1
|
Lotter C, Stierli MA, Puligilla RD, Huwyler J. Dual targeted lipid nanoparticles for enhanced DNA delivery and transfection of breast cancer cells. Eur J Pharm Biopharm 2025; 209:114674. [PMID: 39988265 DOI: 10.1016/j.ejpb.2025.114674] [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: 12/20/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Lipid nanoparticles (LNPs) have gained much attention as non-viral gene delivery systems due to their large payload capacity, reduced immunogenicity, and cost-effective manufacturing. Surface modification of LNPs by covalent attachment of receptor ligands can improve their tissue specificity and reduce off-target effects. In the present work, DNA-LNPs were therefore designed to target breast cancer, particularly the invasive HER2-positive subtype. Targeting was mediated by trastuzumab (Herceptin®) a monoclonal antibody binding to the extracellular domain of the human epidermal growth factor receptor protein (HER2). To overcome intrinsic trastuzumab resistance for some patients with HER2 positive breast cancer, a dual-targeting strategy was employed by combining Herceptin with folate to enhance LNP uptake by cancer cells. Dual-targeted LNPs encapsulating plasmid DNA, coding for a fluorescent reporter protein (tdTomato or EGFP), were prepared using folate-conjugated PEGylated lipids. Subsequently, thiolated Herceptin was conjugated to the surface of the LNPs. At an N/P ratio of 6, small and uniform targeted LNPs were obtained, with a slightly negative ζ-potential. Cellular uptake and transgene expression were characterized invitro using three breast cancer cell lines (MCF7, MDA-mb453, SKBR3), which express varying level of the HER2 receptor. Cellular uptake correlated with HER2 expression levels and was significantly increased when Herceptin was combined with folate. In all tested breast cancer cell lines, dual-targeted LNPs led to an enhanced transgene expression compared to single-targeted LNPs. Furthermore, invivo zebrafish xenograft studies confirmed superior targeting and transfection efficiency of Dual-LNPs under physiological conditions. Our findings highlight the superior performance of dual-targeted LNPs to deliver a DNA expression plasmid to HER2 positive breast cancer cells, emphasizing their potential as an improved targeting and transfection strategy.
Collapse
Affiliation(s)
- Claudia Lotter
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Megan Anna Stierli
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Ramya Deepthi Puligilla
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland.
| |
Collapse
|
2
|
Choi AS, Moon TJ, Abuhashim W, Bhalotia A, Qian H, Paulsen KE, Lorkowski M, Ndamira C, Gopalakrishnan R, Krishnamurthy A, Schiemann WP, Karathanasis E. Can targeted nanoparticles distinguish cancer metastasis from inflammation? J Control Release 2023; 362:812-819. [PMID: 37011838 PMCID: PMC10548349 DOI: 10.1016/j.jconrel.2023.03.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/11/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Targeting ligands have been widely used to increase the intratumoral accumulation of nanoparticles and their uptake by cancer cells. However, these ligands aim at targets that are often also upregulated in inflamed tissues. Here, we assessed the ability of targeted nanoparticles to distinguish metastatic cancer from sites of inflammation. Using common targeting ligands and a 60-nm liposome as a representative nanoparticle, we generated three targeted nanoparticle (NP) variants that targeted either fibronectin, folate, or αvβ3 integrin, whose deposition was compared against that of standard untargeted NP. Using fluorescently labeled NPs and ex vivo fluorescence imaging of organs, we assessed the deposition of the NPs into the lungs of mice modeling 4 different biological landscapes, including healthy lungs, aggressive metastasis in lungs, dormant/latent metastasis in lungs, and lungs with general pulmonary inflammation. Among the four NP variants, fibronectin-targeting NP and untargeted NP exhibited the highest deposition in lungs harboring aggressive metastases. However, the deposition of all targeted NP variants in lungs with metastasis was similar to the deposition in lungs with inflammation. Only the untargeted NP was able to exhibit higher deposition in metastasis than inflammation. Moreover, flow-cytometry analysis showed all NP variants accumulated predominantly in immune cells rather than cancer cells. For example, the number of NP+ macrophages and dendritic cells was 16-fold greater than NP+ cancer cells in the case of fibronectin-targeting NP. Overall, targeted NPs were unable to distinguish cancer metastasis from general inflammation, which may have clinical implications to the nanoparticle-mediated delivery of cancer drugs.
Collapse
Affiliation(s)
- Andrew S Choi
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Taylor J Moon
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Walid Abuhashim
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Anubhuti Bhalotia
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Huikang Qian
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Kai E Paulsen
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Morgan Lorkowski
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Crystal Ndamira
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Ramamurthy Gopalakrishnan
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Animesha Krishnamurthy
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - William P Schiemann
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America; Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America; Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America.
| |
Collapse
|
3
|
Nel J, Elkhoury K, Velot É, Bianchi A, Acherar S, Francius G, Tamayol A, Grandemange S, Arab-Tehrany E. Functionalized liposomes for targeted breast cancer drug delivery. Bioact Mater 2023; 24:401-437. [PMID: 36632508 PMCID: PMC9812688 DOI: 10.1016/j.bioactmat.2022.12.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/05/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Despite the exceptional progress in breast cancer pathogenesis, prognosis, diagnosis, and treatment strategies, it remains a prominent cause of female mortality worldwide. Additionally, although chemotherapies are effective, they are associated with critical limitations, most notably their lack of specificity resulting in systemic toxicity and the eventual development of multi-drug resistance (MDR) cancer cells. Liposomes have proven to be an invaluable drug delivery system but of the multitudes of liposomal systems developed every year only a few have been approved for clinical use, none of which employ active targeting. In this review, we summarize the most recent strategies in development for actively targeted liposomal drug delivery systems for surface, transmembrane and internal cell receptors, enzymes, direct cell targeting and dual-targeting of breast cancer and breast cancer-associated cells, e.g., cancer stem cells, cells associated with the tumor microenvironment, etc.
Collapse
Affiliation(s)
- Janske Nel
- Université de Lorraine, LIBio, F-54000, Nancy, France
| | | | - Émilie Velot
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Arnaud Bianchi
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Samir Acherar
- Université de Lorraine, CNRS, LCPM, F-54000, Nancy, France
| | | | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | | | | |
Collapse
|
4
|
Recent progress in nanocarrier-based drug delivery systems for antitumour metastasis. Eur J Med Chem 2023; 252:115259. [PMID: 36934485 DOI: 10.1016/j.ejmech.2023.115259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Tumour metastasis is one of the major factors leading to poor prognosis as well as lower survival among cancer patients. A number of studies investigating the inhibition of tumour metastasis have been conducted. It is difficult to achieve satisfactory results with surgery alone for distant metastatic tumours, and chemotherapy can boost the healing rate and prognosis of patients. However, the poor therapeutic efficacy of chemotherapy drugs due to their low solubility, lack of tumour targeting, instability in vivo, high toxicity and multidrug resistance hinder their application. Immunotherapy is beneficial to the treatment of metastatic cancers, but it also has disadvantages such as adverse reactions and acquired resistance. Fortunately, delivery of chemotherapeutic drugs with nanocarriers can reduce systemic reactions caused by chemotherapeutic agents and inhibit metastasis. This review discusses the underlying mechanisms of metastasis, therapeutic approaches for antitumour metastasis, the advantages of nanodrug delivery systems and their application in reducing metastasis.
Collapse
|
5
|
Yang J, Luly KM, Green JJ. Nonviral nanoparticle gene delivery into the CNS for neurological disorders and brain cancer applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1853. [PMID: 36193561 PMCID: PMC10023321 DOI: 10.1002/wnan.1853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/24/2022] [Accepted: 08/11/2022] [Indexed: 03/15/2023]
Abstract
Nonviral nanoparticles have emerged as an attractive alternative to viral vectors for gene therapy applications, utilizing a range of lipid-based, polymeric, and inorganic materials. These materials can either encapsulate or be functionalized to bind nucleic acids and protect them from degradation. To effectively elicit changes to gene expression, the nanoparticle carrier needs to undergo a series of steps intracellularly, from interacting with the cellular membrane to facilitate cellular uptake to endosomal escape and nucleic acid release. Adjusting physiochemical properties of the nanoparticles, such as size, charge, and targeting ligands, can improve cellular uptake and ultimately gene delivery. Applications in the central nervous system (CNS; i.e., neurological diseases, brain cancers) face further extracellular barriers for a gene-carrying nanoparticle to surpass, with the most significant being the blood-brain barrier (BBB). Approaches to overcome these extracellular challenges to deliver nanoparticles into the CNS include systemic, intracerebroventricular, intrathecal, and intranasal administration. This review describes and compares different biomaterials for nonviral nanoparticle-mediated gene therapy to the CNS and explores challenges and recent preclinical and clinical developments in overcoming barriers to nanoparticle-mediated delivery to the brain. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Joanna Yang
- Departments of Biomedical Engineering, Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathryn M Luly
- Departments of Biomedical Engineering, Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jordan J Green
- Departments of Biomedical Engineering, Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Chavda VP, Nalla LV, Balar P, Bezbaruah R, Apostolopoulos V, Singla RK, Khadela A, Vora L, Uversky VN. Advanced Phytochemical-Based Nanocarrier Systems for the Treatment of Breast Cancer. Cancers (Basel) 2023; 15:1023. [PMID: 36831369 PMCID: PMC9954440 DOI: 10.3390/cancers15041023] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
As the world's most prevalent cancer, breast cancer imposes a significant societal health burden and is among the leading causes of cancer death in women worldwide. Despite the notable improvements in survival in countries with early detection programs, combined with different modes of treatment to eradicate invasive disease, the current chemotherapy regimen faces significant challenges associated with chemotherapy-induced side effects and the development of drug resistance. Therefore, serious concerns regarding current chemotherapeutics are pressuring researchers to develop alternative therapeutics with better efficacy and safety. Due to their extremely biocompatible nature and efficient destruction of cancer cells via numerous mechanisms, phytochemicals have emerged as one of the attractive alternative therapies for chemotherapeutics to treat breast cancer. Additionally, phytofabricated nanocarriers, whether used alone or in conjunction with other loaded phytotherapeutics or chemotherapeutics, showed promising results in treating breast cancer. In the current review, we emphasize the anticancer activity of phytochemical-instigated nanocarriers and phytochemical-loaded nanocarriers against breast cancer both in vitro and in vivo. Since diverse mechanisms are implicated in the anticancer activity of phytochemicals, a strong emphasis is placed on the anticancer pathways underlying their action. Furthermore, we discuss the selective targeted delivery of phytofabricated nanocarriers to cancer cells and consider research gaps, recent developments, and the druggability of phytoceuticals. Combining phytochemical and chemotherapeutic agents with nanotechnology might have far-reaching impacts in the future.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Lakshmi Vineela Nalla
- Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Guntur 522302, Andhra Pradesh, India
| | - Pankti Balar
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu 610064, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33613, USA
| |
Collapse
|
7
|
Vikas, Mehata AK, Suseela MNL, Behera C, Kumari P, Mahto SK, Muthu MS. Chitosan-alginate nanoparticles of cabazitaxel: Design, dual-receptor targeting and efficacy in lung cancer model. Int J Biol Macromol 2022; 221:874-890. [PMID: 36089091 DOI: 10.1016/j.ijbiomac.2022.09.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Cabazitaxel (CZT) loaded chitosan-alginate based (CSA) nanoparticles were developed with dual targeting functions of both folate receptor and epidermal growth factor receptor (EGFR) using ionic gelation technique. The chitosan-folate conjugate was synthesized, and characterized by using FTIR, NMR and Mass spectroscopy. The physicochemical parameters and morphology of all CSA nanoparticles were examined. The degree of conjugation of folic acid and cetuximab (CTXmab) was determined by UV-Visible spectroscopy and Bradford assay, respectively. Moreover, XPS analysis also supported the presence of the ligands on nanoparticles. The cellular-uptake study performed on A-549 cells demonstrated a significant enhancement in the uptake of dual-receptor targeted CSA nanoparticles than non-targeted and single-receptor targeted CSA nanoparticles. Further, CZT-loaded dual receptors targeted CSA nanoparticles also showed significantly lower IC50 values (~38 folds) than the CZT control against A-549 cells. Further, in-vivo histopathological evaluations of dual receptor-targeted CSA nanoparticles have demonstrated better safety in Wistar rats. Moreover, its treatment on the Benzo(a)pyrene (B(a)P) induced lung cancer mice model has showed the enhanced anticancer efficacy of CZT with a prolonged survival rate.
Collapse
Affiliation(s)
- Vikas
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - M Nikitha Lakshmi Suseela
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Chittaranjan Behera
- PK-PD Tox & Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Pooja Kumari
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Sanjeev Kumar Mahto
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India.
| |
Collapse
|
8
|
Joun I, Nixdorf S, Deng W. Advances in lipid-based nanocarriers for breast cancer metastasis treatment. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:893056. [PMID: 36062261 PMCID: PMC9433809 DOI: 10.3389/fmedt.2022.893056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
Breast cancer (BC) is the most common cancer affecting women worldwide, with over 2 million women diagnosed every year, and close to 8 million women currently alive following a diagnosis of BC in the last 5-years. The side effects such as chemodrug toxicity to healthy tissues and drug resistance severely affect the quality of life of BC patients. To overcome these limitations, many efforts have been made to develop nanomaterial-based drug delivery systems. Among these nanocarriers, lipid-based delivery platforms represented one of the most successful candidates for cancer therapy, improving the safety profile and therapeutic efficacy of encapsulated drugs. In this review we will mainly discuss and summarize the recent advances in such delivery systems for BC metastasis treatment, with a particular focus on targeting the common metastatic sites in bone, brain and lung. We will also provide our perspectives on lipid-based nanocarrier development for future clinical translation.
Collapse
Affiliation(s)
- Ingrid Joun
- School of Chemical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Sheri Nixdorf
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| | - Wei Deng
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
- *Correspondence: Wei Deng
| |
Collapse
|
9
|
Wang X, Wu C, Liu S, Peng D. Combinatorial therapeutic strategies for enhanced delivery of therapeutics to brain cancer cells through nanocarriers: current trends and future perspectives. Drug Deliv 2022; 29:1370-1383. [PMID: 35532094 PMCID: PMC9090367 DOI: 10.1080/10717544.2022.2069881] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Brain cancer is the most aggressive one among various cancers. It has a drastic impact on people's lives because of the failure in treatment efficacy of the currently employed strategies. Various strategies used to relieve pain in brain cancer patients and to prolong survival time include radiotherapy, chemotherapy, and surgery. Nevertheless, several inevitable limitations are accompanied by such treatments due to unsatisfactory curative effects. Generally, the treatment of cancers is very challenging due to many reasons including drugs’ intrinsic factors and physiological barriers. Blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) are the two additional hurdles in the way of therapeutic agents to brain tumors delivery. Combinatorial and targeted therapies specifically in cancer show a very promising role where nanocarriers’ based formulations are designed primarily to achieve tumor-specific drug release. A dual-targeting strategy is a versatile way of chemotherapeutics delivery to brain tumors that gets the aid of combined ligands and mediators that cross the BBB and reaches the target site efficiently. In contrast to single targeting where one receptor or mediator is targeted, the dual-targeting strategy is expected to produce a multiple-fold increase in therapeutic efficacy for cancer therapy, especially in brain tumors. In a nutshell, a dual-targeting strategy for brain tumors enhances the delivery efficiency of chemotherapeutic agents via penetration across the blood-brain barrier and enhances the targeting of tumor cells. This review article highlights the ongoing status of the brain tumor therapy enhanced by nanoparticle based delivery with the aid of dual-targeting strategies. The future perspectives in this regard have also been highlighted.
Collapse
Affiliation(s)
- Xiande Wang
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin'an People's Hospital, The First People's Hospital of Hangzhou Lin'an District, Hangzhou, China
| | - Cheng Wu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Shiming Liu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Deqing Peng
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
10
|
Khan N, Ruchika, Kumar Dhritlahre R, Saneja A. Recent advances in dual-ligand targeted nanocarriers for cancer therapy. Drug Discov Today 2022; 27:2288-2299. [DOI: 10.1016/j.drudis.2022.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/05/2022] [Accepted: 04/11/2022] [Indexed: 12/30/2022]
|
11
|
Chen W, Yuan Y, Li C, Mao H, Liu B, Jiang X. Modulating Tumor Extracellular Matrix by Simultaneous Inhibition of Two Cancer Cell Receptors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109376. [PMID: 34967049 DOI: 10.1002/adma.202109376] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/25/2021] [Indexed: 06/14/2023]
Abstract
The extracellular matrix (ECM) is involved in fundamental cellular processes and pathological progression of many diseases. While most research and current knowledge focuses on the processes of biological and mechanical changes in ECM signaling residing cancer cells to respond, little is known of the converse-of how cancer cells initiate the changes of ECM properties. Here, it is reported that blocking the cancer cell signaling leads to disruption of tumor ECM. Using recombinant proteins (RPs) and recombinant protein-drug conjugates (RPDCs) that simultaneously target both epidermal growth factor receptor and integrin, it is demonstrated that multireceptor-mediated active modulation of tumor ECM can inhibit and even reverse tumor remodeling of the physiological and structural microenvironment. These results not only provide insights into the regulatory roles of cancer cells in developing a protumoral microenvironment, but also introduce a new therapeutic platform or strategy to treat cancers.
Collapse
Affiliation(s)
- Weizhi Chen
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yang Yuan
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Cheng Li
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xiqun Jiang
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
12
|
Covarrubias G, Moon TJ, Loutrianakis G, Sims HM, Umapathy MP, Lorkowski ME, Bielecki PA, Wiese ML, Atukorale PU, Karathanasis E. Comparison of the uptake of untargeted and targeted immunostimulatory nanoparticles by immune cells in the microenvironment of metastatic breast cancer. J Mater Chem B 2022; 10:224-235. [PMID: 34846443 PMCID: PMC8732314 DOI: 10.1039/d1tb02256c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
To alter the immunosuppressive tumor microenvironment (TME), we developed an immunostimulatory nanoparticle (NP) to reprogram a tumor's dysfunctional and inhibitory antigen-presenting cells (APCs) into properly activated APCs that stimulate tumor-reactive cytotoxic T cells. Importantly, systemic delivery allowed NPs to efficiently utilize the entire microvasculature and gain access into the majority of the perivascular TME, which coincided with the APC-rich tumor areas leading to uptake of the NPs predominantly by APCs. In this work, a 60 nm NP was loaded with a STING agonist, which triggered robust production of interferon β, resulting in activation of APCs. In addition to untargeted NPs, we employed 'mainstream' ligands targeting fibronectin, αvβ3 integrin and P-selectin that are commonly used to direct nanoparticles to tumors. Using the 4T1 mouse model, we assessed the microdistribution of the four NP variants in the tumor immune microenvironment in three different breast cancer landscapes, including primary tumor, early metastasis, and late metastasis. The different NP variants resulted in variable uptake by immune cell subsets depending on the organ and tumor stage. Among the NP variants, therapeutic studies indicated that the untargeted NPs and the integrin-targeting NPs exhibited a remarkable short- and long-term immune response and long-lasting antitumor effect.
Collapse
Affiliation(s)
- Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Georgia Loutrianakis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Haley M Sims
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Mayura P Umapathy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Morgan E Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Peter A Bielecki
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Michelle L Wiese
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Prabhani U Atukorale
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| |
Collapse
|
13
|
Vikas, Viswanadh MK, Mehata AK, Sharma V, Priya V, Varshney N, Mahto SK, Muthu MS. Bioadhesive chitosan nanoparticles: Dual targeting and pharmacokinetic aspects for advanced lung cancer treatment. Carbohydr Polym 2021; 274:118617. [PMID: 34702448 DOI: 10.1016/j.carbpol.2021.118617] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022]
Abstract
The chitosan-folate conjugate was synthesized initially and confirmed by FTIR and NMR spectroscopic studies. Following, docetaxel (DXL) loaded non-targeted, single receptor and dual receptor (folate and EGFR) targeted chitosan nanoparticles were prepared and their shape, particle size, zeta-potential, surface morphology and texture were screened by SEM, TEM, AFM analyses. Surface chemistry analysis by XPS indeed confirmed the successful conjugation of folate and cetuximab on the targeted formulations. In-vitro analysis of dual-targeted chitosan nanoparticles has revealed their superior cytotoxicity against A-549 cells. The IC50 of dual receptor-targeted chitosan NP was almost 34 times lower than DXL control. In-vivo pharmacokinetic study on Wistar rats has demonstrated improved relative bioavailability of all NP in comparison to DXL control. The results illustrated that EGFR and folate dual targeted NP enhanced the cytotoxicity of DXL towards A-549 lung cancer cells and substantially improved DXL pharmacokinetics in rats.
Collapse
Affiliation(s)
- Vikas
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Matte Kasi Viswanadh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Vishal Sharma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Vishnu Priya
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Neelima Varshney
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Sanjeev Kumar Mahto
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India.
| |
Collapse
|
14
|
Covarrubias G, Lorkowski ME, Sims HM, Loutrianakis G, Rahmy A, Cha A, Abenojar E, Wickramasinghe S, Moon TJ, Samia ACS, Karathanasis E. Hyperthermia-mediated changes in the tumor immune microenvironment using iron oxide nanoparticles. NANOSCALE ADVANCES 2021; 3:5890-5899. [PMID: 34746645 PMCID: PMC8507876 DOI: 10.1039/d1na00116g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
Iron oxide nanoparticles (IONPs) have often been investigated for tumor hyperthermia. IONPs act as heating foci in the presence of an alternating magnetic field (AMF). It has been shown that hyperthermia can significantly alter the tumor immune microenvironment. Typically, mild hyperthermia invokes morphological changes within the tumor, which elicits a secretion of inflammatory cytokines and tumor neoantigens. Here, we focused on the direct effect of IONP-induced hyperthermia on the various tumor-resident immune cell subpopulations. We compared direct intratumoral injection to systemic administration of IONPs followed by application of an external AMF. We used the orthotopic 4T1 mouse model, which represents aggressive and metastatic breast cancer with a highly immunosuppressive microenvironment. A non-inflamed and 'cold' microenvironment inhibits peripheral effector lymphocytes from effectively trafficking into the tumor. Using intratumoral or systemic injection, IONP-induced hyperthermia achieved a significant reduction of all the immune cell subpopulations in the tumor. However, the systemic delivery approach achieved superior outcomes, resulting in substantial reductions in the populations of both innate and adaptive immune cells. Upon depletion of the existing dysfunctional tumor-resident immune cells, subsequent treatment with clinically approved immune checkpoint inhibitors encouraged the repopulation of the tumor with 'fresh' infiltrating innate and adaptive immune cells, resulting in a significant decrease of the tumor cell population.
Collapse
Affiliation(s)
- Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland Ohio USA
| | - Morgan E Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland Ohio USA
| | - Haley M Sims
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
| | - Georgia Loutrianakis
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
| | - Abdelrahman Rahmy
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
| | - Anthony Cha
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
| | - Eric Abenojar
- Department of Chemistry, Case Western Reserve University Cleveland Ohio USA
| | | | - Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
| | | | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University Cleveland Ohio USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland Ohio USA
| |
Collapse
|
15
|
Huang WY, Lai CH, Peng SL, Hsu CY, Hsu PH, Chu PY, Feng CL, Lin YH. Targeting Tumor Cells with Nanoparticles for Enhanced Co-Drug Delivery in Cancer Treatment. Pharmaceutics 2021; 13:1327. [PMID: 34575403 PMCID: PMC8465501 DOI: 10.3390/pharmaceutics13091327] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer (GC) is a fatal malignant tumor, and effective therapies to attenuate its progression are lacking. Nanoparticle (NP)-based solutions may enable the design of novel treatments to eliminate GC. Refined, receptor-targetable NPs can selectively target cancer cells and improve the cellular uptake of drugs. To overcome the current limitations and enhance the therapeutic effects, epigallocatechin-3-gallate (EGCG) and low-concentration doxorubicin (DX) were encapsulated in fucoidan and d-alpha-tocopherylpoly (ethylene glycol) succinate-conjugated hyaluronic acid-based NPs for targeting P-selectin-and cluster of differentiation (CD)44-expressing gastric tumors. The EGCG/DX-loaded NPs bound to GC cells and released bioactive combination drugs, demonstrating better anti-cancer effects than the EGCG/DX combination solution. In vivo assays in an orthotopic gastric tumor mouse model showed that the EGCG/DX-loaded NPs significantly increased the activity of gastric tumors without inducing organ injury. Overall, our EGCG/DX-NP system exerted a beneficial effect on GC treatment and may facilitate the development of nanomedicine-based combination chemotherapy against GC in the future.
Collapse
Affiliation(s)
- Wen-Ying Huang
- Department of Applied Cosmetology, Hung-Kuang University, Taichung 433304, Taiwan;
| | - Chih-Ho Lai
- Molecular Infectious Disease Research Center, Department of Microbiology and Immunology, Chang Gung University and Chang Gung Memorial Hospital, Taoyuan 333323, Taiwan;
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung 404, Taiwan;
| | - Che-Yu Hsu
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (C.-Y.H.); (P.-Y.C.)
| | - Po-Hung Hsu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Pei-Yi Chu
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (C.-Y.H.); (P.-Y.C.)
| | - Chun-Lung Feng
- Division of Hepatogastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung 404332, Taiwan;
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (C.-Y.H.); (P.-Y.C.)
- Center for Advanced Pharmaceutics and Drug Delivery Research, Department and Institute of Pharmacology, Institute of Biopharmaceutical Sciences, Faculty of Pharmacy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan
| |
Collapse
|
16
|
Beyaz H, Uludag H, Kavaz D, Rizaner N. Mechanisms of Drug Resistance and Use of Nanoparticle Delivery to Overcome Resistance in Breast Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:163-181. [PMID: 34287795 DOI: 10.1007/5584_2021_648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Breast cancer is the leading cancer type diagnosed among women in the world. Unfortunately, drug resistance to current breast cancer chemotherapeutics remains the main challenge for a higher survival rate. The recent progress in the nanoparticle platforms and distinct features of nanoparticles that enhance the efficacy of therapeutic agents, such as improved delivery efficacy, increased intracellular cytotoxicity, and reduced side effects, hold great promise to overcome the observed drug resistance. Currently, multifaceted investigations are probing the resistance mechanisms associated with clinical drugs, and identifying new breast cancer-associated molecular targets that may lead to improved therapeutic approaches with the nanoparticle platforms. Nanoparticle platforms including siRNA, antibody-specific targeting and the role of nanoparticles in cellular processes and their effect on breast cancer were discussed in this article.
Collapse
Affiliation(s)
- Huseyin Beyaz
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Nicosia, Turkey.
| | - Hasan Uludag
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Doga Kavaz
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Nicosia, Turkey
- Biotechnology Research Center, Cyprus International University, Nicosia, Turkey
| | - Nahit Rizaner
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Nicosia, Turkey
- Biotechnology Research Center, Cyprus International University, Nicosia, Turkey
| |
Collapse
|
17
|
Kong X, Cheng R, Wang J, Fang Y, Hwang KC. Nanomedicines inhibiting tumor metastasis and recurrence and their clinical applications. NANO TODAY 2021; 36:101004. [DOI: 10.1016/j.nantod.2020.101004] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Jahan S, Karim ME, Chowdhury EH. Nanoparticles Targeting Receptors on Breast Cancer for Efficient Delivery of Chemotherapeutics. Biomedicines 2021; 9:114. [PMID: 33530291 PMCID: PMC7910939 DOI: 10.3390/biomedicines9020114] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/25/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
The journey of chemotherapeutic drugs from the site of administration to the site of action is confronted by several factors including low bioavailability, uneven distribution in major organs, limited accessibility of drug molecules to the distant tumor tissues, and lower therapeutic indexes. These unavoidable features of classical chemotherapeutics necessitate an additional high, repetitive dose of drugs to obtain maximum therapeutic responses with the result of unintended adverse side effects. An erratic tumor microenvironment, notable drawbacks of conventional chemotherapy, and multidrug-resistant mechanisms of breast cancer cells warrant precisely designed therapeutics for the treatment of cancers. In recent decades, nanoparticles have been deployed for the delivery of standard anticancer drugs to maximize the therapeutic potency while minimizing the adverse effects to increase the quality and span of life. Several organic and inorganic nanoplatforms that have been designed exploiting the distinctive features of the tumor microenvironment and tumor cells offer favorable physicochemical properties and pharmacokinetic profiles of a parent drug, with delivery of higher amounts of the drug to the pathological site and its controlled release, thereby improving the balance between its efficacy and toxicity. Advances to this front have included design and construction of targeted nanoparticles by conjugating homing devices like peptide, ligand, and Fab on the surface of nanomaterials to navigate nanoparticledrug complexes towards the target tumor cell with minimal destruction of healthy cells. Furthermore, actively targeting nanoparticles can facilitate the delivery and cellular uptake of nanoparticle-loaded drug constructs via binding with specific receptors expressed aberrantly on the surface of a tumor cell. Herein, we present an overview of the principle of targeted delivery approaches, exploiting drug-nanoparticle conjugates with multiple targeting moieties to target specific receptors of breast cancer cells and highlighting therapeutic evaluation in preclinical studies. We conclude that an understanding of the translational gap and challenges would show the possible future directions to foster the development of novel targeted nanotherapeutics.
Collapse
Affiliation(s)
| | | | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Petaling Jaya 47500, Malaysia; (S.J.); (M.E.K.)
| |
Collapse
|
19
|
Biomedical nanoparticle design: What we can learn from viruses. J Control Release 2021; 329:552-569. [PMID: 33007365 PMCID: PMC7525328 DOI: 10.1016/j.jconrel.2020.09.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 01/02/2023]
Abstract
Viruses are nanomaterials with a number of properties that surpass those of many synthetic nanoparticles (NPs) for biomedical applications. They possess a rigorously ordered structure, come in a variety of shapes, and present unique surface elements, such as spikes. These attributes facilitate propitious biodistribution, the crossing of complex biological barriers and a minutely coordinated interaction with cells. Due to the orchestrated sequence of interactions of their stringently arranged particle corona with cellular surface receptors they effectively identify and infect their host cells with utmost specificity, while evading the immune system at the same time. Furthermore, their efficacy is enhanced by their response to stimuli and the ability to spread from cell to cell. Over the years, great efforts have been made to mimic distinct viral traits to improve biomedical nanomaterial performance. However, a closer look at the literature reveals that no comprehensive evaluation of the benefit of virus-mimetic material design on the targeting efficiency of nanomaterials exists. In this review we, therefore, elucidate the impact that viral properties had on fundamental advances in outfitting nanomaterials with the ability to interact specifically with their target cells. We give a comprehensive overview of the diverse design strategies and identify critical steps on the way to reducing them to practice. More so, we discuss the advantages and future perspectives of a virus-mimetic nanomaterial design and try to elucidate if viral mimicry holds the key for better NP targeting.
Collapse
|
20
|
McCallum GA, Shiralkar J, Suciu D, Covarrubias G, Yu JS, Karathanasis E, Durand DM. Chronic neural activity recorded within breast tumors. Sci Rep 2020; 10:14824. [PMID: 32908180 PMCID: PMC7481786 DOI: 10.1038/s41598-020-71670-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
Nerve fibers are known to reside within malignant tumors and the greater the neuronal density the worse prognosis for the patient. Recent discoveries using tumor bearing animal models have eluded to the autonomic nervous system having a direct effect on tumor growth and metastasis. We report the first direct and chronic in vivo measurements of neural activity within tumors. Using a triple-negative mammary cancer mouse model and chronic neural interface techniques, we have recorded neural activity directly within the tumor mass while the tumor grows and metastasizes. The results indicate that there is a strong connection between the autonomic nervous system and the tumor and could help uncover the mechanisms of tumor growth and metastasis.
Collapse
Affiliation(s)
- Grant A McCallum
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Jay Shiralkar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Diana Suciu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jennifer S Yu
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.,Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Dominique M Durand
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
21
|
Covarrubias G, Johansen ML, Vincent J, Erokwu BO, Craig SEL, Rahmy A, Cha A, Lorkowski M, MacAskill C, Scott B, Gargesha M, Roy D, Flask CA, Karathanasis E, Brady-Kalnay SM. PTPmu-targeted nanoparticles label invasive pediatric and adult glioblastoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102216. [PMID: 32413511 DOI: 10.1016/j.nano.2020.102216] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 04/07/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
Abstract
Poor prognosis for glioblastoma (GBM) is a consequence of the aggressive and infiltrative nature of gliomas where individual cells migrate away from the main tumor to distant sites, making complete surgical resection and treatment difficult. In this manuscript, we characterize an invasive pediatric glioma model and determine if nanoparticles linked to a peptide recognizing the GBM tumor biomarker PTPmu can specifically target both the main tumor and invasive cancer cells in adult and pediatric glioma models. Using both iron and lipid-based nanoparticles, we demonstrate by magnetic resonance imaging, optical imaging, histology, and iron quantification that PTPmu-targeted nanoparticles effectively label adult gliomas. Using PTPmu-targeted nanoparticles in a newly characterized orthotopic pediatric SJ-GBM2 model, we demonstrate individual tumor cell labeling both within the solid tumor margins and at invasive and dispersive sites.
Collapse
Affiliation(s)
- Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Mette L Johansen
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH
| | - Jason Vincent
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH
| | | | - Sonya E L Craig
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH
| | - Abdelrahman Rahmy
- Department of Chemistry, Case Western Reserve University, Cleveland, OH
| | - Anthony Cha
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Morgan Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | | | | | | | | | - Chris A Flask
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH; Department of Radiology, Case Western Reserve University, Cleveland, OH; Department of Pediatrics, Case Western Reserve University, Cleveland, OH
| | | | - Susann M Brady-Kalnay
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH; Department of Neurosciences, Case Western Reserve University, Cleveland, OH.
| |
Collapse
|
22
|
Turan O, Bielecki P, Tong K, Covarrubias G, Moon T, Rahmy A, Cooley S, Park Y, Peiris PM, Ghaghada KB, Karathanasis E. Effect of Dose and Selection of Two Different Ligands on the Deposition and Antitumor Efficacy of Targeted Nanoparticles in Brain Tumors. Mol Pharm 2019; 16:4352-4360. [PMID: 31442061 DOI: 10.1021/acs.molpharmaceut.9b00693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Deposition of nanoparticles to tumors often can be enhanced by targeting receptors overexpressed in a tumor. However, a tumor may exhibit a finite number of a biomarker that is accessible and targetable by nanoparticles, limiting the available landing spots. To explore this, we selected two different biomarkers that effectively home nanoparticles in brain tumors. Specifically, we used either an αvβ3 integrin-targeting peptide or a fibronectin-targeting peptide as a ligand on nanoparticles termed RGD-NP and CREKA-NP, respectively. In mouse models of glioblastoma multiforme, we systemically injected the nanoparticles loaded with a cytotoxic drug at different doses ranging from 2 to 8 mg/kg drug. The upper dose threshold of RGD-NP is ∼2 mg/kg. CREKA-NP reached its upper dose threshold at 5 mg/kg. For both targeted nanoparticle variants, higher dose did not ensure higher intratumoral drug levels, but it contributed to elevated off-target deposition and potentially greater toxicity. A cocktail combining RGD-NP and CREKA-NP was then administered at a dose corresponding to the upper dose threshold for each formulation resulting in a 3-fold higher intratumoral deposition than the individual formulations. The combination of the two different targeting schemes at the appropriate dose for each nanoparticle variant facilitated remarkable increase in intratumoral drug levels that was not achievable by a sole targeting nanoparticle alone.
Collapse
Affiliation(s)
- Oguz Turan
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Peter Bielecki
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Kathleen Tong
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Gil Covarrubias
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Taylor Moon
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Abdelrahman Rahmy
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Shane Cooley
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Youngjun Park
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Pubudu M Peiris
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States.,Case Comprehensive Cancer Center , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Ketan B Ghaghada
- Edward B. Singleton Department of Pediatric Radiology , Texas Children's Hospital , Houston , Texas 77030 , United States
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States.,Case Comprehensive Cancer Center , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| |
Collapse
|