1
|
Zhu M, Xie Y, Li Z, Bao H, Miao D, Guo X, Wang S, Chen K, Chen H, Dai J, Yang N, Yu L, Pei J. Antitumour and anti-angiogenesis efficacy of a multifunctional self-oxygenated active-targeting drug delivery system by encapsulating biological and chemotherapeutic drugs. Colloids Surf B Biointerfaces 2025; 250:114549. [PMID: 39965481 DOI: 10.1016/j.colsurfb.2025.114549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
The hypoxic tumour microenvironment (TME), resulting from abnormal tumour angiogenesis, is a major factor contributing to treatment failure in breast cancer patients. In this study, we present a ZnO2-based oestrone-conjugated PEGylated liposome (ZnO2@EPL-CDDP/EGCG) that incorporates cisplatin (CDDP) and epigallocatechin-3-gallate (EGCG). ZnO2 remains stable in neutral environments but decomposes under mildly acidic conditions, releasing Zn²⁺ and H₂O₂. These byproducts inhibit the electron transport chain, stimulate the endogenous reactive oxygen species production for chemodynamic therapy (CDT), and generate oxygen at tumour sites to alleviate hypoxia and enhance anti-angiogenic efficacy. EGCG inhibits tumour angiogenesis by down-regulating hypoxia-inducible factor-1α (HIF-1α) and its downstream pathways, while also exhibiting synergistic anti-tumour effects with CDDP. Oestrone-conjugated and polyethylene glycol (PEG) modifications facilitate targeted accumulation at tumour sites. Our findings indicate that ZnO2@EPL-CDDP/EGCG significantly improves the therapeutic efficacy of both EGCG and CDDP, remodels tumour vasculature, and alleviates hypoxia within the TME. This self-oxygenated, actively targeted drug delivery system notably extends the survival of healthy ICR mice without observed toxicity. This novel approach, which co-encapsulates ZnO2, EGCG, and CDDP in an active-targeting liposomal formulation for the first time, represents a promising strategy for effective cancer treatment.
Collapse
Affiliation(s)
- Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Zhiping Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Shanshan Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Kejia Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Hongzhu Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Jingwen Dai
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Liangping Yu
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China.
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China.
| |
Collapse
|
2
|
Edvall C, Kale N, Tani S, Ambhore S, Hossain R, Ozoude C, Van Horsen K, Mohammad J, Tuvin DM, Kalathingal S, Loganathan J, Choi Y, Sathish V, Brown J, Mallik S. Hypoxia-Responsive Polymersomes for Stemness Reduction in Patient-Derived Solid Tumor Spheroids. ACS APPLIED BIO MATERIALS 2025; 8:2916-2926. [PMID: 40056142 DOI: 10.1021/acsabm.4c01735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
Abstract
Aggressive solid tumors are associated with rapid growth, early hypoxia, a lack of targeted therapies, and a poor prognosis. The hypoxic niches within the rapidly growing solid tumors give rise to a stem-cell-like phenotype with higher metastasis and drug resistance. To overcome the drug resistance of these regions, we used hypoxia-responsive polymersomes with an encapsulated anticancer drug (doxorubicin, Dox) and a stemness modulator (all-trans retinoic acid, ATRA). Reductase enzymes overexpressed in hypoxia reduce the azobenzene linker of the polymers, disrupt the bilayer structure of the polymersomes, and release the encapsulated drugs. We used triple-negative breast cancer (TNBC) as a representative of aggressive and hypoxic solid tumors. We observed that ATRA synergistically enhanced the efficacy of Dox in killing cancer cells. A synergistic combination of the two drug-encapsulated polymersomes reduced the volumes of patient-derived TNBC spheroids by 90%. In contrast, Dox alone decreased the spheroid volumes by 70% and encapsulated ATRA by 19%. Mechanistic studies revealed that ATRA inhibited efflux pumps, leading to a higher concentration of doxorubicin within TNBC cells. In addition, the combination of encapsulated Dox and ATRA significantly decreased stemness expression of the TNBC cells in hypoxia compared to that of Dox alone.
Collapse
Affiliation(s)
- Connor Edvall
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Narendra Kale
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Sakurako Tani
- Department of Physics, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Shubhashri Ambhore
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Rayat Hossain
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Chukwuebuka Ozoude
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Karl Van Horsen
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jiyan Mohammad
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Daniel M Tuvin
- Sanford Broadway Clinic,801 Broadway N, Fargo, North Dakota 58102, United States
| | - Santo Kalathingal
- Agathos Biologics,4837 Amber Valley Pkwy Suite 12, Fargo, North Dakota 58104, United States
| | - Jagadish Loganathan
- Agathos Biologics,4837 Amber Valley Pkwy Suite 12, Fargo, North Dakota 58104, United States
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - James Brown
- Agathos Biologics,4837 Amber Valley Pkwy Suite 12, Fargo, North Dakota 58104, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota 57007, United States
| |
Collapse
|
3
|
Xu W, Jia A, Lei Z, Wang J, Jiang H, Wang S, Wang Q. Stimuli-responsive prodrugs with self-immolative linker for improved cancer therapy. Eur J Med Chem 2024; 279:116928. [PMID: 39362023 DOI: 10.1016/j.ejmech.2024.116928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
Self-immolative prodrugs have gained significant attention as an innovative approach for targeted cancer therapy. These prodrugs are engineered to release the active anticancer agents in response to specific triggers within the tumor microenvironment, thereby improving therapeutic precision while reducing systemic toxicity. This review focuses on the molecular architecture and design principles of self-immolative prodrugs, emphasizing the role of stimuli-responsive linkers and activation mechanisms that enable controlled drug release. Recent advancements in this field include the development of prodrugs that incorporate targeting moieties for enhanced site-specificity. Moreover, the review discusses the incorporation of targeting moieties to achieve site-specific drug delivery, thereby improving the selectivity of treatment. By summarizing key research from the past five years, this review highlights the potential of self-immolative prodrugs to revolutionize cancer treatment strategies and pave the way for their integration into clinical practice.
Collapse
Affiliation(s)
- Wenting Xu
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhixian Lei
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China
| | - Jianing Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Hongfei Jiang
- School of Pharmacy, Qingdao University, Qingdao, 266071, China.
| | - Shuai Wang
- Department of Radiotherapy, School of Medical Imaging, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, Shandong, China.
| | - Qi Wang
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China.
| |
Collapse
|
4
|
Khan AH, Basak A, Zaman A, Das PK. Inherently targeted estradiol-derived carbon dots for selective killing of ER (+) breast cancer cells via oridonin-triggered p53 pathway activation. J Mater Chem B 2024; 12:11708-11720. [PMID: 39435655 DOI: 10.1039/d4tb01415d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
One of the most prevalent cancers globally is breast cancer and approximately two thirds of the breast cancers are hormone receptor positive with estrogen receptors (ER) being a prominent target. Notably, p53 that controls several cellular functions and prevents tumor formation, gets suppressed in breast cancers. Reactivation of p53 can lead to cell cycle arrest as well as apoptosis. Therefore, targeting the estrogen receptor for selective delivery of anticancer drugs that can reactivate p53 in ER (+) breast cancers can be a crucial method in breast cancer therapy. Herein, we have designed and developed estradiol-derived inherently targeted specific carbon dots (E2-CA-CD) from 17β-estradiol and citric acid following a solvothermal method. The synthesized carbon dots were characterized using spectroscopic and microscopic techniques. The water soluble, intrinsically fluorescent E2-CA-CD showed excellent biocompatibility in MCF-7, MDA-MB-231 as well as NIH3T3 cells and demonstrated target specific bioimaging in ER (+) MCF-7 cells due to the overexpressed ER receptors. Furthermore, oridonin, a well-known hydrophobic anticancer drug capable of upregulating the p53 pathway, was loaded on the carbon dots to increase its bioavailability. E2-CA-CD-Ori caused ∼2.2 times higher killing in ER (+) MCF-7 cells compared to ER (-) MDA-MB-231 cells and normal cells NIH3T3. Also, E2-CA-CD-Ori showed ∼3 fold better killing in MCF-7 cells compared to native oridonin. E2-CA-CD-Ori-induced killing of MCF-7 cells took place through the early to late apoptotic pathway along with the elevation of the intracellular ROS level. Importantly, E2-CA-CD-Ori triggered the activation of the p53 pathway in MCF-7 cells, which in turn induced apoptosis involving the upregulation of Bax and downregulation of Bcl-2 leading to the selective and efficient killing of ER (+) MCF-7 cells.
Collapse
Affiliation(s)
- Aftab Hossain Khan
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Ambalika Basak
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Afreen Zaman
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| |
Collapse
|
5
|
de Assis Ramos MM, Ricardo-da-Silva FY, Macedo LDO, Correia CJ, Moreira LFP, Löbenberg R, Breithaupt-Faloppa AC, Bou-Chacra N. A review on lipid and polymeric nano-based 17-β-estradiol delivery systems: advances and challenges. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13633. [PMID: 39619127 PMCID: PMC11604423 DOI: 10.3389/jpps.2024.13633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
17β-estradiol (E2) is an endogenous steroid hormone pivotal for the development of female secondary sexual characteristics and the maintenance of the female reproductive system. Its roles extend beyond these physiological functions, as E2 is employed in hormone replacement therapy to alleviate symptoms associated with menopause. Furthermore, E2 exhibits therapeutic potential in the management of osteoporosis, breast cancer, and various neurological and cardiovascular conditions, partly due to its anti-inflammatory effects via modulation of the MAPK/NFκB signaling pathway. Notwithstanding, the hydrophobic nature of E2 significantly hinders the formulation of efficacious delivery systems for its clinical deployment. Recent advances have highlighted nano-based delivery systems for E2 as a promising solution to this solubility challenge. This review critically examines contemporary nano-delivery strategies for E2, particularly emphasizing lipid and polymeric nanoparticle-based systems. These nanostructures are designed to enhance stability, biocompatibility, controlled release, and targeted delivery of E2, yet the selectivity of E2 delivery for therapeutic purposes remains an ongoing challenge. The novelty of this review lies in its focus on the advances in nano-based E2 delivery systems over the past decade, a topic not extensively covered in prior literature. We present a comprehensive analysis of the encapsulation of E2 within polymeric and lipid nanoparticles, underscoring the untapped potential of these strategies. This review identifies a significant research gap, advocating for intensified experimental investigations that could pave the way for the translation of nano-based E2 therapies from bench to bedside.
Collapse
Affiliation(s)
- Mayara Munhoz de Assis Ramos
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Yamamoto Ricardo-da-Silva
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiza de Oliveira Macedo
- Departamento de Farmácia, Faculdade de Ciências Farmacêuticas da Universidade de São Paulo, São Paulo, Brazil
| | - Cristiano Jesus Correia
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Felipe Pinho Moreira
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raimar Löbenberg
- Division of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Nadia Bou-Chacra
- Departamento de Farmácia, Faculdade de Ciências Farmacêuticas da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Guan C, Han Y, Ling Z, Meng X, Zhang B, Dong W, Zhang D, Chen K. Nanomaterials: breaking the bottleneck of breast cancer drug resistance. Front Immunol 2024; 15:1492546. [PMID: 39606228 PMCID: PMC11599193 DOI: 10.3389/fimmu.2024.1492546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Drug resistance poses a significant challenge in the treatment of breast cancer. In recent years, a variety of nanomaterials have been discovered and synthesized that can selectively target tumor cells and play a crucial role in the advancement of breast cancer therapies. As our understanding of tumor heterogeneity deepens, the emerging potential of nanomaterials in addressing drug resistance has garnered considerable attention. These materials not only selectively target tumor cells but also possess unique properties that make them promising options for cancer treatment, including low toxicity, excellent biocompatibility, ease of preparation, the ability to carry antitumor drugs, and customizable surface functions. In this review, we will comprehensively summarize two key developments in breast cancer treatment: the application of antitumor drugs and nanomaterials. We will explore the mechanisms by which nanomaterials improve drug resistance in breast cancer, targeted nanotherapy strategies to mitigate this resistance, and recent research advancements in anticancer nanomaterials. This overview aims to highlight the significant role of nanomaterials in breast cancer treatment and provide a theoretical framework for identifying optimal treatment strategies in the future.
Collapse
Affiliation(s)
- Chao Guan
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Yahao Han
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| | - Zhenzheng Ling
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Xiang Meng
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Baolin Zhang
- The Fourth Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Wanwei Dong
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| | - Di Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Keyan Chen
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Ciepła J, Smolarczyk R. Tumor hypoxia unveiled: insights into microenvironment, detection tools and emerging therapies. Clin Exp Med 2024; 24:235. [PMID: 39361163 PMCID: PMC11449960 DOI: 10.1007/s10238-024-01501-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Hypoxia is one of the defining characteristics of the tumor microenvironment (TME) in solid cancers. It has a major impact on the growth and spread of malignant cells as well as their resistance to common treatments like radiation and chemotherapy. Here, we explore the complex functions of hypoxia in the TME and investigate its effects on angiogenesis, immunological evasion, and cancer cell metabolism. For prognostic and therapeutic reasons, hypoxia identification is critical, and recent developments in imaging and molecular methods have enhanced our capacity to precisely locate underoxygenated areas inside tumors. Furthermore, targeted therapies that take advantage of hypoxia provide a potential new direction in the treatment of cancer. Therapeutic approaches that specifically target hypoxic conditions in tumors without causing adverse effects are being led by hypoxia-targeted nanocarriers and hypoxia-activated prodrugs (HAPs). This review provides an extensive overview of this dynamic and clinically significant area of oncology research by synthesizing current knowledge about the mechanisms of hypoxia in cancer, highlighting state-of-the-art detection methodologies, and assessing the potential and efficacy of hypoxia-targeted therapies.
Collapse
Affiliation(s)
- Joanna Ciepła
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| |
Collapse
|
8
|
Mamnoon B, Moses AS, Sundaram S, Raitmayr CJ, Morgan T, Baldwin MK, Myatt L, Taratula O, Taratula OR. Glutathione-Responsive Methotrexate Polymersomes for Potential Management of Ectopic Pregnancy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302969. [PMID: 37452511 PMCID: PMC10787806 DOI: 10.1002/smll.202302969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/22/2023] [Indexed: 07/18/2023]
Abstract
The first-line treatment for ectopic pregnancy (EP), the chemotherapeutic methotrexate (MTX), has a failure rate of more than 10%, which can lead to severe complications or death. Inadequate accumulation of administered MTX at the ectopic implantation site significantly contributes to therapeutic failure. This study reports the first glutathione-responsive polymersomes for efficient delivery of MTX to the implantation site and its triggered release in placental cells. Fluorescence and photoacoustic imaging have confirmed that the developed polymersomes preferentially accumulate after systemic administration in the implantation site of pregnant mice at early gestational stages. The high concentrations of intracellular glutathione (GSH) reduce an incorporated disulfide bond within polymersomes upon internalization into placental cells, resulting in their disintegration and efficient drug release. Consequently, MTX delivered by polymersomes induces pregnancy demise in mice, as opposed to free MTX at the same dose regimen. To achieve the same therapeutic efficacy with free MTX, a sixfold increase in dosage is required. In addition, mice successfully conceive and birth healthy pups following a prior complete pregnancy demise induced by methotrexate polymersomes. Therefore, the developed MTX nanomedicine can potentially improve EP management and reduce associated mortality rates and related cost.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Subisha Sundaram
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Constanze J Raitmayr
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Terry Morgan
- Department of Pathology and Laboratory Medicine, and the Center for Developmental Health, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Maureen K Baldwin
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| |
Collapse
|
9
|
Negut I, Bita B. Polymersomes as Innovative, Stimuli-Responsive Platforms for Cancer Therapy. Pharmaceutics 2024; 16:463. [PMID: 38675124 PMCID: PMC11053450 DOI: 10.3390/pharmaceutics16040463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This review addresses the urgent need for more targeted and less toxic cancer treatments by exploring the potential of multi-responsive polymersomes. These advanced nanocarriers are engineered to deliver drugs precisely to tumor sites by responding to specific stimuli such as pH, temperature, light, hypoxia, and redox conditions, thereby minimizing the side effects associated with traditional chemotherapy. We discuss the design, synthesis, and recent applications of polymersomes, emphasizing their ability to improve therapeutic outcomes through controlled drug release and targeted delivery. Moreover, we highlight the critical areas for future research, including the optimization of polymersome-biological interactions and biocompatibility, to facilitate their clinical adoption. Multi-responsive polymersomes emerge as a promising development in nanomedicine, offering a pathway to safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Irina Negut
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
| | - Bogdan Bita
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
- National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania
| |
Collapse
|
10
|
Ding L, Agrawal P, Singh SK, Chhonker YS, Sun J, Murry DJ. Polymer-Based Drug Delivery Systems for Cancer Therapeutics. Polymers (Basel) 2024; 16:843. [PMID: 38543448 PMCID: PMC10974363 DOI: 10.3390/polym16060843] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 11/12/2024] Open
Abstract
Chemotherapy together with surgery and/or radiotherapy are the most common therapeutic methods for treating cancer. However, the off-target effects of chemotherapy are known to produce side effects and dose-limiting toxicities. Novel delivery platforms based on natural and synthetic polymers with enhanced pharmacokinetic and therapeutic potential for the treatment of cancer have grown tremendously over the past 10 years. Polymers can facilitate selective targeting, enhance and prolong circulation, improve delivery, and provide the controlled release of cargos through various mechanisms, including physical adsorption, chemical conjugation, and/or internal loading. Notably, polymers that are biodegradable, biocompatible, and physicochemically stable are considered to be ideal delivery carriers. This biomimetic and bio-inspired system offers a bright future for effective drug delivery with the potential to overcome the obstacles encountered. This review focuses on the barriers that impact the success of chemotherapy drug delivery as well as the recent developments based on natural and synthetic polymers as platforms for improving drug delivery for treating cancer.
Collapse
Affiliation(s)
- Ling Ding
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Prachi Agrawal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (P.A.); (J.S.)
| | - Sandeep K. Singh
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Yashpal S. Chhonker
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Jingjing Sun
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (P.A.); (J.S.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daryl J. Murry
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
11
|
Mehata AK, Singh V, Vikas, Srivastava P, Koch B, Kumar M, Muthu MS. Chitosan nanoplatform for the co-delivery of palbociclib and ultra-small magnesium nanoclusters: dual receptor targeting, therapy and imaging. Nanotheranostics 2024; 8:179-201. [PMID: 38444739 PMCID: PMC10911970 DOI: 10.7150/ntno.94364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/26/2024] [Indexed: 03/07/2024] Open
Abstract
Theranostic nanoparticles have gained significant attention in cancer diagnosis and therapy. In this study, estrone (ES) and folic acid (FA) functionalized single and dual receptor targeted theranostic chitosan nanoparticles were developed for breast cancer imaging and therapy. These nanoparticles (NPs) were loaded with palbociclib (PB) and ultra-small magnesium nanoclusters (UMN). The developed nontargeted theranostic NPs (PB-UMN-CS-NPs), estrogen receptor targeted theranostic NPs (PB-UMN-CS-ES-NPs), folate receptor targeted theranostic NPs (PB-UMN-CS-FA-NPs), and dual targeted theranostic NPs (PB-UMN-CS-ES-FA-NPs) have particle sizes of 178.4 ± 1.21 nm, 181.6± 1.35 nm, 185.1± 1.33 nm, and 198.2± 1.43 nm with surface charges of +19.02± 0.382 mV, +13.89±0.410 mV, +16.72±0.527 mV and +15.23±0.377 mV, respectively. Cytotoxicity studies on estrogen receptor (ER) and folate receptor (FR) expressing breast cancer cells revealed that dual-targeted theranostic NPs (PB-UMN-CS-FA-ES-NPs) were more effective, inhibiting cell growth by 54.17 and 42.23 times in MCF-7 and T-47D cells compared to free PB, respectively. Additionally, developed NPs were capable of inhibiting the cell cycle progression of MCF-7 cells from the G1 phase to the S phase more efficiently compared to free PB. Ultrasound and photoacoustic (USG/PA) imaging demonstrated that dual targeted theranostic NPs were capable of effectively reducing hypoxic tumor volume and significantly suppressing tumor vascularity compared to free PB, nontargeted, FR targeted and ER targeted NPs. Moreover, in vivo optical imaging demonstrated tumor specific accumulation of the dual-targeted theranostic NPs. Furthermore, in vitro hemocompatibility and histopathological studies confirmed the biocompatibility of developed nanoformulations.
Collapse
Affiliation(s)
- Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, IIT (Banaras Hindu University), Varanasi-221005, UP, India
| | - Virendra Singh
- Cancer Biology Laboratory, Department of Zoology Institute of Science, (Banaras Hindu University), Varanasi-221005, UP, India
| | - Vikas
- Department of Pharmaceutical Engineering and Technology, IIT (Banaras Hindu University), Varanasi-221005, UP, India
| | - Prachi Srivastava
- Nano2Micro Material Design Lab, Chemical Engineering and Technology, IIT BHU, Varanasi-221005, UP, India
| | - Biplob Koch
- Cancer Biology Laboratory, Department of Zoology Institute of Science, (Banaras Hindu University), Varanasi-221005, UP, India
| | - Manoj Kumar
- Nano2Micro Material Design Lab, Chemical Engineering and Technology, IIT BHU, Varanasi-221005, UP, India
| | - Madaswamy S. Muthu
- Department of Pharmaceutical Engineering and Technology, IIT (Banaras Hindu University), Varanasi-221005, UP, India
| |
Collapse
|
12
|
Kayani A, Raza A, Si J, Dutta D, Zhou Q, Ge Z. Polymersome Membrane Engineering with Active Targeting or Controlled Permeability for Responsive Drug Delivery. Biomacromolecules 2023; 24:4622-4645. [PMID: 37870458 DOI: 10.1021/acs.biomac.3c00839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Polymersomes have been extensively investigated for drug delivery as nanocarriers for two decades due to a series of advantages including high stability under physiological conditions, simultaneous encapsulation of hydrophilic and hydrophobic drugs inside inner cavities and membranes, respectively, and facile adjustment of membrane and surface properties, as well as controlled drug release through incorporation of stimuli-responsive components. Despite these features, polymersome nanocarriers frequently suffer from nontargeting delivery and poor membrane permeability. In recent years, polymersomes have been functionalized for more efficient drug delivery. The surface shells were explored to be modified with diverse active targeting groups to improve disease-targeting delivery. The membrane permeability of the polymersomes was adjusted by incorporation of the stimuli-responsive components for smart controlled transportation of the encapsulated drugs. Therefore, being the polymersome-biointerface, tailorable properties can be introduced by its carefully modulated engineering. This review elaborates on the role of polymersome membranes as a platform to incorporate versatile features. First, we discuss how surface functionalization facilitates the directional journey to the targeting sites toward specific diseases, cells, or intracellular organelles via active targeting. Moreover, recent advances in the past decade related to membrane permeability to control drug release are also summarized. We finally discuss future development to promote polymersomes as in vivo drug delivery nanocarriers.
Collapse
Affiliation(s)
- Anum Kayani
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Arsalan Raza
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jiale Si
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Debabrata Dutta
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Qinghao Zhou
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
13
|
Fang LR, Wang YH, Xiong ZZ, Wang YM. Research progress of nanomaterials in tumor-targeted drug delivery and imaging therapy. OPENNANO 2023; 14:100184. [DOI: 10.1016/j.onano.2023.100184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Duan H, Wang L, Wang S, He Y. Surface modification potentials of cell membrane-based materials for targeted therapies: a chemotherapy-focused review. Nanomedicine (Lond) 2023; 18:1281-1303. [PMID: 37753724 DOI: 10.2217/nnm-2023-0164] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023] Open
Abstract
Nanotechnology has significant potential for cancer management at all stages, including prevention, diagnosis and treatment. In therapeutic applications, nanoparticles (NPs) have biological stability, targeting and body-clearance issues. To overcome these difficulties, biomimetic or cell membrane-coating methods using immune cell membranes are advised. Macrophage or neutrophil cell membrane-coated NPs may impede cancer progression in malignant tissue. Immune cell surface proteins and their capacity to maintain activity after membrane extraction and NP coating determine NP functioning. Immune cell surface proteins may offer NPs higher cellular interactions, blood circulation, antigen recognition for targeting, progressive drug release and reduced in vivo toxicity. This article examines nano-based systems with immune cell membranes, their surface modification potential, and their application in cancer treatment.
Collapse
Affiliation(s)
- Hongliang Duan
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun, 130000, China
| | - LiJuan Wang
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, 130000, China
| | - Sen Wang
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun, 130000, China
| | - Yangfang He
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, 130000, China
| |
Collapse
|
15
|
Mehata AK, Singh V, Singh N, Mandal A, Dash D, Koch B, Muthu MS. Chitosan- g-estrone Nanoparticles of Palbociclib Vanished Hypoxic Breast Tumor after Targeted Delivery: Development and Ultrasound/Photoacoustic Imaging. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37433149 DOI: 10.1021/acsami.3c03184] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Breast cancer is the leading cause of death among women globally. Approximately 80% of all breast cancers diagnosed are overexpressed with estrogen receptors (ERs). In this study, we have developed an estrone (Egen)-grafted chitosan-based polymeric nanocarrier for the targeted delivery of palbociclib (PLB) to breast cancer. The nanoparticles (NPs) were prepared by solvent evaporation using the ionic gelation method and characterized for particle size, zeta potential, polydispersity, surface morphology, surface chemistry, drug entrapment efficiency, cytotoxicity assay, cellular uptake, and apoptosis study. The developed PLB-CS NPs and PLB-CS-g-Egen NPs had a particle size of 116.3 ± 1.53 nm and 141.6 ± 1.97 nm, respectively. The zeta potential of PLB-CS NPs and PLB-CS-g-Egen NPs was found to be 18.70 ± 0.416 mV and 12.45 ± 0.574 mV, respectively. The morphological analysis demonstrated that all NPs were spherical in shape and had a smooth surface. An in vitro cytotoxicity assay was performed in estrogen receptor (ER)-expressing MCF7 cells and T47D cells, which suggested that targeted NPs were 57.34- and 30.32-fold more cytotoxic compared to the pure PLB, respectively. Additionally, cell cycle analysis confirmed that cell cycle progression from the G1 into S phase was blocked more efficiently by targeted NPs compared to nontargeted NPs and PLB in MCF7 cells. In vivo pharmacokinetic studies demonstrated that entrapment of the PLB in the NPs improved the half-life and bioavailability by ∼2-3-fold. Further, ultrasound and photoacoustic imaging of DMBA induced breast cancer in the Sprague-Dawley (SD) rat showed that targeted NPs completely vanished breast tumor, reduced hypoxic tumor volume, and suppressed tumor angiogenesis more efficiently compared to the nontargeted NPs and free PLB. Further, in vitro hemocompatibility and histopathology studies suggested that NPs were biocompatible and safe for clinical use.
Collapse
Affiliation(s)
- Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Virendra Singh
- Cancer Biology Laboratory, Department of Zoology Institute of Science, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Nitesh Singh
- Department of Biochemistry, Institute of Medical Sciences, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Abhijit Mandal
- Department of Radiotherapy and Radiation Medicine, Institute of Medical Sciences, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Biplob Koch
- Cancer Biology Laboratory, Department of Zoology Institute of Science, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (BHU), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
16
|
Zhang Y, Pan H, Yu C, Liu R, Xing B, Jia B, He J, Jia X, Feng X, Zhang Q, Dang W, Hu Z, Deng X, Guo P, Liu Z, Pan W. Phytoestrogen-derived multifunctional ligands for targeted therapy of breast cancer. Asian J Pharm Sci 2023; 18:100827. [PMID: 37588993 PMCID: PMC10425851 DOI: 10.1016/j.ajps.2023.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/20/2023] [Accepted: 05/31/2023] [Indexed: 08/18/2023] Open
Abstract
Nano-targeted delivery systems have been widely used for breast tumor drug delivery. Estrogen receptors are considered to be significant drug delivery target receptors due to their overexpression in a variety of tumor cells. However, targeted ligands have a significant impact on the safety and effectiveness of active delivery systems, limiting the clinical transformation of nanoparticles. Phytoestrogens have shown good biosafety characteristics and some affinity with the estrogen receptor. In the present study, molecular docking was used to select tanshinone IIA (Tan IIA) among phytoestrogens as a target ligand to be used in nanodelivery systems with some modifications. Modified Tan IIA (Tan-NH2) showed a good biosafety profile and demonstrated tumor-targeting, anti-tumor and anti-tumor metastasis effects. Moreover, the ligand was utilized with the anti-tumor drug Dox-loaded mesoporous silica nanoparticles via chemical modification to generate a nanocomposite Tan-Dox-MSN. Tan-Dox-MSN had a uniform particle size, good dispersibility and high drug loading capacity. Validation experiments in vivo and in vitro showed that it also had a better targeting ability, anti-tumor effect and lower toxicity in normal organs. These results supported the idea that phytoestrogens with high affinity for the estrogen receptor could improve the therapeutic efficacy of nano-targeted delivery systems in breast tumors.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hao Pan
- School of Pharmacy, Liaoning University, Shenyang 110036, China
| | - Changxiang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bin Xing
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bei Jia
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiachen He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xintao Jia
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaojiao Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qingqing Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wenli Dang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zheming Hu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiuping Deng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pan Guo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhidong Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Weisan Pan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
17
|
Hughes KA, Misra B, Maghareh M, Bobbala S. Use of stimulatory responsive soft nanoparticles for intracellular drug delivery. NANO RESEARCH 2023; 16:6974-6990. [PMID: 36685637 PMCID: PMC9840428 DOI: 10.1007/s12274-022-5267-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 05/24/2023]
Abstract
Drug delivery has made tremendous advances in the last decade. Targeted therapies are increasingly common, with intracellular delivery highly impactful and sought after. Intracellular drug delivery systems have limitations due to imprecise and non-targeted release profiles. One way this can be addressed is through using stimuli-responsive soft nanoparticles, which contain materials with an organic backbone such as lipids and polymers. The choice of biomaterial is essential for soft nanoparticles to be responsive to internal or external stimuli. The nanoparticle must retain its integrity and payload in non-targeted physiological conditions while responding to particular intracellular environments where payload release is desired. Multiple internal and external factors could stimulate the intracellular release of drugs from nanoparticles. Internal stimuli include pH, oxidation, and enzymes, while external stimuli include ultrasound, light, electricity, and magnetic fields. Stimulatory responsive soft nanoparticulate systems specifically utilized to modulate intracellular delivery of drugs are explored in this review.
Collapse
Affiliation(s)
- Krystal A. Hughes
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Maryam Maghareh
- Department of Clinical Pharmacy, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| |
Collapse
|
18
|
Hamdy NM, Eskander G, Basalious EB. Insights on the Dynamic Innovative Tumor Targeted-Nanoparticles-Based Drug Delivery Systems Activation Techniques. Int J Nanomedicine 2022; 17:6131-6155. [PMID: 36514378 PMCID: PMC9741821 DOI: 10.2147/ijn.s386037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer conventional chemotherapeutic drugs novel formula progress, nowadays, uses nano technology for targeted drug delivery, specifically tailored to overcome therapeutic agents' delivery challenges. Polymer drug delivery systems (DDS) play a crucial role in minimizing off-target side effects arising when using standard cytotoxic drugs. Using nano-formula for targeted localized action, permits using larger effective cytotoxic doses on a single special spot, that can seriously cause harm if it was administered systemically. Therefore, various nanoparticles (NPs) specifically have attached groups for targeting capabilities, not seen in bulk materials, which then need activation. In this review, we will present a simple innovative, illustrative, in a cartoon-way, enumeration of NP anti-cancer drug targeting delivery system activation-types. Area(s) covered in this review are the mechanisms of various NP activation techniques.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Georgette Eskander
- Faculty of Pharmacy, Ain Shams University, Postgraduate Student, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
19
|
Bergandi L, Lucia U, Grisolia G, Salaroglio IC, Gesmundo I, Granata R, Borchiellini R, Ponzetto A, Silvagno F. Thermomagnetic Resonance Effect of the Extremely Low Frequency Electromagnetic Field on Three-Dimensional Cancer Models. Int J Mol Sci 2022; 23:ijms23147955. [PMID: 35887313 PMCID: PMC9318636 DOI: 10.3390/ijms23147955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
In our recent studies, we have developed a thermodynamic biochemical model able to select the resonant frequency of an extremely low frequency electromagnetic field (ELF-EMF) specifically affecting different types of cancer, and we have demonstrated its effects in vitro. In this work, we investigate the cellular response to the ELF electromagnetic wave in three-dimensional (3D) culture models, which mimic the features of tumors in vivo. Cell membrane was modelled as a resistor–capacitor circuit and the specific thermal resonant frequency was calculated and tested on two-dimensional (2D) and three-dimensional (3D) cell cultures of human pancreatic cancer, glioblastoma and breast cancer. Cell proliferation and the transcription of respiratory chain and adenosine triphosphate synthase subunits, as well as uncoupling proteins, were assessed. For the first time, we demonstrate that an ELF-EMF hampers growth and potentiates both the coupled and uncoupled respiration of all analyzed models. Interestingly, the metabolic shift was evident even in the 3D aggregates, making this approach particularly valuable and promising for future application in vivo, in aggressive cancer tissues characterized by resistance to treatments.
Collapse
Affiliation(s)
- Loredana Bergandi
- Department of Oncology, University of Torino, Via Santena 5 bis, 10126 Torino, Italy; (L.B.); (I.C.S.)
| | - Umberto Lucia
- Department of Energy, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (U.L.); (G.G.); (R.B.)
| | - Giulia Grisolia
- Department of Energy, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (U.L.); (G.G.); (R.B.)
| | - Iris Chiara Salaroglio
- Department of Oncology, University of Torino, Via Santena 5 bis, 10126 Torino, Italy; (L.B.); (I.C.S.)
| | - Iacopo Gesmundo
- Department of Medical Sciences, University of Torino, Corso A.M. Dogliotti 14, 10126 Torino, Italy; (I.G.); (R.G.); (A.P.)
| | - Riccarda Granata
- Department of Medical Sciences, University of Torino, Corso A.M. Dogliotti 14, 10126 Torino, Italy; (I.G.); (R.G.); (A.P.)
| | - Romano Borchiellini
- Department of Energy, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (U.L.); (G.G.); (R.B.)
| | - Antonio Ponzetto
- Department of Medical Sciences, University of Torino, Corso A.M. Dogliotti 14, 10126 Torino, Italy; (I.G.); (R.G.); (A.P.)
| | - Francesca Silvagno
- Department of Oncology, University of Torino, Via Santena 5 bis, 10126 Torino, Italy; (L.B.); (I.C.S.)
- Correspondence:
| |
Collapse
|
20
|
Hariharan K, Patel P, Mehta T. Surface modifications of Gold Nanoparticles: Stabilization and Recent Applications in Cancer Therapy. Pharm Dev Technol 2022; 27:665-683. [PMID: 35850605 DOI: 10.1080/10837450.2022.2103825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Gold nanoparticles (GNP) are noble metal nanocarriers that have been recently researched upon for pharmaceutical applications, imaging, and diagnosis. These metallic nanocarriers are easy to synthesize using chemical reduction techniques as their surface can be easily modified. Also, the properties of GNP are significantly affected by its size and shape which mandates its stabilization using suitable techniques of surface modification. Over the past decade, research has focused on surface modification of GNP and its stabilization using polymers, polysaccharides, proteins, dendrimers, and phase-stabilizers like gel phase or ionic liquid phase. The use of GNP for pharmaceutical applications requires its surface modification using biocompatible and inert surface modifiers. The stabilizers used, interact with the surface of GNP to provide either electrostatic stabilization or steric stabilization. This review extensively discusses the surface modification techniques for GNP and the related molecular level interactions involved in the same. The influence of various factors like the concentration of stabilizers used their characteristics like chain length and thickness, pH of the surrounding media, etc., on the surface of GNP and resulting to stability have been discussed in detail. Further, this review highlights the recent applications of surface-modified GNP in the management of tumor microenvironment and cancer therapy.
Collapse
Affiliation(s)
- Kartik Hariharan
- Institute of Pharmacy, Nirma University, SG Highway, Gota, Ahmedabad-382481, Gujarat, India
| | - Parth Patel
- Institute of Pharmacy, Nirma University, SG Highway, Gota, Ahmedabad-382481, Gujarat, India
| | - Tejal Mehta
- Institute of Pharmacy, Nirma University, SG Highway, Gota, Ahmedabad-382481, Gujarat, India
| |
Collapse
|
21
|
Wang S, Ma Y, Ma C, Liu K, Huo Z, Shang Y. A supramolecular nanofiber formed by enzyme-instructed self-assembly for SKBR-3 cell selective inhibition. Chem Asian J 2022; 17:e202200301. [PMID: 35510693 DOI: 10.1002/asia.202200301] [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] [Received: 03/23/2022] [Revised: 04/27/2022] [Indexed: 11/06/2022]
Abstract
Cell-targeted peptides are recommended for precision cancer treatment due to their comparable targeting properties, small molecular size and good biocompatibility. However, unpredictable bioactivity, low penetration rate and poor stability greatly limit its efficacy. Supramolecular self-assembly based on synthetic peptide has great potential to solve related problems and achieve better therapeutic effects. Herein, we report and compare the effects of two different assembly pathway, heating-cooling and enzyme instruction, on the penetrability of SKBR-3 cell targeted peptides. It was found that enzyme-instructed self-assembly (EISA) resulted in hydrogels composed of uniform supramolecular nanofibers, whereas heating-cooling resulted in solutions and precipitations composed of slightly different nanoparticles. The nanofibers formed by EISA showed enhanced cellular uptake (2.54 μM), which was significantly higher than the 1.06 μM of the nanoparticles formed by temperature regulation. Thus, EISA is a promising strategy to improve the cell penetration rate of targeted peptides, and could provide a better solution for precision cancer treatment.
Collapse
Affiliation(s)
- Shijiang Wang
- Shandong Cancer Hospital and Institute, Department of Radiotherapy, CHINA
| | - Yan Ma
- Shandong Cancer Hospital and Institute, Department of Gastrointestinal oncology, CHINA
| | - Changsheng Ma
- Shandong Cancer Hospital and Institute, Department of Radiotherapy, CHINA
| | - Kai Liu
- Shandong Cancer Hospital and Institute, Department of Gastrointestinal oncology, CHINA
| | - Zhijun Huo
- Shandong Cancer Hospital and Institute, Breast Cancer Center, CHINA
| | - Yuna Shang
- Tianjin Normal University, College of Chemistry, 393# Binshuixi road, 300387, Tianjin, CHINA
| |
Collapse
|
22
|
Banthia P, Gambhir L, Sharma A, Daga D, Kapoor N, Chaudhary R, Sharma G. Nano to rescue: repository of nanocarriers for targeted drug delivery to curb breast cancer. 3 Biotech 2022; 12:70. [PMID: 35223356 PMCID: PMC8841383 DOI: 10.1007/s13205-022-03121-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/16/2022] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is a heterogeneous disease with different intrinsic subtypes. The conventional treatment of surgical resection, chemotherapy, immunotherapy and radiotherapy has not shown significant improvement in the survival rate of breast cancer patients. The therapeutics used cause bystander toxicities deteriorating healthy tissues. The breakthroughs of nanotechnology have been a promising feat in selective targeting of tumor site thus increasing the therapeutic gain. By the application of nanoenabled carriers, nanomedicines ensure targeted delivery, stability, enhanced cellular uptake, biocompatibility and higher apoptotic efficacy. The present review focuses on breakthrough of nanoscale intervention in targeted drug delivery as novel class of therapeutics. Nanoenabled carriers like polymeric and metallic nanoparticles, dendrimers, quantum dots, liposomes, solid lipid nanoparticles, carbon nanotubes, drug-antibody conjugates and exosomes revolutionized the targeted therapeutic delivery approach. These nanoassemblies have shown additional effect of improving the solubility of drugs such as paclitaxel, reducing the dose and toxicity. The present review provides an insight on the different drug conjugates employed/investigated to curb breast cancer using nanocarrier mediated targeted drug delivery. However, identification of appropriate biomarkers to target, clearer insight of the biological processes, batch uniformity, reproducibility, nanomaterial toxicity and stabilities are the hurdles faced by nanodrugs. The potential of nano-therapeutics delivery necessitates the agglomerated efforts of research community to bridge the route of nanodrugs for scale-up, commercialization and clinical applications.
Collapse
Affiliation(s)
- Poonam Banthia
- School of Applied Sciences, Suresh Gyan Vihar University, Jaipur, Rajasthan India
| | - Lokesh Gambhir
- School of Applied Sciences, Suresh Gyan Vihar University, Jaipur, Rajasthan India
| | - Asha Sharma
- Department of Zoology, Swargiya P. N. K. S. Govt. PG College, Dausa, Rajasthan India
| | - Dhiraj Daga
- Department of Radiation Oncology, JLN Medical College, Ajmer, Rajasthan India
| | - Neha Kapoor
- School of Applied Sciences, Suresh Gyan Vihar University, Jaipur, Rajasthan India
| | - Rishabh Chaudhary
- Department of Emergency Medicine, Institute of Bioelectronic Medicine, Feinstein Institute of Medical Research, Northwell Health, New Hyde Park, NY USA
| | - Gaurav Sharma
- School of Applied Sciences, Suresh Gyan Vihar University, Jaipur, Rajasthan India
| |
Collapse
|
23
|
Le TN, Lin CJ, Shen YC, Lin KY, Lee CK, Huang CC, Rao NV. Hyaluronic Acid Derived Hypoxia-Sensitive Nanocarrier for Tumor Targeted Drug Delivery. ACS APPLIED BIO MATERIALS 2021; 4:8325-8332. [PMID: 35005953 DOI: 10.1021/acsabm.1c00847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hyaluronic acid (HA) is conjugated with BHQ3 moiety with azo bonds to prepare hypoxia-responsive polymer conjugate. Because of the amphiphilic nature, the polymer conjugate self-assembles to HA-BHQ3 nanoparticles (NPs). The anticancer drug doxorubicin (DOX) is loaded into the NPs. In the physiological environment, DOX is released slowly. In contrast, under hypoxic conditions, the azo bond in BHQ3 is cleaved, thus significantly enhancing the DOX release rate. For instance, after 24 h, 25% of DOX is released under normal conditions, while 74% of DOX is released under hypoxic conditions. In vitro cytotoxicity demonstrates higher toxicity in the hypoxic conditions. DOX@HA-BHQ3 NPs exhibit greater toxicity levels against 4T1 cells in hypoxic conditions. The fluorescent microscope images confirm the oxygen-dependent intracellular DOX release from the NPs. The in vivo biodistribution results suggest the tumor targetability of HA-BHQ3 NPs in 4T1 tumor-bearing mice.
Collapse
Affiliation(s)
- Trong-Nghia Le
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Chin-Jung Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Yen Chen Shen
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Kuan-Yu Lin
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Cheng-Kang Lee
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - N Vijayakameswara Rao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| |
Collapse
|
24
|
Foglietta F, Serpe L, Canaparo R. The Effective Combination between 3D Cancer Models and Stimuli-Responsive Nanoscale Drug Delivery Systems. Cells 2021; 10:3295. [PMID: 34943803 PMCID: PMC8699241 DOI: 10.3390/cells10123295] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Stimuli-responsive drug-delivery systems (DDSs) have emerged as a potential tool for applications in healthcare, mainly in the treatment of cancer where versatile nanocarriers are co-triggered by endogenous and exogenous stimuli. Two-dimensional (2D) cell cultures are the most important in vitro model used to evaluate the anticancer activity of these stimuli-responsive DDSs due to their easy manipulation and versatility. However, some limitations suggest that these in vitro models poorly predict the outcome of in vivo studies. One of the main drawbacks of 2D cell cultures is their inadequate representation of the 3D environment's physiological complexity, which sees cells interact with each other and the extracellular matrix (ECM) according to their specific cellular organization. In this regard, 3D cancer models are a promising approach that can overcome the main shortcomings of 2D cancer cell cultures, as these in vitro models possess many peculiarities by which they mimic in vivo tumors, including physiologically relevant cell-cell and cell-ECM interactions. This is, in our opinion, even more relevant when a stimuli-responsive DDS is being investigated. In this review, we therefore report and discuss endogenous and exogenous stimuli-responsive DDSs whose effectiveness has been tested using 3D cancer cell cultures.
Collapse
Affiliation(s)
| | - Loredana Serpe
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy; (F.F.); (R.C.)
| | | |
Collapse
|
25
|
Mamnoon B, Feng L, Froberg J, Choi Y, Sathish V, Taratula O, Taratula O, Mallik S. Targeting Estrogen Receptor-Positive Breast Microtumors with Endoxifen-Conjugated, Hypoxia-Sensitive Polymersomes. ACS OMEGA 2021; 6:27654-27667. [PMID: 34722965 PMCID: PMC8552235 DOI: 10.1021/acsomega.1c02250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Endoxifen is the primary active metabolite of tamoxifen, a nonsteroidal-selective estrogen receptor modulator (SERM) and widely used medication to treat estrogen receptor-positive (ER+) breast cancer. In this study, endoxifen was conjugated to the surface of polymeric nanoparticles (polymersomes) for targeted delivery of doxorubicin (DOX) to estrogen receptor-positive breast cancer cells (MCF7). Rapid cell growth and insufficient blood supply result in low oxygen concentration (hypoxia) within the solid breast tumors. The polymersomes developed here are prepared from amphiphilic copolymers of polylactic acid (PLA) and poly(ethylene glycol) (PEG) containing diazobenzene as the hypoxia-responsive linker. We prepared two nanoparticle formulations: DOX-encapsulated hypoxia-responsive polymersomes (DOX-HRPs) and endoxifen-conjugated, DOX-encapsulated hypoxia-responsive polymersomes (END-DOX-HRPs). Cellular internalization studies demonstrated eight times higher cytosolic and nuclear localization after incubating breast cancer cells with END-DOX-HRPs (targeted polymersomes) in contrast to DOX-HRPs (nontargeted polymersomes). Cytotoxicity studies on monolayer cell cultures exhibited that END-DOX-HRPs were three times more toxic to ER+ MCF7 cells than DOX-HRPs and free DOX in hypoxia. The cell viability studies on three-dimensional hypoxic cultures also demonstrated twice as much toxicity when the spheroids were treated with targeted polymersomes instead of nontargeted counterparts. This is the first report of surface-decorated polymeric nanoparticles with endoxifen ligands for targeted drug delivery to ER+ breast cancer microtumors. The newly designed endoxifen-conjugated, hypoxia-responsive polymersomes might have translational potential for ER+ breast cancer treatment.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| | - Li Feng
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| | - Jamie Froberg
- Department
of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Yongki Choi
- Department
of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Venkatachalem Sathish
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| | - Oleh Taratula
- Department
of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Olena Taratula
- Department
of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Sanku Mallik
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
26
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
27
|
Development of Polymer-Assisted Nanoparticles and Nanogels for Cancer Therapy: An Update. Gels 2021; 7:gels7020060. [PMID: 34067587 PMCID: PMC8162331 DOI: 10.3390/gels7020060] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
With cancer remaining as one of the main causes of deaths worldwide, many studies are undergoing the effort to look for a novel and potent anticancer drug. Nanoparticles (NPs) are one of the rising fields in research for anticancer drug development. One of the key advantages of using NPs for cancer therapy is its high flexibility for modification, hence additional properties can be added to the NPs in order to improve its anticancer action. Polymer has attracted considerable attention to be used as a material to enhance the bioactivity of the NPs. Nanogels, which are NPs cross-linked with hydrophilic polymer network have also exhibited benefits in anticancer application. The characteristics of these nanomaterials include non-toxic, environment-friendly, and variable physiochemical properties. Some other unique properties of polymers are also attributed by diverse methods of polymer synthesis. This then contributes to the unique properties of the nanodrugs. This review article provides an in-depth update on the development of polymer-assisted NPs and nanogels for cancer therapy. Topics such as the synthesis, usage, and properties of the nanomaterials are discussed along with their mechanisms and functions in anticancer application. The advantages and limitations are also discussed in this article.
Collapse
|
28
|
Metzger W, Rösch B, Sossong D, Bubel M, Pohlemann T. Flow cytometric quantification of apoptotic and proliferating cells applying an improved method for dissociation of spheroids. Cell Biol Int 2021; 45:1633-1643. [PMID: 33913594 DOI: 10.1002/cbin.11618] [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] [Received: 01/06/2021] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 01/31/2023]
Abstract
Spheroids are a promising tool for many cell culture applications, but their microscopic analysis is limited. Flow cytometry on a single cell basis, which requires a gentle but also efficient dissociation of spheroids, could be an alternative analysis. Mono-culture and coculture spheroids consisting of human fibroblasts and human endothelial cells were generated by the liquid overlay technique and were dissociated using AccuMax as a dissociation agent combined with gentle mechanical forces. This study aimed to quantify the number of apoptotic and proliferative cells. We were able to dissociate spheroids of differing size, age, and cellular composition in a single-step dissociation protocol within 10 min. The number of single cells was higher than 95% and in most cases, the viability of the cells after dissociation was higher than 85%. Coculture spheroids exhibited a higher sensitivity as shown by lower viability, higher amount of cellular debris, and a higher amount of apoptotic cells. Considerable expression of the proliferation marker Ki67 could only be seen in 1-day-old spheroids but was already downregulated on Day 3. In summary, our dissociation protocol enabled a fast and gentle dissociation of spheroids for the subsequent flow cytometric analysis. The chosen cell type had a strong influence on cell viability and apoptosis. Initially high rates of proliferative cells decreased rapidly and reached values of healthy tissue 3 days after generation of the spheroids. In conclusion, the flow cytometry of dissociated spheroids could be a promising analytical tool, which could be ideally combined with microscopic techniques.
Collapse
Affiliation(s)
- Wolfgang Metzger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Barbara Rösch
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Daniela Sossong
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Monika Bubel
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
29
|
Wei C, Wang P, Huang Z, He D, Zhu W, Liu H, Chen Z, Wang W, Li Y, Shen J, Qin L. Construction of Surface-Modified Polydopamine Nanoparticles for Sequential Drug Release and Combined Chemo-Photothermal Cancer Therapy. Mol Pharm 2021; 18:1327-1343. [PMID: 33530691 DOI: 10.1021/acs.molpharmaceut.0c01164] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Single chemotherapy often causes severe adverse effects and drug resistance to limit therapeutic efficacy. As a noninvasive approach, photothermal therapy (PTT) represents an attractive option for cancer therapy due to the benefits of remote control and precise treatment methods. Nanomedicines constructed with combined chemo-photothermal properties may exert synergistic effects and improved antitumor efficacy. In this study, we developed polydopamine (PDA)-coated nanoparticles grafted with folic acid (FA) and polyethylene glycol to transport doxorubicin (DOX) for targeted cancer therapy. The results showed that this delivery vehicle has a nanoscale particle size and narrow size distribution. No particle aggregation or significant drug leakage was observed during the stability test. This system presented excellent photothermal conversion capability under near-infrared light (NIR) laser irradiation due to the PDA layer covering. In vitro dissolution profiles demonstrated that sequential and triggered DOX release from nanoparticles was pH-, NIR irradiation-, and redox level-dependent and could be best fitted with the Ritger-Peppas equation. FA modification effectively promoted the intracellular uptake of nanoparticles by HepG2 cells and therefore significantly inhibited cell recovery and induced tumor cell apoptosis. Compared to the free DOX group, nanoparticles reduced the DOX concentration in the heart to avoid drug-related cardiotoxicity. More importantly, the in vivo antitumor efficacy results showed that compared with the single chemotherapy strategy, the nanoparticle group exerted combined and satisfactory tumor growth inhibition effects with good biocompatibility. In summary, this nanocarrier delivery system can organically combine chemotherapy and PTT to achieve effective and precise cancer treatment.
Collapse
Affiliation(s)
- Cui Wei
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pengfei Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhenpeng Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dahua He
- Department of Pharmacy, Guangdong Women and Children Hospital, Guangzhou 510010, China
| | - Wanye Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huan Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhihao Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wanting Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yusheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Juan Shen
- Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Linghao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
30
|
Shi Z, Wang Y, Xiao T, Dong S, Lan T. Preparation and Thermal Decomposition Kinetics of a New Type of a Magnetic Targeting Drug Carrier. ACS OMEGA 2021; 6:3427-3433. [PMID: 33553961 PMCID: PMC7860512 DOI: 10.1021/acsomega.0c06075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/06/2021] [Indexed: 05/15/2023]
Abstract
We have designed a new magnetic targeting drug carrier Fe3O4-PVA with a core of triiron tetroxide (Fe3O4) and a shell made of polyvinyl alcohol (PVA) to improve the hydrophilicity of Fe3O4. With adriamycin hydrochloride as a model drug, this study goes on to measure the drug carrier performance of Fe3O4-PVA. In addition, the thermal stability and enthalpy of thermal decomposition of Fe3O4-PVA were measured using a differential scanning calorimeter with a non-isothermal decomposition method. The kinetics of thermal decomposition of Fe3O4-PVA were also investigated. Over the course of this study, it was determined that the resulting drug carrier Fe3O4-PVA exhibited high drug loading levels and excellent release levels.
Collapse
Affiliation(s)
- Zhen Shi
- College
of Materials Science and Engineering, Qiqihar
University, Qiqihar 161006, Heilongjiang, China
- Heilongjiang
Provincial Key Laboratory of Polymeric Composite Materials, Qiqihar 161006, China
| | - Yazhen Wang
- College
of Materials Science and Engineering, Qiqihar
University, Qiqihar 161006, Heilongjiang, China
- Heilongjiang
Provincial Key Laboratory of Polymeric Composite Materials, Qiqihar 161006, China
- College
of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Tianyuan Xiao
- Heilongjiang
Provincial Key Laboratory of Polymeric Composite Materials, Qiqihar 161006, China
| | - Shaobo Dong
- Heilongjiang
Provincial Key Laboratory of Polymeric Composite Materials, Qiqihar 161006, China
| | - Tianyu Lan
- Heilongjiang
Provincial Key Laboratory of Polymeric Composite Materials, Qiqihar 161006, China
| |
Collapse
|
31
|
Jahan S, Karim ME, Chowdhury EH. Nanoparticles Targeting Receptors on Breast Cancer for Efficient Delivery of Chemotherapeutics. Biomedicines 2021; 9:114. [PMID: 33530291 PMCID: PMC7910939 DOI: 10.3390/biomedicines9020114] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/25/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
The journey of chemotherapeutic drugs from the site of administration to the site of action is confronted by several factors including low bioavailability, uneven distribution in major organs, limited accessibility of drug molecules to the distant tumor tissues, and lower therapeutic indexes. These unavoidable features of classical chemotherapeutics necessitate an additional high, repetitive dose of drugs to obtain maximum therapeutic responses with the result of unintended adverse side effects. An erratic tumor microenvironment, notable drawbacks of conventional chemotherapy, and multidrug-resistant mechanisms of breast cancer cells warrant precisely designed therapeutics for the treatment of cancers. In recent decades, nanoparticles have been deployed for the delivery of standard anticancer drugs to maximize the therapeutic potency while minimizing the adverse effects to increase the quality and span of life. Several organic and inorganic nanoplatforms that have been designed exploiting the distinctive features of the tumor microenvironment and tumor cells offer favorable physicochemical properties and pharmacokinetic profiles of a parent drug, with delivery of higher amounts of the drug to the pathological site and its controlled release, thereby improving the balance between its efficacy and toxicity. Advances to this front have included design and construction of targeted nanoparticles by conjugating homing devices like peptide, ligand, and Fab on the surface of nanomaterials to navigate nanoparticledrug complexes towards the target tumor cell with minimal destruction of healthy cells. Furthermore, actively targeting nanoparticles can facilitate the delivery and cellular uptake of nanoparticle-loaded drug constructs via binding with specific receptors expressed aberrantly on the surface of a tumor cell. Herein, we present an overview of the principle of targeted delivery approaches, exploiting drug-nanoparticle conjugates with multiple targeting moieties to target specific receptors of breast cancer cells and highlighting therapeutic evaluation in preclinical studies. We conclude that an understanding of the translational gap and challenges would show the possible future directions to foster the development of novel targeted nanotherapeutics.
Collapse
Affiliation(s)
| | | | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Petaling Jaya 47500, Malaysia; (S.J.); (M.E.K.)
| |
Collapse
|
32
|
Shi Z, Wang Y, Dong S, Lan T. Comparison of the performance of magnetic targeting drug carriers prepared using two synthesis methods. RSC Adv 2021; 11:20670-20678. [PMID: 35479366 PMCID: PMC9033997 DOI: 10.1039/d1ra04256d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
In this paper, two methods were used to prepare the magnetic targeting drug carrier Fe3O4–PVA@SH, the step-by-step method and the one-pot method. The loading and release properties of the compound were measured. The results show that the Fe3O4–PVA@SH prepared using both methods exhibited excellent drug delivery properties in an environment that simulates human body fluid (pH 7.2) and a lysosomal in vitro simulation (pH 4.7). In applications such as drug delivery, magnetic targeted drug carriers prepared by both methods demonstrated superparamagnetism, high fat solubility, high hydroxyl content, and good water solubility. Roadmap for the synthesis of Fe3O4–PVA@SH using the step-by-step method and one-pot method.![]()
Collapse
Affiliation(s)
- Zhen Shi
- College of Materials Science and Engineering
- Qiqihar University
- Qiqihar 161006
- China
- Heilongjiang Provincial Key Laboratory of Polymeric Composite Materials
| | - Yazhen Wang
- College of Materials Science and Engineering
- Qiqihar University
- Qiqihar 161006
- China
- Heilongjiang Provincial Key Laboratory of Polymeric Composite Materials
| | - Shaobo Dong
- Heilongjiang Provincial Key Laboratory of Polymeric Composite Materials
- Qiqihar 161006
- China
| | - Tianyu Lan
- Heilongjiang Provincial Key Laboratory of Polymeric Composite Materials
- Qiqihar 161006
- China
| |
Collapse
|
33
|
Mamnoon B, Loganathan J, Confeld MI, De Fonseka N, Feng L, Froberg J, Choi Y, Tuvin DM, Sathish V, Mallik S. Targeted polymeric nanoparticles for drug delivery to hypoxic, triple-negative breast tumors. ACS APPLIED BIO MATERIALS 2020; 4:1450-1460. [PMID: 33954285 DOI: 10.1021/acsabm.0c01336] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High recurrence and metastasis to vital organs are the major characteristics of triple-negative breast cancer (TNBC). Low vascular oxygen tension promotes resistance to chemo- and radiation therapy. Neuropilin-1 (NRP-1) receptor is highly expressed on TNBC cells. The tumor-penetrating iRGD peptide interacts with the NRP-1 receptor, triggers endocytosis and transcytosis, and facilitates penetration. Herein, we synthesized a hypoxia-responsive diblock PLA-diazobenzene-PEG copolymer and prepared self-assembled hypoxia-responsive polymersomes (Ps) in an aqueous buffer. The iRGD peptide was incorporated into the polymersome structure to make hypoxia-responsive iRGD-conjugated polymersomes (iPs). Doxorubicin (DOX) was encapsulated in the polymersomes to prepare both targeted and non-targeted hypoxia-responsive polymersomes (DOX-iPs and DOX-Ps, respectively). The polymeric nanoparticles released less than 30% of their encapsulated DOX within 12 hours under normoxic conditions (21% oxygen), whereas under hypoxia (2% Oxygen), doxorubicin release remarkably increased to over 95%. The targeted polymersomes significantly decreased TNBC cells' viability in monolayer and spheroid cultures under hypoxia compared to normoxia. Animal studies displayed that targeted polymersomes significantly diminished tumor growth in xenograft nude mice. Overall, the targeted polymersomes exhibited potent anti-tumor activity in monolayer, spheroid, and animal models of TNBC. With further developments, the targeted nanocarriers discussed here might have the translational potential as drug carriers for the treatment of TNBC.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Jagadish Loganathan
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Matthew I Confeld
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Nimesha De Fonseka
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Li Feng
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Jamie Froberg
- Department of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Daniel M Tuvin
- Sanford Broadway Clinic, Fargo, North Dakota 58102, United States
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
34
|
Thomas RG, Surendran SP, Jeong YY. Tumor Microenvironment-Stimuli Responsive Nanoparticles for Anticancer Therapy. Front Mol Biosci 2020; 7:610533. [PMID: 33392264 PMCID: PMC7775573 DOI: 10.3389/fmolb.2020.610533] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer is a disease that affects a large number of people all over the world. For treating cancer, nano-drug delivery system has been introduced recently with objective of increasing therapeutic efficiency of chemotherapeutic drug. The main characteristics of this system are the encapsulation of the insoluble chemotherapeutic cargo, increasing the period of circulation in the body, as well as the delivery of the drug at that specific site. Currently, the nano-drug delivery system based on the stimuli response is becoming more popular because of the extra features for controlling the drug release based on the internal atmosphere of cancer. This review provides a summary of different types of internal (pH, redox, enzyme, ROS, hypoxia) stimuli-responsive nanoparticle drug delivery systems as well as perspective for upcoming times.
Collapse
Affiliation(s)
- Reju George Thomas
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
- BioMolecular Theranostics (BiT) Laboratory, Department of Biomedical Sciences, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| | - Suchithra Poilil Surendran
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
- BioMolecular Theranostics (BiT) Laboratory, Department of Biomedical Sciences, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
- BioMolecular Theranostics (BiT) Laboratory, Department of Biomedical Sciences, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| |
Collapse
|