1
|
Lammers T. Nanomedicine Tumor Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312169. [PMID: 38361435 DOI: 10.1002/adma.202312169] [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: 11/14/2023] [Revised: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Nanomedicines are extensively explored for cancer therapy. By delivering drug molecules more efficiently to pathological sites and by attenuating their accumulation in healthy organs and tissues, nanomedicine formulations aim to improve the balance between drug efficacy and toxicity. More than 20 cancer nanomedicines are approved for clinical use, and hundreds of formulations are in (pre)clinical development. Over the years, several key pitfalls have been identified as bottlenecks in nanomedicine tumor targeting and translation. These go beyond materials- and production-related issues, and particularly also encompass biological barriers and pathophysiological heterogeneity. In this manuscript, the author describes the most important principles, progress, and products in nanomedicine tumor targeting, delineates key current problems and challenges, and discusses the most promising future prospects to create clinical impact.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohyhrid Medical Systems, University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
2
|
Kashyap A, Kumari M, Singh A, Mukherjee K, Maity D. Current development of theragnostic nanoparticles for women's cancer treatment. Biomed Mater 2024; 19:042001. [PMID: 38471150 DOI: 10.1088/1748-605x/ad3311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/12/2024] [Indexed: 03/14/2024]
Abstract
In the biomedical industry, nanoparticles (NPs-exclusively small particles with size ranging from 1-100 nanometres) are recently employed as powerful tools due to their huge potential in sophisticated and enhanced cancer theragnostic (i.e. therapeutics and diagnostics). Cancer is a life-threatening disease caused by carcinogenic agents and mutation in cells, leading to uncontrolled cell growth and harming the body's normal functioning while affecting several factors like low levels of reactive oxygen species, hyperactive antiapoptotic mRNA expression, reduced proapoptotic mRNA expression, damaged DNA repair, and so on. NPs are extensively used in early cancer diagnosis and are functionalized to target receptors overexpressing cancer cells for effective cancer treatment. This review focuses explicitly on how NPs alone and combined with imaging techniques and advanced treatment techniques have been researched against 'women's cancer' such as breast, ovarian, and cervical cancer which are substantially occurring in women. NPs, in combination with numerous imaging techniques (like PET, SPECT, MRI, etc) have been widely explored for cancer imaging and understanding tumor characteristics. Moreover, NPs in combination with various advanced cancer therapeutics (like magnetic hyperthermia, pH responsiveness, photothermal therapy, etc), have been stated to be more targeted and effective therapeutic strategies with negligible side effects. Furthermore, this review will further help to improve treatment outcomes and patient quality of life based on the theragnostic application-based studies of NPs in women's cancer treatment.
Collapse
Affiliation(s)
- Ananya Kashyap
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Madhubala Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Arnika Singh
- Department of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Koel Mukherjee
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Dipak Maity
- Integrated Nanosystems Development Institute, Indiana University Indianapolis, IN 46202, United States of America
- Department of Chemistry and Chemical Biology, Indiana University Indianapolis, IN 46202, United States of America
| |
Collapse
|
3
|
Saman S, Srivastava N, Yasir M, Chauhan I. A Comprehensive Review on Current Treatments and Challenges Involved in the Treatment of Ovarian Cancer. Curr Cancer Drug Targets 2024; 24:142-166. [PMID: 37642226 DOI: 10.2174/1568009623666230811093139] [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: 01/20/2023] [Revised: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 08/31/2023]
Abstract
Ovarian cancer (OC) is the second most common gynaecological malignancy. It typically affects females over the age of 50, and since 75% of cases are only discovered at stage III or IV, this is a sign of a poor diagnosis. Despite intraperitoneal chemotherapy's chemosensitivity, most patients relapse and face death. Early detection is difficult, but treatment is also difficult due to the route of administration, resistance to therapy with recurrence, and the need for precise cancer targeting to minimize cytotoxicity and adverse effects. On the other hand, undergoing debulking surgery becomes challenging, and therapy with many chemotherapeutic medications has manifested resistance, a condition known as multidrug resistance (MDR). Although there are other therapeutic options for ovarian cancer, this article solely focuses on co-delivery techniques, which work via diverse pathways to overcome cancer cell resistance. Different pathways contribute to MDR development in ovarian cancer; however, usually, pump and non-pump mechanisms are involved. Striking cancerous cells from several angles is important to defeat MDR. Nanocarriers are known to bypass the drug efflux pump found on cellular membranes to hit the pump mechanism. Nanocarriers aid in the treatment of ovarian cancer by enhancing the delivery of chemotherapeutic drugs to the tumour sites through passive or active targeting, thereby reducing unfavorable side effects on the healthy tissues. Additionally, the enhanced permeability and retention (EPR) mechanism boosts the bioavailability of the tumour site. To address the shortcomings of conventional delivery, the current review attempts to explain the current conventional treatment with special reference to passively and actively targeted drug delivery systems (DDSs) towards specific receptors developed to treat ovarian cancer. In conclusion, tailored nanocarriers would optimize medication delivery into the intracellular compartment before optimizing intra-tumour distribution. Other novel treatment possibilities for ovarian cancer include tumour vaccines, gene therapy, targeting epigenetic alteration, and biologically targeted compounds. These characteristics might enhance the therapeutic efficacy.
Collapse
Affiliation(s)
- Saika Saman
- Department of Pharmaceutics, Faculty of Pharmacy, Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Sector 125, Noida, 201313, India
| | - Nimisha Srivastava
- Department of Pharmaceutics, Faculty of Pharmacy, Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Sector 125, Noida, 201313, India
| | - Mohd Yasir
- Department of Pharmacy (Pharmaceutics), College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Iti Chauhan
- Department of Pharmacy, I.T.S College of Pharmacy, Muradnagar, Ghaziabad, India
| |
Collapse
|
4
|
Keyvani V, Mollazadeh S, Riahi E, Mahmoudian RA, Tabari M, Lagzian E, Ghorbani E, Akbarzade H, Gholami AS, Gataa IS, Hassanian SM, Ferns GA, Khazaei M, Avan A, Anvari K. The Application of Nanotechnological Therapeutic Platforms against Gynecological Cancers. Curr Pharm Des 2024; 30:975-987. [PMID: 38500284 DOI: 10.2174/0113816128291955240306112558] [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: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
Gynecological cancers (GCs), ovarian, cervical, and endometrial/uterine cancers, are often associated with poor outcomes. Despite the development of several therapeutic modalities against GCs, the effectiveness of the current therapeutic approaches is limited due to their side effects, low therapeutic index, short halflife, and resistance to therapy. To overcome these limitations, nano delivery-based approaches have been introduced with the potential of targeted delivery, reduced toxicity, controlled release, and improved bioavailability of various cargos. This review summarizes the application of different nanoplatforms, such as lipid-based, metal- based, and polymeric nanoparticles, to improve the chemo/radio treatments of GC. In the following work, the use of nanoformulated agents to fight GCs has been mentioned in various clinical trials. Although nanosystems have their own challenges, the knowledge highlighted in this article could provide deep insight into translations of NPs approaches to overcome GCs.
Collapse
Affiliation(s)
- Vahideh Keyvani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Espanta Riahi
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Mashhad, Iran
- Department of Biology, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Reihaneh Alsadat Mahmoudian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoomeh Tabari
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Lagzian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Akbarzade
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir-Sadra Gholami
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane 4059, Australia
| | - Kazem Anvari
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Liu B, Zhang J, Liu Z, Wang P, Zhang Y, He H, Yin T, Gou J, Tang X. Research on the preparation process of the cytarabine/daunorubicin dual-encapsulation liposome and its physicochemical properties and performances in vitro/vivo. Int J Pharm 2023; 646:123500. [PMID: 37820944 DOI: 10.1016/j.ijpharm.2023.123500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/20/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
As the only Food and Drug Administration (FDA)-approved dual-encapsulation liposome injection for treating Acute myeloid leukemia (AML), CPX-351 outperforms the standard chemotherapy treatment "DA 7 + 3″ in terms of clinical effectiveness. Although research on dual-loaded liposomes has increased in recent years, little attention has been paid to their preparation, which can affect their quality, efficacy, and safety. This study explored various preparation processes to create the cytarabine/daunorubicin co-loaded liposome (the Cyt/Daun liposome) and eventually settled on two methods: the sequential loading approach, thin film hydration-extrusion-copper ion gradient, and the simultaneous encapsulation technique, copper ion gradient-concentration gradient. Different preparation methods resulted in different particle sizes and encapsulation efficiencies; the two aforementioned preparation processes generated dual-loaded liposomes with comparable physicochemical properties. The sequential encapsulation technique was selected for the subsequent research owing to its higher encapsulation efficiency prior to purification; the prepared Cyt/Daun liposomes had small and uniform particle size (108.6 ± 1.02 nm, Polydispersity index (PDI) 0.139 ± 0.01), negative charge (-(60.2 ± 1.15) mV), high drug encapsulation efficiency (Cyt 88.2 ± 0.24 %, Duan 94.2 ± 0.45 %) and good plasma stability. To improve its storage stability, the Cyt/Daun liposome was lyophilized (-40 °C for 4 h, maintained for 130 min, and dried for 1200 min) using sucrose-raffinose (mass ratio 7:3; glycolipid ratio 4:1, w/w) as a lyoprotectant. The lyophilized liposomes were purple cakes, redissolved rapidly with insignificant alterations in particle size and encapsulation efficiency, and possessed well storage stability. The pharmacokinetic and tissue distribution studies demonstrated that the Cyt/Daun liposome could achieve long circulation and maintain synergic proportions of drugs within 24 h, increasing the accumulation of drugs at tumor sites. Furthermore, the in vitro/in vivo pharmacodynamic studies confirmed its good anti-tumor activity and safety.
Collapse
Affiliation(s)
- Boyuan Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Jiaoyang Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Zixu Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Ping Wang
- School of Pharmacy, Jilin University, Changchun 130021, Jilin, PR China
| | - Yu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Jingxin Gou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
6
|
Dana PM, Sadoughi F, Reiter RJ, Mohammadi S, Heidar Z, Mirzamoradi M, Asemi Z. Melatonin as an adjuvant treatment modality with doxorubicin [Biochimie 200 (2022) 1-7]. Biochimie 2022; 200:1-7. [PMID: 35569703 DOI: 10.1016/j.biochi.2022.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/30/2022] [Accepted: 05/09/2022] [Indexed: 01/12/2023]
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R, Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA
| | - Sotoudeh Mohammadi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Heidar
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Mirzamoradi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R, Iran
| |
Collapse
|
7
|
Wang Z, Li J, Lin G, He Z, Wang Y. Metal complex-based liposomes: Applications and prospects in cancer diagnostics and therapeutics. J Control Release 2022; 348:1066-1088. [PMID: 35718211 DOI: 10.1016/j.jconrel.2022.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Metal complexes are of increasing interest as pharmaceutical agents in cancer diagnostics and therapeutics, while some of them suffer from issues such as limited water solubility and severe systemic toxicity. These drawbacks severely hampered their efficacy and clinical applications. Liposomes hold promise as delivery vehicles for constructing metal complex-based liposomes to maximize the therapeutic efficacy and minimize the side effects of metal complexes. This review provides an overview on the latest advances of metal complex-based liposomal delivery systems. First, the development of metal complex-mediated liposomal encapsulation is briefly introduced. Next, applications of metal complex-based liposomes in a variety of fields are overviewed, where drug delivery, cancer imaging (single photon emission computed tomography (SPECT), positron emission tomography (PET), and magnetic resonance imaging (MRI)), and cancer therapy (chemotherapy, phototherapy, and radiotherapy) were involved. Moreover, the potential toxicity, action of toxic mechanisms, immunological effects of metal complexes as well as the advantages of metal complex-liposomes in this content are also discussed. In the end, the future expectations and challenges of metal complex-based liposomes in clinical cancer therapy are tentatively proposed.
Collapse
Affiliation(s)
- Zhaomeng Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Guimei Lin
- School of Pharmacy, Shandong University, Jinan 250000, PR China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
8
|
Maleki Dana P, Sadoughi F, J Reiter R, Mohammadi S, Heidar Z, Mirzamoradi M, Asemi Z. Melatonin as an adjuvant treatment modality with doxorubicin. Biochimie 2022; 202:49-55. [PMID: 35752222 DOI: 10.1016/j.biochi.2022.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Combination chemotherapy seems to be a beneficial choice for some cancer patients particularly when the drugs target different processes of oncogenesis; patients treated with combination therapies sometimes have a better prognosis than those treated with single drug chemotherapy. However, research has shown that this is not always the case, and this approach may only increase toxicity without having a significant effect in augmenting the antitumor actions of the drugs. Doxorubicin (Dox) is one of the most common chemotherapy drugs used to treat many types of cancer, but it also has serious side effects, such as cardiotoxicity, skin necrosis, testicular toxicity, and nephrotoxicity. Many studies have examined the efficiacy of melatonin (MLT) as an anticancer agent. In fact, MLT is an anti-cancer agent that has various functions in inhibiting cancer cell proliferation, inducing apoptosis, and suppressing metastasis. Herein, we provide a comprehensive evaluation of the literature concerned with the role of MLT as an adjuvant in Dox-based chemotherapies and discuss how MLT may enhance the antitumor effects of Dox (e.g., by inducing apoptosis and suppressing metastasis) while rescuring other organs from its adverse effects, such as cardio- and nephrotoxicity.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA.
| | - Sotoudeh Mohammadi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zahra Heidar
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Mirzamoradi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| |
Collapse
|
9
|
Bhattacharjee S. Craft of Co-encapsulation in Nanomedicine: A Struggle To Achieve Synergy through Reciprocity. ACS Pharmacol Transl Sci 2022; 5:278-298. [PMID: 35592431 PMCID: PMC9112416 DOI: 10.1021/acsptsci.2c00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/19/2022]
Abstract
Achieving synergism, often by combination therapy via codelivery of chemotherapeutic agents, remains the mainstay of treating multidrug-resistance cases in cancer and microbial strains. With a typical core-shell architecture and surface functionalization to ensure facilitated targeting of tissues, nanocarriers are emerging as a promising platform toward gaining such synergism. Co-encapsulation of disparate theranostic agents in nanocarriers-from chemotherapeutic molecules to imaging or photothermal modalities-can not only address the issue of protecting the labile drug payload from a hostile biochemical environment but may also ensure optimized drug release as a mainstay of synergistic effect. However, the fate of co-encapsulated molecules, influenced by temporospatial proximity, remains unpredictable and marred with events with deleterious impact on therapeutic efficacy, including molecular rearrangement, aggregation, and denaturation. Thus, more than just an art of confining multiple therapeutics into a 3D nanoscale space, a co-encapsulated nanocarrier, while aiming for synergism, should strive toward achieving a harmonious cohabitation of the encapsulated molecules that, despite proximity and opportunities for interaction, remain innocuous toward each other and ensure molecular integrity. This account will inspect the current progress in co-encapsulation in nanocarriers and distill out the key points toward accomplishing such synergism through reciprocity.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
10
|
Kesavan A, Chandrasekhar Reddy U, Kurian J, Muraleedharan KM. Cancer cell uptake and distribution of oxanorbornane-based synthetic lipids and their prospects as novel drug delivery systems. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
11
|
Versatile Encapsulation and Synthesis of Potent Liposomes by Thermal Equilibration. MEMBRANES 2022; 12:membranes12030319. [PMID: 35323794 PMCID: PMC8954264 DOI: 10.3390/membranes12030319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/05/2023]
Abstract
The wide-scale use of liposomal delivery systems is challenged by difficulties in obtaining potent liposomal suspensions. Passive and active loading strategies have been proposed to formulate drug encapsulated liposomes but are limited by low efficiencies (passive) or high drug specificities (active). Here, we present an efficient and universal loading strategy for synthesizing therapeutic liposomes. Integrating a thermal equilibration technique with our unique liposome synthesis approach, co-loaded targeting nanovesicles can be engineered in a scalable manner with potencies 200-fold higher than typical passive encapsulation techniques. We demonstrate this capability through simultaneous co-loading of hydrophilic and hydrophobic small molecules and targeted delivery of liposomal Doxorubicin to metastatic breast cancer cell line MDA-MB-231. Molecular dynamic simulations are used to explain interactions between Doxorubicin and liposome membrane during thermal equilibration. By addressing the existing challenges, we have developed an unparalleled approach that will facilitate the formulation of novel theranostic and pharmaceutical strategies.
Collapse
|
12
|
Yang Y, Zuo S, Li L, Kuang X, Li J, Sun B, Wang S, He Z, Sun J. Iron-doxorubicin prodrug loaded liposome nanogenerator programs multimodal ferroptosis for efficient cancer therapy. Asian J Pharm Sci 2021; 16:784-793. [PMID: 35027953 PMCID: PMC8737402 DOI: 10.1016/j.ajps.2021.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/01/2021] [Accepted: 05/24/2021] [Indexed: 01/18/2023] Open
Affiliation(s)
- Yinxian Yang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shiyi Zuo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiao Kuang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinbo Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors.
| | - Shujun Wang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors.
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors.
| |
Collapse
|
13
|
Barbălată CI, Porfire AS, Sesarman A, Rauca VF, Banciu M, Muntean D, Știufiuc R, Moldovan A, Moldovan C, Tomuță I. A Screening Study for the Development of Simvastatin-Doxorubicin Liposomes, a Co-Formulation with Future Perspectives in Colon Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13101526. [PMID: 34683821 PMCID: PMC8537800 DOI: 10.3390/pharmaceutics13101526] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
An increasing number of studies published so far have evidenced the benefits of Simvastatin (SIM) and Doxorubicin (DOX) co-treatment in colorectal cancer. In view of this, the current study aimed to investigate the pharmaceutical development of liposomes co-encapsulating SIM and DOX, by implementing the Quality by Design (QbD) concept, as a means to enhance the antiproliferative effect of the co-formulation on C26 murine colon cancer cells co-cultured with macrophages. It is known that the quality profile of liposomes is dependent on the critical quality attributes (CQAs) of liposomes (drug entrapped concentration, encapsulation efficiency, size, zeta potential, and drug release profile), which are, in turn, directly influenced by various formulation factors and processing parameters. By using the design of experiments, it was possible to outline the increased variability of CQAs in relation to formulation factors and identify by means of statistical analysis the material attributes that are critical (phospholipids, DOX and SIM concentration) for the quality of the co-formulation. The in vitro studies performed on a murine colon cancer cell line highlighted the importance of delivering the optimal drug ratio at the target site, since the balance antiproliferative vs. pro-proliferative effects can easily be shifted when the molar ratio between DOX and SIM changes.
Collapse
Affiliation(s)
- Cristina Ioana Barbălată
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| | - Alina Silvia Porfire
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
- Correspondence:
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Dana Muntean
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| | - Rareș Știufiuc
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Alin Moldovan
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Cristian Moldovan
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Ioan Tomuță
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| |
Collapse
|
14
|
Zhang L, Zhang S, Jiang M, Lu L, Ding Y, Ma N, Zhao Y, Xuchen S, Zhang N. Novel Timosaponin AIII-Based Multifunctional Liposomal Delivery System for Synergistic Therapy Against Hepatocellular Carcinoma Cancer. Int J Nanomedicine 2021; 16:5531-5550. [PMID: 34429598 PMCID: PMC8379713 DOI: 10.2147/ijn.s313759] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction As high cholesterol level has been reported to be associated with cancer cell growth and cholesterol is vulnerable to oxidation, the conventional liposomes including cholesterol in the formulation seem to be challenged. Timosaponin AIII (TAIII), as a steroid saponin from Anemarrhena asphodeloides Bunge, possesses a similar structure with cholesterol and exhibits a wide range of antitumor activities, making it possible to develop a TAIII-based liposome where TAIII could potentially stabilize the phospholipid bilayer as a substitution of cholesterol and work as a chemotherapeutic drug as well. Meanwhile, TAIII could enhance the uptake of doxorubicin hydrochloride (DOX) in human hepatocellular carcinoma (HCC) cells and exhibit synergistic effect. Thus, we designed a novel thermally sensitive multifunctional liposomal system composed of TAIII and lipids to deliver DOX for enhanced HCC treatment. Methods The synergistic effects of DOX and TAIII were explored on HCC cells and the tumor inhibition rate of TAIII-based liposomes carrying DOX was evaluated on both subcutaneous and orthotopic transplantation tumor models. TAIII-based multifunctional liposomes were characterized. Results Synergistic HCC cytotoxicity was achieved at molar ratios of 1:1, 1:2 and 1:4 of DOX/TAIII. TAIII-based liposomes carrying a low DOX dose of 2 mg/kg exhibited significantly enhanced antitumor activity than 5 mg/kg of DOX without detected cardiotoxicity on both subcutaneous and orthotopic transplantation tumor models. TAIII-based liposomes were characterized with smaller size than cholesterol liposomes but exhibited favorable stability. Mild hyperthermia generated by laser irradiation accelerated the release of DOX and TAIII from liposomes at tumor site, and cell permeability of TAIII enhanced uptake of DOX in HCC cells. Conclusion The innovative application of TAIII working as bilayer stabilizer and chemotherapeutic drug affords a stable multifunctional liposomal delivery system for synergistic therapy against HCC, which may be referred for the development of other types of saponins with similar property.
Collapse
Affiliation(s)
- Lijuan Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shengan Zhang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Min Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Lu Lu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yue Ding
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Ninghui Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yuan Zhao
- Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Sihan Xuchen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Nailian Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
15
|
Heng Y, Liang Y, Zhang J, Li L, Zhang W, Jiang Y, Wang S, Jia L. Camptothecin Inhibits Neddylation to Activate the Protective Autophagy Through NF- κB/AMPK/mTOR/ULK1 Axis in Human Esophageal Cancer Cells. Front Oncol 2021; 11:671180. [PMID: 33898327 PMCID: PMC8061413 DOI: 10.3389/fonc.2021.671180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
The neddylation pathway is overactivated in esophageal cancer. Our previous studies indicated that inactivation of neddylation by the NAE inhibitor induced apoptosis and autophagy in cancer cells. Camptothecin (CPT), a well-known anticancer agent, could induce apoptosis and autophagy in cancer cells. However, whether CPT could affect the neddylation pathway and the molecular mechanisms of CPT-induced autophagy in esophageal cancer remains elusive. We found that CPT induced apoptosis and autophagy in esophageal cancer. Mechanistically, CPT inhibited the activity of neddylation and induced the accumulation of p-IkBa to block NF-κB pathway. Furthermore, CPT induced the generation of ROS to modulate the AMPK/mTOR/ULK1 axis to finally promote protective autophagy. In our study, we elucidate a novel mechanism of the NF-κB/AMPK/mTOR/ULK1 pathway in CPT-induced protective autophagy in esophageal cancer cells, which provides a sound rationale for combinational anti-ESCC therapy with CPT and inhibition AMPK/ULK1 pathway.
Collapse
Affiliation(s)
- Yongqing Heng
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yupei Liang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junqian Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihui Li
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjuan Zhang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yanyu Jiang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiwen Wang
- Department of Laboratory Medicine, Huadong Hospital, Affiliated to Fudan University, Shanghai, China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Tian JY, Chi CL, Bian G, Xing D, Guo FJ, Wang XQ. PSMA conjugated combinatorial liposomal formulation encapsulating genistein and plumbagin to induce apoptosis in prostate cancer cells. Colloids Surf B Biointerfaces 2021; 203:111723. [PMID: 33839474 DOI: 10.1016/j.colsurfb.2021.111723] [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: 12/08/2020] [Revised: 02/27/2021] [Accepted: 03/24/2021] [Indexed: 01/27/2023]
Abstract
Although the biomedical sciences have achieved tremendous success in developing novel approaches to managing prostate cancer, this disease remains one of the major health concerns among men worldwide. Liposomal formulations of single drugs have shown promising results in cancer treatment; however, the use of multi drugs has shown a better therapeutic index than individual drugs. The identification of cancer-specific receptors has added value to design targeted drug delivering nanocarriers. We have developed genistein and plumbagin co-encapsulating liposomes (∼120 nm) with PSMA specific antibodies to target prostate cancer cells selectively in this work. These liposomes showed >90 % decrease in PSMA expressing prostate cancer cell proliferation without any appreciable toxicity to healthy cells and human red blood cells. Release of plumbagin and genistein was found to decrease the expression of PI3/AKT3 signaling proteins and Glut-1 receptors (inhibited glucose uptake and metabolism), respectively. The decrease in migration potential of cells and induced apoptosis established the observed anti-proliferative effect in prostate cancer cell lines. The discussed strategy of developing novel, non-toxic, and PSMA specific antibody conjugated liposomes carrying genistein and plumbagin drugs may also be used for encapsulating other drugs and inhibit the growth of different types of cancers.
Collapse
Affiliation(s)
- Jing-Yan Tian
- Department of Urology, Second Division of The First Hospital of Jilin University, 3302 Jilin Rd, Changchun, 130031, Jilin, People's Republic of China
| | - Chang-Liang Chi
- Department of Urology, Second Division of The First Hospital of Jilin University, 3302 Jilin Rd, Changchun, 130031, Jilin, People's Republic of China
| | - Ge Bian
- Department of Urology, Second Division of The First Hospital of Jilin University, 3302 Jilin Rd, Changchun, 130031, Jilin, People's Republic of China
| | - Dong Xing
- Department of Urology, Second Division of The First Hospital of Jilin University, 3302 Jilin Rd, Changchun, 130031, Jilin, People's Republic of China
| | - Feng-Jun Guo
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, 218 Ziqiang Rd, Changchun, 130041, Jilin, People's Republic of China.
| | - Xiao-Qing Wang
- Department of Urology, Second Division of The First Hospital of Jilin University, 3302 Jilin Rd, Changchun, 130031, Jilin, People's Republic of China.
| |
Collapse
|
17
|
Liposomes co-encapsulating doxorubicin and glucoevatromonoside derivative induce synergic cytotoxic response against breast cancer cell lines. Biomed Pharmacother 2021; 136:111123. [PMID: 33486211 DOI: 10.1016/j.biopha.2020.111123] [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: 09/20/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 11/23/2022] Open
Abstract
Cancer is one of the main causes of death in the world and thus a global public health problem. Among the treatments available for cancer are surgery, radiotherapy, and chemotherapy. Currently, there is increased interest in the combination of two or more antitumor agents to achieve a synergistic effect in cancer therapy. Doxorubicin (DOX), a chemotherapeutic which has a potent antineoplastic action, has been used in the treatment of various tumors. However, the use of DOX is limited, mainly due to the cardiotoxicity. Therefore, nanostructured systems, such as liposomes, have been developed to carry this drug and target the tumor region, since tumor tissues present enhanced permeability and retention for nanosystems. Cardiac glycosides, such as digitoxin, have recently shown great antitumor potential despite the low therapeutic index which may limit their use. Furthermore, some compounds of this class have low water solubility, which makes their in vivo administration difficult. In this context, liposomes represent a valid strategy to carry simultaneously antitumor drugs allowing their intravenous administration. In this study, liposomes loaded with glucoevatromonoside containing peracetylated glucose hydroxyl groups (GEVPG) and DOX at molar ratio of 1:1 (SpHL-GEVPG:DOX 1:1) were developed, and their chemical and physicochemical properties were evaluated. This formulation presented a combination index (CI) lower than 1 at inhibitory concentration of 90 % growth (IC90) for three human breast tumor lines evaluated (0.52 ± 0.39 for MDA-MB-231, 0.19 ± 0.13 for MCF-7, and 0.99 ± 0.09 for SKBR-3). These results indicate a synergistic cytotoxic effect of the GEVPG and DOX combination encapsulated in liposomes. In addition, SpHL-GEVPG:DOX 1:1 presented selectivity towards these cancer cells. Long-term in vitro cytotoxicity studies demonstrated that MDA-MB-231 surviving cells after treatment with SpHL-GEVPG:DOX 1:1 did not recover proliferation capacity after 21 d. From the studies of cell cycle and death pathway evaluation, it was observed that SpHL-GEVPG:DOX 1:1 arrested the cell cycle in the G2/M phase and similarly induced apoptosis and necrosis. However, SpHL-GEVPG:DOX at molar ratio of 1:1 showed lower induction of both apoptotic and necrotic pathways compared to free DOX and SpHL-DOX, suggesting that the mechanism of death involved may not be related to necrosis or apoptosis. Lastly, SpHL-GEVPG:DOX 1:1 showed a good storage stability for 90 d at 4 °C. Therefore, the results of the present work indicate the potential use of SpHL-GEVPG:DOX 1:1 as a new anticancer formulation.
Collapse
|
18
|
Nanotechnology in ovarian cancer: Diagnosis and treatment. Life Sci 2020; 266:118914. [PMID: 33340527 DOI: 10.1016/j.lfs.2020.118914] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/05/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022]
Abstract
To overcome the drawbacks of conventional delivery, this review spotlights a number of nanoscale drug delivery systems, including nanoparticles, liposomes, nano micelles, branched dendrimers, nanocapsules, and nanostructured lipid formulations for the targeted therapy of ovarian cancer. These nanoformulations offer numerous advantages to promote therapeutic drug delivery such as nontoxicity, biocompatibility, good biodegradability, increased therapeutic impact than free drugs, and non-inflammatory effects. Importantly, the development of specific ligands functionalized nanoformulations enable preferential targeting of ovarian tumors and eventually amplify the therapeutic potential compared to nonfunctionalized counterparts. Ovarian cancer is typically identified by biomarker assessment such as CA125, HE4, Mucin 1, and prostatic. There is, nevertheless, a tremendous demand for less costly, faster, and compact medical tools, both for timely detection and ovarian cancer control. This paper explored multiple types of tumor marker-based on nanomaterial biosensors. Initially, we mention different forms of ovarian cancer biomarkers involving CA125, human epididymis protein 4 (HE4), mucin 1 (MUC1), and prostate. It is accompanied by a brief description of new nanotechnology methods for diagnosis. Nanobiosensors for evaluating ovarian cancer biomarkers can be categorized based on electrochemical, optical, paper-based, giant magnetoresistive, and lab-on-a-chip devices.
Collapse
|
19
|
Design principles of drug combinations for chemotherapy. J Control Release 2020; 323:36-46. [DOI: 10.1016/j.jconrel.2020.04.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022]
|
20
|
Gupta S, Pathak Y, Gupta MK, Vyas SP. Nanoscale drug delivery strategies for therapy of ovarian cancer: conventional vs targeted. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:4066-4088. [PMID: 31625408 DOI: 10.1080/21691401.2019.1677680] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ovarian cancer is the second most common gynaecological malignancy. It usually occurs in women older than 50 years, and because 75% of cases are diagnosed at stage III or IV it is associated with poor diagnosis. Despite the chemosensitivity of intraperitoneal chemotherapy, the majority of patients is relapsed and eventually dies. In addition to the challenge of early detection, its treatment presents several challenges like the route of administration, resistance to therapy with recurrence and specific targeting of cancer to reduce cytotoxicity and side effects. In ovarian cancer therapy, nanocarriers help overcome problems of poor aqueous solubility of chemotherapeutic drugs and enhance their delivery to the tumour sites either by passive or active targeting, and thus reducing adverse side effects to the healthy tissues. Moreover, the bioavailability to the tumour site is increased by the enhanced permeability and retention (EPR) mechanism. The present review aims to describe the current conventional treatment with special reference to passively and actively targeted drug delivery systems (DDSs) towards specific receptors designed against ovarian cancer to overcome the drawbacks of conventional delivery. Conclusively, targeted nanocarriers would optimise the intra-tumour distribution, followed by drug delivery into the intracellular compartment. These features may contribute to greater therapeutic effect.
Collapse
Affiliation(s)
- Swati Gupta
- Amity Institute of Pharmacy, Amity University Uttar Pradesh , Noida , India
| | - Yashwant Pathak
- College of Pharmacy, University of South Florida Health , Tampa , FL , USA.,Faculty of Pharmacy, University of Airlangga , Surabaya , Indonesia
| | - Manish K Gupta
- TERI-Deakin Nanobiotechnology Centre, The Energy and Resources Institute (TERI), Gual Pahari, TERI Gram , Gurugram , India
| | - Suresh P Vyas
- Department of Pharmaceutical Sciences, Dr H.S. Gour University , Sagar , India
| |
Collapse
|
21
|
Zhao Y, Fletcher NL, Gemmell A, Houston ZH, Howard CB, Blakey I, Liu T, Thurecht KJ. Investigation of the Therapeutic Potential of a Synergistic Delivery System through Dual Controlled Release of Camptothecin–Doxorubicin. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yongmei Zhao
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
- School of PharmacyNantong University Nantong 226019 China
| | - Nicholas L. Fletcher
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
| | - Anna Gemmell
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
| | - Zachary H. Houston
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
| | - Christopher B. Howard
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
| | - Idriss Blakey
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
| | - Tianqing Liu
- QIMR Berghofer Medical Research Institute 300 Herston Road Brisbane QLD 4006 Australia
| | - Kristofer J. Thurecht
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and ARC Training Centre in Biomedical Imaging TechnologyThe University of Queensland Brisbane QLD 4072 Australia
| |
Collapse
|
22
|
Taneja N, Gota V, Gurjar M, Singh KK. Development and validation of high-performance liquid chromatographic method for quantification of Irinotecan and its active metabolite SN-38 in colon tumor bearing NOD/SCID mice plasma samples: application to pharmacokinetic study. ACTA CHROMATOGR 2019. [DOI: 10.1556/1326.2018.00370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Neetika Taneja
- C. U. Shah College of Pharmacy, S.N. D. T. Women's University, Santacruz (W), Mumbai 400049, India
| | - Vikram Gota
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India
| | - Murari Gurjar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India
| | - Kamalinder K. Singh
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, United Kingdom
| |
Collapse
|
23
|
Navya PN, Kaphle A, Srinivas SP, Bhargava SK, Rotello VM, Daima HK. Current trends and challenges in cancer management and therapy using designer nanomaterials. NANO CONVERGENCE 2019; 6:23. [PMID: 31304563 PMCID: PMC6626766 DOI: 10.1186/s40580-019-0193-2] [Citation(s) in RCA: 344] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 06/17/2019] [Indexed: 05/06/2023]
Abstract
Nanotechnology has the potential to circumvent several drawbacks of conventional therapeutic formulations. In fact, significant strides have been made towards the application of engineered nanomaterials for the treatment of cancer with high specificity, sensitivity and efficacy. Tailor-made nanomaterials functionalized with specific ligands can target cancer cells in a predictable manner and deliver encapsulated payloads effectively. Moreover, nanomaterials can also be designed for increased drug loading, improved half-life in the body, controlled release, and selective distribution by modifying their composition, size, morphology, and surface chemistry. To date, polymeric nanomaterials, metallic nanoparticles, carbon-based materials, liposomes, and dendrimers have been developed as smart drug delivery systems for cancer treatment, demonstrating enhanced pharmacokinetic and pharmacodynamic profiles over conventional formulations due to their nanoscale size and unique physicochemical characteristics. The data present in the literature suggest that nanotechnology will provide next-generation platforms for cancer management and anticancer therapy. Therefore, in this critical review, we summarize a range of nanomaterials which are currently being employed for anticancer therapies and discuss the fundamental role of their physicochemical properties in cancer management. We further elaborate on the topical progress made to date toward nanomaterial engineering for cancer therapy, including current strategies for drug targeting and release for efficient cancer administration. We also discuss issues of nanotoxicity, which is an often-neglected feature of nanotechnology. Finally, we attempt to summarize the current challenges in nanotherapeutics and provide an outlook on the future of this important field.
Collapse
Affiliation(s)
- P N Navya
- Nano-Bio Interfacial Research Laboratory (NBIRL), Department of Biotechnology, Siddaganga Institute of Technology, Tumkur, Karnataka, 572103, India.
- Department of Biotechnology, Bannari Amman Institute of Technology, Sathyamangalam, Erode, Tamil Nadu, 638401, India.
| | - Anubhav Kaphle
- Melbourne Integrative Genomics, School of BioSciences/School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - S P Srinivas
- School of Optometry, Indiana University, Bloomington, Indiana, 47405, USA
| | - Suresh Kumar Bhargava
- Centre for Advanced Materials and Industrial Chemistry, School of Science, RMIT University, Melbourne, VIC, 3001, Australia
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts (UMass) Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Hemant Kumar Daima
- Nano-Bio Interfacial Research Laboratory (NBIRL), Department of Biotechnology, Siddaganga Institute of Technology, Tumkur, Karnataka, 572103, India.
- Centre for Advanced Materials and Industrial Chemistry, School of Science, RMIT University, Melbourne, VIC, 3001, Australia.
- Amity Institute of Biotechnology, Amity University Rajasthan, Kant Kalwar, NH-11C, Jaipur-Delhi Highway, Jaipur, Rajasthan, 303002, India.
| |
Collapse
|
24
|
Mang D, Zhang S, Wu X, Hu X, Mochizuki T, Li G, Zhang Y. Enzyme-mediated dual-targeted-assembly realizes a synergistic anticancer effect. Chem Commun (Camb) 2019; 55:6126-6129. [PMID: 31070616 DOI: 10.1039/c9cc02715g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We designed and synthesized homochiral-peptide-based boron diketonate complexes. Co-administration of the two stereoisomers in cancer cells led to molecular assembly targeting both the plasma membrane and the lysosomes mediated via membrane-bonded enzymes. The dual-targeted-assembly generates a synergistic anticancer effect with amplified cancer spheroid toxicity and enhanced inhibition efficacy on cancer cell migration.
Collapse
Affiliation(s)
- Dingze Mang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan.
| | - Shijin Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan.
| | - Xia Wu
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan.
| | - Xunwu Hu
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan.
| | - Toshiaki Mochizuki
- Imaging Section, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan
| | - Guanying Li
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan.
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan.
| |
Collapse
|
25
|
Franco MS, Oliveira MC. Liposomes Co- encapsulating Anticancer Drugs in Synergistic Ratios as an Approach to Promote Increased Efficacy and Greater Safety. Anticancer Agents Med Chem 2019; 19:17-28. [DOI: 10.2174/1871520618666180420170124] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 03/17/2018] [Accepted: 03/20/2018] [Indexed: 11/22/2022]
Abstract
The era of chemotherapy began in the 1940s, but it was in the 1960s that it was seen as really
promising when the first patients with childhood acute lymphoblastic leukemia were cured with combination
chemotherapy. Today, it is known that due to resistance to single agents, combination therapy is essential for
tumor eradication and cure. In the last decade, studies have shown that anticancer drug combinations can act
synergistically or antagonistically against tumor cells in vitro, depending on the ratios of the individual drugs
forming the combination. From this observation and facing the possibility of maintaining the in vivo synergistic
ratio of combinations came the idea of co-encapsulating anticancer agents in nanosystems. In vivo studies validated
this idea by showing that the co-encapsulation of anticancer agents in liposomes allows the maintenance
of drug ratios in the plasma and the delivery of fixed drug ratios directly to tumor tissue, leading to a better
efficacy compared to the administration of the free drugs combination. Liposomes co-encapsulating
irinotecan/floxuridine are now in Phase II trial, and liposomes co-encapsulating cytarabine/daunorubicin were
recently approved by the FDA for treatment of patients with acute myeloid leukemia.
Collapse
Affiliation(s)
- Marina S. Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Mônica C. Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
26
|
Franco MS, Roque MC, Oliveira MC. Short and Long-Term Effects of the Exposure of Breast Cancer Cell Lines to Different Ratios of Free or Co-Encapsulated Liposomal Paclitaxel and Doxorubicin. Pharmaceutics 2019; 11:pharmaceutics11040178. [PMID: 30979090 PMCID: PMC6523953 DOI: 10.3390/pharmaceutics11040178] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 03/29/2019] [Accepted: 03/31/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Associating paclitaxel (PTX) to doxorubicin (DXR) is one of the main chemotherapy strategies for breast cancer (BC) management. Protocols currently available consist in administering both drugs on their maximum tolerated dose, not taking into account the possible differences in efficacy due to their combination ratio. In the present study, the short and long-term cytotoxic effects as well as migratory effects of PTX, DXR, and its combinations at 10:1; 1:1 and 1:10 PTX:DXR molar ratios either free or co-encapsulated in liposomes were evaluated against three human BC cell lines (MDA-MB-231, MCF-7, and SKBR-3). Method: The MTT assay was used to screen for synergy or antagonism between PTX and DXR and the combination index value was calculated using the CalcuSyn software. Nuclear morphological alterations were evaluated by staining the cells with Hoescht 33342. The investigation of senescence and clonogenicity of BC cell lines exposed to different treatments was also studied. In addition, the ability of these cells to migrate was assessed. Results: Taken together, the results presented herein allow us to suggest that there is no benefit in enhancing the PTX concentration above that of DXR in the combination for any of the three cell lines tested. Conclusion: The developed liposomes co-encapsulating PTX and DXR in different molar ratios retained the biological properties of the mixture of free drugs and are valuable for planning new therapeutic strategies.
Collapse
Affiliation(s)
- Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Marjorie Coimbra Roque
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
27
|
Rawal S, Patel MM. Threatening cancer with nanoparticle aided combination oncotherapy. J Control Release 2019; 301:76-109. [PMID: 30890445 DOI: 10.1016/j.jconrel.2019.03.015] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022]
Abstract
Employing combination therapy has become obligatory in cancer cases exhibiting high tumor load, chemoresistant tumor population, and advanced disease stages. Realization of this fact has now led many of the combination oncotherapies to become an integral part of anticancer regimens. Combination oncotherapy may encompass a combination of anticancer agents belonging to a similar therapeutic category or that of different therapeutic categories (e.g. chemotherapy + gene therapy). Differences in the physicochemical properties, pharmacokinetics and biodistribution pattern of different payloads are the major constraints that are faced by combination chemotherapy. Concordant efforts in the field of nanotechnology and oncology have emerged with several approaches to solve the major issues encountered by combination therapy. Unique colloidal behaviors of various types of nanoparticles and differential targeting strategies have accorded an unprecedented ability to optimize combination oncotherapeutic delivery. Nanocarrier based delivery of the various types of payloads such as chemotherapeutic agents and other anticancer therapeutics such as small interfering ribonucleic acid (siRNA), chemosensitizers, radiosensitizers, and antiangiogenic agents have been addressed in the present review. Various nano-delivery systems like liposomes, polymeric nanoparticles, polymerosomes, dendrimers, micelles, lipid based nanoparticles, prodrug based nanocarriers, polymer-drug conjugates, polymer-lipid hybrid nanoparticles, carbon nanotubes, nanosponges, supramolecular nanocarriers and inorganic nanoparticles (gold nanoparticles, silver nanoparticles, magnetic nanoparticles and mesoporous silica based nanoparticles) that have been extensively explored for the formulation of multidrug delivery is an imperative part of discussion in the review. The present review features the outweighing benefits of combination therapy over mono-oncotherapy and discusses several existent nanoformulation strategies that facilitate a successful combination oncotherapy. Several obstacles that may impede in transforming nanotechnology-based combination oncotherapy from bench to bedside, and challenges associated therein have also been discussed in the present review.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India
| | - Mayur M Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India.
| |
Collapse
|
28
|
Liu J, Chi D, Pan S, Zhao L, Wang X, Wang D, Wang Y. Effective co-encapsulation of doxorubicin and irinotecan for synergistic therapy using liposomes prepared with triethylammonium sucrose octasulfate as drug trapping agent. Int J Pharm 2019; 557:264-272. [DOI: 10.1016/j.ijpharm.2018.12.072] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/07/2018] [Accepted: 12/13/2018] [Indexed: 11/27/2022]
|
29
|
Investigation of the antitumor activity and toxicity of long-circulating and fusogenic liposomes co-encapsulating paclitaxel and doxorubicin in a murine breast cancer animal model. Biomed Pharmacother 2018; 109:1728-1739. [PMID: 30551427 DOI: 10.1016/j.biopha.2018.11.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 11/23/2022] Open
Abstract
To associate paclitaxel (PTX) with doxorubicin (DXR) is one of the main chemotherapy strategies for breast cancer (BC) management. Despite the high response rates for this combination, it presents a cardiotoxic synergism, attributed to pharmacokinetic interactions between PTX and both DXR and its metabolite, doxorubicinol. One of the main strategies to minimize the cardiotoxicity of the combination is to extend the interval of time between DXR and PTX administration. However, it has been previously suggested that their co-administration leads to better efficacy compared to their sequential administration. In the present study, we investigated different molar ratio combinations of PTX:DXR (10:1; 1:1, and 1:10) against the 4T1 murine breast cancer cell line and concluded that there is no benefit of enhancing PTX concentration above that of DXR on the combination. Therefore, we obtained a long-circulating and fusogenic liposomal formulation co-encapsulating PTX and DXR (LCFL-PTX/DXR) at a molar ratio of 1:10, respectively, which maintained the in vitro biological activity of the combination. This formulation was investigated for its antitumor activity and toxicity in Balb/c mice bearing 4T1 breast tumor, and compared to treatments with free PTX, free DXR, and the mixture of free PTX:DXR at 1:10 molar ratio. The higher tumor inhibition ratios were observed for the treatments with free and co-encapsulated PTX:DXR in liposomes (66.87 and 66.52%, respectively, P>0.05) as compared to the control. The great advantage of the treatment with LCFL-PTX/DXR was its improved cardiac toxicity profile. While degeneration was observed in the hearts of all animals treated with the free PTX:DXR combination, no signs of cardiac toxicity were observed for animals treated with the LCFL-PTX/DXR. Thus, LCFL-PTX/DXR enables the co-administration of PTX and DXR, and might be considered valuable for breast cancer management.
Collapse
|
30
|
Chang CE, Hsieh CM, Chen LC, Su CY, Liu DZ, Jhan HJ, Ho HO, Sheu MT. Novel application of pluronic lecithin organogels (PLOs) for local delivery of synergistic combination of docetaxel and cisplatin to improve therapeutic efficacy against ovarian cancer. Drug Deliv 2018; 25:632-643. [PMID: 29463123 PMCID: PMC6058476 DOI: 10.1080/10717544.2018.1440444] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The synergistic combination of docetaxel (DTX) and cisplatin (CIS) by local drug delivery with a pluronic lecithin organogel (PLO) to facilitate high drug concentrations at tumor sites and less nonspecific distribution to normal organs is thought to be beneficial in chemotherapy. In this study, using Capryol-90 (C90) with the addition of lecithin as the oil phase was developed to carry DTX, which was then incorporated into a PLO-containing CIS to formulate a dual-drug injectable PLO for local delivery. An optimal PLO composite, P13L0.15O1.5, composed of PF127:lecithin:C90 at a 13:0.15:1.5 weight ratio was obtained. The sol-gel transition temperature of P13L0.15O1.5 was found to be 33 °C. Tumor inhibition studies illustrated that DTX/CIS-loaded P13L0.15O1.5 could efficiently suppress tumor growth by both intratumoral and peritumoral injections in SKOV-3 xenograft mouse model. Pharmacokinetic studies showed that subcutaneous administration of P13L0.15O1.5 was able to sustain the release of DTX and CIS leading to their slow absorption into the systemic circulation resulting in lower area under the plasma concentration curve at 0-72 h (AUC0-72) and maximum concentration (Cmax) values but longer half-life (T1/2) and mean residence time (MRT) values. An in vivo biodistribution study showed lower DTX and CIS concentrations in organs compared to other treatment groups after IT administration of the dual drug-loaded P13L0.15O1.5. It was concluded that the local co-delivery of DTX and CIS by PLOs may be a promising and effective platform for local anticancer drug delivery with minimal systemic toxicities.
Collapse
Affiliation(s)
- Chia-En Chang
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Chien-Ming Hsieh
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ling-Chun Chen
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC.,b Department of Biotechnology and Pharmaceutical Technology , Yuanpei University of Medical Technology , Hsinchu , Taiwan, ROC
| | - Chia-Yu Su
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Der-Zen Liu
- c Graduate Institute of Biomedical Materials and Engineering , Taipei Medical University , Taipei , Taiwan, ROC
| | - Hua-Jing Jhan
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Hsiu-O Ho
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ming-Thau Sheu
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC.,d Clinical Research Center and Traditional Herbal Medicine Research Center , Taipei Medical University Hospital , Taipei , Taiwan, ROC
| |
Collapse
|
31
|
Franco MS, Oliveira MC. Ratiometric drug delivery using non-liposomal nanocarriers as an approach to increase efficacy and safety of combination chemotherapy. Biomed Pharmacother 2017; 96:584-595. [PMID: 29035823 DOI: 10.1016/j.biopha.2017.10.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 10/18/2022] Open
Abstract
The observation that different drug ratios of the same drug combination can lead to synergistic or antagonistic effects when tested against the same cancer cell line in vitro gave rise to a new trend, the ratiometric delivery. This strategy consists of co-encapsulating a specific synergistic ratio of a drug combination into a nanocarrier so that synergism observed in vitro will be faithfully translated to in vivo, optimizing combination therapy. In this review we focus on how to quantify synergism in vitro, followed by how this affected the evolution of nanocarriers culminating in the ratiometric delivery, and finally we summarize the results of the non-liposomal formulations that were built upon this concept.
Collapse
Affiliation(s)
- Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
32
|
|
33
|
Shah VM, Nguyen DX, Alfatease A, Bracha S, Alani AW. Characterization of pegylated and non-pegylated liposomal formulation for the delivery of hypoxia activated vinblastine-N-oxide for the treatment of solid tumors. J Control Release 2017; 253:37-45. [PMID: 28302582 DOI: 10.1016/j.jconrel.2017.03.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/24/2017] [Accepted: 03/13/2017] [Indexed: 12/27/2022]
Abstract
Solid tumors often contain hypoxic regions which are resistant to standard chemotherapy and radiotherapy. We have developed a liposomal delivery system for a prodrug of vinblastine (CPD100) which converts to the parent compound only in the presence of lower oxygen levels. As a part of this work we have developed and optimized two formulations of CPD100: one composed of sphingomyelin/cholesterol (55/45; mol/mol) (CPD100Li) and the other composed of sphingomyelin/cholesterol/PEG (55/40/5; mol/mol) (CPD100 PEGLi). We evaluated the antiproliferative effect of CPD100 and the two formulations against A549 non-small lung cancer cell. A549 cell line showed to be sensitive to CPD100 and the two formulations displayed a higher hypoxic: air cytotoxicity ratio compared to the pro-drug. CPD100 elimination from the circulation after injection in mouse was characterized by a very short circulation time (~0.44h), lower area under the curve (AUC) (33μgh/mL) and high clearance (916mL/h/kg) and lower volume of distribution (17.4mL/kg).Total drug elimination from the circulation after the administration of liposomal formulation was characterized by prolonged circulation time (5.5h) along with increase in the AUC (56μgh/mL) for CPD100 Li and (9.5h) with AUC (170μgh/mL) for CPD100PEGLi. This was observed along with increase in volume of distribution and decrease in clearance for the liposomes. The systemic exposure of the free drug was much lower than that achieved with the liposomes. When evaluated for the efficacy in A549 xenograft model in mice, both the liposomes demonstrated excellent tumor suppression and reduction for 3months. The blood chemistry panel and the comprehensive blood analysis showed no increase or decrease in the markers and blood count. In summary, the pharmacokinetic analysis along with the efficacy data emphasis on how the delivery vehicle modifies and enhances the accumulation of the drug and at the same time the increased systemic exposure is not related to toxicity.
Collapse
Affiliation(s)
- Vidhi M Shah
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University/OHSU, Portland, OR 97201, United States
| | - Duc X Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University/OHSU, Portland, OR 97201, United States
| | - Adel Alfatease
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University/OHSU, Portland, OR 97201, United States
| | - Shay Bracha
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States
| | - Adam Wg Alani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University/OHSU, Portland, OR 97201, United States.
| |
Collapse
|
34
|
Trusova VM, Deligeorgiev T, Gorbenko G. Liposomal Co-Encapsulation of Two Novel Europium Complexes and Doxorubicin: Fluorescence Study. J Fluoresc 2017; 27:1359-1363. [PMID: 28283899 DOI: 10.1007/s10895-017-2070-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/02/2017] [Indexed: 12/19/2022]
Abstract
The present study was undertaken to design the novel liposomal drug formulation containing doxorubicin and europium coordination complexes. It was shown that co-encapsulation of the drugs facilitates the partitioning and permeation of lanthanides into the lipid bilayer. The obtained results suggest that new drug platform may have potential application in the design of novel antitumor agents.
Collapse
Affiliation(s)
- Valeriya M Trusova
- Department of Nuclear and Medical Physics, V.N. Karazin Kharkov National University, 4 Svobody Sq, Kharkov, 61072, Ukraine. .,, 19-32 Geroyev Truda Str, Kharkov, 61072, Ukraine.
| | - Todor Deligeorgiev
- Department of Applied Organic Chemistry, Faculty of Chemistry, University of Sofia, 1164, Sofia, Bulgaria
| | - Galyna Gorbenko
- Department of Nuclear and Medical Physics, V.N. Karazin Kharkov National University, 4 Svobody Sq, Kharkov, 61072, Ukraine
| |
Collapse
|
35
|
Zununi Vahed S, Salehi R, Davaran S, Sharifi S. Liposome-based drug co-delivery systems in cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:1327-1341. [DOI: 10.1016/j.msec.2016.11.073] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
|
36
|
Ingebrigtsen SG, Škalko-Basnet N, de Albuquerque Cavalcanti Jacobsen C, Holsæter AM. Successful co-encapsulation of benzoyl peroxide and chloramphenicol in liposomes by a novel manufacturing method - dual asymmetric centrifugation. Eur J Pharm Sci 2017; 97:192-199. [DOI: 10.1016/j.ejps.2016.11.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022]
|
37
|
Tuo J, Xie Y, Song J, Chen Y, Guo Q, Liu X, Ni X, Xu D, Huang H, Yin S, Zhu W, Wu J, Hu H. Development of a novel berberine-mediated mitochondria-targeting nano-platform for drug-resistant cancer therapy. J Mater Chem B 2016; 4:6856-6864. [PMID: 32263579 DOI: 10.1039/c6tb01730d] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent studies have shown that targeting doxorubicin to mitochondria of tumor cells can bypass the multi-drug resistance problem and inhibit tumor growth. We previously discovered that the C-9th and C-13th position-alkylated berberine derivatives possess improved mitochondria-targeting activity compared to berberine. Therefore, we hypothesize that these alkylated berberine derivatives could be utilized as potential mitochondrial-targeting ligands by inserting the alkyl chain into the liposomal bilayer membrane during the preparation of liposomes. In this research, a berberine derivate (a 16-carbon aliphatic chain was introduced to the C-9th of berberine, 9-C16 berberine) was employed to prepare mitochondria-targeting doxorubicin-loaded folic acid-conjugated polyethylene glycol(PEGylated) liposomes (MT-FOL-PLS). The results of in vitro cytotoxicity and apoptosis-inducing studies revealed that MT-FOL-PLS showed the strongest cytotoxicity and apoptosis-inducing effects in drug resistant MCF-7/adr cells in comparison with free doxorubicin and regular liposomal doxorubicin. MT-FOL-PLS enhanced cellular uptake of doxorubicin up to 15-fold compared to free doxorubicin, and targeted doxorubicin to mitochondria. In vivo and ex vivo drug distribution studies showed that MT-FOL-PLS increased the drug distribution in tumor and the administration of MT-FOL-PLS to resistant MCF-7/adr cell mouse xenografts stopped tumor growth. Our results confirmed that alkylated berberines can be exploited as mitochondrial-targeting ligands to overcome cancer multi-drug resistance, further advancing the research on active targeting of liposome delivery systems in the treatment of resistant cancer.
Collapse
Affiliation(s)
- Jue Tuo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P. R. China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Wehbe M, Chernov L, Chen K, Bally MB. PRCosomes: pretty reactive complexes formed in liposomes. J Drug Target 2016; 24:787-796. [DOI: 10.1080/1061186x.2016.1186169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lina Chernov
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kent Chen
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
- Center for Drug Research and Development, Vancouver, BC, Canada
| |
Collapse
|
40
|
Zylberberg C, Matosevic S. Pharmaceutical liposomal drug delivery: a review of new delivery systems and a look at the regulatory landscape. Drug Deliv 2016; 23:3319-3329. [PMID: 27145899 DOI: 10.1080/10717544.2016.1177136] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Liposomes were the first nanoscale drug to be approved for clinical use in 1995. Since then, the technology has grown considerably, and pioneering recent work in liposome-based delivery systems has brought about remarkable developments with significant clinical implications. This includes long-circulating liposomes, stimuli-responsive liposomes, nebulized liposomes, elastic liposomes for topical, oral and transdermal delivery and covalent lipid-drug complexes for improved drug plasma membrane crossing and targeting to specific organelles. While the regulatory bodies' opinion on liposomes is well-documented, current guidance that address new delivery systems are not. This review describes, in depth, the current state-of-the-art of these new liposomal delivery systems and provides a critical overview of the current regulatory landscape surrounding commercialization efforts of higher-level complexity systems, the expected requirements and the hurdles faced by companies seeking to bring novel liposome-based systems for clinical use to market.
Collapse
|
41
|
Camacho KM, Menegatti S, Mitragotri S. Low-molecular-weight polymer-drug conjugates for synergistic anticancer activity of camptothecin and doxorubicin combinations. Nanomedicine (Lond) 2016; 11:1139-51. [PMID: 27079141 DOI: 10.2217/nnm.16.33] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND High-molecular-weight (MW) polymers (>50,000 Da) can be conjugated to chemotherapy drugs in order to improve their tumor accumulation, while low MW polymers ≤10,000 Da are often overlooked due to faster plasma clearance. Small polymers, however, may facilitate deeper tumor penetration. MATERIALS & METHODS Here, we investigate the anticancer efficacy of 10 kDa hyaluronic acid or poly(vinyl alcohol) conjugated to synergistic combinations of camptothecin and doxorubicin, with emphasis on chemical linker impacts. RESULTS Our results emphasize drug hydrolyzability for synergy preservation, and also demonstrate superior cancer cell inhibition with low MW polymer-drug conjugates. CONCLUSION This study shows the high therapeutic potential of low MW polymer-drug conjugates for polychemotherapy delivery, and provides further insight into the development of polymer-drug therapeutics.
Collapse
Affiliation(s)
- Kathryn M Camacho
- Department of Chemical Engineering, Center for Bioengineering, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| | - Stefano Menegatti
- Department of Chemical & Biomolecular Engineering, Department of Biomedical Engineering, Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695, USA
| | - Samir Mitragotri
- Department of Chemical Engineering, Center for Bioengineering, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| |
Collapse
|
42
|
Hu T, Cao H, Yang C, Zhang L, Jiang X, Gao X, Yang F, He G, Song X, Tong A, Guo G, Gong C, Li R, Zhang X, Wang X, Zheng Y. LHD-Modified Mechanism-Based Liposome Coencapsulation of Mitoxantrone and Prednisolone Using Novel Lipid Bilayer Fusion for Tissue-Specific Colocalization and Synergistic Antitumor Effects. ACS APPLIED MATERIALS & INTERFACES 2016; 8:6586-601. [PMID: 26907854 DOI: 10.1021/acsami.5b10598] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Coencapsulation liposomes are of interest to researchers because they maximize the synergistic effect of loaded drugs. A combination regimen of mitoxantrone (MTO) and prednisolone (PLP) has been ideal for tumor therapy. MTO and PLP offer synergistic antitumor effects confirmed by several experiments in this research. The deduced synergistic mechanism is regulation of Akt signaling pathway including the targets of p-Akt, p-GSK-3β, p-s6 ribosomal protein, and p-AMPK by MTO reactivating PLP-induced apoptosis. The liposome fusion method is adopted to create coencapsulation liposomes (PLP-MTO-YM). Low molecular weight heparin-sodium deoxycholate conjugate (LHD) then is used as a targeting ligand to prove target binding and inhibition of angiogenesis. LHD-modified liposomes (PLP-MTO-HM) have a high entrapment efficiency around 95% for both MTO and PLP. DSC results indicate that both drugs interacted with liposomes to prevent drug leak during liposome fusion. DiD-C6-HM dyes colocalize well to tumor tissue, and coadministration of DiD-HM and C6-CM did not achieve dye colocalization until 24 h after administration. In both CT26 and B16F10 mouse model, PLP-MTO-HM shows a significantly higher tumor inhibition rate relative to the coadministration of MTO-HM and PLP-CM (p < 0.05 or p < 0.01). Thus, the coencapsulation system (PLP-MTO-HM) offers ideal antitumor effects relative to coadministration therapy due to enhanced synergistic effect, and this suggests a promising future for the tumor targeting vectors.
Collapse
Affiliation(s)
- Tingting Hu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Hua Cao
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Chengli Yang
- School of Pharmacy, Zunyi Medical University , 201#, Dalian Road, Zunyi, Guizhou 563000, People's Republic of China
| | - Lijing Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Xiaohua Jiang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Xiang Gao
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Fan Yang
- Department of Gynecology, West China Second University Hospital, Sichuan University , Chengdu 610041, People's Republic of China
| | - Gu He
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Xiangrong Song
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Aiping Tong
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Gang Guo
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Changyang Gong
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Rui Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Xiaoning Zhang
- Laboratory of Pharmaceutics, School of Medicine, Tsinghua University , 30#, Shuangqing Road, Haidian Dist, Beijing 100084, People's Republic of China
| | - Xinchun Wang
- School of Pharmacy, Shihezi University , No. 221, North Fourth Road, Shihezi, Xinjiang 832000, People's Republic of China
| | - Yu Zheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
43
|
Lu S, Xu L, Kang ET, Mahendran R, Chiong E, Neoh KG. Co-delivery of peptide-modified cisplatin and doxorubicin via mucoadhesive nanocapsules for potential synergistic intravesical chemotherapy of non-muscle-invasive bladder cancer. Eur J Pharm Sci 2016; 84:103-15. [PMID: 26780592 DOI: 10.1016/j.ejps.2016.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/11/2015] [Accepted: 01/13/2016] [Indexed: 11/25/2022]
Abstract
Synergistic effect against UMUC3 bladder cancer cells was demonstrated via a "two-in-one" combination of doxorubicin (Dox) and peptide-modified cisplatin (Pt-ALy) loaded in positively charged mucoadhesive chitosan-polymethacrylic acid (CM) nanocapsules. The in vitro killing efficacy of the dual drug-loaded nanocapsules (CM-Dox-PtALy) against UMUC3 cells after 4h- and 72h-treatment is much higher (with 5-16 times lower IC50) than either Dox- or Pt-ALy-loaded nanocapsules, resulting in combination indexes of much less than 1 (i.e. obvious synergism) at fractions of affected cells ranging from 0.2 to 0.8. The dose reduction index of Pt-ALy for 72h-treatment is higher than for 4h-treatment, suggesting that Dox in CM-Dox-PtALy played a more significant role in the synergy in the former. The drug-loaded CM nanocapsules are readily taken in by the cells as shown by flow cytometry, confocal laser scanning microscopy and inductively coupled plasma mass spectrometry. Microscopy observations indicate that CM nanocapsules attach strongly on the luminal surface of the bladder with no obvious damage of the urothelium, supporting our objective of prolonging the dwell time of the drug-loaded nanocapsules for intravesical applications. Our study indicates that the mucoadhesive CM-Dox-PtALy nanocapsules have a high drug loading and a sustained release profile, and thus, are promising for synergistic intravesical chemotherapy of non-muscle-invasive bladder cancers.
Collapse
Affiliation(s)
- Shengjie Lu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Kent Ridge, Singapore 117576, Singapore
| | - Liqun Xu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Kent Ridge, Singapore 117576, Singapore
| | - En Tang Kang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Kent Ridge, Singapore 117576, Singapore
| | - Ratha Mahendran
- Department of Surgery, National University of Singapore, Kent Ridge, Singapore 117576, Singapore
| | - Edmund Chiong
- Department of Surgery, National University of Singapore, Kent Ridge, Singapore 117576, Singapore
| | - Koon Gee Neoh
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Kent Ridge, Singapore 117576, Singapore.
| |
Collapse
|
44
|
Janni DS, Reddy UC, Saroj S, Muraleedharan KM. A modular approach towards drug delivery vehicles using oxanorbornane-based non-ionic amphiphiles. J Mater Chem B 2016; 4:8025-8032. [DOI: 10.1039/c6tb02192a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The self-assembly of non-ionic amphiphiles with hydroxylated oxanorbornane head-group was controlled using amino acid units as spacers between hydrophilic and lipophilic domains to get spherical supramolecular aggregates suitable for drug delivery applications.
Collapse
Affiliation(s)
- D. Sirisha Janni
- Department of Chemistry
- Indian Institute of Technology Madras
- Chennai
- India
| | | | - Soumya Saroj
- Department of Chemistry
- Indian Institute of Technology Madras
- Chennai
- India
| | | |
Collapse
|
45
|
Liu Y, Ng Y, Toh MR, Chiu GNC. Lipid-dendrimer hybrid nanosystem as a novel delivery system for paclitaxel to treat ovarian cancer. J Control Release 2015; 220:438-446. [PMID: 26551345 DOI: 10.1016/j.jconrel.2015.11.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 10/27/2015] [Accepted: 11/05/2015] [Indexed: 12/18/2022]
Abstract
Combining lipids and dendrimers into one formulation is an emerging platform in the drug delivery field. This study aims to (i) develop and characterize a lipid-dendrimer hybrid (LDH) nanosystem for the hydrophobic anticancer drug paclitaxel, and (ii) evaluate its in vitro and in vivo anti-cancer activity in ovarian cancer models. The LDH nanosystems were prepared from 1,2-dipalmitoyl-sn-glycero-3-phosphocholine and poly (amidoamine) (PAMAM) G4.0. The size and zeta potential of the LDH nanosystem were 37.6 ± 6.1n m and +2.9 ± 0.1 mV, respectively, with vesicular morphology observed under cryo-TEM. The encapsulation efficiency of paclitaxel in the LDH system was 78.0 ± 2.1%. The potency of paclitaxel could be significantly improved by 37-fold when presented in the LDH nanosystem as compared to free drug, whereby paclitaxel and PAMAM G4.0 acted synergistically in killing the ovarian cancer cells. As shown by fluorescence confocal microscopy, majority of the lipids in the LDH nanosystem were located in the plasma membrane, while the dendrimers were distributed intracellularly upon uptake. Despite the use of a 10-fold lower paclitaxel dose, the survival of IGROV-1 ovarian tumor-bearing animals could be significantly prolonged by the paclitaxel-loaded LDH nanosystem, as reflected by a 50% increase in the median survival time. Such hybrid nanosystem emerged from combining two established drug delivery platforms could pave way for the development of multifunctional delivery systems for potential theranostic applications.
Collapse
Affiliation(s)
- Yuanjie Liu
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Yiwei Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Ming R Toh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Gigi N C Chiu
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| |
Collapse
|
46
|
WITHDRAWN: Polymer assembly: Promising carriers as co-delivery systems for cancer therapy. Prog Polym Sci 2015. [DOI: 10.1016/j.progpolymsci.2015.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
47
|
Yi X, Lian X, Dong J, Wan Z, Xia C, Song X, Fu Y, Gong T, Zhang Z. Co-delivery of Pirarubicin and Paclitaxel by Human Serum Albumin Nanoparticles to Enhance Antitumor Effect and Reduce Systemic Toxicity in Breast Cancers. Mol Pharm 2015; 12:4085-98. [DOI: 10.1021/acs.molpharmaceut.5b00536] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Xiaoli Yi
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Xianghong Lian
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Jianxia Dong
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Zhuoya Wan
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Chunyu Xia
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Xu Song
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Yao Fu
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Tao Gong
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| | - Zhirong Zhang
- Key Laboratory
of Drug Targeting
and Drug Delivery Systems, Ministry of Education, Sichuan University, Sichuan, People’s Republic of China
| |
Collapse
|
48
|
Liu C, Luo Q, Tu Y, Wang G, Liu Y, Xie Y. Drug-carrier interaction analysis in the cell penetrating peptide-modified liposomes for doxorubicin loading. J Microencapsul 2015; 32:745-54. [PMID: 26299658 DOI: 10.3109/02652048.2015.1073390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Doxorubicin (DOX) is widely used as an antitumor model drug in liposomes because of its high encapsulation efficiency. The cell-penetrating peptide (CPP) has potential applications in drug delivery systems. However, we discovered that the encapsulation efficiency of DOX decreased with increasing modification density of CPP on liposomes. To explore the interaction mechanisms of CPP-modified liposomes (CPPL) for DOX loading, X-ray diffraction, Fourier transform infrared spectroscopy and Raman spectroscopy were utilised, and theoretical calculations based on molecular dynamics simulation were performed. Results showed that the monomeric intermolecular interaction between CPP and DOX, in which the guanidinium group of CPP was parallel to the planar aromatic chromophore of DOX, depending on the cation-pi interaction and hydrogen bonds, weakened the tendency of DOX transporting into the internal medium from the liposomal external medium. Analysis of the interaction between CPP and DOX at the molecular level provided theoretical guidance for the further development of CPPL.
Collapse
Affiliation(s)
- Chang Liu
- a Department of Pharmaceutics, State Key Laboratory of Natural and Biomimetic Drugs , School of Pharmaceutical Sciences, Peking University , Beijing , People's Republic of China and
| | - Qi Luo
- b Soft Matter Research Center and Department of Chemistry, Zhejiang University , Hangzhou , Zhejiang , People's Republic of China
| | - YingFeng Tu
- a Department of Pharmaceutics, State Key Laboratory of Natural and Biomimetic Drugs , School of Pharmaceutical Sciences, Peking University , Beijing , People's Republic of China and
| | - GuiLing Wang
- a Department of Pharmaceutics, State Key Laboratory of Natural and Biomimetic Drugs , School of Pharmaceutical Sciences, Peking University , Beijing , People's Republic of China and
| | - YingChun Liu
- b Soft Matter Research Center and Department of Chemistry, Zhejiang University , Hangzhou , Zhejiang , People's Republic of China
| | - Ying Xie
- a Department of Pharmaceutics, State Key Laboratory of Natural and Biomimetic Drugs , School of Pharmaceutical Sciences, Peking University , Beijing , People's Republic of China and
| |
Collapse
|
49
|
SONG XINGLI, REN YI, ZHANG JING, WANG GANG, HAN XUEDONG, ZHENG WEI, ZHEN LINLIN. Targeted delivery of doxorubicin to breast cancer cells by aptamer functionalized DOTAP/DOPE liposomes. Oncol Rep 2015; 34:1953-60. [DOI: 10.3892/or.2015.4136] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/06/2015] [Indexed: 11/06/2022] Open
|
50
|
Linton SS, Sherwood SG, Drews KC, Kester M. Targeting cancer cells in the tumor microenvironment: opportunities and challenges in combinatorial nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:208-22. [PMID: 26153136 DOI: 10.1002/wnan.1358] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/01/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022]
Abstract
Cancer therapies of the future will rely on synergy between drugs delivered in combination to achieve both maximum efficacy and decreased toxicity. Nanoscale drug delivery vehicles composed of highly tunable nanomaterials ('nanocarriers') represent the most promising approach to achieve simultaneous, cell-selective delivery of synergistic ratios of combinations of drugs within solid tumors. Nanocarriers are currently being used to co-encapsulate and deliver synergistic ratios of multiple anticancer drugs to target cells within solid tumors. Investigators exploit the unique environment associated with solid tumors, termed the tumor microenvironment (TME), to make 'smart' nanocarriers. These sophisticated nanocarriers exploit the pathological conditions in the TME, thereby creating highly targeted nanocarriers that release their drug payload in a spatially and temporally controlled manner. The translational and commercial potential of nanocarrier-based combinatorial nanomedicines in cancer therapy is now a reality as several companies have initiated human clinical trials.
Collapse
Affiliation(s)
- Samuel S Linton
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, USA
| | - Samantha G Sherwood
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Kelly C Drews
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|