1
|
Moudgil A, Salve R, Gajbhiye V, Chaudhari BP. Challenges and emerging strategies for next generation liposomal based drug delivery: An account of the breast cancer conundrum. Chem Phys Lipids 2023; 250:105258. [PMID: 36375540 DOI: 10.1016/j.chemphyslip.2022.105258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
The global cancer burden is witnessing an upsurge with breast cancer surpassing other cancers worldwide. Furthermore, an escalation in the breast cancer caseload is also expected in the coming years. The conventional therapeutic regimens practiced routinely are associated with many drawbacks to which nanotechnological interventions offer a great advantage. But how eminent could liposomes and their advantages be in superseding these existing therapeutic modalities? A solution is reflected in this review that draws attention to a decade-long journey embarked upon by researchers in this wake. This text is a comprehensive discussion of liposomes, the front runners of the drug delivery systems, and their active and passive targeting approaches for breast cancer management. Active targeting has been studied over the decade by many receptors overexpressed on the breast cancer cells and passive targeting with many drug combinations. The results converge on the fact that the actively targeted formulations exhibit a superior efficacy over their non-targeted counterparts and the all liposomal formulations are efficacious over the free drugs. This undoubtedly underlines the dominion of liposomal formulations over conventional chemotherapy. These investigations have led to the development of different liposomal formulations with active and passive targeting capacities that could be explored in depth. Acknowledging and getting a deeper insight into the liposomal evolution through time also unveiled many imperfections and unchartered territories that can be explored to deliver dexterous liposomal formulations against breast cancer and more in the clinical trial pipeline.
Collapse
Affiliation(s)
- Aliesha Moudgil
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Rajesh Salve
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Bhushan P Chaudhari
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India.
| |
Collapse
|
2
|
Bhattarai S, Mackeyev Y, Venkatesulu BP, Krishnan S, Singh PK. CXC chemokine receptor 4 (CXCR4) targeted gold nanoparticles potently enhance radiotherapy outcomes in breast cancer. NANOSCALE 2021; 13:19056-19065. [PMID: 34757363 PMCID: PMC9124374 DOI: 10.1039/d1nr05385j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) is overexpressed on most breast cancer cell surfaces including triple negative breast cancer (TNBC) which lacks traditional receptor overexpression. We targeted gold nanoparticles (GNPs) to this receptor via conjugation to anti-CXCR4 antibody (cGNPs). Irradiation of cells treated with cGNPs compared to PEGylated GNPs (pGNPs) resulted in more prominent radiosensitization of MDA-MB-231 cells with abundant CXCR4 overexpression than HTB-123 cells with moderate and MCF-7 cells with minimal CXCR4 overexpression. Overexpression of CXCR4 facilitated improved cellular internalization of cGNPs and irradiation of internalized cGNPs resulted in more unrepaired DNA double strand breaks and increased the production of oxygen free radicals compared to irradiation with non-internalized pGNPs. In a murine TNBC xenograft model, CXCR4 targeting potently increased tumor regrowth delay following radiation compared to radiation in the presence of pGNPs or vehicle alone. CXCR4 targeted GNPs enhance the efficacy of TNBC radiotherapy by increasing oxidative stress and DNA damage.
Collapse
Affiliation(s)
- Shanta Bhattarai
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Yuri Mackeyev
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA.
| | | | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA.
| | - Pankaj K Singh
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
3
|
Guo P, Busatto S, Huang J, Morad G, Moses MA. A facile magnetic extrusion method for preparing endosome-derived vesicles for cancer drug delivery. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2008326. [PMID: 34924915 PMCID: PMC8680268 DOI: 10.1002/adfm.202008326] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Indexed: 06/14/2023]
Abstract
To date, the scaled-up manufacturing and efficient drug loading of exosomes are two existing challenges limiting the clinical translation of exosome-based drug delivery. Herein, we developed a facile magnetic extrusion method for preparing endosome-derived vesicles, also known as exosome mimetics (EMs), which share the same biological origin and similar morphology, composition, and biofunctions with native exosomes. The high yield and consistency of this magnetic extrusion method help to overcome the manufacturing bottleneck in exosome research. Moreover, the proposed standardized multi-step method readily facilitates the ammonium sulfate gradient approach to actively load chemodrugs such as doxorubicin into EMs. The engineered EMs developed and tested here exhibit comparable drug delivery properties as do native exosomes and potently inhibit tumor growth by delivering doxorubicin in an orthotopic breast tumor model. These findings demonstrate that EMs can be prepared in a facile and scaled-up manner as a promising biological nanomedicine for cancer drug delivery.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Sara Busatto
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Jing Huang
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Golnaz Morad
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
- Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, United States
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
5
|
Chittasupho C, Aonsri C, Imaram W. Targeted dendrimers for antagonizing the migration and viability of NALM-6 lymphoblastic leukemia cells. Bioorg Chem 2021; 107:104601. [PMID: 33476870 DOI: 10.1016/j.bioorg.2020.104601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/18/2020] [Accepted: 12/24/2020] [Indexed: 01/08/2023]
Abstract
Acute lymphoblastic leukemia (ALL) or white blood cell cancer is one of the major causes that kills many children worldwide. Although various therapeutic agents are available for ALL treatment, the new drug discovery and drug delivery system are needed to improve their effectiveness, to reduce the toxicity and side-effect, and to enhance their selectivity to target cancer cells. CXCR4 is a protein expressed on the surface of various types of cancer cell including ALL. In this work, the CXCR4-targeted PAMAM dendrimer was constructed by conjugating G5 PAMAM with a CXCR4 antagonist, LFC131. The results revealed that the LFC131-conjugated G5 PAMAM selectively targeted CXCR4 expressing leukemic precursor B cells (NALM-6) and the migration of NALM-6 cells induced by SDF-1α was inhibited at non-cytotoxic concentration. Further research based on this findings may contribute to potential anti-metastatic drugs for lymphoblastic leukemia.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Mueang, Chiang Mai, Thailand
| | - Chaiyawat Aonsri
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Witcha Imaram
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand; Special Research Unit for Advanced Magnetic Resonance, Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand.
| |
Collapse
|
6
|
Kunjiappan S, Pavadai P, Vellaichamy S, Ram Kumar Pandian S, Ravishankar V, Palanisamy P, Govindaraj S, Srinivasan G, Premanand A, Sankaranarayanan M, Theivendren P. Surface receptor‐mediated targeted drug delivery systems for enhanced cancer treatment: A state‐of‐the‐art review. Drug Dev Res 2020; 82:309-340. [DOI: 10.1002/ddr.21758] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Selvaraj Kunjiappan
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy M.S. Ramaiah University of Applied Sciences Bengaluru Karnataka India
| | - Sivakumar Vellaichamy
- Department of Pharmaceutics Arulmigu Kalasalingam College of Pharmacy Krishnankoil Tamilnadu India
| | | | | | - Ponnusamy Palanisamy
- School of Mechanical Engineering Vellore Institute of Technology Vellore Tamilnadu India
| | - Saravanan Govindaraj
- Department of Pharmaceutical Chemistry MNR College of Pharmacy Sangareddy Telangana India
| | - Gowshiki Srinivasan
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | - Adhvitha Premanand
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | | | - Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry Swamy Vivekananda College of Pharmacy Elayampalayam, Namakkal Tamilnadu India
| |
Collapse
|
7
|
Advanced fluorescence microscopy reveals disruption of dynamic CXCR4 dimerization by subpocket-specific inverse agonists. Proc Natl Acad Sci U S A 2020; 117:29144-29154. [PMID: 33148803 PMCID: PMC7682396 DOI: 10.1073/pnas.2013319117] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Class A G protein−coupled receptors (GPCRs) can form dimers and oligomers via poorly understood mechanisms. We show here that the chemokine receptor CXCR4, which is a major pharmacological target, has an oligomerization behavior modulated by its active conformation. Combining advanced, single-molecule, and single-cell optical tools with functional assays and computational approaches, we unveil three key features of CXCR4 quaternary organization: CXCR4 dimerization 1) is dynamic, 2) increases with receptor expression level, and 3) can be disrupted by stabilizing an inactive receptor conformation. Ligand binding motifs reveal a ligand binding subpocket essential to modulate both CXCR4 basal activity and dimerization. This is relevant to develop new strategies to design CXCR4-targeting drugs. Although class A G protein−coupled receptors (GPCRs) can function as monomers, many of them form dimers and oligomers, but the mechanisms and functional relevance of such oligomerization is ill understood. Here, we investigate this problem for the CXC chemokine receptor 4 (CXCR4), a GPCR that regulates immune and hematopoietic cell trafficking, and a major drug target in cancer therapy. We combine single-molecule microscopy and fluorescence fluctuation spectroscopy to investigate CXCR4 membrane organization in living cells at densities ranging from a few molecules to hundreds of molecules per square micrometer of the plasma membrane. We observe that CXCR4 forms dynamic, transient homodimers, and that the monomer−dimer equilibrium is governed by receptor density. CXCR4 inverse agonists that bind to the receptor minor pocket inhibit CXCR4 constitutive activity and abolish receptor dimerization. A mutation in the minor binding pocket reduced the dimer-disrupting ability of these ligands. In addition, mutating critical residues in the sixth transmembrane helix of CXCR4 markedly diminished both basal activity and dimerization, supporting the notion that CXCR4 basal activity is required for dimer formation. Together, these results link CXCR4 dimerization to its density and to its activity. They further suggest that inverse agonists binding to the minor pocket suppress both dimerization and constitutive activity and may represent a specific strategy to target CXCR4.
Collapse
|
8
|
Guo P, Huang J, Moses-Gardner A, Smith ER, Moses MA. Quantitative Analysis of Different Cell Entry Routes of Actively Targeted Nanomedicines Using Imaging Flow Cytometry. Cytometry A 2019; 95:843-853. [PMID: 31294926 DOI: 10.1002/cyto.a.23848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/31/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
A rapid, high-throughput, and quantitative method for cell entry route characterization is still lacking in nanomedicine research. Here, we report the application of imaging flow cytometry for quantitatively analyzing cell entry routes of actively targeted nanomedicines. We first engineered ICAM1 antibody-directed fusogenic nanoliposomes (ICAM1-FusoNLPs) and ICAM1 antibody-directed endocytic nanolipogels (ICAM1-EndoNLGs) featuring highly similar surface properties but different cell entry routes: receptor-mediated membrane fusion and receptor-mediated endocytosis, respectively. By using imaging flow cytometry, we characterized their intracellular delivery into human breast cancer MDA-MB-231 cells. We found that ICAM1-FusoNLPs mediated a 2.8-fold increased cell uptake of fluorescent payload, FITC-dextran, with a 2.4-fold increased intracellular distribution area in comparison with ICAM1-EndoNLGs. We also investigated the effects of incubation time and endocytic inhibitors on the cell entry routes of ICAM1-FusoNLP and ICAM1-EndoNLG. Our results indicate that receptor-mediated membrane fusion is a faster and more efficient cell entry route than receptor-mediated endocytosis, bringing with it a significant therapeutic benefit in a proof-of-principle nanomedicine-mediated siRNA transfection experiment. Our studies suggest that cell entry route may be an important design parameter to be considered in the development of next-generation nanomedicines. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, 02115.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts, 02115
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, 02115.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts, 02115
| | - Alexander Moses-Gardner
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, 02115.,Department of Neurosurgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts, 02115
| | - Edward R Smith
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, 02115.,Department of Neurosurgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts, 02115
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, 02115.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts, 02115
| |
Collapse
|
9
|
Guo P, Moses-Gardner A, Huang J, Smith ER, Moses MA. ITGA2 as a potential nanotherapeutic target for glioblastoma. Sci Rep 2019; 9:6195. [PMID: 30996239 PMCID: PMC6470144 DOI: 10.1038/s41598-019-42643-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/28/2019] [Indexed: 01/14/2023] Open
Abstract
High grade gliomas, including glioblastoma (GBM), are the most common and deadly brain cancers in adults. Here, we performed a quantitative and unbiased screening of 70 cancer-related antigens using comparative flow cytometry and, for the first time, identified integrin alpha-2 (ITGA2) as a novel molecular target for GBM. In comparison to epidermal growth factor receptor (EGFR), a well-established GBM target, ITGA2 is significantly more expressed on human GBM cells and significantly less expressed on normal human glial cells. We also found that ITGA2 antibody blockade significantly impedes GBM cell migration but not GBM cell proliferation. To investigate the utility of ITGA2 as a therapeutic target in GBM, we designed and engineered an ITGA2 antibody-directed liposome that can selectively deliver doxorubicin, a standard-of-care chemotherapeutic agent, to GBM cells. This novel approach significantly improved antitumor efficacy. We also demonstrated that these ITGA2 antibody-directed liposomes can effectively breach the blood-brain tumor barrier (BBTB) in vitro via GBM-induced angiogenesis effects. These findings support further research into the use of ITGA2 as a novel nanotherapeutic target for GBM.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States
- Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Alexander Moses-Gardner
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States
- Department of Neurosurgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States
- Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Edward R Smith
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States.
- Department of Neurosurgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States.
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States.
- Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States.
| |
Collapse
|
10
|
Kim B, Pena CD, Auguste DT. Targeted Lipid Nanoemulsions Encapsulating Epigenetic Drugs Exhibit Selective Cytotoxicity on CDH1–/FOXM1+ Triple Negative Breast Cancer Cells. Mol Pharm 2019; 16:1813-1826. [DOI: 10.1021/acs.molpharmaceut.8b01065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Bumjun Kim
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, New York 10031, United States
| | - Caroline D. Pena
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, New York 10031, United States
| | - Debra T. Auguste
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, New York 10031, United States
| |
Collapse
|
11
|
Guo P, Yang J, Liu D, Huang L, Fell G, Huang J, Moses MA, Auguste DT. Dual complementary liposomes inhibit triple-negative breast tumor progression and metastasis. SCIENCE ADVANCES 2019; 5:eaav5010. [PMID: 30906868 PMCID: PMC6426465 DOI: 10.1126/sciadv.aav5010] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/31/2019] [Indexed: 05/10/2023]
Abstract
Distinguishing malignant cells from non-neoplastic ones is a major challenge in triple-negative breast cancer (TNBC) treatment. Here, we developed a complementary targeting strategy that uses precisely matched, multivalent ligand-receptor interactions to recognize and target TNBC tumors at the primary site and metastatic lesions. We screened a panel of cancer cell surface markers and identified intercellular adhesion molecule-1 (ICAM1) and epithelial growth factor receptor (EGFR) as optimal candidates for TNBC complementary targeting. We engineered a dual complementary liposome (DCL) that precisely complements the molecular ratio and organization of ICAM1 and EGFR specific to TNBC cell surfaces. Our in vitro mechanistic studies demonstrated that DCLs, compared to single-targeting liposomes, exhibited increased binding, enhanced internalization, and decreased receptor signaling. DCLs consistently exhibited substantially increased tumor targeting activity and antitumor efficacy in orthotopic and lung metastasis models, indicating that DCLs are a platform technology for the design of personalized nanomedicines for TNBC.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Jiang Yang
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Daxing Liu
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Lan Huang
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Gillian Fell
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jing Huang
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Debra T. Auguste
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| |
Collapse
|
12
|
Large DE, Soucy JR, Hebert J, Auguste DT. Advances in Receptor-Mediated, Tumor-Targeted Drug Delivery. ADVANCED THERAPEUTICS 2019; 2:1800091. [PMID: 38699509 PMCID: PMC11064891 DOI: 10.1002/adtp.201800091] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Receptor-mediated drug delivery presents an opportunity to enhance therapeutic efficiency by accumulating drug within the tissue of interest and reducing undesired, off-target effects. In cancer, receptor overexpression is a platform for binding and inhibiting pathways that shape biodistribution, toxicity, cell binding and uptake, and therapeutic function. This review will identify tumor-targeted drug delivery vehicles and receptors that show promise for clinical translation based on quantitative in vitro and in vivo data. The authors describe the rationale to engineer a targeted drug delivery vehicle based on the ligand, chemical conjugation method, and type of drug delivery vehicle. Recent advances in multivalent targeting and ligand organization on tumor accumulation are discussed. Revolutionizing receptor-mediated drug delivery may be leveraged in the therapeutic delivery of chemotherapy, gene editing tools, and epigenetic drugs.
Collapse
Affiliation(s)
- Danielle E Large
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Jacob Hebert
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Debra T Auguste
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| |
Collapse
|
13
|
Peptide density targets and impedes triple negative breast cancer metastasis. Nat Commun 2018; 9:2612. [PMID: 29973594 PMCID: PMC6031661 DOI: 10.1038/s41467-018-05035-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/11/2018] [Indexed: 12/27/2022] Open
Abstract
The C-X-C chemokine receptor type 4 (CXCR4, CD184) pathway is a key regulator of cancer metastasis. Existing therapeutics that block CXCR4 signaling are dependent on single molecule-receptor interactions or silencing CXCR4 expression. CXCR4 localizes in lipid rafts and forms dimers therefore CXCR4 targeting and signaling may depend on ligand density. Herein, we report liposomes presenting a CXCR4 binding peptide (DV1) as a three-dimensional molecular array, ranging from 9k to 74k molecules μm−2, target triple negative breast cancer (TNBC). TNBC cells exhibit a maxima in binding and uptake of DV1-functionalized liposomes (L-DV1) in vitro at a specific density, which yields a significant reduction in cell migration. This density inhibits metastasis from a primary tumor for 27 days, resulting from peptide density dependent gene regulation. We show that complementing cell membrane receptor expression may be a strategy for targeting cells and regulating signaling. The C-X-C chemokine receptor type 4 (CXCR4) pathway is a key regulator of cancer metastasis. Here, the authors present a method to block CXCR4 and thereby inhibit breast cancer metastasis by developing a liposome that presents CXCR4-binding peptides in a multivalent fashion.
Collapse
|
14
|
Guo P, Huang J, Zhao Y, Martin CR, Zare RN, Moses MA. Nanomaterial Preparation by Extrusion through Nanoporous Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703493. [PMID: 29468837 DOI: 10.1002/smll.201703493] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/09/2018] [Indexed: 05/20/2023]
Abstract
Template synthesis represents an important class of nanofabrication methods. Herein, recent advances in nanomaterial preparation by extrusion through nanoporous membranes that preserve the template membrane without sacrificing it, which is termed as "non-sacrificing template synthesis," are reviewed. First, the types of nanoporous membranes used in nanoporous membrane extrusion applications are introduced. Next, four common nanoporous membrane extrusion strategies: vesicle extrusion, membrane emulsification, precipitation extrusion, and biological membrane extrusion, are examined. These methods have been utilized to prepare a wide range of nanomaterials, including liposomes, emulsions, nanoparticles, nanofibers, and nanotubes. The principle and historical context of each specific technology are discussed, presenting prominent examples and evaluating their positive and negative features. Finally, the current challenges and future opportunities of nanoporous membrane extrusion methods are discussed.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Yaping Zhao
- School of Chemistry and Chemical Engineering, Shanghai Jiaotong University, 800 Dongchuan road, Shanghai, 200240, China
| | - Charles R Martin
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL, 32611, USA
| | - Richard N Zare
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA, 94305, USA
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| |
Collapse
|
15
|
Abstract
To date, the role of elasticity in drug delivery remains elusive due to the inability to measure microscale mechanics and alter rheology without affecting chemistry. Herein, we describe the in vitro cellular uptake and in vivo tumor uptake of nanolipogels (NLGs). NLGs are composed of identical lipid bilayers encapsulating an alginate core, with tunable elasticity. The elasticity of NLGs was evaluated by atomic force microscopy, which demonstrated that they exhibit Young’s moduli ranging from 45 ± 9 to 19,000 ± 5 kPa. Neoplastic and non-neoplastic cells exhibited significantly greater uptake of soft NLGs (Young’s modulus <1.6 MPa) relative to their elastic counterparts (Young’s modulus >13.8 MPa). In an orthotopic breast tumor model, soft NLGs accumulated significantly more in tumors, whereas elastic NLGs preferentially accumulated in the liver. Our findings demonstrate that particle elasticity directs tumor accumulation, suggesting that it may be a design parameter to enhance tumor delivery efficiency. Nanoparticle elasticity is thought to play an important role in drug delivery, but is little studied. Here, the authors use nanolipogels with tunable moduli to study the effect of particle elasticity on in vitro cellular uptake and in vivo tumor uptake, finding that stiffer particles are not as easily internalized.
Collapse
|
16
|
Guo P, Yang J, Bielenberg DR, Dillon D, Zurakowski D, Moses MA, Auguste DT. A quantitative method for screening and identifying molecular targets for nanomedicine. J Control Release 2017; 263:57-67. [PMID: 28341549 DOI: 10.1016/j.jconrel.2017.03.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/10/2017] [Accepted: 03/17/2017] [Indexed: 12/11/2022]
Abstract
Identifying a molecular target is essential for tumor-targeted nanomedicine. Current cancer nanomedicines commonly suffer from poor tumor specificity, "off-target" toxicity, and limited clinical efficacy. Here, we report a method to screen and identify new molecular targets for tumor-targeted nanomedicine based on a quantitative analysis. In our proof-of-principle study, we used comparative flow cytometric screening to identify ICAM-1 as a potential target for metastatic melanoma (MM). We further evaluated ICAM-1 as a MM targeting moiety by characterizing its (1) tumor specificity, (2) expression level, (3) cellular internalization, (4) therapeutic function, and (5) potential clinical impact. Quantitation of ICAM-1 protein expression on cells and validation by immunohistochemistry on human tissue specimens justified the synthesis of antibody-functionalized drug delivery vehicles, which were benchmarked against appropriate controls. We engineered ICAM-1 antibody conjugated, doxorubicin encapsulating immunoliposomes (ICAM-Dox-LPs) to selectively recognize and deliver doxorubicin to MM cells and simultaneously neutralize ICAM-1 signaling via an antibody blockade, demonstrating significant and simultaneous inhibitory effects on MM cell proliferation and migration. This paper describes a novel, quantitative metric system that identifies and evaluates new cancer targets for tumor-targeting nanomedicine.
Collapse
Affiliation(s)
- Peng Guo
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, United States; Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA 02115, United States; Department of Surgery, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Jiang Yang
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA 02115, United States; Department of Surgery, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Diane R Bielenberg
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA 02115, United States; Department of Surgery, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States
| | - David Zurakowski
- Department of Anesthesia, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA 02115, United States; Department of Surgery, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Debra T Auguste
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, United States.
| |
Collapse
|
17
|
Guo P, Huang J, Moses MA. Characterization of dormant and active human cancer cells by quantitative phase imaging. Cytometry A 2017; 91:424-432. [PMID: 28314083 DOI: 10.1002/cyto.a.23083] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 01/14/2023]
Abstract
The switch of tumor cells from a dormant, non-angiogenic phenotype to an active, angiogenic phenotype is a critical step in early cancer progression. To date, relatively little is known about the cellular behaviors of angiogenic and non-angiogenic tumor cell phenotypes. In this study, holographic imaging cytometry, a quantitative phase imaging (QPI) technique was used to continuously and non-invasively analyze, quantify, and compare a panel of fundamental cellular behaviors of angiogenic and non-angiogenic human osteosarcoma cells (KHOS) in a simple and economical way. Results revealed that angiogenic KHOS cells (KHOS-A) have significantly higher cell motility speeds than their non-angiogenic counterpart (KHOS-N) while no difference in their cell proliferation rates and cell cycle lengths were observed. KHOS-A cells were also found to have significantly smaller cell areas and greater cell optical thicknesses when compared with the non-angiogenic KHOS-N cells. No difference in average cell volumes was observed. These studies demonstrate that the morphology and behavior of angiogenic and non-angiogenic cells can be continuously, efficiently, and non-invasively monitored using a simple, quantitative, and economical system that does not require tedious and time-consuming assays to provide useful information about tumor dormancy. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115
| |
Collapse
|
18
|
Tao Y, Li M, Kim B, Auguste DT. Incorporating gold nanoclusters and target-directed liposomes as a synergistic amplified colorimetric sensor for HER2-positive breast cancer cell detection. Am J Cancer Res 2017; 7:899-911. [PMID: 28382162 PMCID: PMC5381252 DOI: 10.7150/thno.17927] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/03/2016] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the second leading cause of cancer-related mortality in women. Successful development of sensitive nanoprobes for breast cancer cell detection is of great importance for breast cancer diagnosis and symptomatic treatment. Herein, inspired by the intrinsic peroxidase property of gold nanoclusters, high loading, and targeting ability of ErbB2/Her2 antibody functionalized liposomes, we report that gold nanoclusters-loaded, target-directed, functionalized liposomes can serve as a robust sensing platform for amplified colorimetric detection of HER2-positive breast cancer cells. This approach allows HER2-positive breast cancer cell identification at high sensitivity with high selectivity. In addition, the colorimetric “readout” offers extra advantages in terms of low-cost, portability, and easy-to-use applications. The practicality of this platform was further proved by successful detection of HER2-positive breast cancer cells in human serum samples and in breast cancer tissue, which indicated our proposed method has potential for application in cancer theranostics.
Collapse
|
19
|
Ayesa U, Gray BD, Pak KY, Chong PLG. Liposomes Containing Lipid-Soluble Zn(II)-Bis-dipicolylamine Derivatives Show Potential To Be Targeted to Phosphatidylserine on the Surface of Cancer Cells. Mol Pharm 2016; 14:147-156. [PMID: 28043132 DOI: 10.1021/acs.molpharmaceut.6b00760] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Here we used a lipid-soluble Zn(II)-bis-dipicolylamine derivative as a membrane component to develop liposomal carriers that have potential to be targeted to phosphatidylserine (PS) rich surfaces on cancer cells and to preferentially kill cancer cells without using anticancer drugs. This DPA derivative (abbreviated as DPA-Cy3[22,22]) contains the fluorophore cyanine 3 (Cy3) and two 22-carbon chains that can be anchored into liposomal membrane bilayers. DPA-Cy3[22,22]/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) unilamellar vesicles (∼150 nm) showed selective binding to PS-containing liposomes as demonstrated by anion exchange chromatography. This binding does not result in vesicle fusion or aggregation. Flow cytometry showed that DPA-Cy3[22,22]/POPC liposomes have preferential binding to MCF-7 breast cancer cells over MCF-12A noncancer cells due to 3-7 times more PS exposures on MCF-7. The extent of liposome binding with MCF-7 cells was increased by two times after cells were pretreated with the apoptotic inducer camptothecin, which increased PS exposure to the cell surface. Moreover, our flow cytometry data also suggest that local cell membrane perturbations may occur upon liposome binding and internalization. This implies that DPA-Cy3[22,22]/POPC liposomes alone may have a PS-dependent cytotoxic effect. This assertion was supported by the cell proliferation assay, which showed that 9.1 mol % DPA-Cy3[22,22]/POPC liposomes exert cytotoxicity on MCF-7 cells 3.5 times higher than that on MCF-12A cells. These results indicate that DPA-Cy3[22,22]-containing liposomes hold great promise as efficient nano drug carriers.
Collapse
Affiliation(s)
- Umme Ayesa
- Department of Medical Genetics and Molecular Biochemistry, The Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania 19140, United States
| | - Brian D Gray
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Parkson Lee-Gau Chong
- Department of Medical Genetics and Molecular Biochemistry, The Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
20
|
Akhtar N, Khan RA. Liposomal systems as viable drug delivery technology for skin cancer sites with an outlook on lipid-based delivery vehicles and diagnostic imaging inputs for skin conditions'. Prog Lipid Res 2016; 64:192-230. [DOI: 10.1016/j.plipres.2016.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/15/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022]
|
21
|
Boonstra MC, de Geus SWL, Prevoo HAJM, Hawinkels LJAC, van de Velde CJH, Kuppen PJK, Vahrmeijer AL, Sier CFM. Selecting Targets for Tumor Imaging: An Overview of Cancer-Associated Membrane Proteins. BIOMARKERS IN CANCER 2016; 8:119-133. [PMID: 27721658 PMCID: PMC5040425 DOI: 10.4137/bic.s38542] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/30/2022]
Abstract
Tumor targeting is a booming business: The global therapeutic monoclonal antibody market accounted for more than $78 billion in 2012 and is expanding exponentially. Tumors can be targeted with an extensive arsenal of monoclonal antibodies, ligand proteins, peptides, RNAs, and small molecules. In addition to therapeutic targeting, some of these compounds can also be applied for tumor visualization before or during surgery, after conjugation with radionuclides and/or near-infrared fluorescent dyes. The majority of these tumor-targeting compounds are directed against cell membrane-bound proteins. Various categories of targetable membrane-bound proteins, such as anchoring proteins, receptors, enzymes, and transporter proteins, exist. The functions and biological characteristics of these proteins determine their location and distribution on the cell membrane, making them more, or less, accessible, and therefore, it is important to understand these features. In this review, we evaluate the characteristics of cancer-associated membrane proteins and discuss their overall usability for cancer targeting, especially focusing on imaging applications.
Collapse
Affiliation(s)
- Martin C Boonstra
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Susanna W L de Geus
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Lukas J A C Hawinkels
- Department of Gastroenterology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.; Antibodies for Research Applications BV, Gouda, the Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.; Antibodies for Research Applications BV, Gouda, the Netherlands
| |
Collapse
|
22
|
Ieranò C, Portella L, Lusa S, Salzano G, D'Alterio C, Napolitano M, Buoncervello M, Macchia D, Spada M, Barbieri A, Luciano A, Barone MV, Gabriele L, Caraglia M, Arra C, De Rosa G, Scala S. CXCR4-antagonist Peptide R-liposomes for combined therapy against lung metastasis. NANOSCALE 2016; 8:7562-7571. [PMID: 26983756 DOI: 10.1039/c5nr06335c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The chemokine CXCL12 activates CXCR4, initiating multiple pathways that control immune cell trafficking, angiogenesis and embryogenesis; CXCR4 is also overexpressed in multiple tumors affecting metastatic dissemination. While there has been great enthusiasm for exploiting the CXCR4-CXCL12 axis as a target in cancer therapy, to date the promise has yet to be fulfilled. A new class of CXCR4-antagonist cyclic peptides was recently developed and the compound named Peptide R was identified as the most active. With the intent to improve the efficacy and biodistribution of Peptide R, stealth liposomes decorated with Peptide R were developed (PL-Peptide R). In vitro PL-Peptide R efficiently inhibited CXCR4-dependent migration and in vivo it significantly reduced lung metastases and increased overall survival in B16-CXCR4 injected C57BL/6 mice. To evaluate if PL-Peptide R could also be a drug delivery system for CXCR4 expressing tumors, the PL-Peptide R was loaded with doxorubicin (DOX) (PL-Peptide R-DOX). PL-Peptide R-DOX efficiently delivered DOX to CXCR4 expressing cell lines with a consequent decrease in the DOX IC50 efficient dose. In vivo, B16-CXCR4 injected C57BL/6 mice treated with PL-Peptide R-DOX developed fewer lung metastases compared to PL-DOX treated mice. This work provides the proof-of-concept to prevent metastasis by using combined nanomedicine.
Collapse
Affiliation(s)
- Caterina Ieranò
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Luigi Portella
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Sara Lusa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Giuseppina Salzano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy. and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Ave, Boston, MA, USA
| | - Crescenzo D'Alterio
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Maria Napolitano
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Maria Buoncervello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Daniele Macchia
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Massimo Spada
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Maria Vittoria Barone
- Department of Translational Medical Science and European Laboratory for the Investigation of Food Induced Disease (ELFID), University of Naples, Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Lucia Gabriele
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138, Naples, Italy
| | - Claudio Arra
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Stefania Scala
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| |
Collapse
|
23
|
SARI , a novel target gene of glucocorticoid receptor, plays an important role in dexamethasone-mediated killing of B lymphoma cells. Cancer Lett 2016; 373:57-66. [DOI: 10.1016/j.canlet.2016.01.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 01/17/2016] [Accepted: 01/18/2016] [Indexed: 12/18/2022]
|
24
|
Chen WL, Yuan ZQ, Liu Y, Yang SD, Zhang CG, Li JZ, Zhu WJ, Li F, Zhou XF, Lin YM, Zhang XN. Liposomes coated with N-trimethyl chitosan to improve the absorption of harmine in vivo and in vitro. Int J Nanomedicine 2016; 11:325-36. [PMID: 26855571 PMCID: PMC4725628 DOI: 10.2147/ijn.s95540] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In this study, harmine liposomes (HM-lip) were prepared through the thin-film hydration-pH-gradient method and then coated with N-trimethyl chitosan (TMC). Particle size, zeta potential, entrapment efficiency, and in vitro release of HM-lip and TMC-coated harmine liposomes (TMC-HM-lip) were also determined. Sprague Dawley rats were further used to investigate the pharmacokinetics in vivo. Retention behavior in mouse gastrointestinal tract (GIT) was studied through high-performance liquid chromatography and near-infrared imaging. Degradation was further evaluated through incubation with Caco-2 cell homogenates, and a Caco-2 monolayer cell model was used to investigate the uptake and transport of drugs. HM-lip and TMC-HM-lip with particle size of 150-170 nm, an entrapment efficiency of about 81%, and a zeta potential of negative and positive, respectively, were prepared. The release of HM from HM-lip and TMC-HM-lip was slower than that from HM solution and was sensitive to pH. TMC-HM-lip exhibited higher oral bioavailability and had prolonged retention time in GIT. HM-lip and TMC-HM-lip could also protect HM against degradation in Caco-2 cell homogenates. The uptake amount of TMC-HM-lip was higher than that of HM and HM-lip. TMC-HM-lip further demonstrated higher apparent permeability coefficient (P(app)) from the apical to the basolateral side than HM and HM-lip because of its higher uptake and capability to open tight junctions in the cell monolayers. TMC-HM-lip can prolong the retention time in the GIT, protect HM against enzyme degradation, and improve transport across Caco-2 cell monolayers, thus enhancing the oral bioavailability of HM.
Collapse
Affiliation(s)
- Wei-Liang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Zhi-Qiang Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yang Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Shu-di Yang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Chun-ge Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Ji-Zhao Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Wen-jing Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Fang Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Xiao-feng Zhou
- Department of Radiobiology, College of Radiological Medicine and Protection, Soochow University, Suzhou, People's Republic of China; Changshu Hospital of Traditional Chinese Medicine, Changshu, People's Republic of China
| | - Yi-mei Lin
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Xue-nong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
25
|
Abstract
This review discusses the potential of CXCR4 chemokine receptor in the design of anticancer and antimetastatic drug delivery systems. The role of CXCR4 in cancer progression and metastasis is discussed in the context of the development of several types of drug delivery strategies. Overview of drug delivery systems targeted to cancers that overexpress CXCR4 is provided, together with the main types of CXCR4-binding ligands used in targeting applications. Drug delivery applications that take advantage of CXCR4 inhibition to achieve enhanced anticancer and antimetastatic activity of combination treatments are also discussed.
Collapse
Affiliation(s)
- Yan Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA ; Department of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
26
|
Guo P, Yang J, Jia D, Moses MA, Auguste DT. ICAM-1-Targeted, Lcn2 siRNA-Encapsulating Liposomes are Potent Anti-angiogenic Agents for Triple Negative Breast Cancer. Theranostics 2016; 6:1-13. [PMID: 26722369 PMCID: PMC4679350 DOI: 10.7150/thno.12167] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/16/2015] [Indexed: 12/21/2022] Open
Abstract
Lipocalin 2 (Lcn2) is a promising therapeutic target as well as a potential diagnostic biomarker for breast cancer. It has been previously shown to promote breast cancer progression by inducing the epithelial to mesenchymal transition in breast cancer cells as well as by enhancing angiogenesis. Lcn2 levels in urine and tissue samples of breast cancer patients has also been correlated with breast cancer status and poor patient prognosis. In this study, we have engineered a novel liposomal small interfering RNA (siRNA) delivery system to target triple negative breast cancer (TNBC) via a recently identified molecular target, intercellular adhesion molecule-1 (ICAM-1). This ICAM-1-targeted, Lcn2 siRNA- encapsulating liposome (ICAM-Lcn2-LP) binds human TNBC MDA-MB-231cells significantly stronger than non-neoplastic MCF-10A cells. Efficient Lcn2 knockdown by ICAM-Lcn2-LPs led to a significant reduction in the production of vascular endothelial growth factor (VEGF) from MDA-MB-231 cells, which, in turn, led to reduced angiogenesis both in vitro and in vivo. Angiogenesis (neovascularization) is a requirement for solid tumor growth and progression, and its inhibition is an important therapeutic strategy for human cancers. Our results indicate that a tumor-specific strategy such as the TNBC-targeted, anti-angiogenic therapeutic approach developed here, may be clinically useful in inhibiting TNBC progression.
Collapse
Affiliation(s)
- Peng Guo
- 1. Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, United States
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Jiang Yang
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Di Jia
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Marsha A. Moses
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Debra T. Auguste
- 1. Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, United States
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
27
|
Khan DR, Webb MN, Cadotte TH, Gavette MN. Use of Targeted Liposome-based Chemotherapeutics to Treat Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:1-5. [PMID: 26309409 PMCID: PMC4533644 DOI: 10.4137/bcbcr.s29421] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 11/05/2022]
Abstract
The use of nanocarriers such as liposomes to deliver anticancer drugs to tumors can significantly enhance the therapeutic index of otherwise unencapsulated cytotoxic agents. This is in part because of the fact that the phospholipid bilayer can protect healthy sensitive tissue from the damaging effects of these types of drugs. Furthermore, the ease with which the phospholipid bilayer surface can be modified to allow for polyethylene glycol incorporation resulting in pegylated liposomes allow for increased circulation times in vivo, and thus an overall increase in the concentration of the drug delivered to the tumor site. This explains the clinical success of the liposomal-based drug Doxil, which has proven to be quite efficacious in the treatment of breast cancer. However, significant challenges remain involving poor drug transfer between the liposome and tumor cells with this type of nontargeted drug delivery system. Thus, future work involves the development of "smart" drugs, or targeted drug delivery intended for improved colocalization between the drug and cancerous cells. While it is not possible to entirely discuss such a rapidly growing field of study involving many different types of chemotherapeutics here, in this review, we discuss some of the recent advancements involving the development of targeted liposome-based chemotherapeutics to treat breast cancer.
Collapse
Affiliation(s)
- David R Khan
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| | - Maggie N Webb
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| | - Thomas H Cadotte
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| | - Madison N Gavette
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| |
Collapse
|
28
|
Abstract
Triple negative breast cancers (TNBCs) have a high mortality rate owing to aggressive proliferation and metastasis and a lack of effective therapeutic options. Herein, we describe the overexpression of intercellular adhesion molecule-1 (ICAM-1) in human TNBC cell lines and tissues, and demonstrate that ICAM-1 is a potential molecular target and biomarker for TNBC therapy and diagnosis. We synthesized ICAM-1 antibody-conjugated iron oxide nanoparticles (ICAM-IONPs) as a magnetic resonance imaging (MRI) probe to evaluate tumor targeting. Quantitative analysis of ICAM-1 surface expression predicted the targeting capability of ICAM-IONPs to TNBC cells. MRI of the TNBC xenograft tumor after systemic administration of ICAM-IONPs, coupled with iron quantification and histology, demonstrated a significant and sustained MRI contrast enhancement and probe accumulation in tumors with ICAM-1 overexpression relative to control. Identification of ICAM-1 as a TNBC target and biomarker may lead to the development of a new strategy and platform for addressing a critical gap in TNBC patient care.
Collapse
|
29
|
Li J, Jiang K, Qiu X, Li M, Hao Q, Wei L, Zhang W, Chen B, Xin X. Overexpression of CXCR4 is significantly associated with cisplatin-based chemotherapy resistance and can be a prognostic factor in epithelial ovarian cancer. BMB Rep 2014; 47:33-8. [PMID: 24209634 PMCID: PMC4163846 DOI: 10.5483/bmbrep.2014.47.1.069] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 04/27/2013] [Accepted: 05/28/2013] [Indexed: 11/24/2022] Open
Abstract
The chemokine receptor 4 (CXCR4) plays an important role in the growth, angiogenesis and metastasis of various cancers, including epithelial ovarian cancer (EOC). However, the correlation between CXCR4 and the clinical response of EOC patients to chemotherapy remains unknown. 124 EOC patients were recruited to assess the relationship between CXCR4 and the response to cisplatin-based chemotherapy. The results showed that patients with a higher CXCR4 expression had a significantly lower chemosensitivity, a poorer progression-free survival and a lower overall survival than those with lower CXCR4 expression. In addition, knockdown of CXCR4 by small interfering RNA suppressed cell proliferation and resulted in G1/S arrest, increased apoptosis and chemosensitivity in both cisplatin-sensitive A2780 cells and cisplatin-resistant cell A2780/cis in vitro. Our data suggest that CXCR4 is one of the key molecules in cisplatin-based chemotherapy for EOC patients and that CXCR4 inhibition is a potential strategy to address the chemoresistance of EOC. [BMB Reports 2014; 47(1): 33-38]
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - BiLiang Chen
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xiaoyan Xin
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
30
|
Nazli C, Demirer GS, Yar Y, Acar HY, Kizilel S. Targeted delivery of doxorubicin into tumor cells via MMP-sensitive PEG hydrogel-coated magnetic iron oxide nanoparticles (MIONPs). Colloids Surf B Biointerfaces 2014; 122:674-683. [PMID: 25183059 DOI: 10.1016/j.colsurfb.2014.07.049] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 01/03/2023]
Abstract
Targeting tumors with nano-scale delivery systems shows promise to improve the therapeutic effects of chemotherapeutic drugs. However, the limited specificity of current nano-scale systems for cancer tissues prevents realization of their full clinical potential. Here, we demonstrate an effective approach to creating as targeted nanocarriers for drug delivery: MIONPs coated with integrin-targeted and matrix-metalloproteinase (MMP)-sensitive PEG hydrogel scaffolds. The functional PEG hydrogel coating has been designed for active loading as well as triggered intra-cellular release of the cancer therapeutic agent doxorubicin (DOX). Our study demonstrated that coated nanocarriers could be taken into cancer cells 11 times more efficiently than uncoated ones. Furthermore, confocal laser scanning microscopy images revealed that these targeted nanocarriers could efficiently deliver and release DOX into the nuclei of HeLa cells within 2h. Coating MIONPs with multifunctional PEG hydrogel could be a promising alternative to existing vehicles for targeted delivery of DOX into tumor tissue.
Collapse
Affiliation(s)
- Caner Nazli
- Koç University, Material Science and Engineering, Istanbul 34450, Turkey
| | - Gozde S Demirer
- Koç University, Chemical and Biological Engineering, Istanbul 34450, Turkey
| | - Yasemin Yar
- Koç University, Material Science and Engineering, Istanbul 34450, Turkey
| | - H Yagci Acar
- Koç University, Material Science and Engineering, Istanbul 34450, Turkey; Koç University, Chemistry Department, Istanbul 34450, Turkey
| | - Seda Kizilel
- Koç University, Material Science and Engineering, Istanbul 34450, Turkey; Koç University, Chemical and Biological Engineering, Istanbul 34450, Turkey.
| |
Collapse
|
31
|
Guo P, You JO, Yang J, Jia D, Moses MA, Auguste DT. Inhibiting metastatic breast cancer cell migration via the synergy of targeted, pH-triggered siRNA delivery and chemokine axis blockade. Mol Pharm 2014; 11:755-65. [PMID: 24467226 PMCID: PMC3993942 DOI: 10.1021/mp4004699] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Because breast cancer patient survival inversely correlates with metastasis, we engineered vehicles to inhibit both the C-X-C chemokine receptor type 4 (CXCR4) and lipocalin-2 (Lcn2) mediated migratory pathways. pH-responsive liposomes were designed to protect and trigger the release of Lcn2 siRNA. Liposomes were modified with anti-CXCR4 antibodies to target metastatic breast cancer (MBC) cells and block migration along the CXCR4-CXCL12 axis. This synergistic approach--coupling the CXCR4 axis blockade with Lcn2 silencing--significantly reduced migration in triple-negative human breast cancer cells (88% for MDA-MB-436 and 92% for MDA-MB-231). The results suggested that drug delivery vehicles engineered to attack multiple migratory pathways may effectively slow progression of MBC.
Collapse
Affiliation(s)
- Peng Guo
- Department of Biomedical Engineering, The City College of New York , 160 Convent Avenue, New York, New York 10031, United States
| | | | | | | | | | | |
Collapse
|
32
|
Gew LT, Misran M. Albumin-fatty acid interactions at monolayer interface. NANOSCALE RESEARCH LETTERS 2014; 9:218. [PMID: 24910574 PMCID: PMC4029981 DOI: 10.1186/1556-276x-9-218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/20/2014] [Indexed: 05/22/2023]
Abstract
The fluid mosaic model of Singer and Nicolson in 1972 shows how proteins are embedded in membranes. To elucidate the interactions between proteins and the surrounding lipids, stearic acid (SA) and bovine serum albumin (BSA) were used as lipid-protein components to mimic the normal membrane bilayer environment using the Langmuir-Blodgett technique. Surface pressure (π)-molecular area (A) isotherms were recorded for the SA monolayer in the presence of BSA on water. The mixed monolayer was successfully transferred onto an oxidized silicon wafer and imaged by tapping mode atomic force microscopy (AFM). Miscibility, compressibility and thermodynamic stability of the mixed system were examined. A large negative deviation of A ex, together with the minimum value of ΔG ex, was observed when the mole fraction of BSA (X BSA) was 0.8, indicating this to be the most stable mixture. In a compressibility analysis, X BSA was observed at below 50 mN m(-1), denoting a liquid-expanded phase and showing the occurrence of a strong interaction of SA with BSA molecules in this phase. AFM observations supported the quantitative data indicating that BSA was strongly attracted onto the membrane surface as predicted.
Collapse
Affiliation(s)
- Lai Ti Gew
- Department of Biological Sciences, Faculty of Science and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, Petaling Jaya, Selangor 46150, Malaysia
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Misni Misran
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
33
|
Liu Y, Gao FP, Zhang D, Fan YS, Chen XG, Wang H. Molecular structural transformation regulated dynamic disordering of supramolecular vesicles as pH-responsive drug release systems. J Control Release 2013; 173:140-7. [PMID: 24188958 DOI: 10.1016/j.jconrel.2013.10.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/02/2013] [Accepted: 10/26/2013] [Indexed: 01/19/2023]
Abstract
The spontaneous release of drug payloads in the whole body always results in the compromised drug bioavailability and ultimate therapeutic efficacy. To achieve enhanced therapeutic efficacy and reduced side effects, pH-responsive targeted drug delivery systems have been studied due to their enhanced tumor accumulation and controllable maximum drug release feature. The present study described a co-assembly constructed by a pH responsive molecule (i.e., malachite green carbinol base (MG)) and liposome for highly efficient doxorubicin (DOX) release in tumor cells (MG-DOX⊂L). The structural transformation of MG effectively regulates the drug release profile in acidic environment. The pH-responsive sensitivity of co-assembly can be fine-tuned by altering the mixing ratios of building blocks with pH responders (i.e., MG molecules). MG-DOX⊂L was beneficial for the DOX release at pH5.0 and showed a higher cytotoxicity in KB cells owing to the pH-responsive drug release in acidic organelles following endocytosis pathway. In vivo tumor targetability and growth inhibition were evaluated in KB cell-xenografted nude mice. We have demonstrated that effective tumor growth inhibition in vivo is attributed to the synergistic contributions from highly efficient cellular entry and responsive intracellular release of DOX from MG-DOX⊂L.
Collapse
Affiliation(s)
- Ya Liu
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing, China; College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao, China
| | - Fu-Ping Gao
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing, China
| | - Di Zhang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing, China
| | - Yun-Shan Fan
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing, China
| | - Xi-Guang Chen
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao, China.
| | - Hao Wang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing, China.
| |
Collapse
|
34
|
Jay SM, Murthy AC, Hawkins JF, Wortzel JR, Steinhauser ML, Alvarez LM, Gannon J, Macrae CA, Griffith LG, Lee RT. An engineered bivalent neuregulin protects against doxorubicin-induced cardiotoxicity with reduced proneoplastic potential. Circulation 2013; 128:152-61. [PMID: 23757312 DOI: 10.1161/circulationaha.113.002203] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Doxorubicin (DOXO) is an effective anthracycline chemotherapeutic, but its use is limited by cumulative dose-dependent cardiotoxicity. Neuregulin-1β is an ErbB receptor family ligand that is effective against DOXO-induced cardiomyopathy in experimental models but is also proneoplastic. We previously showed that an engineered bivalent neuregulin-1β (NN) has reduced proneoplastic potential in comparison with the epidermal growth factor-like domain of neuregulin-1β (NRG), an effect mediated by receptor biasing toward ErbB3 homotypic interactions uncommonly formed by native neuregulin-1β. Here, we hypothesized that a newly formulated, covalent NN would be cardioprotective with reduced proneoplastic effects in comparison with NRG. METHODS AND RESULTS NN was expressed as a maltose-binding protein fusion in Escherichia coli. As established previously, NN stimulated antineoplastic or cytostatic signaling and phenotype in cancer cells, whereas NRG stimulated proneoplastic signaling and phenotype. In neonatal rat cardiomyocytes, NN and NRG induced similar downstream signaling. NN, like NRG, attenuated the double-stranded DNA breaks associated with DOXO exposure in neonatal rat cardiomyocytes and human cardiomyocytes derived from induced pluripotent stem cells. NN treatment significantly attenuated DOXO-induced decrease in fractional shortening as measured by blinded echocardiography in mice in a chronic cardiomyopathy model (57.7±0.6% versus 50.9±2.6%, P=0.004), whereas native NRG had no significant effect (49.4±3.7% versus 50.9±2.6%, P=0.813). CONCLUSIONS NN is a cardioprotective agent that promotes cardiomyocyte survival and improves cardiac function in DOXO-induced cardiotoxicity. Given the reduced proneoplastic potential of NN versus NRG, NN has translational potential for cardioprotection in patients with cancer receiving anthracyclines.
Collapse
Affiliation(s)
- Steven M Jay
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
You JO, Guo P, Auguste DT. A drug-delivery vehicle combining the targeting and thermal ablation of HER2+ breast-cancer cells with triggered drug release. Angew Chem Int Ed Engl 2013; 52:4141-6. [PMID: 23494862 PMCID: PMC5540660 DOI: 10.1002/anie.201209804] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/05/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Jin-Oh You
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | | | | |
Collapse
|
36
|
You JO, Guo P, Auguste DT. A Drug-Delivery Vehicle Combining the Targeting and Thermal Ablation of HER2+ Breast-Cancer Cells with Triggered Drug Release. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201209804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|