1
|
Pellegrino C, Favalli N, Volta L, Benz R, Puglioli S, Bassi G, Zitzmann K, Auernhammer CJ, Nölting S, Magnani CF, Neri D, Beuschlein F, Manz MG. Peptide-guided adaptor-CAR T-Cell therapy for the treatment of SSTR2-expressing neuroendocrine tumors. Oncoimmunology 2024; 13:2412371. [PMID: 39376579 PMCID: PMC11457607 DOI: 10.1080/2162402x.2024.2412371] [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] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Somatostatin receptor type 2 (SSTR2) is one of the five subtypes of somatostatin receptors and is overexpressed on the surface of most gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs), pituitary tumors, paraganglioma, and meningioma, as well as hepatocellular carcinoma and breast cancer. Chimeric antigen receptor (CAR) T-cells are genetically engineered to express an artificial, T-cell activating binder, leading upon ligation to biocidal activity against target-antigen expressing cells. Adaptor-CAR T-cells recognize, via the CAR, a tag on an antigen-binding molecule, building an activating bridge between the CAR and the target cell. We hypothesized that a novel fluorescent-peptide antagonist of SSTR2, called Octo-Fluo, in combination with anti-FITC adaptor CAR (AdFITC(E2)-CAR) T-cells, may function as an on-off tunable activating bridge between the CAR and SSTR2 expressing target cells. In vitro studies confirmed the binding of Octo-Fluo to Bon1-SSTR2 mCherry-Luc cells without evidence of internalization. AdFITC(E2)-CAR T-cells were activated and efficiently induced Bon1-SSTR2 cell death in vitro, in an Octo-Fluo concentration-dependent manner. Similarly, AdFITC(E2)-CAR T-cells in combination with Octo-Fluo efficiently infiltrated the tumor and eliminated Bon1-SSTR2 tumors in immunodeficient mice in therapeutic settings. Both, AdFITC(E2)-CAR T-cell tumor infiltration and biocidal activity were Octo-Fluo concentration-dependent, with high doses of Octo-Fluo, saturating both the CAR and the SSTR2 antigen independently, leading to the loss of tumor infiltration and biocidal activity due to the loss of bridge formation. Our findings demonstrate the potential of using AdFITC(E2)-CAR T-cells with Octo-Fluo as a versatile, on-off tunable bispecific adaptor for targeted CAR T-cell immunotherapy against SSTR2-positive NETs.
Collapse
Affiliation(s)
- Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | | | - Laura Volta
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | - Ramon Benz
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | - Sara Puglioli
- Department of Chemistry, Philochem AG, Otelfingen, Switzerland
| | - Gabriele Bassi
- Department of Chemistry, Philochem AG, Otelfingen, Switzerland
| | - Kathrin Zitzmann
- Department of Medicine II, University-Hospital Munich-Grosshadern, University of Munich, Munich, Germany
| | | | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich (USZ), University of Zürich (UZH), Zürich, Switzerland
| | - Chiara F. Magnani
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| | - Dario Neri
- Department of Chemistry, Philochem AG, Otelfingen, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich (USZ), University of Zürich (UZH), Zürich, Switzerland
- Department of Internal Medicine IV and Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Ludwig Maximilian University, LMU Klinikum, Munich, Germany
- The LOOP Zurich - Medical Research Center, Zürich, Switzerland
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University Hospital Zürich (USZ) and University of Zürich (UZH), Comprehensive Cancer Center, Zürich, Switzerland
| |
Collapse
|
2
|
Faisal S, Tariq MH, Abdullah, Zafar S, Un Nisa Z, Ullah R, Ur Rahman A, Bari A, Ullah K, Khan RU. Bio synthesis, comprehensive characterization, and multifaceted therapeutic applications of BSA-Resveratrol coated platinum nanoparticles. Sci Rep 2024; 14:7875. [PMID: 38570564 PMCID: PMC10991511 DOI: 10.1038/s41598-024-57787-4] [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: 01/20/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
This study examines the manufacturing, characterization, and biological evaluation of platinum nanoparticles, which were synthesized by Enterobacter cloacae and coated with Bovine Serum Albumin (BSA) and Resveratrol (RSV). The formation of PtNPs was confirmed with the change of color from dark yellow to black, which was due to the bioreduction of platinum chloride by E. cloacae. BSA and RSV functionalization enhanced these nanoparticles' biocompatibility and therapeutic potential. TGA, SEM, XRD, and FTIR were employed for characterization, where PtNPs and drug conjugation-related functional groups were studied by FTIR. XRD confirmed the crystalline nature of PtNPs and Pt-BSA-RSV NPs, while TGA and SEM showed thermal stability and post-drug coating morphological changes. Designed composite was also found to be biocompatible in nature in hemolytic testing, indicating their potential in Biomedical applications. After confirmation of PtNPs based nanocaompsite synthesis, they were examined for anti-bacterial, anti-oxidant, anti-inflammatory, and anti-cancer properties. Pt-BSA-RSV NPs showed higher concentration-dependent DPPH scavenging activity, which measured antioxidant capability. Enzyme inhibition tests demonstrated considerable anti-inflammatory activity against COX-2 and 15-LOX enzymes. In in vitro anticancer studies, Pt-BSA-RSV NPs effectively killed human ovarian cancer cells. This phenomenon was demonstrated to be facilitated by the acidic environment of cancer, as the drug release assay confirmed the release of RSV from the NP formulation in the acidic environment. Finally, Molecular docking also demonstrated that RSV has strong potential as an anti-oxidant, antibacterial, anti-inflammatory, and anticancer agent. Overall, in silico and in vitro investigations in the current study showed good medicinal applications for designed nanocomposites, however, further in-vivo experiments must be conducted to validate our findings.
Collapse
Affiliation(s)
- Shah Faisal
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Biotechnology and Microbiology, Bacha Khan University, Charsadda, 24460, Pakistan.
| | - Muhammad Hamza Tariq
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Abdullah
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100, Gliwice, Poland
- Joint Doctoral School, Silesian University of Technology, Akademicka 2A, Gliwice, Poland
| | - Sania Zafar
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, 60000, Pakistan
| | - Zaib Un Nisa
- Department of Chemistry, Abdul Wali Khan University Mardan, Gardan Campus, Mardan, 23200, Pakistan
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Anees Ur Rahman
- Department of Health and Biological Science, Abasyn University, Peshawar, 25000, Pakistan
| | - Ahmed Bari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khair Ullah
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Chinese Academy of Sciences, Beijing, 100049, China
| | - Rahat Ullah Khan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
3
|
Wang L, Quine S, Frickenstein AN, Lee M, Yang W, Sheth VM, Bourlon MD, He Y, Lyu S, Garcia-Contreras L, Zhao YD, Wilhelm S. Exploring and Analyzing the Systemic Delivery Barriers for Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308446. [PMID: 38828467 PMCID: PMC11142462 DOI: 10.1002/adfm.202308446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 06/05/2024]
Abstract
Most nanomedicines require efficient in vivo delivery to elicit diagnostic and therapeutic effects. However, en route to their intended tissues, systemically administered nanoparticles often encounter delivery barriers. To describe these barriers, we propose the term "nanoparticle blood removal pathways" (NBRP), which summarizes the interactions between nanoparticles and the body's various cell-dependent and cell-independent blood clearance mechanisms. We reviewed nanoparticle design and biological modulation strategies to mitigate nanoparticle-NBRP interactions. As these interactions affect nanoparticle delivery, we studied the preclinical literature from 2011-2021 and analyzed nanoparticle blood circulation and organ biodistribution data. Our findings revealed that nanoparticle surface chemistry affected the in vivo behavior more than other nanoparticle design parameters. Combinatory biological-PEG surface modification improved the blood area under the curve by ~418%, with a decrease in liver accumulation of up to 47%. A greater understanding of nanoparticle-NBRP interactions and associated delivery trends will provide new nanoparticle design and biological modulation strategies for safer, more effective, and more efficient nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Skyler Quine
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Michael Lee
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit M. Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Margaret D. Bourlon
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lucila Garcia-Contreras
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73012, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma, 73019, USA
| |
Collapse
|
4
|
Moradbeygi F, Ghasemi Y, Farmani AR, Hemmati S. Glucarpidase (carboxypeptidase G2): Biotechnological production, clinical application as a methotrexate antidote, and placement in targeted cancer therapy. Biomed Pharmacother 2023; 166:115292. [PMID: 37579696 DOI: 10.1016/j.biopha.2023.115292] [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: 06/23/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023] Open
Abstract
Patients receiving high-dose methotrexate (HDMTX) for malignancies are exposed to diverse complications, including nephrotoxicity, hepatotoxicity, mucositis, myelotoxicity, neurological symptoms, and death. Glucarpidase is a recombinant carboxypeptidase G2 (CPG2) that converts MTX into nontoxic metabolites. In this study, the role of vector type, gene optimization, orientation, and host on the expression of CPG2 is investigated. The effectiveness of various therapeutic regimens containing glucarpidase is classified and perspectives on the dose adjustment based on precision medicine are provided. Conjugation with cell-penetrating peptides, human serum albumin, and polymers such as PEG and dextran for delivery, higher stability, and production of the biobetter variants of CPG2 is highlighted. Conjugation of CPG2 to F(ab՜)2 or scFv antibody fragments against tumor-specific antigens and the corresponding prodrugs for tumor-targeted drug delivery using the antibody-directed enzyme prodrug therapy (ADEPT) is communicated. Trials to reduce the off-target effects and the possibility of repeated ADEPT cycles by adding pro-domains sensitive to tumor-overexpressed proteases, antiCPG2 antibodies, CPG2 mutants with immune-system-unrecognizable epitopes, and protective polymers are reported. Intracellular cpg2 gene expression by gene-directed enzyme prodrug therapy (GDEPT) and the concerns regarding the safety and transfection efficacy of the GDEPT vectors are described. A novel bifunctional platform using engineered CAR-T cell micropharmacies, known as Synthetic Enzyme-Armed KillER (SEAKER) cells, expressing CPG2 to activate prodrugs at the tumor niche is introduced. Taken together, integrated data in this review and recruiting combinatorial strategies in novel drug delivery systems define the future directions of ADEPT, GDEPT, and SEAKER cell therapy and the placement of CPG2 therein.
Collapse
Affiliation(s)
- Fatemeh Moradbeygi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Reza Farmani
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Ni R, Huang L, Li Z, Zhang W, Wang Y, Shen Y, Wang J, Lu W. Multifunctional ROS-Responsive and TME-Modulated Lipid-Polymer Hybrid Nanoparticles for Enhanced Tumor Penetration. Int J Nanomedicine 2022; 17:5883-5897. [PMID: 36478745 PMCID: PMC9721131 DOI: 10.2147/ijn.s383517] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/23/2022] [Indexed: 08/09/2024] Open
Abstract
PURPOSE To enhance tumor penetration by formulation design and tumor microenvironment (TME) modulation, herein a novel reactive oxygen species (ROS)-responsive size/shape transformable lipid-polymer hybrid nanoparticle (LPN) has been fabricated for the co-delivery of an anticancer and collagen-inhibition drug. METHODS A ROS-responsive poly(D, L-lactide)-thioketal-polyethylene glycol (PLA-TK-PEG) co-polymer was synthesized. LPNs were then fabricated by encapsulation of losartan (LST)-loaded micelles as the core to support paclitaxel (PTX)-loaded liposomes. The PEG content in the lipid shell of LPNs was then adjusted to obtain the size-/shape-transformable LPNs (M/LST-Lip/PTX-PEG5%). The ROS-responsiveness was observed in vitro by transmission electron microscopy and the tumor-penetration of the LPNs was evaluated in 3D tumor spheroids by confocal laser scanning microscopy. Tumor-targeting, tumor-penetrating, and antitumor efficacies of the NPs in 4T1 tumor-bearing mice were determined by in vivo imaging. RESULTS ROS-responsive micellar core degradation and the transformation of spherical LPNs (120nm) to smaller 40 mm discoid nanoparticles (NP) were observed. The transformable LPNs exhibited enhanced capacity of penetration in contrast to the un-transformable preparations in three-dimensional (3D) tumor spheroids. Furthermore, synergetic penetrating enhancement was achieved by LST-loaded transformable LPNs in 4T1 and fibroblast cell mixed 3D tumor spheroids. The improved tumor penetration of LST-loaded transformable LPNs was observed in vivo, which could be due to their collagen inhibiting and size/shape transformable effect. Due to their enhanced penetrability, LST and PTX-loaded transformable LPNs demonstrated significant in vivo antitumor efficacy in comparison to other preparations. CONCLUSION The results confirmed the efficacy of M/LST-Lip/PTX-PEG5% in tumor targeting, collagen inhibition in TME, and enhanced tumor penetration. This novel drug delivery system can therefore play a substantial role in improving the therapeutic efficacy of antitumor drugs combined with TME-improving agents.
Collapse
Affiliation(s)
- Rui Ni
- School of Pharmacy, Fudan University, Shanghai, People’s Republic of China
- China State Institute of Pharmaceutical Industry, Shanghai, People’s Republic of China
- National Advanced Medical Engineering Research Center, Shanghai, People’s Republic of China
| | - Lele Huang
- National Advanced Medical Engineering Research Center, Shanghai, People’s Republic of China
| | - Zhen Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yajie Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yan Shen
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Jianxin Wang
- School of Pharmacy, Fudan University, Shanghai, People’s Republic of China
| | - Weigen Lu
- China State Institute of Pharmaceutical Industry, Shanghai, People’s Republic of China
| |
Collapse
|
6
|
Hu Y, Chen D, Napoleon JV, Srinivasarao M, Singhal S, Savran CA, Low PS. Efficient capture of circulating tumor cells with low molecular weight folate receptor-specific ligands. Sci Rep 2022; 12:8555. [PMID: 35595733 PMCID: PMC9122947 DOI: 10.1038/s41598-022-12118-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/29/2022] [Indexed: 11/09/2022] Open
Abstract
Retrieval of circulating tumor cells (CTC) has proven valuable for assessing a patient's cancer burden, evaluating response to therapy, and analyzing which drug might treat a cancer best. Although most isolation methods retrieve CTCs based on size, shape, or capture by tumor-specific antibodies, we explore here the use of small molecule tumor-specific ligands linked to magnetic beads for CTC capture. We have designed folic acid-biotin conjugates with different linkers for the capture of folate receptor (FR) + tumor cells spiked into whole blood, and application of the same technology to isolate FR + CTCs from the peripheral blood of both tumor-bearing mice and non-small cell lung patients. We demonstrate that folic acid linked via a rigid linker to a flexible PEG spacer that is in turn tethered to a magnetic bead enables optimal CTC retrieval, reaching nearly 100% capture when 100 cancer cells are spiked into 1 mL of aqueous buffer and ~ 90% capture when the same quantity of cells is diluted into whole blood. In a live animal model, the same methodology is shown to efficiently retrieve CTCs from tumor-bearing mice, yielding cancer cell counts that are proportional to total tumor burden. More importantly, the same method is shown to collect ~ 29 CTCs/8 mL peripheral blood from patients with non-small cell lung cancer. Since the ligand-presentation strategy optimized here should also prove useful in targeting other nanoparticles to other cells, the methods described below should have general applicability in the design of nanoparticles for cell-specific targeting.
Collapse
Affiliation(s)
- Yingwen Hu
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Danyang Chen
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - John V Napoleon
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Madduri Srinivasarao
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cagri A Savran
- School of Mechanical Engineering, Birck Nanotechnology Center, Purdue Center for Cancer Research, Purdue University, 1205 W. State St., West Lafayette, IN, 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
7
|
Phua VJX, Yang CT, Xia B, Yan SX, Liu J, Aw SE, He T, Ng DCE. Nanomaterial Probes for Nuclear Imaging. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:582. [PMID: 35214911 PMCID: PMC8875160 DOI: 10.3390/nano12040582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023]
Abstract
Nuclear imaging is a powerful non-invasive imaging technique that is rapidly developing in medical theranostics. Nuclear imaging requires radiolabeling isotopes for non-invasive imaging through the radioactive decay emission of the radionuclide. Nuclear imaging probes, commonly known as radiotracers, are radioisotope-labeled small molecules. Nanomaterials have shown potential as nuclear imaging probes for theranostic applications. By modifying the surface of nanomaterials, multifunctional radio-labeled nanomaterials can be obtained for in vivo biodistribution and targeting in initial animal imaging studies. Various surface modification strategies have been developed, and targeting moieties have been attached to the nanomaterials to render biocompatibility and enable specific targeting. Through integration of complementary imaging probes to a single nanoparticulate, multimodal molecular imaging can be performed as images with high sensitivity, resolution, and specificity. In this review, nanomaterial nuclear imaging probes including inorganic nanomaterials such as quantum dots (QDs), organic nanomaterials such as liposomes, and exosomes are summarized. These new developments in nanomaterials are expected to introduce a paradigm shift in nuclear imaging, thereby creating new opportunities for theranostic medical imaging tools.
Collapse
Affiliation(s)
- Vanessa Jing Xin Phua
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore; (V.J.X.P.); (S.X.Y.); (S.E.A.); (D.C.E.N.)
| | - Chang-Tong Yang
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore; (V.J.X.P.); (S.X.Y.); (S.E.A.); (D.C.E.N.)
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Bin Xia
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China; (B.X.); (T.H.)
| | - Sean Xuexian Yan
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore; (V.J.X.P.); (S.X.Y.); (S.E.A.); (D.C.E.N.)
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jiang Liu
- Department of Computer Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen 518055, China;
| | - Swee Eng Aw
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore; (V.J.X.P.); (S.X.Y.); (S.E.A.); (D.C.E.N.)
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Tao He
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China; (B.X.); (T.H.)
| | - David Chee Eng Ng
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore; (V.J.X.P.); (S.X.Y.); (S.E.A.); (D.C.E.N.)
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
8
|
Nie W, Wang B, Mi X, Chen J, Yu T, Miao J, Lin Y, Yang T, Ran M, Hong Z, Liu X, Liang X, Qian Z, Gao X. Co-Delivery of Paclitaxel and shMCL-1 by Folic Acid-Modified Nonviral Vector to Overcome Cancer Chemotherapy Resistance. SMALL METHODS 2021; 5:e2001132. [PMID: 34928100 DOI: 10.1002/smtd.202001132] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/05/2021] [Indexed: 02/05/2023]
Abstract
Acquired chemoresistance presents a major clinical impediment, which is an urgent problem to be solved. Interestingly, myeloma cell leukemia-1 (MCL-1) and folate receptor expression levels are higher in chemotherapy-resistant patients than in pretreatment patients. In this study, a multifunctional folic acid (FA)-targeting core-shell structure is presented for simultaneous delivery of shMCL-1 and paclitaxel (PTX). The transfection efficiency of shMCL-1 with the FA-targeting delivery system is higher than with a nontargeting delivery system in Skov3 and A2780T cells. The FA-targeting system significantly inhibits cell growth, blocks cell cycles, and promotes apoptosis of cancer cells in vitro. The mechanisms involved in inhibiting growth are related to Bcl-2/Bax and cdc2/Cyclin B1 pathways. An analysis of RNA sequencing suggests that shMCL-1 reverses chemoresistance through regulating genes such as regulator of chromosome condensation 2 (RCC2). The synergetic effect of shMCL-1 and PTX effectively inhibits tumor growth in both PTX-resistant and normal cancer models by inducing tumor apoptosis, inhibiting proliferation, and limiting tumor angiogenesis. The study results indicate that a FA-targeting delivery system combining shMCL-1 with PTX can simultaneously target tumor sites and restore the sensitivity of chemotherapy-resistant cancer to PTX. These findings have important implications for patients with normal or PTX-resistant cancer.
Collapse
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xue Mi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Junming Miao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunzhu Lin
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Tingting Yang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Mengni Ran
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zehuo Hong
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiaoxiao Liu
- Department of Radiation Oncology, Cancer Center, Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, P. R. China
| | - Xiao Liang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
9
|
Helmy LA, Abdel-Halim M, Hassan R, Sebak A, Farghali HAM, Mansour S, Tammam SN. The other side to the use of active targeting ligands; the case of folic acid in the targeting of breast cancer. Colloids Surf B Biointerfaces 2021; 211:112289. [PMID: 34954516 DOI: 10.1016/j.colsurfb.2021.112289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/08/2021] [Accepted: 12/12/2021] [Indexed: 12/14/2022]
Abstract
Due to its overexpression in cancer cells, the folate receptor (FR) is heavily exploited in the active targeting of nanoparticles (NPs). Its ligand, folic acid (FA) is as a consequence widely used as a NP targeting ligand. Although rather popular and successful in principle, recent data has shown that FA may result in breast cancer initiation and progression, which questions the suitability of FA as NP cancer targeting ligand. In this work, intravenous administration of free FA to healthy female mice resulted in breast tissue dysplasia, hyperplasia and in the increased expression of human epidermal growth factor receptor-2 (HER2), folate receptor (FR), cancer antigen 15-3 (CA15.3), vascular endothelial growth factor (VEGF), signal transducer and activator of transcription 3 (STAT3) and the pro-inflammatory cytokines, tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6) and interleukin-1β. In addition to the reduction in IL2. To evaluate the suitability and safety of FA as NP targeting ligand in breast cancer, small (≈ 150 nm) and large (≈ 500 nm) chitosan NPs were formulated and decorated with two densities of FA. The success of active targeting by FA was confirmed in two breast cancer cell lines (MCF-7 and MDA-MB-231 cells) in comparison to HEK293 cells. FA modified NPs that demonstrated successful active targeting in-vitro were assessed in-vivo. Upon intravenous administration, large NPs modified with a high density of FA accumulated in the breast tissue and resulted in similar effects as those observed with free FA. These results therefore question the suitability of FA as a targeting ligand in breast cancer and shed light on the importance of considering the activity (other than targeting) of the ligands used in NP active targeting.
Collapse
Affiliation(s)
- Lama A Helmy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, Egypt
| | - Mohammed Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, the German University in Cairo, Egypt
| | - Raghda Hassan
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, Egypt
| | - Aya Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, Egypt
| | - Haithem A M Farghali
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary medicine, Cairo University, Egypt
| | - Samar Mansour
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, Egypt; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy- Ain Shams University, Egypt
| | - Salma N Tammam
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, the German University in Cairo, Egypt.
| |
Collapse
|
10
|
Ahmed-Cox A, Pandzic E, Johnston ST, Heu C, McGhee J, Mansfeld FM, Crampin EJ, Davis TP, Whan RM, Kavallaris M. Spatio-temporal analysis of nanoparticles in live tumor spheroids impacted by cell origin and density. J Control Release 2021; 341:661-675. [PMID: 34915071 DOI: 10.1016/j.jconrel.2021.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/18/2021] [Accepted: 12/08/2021] [Indexed: 12/18/2022]
Abstract
Nanoparticles hold great preclinical promise in cancer therapy but continue to suffer attrition through clinical trials. Advanced, three dimensional (3D) cellular models such as tumor spheroids can recapitulate elements of the tumor environment and are considered the superior model to evaluate nanoparticle designs. However, there is an important need to better understand nanoparticle penetration kinetics and determine how different cell characteristics may influence this nanoparticle uptake. A key challenge with current approaches for measuring nanoparticle accumulation in spheroids is that they are often static, losing spatial and temporal information which may be necessary for effective nanoparticle evaluation in 3D cell models. To overcome this challenge, we developed an analysis platform, termed the Determination of Nanoparticle Uptake in Tumor Spheroids (DONUTS), which retains spatial and temporal information during quantification, enabling evaluation of nanoparticle uptake in 3D tumor spheroids. Outperforming linear profiling methods, DONUTS was able to measure silica nanoparticle uptake to 10 μm accuracy in both isotropic and irregularly shaped cancer cell spheroids. This was then extended to determine penetration kinetics, first by a forward-in-time, center-in-space model, and then by mathematical modelling, which enabled the direct evaluation of nanoparticle penetration kinetics in different spheroid models. Nanoparticle uptake was shown to inversely relate to particle size and varied depending on the cell type, cell stiffness and density of the spheroid model. The automated analysis method we have developed can be applied to live spheroids in situ, for the advanced evaluation of nanoparticles as delivery agents in cancer therapy.
Collapse
Affiliation(s)
- Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW 2031, Australia; ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Australian Center for NanoMedicine, UNSW Sydney, NSW 2031, Australia; School of Women and Children's Health, Faculty of Medicine and Health, UNSW Sydney, NSW 2031, Australia
| | - Elvis Pandzic
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Center, UNSW Sydney, NSW 2031, Australia
| | - Stuart T Johnston
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Systems Biology Laboratory, School of Mathematics and Statistics, and Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Celine Heu
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Center, UNSW Sydney, NSW 2031, Australia
| | - John McGhee
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Australian Center for NanoMedicine, UNSW Sydney, NSW 2031, Australia; 3D Visualisation Aesthetics Lab, UNSW Art & Design, UNSW Sydney, NSW 2021, Australia
| | - Friederike M Mansfeld
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW 2031, Australia; ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Australian Center for NanoMedicine, UNSW Sydney, NSW 2031, Australia; School of Women and Children's Health, Faculty of Medicine and Health, UNSW Sydney, NSW 2031, Australia; ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, 3052, Australia
| | - Edmund J Crampin
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Systems Biology Laboratory, School of Mathematics and Statistics, and Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria 3010, Australia; School of Medicine, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Thomas P Davis
- Precision Medicine, Australian Institute of Bioengineering & Nanotechnology, University of Queensland, QLD, 40679, Australia
| | - Renee M Whan
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Center, UNSW Sydney, NSW 2031, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW 2031, Australia; ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Australian Center for NanoMedicine, UNSW Sydney, NSW 2031, Australia; School of Women and Children's Health, Faculty of Medicine and Health, UNSW Sydney, NSW 2031, Australia.
| |
Collapse
|
11
|
Al-Mansoori L, Elsinga P, Goda SK. Bio-vehicles of cytotoxic drugs for delivery to tumor specific targets for cancer precision therapy. Biomed Pharmacother 2021; 144:112260. [PMID: 34607105 DOI: 10.1016/j.biopha.2021.112260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/09/2023] Open
Abstract
Abnormal structural and molecular changes in malignant tissues were thoroughly investigated and utilized to target tumor cells, hence rescuing normal healthy tissues and lowering the unwanted side effects as non-specific cytotoxicity. Various ligands for cancer cell specific markers have been uncovered and inspected for directional delivery of the anti-cancer drug to the tumor site, in addition to diagnostic applications. Over the past few decades research related to the ligand targeted therapy (LTT) increased tremendously aiming to treat various pathologies, mainly cancers with well exclusive markers. Malignant tumors are known to induce elevated levels of a variety of proteins and peptides known as cancer "markers" as certain antigens (e.g., Prostate specific membrane antigen "PSMA", carcinoembryonic antigen "CEA"), receptors (folate receptor, somatostatin receptor), integrins (Integrin αvβ3) and cluster of differentiation molecules (CD13). The choice of an appropriate marker to be targeted and the design of effective ligand-drug conjugate all has to be carefully selected to generate the required therapeutic effect. Moreover, since some tumors express aberrantly high levels of more than one marker, some approaches investigated targeting cancer cells with more than one ligand (dual or multi targeting). We aim in this review to report an update on the cancer-specific receptors and the vehicles to deliver cytotoxic drugs, including recent advancements on nano delivery systems and their implementation in targeted cancer therapy. We will discuss the advantages and limitations facing this approach and possible solutions to mitigate these obstacles. To achieve the said aim a literature search in electronic data bases (PubMed and others) using keywords "Cancer specific receptors, cancer specific antibody, tumor specific peptide carriers, cancer overexpressed proteins, gold nanotechnology and gold nanoparticles in cancer treatment" was carried out.
Collapse
Affiliation(s)
- Layla Al-Mansoori
- Qatar University, Biomedical Research Centre, Qatar University, Doha 2713, Qatar.
| | - Philip Elsinga
- University of Groningen, University Medical Center Groningen (UMCG), Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sayed K Goda
- Cairo University, Faculty of Science, Giza, Egypt; University of Derby, College of Science and Engineering, Derby, UK.
| |
Collapse
|
12
|
Janus-Type Mesoporous Silica Nanoparticles for Sequential Tumoral Cell and Mitochondria Targeting. Methods Mol Biol 2021. [PMID: 34118049 DOI: 10.1007/978-1-0716-1262-0_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The development of nanoparticles has provided a powerful weapon in the fight against cancer due to the discovery of their selective accumulation in tumoral tissues, known as enhanced permeation and retention (EPR) effect (Peer et al, Nat Nanotechnol 2:751-760, 2007). Tumoral tissues require afastformation of blood vessels to sustain this rapid growth.
Collapse
|
13
|
Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorg Chem 2021; 112:104946. [PMID: 33989916 DOI: 10.1016/j.bioorg.2021.104946] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| |
Collapse
|
14
|
Aramini B, Masciale V, Grisendi G, Banchelli F, D'Amico R, Maiorana A, Morandi U, Dominici M, Haider KH. Cancer stem cells and macrophages: molecular connections and future perspectives against cancer. Oncotarget 2021; 12:230-250. [PMID: 33613850 PMCID: PMC7869576 DOI: 10.18632/oncotarget.27870] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) have been considered the key drivers of cancer initiation and progression due to their unlimited self-renewal capacity and their ability to induce tumor formation. Macrophages, particularly tumor-associated macrophages (TAMs), establish a tumor microenvironment to protect and induce CSCs development and dissemination. Many studies in the past decade have been performed to understand the molecular mediators of CSCs and TAMs, and several studies have elucidated the complex crosstalk that occurs between these two cell types. The aim of this review is to define the complex crosstalk between these two cell types and to highlight potential future anti-cancer strategies.
Collapse
Affiliation(s)
- Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Center of Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Center of Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | |
Collapse
|
15
|
Mangal D, Conrad JC, Palmer JC. Nanoparticle dispersion in porous media: Effects of hydrodynamic interactions and dimensionality. AIChE J 2021. [DOI: 10.1002/aic.17147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Deepak Mangal
- Department of Chemical and Biomolecular Engineering University of Houston Houston Texas USA
| | - Jacinta C. Conrad
- Department of Chemical and Biomolecular Engineering University of Houston Houston Texas USA
| | - Jeremy C. Palmer
- Department of Chemical and Biomolecular Engineering University of Houston Houston Texas USA
| |
Collapse
|
16
|
Patel TK, Adhikari N, Amin SA, Biswas S, Jha T, Ghosh B. Small molecule drug conjugates (SMDCs): an emerging strategy for anticancer drug design and discovery. NEW J CHEM 2021. [DOI: 10.1039/d0nj04134c] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanisms of how SMDCs work. Small molecule drugs are conjugated with the targeted ligand using pH sensitive linkers which allow the drug molecule to get released at lower lysosomal pH. It helps to accumulate the chemotherapeutic agents to be localized in the tumor environment upon cleaving of the pH-labile bonds.
Collapse
Affiliation(s)
- Tarun Kumar Patel
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Nilanjan Adhikari
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Sk. Abdul Amin
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Swati Biswas
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Tarun Jha
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| |
Collapse
|
17
|
Chen Y, Li B, Feng J, Fang Q, Cheng J, Xie W, Li C, Cheng S, Zhang Y, Gao H. JAG1, Regulated by microRNA-424-3p, Involved in Tumorigenesis and Epithelial-Mesenchymal Transition of High Proliferative Potential-Pituitary Adenomas. Front Oncol 2020; 10:567021. [PMID: 33425722 PMCID: PMC7787033 DOI: 10.3389/fonc.2020.567021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022] Open
Abstract
Pituitary adenomas (PAs) are a neoplastic proliferation of anterior pituitary. Signature of Notch pathway relies upon the histopathological type of PAs. The details of Notch pathway that are involved in the migration and invasion of Pas are still unclear. This paper filters and testifies the relation between Notch signaling pathway and the migration/invasion in subtypes of PAs. The diversity of genes and pathways is investigated based on transcriptome data of 60 patients by KEGG pathway analysis and GSEA. A series of functional experiments demonstrate the role of candidate genes by overexpression and antibody blocking in GH3 cell line. Volcano map and GSEA results exhibit the differential and the priority of Jagged1 canonical Notch Ligand (JAG1) in the Notch pathway combined with clinical features. JAG1 is involved in epithelial–mesenchymal transition (EMT) in PAs by correlation analysis of RNA-seq data. Progression-free survival (PFS) of patients with high JAG1 was shorter than patients with low JAG1 according to follow-up data (P = 0.006). Furthermore, overexpression and antibody blocking experiments in GH3 cell line indicate that JAG1 could promote cell proliferation, migration, and G1/S transition. Double luciferase reporter assay gives manifests that JAG1 is the target gene of miR-424-3p, and mimics or inhibitor of miR-424-3p can regulate the level of JAG1 which, in turn, affects cell proliferation and the levels of MMP2 and VIM in GH3 cell line, respectively. Our study delves into the relation between the Notch signaling pathway and cell proliferation and EMT in PAs, providing a potential treatment through targeting JAG1.
Collapse
Affiliation(s)
- Yiyuan Chen
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jie Feng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Qiuyue Fang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianhua Cheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weiyan Xie
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Central Nervous System Injury, Capital Medical University, Beijing, China
| | - Sen Cheng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Central Nervous System Injury, Capital Medical University, Beijing, China
| | - Hua Gao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Central Nervous System Injury, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Bizmark N, Schneider J, Priestley RD, Datta SS. Multiscale dynamics of colloidal deposition and erosion in porous media. SCIENCE ADVANCES 2020; 6:6/46/eabc2530. [PMID: 33188022 PMCID: PMC7673751 DOI: 10.1126/sciadv.abc2530] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/23/2020] [Indexed: 05/08/2023]
Abstract
Diverse processes-e.g., environmental pollution, groundwater remediation, oil recovery, filtration, and drug delivery-involve the transport of colloidal particles in porous media. Using confocal microscopy, we directly visualize this process in situ and thereby identify the fundamental mechanisms by which particles are distributed throughout a medium. At high injection pressures, hydrodynamic stresses cause particles to be continually deposited on and eroded from the solid matrix-notably, forcing them to be distributed throughout the entire medium. By contrast, at low injection pressures, the relative influence of erosion is suppressed, causing particles to localize near the inlet of the medium. Unexpectedly, these macroscopic distribution behaviors depend on imposed pressure in similar ways for particles of different charges, although the pore-scale distribution of deposition is sensitive to particle charge. These results reveal how the multiscale interactions between fluid, particles, and the solid matrix control how colloids are distributed in a porous medium.
Collapse
Affiliation(s)
- Navid Bizmark
- Princeton Institute for the Science and Technology of Materials, Princeton University, Princeton, NJ 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Joanna Schneider
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Rodney D Priestley
- Princeton Institute for the Science and Technology of Materials, Princeton University, Princeton, NJ 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Sujit S Datta
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
19
|
Salim L, Desaulniers JP. To Conjugate or to Package? A Look at Targeted siRNA Delivery Through Folate Receptors. Nucleic Acid Ther 2020; 31:21-38. [PMID: 33121373 DOI: 10.1089/nat.2020.0893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RNA interference (RNAi) applications have evolved from experimental tools to study gene function to the development of a novel class of gene-silencing therapeutics. Despite decades of research, it was not until August 2018 that the US FDA approved the first-ever RNAi drug, marking a new era for RNAi therapeutics. Although there are many limitations associated with the inherent structure of RNA, delivery to target cells and tissues remains the most challenging. RNAs are unable to diffuse across cellular membranes due to their large size and polyanionic backbone and, therefore, require a delivery vector. RNAi molecules can be conjugated to a targeting ligand or packaged into a delivery vehicle. Alnylam has used both strategies in their FDA-approved formulations to achieve efficient delivery to the liver. To harness the full potential of RNAi therapeutics, however, we must be able to target additional cells and tissues. One promising target is the folate receptor α, which is overexpressed in a variety of tumors despite having limited expression and distribution in normal tissues. Folate can be conjugated directly to the RNAi molecule or used to functionalize delivery vehicles. In this review, we compare both delivery strategies and discuss the current state of research in the area of folate-mediated delivery of RNAi molecules.
Collapse
Affiliation(s)
- Lidya Salim
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, Canada
| | | |
Collapse
|
20
|
Zhou Y, Mowlazadeh Haghighi S, Liu Z, Wang L, Hruby VJ, Cai M. Development of Ligand-Drug Conjugates Targeting Melanoma through the Overexpressed Melanocortin 1 Receptor. ACS Pharmacol Transl Sci 2020; 3:921-930. [PMID: 33073191 DOI: 10.1021/acsptsci.0c00072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 12/31/2022]
Abstract
Melanoma is a lethal form of skin cancer. Despite recent breakthroughs of BRAF-V600E and PD-1 inhibitors showing remarkable clinical responses, melanoma can eventually survive these targeted therapies and become resistant. To solve the drug resistance issue, we designed and synthesized ligand-drug conjugates that couple cytotoxic drugs, which have a low cancer resistance issue, with the melanocortin 1 receptor (MC1R) agonist melanotan-II (MT-II), which provides specificity to MC1R-overexpressing melanoma. The drug-MT-II conjugates maintain strong binding interactions to MC1R and induce selective drug delivery to A375 melanoma cells through its MT-II moiety in vitro. Furthermore, using camptothecin as the cytotoxic drug, camptothecin-MT-II (compound 1) can effectively inhibit A375 melanoma cell growth with an IC50 of 16 nM. By providing selectivity to melanoma cells through its MT-II moiety, this approach of drug-MT-II conjugates enables us to have many more options for cytotoxic drug selection, which can be the key to solving the cancer resistant problem for melanoma.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Saghar Mowlazadeh Haghighi
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Zekun Liu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Lingzhi Wang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Minying Cai
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
21
|
Wang H, Liu Y, He R, Xu D, Zang J, Weeranoppanant N, Dong H, Li Y. Cell membrane biomimetic nanoparticles for inflammation and cancer targeting in drug delivery. Biomater Sci 2020; 8:552-568. [PMID: 31769765 DOI: 10.1039/c9bm01392j] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanoparticle capture and elimination by the immune system are great obstacles for drug delivery. Camouflaging nanoparticles with cell membrane represents a promising strategy to communicate and negotiate with the immune system. As a novel class of nanotherapeutics, such biomimetic nanoparticles inherit specific biological functionalities of the source cells (e.g., erythrocytes, immune cells, cancer cells and platelets) in order to evade immune elimination, prolong circulation time, and even target a disease region by virtue of the homing tendency of the cell membrane protein. In this review, we begin with an overview of different cell membranes that can be utilized to create a biointerface on nanoparticles. Subsequently, we elaborate on the state-of-the-art of cell membrane biomimetic nanoparticles for drug delivery. In particular, a summary of data on circulation capacity and targeting efficiency by camouflaged nanoparticles is presented. In addition to cancer therapy, inflammation treatment, as an emerging application of biomimetic nanoparticles, is specifically included. The challenges and outlook of this technology are discussed.
Collapse
Affiliation(s)
- Huaiji Wang
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Agazzi ML, Herrera SE, Cortez ML, Marmisollé WA, Azzaroni O. Self-assembled peptide dendrigraft supraparticles with potential application in pH/enzyme-triggered multistage drug release. Colloids Surf B Biointerfaces 2020; 190:110895. [DOI: 10.1016/j.colsurfb.2020.110895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/29/2020] [Accepted: 02/21/2020] [Indexed: 01/24/2023]
|
23
|
Huo C, Xiao J, Xiao K, Zou S, Wang M, Qi P, Liu T, Hu Y. Pre-Treatment with Zirconia Nanoparticles Reduces Inflammation Induced by the Pathogenic H5N1 Influenza Virus. Int J Nanomedicine 2020; 15:661-674. [PMID: 32099358 PMCID: PMC6996547 DOI: 10.2147/ijn.s221667] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
Background New approaches are urgently needed to fight influenza viral infection. Previous research has shown that zirconia nanoparticles can be used as anticancer materials, but their antiviral activity has not been reported. Here, we investigated the antiviral effect of zirconia (ZrO2) nanoparticles (NPs) against a highly pathogenic avian influenza virus. Materials and Methods In this study, the antiviral effects of ZrO2 on H5N1 virus were assessed in vivo, and the molecular mechanism responsible for this protection was investigated. Results Mice treated with 200 nm positively-charged NPs at a dose of 100 mg/kg showed higher survival rates and smaller reductions in weight. 200 nm ZrO2 activated mature dendritic cells and initially promoted the expression of cytokines associated with the antiviral response and innate immunity. In the lungs of H5N1-infected mice, ZrO2 treatment led to less pathological lung injury, significant reduction in influenza A virus replication, and overexpression of pro-inflammatory cytokines. Conclusion This antiviral study using zirconia NPs shows protection of mice against highly pathogenic avian influenza virus and suggests strong application potential for this method, introducing a new tool against a wide range of microbial infections.
Collapse
Affiliation(s)
- Caiyun Huo
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Jin Xiao
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Zhongmu Institutes of China Animal Husbandry Industry Co., Ltd, Beijing, People's Republic of China
| | - Kai Xiao
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Shumei Zou
- National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health and Family Planning Commission, Beijing, People's Republic of China
| | - Ming Wang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China.,Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Zhongmu Institutes of China Animal Husbandry Industry Co., Ltd, Beijing, People's Republic of China
| | - Peng Qi
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Zhongmu Institutes of China Animal Husbandry Industry Co., Ltd, Beijing, People's Republic of China
| | - Tianlong Liu
- Laboratory of Veterinary Pathology and Public Health, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Yanxin Hu
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| |
Collapse
|
24
|
Li X, Zhao Z, Yang Y, Liu Z, Wang J, Xu Y, Zhang Y. Novel β-1,3-d-glucan porous microcapsule enveloped folate-functionalized liposomes as a Trojan horse for facilitated oral tumor-targeted co-delivery of chemotherapeutic drugs and quantum dots. J Mater Chem B 2020; 8:2307-2320. [DOI: 10.1039/c9tb02674f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study, novel β-1,3-d-glucan porous microcapsule enveloped folate-functionalized liposomes were developed for the potential co-delivery of chemotherapeutic drugs and quantum dots with facilitated drug absorption and antitumor efficacy.
Collapse
Affiliation(s)
- Xiaonan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Ziming Zhao
- Department of Pharmaceutics, School of Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Yihua Yang
- Department of Pharmaceutics, School of Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Zhaorong Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Jinglei Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Yalu Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Yanzhuo Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
- Department of Pharmaceutics, School of Pharmacy
| |
Collapse
|
25
|
Naumenko VA, Vlasova KY, Garanina AS, Melnikov PA, Potashnikova DM, Vishnevskiy DA, Vodopyanov SS, Chekhonin VP, Abakumov MA, Majouga AG. Extravasating Neutrophils Open Vascular Barrier and Improve Liposomes Delivery to Tumors. ACS NANO 2019; 13:12599-12612. [PMID: 31609576 DOI: 10.1021/acsnano.9b03848] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Liposomes are the most extensively used nanocarriers in cancer therapy. Despite the advantages these vehicles provide over free drugs, there are still limitations with regards to the efficiency of liposomes delivery to tumors and off-target accumulation. A better understanding of nanodrugs extravasation mechanisms in different tumor types and normal vessels is needed to improve their antitumor activity. We used intravital microscopy to track for fluorescent liposomes behavior in xenograft tumor models (murine breast cancer 4T1 and melanoma B16, human prostate cancer 22Rv1) and normal skin and identified two distinct extravasation patterns. Microleakage, a local perivascular nanoparticle deposition, was found both in malignant and healthy tissues. This type of liposomes leakage does not provide access to tumor cells and is presumably responsible for drug deposition in normal tissues. In contrast, macroleakage penetrated deep into tissues and localized predominantly on the tumor-host interface. Although neutrophils did not uptake liposomes, their extravasation appeared to initiate both micro- and macroleakages. Based on neutrophils and liposomes extravasation dynamics, we hypothesized that microleakage and macroleakage are subsequent steps of the extravasation process corresponding to liposomes transport through endothelial and subendothelial barriers. Of note, extravasation spots were detected more often in the proximity of neutrophils, and across studied tumor types, neutrophils counts correlated with leakage frequencies. Reduced liposomes accumulation in 4T1 tumors upon Ly6G depletion further corroborated neutrophils role in nanoparticles delivery. Elucidating liposomes extravasation routes has a potential to help improve existing strategies and develop effective nanodrugs for cancer therapy.
Collapse
Affiliation(s)
- Victor A Naumenko
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
| | - Kseniya Yu Vlasova
- School of Chemistry , M. V. Lomonosov Moscow State University , Moscow 119991 , Russia
| | | | - Pavel A Melnikov
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Daria M Potashnikova
- School of Biology, Department of Cell Biology and Histology , M. V. Lomonosov Moscow State University , Moscow 119234 , Russia
| | - Daniil A Vishnevskiy
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Stepan S Vodopyanov
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
| | - Vladimir P Chekhonin
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Maxim A Abakumov
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
- Department of Medical Nanobiotechnology , N. I. Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Alexander G Majouga
- National University of Science and Technology (MISIS) , Moscow 119049 , Russia
- D. Mendeleev University of Chemical Technology of Russia , Moscow 125047 , Russia
| |
Collapse
|
26
|
Hsu JC, Cruz ED, Lau KC, Bouché M, Kim J, Maidment ADA, Cormode DP. Renally Excretable and Size-Tunable Silver Sulfide Nanoparticles for Dual-Energy Mammography or Computed Tomography. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2019; 31:7845-7854. [PMID: 33005070 PMCID: PMC7523639 DOI: 10.1021/acs.chemmater.9b01750] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Significant effort has been focused on developing renally-clearable nanoparticle agents since efficient renal clearance is important for eventual clinical translation. Silver sulfide nanoparticles (Ag2S-NP) have recently been identified as contrast agents for dual energy mammography, computed tomography (CT) and fluorescence imaging and probes for drug delivery and photothermal therapy with good biocompatibility. However, most Ag2S-NP reported to date are not renally excretable and are observed in vivo to accumulate and remain in the reticuloendothelial system (RES) organs, i.e. liver and spleen, for a long time, which could negatively impact their likelihood for translation. Herein, we present renally-clearable, 3.1 nm Ag2S-NP with 85% of the injected dose (ID) being excreted within 24 hours of intravenous injection, which is amongst the best clearance of similarly sized nanoparticles reported thus far (mostly between 20-75% of ID). The urinary excretion and low RES accumulation of these nanoparticles in mice were indicated by in vivo CT imaging and biodistribution analysis. In summary, these ultrasmall Ag2S-NP can be effectively eliminated via urine and have high translational potential for various biomedical applications.
Collapse
Affiliation(s)
- Jessica C. Hsu
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma D. Cruz
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Kristen C. Lau
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathilde Bouché
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Johoon Kim
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D. A. Maidment
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - David P. Cormode
- Department of Radiology, University of Pennsylvania 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
27
|
Deneka AY, Boumber Y, Beck T, Golemis EA. Tumor-Targeted Drug Conjugates as an Emerging Novel Therapeutic Approach in Small Cell Lung Cancer (SCLC). Cancers (Basel) 2019; 11:E1297. [PMID: 31484422 PMCID: PMC6769513 DOI: 10.3390/cancers11091297] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/25/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
There are few effective therapies for small cell lung cancer (SCLC), a highly aggressive disease representing 15% of total lung cancers. With median survival <2 years, SCLC is one of the most lethal cancers. At present, chemotherapies and radiation therapy are commonly used for SCLC management. Few protein-targeted therapies have shown efficacy in improving overall survival; immune checkpoint inhibitors (ICIs) are promising agents, but many SCLC tumors do not express ICI targets such as PD-L1. This article presents an alternative approach to the treatment of SCLC: the use of drug conjugates, where a targeting moiety concentrates otherwise toxic agents in the vicinity of tumors, maximizing the differential between tumor killing and the cytotoxicity of normal tissues. Several tumor-targeted drug conjugate delivery systems exist and are currently being actively tested in the setting of SCLC. These include antibody-drug conjugates (ADCs), radioimmunoconjugates (RICs), small molecule-drug conjugates (SMDCs), and polymer-drug conjugates (PDCs). We summarize the basis of action for these targeting compounds, discussing principles of construction and providing examples of effective versus ineffective compounds, as established by preclinical and clinical testing. Such agents may offer new therapeutic options for the clinical management of this challenging disease in the future.
Collapse
Affiliation(s)
- Alexander Y Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
- Department of Biochemistry, Kazan Federal University, 420000 Kazan, Russia.
| | - Yanis Boumber
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Department of Biochemistry, Kazan Federal University, 420000 Kazan, Russia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Tim Beck
- Cleveland Clinic, Cleveland, OH 44195, USA
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
28
|
Wang J, Geng Y, Zhang Y, Wang X, Liu J, Basit A, Miao T, Liu W, Jiang W. Bacterial magnetosomes loaded with doxorubicin and transferrin improve targeted therapy of hepatocellular carcinoma. Nanotheranostics 2019; 3:284-298. [PMID: 31423412 PMCID: PMC6696728 DOI: 10.7150/ntno.34601] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
High metastatic rate and recurrence of tumor because of tumor circulating cells are seriously hinders for clinical tumor therapy. Herein, we develop a novel, active-targeting nanotherapeutic by simultaneously loading doxorubicin (DOX) and transferrin (Tf) onto bacterial magnetosomes (Tf-BMs-DOX) and investigate its antitumor efficacy in vitro and in vivo. Drug release profiles indicated that Tf-BMs/BMs loaded with DOX were capable of sustained drug release, suggesting that reduce drugs required frequency of administration and enhance their therapeutic effect. The results of cellular uptake revealed that Tf-BMs-DOX recognized hepatocellular carcinoma HepG2 cells more specifically compared to HL-7702 normal hepatocytes because of high expression of transferrin receptor (TfR) on the surface of HepG2 cells. Tf-BMs-DOX increased tumor cytotoxicity and apoptosis more significantly than free DOX or BMs-DOX by regulating the expression of tumor-related and apoptosis-related genes. Following intravenous injection in HepG2 cell-bearing mice, Tf-BMs-DOX displayed tumor suppression rate of 56.78%, significantly higher than that of the BMs-DOX (41.53%) and free DOX (31.26%) groups. These results suggest that Tf-BMs-DOX have the potential to actively target to tumor sites, as well as the ability to kill circulating tumor cells via intravenous injection. Our findings provide a promising candidate for the clinical treatment of metastatic cancer.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yuanyuan Geng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yunpeng Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xi Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Junquan Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Abdul Basit
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ting Miao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Weiquan Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Wei Jiang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
29
|
Yu W, He X, Yang Z, Yang X, Xiao W, Liu R, Xie R, Qin L, Gao H. Sequentially responsive biomimetic nanoparticles with optimal size in combination with checkpoint blockade for cascade synergetic treatment of breast cancer and lung metastasis. Biomaterials 2019; 217:119309. [PMID: 31271855 DOI: 10.1016/j.biomaterials.2019.119309] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/15/2019] [Accepted: 06/25/2019] [Indexed: 12/30/2022]
Abstract
Recently, photodynamic therapy (PDT) emerges as a promising way to initiate immune response and being used in combination with chemotherapy. However, the antitumor effect is restricted due to the poor tumor penetration and retention, premature drug release and immunosuppressive environment of tumor sites. And as the size of nanoparticles plays a key role in drug delivery, series of hyaluronidase-responsive size-reducible biomimetic nanoparticles (mCAuNCs@HA) with different initial sizes are synthesized, and the optimal size of 150 nm is screened out because of the best blood circulation, tumor penetration and retention. Then the photosensitizer pheophorbide A and ROS-responsive paclitaxel dimer prodrug (PXTK) are co-loaded to facilitate on-demand drug release. The hydrolysis byproduct cinnamaldehyde in turn stimulates the ROS production by mitochondria, which compensates for the ROS consumed in the hydrolysis process. Anti-PD-L1 peptide (dPPA) is furthered loaded to alleviate the immunosuppressive environment of tumor and enhance the function of cytotoxic T lymphocytes activated by PDT-induced immunogenic cell death. The combination therapy activates CD4+, CD8+ T cells and NK cells and enhances secretion of cytokines (TNF-α and IL-12) with tumor inhibition rate increased to 84.2% and no metastasis is observed, providing a viable combination therapy for better anti-tumor and anti-metastasis efficacy.
Collapse
Affiliation(s)
- Wenqi Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Xueqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Zhihang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Xiaotong Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Wei Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Rui Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Rou Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
30
|
Pal A, Albusairi W, Liu F, Tuhin MTH, Miller M, Liang D, Joo H, Amin TU, Wilson EA, Faridi JS, Park M, Alhamadsheh MM. Hydrophilic Small Molecules That Harness Transthyretin To Enhance the Safety and Efficacy of Targeted Chemotherapeutic Agents. Mol Pharm 2019; 16:3237-3252. [PMID: 31136717 PMCID: PMC6607395 DOI: 10.1021/acs.molpharmaceut.9b00432] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
hydrophobicity of many chemotherapeutic agents usually results
in their nonselective passive distribution into healthy cells and
organs causing collateral toxicity. Ligand-targeted drugs (LTDs) are
a promising class of targeted anticancer agents. The hydrophilicity
of the targeting ligands in LTDs limits its nonselective passive tissue
distribution and toxicity to healthy cells. In addition, the small
size of LTDs allows for better tumor penetration, especially in the
case of solid tumors. However, the short circulation half-life of
LTDs, due to their hydrophilicity and small size, remains a significant
challenge for achieving their full therapeutic potential. Therefore,
extending the circulation half-life of targeted chemotherapeutic agents
while maintaining their hydrophilicity and small size will represent
a significant advance toward effective and safe cancer treatment.
Here, we present a new approach for enhancing the safety and efficacy
of targeted chemotherapeutic agents. By endowing hydrophobic chemotherapeutic
agents with a targeting moiety and a hydrophilic small molecule that
binds reversibly to the serum protein transthyretin, we generated
small hydrophilic drug conjugates that displayed enhanced circulation
half-life in rodents and selectivity to cancer cells. To the best
of our knowledge, this is the first demonstration of a successful
approach that maintains the small size and hydrophilicity of targeted
anticancer agents containing hydrophobic payloads while at the same
time extending their circulation half-life. This was demonstrated
by the superior in vivo efficacy and lower toxicity of our conjugates
in xenograft mouse models of metastatic prostate cancer.
Collapse
|
31
|
Lin X, Zhang X, Wang S, Liang X, Xu Y, Chen M, Gao C, Liu R, Tang J, Dai Z, Sun D. Intraoperative Identification and Guidance of Breast Cancer Microfoci Using Ultrasound and Near-Infrared Fluorescence Dual-Modality Imaging. ACS APPLIED BIO MATERIALS 2019; 2:2252-2261. [PMID: 35030664 DOI: 10.1021/acsabm.9b00206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Xiaona Lin
- Department of Ultrasonic Imaging, Peking University Shenzhen Hospital, Shenzhen 518035, China
| | - Xiaoting Zhang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Shumin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Yunxue Xu
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Min Chen
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Chuang Gao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Jie Tang
- 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
| | - Desheng Sun
- Department of Ultrasonic Imaging, Peking University Shenzhen Hospital, Shenzhen 518035, China
| |
Collapse
|
32
|
Orellana EA, Abdelaal AM, Rangasamy L, Tenneti S, Myoung S, Low PS, Kasinski AL. Enhancing MicroRNA Activity through Increased Endosomal Release Mediated by Nigericin. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:505-518. [PMID: 31071527 PMCID: PMC6506612 DOI: 10.1016/j.omtn.2019.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 12/28/2022]
Abstract
The therapeutic promise of small-RNA therapeutics is limited, not only by the lack of delivery vehicles, but also by the inability of the small RNAs to reach intracellular compartments where they can be biologically active. We previously reported successful delivery of functionally active miRNAs via receptor-mediated endocytosis. This type of targeted therapy still faces a major challenge in the delivery field: endosomal sequestration. Here, a new method has been developed to promote endosomal escape of delivered miRNA. The strategy relies on the difference in solute contents between nascent endosomes and the cytoplasm; early endosomes are rich in sodium ions, whereas the intracellular fluid is rich is potassium ions. Exploiting this difference through favoring the influx of potassium into the endosomes without the exchange of osmotically active sodium, results in an osmotic differential leading to the endosomes swelling and bursting. One molecule that is able to exchange potassium for an osmotically inactive hydrogen ion is the ionophore nigericin. Through generating an intramolecular miRNA delivery vehicle, containing a ligand, in this case folate and nigericin, we enabled the escape of folate-RNA conjugates from their entrapping endosomes into the cytoplasm where they bound the RNA-induced silencing complex and activated the RNAi response.
Collapse
Affiliation(s)
- Esteban A Orellana
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Ahmed M Abdelaal
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | | | - Sunghyun Myoung
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Andrea L Kasinski
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
33
|
Patel S, Kim J, Herrera M, Mukherjee A, Kabanov AV, Sahay G. Brief update on endocytosis of nanomedicines. Adv Drug Deliv Rev 2019; 144:90-111. [PMID: 31419450 PMCID: PMC6986687 DOI: 10.1016/j.addr.2019.08.004] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/06/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022]
Abstract
The complexity of nanoscale interactions between biomaterials and cells has limited the realization of the ultimate vision of nanotechnology in diagnostics and therapeutics. As such, significant effort has been devoted to advancing our understanding of the biophysical interactions of the myriad nanoparticles. Endocytosis of nanomedicine has drawn tremendous interest in the last decade. Here, we highlight the ever-present barriers to efficient intracellular delivery of nanoparticles as well as the current advances and strategies deployed to breach these barriers. We also introduce new barriers that have been largely overlooked such as the glycocalyx and macromolecular crowding. Additionally, we draw attention to the potential complications arising from the disruption of the newly discovered functions of the lysosomes. Novel strategies of exploiting the inherent intracellular defects in disease states to enhance delivery and the use of exosomes for bioanalytics and drug delivery are explored. Furthermore, we discuss the advances in imaging techniques like electron microscopy, super resolution fluorescence microscopy, and single particle tracking which have been instrumental in our growing understanding of intracellular pathways and nanoparticle trafficking. Finally, we advocate for the push towards more intravital analysis of nanoparticle transport phenomena using the multitude of techniques available to us. Unraveling the underlying mechanisms governing the cellular barriers to delivery and biological interactions of nanoparticles will guide the innovations capable of breaching these barriers.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Marco Herrera
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Anindit Mukherjee
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA; Department of Biomedical Engineering, Oregon Health and Science University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA.
| |
Collapse
|
34
|
Tahmasbi Rad A, Chen CW, Aresh W, Xia Y, Lai PS, Nieh MP. Combinational Effects of Active Targeting, Shape, and Enhanced Permeability and Retention for Cancer Theranostic Nanocarriers. ACS APPLIED MATERIALS & INTERFACES 2019; 11:10505-10519. [PMID: 30793580 DOI: 10.1021/acsami.8b21609] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Combinatory modulation of the physical and biochemical characteristics of nanocarrier delivery systems is an emergent topic in the field of nanomedicine. Here, we studied the combined effects of incorporation of active targeting moieties into nanocarriers and their morphology affecting the enhanced permeation and retention effect for nanomedicine cancer therapy. Self-assembled lipid discoidal and vesicular nanoparticles with low-polydispersity sub-50 nm size range and identical chemical compositions were synthesized, characterized, and correlated with in vitro cancer cellular internalization, in vivo tumor accumulation and cancer treatments. The fact that folate-associated bicelle yields the best outcome is indicative of the preference for discoidal carriers over spherical carriers and the improved targeting efficacy due to the targeting ligand/receptor binding. The approach is successfully adopted to design the nanocarriers for photodynamic therapy, which yields a consistent trend in in vitro and in vivo efficacy: folate nanodiscs > folate vesicles > nonfolate nanodiscs > nonfolate vesicles. Folate discs not only have shown a higher tumor uptake and photothermal therapeutic efficiency, but also minimize skin photosensitivity side effects. The advantages of nanodiscoidal bicelles as nanocarriers, including well-defined size, robust formation, easy encapsulation of hydrophobic molecules (therapeutics and/or diagnostics), easy incorporation of targeting molecules, and low toxicity, enable the scalable manufacturing of a generalized in vivo multimodal delivery platform.
Collapse
Affiliation(s)
- Armin Tahmasbi Rad
- Polymer Program, Institute of Materials Sciences , University of Connecticut , 191 Auditorium Road , Storrs , Connecticut 06269 , United States
| | - Ching-Wen Chen
- Department of Chemistry , National Chung Hsing University , Taichung 402 , Taiwan , ROC
| | - Wafa Aresh
- Polymer Program, Institute of Materials Sciences , University of Connecticut , 191 Auditorium Road , Storrs , Connecticut 06269 , United States
| | | | - Ping-Shan Lai
- Department of Chemistry , National Chung Hsing University , Taichung 402 , Taiwan , ROC
| | - Mu-Ping Nieh
- Polymer Program, Institute of Materials Sciences , University of Connecticut , 191 Auditorium Road , Storrs , Connecticut 06269 , United States
| |
Collapse
|
35
|
Zhao Z, Ukidve A, Krishnan V, Mitragotri S. Effect of physicochemical and surface properties on in vivo fate of drug nanocarriers. Adv Drug Deliv Rev 2019; 143:3-21. [PMID: 30639257 DOI: 10.1016/j.addr.2019.01.002] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/07/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023]
Abstract
Over the years, a plethora of materials - natural and synthetic - have been engineered at a nanoscopic level and explored for drug delivery. Nanocarriers based on such materials could improve the payload's pharmacokinetics and achieve the desired pharmacological response at the target tissue. Despite the development of rationally designed drug nanocarriers, only a handful of such formulations have been successfully translated into the clinic. The physicochemical properties (size, shape, surface chemistry, porosity, elasticity, and many others) of these nanocarriers influence its biological identity, which in presence of biological barriers in vivo, could significantly modulate the therapeutic index of its cargo and alter the desired outcome. Further, complexities associated with developing effective drug nanocarriers have led to conflicting views of its safety, permeation of biological barriers and cellular uptake. Here, in this review, we emphasize the effect of physicochemical properties of nanocarriers on their interactions with the biological milieu. The review will discuss in depth, how modulating the physicochemical properties would influence a drug nanocarrier's behavior in vivo and the mechanisms underlying these effects. The goal of this review is to summarize the design considerations based on these properties and to provide a conceptual template for achieving improved therapeutic efficacy with enhanced patient compliance.
Collapse
|
36
|
Zhuang C, Guan X, Ma H, Cong H, Zhang W, Miao Z. Small molecule-drug conjugates: A novel strategy for cancer-targeted treatment. Eur J Med Chem 2019; 163:883-895. [DOI: 10.1016/j.ejmech.2018.12.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 10/27/2022]
|
37
|
Figliola C, Marchal E, Groves BR, Thompson A. A step-wise synthetic approach is necessary to access γ-conjugates of folate: folate-conjugated prodigiosenes. RSC Adv 2019; 9:14078-14092. [PMID: 35519339 PMCID: PMC9064012 DOI: 10.1039/c9ra01435g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/23/2019] [Indexed: 11/21/2022] Open
Abstract
Despite the vast literature that describes reacting folic acid with a pharmacophore, this route is ineffective in providing the correct regioisomer of the resulting conjugate. We herein present a step-wise route to the preparation of nine folate conjugates of the tripyrrolic prodigiosene skeleton. The strict requirement for step-wise construction of the folate core is demonstrated, so as to achieve conjugation at only the desired γ-carboxylic acid and thus maintain the α-carboxylic site for folate receptor (FRα) recognition. Linkages via ethylenediamine, polyethylene glycol and glutathione are demonstrated. Despite the vast literature that describes reacting folic acid with a pharmacophore, this route is ineffective in providing the correct regioisomer of the resulting conjugate.![]()
Collapse
|
38
|
Shi H, Leonhard WN, Sijbrandi NJ, van Steenbergen MJ, Fens MHAM, van de Dikkenberg JB, Toraño JS, Peters DJM, Hennink WE, Kok RJ. Folate-dactolisib conjugates for targeting tubular cells in polycystic kidneys. J Control Release 2018; 293:113-125. [PMID: 30472374 DOI: 10.1016/j.jconrel.2018.11.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/22/2022]
Abstract
The aim of the present study was to develop folic acid (FA) conjugates which can deliver the kinase inhibitor dactolisib to the kidneys via folate receptor-mediated uptake in tubular epithelial cells. Dactolisib is a dual inhibitor of phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) and is considered an attractive agent for treatment of polycystic kidney disease. The ethylenediamine platinum(II) linker, herein called Lx, was employed to couple dactolisib via coordination chemistry to thiol-containing FA-spacer adducts to yield FA-Lx-dactolisib conjugates. The dye lissamine was coupled via similar linker chemistry to folate to yield fluorescent FA-Lx-lissamine conjugates. Three different spacers (PEG5-Cys, PEG27-Cys or an Asp-Arg-Asp-Asp-Cys peptide spacer) were used to compare the influence of hydrophilicity and charged groups in the spacer on interaction with target cells and in vivo organ distribution of the final conjugates. The purity and identity of the final products were confirmed by UPLC and LC-MS analysis, respectively. FA-Lx-dactolisib conjugates were stable in serum and culture medium, while dactolisib was released from the conjugates in the presence of glutathione. All three type of conjugates were internalized efficiently by HK-2 cells and uptake could be blocked by an excess of folic acid in the medium, demonstrating FR mediated uptake. FA-Lx-dactolisib conjugates showed nanomolar inhibition of the PI3K pathway (Akt phosphorylation) and mTOR pathway (S6 phosphorylation) in cultured kidney epithelial cells (HK-2 cells). After intraperitoneal administration, all three types conjugates accumulated extensively in kidneys of iKsp-Pkd1del mice with polycystic kidney disease. In conclusion, folate conjugates were successfully prepared by platinum(II) coordination chemistry and accumulated in a target-specific manner in kidney cells and polycystic kidneys. The folate conjugate of dactolisib thus may have potential for targeted therapy of polycystic kidney disease.
Collapse
Affiliation(s)
- Haili Shi
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wouter N Leonhard
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Mies J van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Joep B van de Dikkenberg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Javier Sastre Toraño
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Robbert Jan Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
39
|
Mahalingam SM, Chu H, Liu X, Leamon CP, Low PS. Carbonic Anhydrase IX-Targeted Near-Infrared Dye for Fluorescence Imaging of Hypoxic Tumors. Bioconjug Chem 2018; 29:3320-3331. [PMID: 30185025 DOI: 10.1021/acs.bioconjchem.8b00509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Use of tumor-targeted fluorescence dyes to help surgeons identify otherwise undetected tumor nodules, decrease the incidence of cancer-positive margins, and facilitate localization of malignant lymph nodes has demonstrated considerable promise for improving cancer debulking surgery. Unfortunately, the repertoire of available tumor-targeted fluorescent dyes does not permit identification of all cancer types, raising the need to develop additional tumor-specific fluorescent dyes to ensure localization of all malignant lesions during cancer surgeries. By comparing the mRNA levels of the hypoxia-induced plasma membrane protein carbonic anhydrase IX (CA IX) in 13 major human cancers with the same mRNA levels in corresponding normal tissues, we document that CA IX constitutes a nearly universal marker for the design of tumor-targeted fluorescent dyes. Motivated by this expression profile, we synthesize two new CA IX-targeted near-infrared (NIR) fluorescent imaging agents and characterize their physical and biological properties both in vitro and in vivo. We report that conjugation of either acetazolamide or 6-aminosaccharin (i.e., two CA-IX-specific ligands) to the NIR fluorescent dye, S0456, via an extended phenolic spacer creates a brightly fluorescent dye that binds CA IX with high affinity and allows rapid visualization of hypoxic regions of solid tumors at depths >1 cm beneath a tissue surface. Taken together, these data suggest that a CA IX-targeted NIR dye can constitute a useful addition to a cocktail of tumor-targeted NIR dyes designed to image all human cancers.
Collapse
Affiliation(s)
| | - Haiyan Chu
- Endocyte Inc. , 3000 Kent Avenue , West Lafayette , Indiana 47906 , United States
| | | | - Christopher P Leamon
- Endocyte Inc. , 3000 Kent Avenue , West Lafayette , Indiana 47906 , United States
| | | |
Collapse
|
40
|
Orellana EA, Tenneti S, Rangasamy L, Lyle LT, Low PS, Kasinski AL. FolamiRs: Ligand-targeted, vehicle-free delivery of microRNAs for the treatment of cancer. Sci Transl Med 2018; 9:9/401/eaam9327. [PMID: 28768807 DOI: 10.1126/scitranslmed.aam9327] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/16/2017] [Indexed: 12/20/2022]
Abstract
MicroRNAs are small RNAs that negatively regulate gene expression posttranscriptionally. Because changes in microRNA expression can promote or maintain disease states, microRNA-based therapeutics are being evaluated extensively. Unfortunately, the therapeutic potential of microRNA replacement is limited by deficient delivery vehicles. In this work, microRNAs are delivered in the absence of a protective vehicle. The method relies on direct attachment of microRNAs to folate (FolamiR), which mediates delivery of the conjugated microRNA into cells that overexpress the folate receptor. We show that the tumor-suppressive FolamiR, FolamiR-34a, is quickly taken up both by triple-negative breast cancer cells in vitro and in vivo and by tumors in an autochthonous model of lung cancer and slows their progression. This method delivers microRNAs directly to tumors in vivo without the use of toxic vehicles, representing an advance in the development of nontoxic, cancer-targeted therapeutics.
Collapse
Affiliation(s)
- Esteban A Orellana
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,PULSe Graduate Program, Purdue University, West Lafayette, IN 47907, USA
| | - Srinivasarao Tenneti
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA.,Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH 43202, USA
| | | | - L Tiffany Lyle
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Andrea L Kasinski
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA. .,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
41
|
The diosgenin prodrug nanoparticles with pH-responsive as a drug delivery system uniquely prevents thrombosis without increased bleeding risk. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:673-684. [DOI: 10.1016/j.nano.2017.12.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 12/08/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022]
|
42
|
Poh S, Chelvam V, Ayala-López W, Putt KS, Low PS. Selective liposome targeting of folate receptor positive immune cells in inflammatory diseases. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1033-1043. [DOI: 10.1016/j.nano.2018.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/18/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022]
|
43
|
Niu Y, Zhu J, Li Y, Shi H, Gong Y, Li R, Huo Q, Ma T, Liu Y. Size shrinkable drug delivery nanosystems and priming the tumor microenvironment for deep intratumoral penetration of nanoparticles. J Control Release 2018; 277:35-47. [PMID: 29545106 DOI: 10.1016/j.jconrel.2018.03.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/11/2018] [Indexed: 02/08/2023]
Abstract
The penetration of nanomedicine into solid tumor still constitutes a great challenge for cancer therapy, which lead to the failure of thorough clearance of tumor cells. Aiming at solving this issue, lots of encouraging progress has been made in the development of multistage nanoparticles triggered by various stimuli in the past few years. Besides, the therapeutical effects of nanoagents are also greatly impacted by the complex tumor microenvironment, and remodeling tumor microenvironment has become another important approach for promoting nanoparticles penetration. In this review, we summarize and analyze recent research progress and challenges in promoting nanoparticle penetration based on two kinds of different strategies, which include size shrinkable nanoparticles and priming tumor microenvironments. Especially, many recent reported multi-strategy approaches based on particle size reduction in conjugated with other therapeutic strategies are discussed. And we expect to provide some useful enlightenments and proposals on nanotechnology-based drug delivery systems for more effective therapy of solid tumors.
Collapse
Affiliation(s)
- Yimin Niu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Yang Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Huihui Shi
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yaxiang Gong
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Rui Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Huo
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Tao Ma
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Yang Liu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
44
|
Roointan A, Kianpour S, Memari F, Gandomani M, Gheibi Hayat SM, Mohammadi-Samani S. Poly(lactic-co-glycolic acid): The most ardent and flexible candidate in biomedicine! INT J POLYM MATER PO 2018. [DOI: 10.1080/00914037.2017.1405350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Amir Roointan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Kianpour
- Department of Pharmaceutical Biotechnology, Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Memari
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Molood Gandomani
- Department of Bioengineering, Biotechnology Research Center, Cyprus international University, Nicosia, Cyprus
| | - Seyed Mohammad Gheibi Hayat
- Student Research Committee, Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soliman Mohammadi-Samani
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
45
|
Zhou J, Li M, Lim WQ, Luo Z, Phua SZF, Huo R, Li L, Li K, Dai L, Liu J, Cai K, Zhao Y. A Transferrin-Conjugated Hollow Nanoplatform for Redox-Controlled and Targeted Chemotherapy of Tumor with Reduced Inflammatory Reactions. Theranostics 2018; 8:518-532. [PMID: 29290824 PMCID: PMC5743564 DOI: 10.7150/thno.21194] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/17/2017] [Indexed: 02/05/2023] Open
Abstract
Purpose: In this study, we report the design, development and evaluation of a hollow drug delivery nanoplatform for cancer therapy in vitro and in vivo. This composite nanosystem was prepared by modifying hollow mesoporous silica nanoparticles (HMSNs) with transferrin (Tf) targeting moieties via redox-liable linkage, and was capable of delivering therapeutic cargos (doxorubicin) specifically to the tumor site and subsequently releasing them in an on-demand manner. Moreover, the Tf corona could simultaneously reduce the inflammatory response after intravenous administration in vivo. Methods: Nanostructural morphology of the drug delivery system was observed by scanning electron microscope and transmission electron microscope. The preparation process was monitored primarily using Fourier-transform infrared spectroscopy, dynamic light scattering, nitrogen adsorption/desorption isotherm, and thermogravimetric analysis. The release profile in solution was monitored by fluorescence spectroscopy. In vitro drug delivery efficacy was evaluated on MDA-MB-231 breast cancer cell line using confocal laser scanning microscopy, MTT assay and flow cytometry. In vitro inflammatory response was evaluated on RAW264.7 macrophage cells. In vivo therapeutic experiments were carried out using in situ mouse breast cancer models. Results: The experimental results evidently demonstrate that the developed nanocarrier could effectively deliver anticancer drugs to the tumor site in a targeted manner and release them in response to the elevated glutathione level inside tumor cells, resulting in improved anticancer efficacy both in vitro and in vivo. Moreover, the Tf conjugation significantly ameliorated the inflammatory reaction triggered by the administration of the nanocarrier. Conclusions: This manuscript demonstrated that the Tf-conjugated HMSNs could enhance the delivery efficiency of anticancer drugs, while simultaneously alleviating the adverse side effects. The current study presents a promising integrated delivery system toward effective and safe cancer treatment.
Collapse
Affiliation(s)
- Jun Zhou
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Wei Qi Lim
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Soo Zeng Fiona Phua
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
| | - Runlan Huo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Liqi Li
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Ke Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Liangliang Dai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Junjie Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798
| |
Collapse
|
46
|
Sharma A, Kim EJ, Shi H, Lee JY, Chung BG, Kim JS. Development of a theranostic prodrug for colon cancer therapy by combining ligand-targeted delivery and enzyme-stimulated activation. Biomaterials 2017; 155:145-151. [PMID: 29175083 DOI: 10.1016/j.biomaterials.2017.11.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 01/03/2023]
Abstract
The high incidence of colorectal cancer worldwide is currently a major health concern. Although conventional chemotherapy and surgery are effective to some extent, there is always a risk of relapse due to associated side effects, including post-surgical complications and non-discrimination between cancer and normal cells. In this study, we developed a small molecule-based theranostic system, Gal-Dox, which is preferentially taken up by colon cancer cells through receptor-mediated endocytosis. After cancer-specific activation, the active drug Dox (doxorubicin) is released with a fluorescence turn-on response, allowing both drug localization and site of action to be monitored. The therapeutic potency of Gal-Dox was also evaluated, both in vivo and ex vivo, thus illustrating the potential of Gal-Dox as a colorectal cancer theranostic with great specificity.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Eun-Joong Kim
- Department of Mechanical Engineering, Sogang University, Seoul 04107, South Korea
| | - Hu Shi
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jin Yong Lee
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, South Korea.
| | - Bong Geun Chung
- Department of Mechanical Engineering, Sogang University, Seoul 04107, South Korea.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, South Korea.
| |
Collapse
|
47
|
Ji T, Lang J, Wang J, Cai R, Zhang Y, Qi F, Zhang L, Zhao X, Wu W, Hao J, Qin Z, Zhao Y, Nie G. Designing Liposomes To Suppress Extracellular Matrix Expression To Enhance Drug Penetration and Pancreatic Tumor Therapy. ACS NANO 2017; 11:8668-8678. [PMID: 28806504 DOI: 10.1021/acsnano.7b01026] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
During pancreatic tumor development, pancreatic stellate cells (PSCs) proliferate exuberantly to secrete extracellular matrix (ECM) in the tumor stroma, which presents major barriers for drug delivery and penetration in tumor tissue. Thus, down-regulating ECM levels via regulation of the PSCs may allow enhanced penetration of therapeutic drugs and thereby enhancing their therapeutic efficacy. To regulate the PSCs, a matrix metalloproteinase-2 (MMP-2) responsive peptide-hybrid liposome (MRPL) was constructed via coassembly of a tailor-designed MMP-2 responsive amphiphilic peptide and phospholipids. By utilizing the MMP-2-rich pathological environment, the pirfenidone (PFD) loaded MRPL (MRPL-PFD) can specifically release PFD at the pancreatic tumor site and down-regulate the multiple components of ECM expressed by the PSCs. This resulted in a significant increase in the penetration of gemcitabine into the tumor tissue and enhanced the efficacy of gemcitabine for pancreatic tumor. Our design tailored for antifibrosis of pancreatic cancer may provide a practical approach to build functional liposomes through supramolecular assembly, and regulation of ECM may be a promising adjuvant therapeutic strategy for pancreatic and other ECM-rich tumors.
Collapse
Affiliation(s)
- Tianjiao Ji
- The First Affiliated Hospital, Zhengzhou University , Zhengzhou 450052, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jiayan Lang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- College of Pharmaceutical Science, Jilin University , Changchun 130021, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Feifei Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Lijing Zhang
- The First Affiliated Hospital, Zhengzhou University , Zhengzhou 450052, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Wenjing Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- College of Pharmaceutical Science, Jilin University , Changchun 130021, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jihui Hao
- Department of Pancreatic Carcinoma Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy , Tianjin 300060, China
| | - Zhihai Qin
- The First Affiliated Hospital, Zhengzhou University , Zhengzhou 450052, China
| | - Ying Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
48
|
Affiliation(s)
- Madduri Srinivasarao
- Purdue Institute for Drug
Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S. Low
- Purdue Institute for Drug
Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
49
|
Xu Y, Asghar S, Yang L, Chen Z, Li H, Shi W, Li Y, Shi Q, Ping Q, Xiao Y. Nanoparticles based on chitosan hydrochloride/hyaluronic acid/PEG containing curcumin: In vitro evaluation and pharmacokinetics in rats. Int J Biol Macromol 2017; 102:1083-1091. [DOI: 10.1016/j.ijbiomac.2017.04.105] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 12/20/2022]
|
50
|
López V, Villegas MR, Rodríguez V, Villaverde G, Lozano D, Baeza A, Vallet-Regí M. Janus Mesoporous Silica Nanoparticles for Dual Targeting of Tumor Cells and Mitochondria. ACS APPLIED MATERIALS & INTERFACES 2017; 9:26697-26706. [PMID: 28759196 DOI: 10.1021/acsami.7b06906] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The development of targeted nanocarriers able to be selectively internalized within tumor cells, and therefore to deliver anti-tumor drugs specifically to diseased cells, constitutes one of the most important goals in nano-oncology. Herein, the development of Janus mesoporous silica particles asymmetrically decorated with two targeting moieties, one of them selective for folate membrane cell receptors (folic acid) and the other one able to bind to mitochondria membrane (triphenylphosphine, TPP), is described in order to achieve sequential cell to organelle vectorization. The asymmetric decoration of each side of the particle allows fine control in the targeting attachment process in comparison with the use of symmetric nanocarriers. The presence of folic acid induces a higher increase in particle accumulation inside tumor cells, and once there, these nanocarriers are guided close to mitochondria by the action of the TPP moiety. This strategy can be applied for improving the therapeutic efficacy of current nanomedicines.
Collapse
Affiliation(s)
- Victoria López
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
| | - Maria Rocío Villegas
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine , Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Verónica Rodríguez
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
| | - Gonzalo Villaverde
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine , Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Daniel Lozano
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine , Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Alejandro Baeza
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine , Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid , 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine , Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| |
Collapse
|