1
|
Jia S, Bode AM, Chen X, Luo X. Unlocking the potential: Targeting metabolic pathways in the tumor microenvironment for Cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189166. [PMID: 39111710 DOI: 10.1016/j.bbcan.2024.189166] [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: 02/26/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/13/2024]
Abstract
Cancer incidence and mortality are increasing and impacting global life expectancy. Metabolic reprogramming in the tumor microenvironment (TME) is intimately related to tumorigenesis, progression, metastasis and drug resistance. Tumor cells drive metabolic reprogramming of other cells in the TME through metabolic induction of cytokines and metabolites, and metabolic substrate competition. Consequently, this boosts tumor cell growth by providing metabolic support and facilitating immunosuppression and angiogenesis. The metabolic interplay in the TME presents potential therapeutic targets. Here, we focus on the metabolic reprogramming of four principal cell subsets in the TME: CAFs, TAMs, TILs and TECs, and their interaction with tumor cells. We also summarize medications and therapies targeting these cells' metabolic pathways, particularly in the context of immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Siyuan Jia
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China.
| | - Xiangjian Luo
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
2
|
Wang X, Yin X. Panobinostat inhibits breast cancer progression via Vps34-mediated exosomal pathway. Hum Cell 2023; 36:366-376. [PMID: 36329365 DOI: 10.1007/s13577-022-00812-3] [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: 05/10/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Exosomes play crucial roles in intercellular communication, including tumor metastasis. Panobinostat (LBH589), a histone deacetylases (HDAC) inhibitor, is an emerging anti-tumor drug with promising efficacy in cancer therapy. This study was set out from recent evidence that exosome was a mechanism of intercellular drug transfer with significant pharmacological consequences. It enlightened us LBH589 might regulate tumor growth through exosomal secretion. Here we demonstrated LBH589 induced autophagy and facilitated secretory autophagy. Furthermore, LBH589 dose- and time-dependently stimulated exosomal release mediated by Vps34/Rab5C pathway, documented by the ablation of Vps34 and/or Rab5C in breast cancer cells. Additionally, the findings also presented LBH589 inhibited breast cancer progression via exosomes. Altogether, we revealed a novel mechanism of LBH589 in exosome-mediated anti-tumor effects in breast cancer. The schematic diagram of signaling pathways involved in the suppression of breast cancer progression by LBH589 via exosomes.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology, School of Basic Medicine Qingdao University, 308 Ningxia Road, Qingdao, 266071, China.
| | - Xuzhi Yin
- Department of Commercial Operation, Akesobio, Guangzhou, 528437, China
| |
Collapse
|
3
|
Nowacka M, Ginter-Matuszewska B, Świerczewska M, Sterzyńska K, Nowicki M, Januchowski R. Effect of ALDH1A1 Gene Knockout on Drug Resistance in Paclitaxel and Topotecan Resistant Human Ovarian Cancer Cell Lines in 2D and 3D Model. Int J Mol Sci 2022; 23:3036. [PMID: 35328460 PMCID: PMC8950618 DOI: 10.3390/ijms23063036] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the most common cause of gynecological cancer death. Cancer Stem Cells (CSCs) characterized by drug transporters and extracellular matrix (ECM) molecules expression are responsible for drug resistance development. The goal of our study was to examine the role of aldehyde dehydrogenase 1A1 (ALDH1A1) expression in paclitaxel (PAC) and topotecan (TOP) resistant ovarian cancer cell lines. In both cell lines, we knocked out the ALDH1A1 gene using the CRISPR/Cas9 technique. Additionally, we derived an ALDH1A1 positive TOP-resistant cell line with ALDH1A1 expression in all cells via clonal selection. The effect of ALDH1A1 gene knockout or clonal selection on the expression of ALDH1A1, drug transporters (P-gp and BCRP), and ECM (COL3A1) was determined by Q-PCR, Western blot and immunofluorescence. Using MTT assay, we compared drug resistance in two-dimensional (2D) and three-dimensional (3D) cell culture conditions. We did not observe any effect of ALDH1A1 gene knockout on MDR1/P-gp expression and drug resistance in the PAC-resistant cell line. The knockout of ALDH1A1 in the TOP-resistant cell line resulted in a moderate decrease of BCRP and COL3A1 expression and weakened TOP resistance. The clonal selection of ALDH1A1 cells resulted in very strong downregulation of BCPR and COL3A1 expression and overexpression of MDR1/P-gp. This finally resulted in decreased resistance to TOP but increased resistance to PAC. All spheroids were more resistant than cells growing as monolayers, but the resistance mechanism differs. The spheroids' resistance may result from the presence of cell zones with different proliferation paces, the density of the spheroid, ECM expression, and drug capacity to diffuse into the spheroid.
Collapse
Affiliation(s)
- Marta Nowacka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland; (M.Ś.); (K.S.); (M.N.)
| | - Barbara Ginter-Matuszewska
- Department of Infectious Diseases, Hepatology and Acquired Immunodeficiency, Poznan University of Medical Sciences, 61-003 Poznan, Poland;
| | - Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland; (M.Ś.); (K.S.); (M.N.)
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland; (M.Ś.); (K.S.); (M.N.)
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland; (M.Ś.); (K.S.); (M.N.)
| | - Radosław Januchowski
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zyty 28 St., 65-046 Zielona Gora, Poland;
| |
Collapse
|
4
|
Revisiting Flubendazole Through Nanocrystal Technology: Statistical Design, Characterization and Its Potential Inhibitory Effect on Xenografted Lung Tumor Progression in Mice. J CLUST SCI 2022. [DOI: 10.1007/s10876-022-02220-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
5
|
Chaudhary B, Kumar P, Arya P, Singla D, Kumar V, Kumar D, S R, Wadhwa S, Gulati M, Singh SK, Dua K, Gupta G, Gupta MM. Recent Developments in the Study of the Microenvironment of Cancer and Drug Delivery. Curr Drug Metab 2022; 23:1027-1053. [PMID: 36627789 DOI: 10.2174/1389200224666230110145513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/20/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023]
Abstract
Cancer is characterized by disrupted molecular variables caused by cells that deviate from regular signal transduction. The uncontrolled segment of such cancerous cells annihilates most of the tissues that contact them. Gene therapy, immunotherapy, and nanotechnology advancements have resulted in novel strategies for anticancer drug delivery. Furthermore, diverse dispersion of nanoparticles in normal stroma cells adversely affects the healthy cells and disrupts the crosstalk of tumour stroma. It can contribute to cancer cell progression inhibition and, conversely, to acquired resistance, enabling cancer cell metastasis and proliferation. The tumour's microenvironment is critical in controlling the dispersion and physiological activities of nano-chemotherapeutics which is one of the targeted drug therapy. As it is one of the methods of treating cancer that involves the use of medications or other substances to specifically target and kill off certain subsets of malignant cells. A targeted therapy may be administered alone or in addition to more conventional methods of care like surgery, chemotherapy, or radiation treatment. The tumour microenvironment, stromatogenesis, barriers and advancement in the drug delivery system across tumour tissue are summarised in this review.
Collapse
Affiliation(s)
- Benu Chaudhary
- Department of Pharmacology, Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Parveen Kumar
- Department of Life Science, Shri Ram College of Pharmacy, Karnal, Haryana, India
| | - Preeti Arya
- Department of Pharmacology, Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Deepak Singla
- Department of Pharmacology, Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Virender Kumar
- Department of Pharmacology, Swami Dayanand Post Graduate Institute of Pharmaceutical Sciences, Rohtak, Haryana, India
| | - Davinder Kumar
- Department of Pharmacology, Swami Dayanand Post Graduate Institute of Pharmaceutical Sciences, Rohtak, Haryana, India
| | - Roshan S
- Department of Pharmacology, Deccan School of Pharmacy, Hyderabad, India
| | - Sheetu Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Madan Mohan Gupta
- Faculty of Medical Sciences, School of Pharmacy, The University of the West Indies, St. Augustine, Trinidad & Tobago, West Indies
| |
Collapse
|
6
|
Farrakhova D, Maklygina Y, Romanishkin I, Yakovlev D, Plyutinskaya A, Bezdetnaya L, Loschenov V. Fluorescence imaging analysis of distribution of indocyanine green in molecular and nanoform in tumor model. Photodiagnosis Photodyn Ther 2021; 37:102636. [PMID: 34808398 DOI: 10.1016/j.pdpdt.2021.102636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The efficient intraoperative identification of tumors requires the development of highly specific near-infrared (NIR) probes as contrast agents. One of the most effective dyes existing in clinic oncology is Indocyanine Green (ICG). However, ICG has a rapid excretion, thus ruling out its extended accumulation in pathological tissues therefore limiting its clinical applications. ICG colloid solution (ICG NPs) consists predominantly of J-aggregates and to a lesser extent of H-aggregates and monomers. In the present study we assessed the spectral properties of ICG nanoforms in preclinical models. METHODS We used optical spectroscopy and video fluorescence navigation to monitor accumulation and distribution of ICG monomers and ICG NPs in various tissues in mice with xenografted laryngopharyngeal carcinoma after intravenous drugs injection. RESULTS After i.v. injection, the molecular form of ICG was not retained in the tumor and its circulation cycle averaged 5 min. Alternatively, the nanoform of the drug had a different pharmacokinetics, reaching maximum accumulation 24 h after intravenous injection. Moreover, once in the circulation, we observed a progressive accumulation in the tumor of both ICG H-aggregates and ICG monomers, but not J-aggregates. CONCLUSION Spectral characteristics of ICG NPs indicated the presence of several fractions, namely, J- and H-aggregates along with molecular forms. These fractions had different fluorescence spectra, allowing us to track the transformation of the drug in vivo conditions. After ICG NPs administration, J-aggregates induce accumulation of monomeric forms in the tumor, enabling extended intraoperative diagnostic, and as such further studies of J-aggregates for theranostic applications in oncological surgery are of great interest.
Collapse
Affiliation(s)
- Dina Farrakhova
- Prokhorov General Physics Institute of the Russian Academy of Science, Vavilova str.38, Moscow 119991, Russia.
| | - Yulia Maklygina
- Prokhorov General Physics Institute of the Russian Academy of Science, Vavilova str.38, Moscow 119991, Russia
| | - Igor Romanishkin
- Prokhorov General Physics Institute of the Russian Academy of Science, Vavilova str.38, Moscow 119991, Russia
| | - Dmitry Yakovlev
- Prokhorov General Physics Institute of the Russian Academy of Science, Vavilova str.38, Moscow 119991, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Science, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia
| | - Anna Plyutinskaya
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 2nd Botkin Ave. 3, Moscow 125284, Russia
| | - Lina Bezdetnaya
- Centre de Recherche en Automatique de Nancy, CNRS, Université de Lorraine, Campus Sciences Boulevard des Aiguillettes BP 70239 54506 Vandoeuvre Les Nancy Cedex, Vandoeuvre-lès-Nancy 54519, France; Institut de Cancérologie de Lorraine, 6 Av. de Bourgogne, Vandoeuvre-lès-Nancy 54519, France
| | - Victor Loschenov
- Prokhorov General Physics Institute of the Russian Academy of Science, Vavilova str.38, Moscow 119991, Russia; National Research Nuclear University "MEPhI", Kashirskoe shosse, 31, Moscow 115409, Russia
| |
Collapse
|
7
|
Pengnam S, Plianwong S, Yingyongnarongkul BE, Patrojanasophon P, Opanasopit P. Delivery of small interfering RNAs by nanovesicles for cancer therapy. Drug Metab Pharmacokinet 2021; 42:100425. [PMID: 34954489 DOI: 10.1016/j.dmpk.2021.100425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022]
Abstract
Small interfering ribonucleic acids (siRNAs) are originally recognized as an intermediate of the RNA interference (RNAi) pathway. They can inhibit or silence various cellular pathways by knocking down specific messenger RNA molecules. In cancer cells, siRNAs can suppress the expression of several multidrug-resistant genes, leading to the increased deposition of chemotherapeutic drugs at the tumor site. siRNA therapy can be used to selectively increase apoptosis of cancer cells or activate an immune response to the cancer. However, delivering siRNAs to the targeted location is the main limitation in achieving safe and effective delivery of siRNAs. This review highlights some representative examples of nonviral delivery systems, especially nanovesicles such as exosomes, liposomes, and niosomes. Nanovesicles can improve the delivery of siRNAs by increasing their intracellular delivery, and they have demonstrated excellent potential for cancer therapy. This review focuses on recent discoveries of siRNA targets for cancer therapy and the use of siRNAs to successfully silence these targets. In addition, this review summarizes the recent progress in designing nanovesicles (liposomes or niosomes) for siRNA delivery to cancer cells and the effects of a combination of anticancer drugs and siRNA therapy in cancer therapy.
Collapse
Affiliation(s)
- Supusson Pengnam
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | | | - Boon-Ek Yingyongnarongkul
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| |
Collapse
|
8
|
Wang J, Zhou M, Chen F, Liu X, Gao J, Wang W, Wang H, Yu H. Stimuli-Sheddable Nanomedicine Overcoming Pathophysiological Barriers for Potentiating Immunotherapy of Cancer. J Biomed Nanotechnol 2021; 17:1486-1509. [PMID: 34544528 DOI: 10.1166/jbn.2021.3134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immunotherapy displays potent potential for clinical cancer management by activating the protective immune response; however, the microenvironment of the immunosuppressive tumor restricts the efficiency of immunotherapies. Along with the complex pathophysiological barrier of the solid tumors, successful immunotherapeutic delivery remains a formidable challenge for conventional nanomedicine. Stimuli-sheddable nano vectors may facilitate the delivery of cargoes to tumors with minimal premature cargo leakage in blood circulation while enhancing the tumor penetration of nanomedicines by deshielding the polyethylene glycol (PEG) corona upon endogenous activity such as acidity, enzymes and glutathione, or external stimuli, such as laser irradiation. Throughout this study, researchers overviewed the recent advances of nanomedicine-based cancer immunotherapy using the stimuli-responsive deshielding nano vectors, which allowed researchers to integrate multiple therapeutic regimens for inducing immunogenic cell death. This aided in blocking the immune checkpoints, repolarizing the macrophages, and regulating the kynurenine metabolism. Furthermore, researchers discussed the critical issues in the development of stimuli-sheddable nanoimmunodulators, primarily aimed at speeding up their clinical translation. Finally, researchers provided novel perspectives for improving cancer management with the stimuli-sheddable nanomedicine.
Collapse
Affiliation(s)
- Jiaxin Wang
- College of Chemistry and Chemical Engineering, Inner Magnolia University, Huhhot, 010021, China
| | - Mengxue Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Fangmin Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jin Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Hui Wang
- College of Chemistry and Chemical Engineering, Inner Magnolia University, Huhhot, 010021, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
9
|
Zhang BB, Chen XJ, Fan XD, Zhu JJ, Wei YH, Zheng HS, Zheng HY, Wang BH, Piao JG, Li FZ. Lipid/PAA-coated mesoporous silica nanoparticles for dual-pH-responsive codelivery of arsenic trioxide/paclitaxel against breast cancer cells. Acta Pharmacol Sin 2021; 42:832-842. [PMID: 33824461 PMCID: PMC8182795 DOI: 10.1038/s41401-021-00648-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 03/12/2021] [Indexed: 02/01/2023] Open
Abstract
Nanomedicine has attracted increasing attention and emerged as a safer and more effective modality in cancer treatment than conventional chemotherapy. In particular, the distinction of tumor microenvironment and normal tissues is often used in stimulus-responsive drug delivery systems for controlled release of therapeutic agents at target sites. In this study, we developed mesoporous silica nanoparticles (MSNs) coated with polyacrylic acid (PAA), and pH-sensitive lipid (PSL) for synergistic delivery and dual-pH-responsive sequential release of arsenic trioxide (ATO) and paclitaxel (PTX) (PL-PMSN-PTX/ATO). Tumor-targeting peptide F56 was used to modify MSNs, which conferred a target-specific delivery to cancer and endothelial cells under neoangiogenesis. PAA- and PSL-coated nanoparticles were characterized by TGA, TEM, FT-IR, and DLS. The drug-loaded nanoparticles displayed a dual-pH-responsive (pHe = 6.5, pHendo = 5.0) and sequential drug release profile. PTX within PSL was preferentially released at pH = 6.5, whereas ATO was mainly released at pH = 5.0. Drug-free carriers showed low cytotoxicity toward MCF-7 cells, but ATO and PTX co-delivered nanoparticles displayed a significant synergistic effect against MCF-7 cells, showing greater cell-cycle arrest in treated cells and more activation of apoptosis-related proteins than free drugs. Furthermore, the extracellular release of PTX caused an expansion of the interstitial space, allowing deeper penetration of the nanoparticles into the tumor mass through a tumor priming effect. As a result, FPL-PMSN-PTX/ATO exhibited improved in vivo circulation time, tumor-targeted delivery, and overall therapeutic efficacy.
Collapse
Affiliation(s)
- Bing-Bing Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiao-Jie Chen
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xu-Dong Fan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jing-Jing Zhu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ying-Hui Wei
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hang-Sheng Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hong-Yue Zheng
- Libraries of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bin-Hui Wang
- The Affiliated Municipal Hospital of Taizhou University, Taizhou, 318000, China.
| | - Ji-Gang Piao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Fan-Zhu Li
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
10
|
Rahmanian M, Seyfoori A, Ghasemi M, Shamsi M, Kolahchi AR, Modarres HP, Sanati-Nezhad A, Majidzadeh-A K. In-vitro tumor microenvironment models containing physical and biological barriers for modelling multidrug resistance mechanisms and multidrug delivery strategies. J Control Release 2021; 334:164-177. [PMID: 33895200 DOI: 10.1016/j.jconrel.2021.04.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
The complexity and heterogeneity of the three-dimensional (3D) tumor microenvironment have brought challenges to tumor studies and cancer treatment. The complex functions and interactions of cells involved in tumor microenvironment have led to various multidrug resistance (MDR) and raised challenges for cancer treatment. Traditional tumor models are limited in their ability to simulate the resistance mechanisms and not conducive to the discovery of multidrug resistance and delivery processes. New technologies for making 3D tissue models have shown the potential to simulate the 3D tumor microenvironment and identify mechanisms underlying the MDR. This review overviews the main barriers against multidrug delivery in the tumor microenvironment and highlights the advances in microfluidic-based tumor models with the success in simulating several drug delivery barriers. It also presents the progress in modeling various genetic and epigenetic factors involved in regulating the tumor microenvironment as a noticeable insight in 3D microfluidic tumor models for recognizing multidrug resistance and delivery mechanisms. Further correlation between the results obtained from microfluidic drug resistance tumor models and the clinical MDR data would open up avenues to gain insight into the performance of different multidrug delivery treatment strategies.
Collapse
Affiliation(s)
- Mehdi Rahmanian
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Amir Seyfoori
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Mohsen Ghasemi
- Genetics Department, Breast Cancer Research Center (BCRC), Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Milad Shamsi
- Center for BioEngineering Research and Education (CBRE), University of Calgary, Calgary, Alberta T2N 1N4, Canada; BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Amir Sanati-Nezhad
- Center for BioEngineering Research and Education (CBRE), University of Calgary, Calgary, Alberta T2N 1N4, Canada; BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| | - Keivan Majidzadeh-A
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran; Genetics Department, Breast Cancer Research Center (BCRC), Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran.
| |
Collapse
|
11
|
Nowacka M, Sterzynska K, Andrzejewska M, Nowicki M, Januchowski R. Drug resistance evaluation in novel 3D in vitro model. Biomed Pharmacother 2021; 138:111536. [PMID: 34311534 DOI: 10.1016/j.biopha.2021.111536] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 01/09/2023] Open
Abstract
Ovarian cancer rates the highest mortality among all gynecological malignancies. The main reason for high mortality is the development of drug resistance. It can be related to changes in the expression of many drug resistance genes as well as expression of extracellular matrix proteins and cell density in the tumor. We developed a simple two-dimensional and three-dimensional model of drug sensitive A2780 and resistant to cisplatin and paclitaxel variants of ovarian cancer cell line. Using MTT assay, we compared drug resistance in two-dimensional and three-dimensional cell culture conditions. Real-time polymerase chain reaction analysis was used to compare the expression of drug resistance genes. The expression of proteins in spheroids was determined by immunohistochemistry. We observed a moderate increase in cisplatin resistance and a significant increase in paclitaxel resistance between two-dimensional and three-dimensional cell culture conditions. Our findings show that changes in the expression of drug resistance genes may play a crucial role in the drug resistance of cancer cells in traditional cell culture. On the other hand, the drug resistance in spheroids may result from different mechanisms such as cell density in the spheroid, extracellular matrix proteins expression and drug capacity to diffuse into the spheroid.
Collapse
Affiliation(s)
- Marta Nowacka
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Karolina Sterzynska
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Malgorzata Andrzejewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Radoslaw Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Gora, Zyty 28 St, 65-046 Zielona Gora, Poland.
| |
Collapse
|
12
|
Morey J, Llinás P, Bueno-Costa A, León AJ, Piña MN. Raltitrexed-Modified Gold and Silver Nanoparticles for Targeted Cancer Therapy: Cytotoxicity Behavior In Vitro on A549 and HCT-116 Human Cancer Cells. MATERIALS 2021; 14:ma14030534. [PMID: 33499297 PMCID: PMC7866044 DOI: 10.3390/ma14030534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
Two different raltitrexed gold and silver nanoparticles for the delivery of an antitumoral drug into cancer cells were synthesized and characterized. A cysteine linker was used for the covalent bonding of raltitrexed to the surface of nanoparticles. To evaluate the efficacy of the antifolate-derivative nanoparticles, their cytotoxicity was assayed in vitro with A549 human lung adenocarcinoma and HCT-116 colorectal carcinoma human cells. Modified nanoparticles are a biocompatible material, and administration of silver raltitrexed nanoparticles strongly inhibited the viability of the cancer cells; gold raltitrexed nanoparticles do not show any type of cytotoxic effect. The results suggest that silver raltitrexed nanoparticles could be a potential delivery system for certain cancer cells.
Collapse
Affiliation(s)
- Jeroni Morey
- Department of Chemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (J.M.); (A.J.L.)
| | - Pere Llinás
- Department of Biochemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (P.L.); (A.B.-C.)
| | - Alberto Bueno-Costa
- Department of Biochemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (P.L.); (A.B.-C.)
| | - Alberto J. León
- Department of Chemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (J.M.); (A.J.L.)
| | - M. Nieves Piña
- Department of Chemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (J.M.); (A.J.L.)
- Correspondence: ; Tel.: +34-971-172847
| |
Collapse
|
13
|
Piscatelli JA, Ban J, Lucas AT, Zamboni WC. Complex Factors and Challenges that Affect the Pharmacology, Safety and Efficacy of Nanocarrier Drug Delivery Systems. Pharmaceutics 2021; 13:114. [PMID: 33477395 PMCID: PMC7830329 DOI: 10.3390/pharmaceutics13010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/01/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Major developments in nanomedicines, such as nanoparticles (NPs), nanosomes, and conjugates, have revolutionized drug delivery capabilities over the past four decades. Although nanocarrier agents provide numerous advantages (e.g., greater solubility and duration of systemic exposure) compared to their small-molecule counterparts, there is considerable inter-patient variability seen in the systemic disposition, tumor delivery and overall pharmacological effects (i.e., anti-tumor efficacy and unwanted toxicity) of NP agents. This review aims to provide a summary of fundamental factors that affect the disposition of NPs in the treatment of cancer and why they should be evaluated during preclinical and clinical development. Furthermore, this chapter will highlight some of the translational challenges associated with elements of NPs and how these issues can only be addressed by detailed and novel pharmacology studies.
Collapse
Affiliation(s)
- Joseph A. Piscatelli
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Jisun Ban
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Andrew T. Lucas
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
Haider T, Sandha KK, Soni V, Gupta PN. Recent advances in tumor microenvironment associated therapeutic strategies and evaluation models. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111229. [DOI: 10.1016/j.msec.2020.111229] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/08/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
|
15
|
Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: Designing a new perspective in nanomedicine delivery. Theranostics 2019; 9:8091-8108. [PMID: 31754383 PMCID: PMC6857058 DOI: 10.7150/thno.37204] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
The enhanced permeability and retention (EPR) effect has underlain the predominant nanomedicine design philosophy for the past three decades. However, growing evidence suggests that it is over-represented in preclinical models, and agents designed solely using its principle of passive accumulation can only be applied to a narrow subset of clinical tumors. For this reason, strategies that can improve upon the EPR effect to facilitate nanomedicine delivery to otherwise non-responsive tumors are required for broad clinical translation. EPR-adaptive nanomedicine delivery comprises a class of chemical and physical techniques that modify tumor accessibility in an effort to increase agent delivery and therapeutic effect. In the present review, we overview the primary benefits and limitations of radiation, ultrasound, hyperthermia, and photodynamic therapy as physical strategies for EPR-adaptive delivery to EPR-insensitive tumor phenotypes, and we reflect upon changes in the preclinical research pathway that should be implemented in order to optimally validate and develop these delivery strategies.
Collapse
Affiliation(s)
- Alexander Dhaliwal
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- MD/PhD Program, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON Canada
| |
Collapse
|
16
|
Au JLS, Abbiati RA, Wientjes MG, Lu Z. Target Site Delivery and Residence of Nanomedicines: Application of Quantitative Systems Pharmacology. Pharmacol Rev 2019; 71:157-169. [PMID: 30846487 DOI: 10.1124/pr.118.016816] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Quantitative systems pharmacology (QSP), an emerging field that entails using modeling and computation to interpret, interrogate, and integrate drug effects spanning from the molecule to the whole organism to forecast treatment outcomes, is expected to enhance the efficiency of drug development. Since late 2017, the U.S. Food and Drug Administration has advocated the use of an analogous approach of model-informed drug development. This review focuses on issues pertaining to nanosized medicines (NP) and the potential utility of QSP to determine NP delivery and residence at extracellular or intracellular targets in vivo. The kinetic processes governing NP disposition and transport, interactions with biologic matrix components, binding and internalization in cells, and intracellular trafficking are determined, sometimes jointly, by NP properties (e.g., dimension, materials, surface charge and modifications, shape, and geometry) and target tissue properties (e.g., perfusion status, vessel pore size and wall thickness, vessel and cell density, composition of extracellular matrix, and void volume fraction). These various determinants, together with the heterogeneous tissue structures and microenvironment factors in solid tumors, lead to environment-, spatial-, and time-dependent changes in NP concentrations that are difficult to predict. Adding to the complexity is the recent discovery that NP surface-coating protein corona, whose composition depends on NP properties and which undergoes continuous evolution with time and local protein environments, is yet another unpredictable variable. Examples are provided to demonstrate the potential utility of QSP-based multiscale modeling to capture the physicochemical and biologic processes in equations to enable computational studies of the key kinetic processes in cancer treatments.
Collapse
Affiliation(s)
- Jessie L-S Au
- Institute of Quantitative Systems Pharmacology, Carlsbad, California (J.L.-S.A., R.A.A., M.G.W., Z.L.); Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma (J.L.-S.A., R.A.A.); Optimum Therapeutics LLC, Carlsbad, California (J.L.-S.A., M.G.W., Z.L.); and College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China (J.L.-S.A.)
| | - Roberto A Abbiati
- Institute of Quantitative Systems Pharmacology, Carlsbad, California (J.L.-S.A., R.A.A., M.G.W., Z.L.); Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma (J.L.-S.A., R.A.A.); Optimum Therapeutics LLC, Carlsbad, California (J.L.-S.A., M.G.W., Z.L.); and College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China (J.L.-S.A.)
| | - M Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, Carlsbad, California (J.L.-S.A., R.A.A., M.G.W., Z.L.); Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma (J.L.-S.A., R.A.A.); Optimum Therapeutics LLC, Carlsbad, California (J.L.-S.A., M.G.W., Z.L.); and College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China (J.L.-S.A.)
| | - Ze Lu
- Institute of Quantitative Systems Pharmacology, Carlsbad, California (J.L.-S.A., R.A.A., M.G.W., Z.L.); Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma (J.L.-S.A., R.A.A.); Optimum Therapeutics LLC, Carlsbad, California (J.L.-S.A., M.G.W., Z.L.); and College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China (J.L.-S.A.)
| |
Collapse
|
17
|
Hecel A, Kolkowska P, Krzywoszynska K, Szebesczyk A, Rowinska-Zyrek M, Kozlowski H. Ag+ Complexes as Potential Therapeutic Agents in Medicine and Pharmacy. Curr Med Chem 2019; 26:624-647. [DOI: 10.2174/0929867324666170920125943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 07/28/2017] [Accepted: 08/09/2017] [Indexed: 12/17/2022]
Abstract
Silver is a non-essential element with promising antimicrobial and anticancer properties. This work is a detailed summary of the newest findings on the bioinorganic chemistry of silver, with a special focus on the applications of Ag+ complexes and nanoparticles. The coordination chemistry of silver is given a reasonable amount of attention, summarizing the most common silver binding sites and giving examples of such binding motifs in biologically important proteins. Possible applications of this metal and its complexes in medicine, particularly as antibacterial and antifungal agents and in cancer therapy, are discussed in detail. The most recent data on silver nanoparticles are also summarized.
Collapse
Affiliation(s)
- Aleksandra Hecel
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50383 Wroclaw, Poland
| | - Paulina Kolkowska
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via A. Moro 2, 53100 Siena, Italy
| | - Karolina Krzywoszynska
- Institute of Cosmetology, Public Higher Medical Professional School in Opole, Katowicka 68, 45060 Opole, Poland
| | - Agnieszka Szebesczyk
- Institute of Cosmetology, Public Higher Medical Professional School in Opole, Katowicka 68, 45060 Opole, Poland
| | | | - Henryk Kozlowski
- Institute of Cosmetology, Public Higher Medical Professional School in Opole, Katowicka 68, 45060 Opole, Poland
| |
Collapse
|
18
|
Au JLS, Lu Z, Abbiati RA, Wientjes MG. Systemic Bioequivalence Is Unlikely to Equal Target Site Bioequivalence for Nanotechnology Oncologic Products. AAPS J 2019; 21:24. [PMID: 30710324 PMCID: PMC6432930 DOI: 10.1208/s12248-019-0296-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/09/2019] [Indexed: 11/30/2022] Open
Abstract
Approval of generic drugs by the US Food and Drug Administration (FDA) requires the product to be pharmaceutically equivalent to the reference listed drug (RLD) and demonstrate bioequivalence (BE) in effectiveness when administered to patients under the conditions in the RLD product labeling. Effectiveness is determined by drug exposure at the target sites. However, since such measurement is usually unavailable, systemic exposure is assumed to equal target site exposure and systemic BE to equal target site BE. This assumption, while it often applies to small molecule drug products that are readily dissolved in biological fluids and systemically absorbed, is unlikely to apply to nanotechnology products (NP) that exist as heterogeneous systems and are subjected to dimension- and material-dependent changes. This commentary provides an overview of the intersecting and spatial-dependent processes and variables governing the delivery and residence of oncologic NP in solid tumors. In order to provide a quantitative perspective of the collective effects of these processes, we used quantitative systems pharmacology (QSP) multi-scale modeling to capture the physicochemical and biological events on several scales (whole-body, organ/suborgan, cell/subcellular, spatial locations, time). QSP is an emerging field that entails using modeling and computation to facilitate drug development; an analogous approach (i.e., model-informed drug development) is advocated by to FDA. The QSP model-based simulations illustrated that small changes in NP attributes (e.g., size variations during manufacturing, interactions with proteins in biological milieu) could lead to disproportionately large differences in target site exposure, rending systemic BE unlikely to equal target site BE.
Collapse
Affiliation(s)
- Jessie L-S Au
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA.
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA.
- College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China.
| | - Ze Lu
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| | - Roberto A Abbiati
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA
| | - M Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| |
Collapse
|
19
|
Chen Z, Liu W, Wang X, Liu Y, Li X. Sequential Drug Release to Modulate Collagen Synthesis and Promote Micelle Penetration in Tumors. ACS Biomater Sci Eng 2019; 5:1343-1353. [DOI: 10.1021/acsbiomaterials.8b01600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Zhoujiang Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, 111 North first Section, second Ring Road, Chengdu 610031, P.R. China
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, 668 Jimei Avenue, Xiamen 361021, P. R. China
| | - Weiping Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, 111 North first Section, second Ring Road, Chengdu 610031, P.R. China
| | - Xin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, 111 North first Section, second Ring Road, Chengdu 610031, P.R. China
| | - Yuan Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, 111 North first Section, second Ring Road, Chengdu 610031, P.R. China
| | - Xiaohong Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, 111 North first Section, second Ring Road, Chengdu 610031, P.R. China
| |
Collapse
|
20
|
El-Sawy HS, Al-Abd AM, Ahmed TA, El-Say KM, Torchilin VP. Stimuli-Responsive Nano-Architecture Drug-Delivery Systems to Solid Tumor Micromilieu: Past, Present, and Future Perspectives. ACS NANO 2018; 12:10636-10664. [PMID: 30335963 DOI: 10.1021/acsnano.8b06104] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The microenvironment characteristics of solid tumors, renowned as barriers that harshly impeded many drug-delivery approaches, were precisely studied, investigated, categorized, divided, and subdivided into a complex diverse of barriers. These categories were further studied with a particular perspective, which makes all barriers found in solid-tumor micromilieu turn into different types of stimuli, and were considered triggers that can increase and hasten drug-release targeting efficacy. This review gathers data concerning the nature of solid-tumor micromilieu. Past research focused on the treatment of such tumors, the recent efforts employed for engineering smart nanoarchitectures with the utilization of the specified stimuli categories, the possibility of combining more than one stimuli for much-greater targeting enhancement, examples of the approved nanoarchitectures that already translated clinically as well as the obstacles faced by the use of these nanostructures, and, finally, an overview of the possible future implementations of smart-chemical engineering for the design of more-efficient drug delivery and theranostic systems and for making nanosystems with a much-higher level of specificity and penetrability features.
Collapse
Affiliation(s)
- Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy , Egyptian Russian University , Badr City , Cairo 63514 , Egypt
| | - Ahmed M Al-Abd
- Department of Pharmaceutical Sciences, College of Pharmacy , Gulf Medical University , Ajman , United Arab Emirates
- Pharmacology Department, Medical Division , National Research Centre , Giza 12622 , Egypt
| | - Tarek A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Khalid M El-Say
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , 140 The Fenway, Room 211/214, 360 Huntington Aveue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
21
|
Katti KV, Khoobchandani M, Thipe VC, Al-Yasiri AY, Katti KK, Loyalka SK, Sakr TM, Lugão AB. Prostate tumor therapy advances in nuclear medicine: green nanotechnology toward the design of tumor specific radioactive gold nanoparticles. J Radioanal Nucl Chem 2018. [DOI: 10.1007/s10967-018-6320-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Azizi M, Ghourchian H, Yazdian F, Alizadehzeinabad H. Albumin coated cadmium nanoparticles as chemotherapeutic agent against MDA-MB 231 human breast cancer cell line. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:787-797. [PMID: 29426245 DOI: 10.1080/21691401.2018.1436064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
With the aim of dedicating toxicity of cadmium nanoparticles (CdNPs) against invasive breast cancer, with minimum damage to surrounding healthy cells, CdNPs were coated with albumin nanocarrier by nanoprecipitation method and named CdNPs@BSA. The characterization was done by TEM image, DLS and UV-Vis, fluorescence, circular dichroism spectroscopy. The cytotoxic efficacy of the CdNPs@BSA against human breast cancer cells (MDA-MB 231 cells) was examined by MTT assay. Apoptosis, as the mechanism of cell death, was verified by inverted microscopy, fluorescent microscopy, gel electrophoresis and flow cytometry. The role of ROS generation in apoptosis was also studied. It was found that the resulted CdNPs@BSA (diameter of 88 nm and zeta potential of about -18.85 mV) was suitable for penetration in tumour micro vessels. In the form of CdNPs@BSA, the 77% of the secondary structure and almost all of the tertiary structure remain intact. Comparing to CdNPs, CdNPs@BSA could significantly suppress the MDA-MB 231 while they were less toxic on WBCs. Therefore, they could be a brilliant candidate to be used as a chemotherapeutic agent against invasive breast cancer cells.
Collapse
Affiliation(s)
- Marzieh Azizi
- a Institute of Biochemistry and Biophysics (IBB) , University of Tehran , Tehran , Iran
| | - Hedayatollah Ghourchian
- a Institute of Biochemistry and Biophysics (IBB) , University of Tehran , Tehran , Iran.,b Nanobiomedicine Center of Excellence, Nanoscience and Nanotechnology Research Center , University of Tehran , Tehran , Iran
| | - Fatemeh Yazdian
- c Biological Sciences, Faculty of New Science and Technology , University of Tehran , Tehran , Iran
| | | |
Collapse
|
23
|
Mannancherril V, Therrien B. Strategies toward the Enhanced Permeability and Retention Effect by Increasing the Molecular Weight of Arene Ruthenium Metallaassemblies. Inorg Chem 2017; 57:3626-3633. [DOI: 10.1021/acs.inorgchem.7b02668] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Vidya Mannancherril
- Institut de Chimie, Université de Neuchâtel, Avenue de Bellevaux 51, 2000 Neuchâtel, Switzerland
| | - Bruno Therrien
- Institut de Chimie, Université de Neuchâtel, Avenue de Bellevaux 51, 2000 Neuchâtel, Switzerland
| |
Collapse
|
24
|
Liang C, Li F, Wang L, Zhang ZK, Wang C, He B, Li J, Chen Z, Shaikh AB, Liu J, Wu X, Peng S, Dang L, Guo B, He X, Au DWT, Lu C, Zhu H, Zhang BT, Lu A, Zhang G. Tumor cell-targeted delivery of CRISPR/Cas9 by aptamer-functionalized lipopolymer for therapeutic genome editing of VEGFA in osteosarcoma. Biomaterials 2017; 147:68-85. [PMID: 28938163 DOI: 10.1016/j.biomaterials.2017.09.015] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/02/2017] [Accepted: 09/08/2017] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OS) is a highly aggressive pediatric cancer, characterized by frequent lung metastasis and pathologic bone destruction. Vascular endothelial growth factor A (VEGFA), highly expressed in OS, not only contributes to angiogenesis within the tumor microenvironment via paracrine stimulation of vascular endothelial cells, but also acts as an autocrine survival factor for tumor cell themselves, thus making it a promising therapeutic target for OS. CRISPR/Cas9 is a versatile genome editing technology and holds tremendous promise for cancer treatment. However, a major bottleneck to achieve the therapeutic potential of the CRISPR/Cas9 is the lack of in vivo tumor-targeted delivery systems. Here, we screened an OS cell-specific aptamer (LC09) and developed a LC09-functionalized PEG-PEI-Cholesterol (PPC) lipopolymer encapsulating CRISPR/Cas9 plasmids encoding VEGFA gRNA and Cas9. Our results demonstrated that LC09 facilitated selective distribution of CRISPR/Cas9 in both orthotopic OS and lung metastasis, leading to effective VEGFA genome editing in tumor, decreased VEGFA expression and secretion, inhibited orthotopic OS malignancy and lung metastasis, as well as reduced angiogenesis and bone lesion with no detectable toxicity. The delivery system simultaneously restrained autocrine and paracrine VEGFA signaling in tumor cells and could facilitate translating CRISPR-Cas9 into clinical cancer treatment.
Collapse
Affiliation(s)
- Chao Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China.
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China.
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chao Wang
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Bing He
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Jie Li
- School of Chinese Medicine, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Zhihao Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Atik Badshah Shaikh
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Xiaohao Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Songlin Peng
- Department of Spine Surgery, Shenzhen People's Hospital, Ji Nan University Second College of Medicine, Shenzhen, China
| | - Lei Dang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Baosheng Guo
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Xiaojuan He
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - D W T Au
- Department of Biology and Chemistry, City University of Hong Kong, Hong Kong Special Administrative Region
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hailong Zhu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region.
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China; Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region; Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
25
|
Desu HR, Thoma LA, Wood GC. αvβ3 Integrin Receptor Targeting and Near-Infrared Imaging of Solid Tumors Using Surface-Modified Nanoliposomes. J Pharm Innov 2017. [DOI: 10.1007/s12247-017-9296-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
26
|
Maia ALC, Ferreira CDA, Barros ALBD, e Silva ATM, Ramaldes GA, Silva Cunha Júnior AD, Oliveira DCDP, Fernandes C, Ferreira Soares DC. Vincristine-loaded hydroxyapatite nanoparticles as a potential delivery system for bone cancer therapy. J Drug Target 2017; 26:592-603. [DOI: 10.1080/1061186x.2017.1401078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ana Luiza Chaves Maia
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Gilson Andrade Ramaldes
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Christian Fernandes
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
27
|
Kasala D, Lee SH, Hong JW, Choi JW, Nam K, Chung YH, Kim SW, Yun CO. Synergistic antitumor effect mediated by a paclitaxel-conjugated polymeric micelle-coated oncolytic adenovirus. Biomaterials 2017; 145:207-222. [DOI: 10.1016/j.biomaterials.2017.08.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 08/17/2017] [Accepted: 08/17/2017] [Indexed: 01/06/2023]
|
28
|
Overcoming obstacles in the tumor microenvironment: Recent advancements in nanoparticle delivery for cancer theranostics. Biomaterials 2017; 156:217-237. [PMID: 29207323 DOI: 10.1016/j.biomaterials.2017.10.024] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
Despite rapid advancements in the field of nanotechnology, there is mounting frustration in the scientific community regarding the translational impact of nanomedicine. Modest therapeutic performance of FDA-approved nanomedicines combined with multiple disappointing clinical trials (such as phase III HEAT trial) have raised questions about the future of nanomedicine. Encouraging breakthroughs, however, have been made in the last few years towards the development of new classes of nanoparticles that can respond to tumor microenvironmental conditions and successfully deliver therapeutic agents to cancer cells. Concurrently, a great deal of effort has also been devoted to alter various parameters of tumor pathophysiology to pre-treat tumors before nanoparticles are administered. Such 'priming' treatments improve access of the systemically administered agents to the tumor and promote drug penetration into the deeper layers of tumor tissue. This review will highlight recent advances in cancer nanomedicine exploiting both nanoparticle design and tumor microenvironment modification; and provide a critical perspective on the future development of nanomedicine delivery in oncology.
Collapse
|
29
|
Kalim M, Chen J, Wang S, Lin C, Ullah S, Liang K, Ding Q, Chen S, Zhan J. Intracellular trafficking of new anticancer therapeutics: antibody-drug conjugates. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2265-2276. [PMID: 28814834 PMCID: PMC5546728 DOI: 10.2147/dddt.s135571] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antibody-drug conjugate (ADC) is a milestone in targeted cancer therapy that comprises of monoclonal antibodies chemically linked to cytotoxic drugs. Internalization of ADC takes place via clathrin-mediated endocytosis, caveolae-mediated endocytosis, and pinocytosis. Conjugation strategies, endocytosis and intracellular trafficking optimization, linkers, and drugs chemistry present a great challenge for researchers to eradicate tumor cells successfully. This inventiveness of endocytosis and intracellular trafficking has given considerable momentum recently to develop specific antibodies and ADCs to treat cancer cells. It is significantly advantageous to emphasize the endocytosis and intracellular trafficking pathways efficiently and to design potent engineered conjugates and biological entities to boost efficient therapies enormously for cancer treatment. Current studies illustrate endocytosis and intracellular trafficking of ADC, protein, and linker strategies in unloading and also concisely evaluate practically applicable ADCs.
Collapse
Affiliation(s)
- Muhammad Kalim
- Department of Biochemistry and Genetics, School of Medicine
| | - Jie Chen
- Department of Biochemistry and Genetics, School of Medicine
| | - Shenghao Wang
- Department of Biochemistry and Genetics, School of Medicine
| | - Caiyao Lin
- Department of Biochemistry and Genetics, School of Medicine
| | - Saif Ullah
- Department of Biochemistry and Genetics, School of Medicine
| | - Keying Liang
- Department of Biochemistry and Genetics, School of Medicine
| | - Qian Ding
- Department of Biochemistry and Genetics, School of Medicine
| | - Shuqing Chen
- Department of Pharmaceutical Analysis, College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Jinbiao Zhan
- Department of Biochemistry and Genetics, School of Medicine
| |
Collapse
|
30
|
Lucas AT, Price LS, Schorzman A, Zamboni WC. Complex effects of tumor microenvironment on the tumor disposition of carrier-mediated agents. Nanomedicine (Lond) 2017; 12:2021-2042. [PMID: 28745129 DOI: 10.2217/nnm-2017-0101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Major advances in carrier-mediated agents, including nanoparticle, conjugates and antibody-drug conjugates, have created revolutionary drug delivery systems in cancer over the past two decades. While these agents provide several advantages, such as greater duration of exposure and solubility, compared with their small-molecule counterparts, there is substantial variability in delivery of these agents to tissues and especially tumors. This review provides an overview of tumor microenvironment factors that affect the pharmacokinetics and pharmacodynamics of carrier-mediated agents observed in preclinical models and patients.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lauren Sl Price
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Allison Schorzman
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C Zamboni
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
31
|
Azizi M, Ghourchian H, Yazdian F, Bagherifam S, Bekhradnia S, Nyström B. Anti-cancerous effect of albumin coated silver nanoparticles on MDA-MB 231 human breast cancer cell line. Sci Rep 2017; 7:5178. [PMID: 28701707 PMCID: PMC5508052 DOI: 10.1038/s41598-017-05461-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 05/10/2017] [Indexed: 01/23/2023] Open
Abstract
With the aim of making specific targeting of silver nanoparticles as a drug for tumor cells and developing new anticancer agents, a novel nano-composite was developed. Albumin coated silver nanoparticles (ASNPs) were synthesized, and their anti-cancerous effects were evaluated against MDA-MB 231, a human breast cancer cell line. The synthesized ASNPs were characterized by spectroscopic methods. The morphological changes of the cells were observed by inverted, florescent microscopy and also by DNA ladder pattern on gel electrophoresis; the results revealed that the cell death process occurred through the apoptosis mechanism. It was found that ASNPs with a size of 90 nm and negatively charged with a zeta-potential of about −20 mV could be specifically taken up by tumor cells. The LD50 of ASNPs against MDA-MB 231 (5 μM), was found to be 30 times higher than that for white normal blood cells (152 μM). The characteristics of the synthesized ASNPs included; intact structure of coated albumin, higher cytotoxicity against cancer cells than over normal cells, and cell death based on apoptosis and reduction of gland tumor sizes in mice. This work indicates that ASNPs could be a good candidate for chemotherapeutic drug.
Collapse
Affiliation(s)
- Marzieh Azizi
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,Department of Chemistry, University of Oslo, Oslo, Norway
| | | | - Fatemeh Yazdian
- Faculty of New Science and Technology, University of Tehran, Tehran, Iran
| | - Shahla Bagherifam
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.,Department of Chemistry, University of Oslo, Oslo, Norway
| | | | - Bo Nyström
- Department of Chemistry, University of Oslo, Oslo, Norway
| |
Collapse
|
32
|
Dai W, Wang X, Song G, Liu T, He B, Zhang H, Wang X, Zhang Q. Combination antitumor therapy with targeted dual-nanomedicines. Adv Drug Deliv Rev 2017; 115:23-45. [PMID: 28285944 DOI: 10.1016/j.addr.2017.03.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
Combination therapy is one of the important treatment strategies for cancer at present. However, the outcome of current combination therapy based on the co-administration of conventional dosage forms is suboptimal, due to the short half-lives of chemodrugs, their deficient tumor selectivity and so forth. Nanotechnology-based targeted delivery systems show great promise in addressing the associated problems and providing superior therapeutic benefits. In this review, we focus on the combination of therapeutic strategies between different nanomedicines or drug-loaded nanocarriers, rather than the co-delivery of different drugs via a single nanocarrier. We introduce the general concept of various targeting strategies of nanomedicines, present the principles of combination antitumor therapy with dual-nanomedicines, analyze their advantages and limitations compared with co-delivery strategies, and overview the recent advances of combination therapy based on targeted nanomedicines. Finally, we reviewed the challenges and future perspectives regarding the selection of therapeutic agents, targeting efficiency and the gap between the preclinical and clinical outcome.
Collapse
Affiliation(s)
- Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyou Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Tongzhou Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China.
| |
Collapse
|
33
|
Engineering hepatitis B virus core particles for targeting HER2 receptors in vitro and in vivo. Biomaterials 2016; 120:126-138. [PMID: 28056402 PMCID: PMC5300899 DOI: 10.1016/j.biomaterials.2016.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/11/2022]
Abstract
Hepatitis B Virus core (HBc) particles have been studied for their potential as drug delivery vehicles for cancer therapy. HBc particles are hollow nano-particles of 30–34 nm diameter and 7 nm thick envelopes, consisting of 180–240 units of 21 kDa core monomers. They have the capacity to assemble/dis-assemble in a controlled manner allowing encapsulation of various drugs and other biomolecules. Moreover, other functional motifs, i.e. receptors, receptor binding sequences, peptides and proteins can be expressed. This study focuses on the development of genetically modified HBc particles to specifically recognise and target human epidermal growth factor receptor-2 (HER2)-expressing cancer cells, in vitro and in vivo, for future cancer therapy. The non-specific binding capacity of wild type HBc particles was reduced by genetic deletion of the sequence encoding arginine-rich domains. A specific HER2-targeting was achieved by expressing the ZHER2 affibodies on the HBc particles surface. In vitro studies showed specific uptake of ZHER2-ΔHBc particles in HER2 expressing cancer cells. In vivo studies confirmed positive uptake of ZHER2-ΔHBc particles in HER2-expressing tumours, compared to non-targeted ΔHBc particles in intraperitoneal tumour-bearing mice models. The present results highlight the potential of these nanocarriers in targeting HER2-positive metastatic abdominal cancer following intra-peritoneal administration.
Collapse
|
34
|
Zeng Y, Zhou Z, Fan M, Gong T, Zhang Z, Sun X. PEGylated Cationic Vectors Containing a Protease-Sensitive Peptide as a miRNA Delivery System for Treating Breast Cancer. Mol Pharm 2016; 14:81-92. [PMID: 28043137 DOI: 10.1021/acs.molpharmaceut.6b00726] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Several targeted drug delivery systems have recently been developed to increase the bioavailability of a drug at its site of action, allowing simultaneous reduction of the total necessary drug dose as well as side effects. Here, we designed a cationic gene vector containing matrix metalloproteinase-2 (MMP2)-cleavable substrate peptides that specifically target tumor sites where MMP2 levels are high. The targeted delivery system is fabricated by linking enzyme-cleavable polyethylene glycol (PEG) derivatives to cationic β-cyclodextrin-polyethylenimine conjugates, which reduce the toxicity of polyethylenimine and condense the therapeutic cargo. In the present study, tumor suppressor microRNA miR-34a, which suppresses onset and progression of many types of cancers, was investigated for its therapeutic potential for treating breast cancer. The PEG coating markedly reduces nonspecific interaction between cationic particles and serum proteins, permitting accumulation at the target site; subsequent peptide cleavage by MMP2 facilitates miR-34a delivery into tumor cells. The nanopreparation shows excellent stability, and its internalization, tumor targeting, and antitumor efficacy in vitro and in vivo are better than those of a nanopreparation containing MMP2-uncleavable peptide.
Collapse
Affiliation(s)
- Ye Zeng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Zixuan Zhou
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Minmin Fan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
35
|
To exploit the tumor microenvironment: Since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release 2016; 244:108-121. [DOI: 10.1016/j.jconrel.2016.11.015] [Citation(s) in RCA: 752] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/26/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022]
|
36
|
Zhang RX, Wong HL, Xue HY, Eoh JY, Wu XY. Nanomedicine of synergistic drug combinations for cancer therapy - Strategies and perspectives. J Control Release 2016; 240:489-503. [PMID: 27287891 PMCID: PMC5064882 DOI: 10.1016/j.jconrel.2016.06.012] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/27/2016] [Accepted: 06/06/2016] [Indexed: 12/26/2022]
Abstract
Nanomedicine of synergistic drug combinations has shown increasing significance in cancer therapy due to its promise in providing superior therapeutic benefits to the current drug combination therapy used in clinical practice. In this article, we will examine the rationale, principles, and advantages of applying nanocarriers to improve anticancer drug combination therapy, review the use of nanocarriers for delivery of a variety of combinations of different classes of anticancer agents including small molecule drugs and biologics, and discuss the challenges and future perspectives of the nanocarrier-based combination therapy. The goal of this review is to provide better understanding of this increasingly important new paradigm of cancer treatment and key considerations for rational design of nanomedicine of synergistic drug combinations for cancer therapy.
Collapse
Affiliation(s)
- Rui Xue Zhang
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, Canada M5S 2S2
| | - Ho Lun Wong
- Temple University School of Pharmacy, 3304 North Broad Street, Philadelphia, PA 19140, USA
| | - Hui Yi Xue
- Temple University School of Pharmacy, 3304 North Broad Street, Philadelphia, PA 19140, USA
| | - June Young Eoh
- Temple University School of Pharmacy, 3304 North Broad Street, Philadelphia, PA 19140, USA
| | - Xiao Yu Wu
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, Canada M5S 2S2
| |
Collapse
|
37
|
Cho H, Gao J, Kwon GS. PEG-b-PLA micelles and PLGA-b-PEG-b-PLGA sol-gels for drug delivery. J Control Release 2016; 240:191-201. [PMID: 26699425 PMCID: PMC4909590 DOI: 10.1016/j.jconrel.2015.12.015] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
Poly(ethylene glycol)-block-poly(D,L-lactic acid) (PEG-b-PLA) micelles and poly(D,L-lactic-co-glycolic acid)-block-polyethylene glycol)-block-poly(D,L-lactic-co-glycolic acid) (PLGA-b-PEG-b-PLGA) sol-gels have been extensively researched for systemic and localized drug delivery applications, respectively, and they have both progressed into humans for paclitaxel, an important yet poorly water-soluble chemotherapeutic agent. In this review article, preclinical and clinical research on PEG-b-PLA micelles and PLGA-b-PEG-b-PLGA sol-gels that has focused on paclitaxel will be updated, and recent research on other poorly water-soluble anticancer agents and delivery of drug combinations (i.e. multi-drug delivery) that seeks synergistic anticancer efficacy will be summarized. PEG-b-PLA micelles are a first-generation platform for the systemic multi-delivery of poorly water soluble anticancer agents. PLGA-b-PEG-b-PLGA sol-gels are a first-generation platform for the localized multi-drug delivery of water-soluble and/or poorly water-soluble anticancer agents. In summary, PEG-b-PLA micelles and PLGA-b-PEG-b-PLGA sol-gels may safely enable pre-clinical evaluation and clinical translation of poorly water-soluble anticancer agents, especially for promising, rapidly emerging anticancer combinations.
Collapse
Affiliation(s)
- Hyunah Cho
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, 4588 Parkview Place, St. Louis, MO 63110, United States
| | - Jieming Gao
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, Republic of Korea.
| |
Collapse
|
38
|
Kaur G, Willsmore T, Gulati K, Zinonos I, Wang Y, Kurian M, Hay S, Losic D, Evdokiou A. Titanium wire implants with nanotube arrays: A study model for localized cancer treatment. Biomaterials 2016; 101:176-88. [PMID: 27289379 DOI: 10.1016/j.biomaterials.2016.05.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/21/2016] [Accepted: 05/27/2016] [Indexed: 12/11/2022]
Abstract
Adverse complications associated with systemic administration of anti-cancer drugs are a major problem in cancer therapy in current clinical practice. To increase effectiveness and reduce side effects, localized drug delivery to tumour sites requiring therapy is essential. Direct delivery of potent anti-cancer drugs locally to the cancer site based on nanotechnology has been recognised as a promising alternative approach. Previously, we reported the design and fabrication of nano-engineered 3D titanium wire based implants with titania (TiO2) nanotube arrays (Ti-TNTs) for applications such as bone integration by using in-vitro culture systems. The aim of present study is to demonstrate the feasibility of using such Ti-TNTs loaded with anti-cancer agent for localized cancer therapy using pre-clinical cancer models and to test local drug delivery efficiency and anti-tumour efficacy within the tumour environment. TNF-related apoptosis-inducing ligand (TRAIL) which has proven anti-cancer properties was selected as the model drug for therapeutic delivery by Ti-TNTs. Our in-vitro 2D and 3D cell culture studies demonstrated a significant decrease in breast cancer cell viability upon incubation with TRAIL loaded Ti-TNT implants (TRAIL-TNTs). Subcutaneous tumour xenografts were established to test TRAIL-TNTs implant performance in the tumour environment by monitoring the changes in tumour burden over a selected time course. TRAIL-TNTs showed a significant regression in tumour burden within the first three days of implant insertion at the tumour site. Based on current experimental findings these Ti-TNTs wire implants have shown promising capacity to load and deliver anti-cancer agents maintaining their efficacy for cancer treatment.
Collapse
Affiliation(s)
- Gagandeep Kaur
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, 5005, Australia; School of Medicine, Discipline of Surgery, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tamsyn Willsmore
- School of Medicine, Discipline of Surgery, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Karan Gulati
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Irene Zinonos
- School of Medicine, Discipline of Surgery, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Ye Wang
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Mima Kurian
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Shelley Hay
- School of Medicine, Discipline of Surgery, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Dusan Losic
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Andreas Evdokiou
- School of Medicine, Discipline of Surgery, The University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
39
|
Au JLS, Yeung BZ, Wientjes MG, Lu Z, Wientjes MG. Delivery of cancer therapeutics to extracellular and intracellular targets: Determinants, barriers, challenges and opportunities. Adv Drug Deliv Rev 2016; 97:280-301. [PMID: 26686425 PMCID: PMC4829347 DOI: 10.1016/j.addr.2015.12.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/24/2015] [Accepted: 12/02/2015] [Indexed: 02/08/2023]
Abstract
Advances in molecular medicine have led to identification of worthy cellular and molecular targets located in extracellular and intracellular compartments. Effectiveness of cancer therapeutics is limited in part by inadequate delivery and transport in tumor interstitium. Parts I and II of this report give an overview on the kinetic processes in delivering therapeutics to their intended targets, the transport barriers in tumor microenvironment and extracellular matrix (TME/ECM), and the experimental approaches to overcome such barriers. Part III discusses new concepts and findings concerning nanoparticle-biocorona complex, including the effects of TME/ECM. Part IV outlines the challenges in animal-to-human translation of cancer nanotherapeutics. Part V provides an overview of the background, current status, and the roles of TME/ECM in immune checkpoint inhibition therapy, the newest cancer treatment modality. Part VI outlines the development and use of multiscale computational modeling to capture the unavoidable tumor heterogeneities, the multiple nonlinear kinetic processes including interstitial and transvascular transport and interactions between cancer therapeutics and TME/ECM, in order to predict the in vivo tumor spatiokinetics of a therapeutic based on experimental in vitro biointerfacial interaction data. Part VII provides perspectives on translational research using quantitative systems pharmacology approaches.
Collapse
Affiliation(s)
- Jessie L-S Au
- Optimum Therapeutics LLC, 1815 Aston Avenue, Carlsbad, CA 92008, USA; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA; Medical University of South Carolina, Charleston, SC 29425, USA; Taipei Medical University, Taipei, Taiwan, ROC.
| | - Bertrand Z Yeung
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | | | - Ze Lu
- Optimum Therapeutics LLC, 1815 Aston Avenue, Carlsbad, CA 92008, USA
| | | |
Collapse
|
40
|
Singh MS, Peer D. RNA nanomedicines: the next generation drugs? Curr Opin Biotechnol 2016; 39:28-34. [PMID: 26773301 DOI: 10.1016/j.copbio.2015.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/19/2015] [Indexed: 02/08/2023]
Abstract
RNA therapeutics could represent the next generation personalized medicine. The variety of RNA molecules that can inhibit the expression of any mRNA using, for example, RNA interference (RNAi) strategies, or increase the expression of a given protein using modified mRNA together with new gene editing strategies open new avenues for manipulating the fate of diseased cells while leaving healthy cells untouched. In addition, these therapeutic RNA molecules can maximize the treatment of diseases and minimize its adverse effects. Yet, the promise of RNA therapeutics is hindered by the lack of efficient delivery strategies to selectively target these molecules into specific cells. Herein, we will focus on the challenges and opportunities of the delivery of therapeutic RNAi molecules into cancer cells with special emphasis on solid tumors. Solid tumors represent more than 80 percent of cancers and some are very challenging to treat, not merely due to physiological barriers but also since the tumor microenvironment (TME) is a complex milieu of accessory cells besides the cancerous cells. In this review, we will highlight various limiting factors to successful delivery, current clinical achievements and future outlook focusing on RNAi therapeutics to the TME.
Collapse
Affiliation(s)
- Manu Smriti Singh
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
41
|
Fan G, Fan M, Wang Q, Jiang J, Wan Y, Gong T, Zhang Z, Sun X. Bio-inspired polymer envelopes around adenoviral vectors to reduce immunogenicity and improve in vivo kinetics. Acta Biomater 2016; 30:94-105. [PMID: 26546972 DOI: 10.1016/j.actbio.2015.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 10/27/2015] [Accepted: 11/03/2015] [Indexed: 10/22/2022]
Abstract
Adenoviral vectors have attracted substantial interest for systemic tumor gene therapy, but further work is needed to reduce their immunogenicity and alter their biodistribution before they can be used in the clinic. Here we describe a bio-inspired, cleavable PEGylated β-cyclodextrin-polyethyleneimine conjugate (CDPCP) that spontaneously coats adenovirus in solution. This cleavable PEG coating reduces the innate and adaptive immunogenicity of adenovirus particles, as well as improves their biodistribution away from the liver and into the tumor. Insertion of a matrix metalloproteinase substrate sequence into the conjugate allows PEG cleavage at the tumor site, simultaneously reducing liver biodistribution and increasing transgene expression in tumors, thereby avoiding the "PEG dilemma". Cationic β-cyclodextrin-PEI not only provides electrostatic attraction to promote envelope attachment to the viral capsid, but it also improves vector internalization and transduction after PEG cleavage. These results suggest that CDPCP may help expand the use of adenoviral vectors in cancer gene therapy. STATEMENT OF SIGNIFICANCE The synthesized β-cyclodextrin-PEI-MMP-cleavable-PEG polymer (CDPCP), held great potential for gene therapy when applied for adenovirus coating. The β-cyclodextrin-PEI provided a powerful electrostatic attraction to attach the whole polymer onto the viral capsid, while the MMPs-cleavable PEG reduced innate and adaptive immunogenicity and improved the biodistribution of adenovirus vectors due to the tumor-specific enzyme triggered PEG cleavage. More importantly, an ingenious cooperation between the two components could solve the PEG dilemma. The CDPCP/Ad complexes exhibited a comprehensive and valued profile to be a candidate vector for future tumor gene therapy, we believe the current investigation on this kind of biomaterial may be of particular interest to the readership of Acta biomaterialia.
Collapse
|
42
|
England CG, Gobin AM, Frieboes HB. Evaluation of uptake and distribution of gold nanoparticles in solid tumors. EUROPEAN PHYSICAL JOURNAL PLUS 2015; 130:231. [PMID: 27014559 PMCID: PMC4800753 DOI: 10.1140/epjp/i2015-15231-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Although nanotherapeutics offer a targeted and potentially less toxic alternative to systemic chemotherapy in cancer treatment, nanotherapeutic transport is typically hindered by abnormal characteristics of tumor tissue. Once nanoparticles targeted to tumor cells arrive in the circulation of tumor vasculature, they must extravasate from irregular vessels and diffuse through the tissue to ideally reach all malignant cells in cytotoxic concentrations. The enhanced permeability and retention effect can be leveraged to promote extravasation of appropriately sized particles from tumor vasculature; however, therapeutic success remains elusive partly due to inadequate intra-tumoral transport promoting heterogeneous nanoparticle uptake and distribution. Irregular tumor vasculature not only hinders particle transport but also sustains hypoxic tissue kregions with quiescent cells, which may be unaffected by cycle-dependent chemotherapeutics released from nanoparticles and thus regrow tumor tissue following nanotherapy. Furthermore, a large proportion of systemically injected nanoparticles may become sequestered by the reticuloendothelial system, resulting in overall diminished efficacy. We review recent work evaluating the uptake and distribution of gold nanoparticles in pre-clinical tumor models, with the goal to help improve nanotherapy outcomes. We also examine the potential role of novel layered gold nanoparticles designed to address some of these critical issues, assessing their uptake and transport in cancerous tissue.
Collapse
Affiliation(s)
- Christopher G England
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
| | - André M Gobin
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Hermann B Frieboes
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA; Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
43
|
Zeng L, Chen Y, Huang H, Wang J, Zhao D, Ji L, Chao H. Cyclometalated Ruthenium(II) Anthraquinone Complexes Exhibit Strong Anticancer Activity in Hypoxic Tumor Cells. Chemistry 2015; 21:15308-19. [PMID: 26338207 DOI: 10.1002/chem.201502154] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 12/31/2022]
Abstract
Hypoxia is the critical feature of the tumor microenvironment that is known to lead to resistance to many chemotherapeutic drugs. Six novel ruthenium(II) anthraquinone complexes were designed and synthesized; they exhibit similar or superior cytotoxicity compared to cisplatin in hypoxic HeLa, A549, and multidrug-resistant (A549R) tumor cell lines. Their anticancer activities are related to their lipophilicity and cellular uptake; therefore, these physicochemical properties of the complexes can be changed by modifying the ligands to obtain better anticancer candidates. Complex 1, the most potent member of the series, is highly active against hypoxic HeLa cancer cells (IC50 =0.53 μM). This complex likely has 46-fold better activity than cisplatin (IC50 =24.62 μM) in HeLa cells. This complex tends to accumulate in the mitochondria and the nucleus of hypoxic HeLa cells. Further mechanistic studies show that complex 1 induced cell apoptosis during hypoxia through multiple pathways, including those of DNA damage, mitochondrial dysfunction, and the inhibition of DNA replication and HIF-1α expression, making it an outstanding candidate for further in vivo studies.
Collapse
Affiliation(s)
- Leli Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | - Huaiyi Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | - Jinquan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | - Donglei Zhao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China).
| |
Collapse
|
44
|
Al-Abd AM, Aljehani ZK, Gazzaz RW, Fakhri SH, Jabbad AH, Alahdal AM, Torchilin VP. Pharmacokinetic strategies to improve drug penetration and entrapment within solid tumors. J Control Release 2015; 219:269-277. [PMID: 26342660 DOI: 10.1016/j.jconrel.2015.08.055] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/09/2015] [Accepted: 08/28/2015] [Indexed: 02/08/2023]
Abstract
Despite the discovery of a large number of anticancer agents, cancer still remains among the leading causes of death since the middle of the twentieth century. Solid tumors possess a high degree of genetic instability and emergence of treatment resistance. Tumor resistance has emerged for almost all approved anticancer drugs and will most probably emerge for newly discovered anticancer agents as well. The use of pharmacokinetic approaches to increase anticancer drug concentrations within the solid tumor compartment and prolong its entrapment might diminish the possibility of resistance emergence at the molecular pharmacodynamic level and might even reverse tumor resistance. Several novel treatment modalities such as metronomic therapy, angiogenesis inhibitors, vascular disrupting agents and tumor priming have been introduced to improve solid tumor treatment outcomes. In the current review we will discuss the pharmacokinetic aspect of these treatment modalities in addition to other older treatment modalities, such as extracellular matrix dissolving agents, extracellular matrix synthesis inhibitors, chemoembolization and cellular efflux pump inhibition. Many of these strategies showed variable degrees of success/failure; however, reallocating these modalities based on their influence on the intratumoral pharmacokinetics might improve their understanding and treatment outcomes.
Collapse
Affiliation(s)
- Ahmed M Al-Abd
- Department of Pharmacology, Medical Division, National Research Centre, Dokki, Giza, Egypt; Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA; Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zekra K Aljehani
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rana W Gazzaz
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sarah H Fakhri
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aisha H Jabbad
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
45
|
Wang J, Lu Z, Wang J, Cui M, Yeung BZ, Cole DJ, Wientjes MG, Au JLS. Paclitaxel tumor priming promotes delivery and transfection of intravenous lipid-siRNA in pancreatic tumors. J Control Release 2015; 216:103-10. [PMID: 26272765 DOI: 10.1016/j.jconrel.2015.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/14/2015] [Accepted: 08/05/2015] [Indexed: 01/05/2023]
Abstract
The major barrier for using small interfering RNA (siRNA) as cancer therapeutics is the inadequate delivery and transfection in solid tumors. We have previously shown that paclitaxel tumor priming, by inducing apoptosis, expands the tumor interstitial space, improves the penetration and dispersion of nanoparticles and siRNA-lipoplexes in 3-dimensional tumor histocultures, and promotes the delivery and transfection efficiency of siRNA-lipoplexes under the locoregional setting in vivo (i.e., intraperitoneal treatment of intraperitoneal tumors). The current study evaluated whether tumor priming is functional for systemically delivered siRNA via intravenous injection, which would subject siRNA to several additional delivery barriers and elimination processes. We used the same pegylated cationic (PCat)-siRNA lipoplexes as in the intraperitoneal study to treat mice bearing subcutaneous human pancreatic Hs766T xenograft tumors. The target gene was survivin, an inducible chemoresistance gene. The results show single agent paclitaxel delayed tumor growth but also significantly induced the survivin protein level in residual tumors, whereas addition of PCat-siSurvivin completely reversed the paclitaxel-induced survivin and enhanced the paclitaxel activity (p<0.05). In comparison, PCat-siSurvivin alone did not yield survivin knockdown or antitumor activity, indicating the in vivo effectiveness of intravenous siRNA-mediated gene silencing requires paclitaxel cotreatment. Additional in vitro studies showed that paclitaxel promoted the cytoplasmic release of siGLO, a 22 nucleotide double-stranded RNA that has no mRNA targets, from its PCat lipoplex and/or endosomes/lysosomes. Taken together, our earlier and current data show paclitaxel tumor priming, by promoting the interstitial transport and cytoplasmic release, is critical to promote the delivery and transfection of siRNA in vivo. In addition, because paclitaxel has broad spectrum activity and is used to treat multiple types of solid tumors including the hard-to-treat pancreatic cancer, the synergistic paclitaxel+siSurvivin combination represents a potentially useful chemo-gene therapy.
Collapse
Affiliation(s)
- Jie Wang
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Ze Lu
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Junfeng Wang
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Minjian Cui
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA
| | - Bertrand Z Yeung
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | - David J Cole
- Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Jessie L-S Au
- Optimum Therapeutics LLC, 1815 Aston Avenue, STE 107, Carlsbad, CA 92008, USA; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA; Medical University of South Carolina, Charleston, SC 29425, USA; Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
46
|
Soheilian R, Choi YS, David AE, Abdi H, Maloney CE, Erb RM. Toward Accumulation of Magnetic Nanoparticles into Tissues of Small Porosity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:8267-8274. [PMID: 26145706 DOI: 10.1021/acs.langmuir.5b01458] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Magnetic concentration of drug-laden magnetic nanoparticles has been proven to increase the delivery efficiency of treatment by 2-fold. In these techniques, particles are concentrated by the presence of a magnetic source that delivers a very high magnetic field and a strong magnetic field gradient. We have found that such magnetic conditions cause even 150 nm particles to aggregate significantly into assemblies that exceed several micrometers in length within minutes. Such assembly sizes exceed the effective intercellular pore size of tumor tissues preventing these drug-laden magnetic nanoparticles from reaching their target sites. We demonstrate that by using dynamic magnetic fields instead, we can break up these magnetic nanoparticles while simultaneously concentrating them at target sites. The dynamic fields we investigate involve precessing the field direction while maintaining a field gradient. Manipulating the field direction drives the particles into attractive and repulsive configurations that can be tuned to assemble or disassemble these particle clusters. Here, we develop a simple analytic model to describe the kinetic thresholds of disassembly and we compare both experimental and numerical results of magnetic particle suspensions subjected to dynamic fields. Finally we apply these methods to demonstrate penetration in a porous scaffold with a similar pore size to that expected of a tumor tissue.
Collapse
Affiliation(s)
- Rasam Soheilian
- †Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Young Suk Choi
- ‡Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Allan E David
- ‡Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Hamed Abdi
- †Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Craig E Maloney
- †Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Randall M Erb
- †Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
47
|
Wauthoz N, Bastiat G, Moysan E, Cieślak A, Kondo K, Zandecki M, Moal V, Rousselet MC, Hureaux J, Benoit JP. Safe lipid nanocapsule-based gel technology to target lymph nodes and combat mediastinal metastases from an orthotopic non-small-cell lung cancer model in SCID-CB17 mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1237-45. [DOI: 10.1016/j.nano.2015.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/12/2015] [Accepted: 02/14/2015] [Indexed: 12/19/2022]
|
48
|
Au JLS, Lu Z, Wientjes MG. Versatility of Particulate Carriers: Development of Pharmacodynamically Optimized Drug-Loaded Microparticles for Treatment of Peritoneal Cancer. AAPS JOURNAL 2015; 17:1065-79. [PMID: 26089090 DOI: 10.1208/s12248-015-9785-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023]
Abstract
Intraperitoneal (IP) chemotherapy confers significant survival benefits in cancer patients. However, several problems, including local toxicity and ineffectiveness against bulky tumors, have prohibited it from becoming a standard-of-care. We have developed drug-loaded, tumor-penetrating microparticles (TPM) to address these problems. TPM comprises two components and uses the versatile PLGA or poly(lacticco-glycolic acid) copolymer to provide tumor-selective adherence and pharmacodynamically optimized fractionated dosing to achieve the desired tumor priming (which promotes particle penetration into tumors) plus immediate and sustained antitumor activity. Preclinical studies show that TPM is less toxic and more effective against several IP metastatic tumors with different characteristics (fast vs. slow growing, porous vs. densely packed structures, wide-spread vs. solitary tumors, early vs. late stage, with or without peritoneal carcinomatosis or ascites), compared to the intravenous paclitaxel/Cremophor micellar solution that has been used off-label in previous IP studies. TPM further requires less frequent dosing. These encouraging preclinical results have motivated the follow-up clinical development of TPM. We are working with National Institutes of Health on the IND-enabling studies.
Collapse
Affiliation(s)
- Jessie L-S Au
- Optimum Therapeutics LLC, 1815 Aston Avenue, Suite 107, Carlsbad, California, 92008, USA,
| | | | | |
Collapse
|
49
|
Detection of Phosphatidylcholine-Coated Gold Nanoparticles in Orthotopic Pancreatic Adenocarcinoma using Hyperspectral Imaging. PLoS One 2015; 10:e0129172. [PMID: 26046360 PMCID: PMC4457854 DOI: 10.1371/journal.pone.0129172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/05/2015] [Indexed: 02/08/2023] Open
Abstract
Nanoparticle uptake and distribution to solid tumors are limited by reticuloendothelial system systemic filtering and transport limitations induced by irregular intra-tumoral vascularization. Although vascular enhanced permeability and retention can aid targeting, high interstitial fluid pressure and dense extracellular matrix may hinder local penetration. Extravascular diffusivity depends upon nanoparticle size, surface modifications, and tissue vascularization. Gold nanoparticles functionalized with biologically-compatible layers may achieve improved uptake and distribution while enabling cytotoxicity through synergistic combination of chemotherapy and thermal ablation. Evaluation of nanoparticle uptake in vivo remains difficult, as detection methods are limited. We employ hyperspectral imaging of histology sections to analyze uptake and distribution of phosphatidylcholine-coated citrate gold nanoparticles (CGN) and silica-gold nanoshells (SGN) after tail-vein injection in mice bearing orthotopic pancreatic adenocarcinoma. For CGN, the liver and tumor showed 26.5±8.2 and 23.3±4.1 particles/100μm2 within 10μm from the nearest source and few nanoparticles beyond 50μm, respectively. The spleen had 35.5±9.3 particles/100μm2 within 10μm with penetration also limited to 50μm. For SGN, the liver showed 31.1±4.1 particles/100μm2 within 10μm of the nearest source with penetration hindered beyond 30μm. The spleen and tumor showed uptake of 22.1±6.2 and 15.8±6.1 particles/100μm2 within 10μm, respectively, with penetration similarly hindered. CGH average concentration (nanoparticles/μm2) was 1.09±0.14 in the liver, 0.74±0.12 in the spleen, and 0.43±0.07 in the tumor. SGN average concentration (nanoparticles/μm2) was 0.43±0.07 in the liver, 0.30±0.06 in the spleen, and 0.20±0.04 in the tumor. Hyperspectral imaging of histology sections enables analysis of phosphatidylcholine-coated gold-based nanoparticles in pancreatic tumors with the goal to improve nanotherapeutic efficacy.
Collapse
|
50
|
Arya N, Katti DS. Poly(d,l-lactide-co-glycolide)-chitosan composite particles for the treatment of lung cancer. Int J Nanomedicine 2015; 10:2997-3011. [PMID: 25945047 PMCID: PMC4406260 DOI: 10.2147/ijn.s78120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Tumor heterogeneity makes combination chemotherapy one of the preferred modes of treatment regimens. In this work, sequential exposure of two anticancer agents, paclitaxel (Tx) followed by topotecan (TPT), was shown to have a synergistic effect on non-small cell lung cancer (NSCLC) cell line, NCI-H460. In order to improve patient compliance, the aforementioned concept was translated into a drug delivery system comprising of poly(d,l-lactide-co-glycolide) (PLGA)-chitosan composite particles. TPT-containing chitosan micro-/nanoparticles were prepared by the facile technique of electrospraying and encapsulated within PLGA microparticles using emulsion-solvent evaporation technique for delayed release of TPT. The formulation containing Tx- and TPT-loaded composite particles demonstrated synergism when exposed to NCI-H460 cellular aggregates (tumoroids) generated in vitro. Overall, the results of this study demonstrated the potential of the formulation containing Tx and PLGA-chitosan (TPT-loaded) composite particles for the treatment of lung cancer.
Collapse
Affiliation(s)
- Neha Arya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology - Kanpur, Kanpur, Uttar Pradesh, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology - Kanpur, Kanpur, Uttar Pradesh, India
| |
Collapse
|