51
|
Ofridam F, Tarhini M, Lebaz N, Gagnière É, Mangin D, Elaissari A. pH
‐sensitive polymers: Classification and some fine potential applications. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5230] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Fabrice Ofridam
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Mohamad Tarhini
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, ISA UMR 5280 Villeurbanne France
| | - Noureddine Lebaz
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Émilie Gagnière
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Denis Mangin
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Abdelhamid Elaissari
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, ISA UMR 5280 Villeurbanne France
| |
Collapse
|
52
|
Zhao T, Zhou H, Wu W, Song X, Gong T. A multistage oral delivery system of PTX for improving oral bioavailability and enhancing anticancer efficacy. Drug Dev Ind Pharm 2021; 47:259-267. [PMID: 33501858 DOI: 10.1080/03639045.2021.1879831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Bromotetrandrine (W198) was reported as a P-glycoprotein (P-gp) inhibitor. We aimed to prepare oral W198 micelles following by paclitaxel (PTX) micelles (W198/PTX micelles) to improve the clinical application of PTX. SIGNIFICANCE The poor water solubility, intestinal permeability, and multidrug resistance (MDR) of PTX can be improved in the multistage oral delivery system. METHODS The novel W198/PTX oral micelles were developed by water-bath ultrasound method and were evaluated in vivo and in vitro in 4T1 orthotopic tumor-bearing mice model. RESULTS PTX micelles and W198 micelles were prepared to be round and uniform. W198 micelles pre-administrated group showed higher cellular uptake efficiency of PTX on Caco-2 cells and more prominent cytotoxicity compared with W198-untreated group on 4T1 cells. The oral bioavailability of W198/PTX micelles group was nearly 5.7-folds higher than the PTX micelles only group. In addition, W198/PTX micelles showed enhanced anticancer efficacy. CONCLUSIONS We established a multistage oral delivery system to improve oral bioavailability and anticancer efficacy of PTX.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Hongli Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Wanyan Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Xu Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China.,Institute of Regulatory Science for Medical Devices, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P. R. China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
53
|
Mosca L, Ilari A, Fazi F, Assaraf YG, Colotti G. Taxanes in cancer treatment: Activity, chemoresistance and its overcoming. Drug Resist Updat 2021; 54:100742. [PMID: 33429249 DOI: 10.1016/j.drup.2020.100742] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Since 1984, when paclitaxel was approved by the FDA for the treatment of advanced ovarian carcinoma, taxanes have been widely used as microtubule-targeting antitumor agents. However, their historic classification as antimitotics does not describe all their functions. Indeed, taxanes act in a complex manner, altering multiple cellular oncogenic processes including mitosis, angiogenesis, apoptosis, inflammatory response, and ROS production. On the one hand, identification of the diverse effects of taxanes on oncogenic signaling pathways provides opportunities to apply these cytotoxic drugs in a more rational manner. On the other hand, this may facilitate the development of novel treatment modalities to surmount anticancer drug resistance. In the latter respect, chemoresistance remains a major impediment which limits the efficacy of antitumor chemotherapy. Taxanes have shown impact on key molecular mechanisms including disruption of mitotic spindle, mitosis slippage and inhibition of angiogenesis. Furthermore, there is an emerging contribution of cellular processes including autophagy, oxidative stress, epigenetic alterations and microRNAs deregulation to the acquisition of taxane resistance. Hence, these two lines of findings are currently promoting a more rational and efficacious taxane application as well as development of novel molecular strategies to enhance the efficacy of taxane-based cancer treatment while overcoming drug resistance. This review provides a general and comprehensive picture on the use of taxanes in cancer treatment. In particular, we describe the history of application of taxanes in anticancer therapeutics, the synthesis of the different drugs belonging to this class of cytotoxic compounds, their features and the differences between them. We further dissect the molecular mechanisms of action of taxanes and the molecular basis underlying the onset of taxane resistance. We further delineate the possible modalities to overcome chemoresistance to taxanes, such as increasing drug solubility, delivery and pharmacokinetics, overcoming microtubule alterations or mitotic slippage, inhibiting drug efflux pumps or drug metabolism, targeting redox metabolism, immune response, and other cellular functions.
Collapse
Affiliation(s)
- Luciana Mosca
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council (IBPM-CNR), c/o Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Francesco Fazi
- Dept. Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University, Via A. Scarpa 14-16, 00161 Rome, Italy
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council (IBPM-CNR), c/o Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
54
|
Abreu TR, Biscaia M, Gonçalves N, Fonseca NA, Moreira JN. In Vitro and In Vivo Tumor Models for the Evaluation of Anticancer Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:271-299. [PMID: 33543464 DOI: 10.1007/978-3-030-58174-9_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multiple studies about tumor biology have revealed the determinant role of the tumor microenvironment in cancer progression, resulting from the dynamic interactions between tumor cells and surrounding stromal cells within the extracellular matrix. This malignant microenvironment highly impacts the efficacy of anticancer nanoparticles by displaying drug resistance mechanisms, as well as intrinsic physical and biochemical barriers, which hamper their intratumoral accumulation and biological activity.Currently, two-dimensional cell cultures are used as the initial screening method in vitro for testing cytotoxic nanocarriers. However, this fails to mimic the tumor heterogeneity, as well as the three-dimensional tumor architecture and pathophysiological barriers, leading to an inaccurate pharmacological evaluation.Biomimetic 3D in vitro tumor models, on the other hand, are emerging as promising tools for more accurately assessing nanoparticle activity, owing to their ability to recapitulate certain features of the tumor microenvironment and thus provide mechanistic insights into nanocarrier intratumoral penetration and diffusion rates.Notwithstanding, in vivo validation of nanomedicines remains irreplaceable at the preclinical stage, and a vast variety of more advanced in vivo tumor models is currently available. Such complex animal models (e.g., genetically engineered mice and patient-derived xenografts) are capable of better predicting nanocarrier clinical efficiency, as they closely resemble the heterogeneity of the human tumor microenvironment.Herein, the development of physiologically more relevant in vitro and in vivo tumor models for the preclinical evaluation of anticancer nanoparticles will be discussed, as well as the current limitations and future challenges in clinical translation.
Collapse
Affiliation(s)
- Teresa R Abreu
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal.,UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Mariana Biscaia
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal
| | - Nélio Gonçalves
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal
| | - Nuno A Fonseca
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal.,TREAT U, SA, Parque Industrial de Taveiro, Lote 44, Coimbra, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal. .,UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal.
| |
Collapse
|
55
|
|
56
|
Tawfik SM, Azizov S, Elmasry MR, Sharipov M, Lee YI. Recent Advances in Nanomicelles Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 11:E70. [PMID: 33396938 PMCID: PMC7823398 DOI: 10.3390/nano11010070] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/26/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023]
Abstract
The efficient and selective delivery of therapeutic drugs to the target site remains the main obstacle in the development of new drugs and therapeutic interventions. Up until today, nanomicelles have shown their prospective as nanocarriers for drug delivery owing to their small size, good biocompatibility, and capacity to effectively entrap lipophilic drugs in their core. Nanomicelles are formed via self-assembly in aqueous media of amphiphilic molecules into well-organized supramolecular structures. Molecular weights and structure of the core and corona forming blocks are important properties that will determine the size of nanomicelles and their shape. Selective delivery is achieved via novel design of various stimuli-responsive nanomicelles that release drugs based on endogenous or exogenous stimulations such as pH, temperature, ultrasound, light, redox potential, and others. This review summarizes the emerging micellar nanocarriers developed with various designs, their outstanding properties, and underlying principles that grant targeted and continuous drug delivery. Finally, future perspectives, and challenges for nanomicelles are discussed based on the current achievements and remaining issues.
Collapse
Affiliation(s)
- Salah M. Tawfik
- Department of Materials Convergence and System Engineering, Changwon National University, Changwon 51140, Korea; (S.M.T.); (S.A.); (M.R.E.); (M.S.)
- Surfactant Laboratory, Department of Petrochemicals, Egyptian Petroleum Research Institute (EPRI), Nasr City, Cairo 11727, Egypt
| | - Shavkatjon Azizov
- Department of Materials Convergence and System Engineering, Changwon National University, Changwon 51140, Korea; (S.M.T.); (S.A.); (M.R.E.); (M.S.)
- Laboratory of Polysaccharide Chemistry, Institute of Bioorganic Chemistry, Uzbekistan Academy of Science, Tashkent 100125, Uzbekistan
| | - Mohamed R. Elmasry
- Department of Materials Convergence and System Engineering, Changwon National University, Changwon 51140, Korea; (S.M.T.); (S.A.); (M.R.E.); (M.S.)
| | - Mirkomil Sharipov
- Department of Materials Convergence and System Engineering, Changwon National University, Changwon 51140, Korea; (S.M.T.); (S.A.); (M.R.E.); (M.S.)
| | - Yong-Ill Lee
- Department of Materials Convergence and System Engineering, Changwon National University, Changwon 51140, Korea; (S.M.T.); (S.A.); (M.R.E.); (M.S.)
| |
Collapse
|
57
|
van Eerden RAG, Mathijssen RHJ, Koolen SLW. Recent Clinical Developments of Nanomediated Drug Delivery Systems of Taxanes for the Treatment of Cancer. Int J Nanomedicine 2020; 15:8151-8166. [PMID: 33132699 PMCID: PMC7592152 DOI: 10.2147/ijn.s272529] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/15/2020] [Indexed: 12/16/2022] Open
Abstract
Conventional taxanes are used as cornerstone of the chemotherapeutical treatment for a variety of malignancies. Nevertheless, a large proportion of patients do not benefit from their treatment while they do suffer from severe adverse events related to the solvent or to the active compound. Cremophor EL and polysorbate 80 free formulations, conjugates, oral formulations and different types of drug delivery systems are some examples of the several attempts to improve the treatment with taxanes. In this review article, we discuss recent clinical developments of nanomediated drug delivery systems of taxanes for the treatment of cancer. Targeting mechanisms of drug delivery systems and characteristics of the most commonly used taxane-containing drug delivery systems in the clinical setting will be discussed in this review.
Collapse
Affiliation(s)
- Ruben A G van Eerden
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.,Department of Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
58
|
Rehan F, Ahemad N, Islam RA, Gupta M, Gan SH, Chowdhury EH. Optimization and Formulation of Nanostructured and Self-Assembled Caseinate Micelles for Enhanced Cytotoxic Effects of Paclitaxel on Breast Cancer Cells. Pharmaceutics 2020; 12:pharmaceutics12100984. [PMID: 33080962 PMCID: PMC7589039 DOI: 10.3390/pharmaceutics12100984] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Paclitaxel (PTX) is a widely used anti-cancer drug for treating various types of solid malignant tumors including breast, ovarian and lung cancers. However, PTX has a low therapeutic response and is linked with acquired resistance, as well as a high incidence of adverse events, such as allergic reactions, neurotoxicity and myelosuppression. The situation is compounded when its complex chemical structure contributes towards hydrophobicity, shortening its circulation time in blood, causing off-target effects and limiting its therapeutic activity against cancer cells. Formulating a smart nano-carrier may overcome the solubility and toxicity issues of the drug and enable its more selective delivery to the cancerous cells. Among the nano-carriers, natural polymers are of great importance due to their excellent biodegradability, non-toxicity and good accessibility. The aim of the present research is to develop self-assembled sodium caseinate nanomicelles (NaCNs) with PTX loaded into the hydrophobic core of NaCNs for effective uptake of the drug in cancer cells and its subsequent intracellular release. METHODS The PTX-loaded micelle was characterized with high-performance liquid chromatography (HPLC), Fourier Transform Infrared Spectra (FTIR), High Resolution-Transmission Electron Microscope (HR-TEM), Field Emission Scanning Electron Microscope (FESEM) and Energy Dispersive X-Ray (EDX). Following treatment with PTX-loaded NaCNs, cell viability, cellular uptake and morphological changes were analyzed using MCF-7 and MDA-MB 231 human breast cancer cell lines. RESULTS We found that PTX-loaded NaCNs efficiently released PTX in an acidic tumor environment, while showing an enhanced cytotoxicity, cellular uptake and in-vivo anti-tumor efficacy in a mouse model of breast cancer when compared to free drug and blank micelles. Additionally, the nanomicelles also presented improved colloidal stability for three months at 4 °C and -20 °C and when placed at a temperature of 37 °C. CONCLUSIONS We conclude that the newly developed NaCNs is a promising carrier of PTX to enhance tumor accumulation of the drug while addressing its toxicity issues as well.
Collapse
Affiliation(s)
- Farah Rehan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
- Tropical Medicine and Biology Multidisciplinary Platform, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia
- Global Asia in the 21st century Research Platform, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia
| | - Rowshan Ara Islam
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia;
| | - Manish Gupta
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
- School of Pharmaceutical and Population Health Informatics, DIT University, Mussoorie-Diversion Road, Dehradun, Uttarakhand-248009, India
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
| | - Ezharul Hoque Chowdhury
- Tropical Medicine and Biology Multidisciplinary Platform, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia;
- Correspondence:
| |
Collapse
|
59
|
Doufène K, Malki Y, Vincent LA, Cuq P, Devoisselle JM, Masurier N, Aubert-Pouëssel A. Vegetable Oil-based Hybrid Submicron Particles Loaded with JMV5038: A Promising Formulation against Melanoma. J Pharm Sci 2020; 110:1197-1205. [PMID: 33069708 DOI: 10.1016/j.xphs.2020.10.019] [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: 08/11/2020] [Revised: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 11/29/2022]
Abstract
The aim of this work was to carry out a preformulation study on JMV5038 as a new potent cytotoxic agent, and to develop its formulation within vegetable oil-based hybrid submicron particles (HNP) in order to obtain a versatile dosage form against melanoma. JMV5038 was first characterized through physico-chemical tests and it exhibited high melting point and logP value, an important pH-sensitivity that led to the formation of well-identified degradation products at low pH, as well as a substantial solubility value in silylated castor oil (ICO). Then, JMV5038-loaded HNP were formulated through a thermostabilized emulsion process based on the sol-gel cross-linking of ICO. They showed high loading efficiency and their in vitro release kinetic assessed in a biorelevant PBS/octanol biphasic system showed a constant sustained release over one month. The cytotoxic activity and cytocompatibility of HNP were evaluated on A375 melanoma cells and NIH 3T3 cells, respectively. JMV5038-loaded HNP exhibited a slightly enhanced cytotoxic activity of JMV5038 on melanoma cells while demonstrating their safety on NIH 3T3 cells. In conclusion, JMV5038-loaded HNP proved to be an efficient and safe drug subcutaneous delivery system that will be interesting to evaluate through preclinical studies.
Collapse
Affiliation(s)
- Koceïla Doufène
- Institut Charles Gerhardt Montpellier (ICGM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Yohan Malki
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Laure-Anaïs Vincent
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Pierre Cuq
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Marie Devoisselle
- Institut Charles Gerhardt Montpellier (ICGM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Nicolas Masurier
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Anne Aubert-Pouëssel
- Institut Charles Gerhardt Montpellier (ICGM), University of Montpellier, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
60
|
Abstract
Polymeric micelles have gained interest as novel drug delivery systems for the treatment and diagnosis of cancer, as they offer several advantages over conventional drug therapies. This includes drug targeting to tumor tissue, in vivo biocompatibility and biodegradability, prolonged circulation time, enhanced accumulation, retention of the drug loaded micelle in the tumor and decreased side effects. This article provides an overview on the current state of micellar formulations as nanocarriers for anticancer drugs and their effectiveness in cancer therapeutics, including their clinical status. The type of copolymers used, their physicochemical properties and characterization as well as recent developments in the design of functional polymeric micelles are highlighted. The article also presents the design and outcomes of various types of stimuli-responsive polymeric micelles.
Collapse
|
61
|
Mi P, Miyata K, Kataoka K, Cabral H. Clinical Translation of Self‐Assembled Cancer Nanomedicines. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000159] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center West China Hospital, Sichuan University No. 17 People's South Road Chengdu 610041 China
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| | - Kazunori Kataoka
- Institute for Future Initiatives The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐0033 Japan
- Innovation Center of NanoMedicine Kawasaki Institute of Industrial Promotion 3‐25‐14, Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| |
Collapse
|
62
|
Hwang D, Ramsey JD, Kabanov AV. Polymeric micelles for the delivery of poorly soluble drugs: From nanoformulation to clinical approval. Adv Drug Deliv Rev 2020; 156:80-118. [PMID: 32980449 DOI: 10.1016/j.addr.2020.09.009] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 01/04/2023]
Abstract
Over the last three decades, polymeric micelles have emerged as a highly promising drug delivery platform for therapeutic compounds. Particularly, poorly soluble small molecules with high potency and significant toxicity were encapsulated in polymeric micelles. Polymeric micelles have shown improved pharmacokinetic profiles in preclinical animal models and enhanced efficacy with a superior safety profile for therapeutic drugs. Several polymeric micelle formulations have reached the clinical stage and are either in clinical trials or are approved for human use. This furthers interest in this field and underscores the need for additional learning of how to best design and apply these micellar carriers to improve the clinical outcomes of many drugs. In this review, we provide detailed information on polymeric micelles for the solubilization of poorly soluble small molecules in topics such as the design of block copolymers, experimental and theoretical analysis of drug encapsulation in polymeric micelles, pharmacokinetics of drugs in polymeric micelles, regulatory approval pathways of nanomedicines, and current outcomes from micelle formulations in clinical trials. We aim to describe the latest information on advanced analytical approaches for elucidating molecular interactions within the core of polymeric micelles for effective solubilization as well as for analyzing nanomedicine's pharmacokinetic profiles. Taking into account the considerations described within, academic and industrial researchers can continue to elucidate novel interactions in polymeric micelles and capitalize on their potential as drug delivery vehicles to help improve therapeutic outcomes in systemic delivery.
Collapse
Affiliation(s)
- Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M. V. Lomonosov Moscow State University, Moscow 119992, Russia.
| |
Collapse
|
63
|
Ahmad KS, Talat M, Jaffri SB, Shaheen N. Innovatory role of nanomaterials as bio-tools for treatment of cancer. REV INORG CHEM 2020. [DOI: 10.1515/revic-2020-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Conventional treatment modes like chemotherapy, thermal and radiations aimed at cancerous cells eradication are marked by destruction pointing the employment of nanomaterials as sustainable and auspicious materials for saving human lives. Cancer has been deemed as the second leading cause of death on a global scale. Nanomaterials employment in cancer treatment is based on the utilization of their inherent physicochemical characteristics in addition to their modification for using as nano-carriers and nano-vehicles eluted with anti-cancer drugs. Current work has reviewed the significant role of different types of nanomaterials in cancer therapeutics and diagnostics in a systematic way. Compilation of review has been done by analyzing voluminous investigations employing ERIC, MEDLINE, NHS Evidence and Web of Science databases. Search engines used were Google scholar, Jstore and PubMed. Current review is suggestive of the remarkable performance of nanomaterials making them candidates for cancer treatment for substitution of destructive treatment modes through investigation of their physicochemical characteristics, utilization outputs and long term impacts in patients.
Collapse
Affiliation(s)
- Khuram Shahzad Ahmad
- Department of Environmental Sciences , Fatima Jinnah Women University , The Mall, 46000 Rawalpindi , Pakistan
| | - Muntaha Talat
- Department of Environmental Sciences , Fatima Jinnah Women University , The Mall, 46000 Rawalpindi , Pakistan
| | - Shaan Bibi Jaffri
- Department of Environmental Sciences , Fatima Jinnah Women University , The Mall, 46000 Rawalpindi , Pakistan
| | | |
Collapse
|
64
|
Abstract
Polymeric micelles have gained interest as novel drug delivery systems for the treatment and diagnosis of cancer, as they offer several advantages over conventional drug therapies. This includes drug targeting to tumor tissue, in vivo biocompatibility and biodegradability, prolonged circulation time, enhanced accumulation, retention of the drug loaded micelle in the tumor and decreased side effects. This article provides an overview on the current state of micellar formulations as nanocarriers for anticancer drugs and their effectiveness in cancer therapeutics, including their clinical status. The type of copolymers used, their physicochemical properties and characterization as well as recent developments in the design of functional polymeric micelles are highlighted. The article also presents the design and outcomes of various types of stimuli-responsive polymeric micelles.
Collapse
|
65
|
Abbasi S, Sato Y, Kajimoto K, Harashima H. New Design Strategies for Controlling the Rate of Hydrophobic Drug Release from Nanoemulsions in Blood Circulation. Mol Pharm 2020; 17:3773-3782. [DOI: 10.1021/acs.molpharmaceut.0c00542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Tonomachi 3-25-14, Kawasaki 210-0821, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo 060 0812, Hokkaido, Japan
| | - Kazuaki Kajimoto
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, 2217-14 Hayashi-cho, Takamatsu 761-0395, Kagawa, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo 060 0812, Hokkaido, Japan
| |
Collapse
|
66
|
Bhatia R, Sharma A, Narang RK, Rawal RK. Recent Nanocarrier Approaches for Targeted Drug Delivery in Cancer Therapy. Curr Mol Pharmacol 2020; 14:350-366. [PMID: 32744982 DOI: 10.2174/1874467213666200730114943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 01/16/2023]
Abstract
Cancer is one of the most serious health concerns in the 21st century whose prevalence is beyond boundaries and can affect any organ of the human body. The conventional chemotherapeutic treatment strategies lack specificity to tumors and are associated with toxic effects on the immune system and other organ systems. In the past decades, there has been continuous progress in the development of smart nanocarrier systems for target-specific delivery of drugs against a variety of tumors, including intracellular gene-specific targeting. These nanocarriers are able to recognize the tumor cells and deliver the therapeutic agent in fixed proportions, causing no or very less harm to healthy cells. Nanosystems have modified physicochemical properties, improved bioavailability, and long retention in blood, which enhances their potency. A huge number of nanocarrier based formulations have been developed and are in clinical trials. Nanocarrier systems include polymeric micelles, liposomes, dendrimers, carbon nanotubes, gold nanoparticles, etc. Recent advancements in nanocarrier systems include mesoporous silica nanoparticles (MSNs), metal organic frameworks, and quantum dots. In the present review, various nanocarrier based drug delivery systems, along with their applications in the management of cancer, have been described with special emphasis on MSNs.
Collapse
Affiliation(s)
- Rohit Bhatia
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Amit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Raj K Narang
- Department of Pharmaceutics, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Ravindra K Rawal
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana-133207, Haryana, India
| |
Collapse
|
67
|
Ozturk N, Kara A, Gulyuz S, Ozkose UU, Tasdelen MA, Bozkir A, Yilmaz O, Vural I. Exploiting ionisable nature of PEtOx- co-PEI to prepare pH sensitive, doxorubicin-loaded micelles. J Microencapsul 2020; 37:467-480. [PMID: 32627670 DOI: 10.1080/02652048.2020.1792566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIMS This study was conducted to evaluate block copolymers containing two different poly(ethyleneimine) (PEI) amounts, as new pH-sensitive micellar delivery systems for doxorubicin. METHODS Micelles were prepared with block copolymers consisting of poly(2-ethyl-2-oxazoline)-co-poly(ethyleneimine) (PEtOx-co-PEI) and poly(ε-caprolactone) (PCL) as hydrophilic and hydrophobic blocks, respectively. Doxorubicin loading, micelle size, pH-dependent drug release, and in vitro cytotoxicity on MCF-7 cells were investigated. RESULTS The average size of drug-loaded micelles was under 100 nm and drug loading was between 10.7% and 48.3% (w/w). pH-sensitive drug release was more pronounced (84.7% and 68.9% (w/w) of drug was released at pH 5.0 and pH 7.4, respectively) for the micelles of the copolymer with the lowest PEI amount. The cell viability of doxorubicin-loaded micelles which were prepared by the copolymer with the lowest PEI amount was 28-33% at 72 h. CONCLUSIONS PEtOx-co-PEI-b-PCL micelles of this copolymer were found to be stable and effective pH-sensitive nano-sized carriers for doxorubicin delivery.
Collapse
Affiliation(s)
- Naile Ozturk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.,Department of Pharmaceutical Technology, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Asli Kara
- Department of Medical Services and Techniques, Sungurlu Vocational High School, Hitit University, Corum, Turkey.,Department of Nanotechnology and Nanomedicine, Hacettepe University Institute of Science, Ankara, Turkey
| | - Sevgi Gulyuz
- Materials Institute, Marmara Research Center, TUBITAK, Kocaeli, Turkey.,Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkey
| | - Umut Ugur Ozkose
- Materials Institute, Marmara Research Center, TUBITAK, Kocaeli, Turkey.,Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkey.,Department of Chemistry, Faculty of Science and Letters, Piri Reis University, Istanbul, Turkey
| | - Mehmet Atilla Tasdelen
- Department of Polymer Engineering, Faculty of Engineering, Yalova University, Yalova, Turkey
| | - Asuman Bozkir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Ozgur Yilmaz
- Materials Institute, Marmara Research Center, TUBITAK, Kocaeli, Turkey
| | - Imran Vural
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
68
|
Ullah R, Wazir J, Khan FU, Diallo MT, Ihsan AU, Mikrani R, Aquib M, Zhou X. Factors Influencing the Delivery Efficiency of Cancer Nanomedicines. AAPS PharmSciTech 2020; 21:132. [PMID: 32409932 DOI: 10.1208/s12249-020-01691-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/13/2020] [Indexed: 01/01/2023] Open
Abstract
The superiority of nanomedicine over conventional medicines in the treatment of cancer has gained immediate recognition worldwide. As traditional cancer therapies are nonspecific and detrimental to healthy cells, the ability of nanomedicine to release drugs to target tumor cells specifically instead of healthy cells has brought new hope to cancer patients. This review focuses on the effects of various factors of nanoparticles such as transport, concentration in cells, tumor microenvironment, interaction with protein, penetration, uptake by tumor cells, cancer cell mutations, and intracellular trafficking of the nanoparticle. Besides the history of nanomedicine, future perspectives of nanomedicines are also explored in this text.
Collapse
|
69
|
Sheybanifard M, Beztsinna N, Bagheri M, Buhl EM, Bresseleers J, Varela-Moreira A, Shi Y, van Nostrum CF, van der Pluijm G, Storm G, Hennink WE, Lammers T, Metselaar JM. Systematic evaluation of design features enables efficient selection of Π electron-stabilized polymeric micelles. Int J Pharm 2020; 584:119409. [PMID: 32389790 DOI: 10.1016/j.ijpharm.2020.119409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 11/26/2022]
Abstract
Polymeric micelles (PM) based on poly(ethylene glycol)-b-poly(N-2-benzoyloxypropyl methacrylamide) (mPEG-b-p(HPMA-Bz)) loaded with paclitaxel (PTX-PM) have shown promising results in overcoming the suboptimal efficacy/toxicity profile of paclitaxel. To get insight into the stability of PTX-PM formulations upon storage and to optimize their in vivo tumor-targeted drug delivery properties, we set out to identify a lead PTX-PM formulation with the optimal polymer composition. To this end, PM based on four different mPEG5k-b-p(HPMA-Bz) block copolymers with varying molecular weight of the hydrophobic block (17-3 kDa) were loaded with different amounts of PTX. The hydrodynamic diameter was 52 ± 1 nm for PM prepared using polymers with longer hydrophobic blocks (mPEG5k-b-p(HPMA-Bz)17k and mPEG5k-b-p(HPMA-Bz)10k) and 39 ± 1 nm for PM composed of polymers with shorter hydrophobic blocks (mPEG5k-b-p(HPMA-Bz)5k and mPEG5k-b-p(HPMA-Bz)3k). The best storage stability and the slowest PTX release was observed for PM with larger hydrophobic blocks. On the other hand, smaller sized PM of shorter mPEG5k-b-p(HPMA-Bz)5k showed a better tumor penetration in 3D spheroids. Considering better drug retention capacity of the mPEG5k-b-p(HPMA-Bz)17k and smaller size of the mPEG5k-b-p(HPMA-Bz)5k as two desirable design features, we argue that PM based on these two polymers are the lead candidates for further in vivo studies.
Collapse
Affiliation(s)
- Maryam Sheybanifard
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Nataliia Beztsinna
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands
| | - Mahsa Bagheri
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH University Hospital, Aachen, Germany
| | - Jaleesa Bresseleers
- ChemConnection BV - Ardena Oss, 5349 AB Oss, the Netherlands; Department of Bio-Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Aida Varela-Moreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands; Laboratory of Clinical Chemistry and Hematology (LKCH), University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands
| | - Gabri van der Pluijm
- Leiden University Medical Center, Department of Urology, J-3-108, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, the Netherlands; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - Josbert M Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands.
| |
Collapse
|
70
|
Liu Y, Fens MH, Lou B, van Kronenburg NC, Maas-Bakker RF, Kok RJ, Oliveira S, Hennink WE, van Nostrum CF. π-π-Stacked Poly(ε-caprolactone)- b-poly(ethylene glycol) Micelles Loaded with a Photosensitizer for Photodynamic Therapy. Pharmaceutics 2020; 12:pharmaceutics12040338. [PMID: 32283871 PMCID: PMC7238042 DOI: 10.3390/pharmaceutics12040338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
To improve the in vivo stability of poly(ε-caprolactone)-b-poly(ethylene glycol) (PCL-PEG)-based micelles and cargo retention by π-π stacking interactions, pendant aromatic rings were introduced by copolymerization of ε-caprolactone with benzyl 5-methyl-2-oxo-1,3-dioxane-5-carboxylate (TMC-Bz). It was shown that the incorporation of aromatic rings yielded smaller micelles (18–30 nm) with better colloidal stability in PBS than micelles without aromatic groups. The circulation time of i.v. injected micelles containing multiple pendant aromatic groups was longer (t½-α: ~0.7 h; t½-β: 2.9 h) than that of micelles with a single terminal aromatic group (t½ < 0.3 h). In addition, the in vitro partitioning of the encapsulated photosensitizer (meta-tetra(hydroxyphenyl)chlorin, mTHPC) between micelles and human plasma was favored towards micelles for those that contained the pendant aromatic groups. However, this was not sufficient to fully retain mTHPC in the micelles in vivo, as indicated by similar biodistribution patterns of micellar mTHPC compared to free mTHPC, and unequal biodistribution patterns of mTHPC and the host micelles. Our study points out that more detailed in vitro methods are necessary to more reliably predict in vivo outcomes. Furthermore, additional measures beyond π-π stacking are needed to stably incorporate mTHPC in micelles in order to benefit from the use of micelles as targeted delivery systems.
Collapse
Affiliation(s)
- Yanna Liu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Marcel H.A.M. Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Bo Lou
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Nicky C.H. van Kronenburg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Roel F.M. Maas-Bakker
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Robbert J. Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Cornelus F. van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
- Correspondence: ; Tel.: +31-620274607
| |
Collapse
|
71
|
Sartaj A, Baboota S, Ali J. Nanomedicine: A Promising Avenue for the Development of Effective Therapy for Breast Cancer. Curr Cancer Drug Targets 2020; 20:603-615. [PMID: 32228423 DOI: 10.2174/1568009620666200331124113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/26/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Breast cancer is the most probable cancer among women. However, the available treatment is based on targeting different stages of breast cancer viz., radiation therapy, hormonal therapy, chemotherapy, and surgical interventions, which have some limitations. The available chemotherapeutics are associated with problems like low solubility, low permeability, high first-pass metabolism, and P-glycoprotein efflux. Hence, the aforementioned restrictions lead to ineffective treatment. Multiple chemotherapeutics can also cause resistance in tumors. So, the purpose is to develop an effective therapeutic regimen for the treatment of breast cancer by applying a nanomedicinal approach. METHODS This review has been conducted on a systematic search strategy, based on relevant literature available on Pub Med, MedlinePlus, Google Scholar, and Sciencedirect up to November 2019 using keywords present in abstract and title of the review. As per our inclusion and exclusion criteria, 226 articles were screened. Among 226, a total of 40 articles were selected for this review. RESULTS The significant findings with the currently available treatment is that the drug, besides its distribution to the target-specific site, also distributes to healthy cells, which results in severe side effects. Moreover, the drug is less bioavailable at the site of action; therefore, to overcome this, a high dose is required, which again causes side effects and lower the benefits. Nanomedicinal approaches give an alternative approach to avoid the associated problems of available chemotherapeutics treatment of breast cancer. CONCLUSION The nanomedicinal strategies are useful over the conventional treatment of breast cancer and deliver a target-specific drug-using different novel drug delivery approaches.
Collapse
Affiliation(s)
- Ali Sartaj
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| |
Collapse
|
72
|
Yang R, Zheng Y, Shuai X, Fan F, He X, Ding M, Li J, Tan H, Fu Q. Crosslinking Induced Reassembly of Multiblock Polymers: Addressing the Dilemma of Stability and Responsivity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902701. [PMID: 32328415 PMCID: PMC7175344 DOI: 10.1002/advs.201902701] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/07/2020] [Accepted: 02/13/2020] [Indexed: 05/26/2023]
Abstract
Physical or chemical crosslinking of polymeric micelles has emerged as a straightforward approach to overcome the intrinsic instability of assemblies. However, the crosslinking process may compromise the responsivity of nanosystems and result in inefficient release of payloads. To address this dilemma, a crosslinking induced reassembly (CIRA) strategy is reported here to simultaneously increase the kinetic and thermodynamic stability and redox-responsivity of polymeric micelles. It is found that the click crosslinking of a model multiblock polyurethane at the micellar interface induces microphase separation between the soft and hard segments. The aggregation of hard domains gathers liable disulfide linkages around the interlayer of micelles, which could facilitate the attack of reducing agents and act as an intelligent on-off switch for high stability and triggered release. As a result, the CIRA approach enables an enhanced tumor targeting, improved biodistribution and excellent therapeutic efficacy in vivo. This work provides a facile and versatile platform for controlled delivery applications.
Collapse
Affiliation(s)
- Rui Yang
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Yi Zheng
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Xiaoyu Shuai
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Fan Fan
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Xueling He
- Laboratory Animal Center of Sichuan UniversityChengdu610041China
| | - Mingming Ding
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Jianshu Li
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Hong Tan
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| | - Qiang Fu
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengdu610065China
| |
Collapse
|
73
|
Avramović N, Mandić B, Savić-Radojević A, Simić T. Polymeric Nanocarriers of Drug Delivery Systems in Cancer Therapy. Pharmaceutics 2020; 12:E298. [PMID: 32218326 PMCID: PMC7238125 DOI: 10.3390/pharmaceutics12040298] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 01/10/2023] Open
Abstract
Conventional chemotherapy is the most common therapeutic method for treating cancer by the application of small toxic molecules thatinteract with DNA and causecell death. Unfortunately, these chemotherapeutic agents are non-selective and can damage both cancer and healthy tissues,producing diverse side effects, andthey can have a short circulation half-life and limited targeting. Many synthetic polymers have found application as nanocarriers of intelligent drug delivery systems (DDSs). Their unique physicochemical properties allow them to carry drugs with high efficiency,specificallytarget cancer tissue and control drug release. In recent years, considerable efforts have been made to design smart nanoplatforms, including amphiphilic block copolymers, polymer-drug conjugates and in particular pH- and redox-stimuli-responsive nanoparticles (NPs). This review is focused on a new generation of polymer-based DDSs with specific chemical functionalities that improve their hydrophilicity, drug loading and cellular interactions.Recentlydesigned multifunctional DDSs used in cancer therapy are highlighted in this review.
Collapse
Affiliation(s)
- Nataša Avramović
- Institute of Medical Chemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Boris Mandić
- Faculty of Chemistry, University of Belgrade, Studentski trg 12–16, 11000 Belgrade, Serbia;
| | - Ana Savić-Radojević
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.S.-R.); (T.S.)
| | - Tatjana Simić
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (A.S.-R.); (T.S.)
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| |
Collapse
|
74
|
Chariou PL, Ortega-Rivera OA, Steinmetz NF. Nanocarriers for the Delivery of Medical, Veterinary, and Agricultural Active Ingredients. ACS NANO 2020; 14:2678-2701. [PMID: 32125825 PMCID: PMC8085836 DOI: 10.1021/acsnano.0c00173] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nanocarrier-based delivery systems can be used to increase the safety and efficacy of active ingredients in medical, veterinary, or agricultural applications, particularly when such ingredients are unstable, sparingly soluble, or cause off-target effects. In this review, we highlight the diversity of nanocarrier materials and their key advantages compared to free active ingredients. We discuss current trends based on peer-reviewed research articles, patent applications, clinical trials, and the nanocarrier formulations already approved by regulatory bodies. Although most nanocarriers have been engineered to combat cancer, the number of formulations developed for other purposes is growing rapidly, especially those for the treatment of infectious diseases and parasites affecting humans, livestock, and companion animals. The regulation and prohibition of many pesticides have also fueled research to develop targeted pesticide delivery systems based on nanocarriers, which maximize efficacy while minimizing the environmental impact of agrochemicals.
Collapse
|
75
|
Ali I, Alsehli M, Scotti L, Tullius Scotti M, Tsai ST, Yu RS, Hsieh MF, Chen JC. Progress in Polymeric Nano-Medicines for Theranostic Cancer Treatment. Polymers (Basel) 2020; 12:E598. [PMID: 32155695 PMCID: PMC7182942 DOI: 10.3390/polym12030598] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is a life-threatening disease killing millions of people globally. Among various medical treatments, nano-medicines are gaining importance continuously. Many nanocarriers have been developed for treatment, but polymerically-based ones are acquiring importance due to their targeting capabilities, biodegradability, biocompatibility, capacity for drug loading and long blood circulation time. The present article describes progress in polymeric nano-medicines for theranostic cancer treatment, which includes cancer diagnosis and treatment in a single dosage form. The article covers the applications of natural and synthetic polymers in cancer diagnosis and treatment. Efforts were also made to discuss the merits and demerits of such polymers; the status of approved nano-medicines; and future perspectives.
Collapse
Affiliation(s)
- Imran Ali
- Department of Chemistry, College of Sciences, Taibah University, Al-Medina Al-Munawara 41477, Saudi Arabia;
- Department of Chemistry, Jamia Millia Islamia (Central University), New Delhi 110025, India
| | - Mosa Alsehli
- Department of Chemistry, College of Sciences, Taibah University, Al-Medina Al-Munawara 41477, Saudi Arabia;
| | - Luciana Scotti
- Cheminformatics Laboratory—Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, João Pessoa 58051-970, PB, Brazil; (L.S.); (M.T.S.)
| | - Marcus Tullius Scotti
- Cheminformatics Laboratory—Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, João Pessoa 58051-970, PB, Brazil; (L.S.); (M.T.S.)
| | - Shang-Ting Tsai
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Road, Chung Li District, Taoyuan 32023, Taiwan; (S.-T.T.); (R.-S.Y.); (M.F.H.)
- Center for Minimally-Invasive Medical Devices and Technologies, Chung Yuan Christian University, 200 Chung Pei Road, Chung Li District, Taoyuan 32023, Taiwan
| | - Ruei-Siang Yu
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Road, Chung Li District, Taoyuan 32023, Taiwan; (S.-T.T.); (R.-S.Y.); (M.F.H.)
- Department of Pharmacy, Kaohsiung Armed Forces General Hospital, No.2, Zhongzheng 1st Rd., Lingya Dist., Kaohsiung 80284, Taiwan
| | - Ming Fa Hsieh
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Road, Chung Li District, Taoyuan 32023, Taiwan; (S.-T.T.); (R.-S.Y.); (M.F.H.)
- Center for Minimally-Invasive Medical Devices and Technologies, Chung Yuan Christian University, 200 Chung Pei Road, Chung Li District, Taoyuan 32023, Taiwan
| | - Jung-Chih Chen
- Institute of Biomedical Engineering, National Chiao Tung University, 1001 University Rd., Hsinchu 300, Taiwan;
| |
Collapse
|
76
|
Demazeau M, Gibot L, Mingotaud AF, Vicendo P, Roux C, Lonetti B. Rational design of block copolymer self-assemblies in photodynamic therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:180-212. [PMID: 32082960 PMCID: PMC7006492 DOI: 10.3762/bjnano.11.15] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/04/2019] [Indexed: 05/10/2023]
Abstract
Photodynamic therapy is a technique already used in ophthalmology or oncology. It is based on the local production of reactive oxygen species through an energy transfer from an excited photosensitizer to oxygen present in the biological tissue. This review first presents an update, mainly covering the last five years, regarding the block copolymers used as nanovectors for the delivery of the photosensitizer. In particular, we describe the chemical nature and structure of the block copolymers showing a very large range of existing systems, spanning from natural polymers such as proteins or polysaccharides to synthetic ones such as polyesters or polyacrylates. A second part focuses on important parameters for their design and the improvement of their efficiency. Finally, particular attention has been paid to the question of nanocarrier internalization and interaction with membranes (both biomimetic and cellular), and the importance of intracellular targeting has been addressed.
Collapse
Affiliation(s)
- Maxime Demazeau
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, 118 route de Narbonne, 31062, Toulouse, France
| | - Laure Gibot
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, 118 route de Narbonne, 31062, Toulouse, France
| | - Anne-Françoise Mingotaud
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, 118 route de Narbonne, 31062, Toulouse, France
| | - Patricia Vicendo
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, 118 route de Narbonne, 31062, Toulouse, France
| | - Clément Roux
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, 118 route de Narbonne, 31062, Toulouse, France
| | - Barbara Lonetti
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, 118 route de Narbonne, 31062, Toulouse, France
| |
Collapse
|
77
|
Norouzi M, Amerian M, Amerian M, Atyabi F. Clinical applications of nanomedicine in cancer therapy. Drug Discov Today 2020; 25:107-125. [DOI: 10.1016/j.drudis.2019.09.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/03/2019] [Accepted: 09/24/2019] [Indexed: 12/23/2022]
|
78
|
Harwansh RK, Deshmukh R, Barkat MA, Rahman MA. Bioinspired Polymeric-based Core-shell Smart Nano-systems. Pharm Nanotechnol 2019; 7:181-205. [PMID: 31486750 DOI: 10.2174/2211738507666190429104550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/03/2018] [Accepted: 04/10/2019] [Indexed: 12/20/2022]
Abstract
Smart nanosystems (SNs) have the potential to revolutionize drug delivery. Conventional drug delivery systems have poor drug-loading, early burst release, limited therapeutic effects, etc. Thus, to overcome these problems, researchers have taken advantage of the host-guest interactions as bioinspired nanosystems which can deliver nanocarriers more efficiently with the maximum drug loading capacity and improved therapeutic efficacy as well as bioavailability. SNs employ nanomaterials to form cage molecules by entrapping new nanocarriers called smart nanosystems in their cargo and design. The activities of SNs are based on responsive materials that interact with the stimuli either by changing their properties or conformational structures. The aptitude of living systems to respond to stimuli and process information has encouraged researchers to build up integrated nanosystems exhibiting similar function and therapeutic response. Various smart materials, including polymers, have been exhaustively employed in fabricating different stimuli-responsive nanosystems which can deliver bioactive molecules to a specific site for a certain period with minimal side effects. SNs have been widely explored to deliver diverse kinds of therapeutic agents ranging from bioactive compounds, genes, and biopharmaceuticals like proteins and peptides, to diagnostic imaging agents for biomedical applications. Nanotechnology-based different nanosystems are promising for health care issues. The advancement of SNs with physical science and engineering technology in synthesizing nanostructures and their physicochemical characterization should be exploited in medicine and healthcare for reducing mortality rate, morbidity, disease prevalence and general societal burden.
Collapse
Affiliation(s)
- Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura -281406, India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura -281406, India
| | - Md Abul Barkat
- Department of Pharmaceutics, School of Medical and Allied Sciences, K.R. Mangalam University, Sohna, Gurgaon, India
| | | |
Collapse
|
79
|
Hurley SK, Cutrone NM, Fath KR, Pajovich HT, Garcia J, Smith AM, Banerjee IA. Self-assembled phenylisoxazole-peptide hybrid assemblies and their interactions with breast and ovarian tumor cells. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1525542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Sara K. Hurley
- Department of Chemistry, Fordham University, Bronx, NY, USA
| | | | - Karl R. Fath
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | - Jeremy Garcia
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | | |
Collapse
|
80
|
Feng S, Zhou J, Li Z, Appelman HD, Zhao L, Zhu J, Wang TD. Sorafenib encapsulated in nanocarrier functionalized with glypican-3 specific peptide for targeted therapy of hepatocellular carcinoma. Colloids Surf B Biointerfaces 2019; 184:110498. [PMID: 31536939 DOI: 10.1016/j.colsurfb.2019.110498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/23/2019] [Accepted: 09/08/2019] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer in the world with increasing incidence. Chemotherapy is required for HCC patients after receiving surgical resection. Serious off-target induced side effects and systemic toxicity limit the clinical utility of drugs. Targeting therapeutic nanomedicine is an innovative strategy for enhancing drug delivery efficiency and reducing side effects. Here, we successfully formulated nanocarriers to encapsulate sorafenib, an FDA approved drug for treatment of HCC. Sorafenib is encapsulated with an entrapment efficiency >80% over 20 days. The effective aqueous solubility is improved over 1900-fold. The release ratio in vitro is characterized by a half-life of T1/2 = 22.7 h. The peak target-to-background ratio for nanocarrier uptake by tumor occurs at 24 h post-injection, and is significantly greater for the target peptide versus controls. Ex vivo biodistribution confirms the in vivo results. Tumor regression is significantly greater for the target peptide versus controls after 21 days of therapy. No acute toxicity is found by blood chemistry or necropsy. In summary, a peptide specific for GPC3 has been identified, and used to modify the surface of a nanocarrier that encapsulates sorafenib with high entrapment efficiency. Regression of HCC xenograft tumors showed promise for targeted drug delivery.
Collapse
Affiliation(s)
- Shuo Feng
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Juan Zhou
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Henry D Appelman
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China.
| | - Thomas D Wang
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
81
|
Barkat MA, Beg S, Pottoo FH, Ahmad FJ. Nanopaclitaxel therapy: an evidence based review on the battle for next-generation formulation challenges. Nanomedicine (Lond) 2019; 14:1323-1341. [PMID: 31124758 DOI: 10.2217/nnm-2018-0313] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The poor solubility of paclitaxel (PTX), the most commonly used anticancer drug (Taxol®), has long hindered the development of successful formulations. In 2005, the launch of Abraxane®, a human albumin-based preparation of PTX, competed with Taxol® in the commercial market. The success of Abraxane pushed other generic preparations aside, sparking competition among the global pharmaceutical companies to develop the novel and superior PTX nanotechnology-driven formulations. Unsurprisingly, the success underlying with cancer treatment using nano PTX therapy has now entered into a new era of drug development, patentability, preclinical and clinical evaluation, leading eventually to a significant increase in the regulatory approval of the products. The present article aims to provide recent progress in the development of nano PTX formulations by various pharmaceutical companies for safe and effective drug therapies for patients benefit.
Collapse
Affiliation(s)
- Md Abul Barkat
- Department of Pharmaceutics, School of Medical & Allied Sciences, KR Mangalam University, Gurgaon, Sohna, Haryana, India.,Nanomedicine Research Lab, Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Sarwar Beg
- Nanomedicine Research Lab, Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Faheem H Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University (Formerly University of Dammam), 31441, Dammam, Saudi Arabia
| | - Farhan J Ahmad
- Nanomedicine Research Lab, Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
82
|
Khalifa AM, Elsheikh MA, Khalifa AM, Elnaggar YSR. Current strategies for different paclitaxel-loaded Nano-delivery Systems towards therapeutic applications for ovarian carcinoma: A review article. J Control Release 2019; 311-312:125-137. [PMID: 31476342 DOI: 10.1016/j.jconrel.2019.08.034] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022]
Abstract
Ovarian carcinoma (OC) is one of the leading causes of death among gynecologic malignancies all over the world. It is characterized by high mortality rate because of the lack of early diagnosis. The first-line chemotherapeutic regimen for late stage epithelial ovarian cancer is paclitaxel in combination to carboplatin. However, in most of cases, relapse occurs within six months despite the initial success of this chemotherapeutic combination. A lot of challenges have been encountered with the conventional delivery of paclitaxel in addition to the occurrence of severe off-target toxicity. One major problem is poor paclitaxel solubility which was improved by addition of Cremophor EL that unfortunately resulted in hypersensitivity side effects. Another obstacle is the multi drug resistance which is the main cause of OC recurrence. Accordingly, incorporation of paclitaxel, solely or in combination to other drugs, in nanocarrier systems has grabbed attention of many researchers to circumvent all these hurdles. The current review is the first article that provides a comprehensive overview on multi-faceted implementations of paclitaxel loaded nanoplatforms to solve delivery obstacles of paclitaxel in management of ovarian carcinoma. Moreover, challenges in physicochemical properties, biological activity and targeted delivery of PTX were depicted with corresponding solutions using nanotechnology. Different categories of nanocarriers employed were collected included lipid, protein, polymeric, solid nanoemulsion and hybrid systems. Future perspectives including imperative research considerations in ovarian cancer therapy were proposed as well.
Collapse
Affiliation(s)
- Alaa M Khalifa
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Manal A Elsheikh
- Department of pharmaceutics, Faculty of Pharmacy, Damanhur University, Damanhur, Egypt
| | - Amr M Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | - Yosra S R Elnaggar
- Head of International Publication and Nanotechnology Consultation Center INCC, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Egypt; Department of Pharmaceutics Faculty of Pharmacy, Alexandria University, Egypt.
| |
Collapse
|
83
|
Parveen S, Arjmand F, Tabassum S. Clinical developments of antitumor polymer therapeutics. RSC Adv 2019; 9:24699-24721. [PMID: 35528643 PMCID: PMC9069890 DOI: 10.1039/c9ra04358f] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/18/2019] [Indexed: 01/04/2023] Open
Abstract
Polymer therapeutics encompasses polymer-drug conjugates that are nano-sized, multicomponent constructs already in the clinic as antitumor compounds, either as single agents or in combination with other organic drug scaffolds. Nanoparticle-based polymer-conjugated therapeutics are poised to become a leading delivery strategy for cancer treatments as they exhibit prolonged half-life, higher stability and selectivity, water solubility, longer clearance time, lower immunogenicity and antigenicity and often also specific targeting to tissues or cells. Compared to free drugs, polymer-tethered drugs preferentially accumulate in the tumor sites unlike conventional chemotherapy which does not discriminate between the cancer cells and healthy cells, thereby causing severe side-effects. It is also desirable that the drug reaches its site of action at a particular concentration and the therapeutic dose remains constant over a sufficiently long period of time. This can be achieved by opting for new formulations possessing polymeric systems of drug carriers. However, many challenges still remain unanswered in polymeric drug conjugates which need to be readdressed and therefore, can broaden the scope of this field. This review highlights some of the antitumor polymer therapeutics including polymer-drug conjugates, polymeric micelles, polymeric liposomes and other polymeric nanoparticles that are currently under investigation.
Collapse
Affiliation(s)
- Shazia Parveen
- Chemistry Department, Faculty of Science, Taibah University Yanbu Branch 46423 Yanbu Saudi Arabia +966 504522069
| | - Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University Aligarh-202002 India
| | - Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University Aligarh-202002 India
| |
Collapse
|
84
|
Takahashi M, Miki S, Fujimoto K, Fukuoka K, Matsushita Y, Maida Y, Yasukawa M, Hayashi M, Shinkyo R, Kikuchi K, Mukasa A, Nishikawa R, Tamura K, Narita Y, Hamada A, Masutomi K, Ichimura K. Eribulin penetrates brain tumor tissue and prolongs survival of mice harboring intracerebral glioblastoma xenografts. Cancer Sci 2019; 110:2247-2257. [PMID: 31099446 PMCID: PMC6609810 DOI: 10.1111/cas.14067] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/24/2019] [Accepted: 05/13/2019] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is one of the most devastating human malignancies for which a novel efficient treatment is urgently required. This pre-clinical study shows that eribulin, a specific inhibitor of telomerase reverse transcriptase (TERT)-RNA-dependent RNA polymerase, is an effective anticancer agent against glioblastoma. Eribulin inhibited the growth of 4 TERT promoter mutation-harboring glioblastoma cell lines in vitro at subnanomolar concentrations. In addition, it suppressed the growth of glioblastoma cells transplanted subcutaneously or intracerebrally into mice, and significantly prolonged the survival of mice harboring brain tumors at a clinically equivalent dose. A pharmacokinetics study showed that eribulin quickly penetrated brain tumors and remained at a high concentration even when it was washed away from plasma, kidney or liver 24 hours after intravenous injection. Moreover, a matrix-assisted laser desorption/ionization mass spectrometry imaging analysis revealed that intraperitoneally injected eribulin penetrated the brain tumor and was distributed evenly within the tumor mass at 1 hour after the injection whereas only very low levels of eribulin were detected in surrounding normal brain. Eribulin is an FDA-approved drug for refractory breast cancer and can be safely repositioned for treatment of glioblastoma patients. Thus, our results suggest that eribulin may serve as a novel therapeutic option for glioblastoma. Based on these data, an investigator-initiated registration-directed clinical trial to evaluate the safety and efficacy of eribulin in patients with recurrent GBM (UMIN000030359) has been initiated.
Collapse
Affiliation(s)
- Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Shunichiro Miki
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenji Fujimoto
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Kohei Fukuoka
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuko Matsushita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshiko Maida
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Mami Yasukawa
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Mitsuhiro Hayashi
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Raku Shinkyo
- Tsukuba Research Laboratory, Eisai, Tsukuba, Japan
| | | | - Akitake Mukasa
- Department of Neurosurgery, The University of Tokyo, Tokyo, Japan
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenkichi Masutomi
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
85
|
Lynn GM, Laga R, Jewell CM. Induction of anti-cancer T cell immunity by in situ vaccination using systemically administered nanomedicines. Cancer Lett 2019; 459:192-203. [PMID: 31185250 DOI: 10.1016/j.canlet.2019.114427] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
Patients with inadequate anti-cancer T cell responses experience limited benefit from immune checkpoint inhibitors and other immunotherapies that require T cells. Therefore, treatments that induce de novo anti-cancer T cell immunity are needed. One strategy - referred to as in situ vaccination - is to deliver chemotherapeutic or immunostimulatory drugs into tumors to promote cancer cell death and provide a stimulatory environment for priming T cells against antigens already present in the tumor. However, achieving sufficient drug concentrations in tumors without causing dose-limiting toxicities remains a major challenge. To address this challenge, nanomedicines based on nano-sized carriers ('nanocarriers') of chemotherapeutics and immunostimulants are being developed to improve drug accumulation in tumors following systemic (intravenous) administration. Herein, we present the rationale for using systemically administrable nanomedicines to induce anti-cancer T cell immunity via in situ vaccination and provide an overview of synthetic nanomedicines currently used clinically. We also describe general strategies for improving nanomedicine design to increase tumor uptake, including use of micelle- and star polymer-based nanocarriers. We conclude with perspectives for how nanomedicine properties, host factors and treatment combinations can be leveraged to maximize efficacy.
Collapse
Affiliation(s)
- Geoffrey M Lynn
- Fischell Department of Bioengineering, A. James Clark Hall, Room 5110, 8278 Paint Branch Drive, College Park, MD, 20742, USA; Avidea Technologies, Baltimore, MD, 21205, USA
| | - Richard Laga
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague, Czech Republic
| | - Christopher M Jewell
- Fischell Department of Bioengineering, A. James Clark Hall, Room 5110, 8278 Paint Branch Drive, College Park, MD, 20742, USA; United States Department of Veterans Affairs, VA Maryland Health Care System, 10 North Greene Street, Baltimore, MD, 21201, USA; Robert E. Fischell Institute for Biomedical Devices, A. James Clark Hall, Room 5110, 8278 Paint Branch Drive, College Park, MD, 20742, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Cancer Center, Executive Office, Suite N9E17, 22 S. Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
86
|
Houdaihed L, Evans JC, Allen C. In Vivo Evaluation of Dual-Targeted Nanoparticles Encapsulating Paclitaxel and Everolimus. Cancers (Basel) 2019; 11:E752. [PMID: 31146485 PMCID: PMC6628352 DOI: 10.3390/cancers11060752] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 01/01/2023] Open
Abstract
A synergistic combination of paclitaxel (PTX) and everolimus (EVER) can allow for lower drug doses, reducing the toxicities associated with PTX, while maintaining therapeutic efficacy. Polymeric nanoparticles (NPs) of high stability provide opportunities to modify the toxicity profile of the drugs by ensuring their delivery to tumor at the synergistic ratio while limiting systemic drug exposure and the toxicities that result. The current study goal is to study the in vivo fate of human epidermal factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR) dual-targeted PTX+EVER-loaded NPs (Dual-NPs) in an MDA-MB-231-H2N BC tumor-bearing mouse model. The pharmacokinetic parameters, plasma area under the curve (AUC) and half-life (t1/2), were found to be 20-fold and 3 to 4-fold higher, respectively, for the drugs when administered in the Dual-NPs in comparison to the free-drug combination (i.e., PTX+EVER) at an equivalent dose of PTX. While maintaining anti-tumor efficacy, the levels of body weight loss were significantly lower (p < 0.0001) and the overall degree of neurotoxicity was reduced with Dual-NP treatment in comparison to the free-drug combination when administered at an equivalent dose of PTX. This study suggests that Dual-NPs present a promising platform for the delivery of the PTX and EVER combination with the potential to reduce severe PTX-induced toxicities and in turn, improve quality of life for patients with BC.
Collapse
Affiliation(s)
- Loujin Houdaihed
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | | | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
87
|
Ahangar P, Aziz M, Rosenzweig DH, Weber MH. Advances in personalized treatment of metastatic spine disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:223. [PMID: 31297388 DOI: 10.21037/atm.2019.04.41] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spine is one of the most common sites of bony metastases, and its involvement leads to significant patient morbidity. Surgical management in these patients is aimed at improving quality of life and functional status throughout the course of the disease. Resection of metastases often leads to critical size bone defects, presenting a challenge to achieving adequate bone regeneration to fill the void. Current treatment options for repairing these defects are bone grafting and commercial bone cements; however, each has associated limitations. Additionally, tumor recurrence and tumor-induced bone loss make bone regeneration particularly difficult. Systemic therapeutic delivery, such as bisphosphonates, have become standard of care to combat bone loss despite unfavorable systemic side-effects and lack of local efficacy. Developments from tissue engineering have introduced novel materials with osteoinductive and osteoconductive properties which also act as structural support scaffolds for bone regeneration. These new materials can also act as a therapeutic reservoir to sustainably release drugs locally as an alternative to systemic therapy. In this review, we outline recent advancements in tissue engineering and the role of translational research in developing implants that can fully repair bone defects while also delivering local therapeutics to curb tumor recurrence and improve patient quality of life.
Collapse
Affiliation(s)
- Pouyan Ahangar
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada
| | - Mina Aziz
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada.,Clinical Investigator Program, McGill University, Montreal, QC, Canada
| | - Derek H Rosenzweig
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada
| | - Michael H Weber
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada
| |
Collapse
|
88
|
Brannigan RP, Kimmins SD, Bobbi E, Caulfield S, Heise A. Synthesis of Novel
bis
‐Triazolinedione Crosslinked Amphiphilic Polypept(o)ide Nanostructures. MACROMOL CHEM PHYS 2019. [DOI: 10.1002/macp.201900067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Ruairí P. Brannigan
- Department of ChemistryRoyal College of Surgeons in Ireland (RCSI) 123 St Stephen's Green Dublin 2 D02 YN77 Ireland
| | - Scott D. Kimmins
- Department of ChemistryRoyal College of Surgeons in Ireland (RCSI) 123 St Stephen's Green Dublin 2 D02 YN77 Ireland
| | - Elena Bobbi
- Department of ChemistryRoyal College of Surgeons in Ireland (RCSI) 123 St Stephen's Green Dublin 2 D02 YN77 Ireland
| | - Séamus Caulfield
- Department of ChemistryRoyal College of Surgeons in Ireland (RCSI) 123 St Stephen's Green Dublin 2 D02 YN77 Ireland
| | - Andreas Heise
- Department of ChemistryRoyal College of Surgeons in Ireland (RCSI) 123 St Stephen's Green Dublin 2 D02 YN77 Ireland
| |
Collapse
|
89
|
Zang X, Lee JB, Deshpande K, Garbuzenko OB, Minko T, Kagan L. Prevention of paclitaxel-induced neuropathy by formulation approach. J Control Release 2019; 303:109-116. [PMID: 30981814 DOI: 10.1016/j.jconrel.2019.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/16/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major adverse effect of paclitaxel. Several liposome-based products have been approved and demonstrated superior efficacy and safety profiles for other drugs. The first objective of this work was to evaluate the effect of liposome formulation of paclitaxel (L-PTX) on neurotoxicity in-vitro and in-vivo in comparison to the standard Taxol® formulation. The second aim was to investigate the effect of formulation on paclitaxel biodistribution following intravenous administration in an animal model. Free paclitaxel was toxic to cell of neuronal origin (IC50 = 18.4 μg/mL) at a lower concentration than to lung cancer cells (IC50 = 59.1 μg/mL), and L-PTX demonstrated a comparable toxicity in both cell lines (IC50 = 31.8 and 33.7 μg/mL). Administration of L-PTX at 2 mg/kg per dose for a total of 4 doses on day 0, 2, 4, and 6 to rats did not result in increased sensitivity in response to mechanical or thermal stimulation of hind paws, in comparison to Taxol® administration at the same dose level that resulted in neuropathy. Paclitaxel biodisposition was evaluated for two formulations in plasma, liver, lung, brain, spinal cord, skin and muscle of rats after single intravenous dose at 6 mg/kg. The exposure to paclitaxel in brain, spinal cord, muscle, and skin was lower in the L-PTX group compared to Taxol® group. PEGylated liposomes containing paclitaxel were successfully developed and demonstrated reduced neurotoxicity in-vitro in neuronal cells and prevented development of peripheral neuropathy in-vivo. This proof of concept study showed that formulation in nanoparticles is a promising approach for reducing (or preventing) neurotoxicity caused by cancer drugs.
Collapse
Affiliation(s)
- Xiaowei Zang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Jong Bong Lee
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Kiran Deshpande
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Olga B Garbuzenko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America.
| |
Collapse
|
90
|
Development and Biological Analysis of a Novel Orthotopic Peritoneal Dissemination Mouse Model Generated Using a Pancreatic Ductal Adenocarcinoma Cell Line. Pancreas 2019; 48:315-322. [PMID: 30747829 PMCID: PMC6426353 DOI: 10.1097/mpa.0000000000001253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Peritoneal dissemination (PD) is an important cause of morbidity and mortality among patients with pancreatic ductal adenocarcinoma (PDAC). We sought to develop and characterized a novel PD mouse model by using a previously established PDAC cell line TCC-Pan2. METHODS TCC-Pan2 cell line was characterized for growth rate, tumor markers, histology, and somatic mutations. TCC-Pan2 cells were implanted orthotopically to produce PD. TCC-Pan2 cells from these metastatic foci were expanded in vitro and then implanted orthotopically in mice. This PD model was used for comparing the antitumor effect of paclitaxel and NK105. RESULTS Orthotopically implanted TCC-Pan2 cells caused tumor formation and PD with high frequency in mice. A potent metastatic subline-Pan2M-was obtained. NK105 exerted a stronger antitumor effect than paclitaxel against Pan2M cells harboring a luciferase gene (Pan2MmLuc). Notably, the survival rate on day 80 in the Pan2MmLuc mouse model was 100% for the NK105 group and 0% for the paclitaxel group. CONCLUSION TCC-Pan2 cell line and Pan2MmLuc PD model can serve as useful tools for monitoring the responses to antineoplastic agents and for studying PDAC biology.
Collapse
|
91
|
Fujiwara Y, Mukai H, Saeki T, Ro J, Lin YC, Nagai SE, Lee KS, Watanabe J, Ohtani S, Kim SB, Kuroi K, Tsugawa K, Tokuda Y, Iwata H, Park YH, Yang Y, Nambu Y. A multi-national, randomised, open-label, parallel, phase III non-inferiority study comparing NK105 and paclitaxel in metastatic or recurrent breast cancer patients. Br J Cancer 2019; 120:475-480. [PMID: 30745582 PMCID: PMC6461876 DOI: 10.1038/s41416-019-0391-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/09/2019] [Accepted: 01/17/2019] [Indexed: 12/02/2022] Open
Abstract
Background NK105 is a novel nanoparticle drug delivery formulation that encapsulates paclitaxel (PTX) in polymeric micelles. We conducted an open-label phase III non-inferiority trial to compare the efficacy and safety of NK105 and PTX in metastatic or recurrent breast cancer. Methods Patients were randomly assigned in a 1:1 ratio to receive either NK105 (65 mg/m2) or PTX (80 mg/m2) on days 1, 8 and 15 of a 28-day cycle. The primary endpoint was progression-free survival (PFS), with a non-inferiority margin of 1.215. Results A total of 436 patients were randomised and 211 patients in each group were included in the efficacy analysis. The median PFS was 8.4 and 8.5 months for NK105 and PTX, respectively (adjusted hazard ratio: 1.255; 95% confidence interval: 0.989–1.592). The median overall survival and overall response rates were 31.2 vs. 36.2 months and 31.6% vs. 39.0%, respectively. The two groups exhibited similar safety profiles. The incidence of peripheral sensory neuropathy (PSN) was 1.4% vs. 7.5% (≥Grade 3) for NK105 and PTX, respectively. The patient-reported outcomes of PSN were significantly favourable for NK105 (P < 0.0001). Conclusions The primary endpoint was not met, but NK105 had a better PSN toxicity profile than PTX. Clinical trial registration ClinicalTrials.gov: NCT01644890
Collapse
Affiliation(s)
- Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hirofumi Mukai
- Division of Breast and Medical Oncology, National Cancer Center Hospital East, Chiba, Japan.
| | - Toshiaki Saeki
- Department of Breast Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Jungsil Ro
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Yung-Chang Lin
- Division of Haematology and Oncology, Chang-Gung Memorial Hospital, Linko, Taoyuan, Taiwan
| | | | - Keun Seok Lee
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | | | - Shoichiro Ohtani
- Department of Breast Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Sung Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Katsumasa Kuroi
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Koichiro Tsugawa
- Division of Breast and Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yutaka Tokuda
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yeon Hee Park
- Division of Hematology-Oncology, Samsung Medical Center, Seoul, Korea
| | - Youngsen Yang
- Division of Hematology-Oncology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Internal Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Yoshihiro Nambu
- Pharmaceuticals Group, Nippon Kayaku Co., Ltd., Tokyo, Japan
| |
Collapse
|
92
|
Piao L, Li Y, Zhang H, Jiang J. Stereocomplex micelle loaded with paclitaxel for enhanced therapy of breast cancer in an orthotopic mouse model. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 30:233-246. [PMID: 30606090 DOI: 10.1080/09205063.2019.1565612] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Micelles are promising a nano drug carrier for cancer therapy. However, their application is often limited due to the instability of them in vivo. Herein, we reported the development of stereocomplex micelle (SCM) based on amphiphilic dextran-block-polylactide (Dex-b-PLA) that could improve the stability of micelles, reduce the early release of loaded drugs and target the breast cancer through the enhanced permeability and retention (EPR) effect for enhanced breast cancer therapy. The SCM were fabricated from the equimolar mixture of the enantiomeric Dex-b-PLA copolymers. Paclitaxel (PTX) as a model anti breast cancer drug was loaded in the SCM, noted as SCM/PTX. Transmission electron microscopy (TEM) and dynamic laser scattering (DLS) showed the diameter of SCM/PTX was below100 nm, which was suitable sizes for the EPR effect. The release kinetics of SCM/PTX exhibited that the release of PTX was obviously slow down and showed constant release. In the in vitro antitumor test, the SCM/PTX could effectively suppress the viability of 4T1 cells, which was demonstrated by the MTT assay. Moreover, the SCM/PTX could reduce the distribution of PTX at normal organs and obviously increase the accumulation of PTX at tumor sites. The circulation time of SCM/PTX was also obviously enhanced compared to free PTX. In the in vivo antitumor test, the SCM/PTX effectively inhibited the progression of 4T1 breast cancer in the orthotopic mouse model, as demonstrated by decreased tumor growth and increased apoptosis and necrosis areas within tumor tissues. In addition, the toxic side effects of PTX was also alleviated in the SCM/PTX group. This study introduced a stable micelle system that passive targeted the tumor for enhanced breast cancer therapy.
Collapse
Affiliation(s)
- Longyi Piao
- a Department of Oncology , the Jilin Central Hospital of Jilin University , Jilin , China
| | - Yongmeng Li
- a Department of Oncology , the Jilin Central Hospital of Jilin University , Jilin , China
| | - Hanwen Zhang
- a Department of Oncology , the Jilin Central Hospital of Jilin University , Jilin , China
| | - Jing Jiang
- a Department of Oncology , the Jilin Central Hospital of Jilin University , Jilin , China
| |
Collapse
|
93
|
Soni V, Pandey V, Asati S, Gour V, Tekade RK. Biodegradable Block Copolymers and Their Applications for Drug Delivery. BASIC FUNDAMENTALS OF DRUG DELIVERY 2019:401-447. [DOI: 10.1016/b978-0-12-817909-3.00011-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
94
|
Wu K, Liu J, lei L, Shen Y, Guo S. A stent film of paclitaxel presenting extreme accumulation of paclitaxel in tumor tissue and excellent antitumor efficacy after implantation beneath the subcutaneous tumor xenograft in mice. Int J Pharm 2018; 553:29-36. [DOI: 10.1016/j.ijpharm.2018.09.060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/12/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
|
95
|
Jiang D, Mu W, Pang X, Liu Y, Zhang N, Song Y, Garg S. Cascade Cytosol Delivery of Dual-Sensitive Micelle-Tailored Vaccine for Enhancing Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:37797-37811. [PMID: 30360105 DOI: 10.1021/acsami.8b09946] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Enhancing cytosol delivery of exogenous antigens in antigen presenting cells can improve cross-presentation and CD8+ T cell-mediated immune response. The antigen cytosol delivery speed, which has great importance on the rate of MHC class I molecules (MHC I) antigen presentation pathway and cytotoxic T lymphocytes (CTLs) induction, has not been well studied. We hypothesized that micelle-tailored vaccine with multiple cascaded lysosomal responsive capabilities could accelerate lysosomal escape and enhance cancer immunotherapy. To test our hypothesis, we created a novel micellar cancer vaccine (ovalbumin-loaded pH/redox dual-sensitive micellar vaccine, OLM-D) by cleavable conjugation of an antigen with house-made amphiphilic poly(l-histidine)-poly(ethylene glycol) (PLH-PEG) in current study. OLM-D was supposed to achieve cascade cytosol delivery of ovalbumin through three steps in terms of (i) initial redox triggered ovalbumin release, (ii) promoted proton inflow and micelle disassembly, and (iii) speeded proton sponge effect and lysosome bulging/broke. Redox-sensitive antigen release and consequently accelerative OLM-D disassembly were confirmed by sodium dodecylsulphate polyacrylamide gel electrophoresis (SDS-PAGE), transmission electronic microscopy (TEM), particle sizes, zeta potentials, and in vitro Ova release evaluation. The speeded cytosol delivery of ovalbumin was visualized under a confocal laser scanning microscope (CLSM). The ability of OLM-D to increase the MHC I molecule combination rate and antigen cross-presentation efficiency was identified by antigen presentation assay and maturation assay in bone marrow-derived dendritic cells (BMDCs). In vivo, the capability of OLM-D to accumulate in draining lymph nodes (LNs) after injection was visualized by real-time near infrared fluorescence imaging (NIRF) and the distribution order in different LNs was first observed (a, d, c, b). Enhanced cancer immunity of OLM-D was confirmed by increased CD3+CD8+ T cell quantity, CD3+CD8+25D11.6+ T cells quantity, and IFN-γ, IL-2 secretion post subcutaneous or intraperitoneal injection ( p < 0.05). Taken together, our results indicated that OLM-D provided a promising cascade cytosol delivery strategy, which held great potential to guide further design of nano-particulate cancer vaccines for efficient cancer immunotherapy.
Collapse
Affiliation(s)
- Dandan Jiang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 Wenhuaxi Road , Jinan , Shandong Province 250012 , China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 Wenhuaxi Road , Jinan , Shandong Province 250012 , China
| | - Xiuping Pang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 Wenhuaxi Road , Jinan , Shandong Province 250012 , China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 Wenhuaxi Road , Jinan , Shandong Province 250012 , China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 Wenhuaxi Road , Jinan , Shandong Province 250012 , China
| | - Yunmei Song
- School of Pharmacy and Medical Sciences , University of South Australia , Adelaide , South Australia 5001 , Australia
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences , University of South Australia , Adelaide , South Australia 5001 , Australia
| |
Collapse
|
96
|
Selting KA, Bechtel SM, Espinosa J, Henry CJ, Tate D, Bryan JN, Rajewski L, Flesner BK, Decedue C, Baltezor M. Evaluation of intravenous and subcutaneous administration of a novel, excipient-free, nanoparticulate formulation of paclitaxel in dogs with spontaneously occurring neoplasia. Vet Comp Oncol 2018; 16:650-657. [PMID: 30182530 DOI: 10.1111/vco.12435] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 11/29/2022]
Abstract
Carriers used to solubilize taxane chemotherapy drugs cause severe hypersensitivity. Nanoparticle formulations can provide improved dissolution and bioavailability of taxanes. Thus, a nanoparticulate, excipient-free formulation of paclitaxel (CTI52010) was evaluated in tumour-bearing dogs with intravenous and subcutaneous delivery. Tumour-bearing dogs were treated with intravenous CTI52010 using a modified rapid dose escalation scheme. Subcutaneous administration was then planned for a small cohort of dogs for comparison. For both groups, serial blood samples were collected after first dosing for pharmacokinetic analysis by LCMSMS. Tumour response was measured using RECIST criteria. Toxicity was recorded using VCOG-CTCAEv1.1. Fifteen dogs were treated with intravenous delivery at increasing dosages (80-136 mg/m2 ), with one objective response in the urethral component of a prostatic carcinoma (probable transitional cell carcinoma) and four dogs with durable stable disease (two carcinomas, two sarcomas). Pharmacokinetic data indicate a rapid initial clearing of the drug from serum followed by an extended elimination half-life, similar to normal dogs and suggesting reticuloendothelial clearance. Parameters and toxicity were highly variable and a maximally tolerated dosage could not be reliably confirmed. Three dogs were treated with subcutaneous delivery and no drug was detected in circulation, resulting in termination of the study. This novel formulation of paclitaxel is well tolerated in dogs and no unique toxicity or hypersensitivity was noted. The preliminary responses suggest biologic activity. The lack of systemic absorption after subcutaneous administration suggests a possible role for intratumoural anticancer therapy.
Collapse
Affiliation(s)
- Kim A Selting
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri
| | - Sandra M Bechtel
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri
| | | | - Carolyn J Henry
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri.,Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - Deborah Tate
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri
| | - Jeffrey N Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri
| | - Lian Rajewski
- Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas
| | - Brian K Flesner
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri
| | - Charles Decedue
- Crititech, Inc., Lawrence, Kansas.,Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas
| | - Michael Baltezor
- Crititech, Inc., Lawrence, Kansas.,Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas
| |
Collapse
|
97
|
Deshantri AK, Varela Moreira A, Ecker V, Mandhane SN, Schiffelers RM, Buchner M, Fens MHAM. Nanomedicines for the treatment of hematological malignancies. J Control Release 2018; 287:194-215. [PMID: 30165140 DOI: 10.1016/j.jconrel.2018.08.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022]
Abstract
Hematological malignancies (HM) are a collection of malignant transformations originating from cells in the primary or secondary lymphoid organs. Leukemia, lymphoma, and multiple myeloma comprise the three major types of HM. Current treatment consists of bone marrow transplantation, radiotherapy, immunotherapy and chemotherapy. Although, many chemotherapeutic drugs are clinically available for the treatment of HM, the use of these agents is limited due to dose-related toxicity and lack of specificity to tumor tissue. Moreover, the poor pharmacokinetic profile of most of the chemotherapeutics requires high dosage and frequent administration to maintain therapeutic levels at the target site, both increasing adverse effects. This underlines an urgent need for a suitable drug delivery system to improve efficacy, safety, and pharmacokinetic properties of conventional therapeutics. Nanomedicines have proven to enhance these properties for anticancer therapeutics. The most extensively studied nanomedicine systems are lipid-based nanoparticles and polymeric nanoparticles. Typically, nanomedicines are small sub-micron sized particles in the size range of 20-200 nm. The biocompatible and biodegradable nature of nanomedicines makes them attractive vehicles to improve drug delivery. Their small size allows them to extravasate and accumulate at malignant sites passively by means of the enhanced permeability and retention (EPR) effect, resulting from rapid angiogenesis and inflammation. Moreover, the specificity to the target tissue can be further enhanced by surface modification of nanoparticles. This review describes currently available therapies as well as limitations and potential advantages of nanomedicine formulations for treatment of various types of HM. Additionally, recent investigational and approved nanomedicine formulations and their limited applications in HM are discussed.
Collapse
Affiliation(s)
- Anil K Deshantri
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd, India
| | - Aida Varela Moreira
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Veronika Ecker
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sanjay N Mandhane
- Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd, India
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maike Buchner
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Marcel H A M Fens
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
98
|
Behroozi F, Abdkhodaie MJ, Abandansari HS, Satarian L, Molazem M, Al-Jamal KT, Baharvand H. Engineering folate-targeting diselenide-containing triblock copolymer as a redox-responsive shell-sheddable micelle for antitumor therapy in vivo. Acta Biomater 2018; 76:239-256. [PMID: 29928995 DOI: 10.1016/j.actbio.2018.05.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 11/20/2022]
Abstract
UNLABELLED The oxidation-reduction (redox)-responsive micelle system is based on a diselenide-containing triblock copolymer, poly(ε-caprolactone)-bis(diselenide-methoxy poly(ethylene glycol)/poly(ethylene glycol)-folate) [PCL-(SeSe-mPEG/PEG-FA)2]. This has helped in the development of tumor-targeted delivery for hydrophobic anticancer drugs. The diselenide bond, as a redox-sensitive linkage, was designed in such a manner that it is located at the hydrophilic-hydrophobic hinge to allow complete collapse of the micelle and thus efficient drug release in redox environments. The amphiphilic block copolymers self-assembled into micelles at concentrations higher than the critical micelle concentration (CMC) in an aqueous environment. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) analyses showed that the micelles were spherical with an average diameter of 120 nm. The insoluble anticancer drug paclitaxel (PTX) was loaded into micelles, and its triggered release behavior under different redox conditions was verified. Folate-targeting micelles showed an enhanced uptake in 4T1 breast cancer cells and in vitro cytotoxicity by flow cytometry and (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay, respectively. Delayed tumor growth was confirmed in the subcutaneously implanted 4T1 breast cancer in mice after intraperitoneal injection. The proposed redox-responsive copolymer offers a new type of biomaterial for drug delivery into cancer cells in vivo. STATEMENT OF SIGNIFICANCE On-demand drug actuation is highly desired. Redox-responsive polymeric DDSs have been shown to be able to respond and release their cargo in a selective manner when encountering a significant change in the potential difference, such as that present between cancerous and healthy tissues. This study offers an added advantage to the field of redox-responsive polymers by reporting a new type of shell-sheddable micelle based on an amphiphilic triblock co-polymer, containing diselenide as a redox-sensitive linkage. The linkage was smartly located at the hydrophilic-hydrophilic bridge in the co-polymer offering complete collapse of the micelle when exposed to the right trigger. The system was able to delay tumor growth and reduce toxicity in a breast cancer tumor model following intraperitoneal injection in mice.
Collapse
Affiliation(s)
- Farnaz Behroozi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohammad-Jafar Abdkhodaie
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran; Environmental Applied Science and Management, Ryerson University, Toronto, Canada.
| | - Hamid Sadeghi Abandansari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Leila Satarian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Molazem
- Department of Radiology and Surgery, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London, UK
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
99
|
Zhai J, Luwor RB, Ahmed N, Escalona R, Tan FH, Fong C, Ratcliffe J, Scoble JA, Drummond CJ, Tran N. Paclitaxel-Loaded Self-Assembled Lipid Nanoparticles as Targeted Drug Delivery Systems for the Treatment of Aggressive Ovarian Cancer. ACS APPLIED MATERIALS & INTERFACES 2018; 10:25174-25185. [PMID: 29963859 DOI: 10.1021/acsami.8b08125] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Chemotherapy using cytotoxic agents, such as paclitaxel (PTX), is one of the most effective treatments for advanced ovarian cancer. However, due to nonspecific targeting of the drug and the presence of toxic solvents required for dissolving PTX prior to injection, there are several serious side effects associated with this treatment. In this study, we explored self-assembled lipid-based nanoparticles as PTX carriers, which were able to improve its antitumour efficacy against ovarian cancer. The nanoparticles were also functionalized with epidermal growth factor receptor (EGFR) antibody fragments to explore the benefit of tumor active targeting. The formulated bicontinuous cubic- and sponge-phase nanoparticles, which were stabilized by Pluronic F127 and a lipid poly(ethylene glycol) stabilizer, showed a high capacity of PTX loading. These PTX-loaded nanoparticles also showed significantly higher cytotoxicity than a free drug formulation against HEY ovarian cancer cell lines in vitro. More importantly, the nanoparticle-based PTX treatments, with or without EGFR targeting, reduced the tumor burden by 50% compared to PTX or nondrug control in an ovarian cancer mouse xenograft model. In addition, the PTX-loaded nanoparticles were able to extend the survival of the treatment groups by up to 10 days compared to groups receiving free PTX or nondrug control. This proof-of-concept study has demonstrated the potential of these self-assembled lipid nanomaterials as effective drug delivery nanocarriers for poorly soluble chemotherapeutics, such as PTX.
Collapse
Affiliation(s)
- Jiali Zhai
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , VIC 3000 , Australia
| | - Rodney B Luwor
- Department of Surgery, Royal Melbourne Hospital , University of Melbourne , Melbourne , VIC 3052 , Australia
| | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute , Ballarat , VIC 3353 , Australia
- Federation University Australia , Ballarat , VIC 3010 , Australia
- The Hudson Institute of Medical Research , Clayton , VIC 3168 , Australia
- Department of Obstetrics and Gynaecology , University of Melbourne , Parkville , VIC 3052 , Australia
| | - Ruth Escalona
- Fiona Elsey Cancer Research Institute , Ballarat , VIC 3353 , Australia
- The Hudson Institute of Medical Research , Clayton , VIC 3168 , Australia
- Department of Obstetrics and Gynaecology , University of Melbourne , Parkville , VIC 3052 , Australia
| | - Fiona H Tan
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , VIC 3000 , Australia
- Department of Surgery, Royal Melbourne Hospital , University of Melbourne , Melbourne , VIC 3052 , Australia
| | - Celesta Fong
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , VIC 3000 , Australia
- CSIRO Manufacturing , Clayton , VIC 3168 , Australia
| | | | - Judith A Scoble
- CSIRO Manufacturing , 343 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Calum J Drummond
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , VIC 3000 , Australia
| | - Nhiem Tran
- School of Science, College of Science, Engineering and Health , RMIT University , Melbourne , VIC 3000 , Australia
| |
Collapse
|
100
|
Seah GL, Yu JH, Yang MY, Kim WJ, Kim JH, Park K, Cho JW, Kim JS, Nam YS. Low-power and low-drug-dose photodynamic chemotherapy via the breakdown of tumor-targeted micelles by reactive oxygen species. J Control Release 2018; 286:240-253. [PMID: 30071252 DOI: 10.1016/j.jconrel.2018.07.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/27/2018] [Accepted: 07/29/2018] [Indexed: 12/31/2022]
Abstract
Tumor-targeted delivery of anticancer agents using nanocarriers has been explored to increase the therapeutic index of cancer chemotherapy. However, only a few nanocarriers are clinically available because the physiological complexity often compromises their ability to target, penetrate, and control the release of drugs. Here, we report a method which dramatically increases in vivo therapeutic drug efficacy levels through the photodynamic degradation of tumor-targeted nanocarriers. Folate-decorated poly(ethylene glycol)-polythioketal micelles are prepared to encapsulate paclitaxel and porphyrins. Photo-excitation generates reactive oxygen species within the micelles to cleave the polythioketal backbone efficiently and facilitate drug release only at the illuminated tumor site. Intravenous injection of a murine xenograft model with a low dose of paclitaxel within the micelles, one-milligram drug per kg (mouse), corresponding to an amount less than that of Taxol by one order of magnitude, induces dramatic tumor regression without any acute systemic inflammation responses or organ toxicity under low-power irradiation (55 mW cm-2) at 650 nm.
Collapse
Affiliation(s)
- Geok Leng Seah
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong Heon Yu
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Moon Young Yang
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Woo Jin Kim
- Pathology Research Center, Department of Jeonbuk Inhalation Research, Korea Institute of Toxicology, 30 Baekhak-1-gil, Jeongup, Jeonbuk 56212, Republic of Korea
| | - Jin-Ho Kim
- Samsung Medical Center, Samsung Biomedical Research Institute, Irwon-dong, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Keunchil Park
- Samsung Medical Center, Samsung Biomedical Research Institute, Irwon-dong, Gangnam-gu, Seoul 06351, Republic of Korea; Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-dong, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Jae-Woo Cho
- Pathology Research Center, Department of Jeonbuk Inhalation Research, Korea Institute of Toxicology, 30 Baekhak-1-gil, Jeongup, Jeonbuk 56212, Republic of Korea
| | - Jee Seon Kim
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Yoon Sung Nam
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; KAIST Institute for the NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|