for the treatment of ovarian cancer. However, the application of PTX is limited due to the significant hydrophobicity and poor pharmacokinetics. We previously reported target-directed liposomes carrying tumor-selective conjugated antibody and encapsulated glycosylated PTX (gPTX-L) which successfully overcome the PTX limitation. The tubulin stabilizing activity of gPTX was equivalent to that of PTX while the cytotoxic activity of gPTX was reduced. In human ovarian cancer cell lines, SK-OV-3 and OVK18, the concentration at which cell growth was inhibited by 50% (IC50) for gPTX range from 15–20 nM, which was sensitive enough to address gPTX-L with tumor-selective antibody coupling for ovarian cancer therapy. The cell membrane receptor CD44 is associated with cancer progression and has been recognized as a cancer stem cell marker including ovarian cancer, becoming a suitable candidate to be targeted by gPTX-L therapy. In this study, gPTX-loading liposomes conjugated with anti-CD44 antibody (gPTX-IL) were assessed for the efficacy of targeting CD44-positive ovarian cancer cells. We successfully encapsulated gPTX into liposomes with the loading efficiency (LE) more than 80% in both of gPTX-L and gPTX-IL with a diameter of approximately 100 nm with efficacy of enhanced cytotoxicity in vitro and of convenient treatment in vivo. As the result, gPTX-IL efficiently suppressed tumor growth in vivo. Therefore gPTX-IL could be a promising formulation for effective ovarian cancer therapies.
Collapse
|
37
|
Silva CO, Pinho JO, Lopes JM, Almeida AJ, Gaspar MM, Reis C. Current Trends in Cancer Nanotheranostics: Metallic, Polymeric, and Lipid-Based Systems. Pharmaceutics 2019; 11:E22. [PMID: 30625999 PMCID: PMC6359642 DOI: 10.3390/pharmaceutics11010022] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/28/2018] [Accepted: 01/01/2019] [Indexed: 02/07/2023] Open
Abstract
Theranostics has emerged in recent years to provide an efficient and safer alternative in cancer management. This review presents an updated description of nanotheranostic formulations under development for skin cancer (including melanoma), head and neck, thyroid, breast, gynecologic, prostate, and colon cancers, brain-related cancer, and hepatocellular carcinoma. With this focus, we appraised the clinical advantages and drawbacks of metallic, polymeric, and lipid-based nanosystems, such as low invasiveness, low toxicity to the surrounding healthy tissues, high precision, deeper tissue penetration, and dosage adjustment in a real-time setting. Particularly recognizing the increased complexity and multimodality in this area, multifunctional hybrid nanoparticles, comprising different nanomaterials and functionalized with targeting moieties and/or anticancer drugs, present the best characteristics for theranostics. Several examples, focusing on their design, composition, imaging and treatment modalities, and in vitro and in vivo characterization, are detailed herein. Briefly, all studies followed a common trend in the design of these theranostics modalities, such as the use of materials and/or drugs that share both inherent imaging (e.g., contrast agents) and therapeutic properties (e.g., heating or production reactive oxygen species). This rationale allows one to apparently overcome the heterogeneity, complexity, and harsh conditions of tumor microenvironments, leading to the development of successful targeted therapies.
Collapse
Affiliation(s)
- Catarina Oliveira Silva
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Jacinta Oliveira Pinho
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Joana Margarida Lopes
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - António J Almeida
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Catarina Reis
- iMedUlisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
- IBEB, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| |
Collapse
|
38
|
Neuberger K, Boddupalli A, Bratlie KM. Effects of arginine-based surface modifications of liposomes for drug delivery in Caco-2 colon carcinoma cells. Biochem Eng J 2018. [DOI: 10.1016/j.bej.2018.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Yang H, Liu Z, Li X, Zhang Z, Chen D, Lian H. Artesunate-Loaded and Near-Infrared Dye-Conjugated Albumin Nanoparticles as High-Efficiency Tumor-Targeted Photo-Chemo Theranostic Agent. NANOSCALE RESEARCH LETTERS 2018; 13:319. [PMID: 30311065 PMCID: PMC6181830 DOI: 10.1186/s11671-018-2700-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/30/2018] [Indexed: 05/20/2023]
Abstract
Herein, a tumor-targeted multifunctional theranostic agent was synthetized using a facile method, combining four clinically approved materials: artesunate (Arte), human serum albumin (HSA), folic acid (FA), and indocyanine green (ICG). The obtained nanocomposites (FA-IHA NPs) showed an excellent photo- and physiological stability. The ICG in the FA-IHA NPs was used not only for near infrared (NIR) fluorescence imaging, but also for photothermal and photodynamic (PTT-PDT) therapy under a single NIR irradiation. In addition, the NIR irradiation (808 nm, 1 W/cm2) could trigger Arte release that showed enhanced chemotherapeutic effect. Through fluorescence imaging, the cell uptake and tumor accumulation of FA-IHA NPs were observed in vitro and in vivo, analyzed by confocal microscopy and NIR fluorescence imaging in tumor xenograft mice. Based on the diagnostic results, FA-IHA NPs at 24 h post injection and combined with NIR irradiation (808 nm, 1 W/cm2) could efficiently suppress tumor growth through a photo-chemo combination therapy, with no tumor recurrence in vitro and in vivo. The obtained results suggested that FA-IHA NPs are promising photo-chemo theranostic agents for future clinical translation.
Collapse
Affiliation(s)
- Hainan Yang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Zaijia Liu
- Department of Medical Image, Ezhou Central Hospital, Ezhou, 436000 China
| | - Xufeng Li
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Zhenfeng Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Deji Chen
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Hui Lian
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| |
Collapse
|
40
|
Wei Y, Quan L, Zhou C, Zhan Q. Factors relating to the biodistribution & clearance of nanoparticles & their effects on in vivo application. Nanomedicine (Lond) 2018; 13:1495-1512. [DOI: 10.2217/nnm-2018-0040] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles have promising biomedical applications for drug delivery, tumor imaging and tumor treatment. Pharmacokinetics are important for the in vivo application of nanoparticles. Biodistribution and clearance are largely defined as the key points of pharmacokinetics to maximize therapeutic efficacy and to minimize side effects. Different engineered nanoparticles have different biodistribution and clearance processes. The interactions of organs with nanoparticles, which are determined by the characteristics of the organs and the biochemical/physical properties of the nanoparticles, are a major factor influencing biodistribution and clearance. In this review, the clearance functions of organs and the properties related to pharmacokinetics, including nanoparticle size, shape, biodegradation and surface modifications are discussed.
Collapse
Affiliation(s)
- Yanchun Wei
- Provincial Key Laboratory for Interventional Medical Devices, Huaiyin Institute of Technology, Huai'an, Jiangsu 223001, PR China
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
| | - Li Quan
- Provincial Key Laboratory for Interventional Medical Devices, Huaiyin Institute of Technology, Huai'an, Jiangsu 223001, PR China
| | - Chao Zhou
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
| | - Qiuqiang Zhan
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
- Key Laboratory of Optoelectronic Devices & Systems of Ministry of Education & Guangdong Province, Shenzhen University, Shenzhen 518052, PR China
| |
Collapse
|
41
|
Zhao J, Chen J, Ma S, Liu Q, Huang L, Chen X, Lou K, Wang W. Recent developments in multimodality fluorescence imaging probes. Acta Pharm Sin B 2018; 8:320-338. [PMID: 29881672 PMCID: PMC5989919 DOI: 10.1016/j.apsb.2018.03.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Multimodality optical imaging probes have emerged as powerful tools that improve detection sensitivity and accuracy, important in disease diagnosis and treatment. In this review, we focus on recent developments of optical fluorescence imaging (OFI) probe integration with other imaging modalities such as X-ray computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), single-photon emission computed tomography (SPECT), and photoacoustic imaging (PAI). The imaging technologies are briefly described in order to introduce the strengths and limitations of each techniques and the need for further multimodality optical imaging probe development. The emphasis of this account is placed on how design strategies are currently implemented to afford physicochemically and biologically compatible multimodality optical fluorescence imaging probes. We also present studies that overcame intrinsic disadvantages of each imaging technique by multimodality approach with improved detection sensitivity and accuracy.
Collapse
Affiliation(s)
- Jianhong Zhao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Junwei Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Shengnan Ma
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Qianqian Liu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Lixian Huang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Xiani Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyan Lou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131-0001, USA
| |
Collapse
|
42
|
Xie Z, Guo W, Guo N, Huangfu M, Liu H, Lin M, Xu W, Chen J, Wang T, Wei Q, Han M, Gao J. Targeting tumor hypoxia with stimulus-responsive nanocarriers in overcoming drug resistance and monitoring anticancer efficacy. Acta Biomater 2018; 71:351-362. [PMID: 29545193 DOI: 10.1016/j.actbio.2018.03.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/23/2018] [Accepted: 03/06/2018] [Indexed: 12/13/2022]
Abstract
Although existing nanomedicines have focused on the tumor microenvironment with the goal of improving the effectiveness of conventional chemotherapy, the penetration of a tumor's core still represents a formidable barrier for existing drug delivery systems. Therefore, a novel multifunctional hypoxia-induced size-shrinkable nanoparticle has been designed to increase the penetration of drugs, nucleic acids, or probes into tumors. This cooperative strategy relies on three aspects: (i) the responsiveness of nanoparticles to hypoxia, which shrink when triggered by low oxygen concentrations; (ii) the core of a nanoparticle involves an internal cavity and strong positive charges on the surface to deliver both doxorubicin and siRNA; and (iii) a reactive oxygen species (ROS) probe is incorporated in the nanoparticle to monitor its preliminary therapeutic response in real time, which is expected to realize the enhanced efficacy together with the ability to self-monitor the anticancer activity. A more effective inhibition of tumor growth was observed in tumor-bearing zebrafish, demonstrating the feasibility of this cooperative strategy for in vivo applications. This research highlights a promising value in delivering drugs, nucleic acids, or probes to a tumor's core for cancer imaging and treatment. STATEMENT OF SIGNIFICANCE Hypoxia-induced chemoresistance of tumor cells still represents a formidable barrier, as it is difficult for existing drug delivery systems to penetrate the tumor hypoxia core. This study involves the hypoxia-responsive size-shrinkable nanoparticle co-delivery of DOX and siRNA to enhance the penetration of DOX deep within tumors and subsequently disturb crucial pathways of cancer development induced by hypoxia and to improve sensitization to DOX chemotherapy. Furthermore, the nanopreparation can combine the ROS probe as a self-reporting nanopreparation to realize the function of real-time feedback efficacy, which has a good application prospect in the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Zhiqi Xie
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Wangwei Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Ningning Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Mingyi Huangfu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Huina Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Mengting Lin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - WenHong Xu
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University, College of Medicine, Hangzhou 310058, PR China
| | - Jiejian Chen
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University, College of Medicine, Hangzhou 310058, PR China
| | - TianTian Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Qichun Wei
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University, College of Medicine, Hangzhou 310058, PR China
| | - Min Han
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China.
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
43
|
Matsuki D, Adewale O, Horie S, Okajima J, Komiya A, Oluwafemi O, Maruyama S, Mori S, Kodama T. Treatment of tumor in lymph nodes using near-infrared laser light-activated thermosensitive liposome-encapsulated doxorubicin and gold nanorods. JOURNAL OF BIOPHOTONICS 2017; 10:1676-1682. [PMID: 28417560 DOI: 10.1002/jbio.201600241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 06/07/2023]
Abstract
Tumor metastasis to lymph nodes is an important contributory factor for cancer-related deaths despite recent developments in cancer therapy. In this study, we demonstrate that tumor in the proper axillary lymph node (PALN) of the mouse can be treated by the application of external laser light to trigger the unloading of doxorubicin (DOX) encapsulated in thermosensitive liposomes (TSLs) administered together with gold nanorods (GNRs). GNRs + DOX-TSLs were injected into a mouse lymph node containing cancer cells (malignant fibrous histiocytoma-like cells) and intranodal DOX release was activated using near-infrared (NIR) laser irradiation. The temperature changes arising from the laser-irradiated GNRs triggered the release of DOX from the TSLs. A greater degree of inhibition of tumor growth was found in the co-therapy group compared to the other groups. The treatment effect was achieved by a combination of chemotherapy and NIR-activated hyperthermia. In vivo bioluminescence imaging and histological analysis confirmed tumor necrosis in response to combined treatment. This work presents a theranostic approach with excellent treatment results that has the potential to be developed into an alternative to surgery for the treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Daisuke Matsuki
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Oladipo Adewale
- Department of Applied Chemistry, University of Johannesburg, Doornfontein Campus P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Sachiko Horie
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Junnosuke Okajima
- Institute of Fluid Science, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, 980-8577, Japan
| | - Atsuki Komiya
- Institute of Fluid Science, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, 980-8577, Japan
| | - Oluwatobi Oluwafemi
- Department of Applied Chemistry, University of Johannesburg, Doornfontein Campus P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Shigenao Maruyama
- Institute of Fluid Science, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, 980-8577, Japan
| | - Shiro Mori
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Department of Oral and Maxillofacial Surgery, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Tetsuya Kodama
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| |
Collapse
|
44
|
Tahara K, Nishio M, Takeuchi H. Evaluation of liposomal behavior in the gastrointestinal tract after oral administration using real-time in vivo imaging. Drug Dev Ind Pharm 2017; 44:608-614. [DOI: 10.1080/03639045.2017.1405972] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu, Japan
| | - Maki Nishio
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu, Japan
| | - Hirofumi Takeuchi
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
45
|
Mazza M, Lozano N, Vieira DB, Buggio M, Kielty C, Kostarelos K. Liposome-Indocyanine Green Nanoprobes for Optical Labeling and Tracking of Human Mesenchymal Stem Cells Post-Transplantation In Vivo. Adv Healthc Mater 2017; 6. [PMID: 28777501 DOI: 10.1002/adhm.201700374] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/25/2017] [Indexed: 01/09/2023]
Abstract
Direct labeling of human mesenchymal stem cells (hMSC) prior to transplantation provides a means to track cells after administration and it is a powerful tool for the assessment of new cell-based therapies. Biocompatible nanoprobes consisting of liposome-indocyanine green hybrid vesicles (liposome-ICG) are used to safely label hMSC. Labeled hMSC recapitulating a 3D cellular environment is transplanted as spheroids subcutaneously and intracranially in athymic nude mice. Cells emit a strong NIR signal used for tracking post-transplantation with the IVIS imaging system up to 2 weeks (subcutaneous) and 1 week (intracranial). The transplanted stem cells are imaged in situ after engraftment deep in the brain up to 1 week in living animals using optical imaging techniques and without the need to genetically modify the cells. This method is proposed for efficient, nontoxic direct cell labeling for the preclinical assessment of cell-based therapies and the design of clinical trials, and potentially for localization of the cell engraftment after transplantation into patients.
Collapse
Affiliation(s)
- Mariarosa Mazza
- Nanomedicine Lab; Faculty of Biology, Medicine and Health; University of Manchester; Manchester M13 9PT UK
| | - Neus Lozano
- Nanomedicine Lab; Faculty of Biology, Medicine and Health; University of Manchester; Manchester M13 9PT UK
| | - Debora Braga Vieira
- Nanomedicine Lab; Faculty of Biology, Medicine and Health; University of Manchester; Manchester M13 9PT UK
| | - Maurizio Buggio
- Nanomedicine Lab; Faculty of Biology, Medicine and Health; University of Manchester; Manchester M13 9PT UK
| | - Cay Kielty
- Wellcome Trust Centre for Cell-Matrix Research; Faculty of Biology, Medicine and Health; University of Manchester; Manchester M13 9PT UK
| | - Kostas Kostarelos
- Nanomedicine Lab; Faculty of Biology, Medicine and Health; University of Manchester; Manchester M13 9PT UK
| |
Collapse
|
46
|
Lin L, Liang X, Xu Y, Yang Y, Li X, Dai Z. Doxorubicin and Indocyanine Green Loaded Hybrid Bicelles for Fluorescence Imaging Guided Synergetic Chemo/Photothermal Therapy. Bioconjug Chem 2017; 28:2410-2419. [DOI: 10.1021/acs.bioconjchem.7b00407] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Li Lin
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Xiaolong Liang
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yunxue Xu
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yongbo Yang
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Xiaoda Li
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Zhifei Dai
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
47
|
Nguyen HT, Tran TH, Thapa RK, Phung CD, Shin BS, Jeong JH, Choi HG, Yong CS, Kim JO. Targeted co-delivery of polypyrrole and rapamycin by trastuzumab-conjugated liposomes for combined chemo-photothermal therapy. Int J Pharm 2017; 527:61-71. [DOI: 10.1016/j.ijpharm.2017.05.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/03/2017] [Accepted: 05/16/2017] [Indexed: 01/09/2023]
|
48
|
Beztsinna N, Tsvetkova Y, Jose J, Rhourri-Frih B, Al Rawashdeh W, Lammers T, Kiessling F, Bestel I. Photoacoustic imaging of tumor targeting with riboflavin-functionalized theranostic nanocarriers. Int J Nanomedicine 2017; 12:3813-3825. [PMID: 28572726 PMCID: PMC5441666 DOI: 10.2147/ijn.s125192] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Photoacoustic imaging is an emerging method in the molecular imaging field, providing high spatiotemporal resolution and sufficient imaging depths for many clinical applications. Therefore, the aim of this study was to use photoacoustic imaging as a tool to evaluate a riboflavin (RF)-based targeted nanoplatform. RF is internalized by the cells through a specific pathway, and its derivatives were recently shown as promising tumor-targeting vectors for the drug delivery systems. Here, the RF amphiphile synthesized from a PEGylated phospholipid was successfully inserted into a long-circulating liposome formulation labeled with the clinically approved photoacoustic contrast agent – indocyanine green (ICG). The obtained liposomes had a diameter of 124 nm (polydispersity index =0.17) and had a negative zeta potential of −26 mV. Studies in biological phantoms indicated a stable and concentration-dependent photoacoustic signal (Vevo® LAZR) of the ICG-containing RF-functionalized liposomes. In A431 cells, a high uptake of RF-functionalized liposomes was found and could be blocked competitively. First, studies in mice revealed ~3 times higher photoacoustic signal in subcutaneous A431 tumor xenografts (P<0.05) after injection of RF-functionalized liposomes compared to control particles. In this context, the application of a spectral unmixing protocol confirmed the initial quantitative data and improved the localization of liposomes in the tumor. In conclusion, the synthesized RF amphiphile leads to efficient liposomal tumor targeting and can be favorably detected by photoacoustic imaging with a perspective of theranostic applications.
Collapse
Affiliation(s)
- Nataliia Beztsinna
- Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN), UMR 5248, University of Bordeaux, Pessac, France
| | - Yoanna Tsvetkova
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Jithin Jose
- Fujifilm VisualSonics BV, Amsterdam, the Netherlands
| | - Boutayna Rhourri-Frih
- Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN), UMR 5248, University of Bordeaux, Pessac, France
| | - Wa'el Al Rawashdeh
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany
| | - Isabelle Bestel
- Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN), UMR 5248, University of Bordeaux, Pessac, France
| |
Collapse
|
49
|
Surface modification of lipid-based nanocarriers for cancer cell-specific drug targeting. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0329-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
50
|
Al-Ahmady Z, Lozano N, Mei KC, Al-Jamal WT, Kostarelos K. Engineering thermosensitive liposome-nanoparticle hybrids loaded with doxorubicin for heat-triggered drug release. Int J Pharm 2017; 514:133-141. [PMID: 27863656 DOI: 10.1016/j.ijpharm.2016.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 01/15/2023]
Abstract
The engineering of responsive multifunctional delivery systems that combine therapeutic and diagnostic (theranostic) capabilities holds great promise and interest. We describe the design of thermosensitive liposome-nanoparticle (NP) hybrids that can modulate drug release in response to external heating stimulus. These hybrid systems were successfully engineered by the incorporation of gold, silver, and iron oxide NPs into the lipid bilayer of lysolipid-containing thermosensitive liposomes (LTSL). Structural characterization of LTSL-NP hybrids using cryo-EM and AFM revealed the incorporation of metallic NPs into the lipid membranes without compromising doxorubicin loading and retention capability. The presence of metallic NPs in the lipid bilayer reinforced bilayer retention and offered a nanoparticle concentration-dependent modulation of drug release in response to external heating. In conclusion, LTSL-NP hybrids represent a promising versatile platform based on LTSL liposomes that could further utilize the properties of the embedded NPs for multifunctional theranostic applications.
Collapse
Affiliation(s)
- Zahraa Al-Ahmady
- Nanomedicine Lab, Faculty of Biology, Medicine & Health, University of Manchester, AV Hill Building, Manchester M13 9PT, United Kingdom; UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Neus Lozano
- Nanomedicine Lab, Faculty of Biology, Medicine & Health, University of Manchester, AV Hill Building, Manchester M13 9PT, United Kingdom; UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Kuo-Ching Mei
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom; Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Wafa' T Al-Jamal
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom; University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Biology, Medicine & Health, University of Manchester, AV Hill Building, Manchester M13 9PT, United Kingdom; UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom.
| |
Collapse
|