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
|
Srinivasarao DA, Shah S, Famta P, Vambhurkar G, Jain N, Pindiprolu SKSS, Sharma A, Kumar R, Padhy HP, Kumari M, Madan J, Srivastava S. Unravelling the role of tumor microenvironment responsive nanobiomaterials in spatiotemporal controlled drug delivery for lung cancer therapy. Drug Deliv Transl Res 2025; 15:407-435. [PMID: 39037533 DOI: 10.1007/s13346-024-01673-z] [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] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
Design and development of efficient drug delivery technologies that impart site-specificity is the need of the hour for the effective treatment of lung cancer. The emergence of materials science and nanotechnology partially helped drug delivery scientists to achieve this objective. Various stimuli-responsive materials that undergo degradation at the pathological tumor microenvironment (TME) have been developed and explored for drug delivery applications using nanotechnological approaches. Nanoparticles (NPs), owing to their small size and high surface area to volume ratio, demonstrated enhanced cellular internalization, permeation, and retention at the tumor site. Such passive accumulation of stimuli-responsive materials helped to achieve spatiotemporally controlled and targeted drug delivery within the tumors. In this review, we discussed various stimuli-physical (interstitial pressure, temperature, and stiffness), chemical (pH, hypoxia, oxidative stress, and redox state), and biological (receptor expression, efflux transporters, immune cells, and their receptors or ligands)-that are characteristic to the TME. We mentioned an array of biomaterials-based nanoparticulate delivery systems that respond to these stimuli and control drug release at the TME. Further, we discussed nanoparticle-based combinatorial drug delivery strategies. Finally, we presented our perspectives on challenges related to scale-up, clinical translation, and regulatory approvals.
Collapse
Affiliation(s)
- Dadi A Srinivasarao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Naitik Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Sai Kiran S S Pindiprolu
- Aditya Pharmacy College, Surampalem, 533 437, Andhra Pradesh, India
- Jawaharlal Nehru Technological University, Kakinada, 533 003, Andhra Pradesh, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Meenu Kumari
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
3
|
Wang P, Peng Z, Zhang Y, Zhang X, Chen X, Li F, Chen B, Niu S, Du K, Zhu LM. A chitosan-camouflaged nanomedicine triggered by hierarchically stimuli to release drug for multimodal imaging-guided chemotherapy of breast cancer. Carbohydr Polym 2024; 335:122073. [PMID: 38616095 DOI: 10.1016/j.carbpol.2024.122073] [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: 11/18/2023] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 04/16/2024]
Abstract
Breast cancer remains one of the most intractable diseases, especially the malignant form of metastasis, with which the cancer cells are hard to track and eliminate. Herein, the common known carbohydrate polymer chitosan (CS) was innovatively used as a shelter for the potent tumor-killing agent. The designed nanoparticles (NPs) not only enhance the solubility of hydrophobic paclitaxel (PTX), but also provide a "hide" effect for cytotoxic PTX in physiological condition. Moreover, coupled with the photothermal (PTT) properties of MoS2, results in a potent chemo/PTT platform. The MoS2@PTX-CS-K237 NPs have a uniform size (135 ± 17 nm), potent photothermal properties (η = 31.5 %), and environment-responsive (low pH, hypoxia) and near infrared (NIR) laser irradiation-triggered PTX release. Through a series of in vitro and in vivo experiments, the MoS2@PTX-CS-K237 showed high affinity and specificity for breast cancer cells, impressive tumor killing capacity, as well as the effective inhibitory effect of metastasis. Benefit from the unique optical properties of MoS2, this multifunctional nanomedicine also exhibited favorable thermal/PA/CT multimodality imaging effect on tumor-bearing mice. The system developed in this work represents the advanced design concept of hierarchical stimulus responsive drug release, and merits further investigation as a potential nanotheranostic platform for clinical translation.
Collapse
Affiliation(s)
- Pei Wang
- Department of Radiation Oncology, Cancer Institute, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, PR China
| | - Zhi Peng
- Department of Orthopedic Surgery, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, PR China
| | - Yanyan Zhang
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Xuejing Zhang
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Xia Chen
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Fan Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, PR China
| | - Bo Chen
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, PR China
| | - Shiwei Niu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, PR China.
| | - Kaili Du
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, PR China.
| | - Li-Min Zhu
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China.
| |
Collapse
|
4
|
Li X, Zou J, He Z, Sun Y, Song X, He W. The interaction between particles and vascular endothelium in blood flow. Adv Drug Deliv Rev 2024; 207:115216. [PMID: 38387770 DOI: 10.1016/j.addr.2024.115216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Particle-based drug delivery systems have shown promising application potential to treat human diseases; however, an incomplete understanding of their interactions with vascular endothelium in blood flow prevents their inclusion into mainstream clinical applications. The flow performance of nano/micro-sized particles in the blood are disturbed by many external/internal factors, including blood constituents, particle properties, and endothelium bioactivities, affecting the fate of particles in vivo and therapeutic effects for diseases. This review highlights how the blood constituents, hemodynamic environment and particle properties influence the interactions and particle activities in vivo. Moreover, we briefly summarized the structure and functions of endothelium and simulated devices for studying particle performance under blood flow conditions. Finally, based on particle-endothelium interactions, we propose future opportunities for novel therapeutic strategies and provide solutions to challenges in particle delivery systems for accelerating their clinical translation. This review helps provoke an increasing in-depth understanding of particle-endothelium interactions and inspires more strategies that may benefit the development of particle medicine.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongshan He
- Department of Critical Care Medicine and Department of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, PR China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co., LtD., Jinan 250000, PR China
| | - Xiangrong Song
- Department of Critical Care Medicine and Department of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, PR China.
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China.
| |
Collapse
|
5
|
Sandbhor P, Palkar P, Bhat S, John G, Goda JS. Nanomedicine as a multimodal therapeutic paradigm against cancer: on the way forward in advancing precision therapy. NANOSCALE 2024. [PMID: 38470224 DOI: 10.1039/d3nr06131k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Recent years have witnessed dramatic improvements in nanotechnology-based cancer therapeutics, and it continues to evolve from the use of conventional therapies (chemotherapy, surgery, and radiotherapy) to increasingly multi-complex approaches incorporating thermal energy-based tumor ablation (e.g. magnetic hyperthermia and photothermal therapy), dynamic therapy (e.g. photodynamic therapy), gene therapy, sonodynamic therapy (e.g. ultrasound), immunotherapy, and more recently real-time treatment efficacy monitoring (e.g. theranostic MRI-sensitive nanoparticles). Unlike monotherapy, these multimodal therapies (bimodal, i.e., a combination of two therapies, and trimodal, i.e., a combination of more than two therapies) incorporating nanoplatforms have tremendous potential to improve the tumor tissue penetration and retention of therapeutic agents through selective active/passive targeting effects. These combinatorial therapies can correspondingly alleviate drug response against hypoxic/acidic and immunosuppressive tumor microenvironments and promote/induce tumor cell death through various multi-mechanisms such as apoptosis, autophagy, and reactive oxygen-based cytotoxicity, e.g., ferroptosis, etc. These multi-faced approaches such as targeting the tumor vasculature, neoangiogenic vessels, drug-resistant cancer stem cells (CSCs), preventing intra/extravasation to reduce metastatic growth, and modulation of antitumor immune responses work complementary to each other, enhancing treatment efficacy. In this review, we discuss recent advances in different nanotechnology-mediated synergistic/additive combination therapies, emphasizing their underlying mechanisms for improving cancer prognosis and survival outcomes. Additionally, significant challenges such as CSCs, hypoxia, immunosuppression, and distant/local metastasis associated with therapy resistance and tumor recurrences are reviewed. Furthermore, to improve the clinical precision of these multimodal nanoplatforms in cancer treatment, their successful bench-to-clinic translation with controlled and localized drug-release kinetics, maximizing the therapeutic window while addressing safety and regulatory concerns are discussed. As we advance further, exploiting these strategies in clinically more relevant models such as patient-derived xenografts and 3D organoids will pave the way for the application of precision therapy.
Collapse
Affiliation(s)
- Puja Sandbhor
- Institute for NanoBioTechnology, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Pranoti Palkar
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Sakshi Bhat
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Geofrey John
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Jayant S Goda
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| |
Collapse
|
6
|
Schwartz-Duval A, Mackeyev Y, Mahmud I, Lorenzi PL, Gagea M, Krishnan S, Sokolov KV. Intratumoral Biosynthesis of Gold Nanoclusters by Pancreatic Cancer to Overcome Delivery Barriers to Radiosensitization. ACS NANO 2024; 18:1865-1881. [PMID: 38206058 PMCID: PMC10811688 DOI: 10.1021/acsnano.3c04260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024]
Abstract
Nanoparticle delivery to solid tumors is a prime challenge in nanomedicine. Here, we approach this challenge through the lens of biogeochemistry, the field that studies the flow of chemical elements within ecosystems as manipulated by living cellular organisms and their environments. We leverage biogeochemistry concepts related to gold cycling against pancreatic cancer, considering mammalian organisms as drivers for gold nanoparticle biosynthesis. Sequestration of gold nanoparticles within tumors has been demonstrated as an effective strategy to enhance radiotherapy; however, the desmoplasia of pancreatic cancer impedes nanoparticle delivery. Our strategy overcomes this barrier by applying an atomic-scale agent, ionic gold, for intratumoral gold nanoparticle biosynthesis. Our comprehensive studies showed the cancer-specific synthesis of gold nanoparticles from externally delivered gold ions in vitro and in a murine pancreatic cancer model in vivo; a substantial colocalization of gold nanoparticles (GNPs) with cancer cell nuclei in vitro and in vivo; a strong radiosensitization effect by the intracellularly synthesized GNPs; a uniform distribution of in situ synthesized GNPs throughout the tumor volume; a nearly 40-day total suppression of tumor growth in animal models of pancreatic cancer treated with a combination of gold ions and radiation that was also associated with a significantly higher median survival versus radiation alone (235 vs 102 days, respectively).
Collapse
Affiliation(s)
- Aaron
S. Schwartz-Duval
- Department
of Imaging Physics, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yuri Mackeyev
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center, Houston, Texas 77030, United States
| | - Iqbal Mahmud
- Department
of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Philip L. Lorenzi
- Department
of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Mihai Gagea
- Department
of Veterinary Medicine & Surgery, The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Sunil Krishnan
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center, Houston, Texas 77030, United States
| | - Konstantin V. Sokolov
- Department
of Imaging Physics, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| |
Collapse
|
7
|
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
|
8
|
Sultana N, David AE. Improving Cancer Targeting: A Study on the Effect of Dual-Ligand Density on Targeting of Cells Having Differential Expression of Target Biomarkers. Int J Mol Sci 2023; 24:13048. [PMID: 37685852 PMCID: PMC10487485 DOI: 10.3390/ijms241713048] [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: 07/19/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Silica nanoparticles with hyaluronic acid (HA) and folic acid (FA) were developed to study dual-ligand targeting of CD44 and folate receptors, respectively, in colon cancer. Characterization of particles with dynamic light scattering showed them to have hydrodynamic diameters of 147-271 nm with moderate polydispersity index (PDI) values. Surface modification of the particles was achieved by simultaneous reaction with HA and FA and results showed that ligand density on the surface increased with increasing concentrations in the reaction mixture. The nanoparticles showed minimal to no cytotoxicity with all formulations showing ≥ 90% cell viability at concentrations up to 100 µg/mL. Based on flow cytometry results, SW480 cell lines were positive for both receptors, the WI38 cell line was positive for CD44 receptor, and Caco2 was positive for the folate receptor. Cellular targeting studies demonstrated the potential of the targeted nanoparticles as promising candidates for delivery of therapeutic agents. The highest cellular targeting was achieved with particles synthesized using folate:surface amine (F:A) ratio of 9 for SW480 and Caco2 cells and at F:A = 0 for WI38 cells. The highest selectivity was achieved at F:A = 9 for both SW480:WI38 and SW480:Caco2 cells. Based on HA conjugation, the highest cellular targeting was achieved at H:A = 0.5-0.75 for SW480 cell, at H:A = 0.75 for WI38 cell and at H:A = 0.5 for Caco2 cells. The highest selectivity was achieved at H:A = 0 for both SW480:WI38 and SW480:Caco2 cells. These results demonstrated that the optimum ligand density on the nanoparticle for targeting is dependent on the levels of biomarker expression on the target cells. Ongoing studies will evaluate the therapeutic efficacy of these targeted nanoparticles using in vitro and in vivo cancer models.
Collapse
Affiliation(s)
| | - Allan E. David
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA;
| |
Collapse
|
9
|
Palange AL, Mascolo DD, Ferreira M, Gawne PJ, Spanò R, Felici A, Bono L, Moore TL, Salerno M, Armirotti A, Decuzzi P. Boosting the Potential of Chemotherapy in Advanced Breast Cancer Lung Metastasis via Micro-Combinatorial Hydrogel Particles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205223. [PMID: 36683230 PMCID: PMC10074128 DOI: 10.1002/advs.202205223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Breast cancer cell colonization of the lungs is associated with a dismal prognosis as the distributed nature of the disease and poor permeability of the metastatic foci challenge the therapeutic efficacy of small molecules, antibodies, and nanomedicines. Taking advantage of the unique physiology of the pulmonary circulation, here, micro-combinatorial hydrogel particles (µCGP) are realized via soft lithographic techniques to enhance the specific delivery of a cocktail of cytotoxic nanoparticles to metastatic foci. By cross-linking short poly(ethylene glycol) (PEG) chains with erodible linkers within a shape-defining template, a deformable and biodegradable polymeric skeleton is realized and loaded with a variety of therapeutic and imaging agents, including docetaxel-nanoparticles. In a model of advanced breast cancer lung metastasis, µCGP amplified the colocalization of docetaxel-nanoparticles with pulmonary metastatic foci, prolonged the retention of chemotoxic molecules at the diseased site, suppressed lesion growth, and boosted survival beyond 20 weeks post nodule engraftment. The flexible design and modular architecture of µCGP would allow the efficient deployment of complex combination therapies in other vascular districts too, possibly addressing metastatic diseases of different origins.
Collapse
Affiliation(s)
- Anna Lisa Palange
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
- Present address:
Harvard Medical School, Department of RadiologyMassachusetts General HospitalBostonMA02114USA
| | - Peter J. Gawne
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Alessia Felici
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
- Present address:
Division of Oncology, Department of Medicine and Department of PathologyStanford University School of MedicineStanfordCA94305USA
| | - Luca Bono
- Analytical Chemistry FacilityFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Thomas Lee Moore
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Marco Salerno
- Materials Characterization FacilityFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Andrea Armirotti
- Analytical Chemistry FacilityFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| |
Collapse
|
10
|
Wang A, Yue K, Zhong W, Zhang G, Wang L, Wang H, Zhang H, Zhang X. Ligand-receptor interaction in the specific targeting of biomimetic peptide nanoparticles to lysophosphatidylcholine. Int J Biol Macromol 2023; 227:193-202. [PMID: 36549027 DOI: 10.1016/j.ijbiomac.2022.12.162] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
As nanotechnology is applied clinical medicine, nanoparticle-based therapy is emerging as a novel approach for the treatment of atherosclerosis. Ligand-receptor interaction affects the effectiveness of nanoparticle targeting therapy. In this study, the biomimetic peptide (BP-KFFVLK-WYKDGD) ligand specifically targeting the lysophosphatidylcholine (LPC) receptor in atherosclerotic plaques was constructed. The corresponding ligand-receptor interaction under different pH values was investigated by molecular dynamics simulation and experimental measurements. Results show that the interaction force between the peptide and LPC is greater than that of the peptide and human umbilical vein endothelial cell, clearly demonstrating the specific targeting of the peptide ligand to the LPC receptor. The ligand-receptor binding of peptide and LPC dominantly depends on Coulomb and van der Waals interactions. The YKDG amino acids of the peptide are the main fragment that binds to LPC. Compared with neutral environment at pH 7.4, the interaction forces between the peptide and oxidized low-density lipoprotein (oxLDL) decreased by 18.22 % and 45.87 % under acidic environments at pH 6.5 and 5.5, respectively, because of the change in oxLDL secondary structure and the release of LPC from oxLDL. Nevertheless, the peptide still has a strong binding capacity with oxLDL for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Anqi Wang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China.
| | - Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China
| | - Lei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Hua Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xinxin Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China
| |
Collapse
|
11
|
Zhou S, Li C, Yuan Y, Jiang L, Chen W, Jiang X. Dendritic lipopeptide liposomes decorated with dual-targeted proteins. Biomater Sci 2022; 10:7032-7041. [PMID: 36318065 DOI: 10.1039/d2bm00952h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to their homing effects, cell and cell membrane-derived nanocarriers have been widely used to enhance drug target delivery. Inspired by the protein-anchored cell membrane architecture, we here report a tumor-targeted liposome, dtDLP, which was constructed through the electrostatic interaction between dendritic lipopeptide liposomes and a dual-targeted recombinant protein, achieving superior tumor homing, cellular endocytotic and penetration abilities. The dual-targeted recombinant protein consists of an anti-epidermal growth factor receptor single domain antibody and a peptide ligand for the integrin αvβ3. dtDLPs substantially reduced macrophage phagocytosis and increased drug internalization in both 4T1 cells and HeLa cells by providing more endocytic pathways. In addition, the dtDLPs showed great penetration ability in both multicellular spheroids and tumor tissues. Due to the improved cancer cellular uptake and tumor penetration, the dtDLPs exhibited a superior anticancer effect in both HeLa and 4T1 tumor-bearing mice. This work will be helpful for the design of cell-specific liposomes with admirable tumor targeting, endocytotic and penetration abilities.
Collapse
Affiliation(s)
- Sensen Zhou
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Cheng Li
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Yang Yuan
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Lei Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China. .,State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| |
Collapse
|
12
|
Ma XY, Hill BD, Hoang T, Wen F. Virus-inspired strategies for cancer therapy. Semin Cancer Biol 2022; 86:1143-1157. [PMID: 34182141 PMCID: PMC8710185 DOI: 10.1016/j.semcancer.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/27/2023]
Abstract
The intentional use of viruses for cancer therapy dates back over a century. As viruses are inherently immunogenic and naturally optimized delivery vehicles, repurposing viruses for drug delivery, tumor antigen presentation, or selective replication in cancer cells represents a simple and elegant approach to cancer treatment. While early virotherapy was fraught with harsh side effects and low response rates, virus-based therapies have recently seen a resurgence due to newfound abilities to engineer and tune oncolytic viruses, virus-like particles, and virus-mimicking nanoparticles for improved safety and efficacy. However, despite their great potential, very few virus-based therapies have made it through clinical trials. In this review, we present an overview of virus-inspired approaches for cancer therapy, discuss engineering strategies to enhance their mechanisms of action, and highlight their application for overcoming the challenges of traditional cancer therapies.
Collapse
Affiliation(s)
- Xiao Yin Ma
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brett D Hill
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Trang Hoang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
13
|
Mills JA, Humphries J, Simpson JD, Sonderegger SE, Thurecht KJ, Fletcher NL. Modulating Macrophage Clearance of Nanoparticles: Comparison of Small-Molecule and Biologic Drugs as Pharmacokinetic Modifiers of Soft Nanomaterials. Mol Pharm 2022; 19:4080-4097. [PMID: 36069540 DOI: 10.1021/acs.molpharmaceut.2c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nanomedicines show benefits in overcoming the limitations of conventional drug delivery systems by reducing side effects, toxicity, and exhibiting enhanced pharmacokinetic (PK) profiles to improve the therapeutic window of small-molecule drugs. However, upon administration, many nanoparticles (NPs) prompt induction of host innate immune responses, which in combination with other clearance pathways such as renal and hepatic, eliminate up to 99% of the administered dose. Here, we explore a drug predosing strategy to transiently suppress the mononuclear phagocyte system (MPS), subsequently improving the PK profile and biological behaviors exhibited by a model NP system [hyperbranched polymers (HBPs)] in an immunocompetent mouse model. In vitro assays allowed the identification of five drug candidates that attenuated cellular association. Predosing of lead compounds chloroquine (CQ) and zoledronic acid (ZA) further showed increased HBP retention within the circulatory system of mice, as shown by both fluorescence imaging and positron emission tomography-computed tomography. Flow cytometric evaluation of spleen and liver tissue cells following intravenous administration further demonstrated that CQ and ZA significantly reduced HBP association with myeloid cells by 23 and 16%, respectively. The results of this study support the use of CQ to pharmacologically suppress the MPS to improve NP PKs.
Collapse
Affiliation(s)
- Jessica A Mills
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - James Humphries
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Joshua D Simpson
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Stefan E Sonderegger
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas L Fletcher
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Qian X, Xu X, Wu Y, Wang J, Li J, Chen S, Wen J, Li Y, Zhang Z. Strategies of engineering nanomedicines for tumor retention. J Control Release 2022; 346:193-211. [PMID: 35447297 DOI: 10.1016/j.jconrel.2022.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 01/29/2023]
Abstract
The retention of therapeutic agents in solid tumors at sufficient concentration and duration is crucial for their antitumor effects. Given the important contribution of nanomedicines to oncology, we herein summarized two major strategies of nanomedicines for tumor retention, such as transformation- and interactions-mediated strategies. The transformation-mediated retention strategy was achieved by enlarging particle size of nanomedicines or modulating the morphology into fibrous structures, while the interactions-mediated retention strategy was accomplished by modulating nanomedicines to promote their interactions with versatile cells or components in tumors. Moreover, we provide some considerations and perspectives of tumor-retaining nanomedicines for effective cancer therapy.
Collapse
Affiliation(s)
- Xindi Qian
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shuo Chen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Jingyuan Wen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
17
|
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]
|
18
|
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
|
19
|
Guo Y, Liu X. Radionanomedicine: Advanced Strategy for Precision Theranostics of Breast Cancer. J Biomed Nanotechnol 2022; 18:50-60. [PMID: 35180899 DOI: 10.1166/jbn.2022.3226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Breast carcinoma remains one of the most common and fatal cancers, and even though a series of general therapeutic approaches have been used to treat breast cancer, their outcomes are significantly affected by a variety of side effects. However, nanomedicine could offer novel strategies for dealing with breast carcinoma. In fact, an increasing number of radionanomedicine approaches have recently been used in both diagnostics and therapy. To highlight this trend, the aim of the current review is to systemically summarize the latest advances in radionanomedicine, including single-modular imaging, multiple-modular imaging, and nanomedicine-based theranostics. Barriers to clinical application, the development of next-generation radionanomedicine, and challenges associated with future design are also discussed.
Collapse
Affiliation(s)
- Yang Guo
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaoying Liu
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
20
|
Ganesh D, Jain P, Shanthamurthy CD, Toraskar S, Kikkeri R. Targeting Selectins Mediated Biological Activities With Multivalent Probes. Front Chem 2021; 9:773027. [PMID: 34926401 PMCID: PMC8677667 DOI: 10.3389/fchem.2021.773027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Selectins are type-I transmembrane glycoproteins that are ubiquitously expressed on activated platelets, endothelial cells, and leukocytes. They bind to cell surface glycoproteins and extracellular matrix ligands, regulate the rolling of leukocytes in the blood capillaries, and recruit them to inflammatory sites. Hence, they are potential markers for the early detection and inhibition of inflammatory diseases, thrombosis, cardiovascular disorders, and tumor metastasis. Fucosylated and sialylated glycans, such as sialyl Lewisx, its isoform sialyl Lewisa, and heparan sulfate, are primary selectin ligands. Functionalization of these selectin-binding ligands on multivalent probes, such as nanoparticles, liposomes, and polymers, not only inhibits selectin-mediated biological activity but is also involved in direct imaging of the inflammation site. This review briefly summarizes the selectin-mediated various diseases such as thrombosis, cancer and recent progress in the different types of multivalent probes used to target selectins.
Collapse
Affiliation(s)
- Deepak Ganesh
- Indian Institute of Science Education and Research, Pune, India
| | - Prashant Jain
- Indian Institute of Science Education and Research, Pune, India
| | | | - Suraj Toraskar
- Indian Institute of Science Education and Research, Pune, India
| | | |
Collapse
|
21
|
Drozdov AS, Nikitin PI, Rozenberg JM. Systematic Review of Cancer Targeting by Nanoparticles Revealed a Global Association between Accumulation in Tumors and Spleen. Int J Mol Sci 2021; 22:13011. [PMID: 34884816 PMCID: PMC8657629 DOI: 10.3390/ijms222313011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/13/2022] Open
Abstract
Active targeting of nanoparticles toward tumors is one of the most rapidly developing topics in nanomedicine. Typically, this strategy involves the addition of cancer-targeting biomolecules to nanoparticles, and studies on this topic have mainly focused on the localization of such formulations in tumors. Here, the analysis of the factors determining efficient nanoparticle targeting and therapy, various parameters such as types of targeting molecules, nanoparticle type, size, zeta potential, dose, and the circulation time are given. In addition, the important aspects such as how active targeting of nanoparticles alters biodistribution and how non-specific organ uptake influences tumor accumulation of the targeted nanoformulations are discussed. The analysis reveals that an increase in tumor accumulation of targeted nanoparticles is accompanied by a decrease in their uptake by the spleen. There is no association between targeting-induced changes of nanoparticle concentrations in tumors and other organs. The correlation between uptake in tumors and depletion in the spleen is significant for mice with intact immune systems in contrast to nude mice. Noticeably, modulation of splenic and tumor accumulation depends on the targeting molecules and nanoparticle type. The median survival increases with the targeting-induced nanoparticle accumulation in tumors; moreover, combinatorial targeting of nanoparticle drugs demonstrates higher treatment efficiencies. Results of the comprehensive analysis show optimal strategies to enhance the efficiency of actively targeted nanoparticle-based medicines.
Collapse
Affiliation(s)
- Andrey S. Drozdov
- Laboratory of Nanobiotechnology, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia;
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Julian M. Rozenberg
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| |
Collapse
|
22
|
Ang MJY, Chan SY, Goh YY, Luo Z, Lau JW, Liu X. Emerging strategies in developing multifunctional nanomaterials for cancer nanotheranostics. Adv Drug Deliv Rev 2021; 178:113907. [PMID: 34371084 DOI: 10.1016/j.addr.2021.113907] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer involves a collection of diseases with a common trait - dysregulation in cell proliferation. At present, traditional therapeutic strategies against cancer have limitations in tackling various tumors in clinical settings. These include chemotherapeutic resistance and the inability to overcome intrinsic physiological barriers to drug delivery. Nanomaterials have presented promising strategies for tumor treatment in recent years. Nanotheranostics combine therapeutic and bioimaging functionalities at the single nanoparticle level and have experienced tremendous growth over the past few years. This review highlights recent developments of advanced nanomaterials and nanotheranostics in three main directions: stimulus-responsive nanomaterials, nanocarriers targeting the tumor microenvironment, and emerging nanomaterials that integrate with phototherapies and immunotherapies. We also discuss the cytotoxicity and outlook of next-generation nanomaterials towards clinical implementation.
Collapse
Affiliation(s)
- Melgious Jin Yan Ang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Siew Yin Chan
- Institute of Materials Research and Engineering, Agency for Science, Technology, and Research, Singapore 138634, Singapore
| | - Yi-Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jun Wei Lau
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore.
| |
Collapse
|
23
|
Teunissen AJP, Burnett ME, Prévot G, Klein ED, Bivona D, Mulder WJM. Embracing nanomaterials' interactions with the innate immune system. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1719. [PMID: 33847441 PMCID: PMC8511354 DOI: 10.1002/wnan.1719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapy has firmly established itself as a compelling avenue for treating disease. Although many clinically approved immunotherapeutics engage the adaptive immune system, therapeutically targeting the innate immune system remains much less explored. Nanomedicine offers a compelling opportunity for innate immune system engagement, as many nanomaterials inherently interact with myeloid cells (e.g., monocytes, macrophages, neutrophils, and dendritic cells) or can be functionalized to target their cell-surface receptors. Here, we provide a perspective on exploiting nanomaterials for innate immune system regulation. We focus on specific nanomaterial design parameters, including size, form, rigidity, charge, and surface decoration. Furthermore, we examine the potential of high-throughput screening and machine learning, while also providing recommendations for advancing the field. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Abraham J. P. Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marianne E. Burnett
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Geoffrey Prévot
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emma D. Klein
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniel Bivona
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Willem J. M. Mulder
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
24
|
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
|
25
|
Lorkowski ME, Atukorale PU, Ghaghada KB, Karathanasis E. Stimuli-Responsive Iron Oxide Nanotheranostics: A Versatile and Powerful Approach for Cancer Therapy. Adv Healthc Mater 2021; 10:e2001044. [PMID: 33225633 PMCID: PMC7933107 DOI: 10.1002/adhm.202001044] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/14/2020] [Indexed: 12/16/2022]
Abstract
Recent advancements in unravelling elements of cancer biology involved in disease progression and treatment resistance have highlighted the need for a holistic approach to effectively tackle cancer. Stimuli-responsive nanotheranostics based on iron oxide nanoparticles are an emerging class of versatile nanomedicines with powerful capabilities to "seek, sense, and attack" multiple components of solid tumors. In this work, the rationale for using iron oxide nanoparticles and the basic physical principles that impact their function in biomedical applications are reviewed. Subsequently, recent advances in the integration of iron oxide nanoparticles with various stimulus mechanisms to facilitate the development of stimuli-responsive nanotheranostics for application in cancer therapy are summarized. The integration of an iron oxide core with various surface coating mechanisms results in the generation of hybrid nanoconstructs with capabilities to codeliver a wide variety of highly potent anticancer therapeutics and immune modulators. Finally, emerging future directions and considerations for their clinical translation are touched upon.
Collapse
Affiliation(s)
- 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
| | - Prabhani U. Atukorale
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ketan B. Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children’s Hospital, Houston, Texas, USA
- Department of Radiology, Baylor College of Medicine, Houston, Texas, 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
|
26
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
27
|
Xiao K, Liu Q, Suby N, Xiao W, Agrawal R, Vu M, Zhang H, Luo Y, Li Y, Lam KS. LHRH-Targeted Redox-Responsive Crosslinked Micelles Impart Selective Drug Delivery and Effective Chemotherapy in Triple-Negative Breast Cancer. Adv Healthc Mater 2021; 10:e2001196. [PMID: 33200571 PMCID: PMC7858235 DOI: 10.1002/adhm.202001196] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/20/2020] [Indexed: 02/05/2023]
Abstract
Systemic chemotherapy is efficacious against triple-negative breast cancer (TNBC), but it is often associated with serious side effects. Here, a luteinizing hormone-releasing hormone (LHRH) receptor-targeted and tumor microenvironment-responsive nanoparticle system to selectively deliver chemotherapeutic drugs to TNBC cells, is reported. This delivery system (termed "LHRH-DCMs") contains poly(ethylene glycol) and dendritic cholic acid as a micellar carrier, reversible intra-micellar disulfide bond as a redox-responsive crosslink, and synthetic high-affinity (D-Lys)-LHRH peptide as a targeting moiety. LHRH-DCMs exhibit high drug loading efficiency, optimal particle size, good colloidal stability, and glutathione-responsive drug release. As expected, LHRH-DCMs are more efficiently internalized into human TNBC cells through receptor-mediated endocytosis, resulting in stronger cytotoxicity against these cancer cells than the non-targeted counterpart when encapsulated with paclitaxel (PTX). Furthermore, near-infrared fluorescence and magnetic resonance imaging demonstrate that LHRH-DCMs facilitate the tumor distribution and penetration of payloads in three different animal models of breast cancer, including cell line-derived xenograft (CDX), patient-derived xenograft (PDX), and transgenic mammary carcinoma. Finally, in vivo therapeutic studies show that PTX-LHRH-DCMs outperform both the corresponding nontargeted PTX-DCMs and the current clinical formulation (Taxol) in an orthotopic TNBC model. These results provide new insights into approaches for precise drug delivery of TNBC.
Collapse
Affiliation(s)
- Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Qiangqiang Liu
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Nell Suby
- Department of Obstetrics and Gynecology, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Rinki Agrawal
- Department of Obstetrics and Gynecology, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Michael Vu
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Hongyong Zhang
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Yan Luo
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Yuanpei Li
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, 95817, USA
| |
Collapse
|
28
|
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
|
29
|
Gong N, Sheppard NC, Billingsley MM, June CH, Mitchell MJ. Nanomaterials for T-cell cancer immunotherapy. NATURE NANOTECHNOLOGY 2021; 16:25-36. [PMID: 33437036 DOI: 10.1038/s41565-020-00822-y] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
T-cell-based immunotherapies hold promise for the treatment of many types of cancer, with three approved products for B-cell malignancies and a large pipeline of treatments in clinical trials. However, there are several challenges to their broad implementation. These include insufficient expansion of adoptively transferred T cells, inefficient trafficking of T cells into solid tumours, decreased T-cell activity due to a hostile tumour microenvironment and the loss of target antigen expression. Together, these factors restrict the number of therapeutically active T cells engaging with tumours. Nanomaterials are uniquely suited to overcome these challenges, as they can be rationally designed to enhance T-cell expansion, navigate complex physical barriers and modulate tumour microenvironments. Here, we present an overview of nanomaterials that have been used to overcome clinical barriers to T-cell-based immunotherapies and provide our outlook of this emerging field at the interface of cancer immunotherapy and nanomaterial design.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil C Sheppard
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Akhter MH, Beg S, Tarique M, Malik A, Afaq S, Choudhry H, Hosawi S. Receptor-based targeting of engineered nanocarrier against solid tumors: Recent progress and challenges ahead. Biochim Biophys Acta Gen Subj 2020; 1865:129777. [PMID: 33130062 DOI: 10.1016/j.bbagen.2020.129777] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Background In past few decades, the research on engineered nanocarriers (NCs) has gained significant attention in cancer therapy due to selective delivery of drug molecules on the diseased cells thereby preventing unwanted uptake into healthy cells to cause toxicity. Scope of review The applicability of enhanced permeability and retention (EPR) effect for the delivery of nanomedicines in cancer therapy has gained limited success due to poor accessibility of the drugs to the target cells where non-specific payload delivery to the off target region lack substantial reward over the conventional therapeutic systems. Major conclusions In spite of the fact, nanomedicines fabricated from the biocompatible nanocarriers have reduced targeting potential for meaningful clinical benefits. However, over expression of receptors on the tumor cells provides opportunity to design functional nanomedicine to bind substantially and deliver therapeutics to the cells or tissues of interest by alleviating the bio-toxicity and unwanted effects. This critique will give insight into the over expressed receptor in various tumor and targeting potential of functional nanomedicine as new therapeutic avenues for effective treatment. General significance This review shortly shed light on EPR-based drug targeting using nanomedicinal strategies, their limitation, and advances in therapeutic targeting to the tumor cells.
Collapse
Affiliation(s)
- Md Habban Akhter
- Department of Pharmaceutics, Faculty of Pharmacy, DIT University, Dehradun, India
| | - Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Mohammed Tarique
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Arshi Malik
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Biochemistry, Cancer Metabolism & Epigenetic Unit, Faculty of Science, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Villanueva-Flores F, Castro-Lugo A, Ramírez OT, Palomares LA. Understanding cellular interactions with nanomaterials: towards a rational design of medical nanodevices. NANOTECHNOLOGY 2020; 31:132002. [PMID: 31770746 PMCID: PMC7105107 DOI: 10.1088/1361-6528/ab5bc8] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/28/2019] [Accepted: 11/26/2019] [Indexed: 05/05/2023]
Abstract
Biomedical applications increasingly require fully characterized new nanomaterials. There is strong evidence showing that nanomaterials not only interact with cells passively but also actively, mediating essential molecular processes for the regulation of cellular functions, but we are only starting to understand the mechanisms of those interactions. Systematic studies about cell behavior as a response to specific nanoparticle properties are scarce in the literature even when they are necessary for the rational design of medical nanodevices. Information in the literature shows that the physicochemical properties determine the bioactivity, biocompatibility, and safety of nanomaterials. The information available regarding the interaction and responses of cells to nanomaterials has not been analyzed and discussed in a single document. Hence, in this review, we present the latest advances about cellular responses to nanomaterials and integrate the available information into concrete considerations for the development of innovative, efficient, specific and, more importantly, safe biomedical nanodevices. We focus on how physicochemical nanoparticle properties (size, chemical surface, shape, charge, and topography) influence cell behavior in a first attempt to provide a practical guide for designing medical nanodevices, avoiding common experimental omissions that may lead to data misinterpretation. Finally, we emphasize the importance of the systematic study of nano-bio interactions to acquire sufficient reproducible information that allows accurate control of cell behavior based on tuning of nanomaterial properties. This information is useful to guide the design of specific nanodevices and nanomaterials to elicit desired cell responses, like targeting, drug delivery, cell attachment, differentiation, etc, or to avoid undesired side effects.
Collapse
Affiliation(s)
- Francisca Villanueva-Flores
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| | - Andrés Castro-Lugo
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| | - Octavio T Ramírez
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| | - Laura A Palomares
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| |
Collapse
|
34
|
Wortzel I, Dror S, Kenific CM, Lyden D. Exosome-Mediated Metastasis: Communication from a Distance. Dev Cell 2020; 49:347-360. [PMID: 31063754 DOI: 10.1016/j.devcel.2019.04.011] [Citation(s) in RCA: 857] [Impact Index Per Article: 171.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
Metastasis, a critical phase of tumor progression, remains a primary challenge in treating cancer and a major cause of cancer mortality. Cell-cell communication via extracellular vesicles (exosomes and microvesicles) between primary tumor cells and the microenvironment of distant organs is crucial for pre-metastatic niche (PMN) formation and metastasis. Here, we review work on the contribution of exosome cargo to cancer progression, the role of exosomes in PMN establishment, and the function of exosomes in organotropic metastasis. We also describe the clinical utility of exosomes.
Collapse
Affiliation(s)
- Inbal Wortzel
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Shani Dror
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Candia M Kenific
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
35
|
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
|
36
|
Covarrubias G, He F, Raghunathan S, Turan O, Peiris PM, Schiemann WP, Karathanasis E. Effective treatment of cancer metastasis using a dual-ligand nanoparticle. PLoS One 2019; 14:e0220474. [PMID: 31356633 PMCID: PMC6663022 DOI: 10.1371/journal.pone.0220474] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Metastasis is responsible for the majority of deaths of breast cancer patients. While cytotoxic drugs are available with high potency to kill breast cancer cells, they are not designed to specifically seek and navigate in the dynamic and continuously changing microenvironment of metastatic disease. To effectively delivery chemotherapeutic agents to metastasis, we designed a dual-ligand nanoparticle loaded with doxorubicin by using two different types of ligands targeting EGFR and αvβ3 integrin. Metastatic cancer cells continuously change resulting in heterogeneity even across adjacent micrometastatic regions with variable expression of these targetable receptors. Using a mouse model of breast cancer metastasis, in vivo and ex vivo imaging showed that both EGFR and αvβ3 integrin-targeting were required to reliably direct the nanoparticle to metastasis and capture the spread and exact topology of the disease. Survival studies compared the anticancer efficacy of the standard drug, EGFR-targeting nanoparticle, αvβ3 integrin-targeting nanoparticle and the dual-ligand nanoparticle. While all the other treatments produced moderate therapeutic outcomes, treatment with the dual-ligand nanoparticle yielded significant improvement and event-free survival in a mouse model of breast cancer metastasis.
Collapse
Affiliation(s)
- Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Felicia He
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Shruti Raghunathan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Oguz Turan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Pubudu M. Peiris
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - William P. Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
37
|
Are Integrins Still Practicable Targets for Anti-Cancer Therapy? Cancers (Basel) 2019; 11:cancers11070978. [PMID: 31336983 PMCID: PMC6678560 DOI: 10.3390/cancers11070978] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023] Open
Abstract
Correlative clinical evidence and experimental observations indicate that integrin adhesion receptors, in particular those of the αV family, are relevant to cancer cell features, including proliferation, survival, migration, invasion, and metastasis. In addition, integrins promote events in the tumor microenvironment that are critical for tumor progression and metastasis, including tumor angiogenesis, matrix remodeling, and the recruitment of immune and inflammatory cells. In spite of compelling preclinical results demonstrating that the inhibition of integrin αVβ3/αVβ5 and α5β1 has therapeutic potential, clinical trials with integrin inhibitors targeting those integrins have repeatedly failed to demonstrate therapeutic benefits in cancer patients. Here, we review emerging integrin functions and their proposed contribution to tumor progression, discuss preclinical evidence of therapeutic significance, revisit clinical trial results, and consider alternative approaches for their therapeutic targeting in oncology, including targeting integrins in the other cells of the tumor microenvironment, e.g., cancer-associated fibroblasts and immune/inflammatory cells. We conclude that integrins remain a valid target for cancer therapy; however, agents with better pharmacological properties, alternative models for their preclinical evaluation, and innovative combination strategies for clinical testing (e.g., together with immuno-oncology agents) are needed.
Collapse
|
38
|
Covarrubias G, Cha A, Rahmy A, Lorkowski M, Perera V, Erokwu BO, Flask C, Peiris PM, Schiemann WP, Karathanasis E. Imaging breast cancer using a dual-ligand nanochain particle. PLoS One 2018; 13:e0204296. [PMID: 30335750 PMCID: PMC6193613 DOI: 10.1371/journal.pone.0204296] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/05/2018] [Indexed: 12/26/2022] Open
Abstract
Nanoparticles often only exploit the upregulation of a receptor on cancer cells to enhance intratumoral deposition of therapeutic and imaging agents. However, a single targeting moiety assumes that a tumor is homogenous and static. Tumoral microenvironments are both heterogenous and dynamic, often displaying variable spatial and temporal expression of targetable receptors throughout disease progression. Here, we evaluated the in vivo performance of an iron oxide nanoparticle in terms of targeting and imaging of orthotropic mouse models of aggressive breast tumors. The nanoparticle, a multi-component nanochain, was comprised of 3–5 iron oxide nanoparticles chemically linked in a linear chain. The nanoparticle’s surface was decorated with two types of ligands each targeting two different upregulated biomarkers on the tumor endothelium, P-selectin and fibronectin. The nanochain exhibited improved tumor deposition not only through vascular targeting but also through its elongated structure. A single-ligand nanochain exhibited a ~2.5-fold higher intratumoral deposition than a spherical nanoparticle variant. Furthermore, the dual-ligand nanochain exhibited higher consistency in generating detectable MR signals compared to a single-ligand nanochain. Using a 7T MRI, the dual-ligand nanochains exhibited highly detectable MR signal within 3h after injection in two different animal models of breast cancer.
Collapse
Affiliation(s)
- Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Anthony Cha
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Abdelrahman Rahmy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Morgan Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Vindya Perera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Bernadette O. Erokwu
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Chris Flask
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Pubudu M. Peiris
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - William P. Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
39
|
Skouras A, Papadia K, Mourtas S, Klepetsanis P, Antimisiaris SG. Multifunctional doxorubicin-loaded magnetoliposomes with active and magnetic targeting properties. Eur J Pharm Sci 2018; 123:162-172. [PMID: 30041027 DOI: 10.1016/j.ejps.2018.07.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 01/30/2023]
Abstract
Multifunctional magnetoliposomes (MLs) with active and magnetic targeting potential are evaluated as platform systems for drug targeting applications. USPIO-encapsulating MLs are prepared by freeze drying/extrusion, decorated with one or two ligands for brain or cancer targeting (t-MLs), and actively loaded with Doxorubicin (DOX). MLs have mean diameters between 117 and 171 nm. Ligand attachment yields and DOX-loading efficiency are sufficiently high, 78-95% and 89-92%, respectively, while DOX loading and retention is not affected by co-entrapment of USPIOs, and USPIO loading/retention is not modulated by DOX. Attachment of ligands, also does not affect DOX or USPIO loading. Interestingly, MLs have high magnetophoretic mobility (MM) compared to free USPIOs, which is not affected by surface coating with PEG (up to 8 mol%), but is slightly reduced by Chol incorporation in their membrane, or when functional groups are immobilized on their surface. ML size, (directly related to number of USPIOs entrapped per vesicle), is the most important MM-determining factor. MM increases by 570% when ML size increases from 69 to 348 nm. Targeting potential of t-MLs is verified by enhanced: (i) transport across a cellular model of the blood-brain-barrier, and (ii) anti-proliferative effect towards B16 melanoma cells. The potential of further enhancing t-ML targeting magnetically is verified by additional enhancements of (i) and (ii), when experiments are performed under a permanent magnetic field.
Collapse
Affiliation(s)
- Athanasios Skouras
- Laboratory of Pharmaceutical Technology, Dept. of Pharmacy, School of Health Sciences, University of Patras, Rio 26510, Greece
| | - Konstantina Papadia
- Laboratory of Pharmaceutical Technology, Dept. of Pharmacy, School of Health Sciences, University of Patras, Rio 26510, Greece
| | - Spyridon Mourtas
- Laboratory of Pharmaceutical Technology, Dept. of Pharmacy, School of Health Sciences, University of Patras, Rio 26510, Greece
| | - Pavlos Klepetsanis
- Laboratory of Pharmaceutical Technology, Dept. of Pharmacy, School of Health Sciences, University of Patras, Rio 26510, Greece; Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Rio 26506, Patras, Greece
| | - Sophia G Antimisiaris
- Laboratory of Pharmaceutical Technology, Dept. of Pharmacy, School of Health Sciences, University of Patras, Rio 26510, Greece; Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Rio 26506, Patras, Greece.
| |
Collapse
|