1
|
Stępień M, Zajda J, Keppler BK, Timerbaev AR, Matczuk M. Cisplatin meets liposomes for a smarter delivery: A review. Talanta 2025; 295:128331. [PMID: 40382863 DOI: 10.1016/j.talanta.2025.128331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Effective treatment of tumors remains a significant clinical challenge even for approved anticancer drugs such as cisplatin, whose chemotherapy is hindered by inherent toxicity, rapidly acquired resistance, and nonselective mode of action. In the past years, nanodelivery systems have emerged as a key strategy to overcome these limitations due to their potential to improve drug safety, bioavailability, and efficacy. Among various nanostructures applied as carriers for the delivery of cisplatin, liposomes have undergone intensive testing, with the outcome of being advanced to clinical trials. This fact not only triggers further research endeavors toward developing improved liposomal formulations but also makes it timely to highlight recent trends and strategies, showcasing the evolution and application of cisplatin-liposome systems. The present review is aimed at a critical analysis of fabrication, encapsulation, stability testing, release, and cell/animal experimental procedures, focusing on the analytical methodology used to feature these essential practices and providing insights that may help enhance the efficacy of cisplatin chemotherapy.
Collapse
Affiliation(s)
- Marta Stępień
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Stanisława Noakowskiego 3, 00-664, Warsaw, Poland
| | - Joanna Zajda
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Stanisława Noakowskiego 3, 00-664, Warsaw, Poland.
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Währinger Str. 42, 1090, Vienna, Austria
| | - Andrei R Timerbaev
- Institute of Inorganic Chemistry, University of Vienna, Währinger Str. 42, 1090, Vienna, Austria
| | - Magdalena Matczuk
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Stanisława Noakowskiego 3, 00-664, Warsaw, Poland.
| |
Collapse
|
2
|
Jaiswal N, Mahata N, Chanda N. Nanogold-albumin conjugates: transformative approaches for next-generation cancer therapy and diagnostics. NANOSCALE 2025; 17:11191-11220. [PMID: 40237258 DOI: 10.1039/d4nr05279j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Nanogold-albumin conjugates have garnered significant attention as a highly adaptable theranostic platform, capable of delivering a wide range of therapeutics, from small-molecule drugs to larger biomolecules, while offering promising applications for monitoring and managing cancer. The remarkable theranostic capabilities of these conjugates stem from the combined strengths of gold and albumin, which provide low toxicity, a large surface area, customizable surface chemistry, and unique optical properties, all contributing to their potential in cancer therapy. This review delves into the design and development of two primary types of nanogold-albumin conjugate: supramolecular albumin-coated gold nanoparticles (GNP-BSA/HSA) and albumin-templated ultra-small gold nanoclusters (GNC-BSA/HSA). Each strategy offers distinct advantages, enabling the fine-tuning of conjugate properties to optimize therapeutic delivery and facilitate cancer-specific bio-sensing. The integration of gold and albumin further improves biocompatibility, extends circulation time, and enhances tumor targeting, making these conjugates an attractive option for cancer treatment. The review also focuses on the refinement of surface chemistry to achieve precise targeting of cancer cells, as well as the challenges and future prospects for advancing nanogold-albumin systems in clinical applications.
Collapse
Affiliation(s)
- Namita Jaiswal
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nripen Chanda
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
| |
Collapse
|
3
|
Kotze AC, Knowles AG. Antagonistic interactions between spinosad and macrocyclic lactones in combination against larvae of the sheep blowfly, Lucilia cuprina, in vitro. Vet Parasitol 2025; 334:110389. [PMID: 39764947 DOI: 10.1016/j.vetpar.2024.110389] [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: 09/26/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 02/27/2025]
Abstract
Control of the sheep blowfly relies on insecticides, however resistance is currently impacting on their efficacy. The use of insecticides in combination (mixtures) is considered to be a useful strategy to delay resistance under some circumstances. The present study aimed to examine the combination of spinosad with macrocyclic lactones in order to determine if the two drug classes showed any interactions that would impact on the usefulness of a combination product for flystrike control. We used isobologram analysis to examine drug interactions in in vitro bioassays with the larval stages of the sheep blowfly. In initial single drug assays, ivermectin and abamectin showed similar EC50 against blowfly larvae, while moxidectin and spinosad were 20- and 30-fold less potent, respectively. Combinations of spinosad and each of the macrocyclic lactones showed a pattern of interactions dominated by antagonistic or additive relationships when combined at different ratios. Peaks in antagonism were associated with combinations at ratios equivalent to, or close to, the relative EC50s when used alone. The antagonism was strongest for moxidectin, with combination index values up to 1.82 at the EC50 and 3.2 at the EC90 at a spinosad: moxidectin ratio of 0.78:1. Maximum combination index values for ivermectin and abamectin at the EC90 were 1.61 and 1.42, at ratios of 27:1 and 31:1, respectively, indicating a significant degree of antagonism for both combinations. We suggest that the observed antagonism may be due to interactions at common ion channel receptors, although this requires confirmation. The study highlights the need to examine drug interactions as a component of determining the suitability of specific drug combinations for parasite control.
Collapse
Affiliation(s)
- Andrew C Kotze
- CSIRO Agriculture and Food, Brisbane, QLD 4069, Australia; School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia.
| | | |
Collapse
|
4
|
Saberian E, Jenčová J, Jenča A, Jenča A, Petrášová A, Jenča J, akbarzadehkhayavi A. Combination Therapy of Curcumin and Cisplatin Encapsulated in Niosome Nanoparticles for Enhanced Oral Cancer Treatment. Indian J Clin Biochem 2025; 40:59-66. [PMID: 39835233 PMCID: PMC11741963 DOI: 10.1007/s12291-024-01279-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025]
Abstract
Oral cavity cancer poses a significant health threat due to its aggressive nature and limited responsiveness to traditional therapies like chemotherapy and radiation, highlighting the need for more effective treatment options. To address this, researchers have explored a novel approach using niosome nanoparticles to co-encapsulate curcumin (CUR) and cisplatin (Cis), to enhance therapeutic efficacy. While CUR has anti-cancer properties, its poor bioavailability limits its effectiveness. Cis, on the other hand, is hindered by severe side effects and resistance. A dual-drug delivery system that encapsulates both CUR and Cis in niosome nanoparticles seeks to leverage the synergistic effects of these agents to improve treatment outcomes. The study synthesized Cis and CUR co-loaded nanoparticles (Cis/CUR-NPs) using reverse microemulsion and film dispersion methods, resulting in nanoparticles with an average size of 220.9 nm and a consistent size distribution. In vitro experiments demonstrated that the nanosized Cis/CUR-NPs could release both Cis and CUR, achieving a synergistic effect on OECM-1 cells at an optimal ratio (1:6) of the two drugs. Overall, the findings suggest that Cis/CUR-NPs offer a promising and effective strategy for leveraging the synergistic effects of Cis and CUR in treating oral cancer.
Collapse
Affiliation(s)
- Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Jozef Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | | |
Collapse
|
5
|
Wu Y, Zeng C, Lv J, Li H, Gao J, Liu Z, Yuan Z. Tumor-Targeted cRGD-Coated Liposomes Encapsulating Optimized Synergistic Cepharanthine and IR783 for Chemotherapy and Photothermal Therapy. Int J Nanomedicine 2024; 19:6145-6160. [PMID: 38911506 PMCID: PMC11194009 DOI: 10.2147/ijn.s457008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Background Combination therapy offers superior therapeutic results compared to monotherapy. However, the outcomes of combination therapy often fall short of expectations, mainly because of increased toxicity from drug interactions and challenges in achieving the desired spatial and temporal distribution of drug delivery. Optimizing synergistic drug combination ratios to ensure uniform targeting and distribution across space and time, particularly in vivo, is a significant challenge. In this study, cRGD-coated liposomes encapsulating optimized synergistic cepharanthine (CEP; a chemotherapy drug) and IR783 (a phototherapy agent) were developed for combined chemotherapy and photothermal therapy in vitro and in vivo. Methods An MTT assay was used to evaluate the combination index of CEP and IR783 in five cell lines. The cRGD-encapsulated liposomes were prepared via thin-film hydration, and unencapsulated liposomes served as controls for the loading of CEP and IR783. Fluorescence and photothermal imaging were used to assess the efficacy of CEP and IR783 encapsulated in liposomes at an optimal synergistic ratio, both in vitro and in vivo. Results The combination indices of CEP and IR783 were determined in five cell lines. As a proof-of-concept, the optimal synergistic ratio (1:2) of CEP to IR783 in 4T1 cells was evaluated in vitro and in vivo. The average diameter of the liposomes was approximately 100 nm. The liposomes effectively retained the encapsulated CEP and IR783 in vitro at the optimal synergistic molar ratio for over 7 d. In vivo fluorescence imaging revealed that the fluorescence signal from cRGD-CEP-IR783-Lip was detectable at the tumor site at 4 h post-injection and peaked at 8 h. In vivo photothermal imaging of tumor-bearing mice indicated an increase in tumor temperature by 32°C within 200 s. Concurrently, cRGD-CEP-IR783-Lip demonstrated a significant therapeutic effect and robust biosafety in the in vivo antitumor experiments. Conclusion The combination indices of CEP and IR783 were successfully determined in vitro in five cell lines. The cRGD-coated liposomes encapsulated CEP and IR783 at an optimal synergistic ratio, exhibiting enhanced antitumor effects and targeting upon application in vitro and in vivo. This study presents a novel concept and establishes a research framework for synergistic chemotherapy and phototherapy treatment.
Collapse
Affiliation(s)
- Yumei Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Chunhua Zeng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Jiajia Lv
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Hongyu Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Jie Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Zhidong Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Zeli Yuan
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| |
Collapse
|
6
|
Liu Y, Xu Y, Wang Y, Lv J, Wang K, Tang Z. Hindering the unlimited proliferation of tumor cells synergizes with destroying tumor blood vessels for effective cancer treatment. Biomater Sci 2024; 12:1294-1306. [PMID: 38258411 DOI: 10.1039/d3bm01858j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The rational combination of chemotherapy drugs can improve the curative effect of cancer treatment. As two early recognized tumor hallmarks, the limitless replicative potential of tumor cells is essential for the development of their malignant growth state, and sustained angiogenesis is a prerequisite to the rapid growth of tumors. Based on this, we propose a combination therapy that hinders the unlimited proliferation of tumor cells and destroys tumor blood vessels. Herein, 7-ethyl-10-hydroxycamptothecin (SN38), a typical topoisomerase I inhibitor, was bonded to poly(L-glutamic acid) (PLG) to prepare the nanodrug SN38-NPs, which hinders the unlimited proliferation of tumor cells. A poly(L-glutamic acid)-combretastatin A4 conjugate (CA4-NPs), a representative vascular disrupting agent (VDA), was used to selectively disrupt the tumor blood vessels, cutting off the necessary nutrients and oxygen for the proliferation of tumor cells. In the 4T1 tumor model with an initial volume of about 400 mm3, the combined treatment of SN38-NPs and CA4-NPs showed an excellent cancer treatment effect with a tumor suppression rate of 94.3% and a synergistic interaction (Q = 1.25). Our study provides a new combination therapy approach for chemotherapy, with the hope of further improving the curative effect of anti-cancer therapy.
Collapse
Affiliation(s)
- Ya Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yajun Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Ying Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jianlin Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Kun Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
7
|
Hu Q, Zhang Y, Mukerabigwi JF, Wang H, Cao Y. Polymer Conjugate as the New Promising Drug Delivery System for Combination Therapy against Cancer. Curr Top Med Chem 2024; 24:1101-1119. [PMID: 39005059 DOI: 10.2174/0115680266280603240321064308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 07/16/2024]
Abstract
This review highlights the advantages of combination therapy using polymer conjugates as drug delivery systems for cancer treatment. In this review, the specific structures and materials of polymer conjugates, as well as the different types of combination chemotherapy strategies, are discussed. Specific targeting strategies, such as monoclonal antibody therapy and small molecule ligands, are also explored. Additionally, self-assembled polymer micelles and overcoming multidrug resistance are described as potential strategies for combination therapy. The assessment of combinational therapeutic efficacy and the challenges associated with polymer conjugates are also addressed. The future outlook aims to overcome these challenges and improve the effectiveness of drug delivery systems for combination therapy. The conclusion emphasizes the potential of polymer conjugates in combination therapy while acknowledging the need for further research and development in this field.
Collapse
Affiliation(s)
- Qiang Hu
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yuannian Zhang
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Jean Felix Mukerabigwi
- Department of Chemistry, University of Rwanda, College of Science and Technology, Po. Box: 3900, Kigali, Rwanda
| | - Haili Wang
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yu Cao
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| |
Collapse
|
8
|
Gabizon A, Shmeeda H, Draper B, Parente-Pereira A, Maher J, Carrascal-Miniño A, de Rosales RTM, La-Beck NM. Harnessing Nanomedicine to Potentiate the Chemo-Immunotherapeutic Effects of Doxorubicin and Alendronate Co-Encapsulated in Pegylated Liposomes. Pharmaceutics 2023; 15:2606. [PMID: 38004584 PMCID: PMC10675201 DOI: 10.3390/pharmaceutics15112606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Encapsulation of Doxorubicin (Dox), a potent cytotoxic agent and immunogenic cell death inducer, in pegylated (Stealth) liposomes, is well known to have major pharmacologic advantages over treatment with free Dox. Reformulation of alendronate (Ald), a potent amino-bisphosphonate, by encapsulation in pegylated liposomes, results in significant immune modulatory effects through interaction with tumor-associated macrophages and activation of a subset of gamma-delta T lymphocytes. We present here recent findings of our research work with a formulation of Dox and Ald co-encapsulated in pegylated liposomes (PLAD) and discuss its pharmacological properties vis-à-vis free Dox and the current clinical formulation of pegylated liposomal Dox. PLAD is a robust formulation with high and reproducible remote loading of Dox and high stability in plasma. Results of biodistribution studies, imaging with radionuclide-labeled liposomes, and therapeutic studies as a single agent and in combination with immune checkpoint inhibitors or gamma-delta T lymphocytes suggest that PLAD is a unique product with distinct tumor microenvironmental interactions and distinct pharmacologic properties when compared with free Dox and the clinical formulation of pegylated liposomal Dox. These results underscore the potential added value of PLAD for chemo-immunotherapy of cancer and the relevance of the co-encapsulation approach in nanomedicine.
Collapse
Affiliation(s)
- Alberto Gabizon
- Nano-Oncology Research Center, Oncology Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Hilary Shmeeda
- Nano-Oncology Research Center, Oncology Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
| | - Benjamin Draper
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - Ana Parente-Pereira
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - John Maher
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - Amaia Carrascal-Miniño
- King’s College London, School of Biomedical Engineering & Imaging Sciences, St. Thomas’ Hospital, London SE1 7EH, UK; (A.C.-M.); (R.T.M.d.R.)
| | - Rafael T. M. de Rosales
- King’s College London, School of Biomedical Engineering & Imaging Sciences, St. Thomas’ Hospital, London SE1 7EH, UK; (A.C.-M.); (R.T.M.d.R.)
| | - Ninh M. La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA;
| |
Collapse
|
9
|
Gupta P, Neupane YR, Aqil M, Kohli K, Sultana Y. Lipid-based nanoparticle-mediated combination therapy for breast cancer management: a comprehensive review. Drug Deliv Transl Res 2023; 13:2739-2766. [PMID: 37261602 DOI: 10.1007/s13346-023-01366-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
Breast cancer due to the unpredictable and complex etiopathology combined with the non-availability of any effective drug treatment has become the major root of concern for oncologists globally. The number of women affected by the said disease state is increasing at an alarming rate attributed to environmental and lifestyle changes indicating at the exploration of a novel treatment strategy that can eradicate this aggressive disease. So far, it is treated by promising nanomedicine monotherapy; however, according to the numerous studies conducted, the inadequacy of these nano monotherapies in terms of elevated toxicity and resistance has been reported. This review, therefore, puts forth a new multimodal strategic approach to lipid-based nanoparticle-mediated combination drug delivery in breast cancer, emphasizing the recent advancements. A basic overview about the combination therapy and its index is firstly given. Then, the various nano-based combinations of chemotherapeutics involving the combination delivery of synthetic and herbal agents are discussed along with their examples. Further, the recent exploration of chemotherapeutics co-delivery with small interfering RNA (siRNA) agents has also been explained herein. Finally, a section providing a brief description of the delivery of chemotherapeutic agents with monoclonal antibodies (mAbs) has been presented. From this review, we aim to provide the researchers with deep insight into the novel and much more effective combinational lipid-based nanoparticle-mediated nanomedicines tailored specifically for breast cancer treatment resulting in synergism, enhanced antitumor efficacy, and low toxic effects, subsequently overcoming the hurdles associated with conventional chemotherapy.
Collapse
Affiliation(s)
- Priya Gupta
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yub Raj Neupane
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA, 52242, USA
| | - Mohd Aqil
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
- Lloyd Institute of Management & Technology (Pharm.), Plot No. 11, Knowledge Park-II, Greater Noida, Uttar Pradesh, 201308, India.
| | - Yasmin Sultana
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
10
|
Zashikhina N, Gandalipov E, Dzhuzha A, Korzhikov-Vlakh V, Korzhikova-Vlakh E. Dual drug loaded polypeptide delivery systems for cancer therapy. J Microencapsul 2023:1-19. [PMID: 37824702 DOI: 10.1080/02652048.2023.2270064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The present study was aimed to prepare and examine in vitro novel dual-drug loaded delivery systems. Biodegradable nanoparticles based on poly(L-glutamic acid-co-D-phenylalanine) were used as nanocarriers for encapsulation of two drugs from the paclitaxel, irinotecan, and doxorubicin series. The developed delivery systems were characterised with hydrodynamic diameters less than 300 nm (PDI < 0.3). High encapsulation efficiencies (≥75%) were achieved for all single- and dual-drug formulations. The release studies showed faster release at acidic pH, with the release rate decreasing over time. The release patterns of the co-encapsulated forms of substances differed from those of the separately encapsulated drugs, suggesting differences in drug-polymer interactions. The joint action of encapsulated drugs was analysed using the colon cancer cells, both for the dual-drug delivery sytems and a mixture of single-drug formulations. The encapsulated forms of the drug combinations demonstrated comparable efficacy to the free forms, with the encapsulation enhancing solubility of the hydrophobic drug paclitaxel.
Collapse
Affiliation(s)
- Natalia Zashikhina
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg, Russia
| | - Erik Gandalipov
- International Institute of Solution Chemistry and Advanced Materials Technologies, ITMO University, St. Petersburg, Russia
| | - Apollinariia Dzhuzha
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg, Russia
| | - Viktor Korzhikov-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
11
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
12
|
Liu Y, Yang G, Hui Y, Ranaweera S, Zhao CX. Microfluidic Nanoparticles for Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106580. [PMID: 35396770 DOI: 10.1002/smll.202106580] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/20/2021] [Indexed: 06/14/2023]
Abstract
Nanoparticles (NPs) have attracted tremendous interest in drug delivery in the past decades. Microfluidics offers a promising strategy for making NPs for drug delivery due to its capability in precisely controlling NP properties. The recent success of mRNA vaccines using microfluidics represents a big milestone for microfluidic NPs for pharmaceutical applications, and its rapid scaling up demonstrates the feasibility of using microfluidics for industrial-scale manufacturing. This article provides a critical review of recent progress in microfluidic NPs for drug delivery. First, the synthesis of organic NPs using microfluidics focusing on typical microfluidic methods and their applications in making popular and clinically relevant NPs, such as liposomes, lipid NPs, and polymer NPs, as well as their synthesis mechanisms are summarized. Then, the microfluidic synthesis of several representative inorganic NPs (e.g., silica, metal, metal oxide, and quantum dots), and hybrid NPs is discussed. Lastly, the applications of microfluidic NPs for various drug delivery applications are presented.
Collapse
Affiliation(s)
- Yun Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yue Hui
- Institute of Advanced Technology, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Supun Ranaweera
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Chemical Engineering and Advanced Materials, Faculty of Engineering, Computer and Mathematical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| |
Collapse
|
13
|
Alqahtani AA, Aslam H, Shukrullah S, Fatima H, Naz MY, Rahman S, Mahnashi MH, Irfan M. Nanocarriers for Smart Therapeutic Strategies to Treat Drug-Resistant Tumors: A Review. Assay Drug Dev Technol 2022; 20:191-210. [DOI: 10.1089/adt.2022.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Hira Aslam
- Department of Physics, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Shazia Shukrullah
- Department of Physics, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Hareem Fatima
- Department of Physics, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Muhammad Yasin Naz
- Department of Physics, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Saifur Rahman
- Electrical Engineering Department, College of Engineering, Najran University, Najran, Saudi Arabia
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Muhammad Irfan
- Electrical Engineering Department, College of Engineering, Najran University, Najran, Saudi Arabia
| |
Collapse
|
14
|
van der Merwe L, Svitina H, Willers C, Wrzesinski K, Gouws C. A novel NCI-H69V small cell lung cancer functional mini-tumor model for future treatment screening applications. Biotechnol Prog 2022; 38:e3253. [PMID: 35362670 PMCID: PMC10909478 DOI: 10.1002/btpr.3253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/24/2022] [Accepted: 03/29/2022] [Indexed: 12/09/2022]
Abstract
Small cell lung cancer (SCLC) is aggressive and despite multiple clinical trials, its standard of care is unchanged for the past three decades. In vitro cancer models are crucial in chemotherapy development, and three-dimensional (3D) models aim to bridge the gap between two-dimensional (2D) flat cultures and in vivo testing. Functional 3D spheroids can better represent the in vivo situation and tumor characteristics than 2D models. An NCI-H69V SCLC mini-tumor model was developed in a clinostat-based rotating bioreactor system. Spheroid growth and viability were characterized for 30 days, and the ideal experimental window with mature and metabolically stable spheroids was determined. Application of the model for anticancer treatment screening was validated with the standard chemotherapeutic drug irinotecan, for an exposure period of 72 h. The following parameters were measured: soluble protein content, planar surface area measurements, intracellular adenosine triphosphate and extracellular adenylate kinase levels, and glucose consumption. Histological morphology of the spheroids was observed. The established model proved viable and stable, while treatment with irinotecan caused a decrease in cell growth, viability, and glucose consumption demonstrating reactivity of the model to chemotherapy. Therefore, this NCI-H69V SCLC functional spheroid model could be used for future anticancer compound screening.
Collapse
Affiliation(s)
- Liezaan van der Merwe
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™)North‐West UniversityPotchefstroomSouth Africa
| | - Hanna Svitina
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™)North‐West UniversityPotchefstroomSouth Africa
- Laboratory of Biosynthesis of Nucleic Acids, Department of Functional GenomicsInstitute of Molecular Biology and Genetics of NASUKyivUkraine
| | - Clarissa Willers
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™)North‐West UniversityPotchefstroomSouth Africa
| | - Krzysztof Wrzesinski
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™)North‐West UniversityPotchefstroomSouth Africa
- CelVivo ApSBlommenslystDenmark
| | - Chrisna Gouws
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™)North‐West UniversityPotchefstroomSouth Africa
| |
Collapse
|
15
|
Zheng S, Wang J, Ding N, Chen W, Chen H, Xue M, Chen F, Ni J, Wang Z, Lin Z, Jiang H, Liu X, Wang L. Prodrug polymeric micelles integrating cancer-associated fibroblasts deactivation and synergistic chemotherapy for gastric cancer. J Nanobiotechnology 2021; 19:381. [PMID: 34802453 PMCID: PMC8607732 DOI: 10.1186/s12951-021-01127-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/07/2021] [Indexed: 12/27/2022] Open
Abstract
Background The prognosis of patients with advanced gastric cancer (GC) remains unsatisfactory owing to distant metastasis and resistance to concurrent systemic therapy. Cancer-associated fibroblasts (CAFs), as essential participators in the tumor microenvironment (TME), play a vital role in tumor progression. Thus, CAFs-targeting therapy is appealing for remodeling TME and sensitizing GC to conventional systemic therapy. Methods Amphiphilic SN38 prodrug polymeric micelles (PSN38) and encapsulated the hydrophobic esterase-responsive prodrug of Triptolide (TPL), triptolide-naphthalene sulfonamide (TPL-nsa), were synthesized to form PSN38@TPL-nsa nanoparticles. Then, CAFs were isolated from fresh GC tissues and immortalized. TPL at low dose concentration was used to investigate its effect on CAFs and CAFs-induced GC cells proliferation and migration. The synergistic mechanism and antitumor efficiency of SN38 and TPL co-delivery nanoparticle were investigated both in vitro and in vivo. Results Fibroblast activation protein (FAP), a marker of CAFs, was highly expressed in GC tissues and indicated poorer prognosis. TPL significantly reduced CAFs activity and inhibited CAFs-induced proliferation, migration and chemotherapy resistance of GC cells. In addition, TPL sensitized GC cells to SN38 treatment through attenuated NF-κB activation in both CAFs and GC cells. PSN38@TPL-nsa treatment reduced the expression of collagen, FAP, and α-smooth muscle actin (α-SMA) in tumors. Potent inhibition of primary tumor growth and vigorous anti-metastasis effect were observed after systemic administration of PSN38@TPL-nsa to CAFs-rich peritoneal disseminated tumor and patient-derived xenograft (PDX) model of GC. Conclusion TPL suppressed CAFs activity and CAFs-induced cell proliferation, migration and chemotherapy resistance to SN38 of GC. CAFs-targeted TPL and SN38 co-delivery nanoparticles exhibited potent efficacy of antitumor and reshaping TME, which was a promising strategy to treat advanced GC. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01127-5.
Collapse
Affiliation(s)
- Sheng Zheng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Jiafeng Wang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Ning Ding
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Wenwen Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Hongda Chen
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Meng Xue
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Fei Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Jiaojiao Ni
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Zhuo Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Zhenghua Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310016, China
| | - Xiangrui Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Liangjing Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Gastroenterology, Zhejiang University, Hangzhou, 310058, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
16
|
Triple negative breast cancer and non-small cell lung cancer: Clinical challenges and nano-formulation approaches. J Control Release 2021; 337:27-58. [PMID: 34273417 DOI: 10.1016/j.jconrel.2021.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Abstract
Triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC) are amongst the most aggressive forms of solid tumors. TNBC is highlighted by absence of genetic components of progesterone receptor, HER2/neu and estrogen receptor in breast cancer. NSCLC is characterized by integration of malignant carcinoma into respiratory system. Both cancers are associated with poor median and overall survival rates with low progression free survival with high incidences of relapse. These cancers are characterized by tumor heterogeneity, genetic mutations, generation of cancer-stem cells, immune-resistance and chemoresistance. Further, these neoplasms have been reported for tumor cross-talk into second primary cancers for each other. Current chemotherapeutic regimens include usage of multiple agents in tandem to affect tumor cells through multiple mechanisms with various such combinations being clinically tested. However, lack of controlled delivery and effective temporospatial presence of chemotherapeutics has resulted in suboptimal therapeutic response. Consequently, passive targeted albumin bound paclitaxel and PEGylated liposomal doxorubicin have been clinically used and tested with newer drugs for improved therapeutic efficacy in these cancers. Active targeting of nanocarriers against surface overexpressed proteins in both neoplasms have been explored. However, use of single agent nanoparticulate formulations against both cancers have failed to elicit desired outcomes. This review aims to identify clinical unmet need in these cancers while establishing a correlation with tested nano-formulation approaches and issues with preclinical to clinical translation. Lipid and polymer-based drug-drug and drug-gene combinatorial nanocarriers delivering multiple chemotherapeutics simultaneously to desired site of action have been detailed. Finally, emerging opportunities such as pharmacological targets (immune check point and epigentic modulators) as well as gene-based modulation (siRNA/CRISPR/Cas9) and the nano-formulation challenges for effective treatment of both cancers have been explored.
Collapse
|
17
|
Khoshtabiat L, Meshkini A, Matin MM. Fenton-magnetic based therapy by dual-chemodrug-loaded magnetic hydroxyapatite against colon cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112238. [PMID: 34225878 DOI: 10.1016/j.msec.2021.112238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/02/2021] [Accepted: 06/02/2021] [Indexed: 10/21/2022]
Abstract
Fenton-based therapy is emerging as an effective and selective strategy against cancer. However, a low concentration of transition metal ions, insufficient endogenous H2O2, and a high level of antioxidant activity within the cancer cells have hindered the therapeutic efficacy of this strategy. To address these issues, in this study, the Fenton reagent (magnetic hydroxyapatite, mHAP) was accompanied with chemotherapy drugs (cisplatin (CDDP) and methotrexate (MTX)) and static magnetic field (SMF), in such a way to be a pH-, redox-, and magnetic-responsive nanoplatform. In vitro and in vivo experiments revealed higher toxicity of the final construct, MTX.CDDP@mHAP, toward colon cancer cells, as compared with that of free drugs. The most effective antitumor activity was observed as MTX.CDDP@mHAP-treated tumor cells were exposed to SMF (0.9 T) and no noticeable damage was observed in the normal cells and tissues. Active targeting by MTX and magnetic targeting by mHAP under magnetic field increased the tumor selectivity and enhanced the tumor site accumulation and cellular uptake of MTX.CDDP@mHAPs. The released iron ions within the cancer cells trigger the Fenton reaction while the release of chemotherapy drugs, reduction of intracellular glutathione, and application of SMF aggravated the Fenton reaction, subsequently leading to the generation of reactive oxygen species (ROS) and induction of apoptosis. Therefore, Fenton magnetic-based therapy-mediated by MTX.CDDP@mHAP could be considered as a promising strategy against colon cancer with high therapeutic efficiency and biosafety.
Collapse
Affiliation(s)
- Laya Khoshtabiat
- Biochemical Research Center, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Azadeh Meshkini
- Biochemical Research Center, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
18
|
Zhang Z, Yue YX, Xu L, Wang Y, Geng WC, Li JJ, Kong XL, Zhao X, Zheng Y, Zhao Y, Shi L, Guo DS, Liu Y. Macrocyclic-Amphiphile-Based Self-Assembled Nanoparticles for Ratiometric Delivery of Therapeutic Combinations to Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007719. [PMID: 33598992 DOI: 10.1002/adma.202007719] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/11/2021] [Indexed: 06/12/2023]
Abstract
Combination chemotherapy refers to the use of multiple drugs to treat cancer. In this therapy, the optimal ratio of the drugs is essential to achieve drug synergism and the desired therapeutic effects. However, most delivery strategies are unable to precisely control the ratio of the drugs during the drug loading and delivery processes, resulting in inefficient synergy and unpredictable efficacy. Herein, a macrocyclic-amphiphile-based self-assembled nanoparticle (MASN) that achieves precise loading and ratiometric delivery of therapeutic combinations is presented. By integrating multiple macrocyclic cavities within a single nanoparticle, the MASN can load multiple drug molecules via the host-guest interaction, and the ratio of the drugs loaded can be predicted with their initial concentrations and characteristic binding affinity. Moreover, MASNs are readily degraded under a hypoxic microenvironment, allowing spontaneous release of the drugs upon reaching tumor tissues. With precise drug loading and controlled release mechanisms, MASNs achieve ratiometric delivery of multiple commercial drugs to tumors, thereby achieving optimal anti-tumor effects. Since the optimal drug ratio of a therapeutic combination can be quickly determined in vitro, MASNs can translate this optimal ratio to the therapeutic benefits in vivo, providing a potential platform for the rapid development of effective combination cancer therapies involving multiple drugs.
Collapse
Affiliation(s)
- Zhanzhan Zhang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yu-Xin Yue
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Lina Xu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Ying Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Juan-Juan Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Xiang-Lei Kong
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Xinzhi Zhao
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yadan Zheng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yu Zhao
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Linqi Shi
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Yang Liu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
19
|
Maniam G, Mai CW, Zulkefeli M, Fu JY. Co-encapsulation of gemcitabine and tocotrienols in nanovesicles enhanced efficacy in pancreatic cancer. Nanomedicine (Lond) 2021; 16:373-389. [PMID: 33543651 DOI: 10.2217/nnm-2020-0374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/06/2021] [Indexed: 02/08/2023] Open
Abstract
Aim: To synthesize niosomes co-encapsulating gemcitabine (GEM) and tocotrienols, and physicochemically characterize and evaluate the antipancreatic effects of the nanoformulation on Panc 10.05, SW 1990, AsPC-1 and BxPC-3 cells. Materials & methods: Niosomes-entrapping GEM and tocotrienols composed of Span 60, cholesterol and D-α-tocopheryl polyethylene glycol 1000 succinate were produced by Handjani-Vila and film hydration methods. Results: The film hydration produced vesicles measuring 161.9 ± 0.5 nm, approximately 50% smaller in size than Handjani-Vila method, with maximum entrapment efficiencies of 20.07 ± 0.22% for GEM and 34.52 ± 0.10% for tocotrienols. In Panc 10.05 cells, GEM's antiproliferative effect was enhanced 2.78-fold in combination with tocotrienols. Niosomes produced a significant ninefold enhancement in cytotoxicity of the combination, supported by significantly higher cellular uptake of GEM in the cells. Conclusion: This study is a proof of concept on the synthesis of dual-drug niosomes and their efficacy on pancreatic cancer cells in vitro.
Collapse
Affiliation(s)
- Geetha Maniam
- School of Postgraduate Study, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Product Development & Advisory Services Division, Malaysian Palm Oil Board, Bandar Baru Bangi, Selangor, Malaysia
| | - Chun-Wai Mai
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Centre for Cancer & Stem Cells Research, Institute for Research, Development & Innovation, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Mohd Zulkefeli
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Ju-Yen Fu
- Product Development & Advisory Services Division, Malaysian Palm Oil Board, Bandar Baru Bangi, Selangor, Malaysia
| |
Collapse
|
20
|
Anani T, Rahmati S, Sultana N, David AE. MRI-traceable theranostic nanoparticles for targeted cancer treatment. Am J Cancer Res 2021; 11:579-601. [PMID: 33391494 PMCID: PMC7738852 DOI: 10.7150/thno.48811] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Current cancer therapies, including chemotherapy and radiotherapy, are imprecise, non-specific, and are often administered at high dosages - resulting in side effects that severely impact the patient's overall well-being. A variety of multifunctional, cancer-targeted nanotheranostic systems that integrate therapy, imaging, and tumor targeting functionalities in a single platform have been developed to overcome the shortcomings of traditional drugs. Among the imaging modalities used, magnetic resonance imaging (MRI) provides high resolution imaging of structures deep within the body and, in combination with other imaging modalities, provides complementary diagnostic information for more accurate identification of tumor characteristics and precise guidance of anti-cancer therapy. This review article presents a comprehensive assessment of nanotheranostic systems that combine MRI-based imaging (T1 MRI, T2 MRI, and multimodal imaging) with therapy (chemo-, thermal-, gene- and combination therapy), connecting a range of topics including hybrid treatment options (e.g. combined chemo-gene therapy), unique MRI-based imaging (e.g. combined T1-T2 imaging, triple and quadruple multimodal imaging), novel targeting strategies (e.g. dual magnetic-active targeting and nanoparticles carrying multiple ligands), and tumor microenvironment-responsive drug release (e.g. redox and pH-responsive nanomaterials). With a special focus on systems that have been tested in vivo, this review is an essential summary of the most advanced developments in this rapidly evolving field.
Collapse
|
21
|
Zhang J, Zhou L, Zhang Y, He H, Yin T, Gou J, Wang Y, Tang X. Contrastive Studies of Cytarabine/Daunorubicin Dual-Loaded Liposomes Prepared by pH Gradient and Cu 2+ Gradient Method. AAPS PharmSciTech 2020; 21:325. [PMID: 33206247 DOI: 10.1208/s12249-020-01867-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022] Open
Abstract
Conventional combination chemotherapy often leads to unsatisfactory clinical outcomes due to the different distribution characteristics in vivo and the superimposed systemic toxicity of the drug cocktail. Co-encapsulated nano preparations have been gradually developed in recent years. In this work, cytarabine (Ara-C)/daunorubicin (DNR) liposomes were prepared by the pH gradient (ADL-pH) and Cu2+ gradient (ADL-Cu) methods. Ara-C did not show significant release from either ADL-Cu or ADL-pH in vitro during 168 h, which related to its logPoct. Different drug-loading patterns showed different release characteristics of DNR due to the different existence forms, ADL-pH contains the citrate form, while in ADL-Cu, there is the Cu2+ complex. To evaluate the release behavior, daunorubicin liposome (DL) and daunorubicin-Cu2+ complex (DNR-Cu) were prepared. The addition of EDTA in the release medium significantly increased the release rate of DNR from DL-Cu, while lower pH accelerated DNR release from both DL-pH and DL-Cu. The PK confirmed that ADL-Cu and ADL-pH could prolong the drug circulation time, and ADL-Cu had a mean retention time 1.5 times that of ADL-pH. Furthermore, both liposomes allowed the two drugs to maintain a relatively constant plasma concentration ratio for a prolonged time. Cytotoxicity assays showed that Ara-C/DNR with a molar ratio of 5:1 and 3:1 exhibited an excellent synergistic effect, which was more obvious at 5:1. In vitro antitumor results revealed that ADL-Cu exhibited more cytotoxicity than ADL-pH. All factors tested in this work suggest the considerable potential of ADL-Cu and ADL-pH for anticancer treatment.
Collapse
|
22
|
Yu H, Bao Y, Xu C, Chen L, Tang Z. Poly(L-Glutamic Acid)-Drug Conjugates for Chemo- and Photodynamic Combination Therapy. Macromol Biosci 2020; 21:e2000192. [PMID: 33043592 DOI: 10.1002/mabi.202000192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/26/2020] [Indexed: 11/09/2022]
Abstract
Despite the polymeric vascular disrupting agent (poly(L -glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4) nanoparticles can efficiently inhibit cancer growth, their further application is still a challenge owing to the tumor recurrence and metastasis after treatment. In this study, two poly(L -glutamic acid)-drug conjugates for chemo-and photodynamic combination therapy are fabricated. PLG-g-mPEG-CA4 nanoparticles are prepared by combretastatin A4 (CA4) and poly(L -glutamic acid)-graft-methoxy poly(ethylene glycol) (PLG-g-mPEG) using the Yamaguchi esterification reaction. PLG-g-mPEG-TPP (TPP: 5, 10, 15, 20-tetraphenylporphyrin) nanoparticles are constructed using PLG-g-mPEG and amine porphyrin through condensation reaction between carboxyl group of PLG-g-mPEG and amino group of porphyrin. The results showed that PLG-g-mPEG-CA4 nanoparticles have good antitumor ability. PLG-g-mPEG-TPP nanoparticles can produce singlet oxygen under the laser irradiation. Moreover, the combined therapy of PLG-g-mPEG-CA4 and PLG-g-mPEG-TPP nanoparticles has higher antitumor effect than the single chemotherapy or the single photodynamic therapy in vitro. The combination of CA4 nondrug and photodynamic therapy provides a new insight for enhancing the tumor therapeutic effect with vascular disrupting agents and other therapy.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Yanli Bao
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China
| | - Caina Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Li Chen
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| |
Collapse
|
23
|
Bellat V, Verchère A, Ashe SA, Law B. Transcriptomic insight into salinomycin mechanisms in breast cancer cell lines: synergistic effects with dasatinib and induction of estrogen receptor β. BMC Cancer 2020; 20:661. [PMID: 32678032 PMCID: PMC7364656 DOI: 10.1186/s12885-020-07134-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tumors are heterogeneous in nature, composed of different cell populations with various mutations and/or phenotypes. Using a single drug to encounter cancer progression is generally ineffective. To improve the treatment outcome, multiple drugs of distinctive mechanisms but complementary anticancer activities (combination therapy) are often used to enhance antitumor efficacy and minimize the risk of acquiring drug resistance. We report here the synergistic effects of salinomycin (a polyether antibiotic) and dasatinib (a Src kinase inhibitor). METHODS Functionally, both drugs induce cell cycle arrest, intracellular reactive oxygen species (iROS) production, and apoptosis. We rationalized that an overlapping of the drug activities should offer an enhanced anticancer effect, either through vertical inhibition of the Src-STAT3 axis or horizontal suppression of multiple pathways. We determined the toxicity induced by the drug combination and studied the kinetics of iROS production by fluorescence imaging and flow cytometry. Using genomic and proteomic techniques, including RNA-sequencing (RNA-seq), reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Western Blot, we subsequently identified the responsible pathways that contributed to the synergistic effects of the drug combination. RESULTS Compared to either drug alone, the drug combination showed enhanced potency against MDA-MB-468, MDA-MB-231, and MCF-7 human breast cancer (BC) cell lines and tumor spheroids. The drug combination induces both iROS generation and apoptosis in a time-dependent manner, following a 2-step kinetic profile. RNA-seq data revealed that the drug combination exhibited synergism through horizontal suppression of multiple pathways, possibly through a promotion of cell cycle arrest at the G1/S phase via the estrogen-mediated S-phase entry pathway, and partially via the BRCA1 and DNA damage response pathway. CONCLUSION Transcriptomic analyses revealed for the first time, that the estrogen-mediated S-phase entry pathway partially contributed to the synergistic effect of the drug combination. More importantly, our studies led to the discoveries of new potential therapeutic targets, such as E2F2, as well as a novel drug-induced targeting of estrogen receptor β (ESR2) approach for triple-negative breast cancer treatment, currently lacking of targeted therapies.
Collapse
Affiliation(s)
- Vanessa Bellat
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Alice Verchère
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Sally A Ashe
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Benedict Law
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA. .,Lead contact, New York, USA.
| |
Collapse
|
24
|
Ren Z, Sun S, Sun R, Cui G, Hong L, Rao B, Li A, Yu Z, Kan Q, Mao Z. A Metal-Polyphenol-Coordinated Nanomedicine for Synergistic Cascade Cancer Chemotherapy and Chemodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906024. [PMID: 31834662 DOI: 10.1002/adma.201906024] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/12/2019] [Indexed: 05/11/2023]
Abstract
The clinical application of chemotherapy is impeded by the unsatisfactory efficacy and severe side effects. Chemodynamic therapy (CDT) has emerged as an efficient strategy for cancer treatment utilizing Fenton chemistry to destroy cancer cells by converting endogenous H2 O2 into highly toxic reactive oxygen species. Apart from the chemotherapeutic effect, cisplatin is able to act as an artificial enzyme to produce H2 O2 for CDT through cascade reactions, thus remarkably improving the anti-tumor outcomes. Herein, an organic theranostic nanomedicine (PTCG NPs) is constructed with high loading capability using epigallocatechin-3-gallate (EGCG), phenolic platinum(IV) prodrug (Pt-OH), and polyphenol modified block copolymer (PEG-b-PPOH) as the building blocks. The high stability of PTCG NPs during circulation stems from their strong metal-polyphenol coordination interactions, and efficient drug release is realized after cellular internalization. The activated cisplatin elevates the intracellular H2 O2 level through cascade reactions. This is further utilized to produce highly toxic reactive oxygen species catalyzed by an iron-based Fenton reaction. In vitro and in vivo investigations demonstrate that the combination of chemotherapy and chemodynamic therapy achieves excellent anticancer efficacy. Meanwhile, systemic toxicity faced by platinum-based drugs is avoided through this nanoformulation. This work provides a promising strategy to develop advanced nanomedicine for cascade cancer therapy.
Collapse
Affiliation(s)
- Zhigang Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shichao Sun
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Ranran Sun
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Guangying Cui
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Benchen Rao
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ang Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zujiang Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Quancheng Kan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| |
Collapse
|
25
|
Wu Y, Lv S, Li Y, He H, Ji Y, Zheng M, Liu Y, Yin L. Co-delivery of dual chemo-drugs with precisely controlled, high drug loading polymeric micelles for synergistic anti-cancer therapy. Biomater Sci 2019; 8:949-959. [PMID: 31840696 DOI: 10.1039/c9bm01662g] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Simultaneous delivery of multiple chemotherapeutics using polymeric micelles often suffers from unsatisfactory drug loading, drug ratio management, and drug release. Herein, we report a feasible strategy to prepare micelles with ultra-high drug loading and a controllable drug ratio through the introduction of donor-acceptor interactions between drugs and polymeric carriers. An amphiphilic copolymer modified with phenylboronic acid moieties on the hydrophobic segment was synthesized, in which phenylboronic acid functioned as an electron acceptor and formed donor-acceptor coordination with doxorubicin (DOX) and irinotecan (IR). The obtained dual-drug-loaded micelles possessed high drug loading (up to 50%), a tunable drug ratio, and a uniform particle size. Furthermore, both of the encapsulated drug cargoes could be effectively and selectively released in cancer cells with over-produced reactive oxygen species (ROS), and thus the drug-loaded micelles exhibited synergistic anticancer efficacy and reduced systemic toxicity.
Collapse
Affiliation(s)
- Yuchen Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Shixian Lv
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China. and Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Yongjuan Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Hua He
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Yong Ji
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China.
| | - Mingfeng Zheng
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China.
| | - Yong Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| |
Collapse
|
26
|
Xu S, Jin Z, Zhang Z, Huang W, Shen Y, Wang Z, Guo S. Precise ratiometric co-loading, co-delivery and intracellular co-release of paclitaxel and curcumin by aid of their conjugation to the same gold nanorods to exert synergistic effects on MCF-7/ADR cells. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
27
|
Levy A, Leynes C, Baig M, Chew SA. The Application of Biomaterials in the Treatment of Platinum‐Resistant Ovarian Cancer. ChemMedChem 2019; 14:1810-1827. [DOI: 10.1002/cmdc.201900450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Arkene Levy
- Department of Pharmacology, College of Medical Sciences Nova Southeastern University 3200 South University Drive Davie FL 33328 USA
| | - Carolina Leynes
- Department Health and Biomedical Sciences University of Texas Rio Grande Valley One West University Boulevard Brownsville TX 78520 USA
| | - Mirza Baig
- Dr. Kiran C. Patel College of Osteopathic Medicine Nova Southeastern University 3200 South University Drive Davie FL 33328 USA
| | - Sue Anne Chew
- Department Health and Biomedical Sciences University of Texas Rio Grande Valley One West University Boulevard Brownsville TX 78520 USA
| |
Collapse
|
28
|
Kang T, Li Y, Wang Y, Zhu J, Yang L, Huang Y, Xiong M, Liu J, Wang S, Huang M, Wei X, Gou M. Modular Engineering of Targeted Dual-Drug Nanoassemblies for Cancer Chemoimmunotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:36371-36382. [PMID: 31490057 DOI: 10.1021/acsami.9b11881] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Combination of chemotherapeutics and immunomodulators can generate synergistic anticancer efficacy, exerting efficient chemoimmunotherapy for cancer treatment. Nanoparticulate delivery systems hold great promise to promote synergistic anticancer efficacy for the codelivery of drugs. However, there remain challenges to precisely coencapsulate and deliver combinational drugs at designed ratios due to the difference of compatibility between drugs and nanocarriers. In this study, coassembled nanoparticles of lipophilic prodrugs (LPs) were designed to codeliver chemotherapeutics and immunomodulators for cancer treatment. Such nanoassemblies (NAs) could act as platforms to ratiometrically coencapsulate chemotherapeutics and immunomodulators. Based on this method, NAs formed by the self-assembly of iRGD peptide derivatives, paclitaxel (PTX) LPs, and imiquimod (R837) LPs were demonstrated to target the tumor at unified pharmacokinetics, further inducing the effective tumor inhibition and tumor recurrence prevention. This work provided an alternative to prepare chemoimmunotherapeutic NAs with advantages of ratiometric drug coencapsulation and unified pharmacokinetics, which may advance the future cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Tianyi Kang
- Department of Biomedical Engineering , Southern University of Science and Technology , Shenzhen , Guangdong , 518055 P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Shrestha B, Tang L, Romero G. Nanoparticles‐Mediated Combination Therapies for Cancer Treatment. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900076] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Binita Shrestha
- Department of Biomedical Engineering University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Liang Tang
- Department of Biomedical Engineering University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Gabriela Romero
- Department of Chemical Engineering University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| |
Collapse
|
30
|
Selenium-doped calcium carbonate nanoparticles loaded with cisplatin enhance efficiency and reduce side effects. Int J Pharm 2019; 570:118638. [PMID: 31449842 DOI: 10.1016/j.ijpharm.2019.118638] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/09/2019] [Accepted: 08/19/2019] [Indexed: 11/20/2022]
Abstract
Osteosarcoma is the bone tumor that most commonly affects children and teenagers with low survival rate because of metastatic relapse or recurrence. Cisplatin is a first-line chemotherapy for osteosarcoma. However, severe side effects limit its use in clinic. Selenium (Se) is an anticarcinogen that can protect normal tissues from side effects of chemotherapy. In this study, nanoparticles were used to co-deliver cisplatin and Se in a synergistic combination. Se-doped and lipid-coated calcium carbonate nanoparticles loaded with cisplatin (Pt/Se@CaCO3 NPs) were prepared by a reverse microemulsion method. The NPs delivered cisplatin and Se to tumour cells at an optimal synergistic ratio of 1:1 (mol/mol) both in vitro and in an osteosarcoma xenograft model. These results demonstrate that Pt/Se@CaCO3 NPs have great prospects for the osteosarcoma therapy.
Collapse
|
31
|
Zhu Z, Pan H, Li Y, Pan W. Evaluation of the Synergism Mechanism of Tamoxifen and Docetaxel by Nanoparticles. Anticancer Agents Med Chem 2019; 19:1991-2000. [PMID: 31267877 DOI: 10.2174/1871520619666190702120829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/22/2019] [Accepted: 05/02/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Our previous studies have shown that Docetaxel (DTX) and Tamoxifen (TMX) loaded nanoparticles(Co-NPs) could exhibit a synergistic effect on estrogen receptor positive cell lines. In the current study,we have studied the synergistic effect of Co-NPs and underlying possible molecular mechanism. METHODS Cell apoptosis assay, pharmacokinetic experiment and immunohistochemistry experiment were used to explore the synergistic effect and underlying possible mechanism in vitro and in vivo. RESULTS Cell apoptosis assay revealed that Co-NPs could mediate cell sensitization to a cytotoxic agent, resulting in remarkable cell apoptosis. In addition, pharmacokinetic experiment research showed that Co-NPs have longer circulation time in vivo, which could prolong the treatment time of the chemotherapeutic drugs. Immunohistochemistry experiment revealed that the Co-NPs could downregulate the expression of P-gp level to reduce the drugs' efflux. CONCLUSION The possible mechanism of the synergistic effect of DTX and TMX by Co-NPs was attributed to the longer in vivo circulation time, significantly increased rate of cell apoptosis and downregulated expression of P-gp level to the tumor cells.
Collapse
Affiliation(s)
- Zhihong Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hao Pan
- College of Pharmacy, Liaoning University, Shenyang 110036, China
| | - Yuenan Li
- College of Pharmacy, Liaoning University, Shenyang 110036, China
| | - Weisan Pan
- College of Pharmacy, Liaoning University, Shenyang 110036, China
| |
Collapse
|
32
|
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: 129] [Impact Index Per Article: 21.5] [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
|
33
|
Leung AWY, Amador C, Wang LC, Mody UV, Bally MB. What Drives Innovation: The Canadian Touch on Liposomal Therapeutics. Pharmaceutics 2019; 11:pharmaceutics11030124. [PMID: 30884782 PMCID: PMC6471263 DOI: 10.3390/pharmaceutics11030124] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 01/09/2023] Open
Abstract
Liposomes are considered one of the most successful drug delivery systems (DDS) given their established utility and success in the clinic. In the past 40–50 years, Canadian scientists have made ground-breaking discoveries, many of which were successfully translated to the clinic, leading to the formation of biotech companies, the creation of research tools, such as the Lipex Extruder and the NanoAssemblr™, as well as contributing significantly to the development of pharmaceutical products, such as Abelcet®, MyoCet®, Marqibo®, Vyxeos®, and Onpattro™, which are making positive impacts on patients’ health. This review highlights the Canadian contribution to the development of these and other important liposomal technologies that have touched patients. In this review, we try to address the question of what drives innovation: Is it the individual, the teams, the funding, and/or an entrepreneurial spirit that leads to success? From this perspective, it is possible to define how innovation will translate to meaningful commercial ventures and products with impact in the future. We begin with a brief history followed by descriptions of drug delivery technologies influenced by Canadian researchers. We will discuss recent advances in liposomal technologies, including the Metaplex technology from the author’s lab. The latter exemplifies how a nanotechnology platform can be designed based on multidisciplinary groups with expertise in coordination chemistry, nanomedicines, disease, and business to create new therapeutics that can effect better outcomes in patient populations. We conclude that the team is central to the effort; arguing if the team is entrepreneurial and well positioned, the funds needed will be found, but likely not solely in Canada.
Collapse
Affiliation(s)
- Ada W Y Leung
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6T 1Z4, Canada.
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada.
- Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Carolyn Amador
- Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Lin Chuan Wang
- Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Urmi V Mody
- Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Marcel B Bally
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6T 1Z4, Canada.
- Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
- Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
34
|
Enrico C. Nanotechnology-Based Drug Delivery of Natural Compounds and Phytochemicals for the Treatment of Cancer and Other Diseases. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2019. [DOI: 10.1016/b978-0-444-64185-4.00003-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
35
|
|
36
|
Novohradsky V, Zajac J, Vrana O, Kasparkova J, Brabec V. Simultaneous delivery of olaparib and carboplatin in PEGylated liposomes imparts this drug combination hypersensitivity and selectivity for breast tumor cells. Oncotarget 2018; 9:28456-28473. [PMID: 29983873 PMCID: PMC6033346 DOI: 10.18632/oncotarget.25466] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/05/2018] [Indexed: 01/09/2023] Open
Abstract
Combination regiments involving platinum anticancer drugs and agents with unrelated mechanisms of action are a subject of widespread interest. Here, we show that synergistic toxic action in cancer cells of combinations of antitumor platinum drug carboplatin and effective PARP inhibitor olaparib is considerably improved if these combined drugs are encapsulated into liposomes. Notably, the formation of such nano-formulations, called OLICARB, leads to a marked enhancement of activity in human cancer cell lines (including those resistant to conventional platinum antitumor drugs) and selectivity towards tumor cells. We used immunofluorescence analysis of γH2AX expression and examined DNA damage in cancerous cells treated with the investigated compounds. We find that the synergistic toxic effects in cancer cells of both drugs used in combination, nonencapsulated or embedded in the OLICARB nanoparticles, positively correlates with DNA damage. These results also suggest that the enhancement of the toxic effects of carboplatin by olaparib in cancer cells is a consequence of an accumulation of cytotoxic lesions in DNA due to the inhibition of repair of platinated DNA augmented by the synergistic action of olaparib as an effective PARP inhibitor. Our findings also reveal that the combination of olaparib with carboplatin encapsulated in the OLICARB nanoparticles is particularly effective to inhibit the growth of 3D mammospheres. Collectively, the data provide convincing evidence that the encapsulation of carboplatin and olaparib into liposomal constructs to form the OLICARB nanoparticles may represent the viable approach for the treatment of tumors with the aim to eliminate the possible effects of acquired resistance.
Collapse
Affiliation(s)
- Vojtech Novohradsky
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Juraj Zajac
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Oldrich Vrana
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Jana Kasparkova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Viktor Brabec
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| |
Collapse
|
37
|
Houdaihed L, Evans JC, Allen C. Codelivery of Paclitaxel and Everolimus at the Optimal Synergistic Ratio: A Promising Solution for the Treatment of Breast Cancer. Mol Pharm 2018; 15:3672-3681. [PMID: 29863881 DOI: 10.1021/acs.molpharmaceut.8b00217] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Clinical studies examining the combination of paclitaxel (PTX) and everolimus (EVER), an mTOR inhibitor, have failed to result in significant improvements in efficacy and toxicity in patients with breast cancer (BC), relative to treatment with PTX alone. These disappointing clinical trial results have been attributed to poorly designed preclinical studies using the combination of PTX and EVER as well as the significantly different pharmacokinetic profiles of the two drugs. In the current work, the potential synergy between PTX and EVER was examined in a panel of six BC cell lines that differ in terms of their molecular subtype and drug sensitivity. Polymeric nanoparticles (NPs) were used to encapsulate PTX and EVER at an optimal synergistic ratio to achieve specific, colocalized delivery of the combination therapy in BC cell lines. Combinations of PTX and EVER (especially at relatively high doses of EVER) resulted in pronounced synergy in all BC cell lines evaluated. The optimal molar ratio of PTX:EVER was determined to be 1:0.5. The combination was delivered to BC cells at the synergistic ratio via encapsulation within polymeric NPs formed from the poly(ethylene glycol)- b-poly(lactide- co-glycolide) (PEG- b-PLGA) copolymer. The NPs had an average diameter of less than 100 nm and were capable of in vitro retention of the encapsulated PTX and EVER at the optimal synergistic molar ratio for over 7 days. Cytotoxicity data demonstrated that PTX+EVER-loaded NPs were significantly less cytotoxic than the free drug combination in MCF-7 and SKBR3 BC cell lines following 72 h, suggesting that PTX+EVER-loaded NPs remain stable and retain the drug combination loaded within the core after 72 h. The uptake of FITC-labeled NPs in SKBR3 cells was evaluated by flow cytometry, with approximately 41% of cells demonstrating detectable fluorescence after 24 h of exposure. The thorough and systematic approach used in this study to determine and evaluate a synergistic PTX:EVER ratio in conjunction with a potentially promising delivery vector for the drug combination could offer a future clinical benefit for patients with BC.
Collapse
Affiliation(s)
- Loujin Houdaihed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto M5S 3M2 , Canada
| | - James C Evans
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto M5S 3M2 , Canada
| | - Christine Allen
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto M5S 3M2 , Canada
| |
Collapse
|
38
|
Tolcher AW, Mayer LD. Improving combination cancer therapy: the CombiPlex® development platform. Future Oncol 2018; 14:1317-1332. [DOI: 10.2217/fon-2017-0607] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Current combination therapy approaches assume that better outcomes are achieved by combining drugs at their maximally tolerated doses. However, administration of individual agents cannot consistently deliver synergistic drug ratios to tumor cells due to differences in pharmacokinetics of the individual drugs. Further, the toxicity of combination regimens often necessitates administration of suboptimal dosages. Delivery technologies, such as the CombiPlex® platform, can enable efficient and sustained delivery of combination treatments at a synergistic ratio. The CombiPlex platform determines synergistic drug ratios in vitro and identifies an appropriate nanoscale carrier to maintain that ratio in vivo and enhance its delivery to tumor cells. CPX-351, a dual-drug liposomal encapsulation of cytarabine and daunorubicin, is the first clinical proof-of-concept example of the CombiPlex platform.
Collapse
Affiliation(s)
- Anthony W Tolcher
- South Texas Accelerated Research Therapeutics, Suite 4022, 4383 Medical Drive, San Antonio, TX 78230, USA
| | - Lawrence D Mayer
- Jazz Pharmaceuticals, plc, Suite 250, 887 Great Northern Way, Vancouver, British Columbia V5T 4T5, Canada
| |
Collapse
|
39
|
Hu Y, Miller M, Zhang B, Nguyen TT, Nielsen MK, Aroian RV. In vivo and in vitro studies of Cry5B and nicotinic acetylcholine receptor agonist anthelmintics reveal a powerful and unique combination therapy against intestinal nematode parasites. PLoS Negl Trop Dis 2018; 12:e0006506. [PMID: 29775454 PMCID: PMC5979042 DOI: 10.1371/journal.pntd.0006506] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/31/2018] [Accepted: 05/08/2018] [Indexed: 01/21/2023] Open
Abstract
Background The soil-transmitted nematodes (STNs) or helminths (hookworms, whipworms, large roundworms) infect the intestines of ~1.5 billion of the poorest peoples and are leading causes of morbidity worldwide. Only one class of anthelmintic or anti-nematode drugs, the benzimidazoles, is currently used in mass drug administrations, which is a dangerous situation. New anti-nematode drugs are urgently needed. Bacillus thuringiensis crystal protein Cry5B is a powerful, promising new candidate. Drug combinations, when properly made, are ideal for treating infectious diseases. Although there are some clinical trials using drug combinations against STNs, little quantitative and systemic work has been performed to define the characteristics of these combinations in vivo. Methodology/Principal findings Working with the hookworm Ancylostoma ceylanicum-hamster infection system, we establish a laboratory paradigm for studying anti-nematode combinations in vivo using Cry5B and the nicotinic acetylcholine receptor (nAChR) agonists tribendimidine and pyrantel pamoate. We demonstrate that Cry5B strongly synergizes in vivo with both tribendimidine and pyrantel at specific dose ratios against hookworm infections. For example, whereas 1 mg/kg Cry5B and 1 mg/kg tribendimidine individually resulted in only a 0%-6% reduction in hookworm burdens, the combination of the two resulted in a 41% reduction (P = 0.020). Furthermore, when mixed at synergistic ratios, these combinations eradicate hookworm infections at doses where the individual doses do not. Using cyathostomin nematode parasites of horses, we find based on inhibitory concentration 50% values that a strongylid parasite population doubly resistant to nAChR agonists and benzimidazoles is more susceptible or “hypersusceptible” to Cry5B than a cyathostomin population not resistant to nAChR agonists, consistent with previous Caenhorhabditis elegans results. Conclusions/Significance Our study provides a powerful means by which anthelmintic combination therapies can be examined in vivo in the laboratory. In addition, we demonstrate that Cry5B and nAChR agonists have excellent combinatorial properties—Cry5B combined with nAChR agonists gives rise to potent cures that are predicted to be recalcitrant to the development of parasite resistance. These drug combinations highlight bright spots in new anthelmintic development for human and veterinary animal intestinal nematode infections. Intestinal nematodes are roundworm parasites of humans and animals, causing significant morbidity in both. In humans, these parasites are leading causes of morbidity in children, e.g., causing growth stunting, cognitive impairment, and malnutrition. Few drugs are used to treat these parasites in humans and animals and there is increasing evidence that the drugs are losing efficacy and/or have low efficacy. Infectious diseases are best treated with drug combinations and not single drugs. However, there has been little work to characterize in detail how various anti-nematode drugs combine. Here we establish a new laboratory model to study anti-nematode drug combinations using the human hookworm Ancylostoma ceylanicum infection in hamsters. We show that two classes of anti-nematode drugs, Cry5B and the nicotinic acetylcholine receptor agonists tribendimidine and pyrantel, combine (synergize) in a way that is more powerful at specific drug ratios than predicted from their individual impacts. Furthermore, when combined at these ratios, these combinations completely eliminated parasites at doses where normally neither drug has that effect. Horse parasites resistant to pyrantel also appear to be hypersensitive (more sensitive than wild-type parasites) to Cry5B. These characteristics predict that combinations of Cry5B with tribendimidine or pyrantel will be extremely effective therapeutically and relatively recalcitrant to the development of parasite resistance.
Collapse
Affiliation(s)
- Yan Hu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Melanie Miller
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Bo Zhang
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Thanh-Thanh Nguyen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Martin K. Nielsen
- Department of Veterinary Science, University of Kentucky, Lexington, KY, United States of America
| | - Raffi V. Aroian
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
- * E-mail:
| |
Collapse
|
40
|
Cheng Y, Zhao P, Wu S, Yang T, Chen Y, Zhang X, He C, Zheng C, Li K, Ma X, Xiang G. Cisplatin and curcumin co-loaded nano-liposomes for the treatment of hepatocellular carcinoma. Int J Pharm 2018; 545:261-273. [PMID: 29730175 DOI: 10.1016/j.ijpharm.2018.05.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/24/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) continues to be a leading cause of cancer related death in the world. Conventional chemotherapeutic agents such as cisplatin (CDDP) have an unsatisfactory efficacy on HCC due to the poor response, severe toxicity and drug resistance. Curcumin (CUR) could improve the chemosensitivity of HCC to chemotherapy drugs by regulating a variety of signaling pathways. Herein, we describe a combination strategy using co-loaded liposomes to effectively deliver and release CDDP and curcumin (CUR) to HCC for overcoming the unsatisfactory clinical outcome of CDDP monotherapy. In the study, CDDP and CUR co-loaded liposomes (CDDP/CUR-Lip) were prepared by a reverse microemulsion and film dispersion method and their average particle size 294.6 ± 14.8 nm with uniform size distribution. In vitro study showed that the nano sized CDDP/CUR-Lip could synchronously release both CDDP and CUR to achieve the synergistic effect against HCC cells based on the optimal ratio (1:8) of both drugs. Compared with free drug or encapsulated mono-drug therapy, CDDP/CUR-Lip demonstrated the higher anti-tumor activity in vitro against HepG2 cells with the IC50 of 0.62 μM. In addition, CDDP/CUR-Lip also increased intracellular ROS level during the HCC cells treatment. Furthermore, compared with single drug formulation, CDDP/CUR-Lip showed the elongated retention time (t1/2 = 2.38 h) and improved antitumor effect in both mouse hepatoma H22 and human HCC HepG2 xenograft models with reduced side effects. In conclusion, CDDP/CUR-Lip provide an attractive and potential strategy to attain synergistic effect of CDDP and CUR for the treatment of HCC.
Collapse
Affiliation(s)
- Yao Cheng
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Pengxuan Zhao
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shuangping Wu
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tan Yang
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yan Chen
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaojuan Zhang
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chuanchuan He
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chao Zheng
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Kelin Li
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiang Ma
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Guangya Xiang
- Pharmacy School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
41
|
Wan X, Min Y, Bludau H, Keith A, Sheiko SS, Jordan R, Wang AZ, Sokolsky-Papkov M, Kabanov AV. Drug Combination Synergy in Worm-like Polymeric Micelles Improves Treatment Outcome for Small Cell and Non-Small Cell Lung Cancer. ACS NANO 2018; 12:2426-2439. [PMID: 29533606 PMCID: PMC5960350 DOI: 10.1021/acsnano.7b07878] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nanoparticle-based systems for concurrent delivery of multiple drugs can improve outcomes of cancer treatments, but face challenges because of differential solubility and fairly low threshold for incorporation of many drugs. Here we demonstrate that this approach can be used to greatly improve the treatment outcomes of etoposide (ETO) and platinum drug combination ("EP/PE") therapy that is the backbone for treatment of prevalent and deadly small cell lung cancer (SCLC). A polymeric micelle system based on amphiphilic block copolymer poly(2-oxazoline)s (POx) poly(2-methyl-2-oxazoline- block-2-butyl-2-oxazoline- block-2-methyl-2-oxazoline) (P(MeOx- b-BuOx- b-MeOx) is used along with an alkylated cisplatin prodrug to enable co-formulation of EP/PE in a single high-capacity vehicle. A broad range of drug mixing ratios and exceptionally high two-drug loading of over 50% wt. drug in dispersed phase is demonstrated. The highly loaded POx micelles have worm-like morphology, unprecedented for drug loaded polymeric micelles reported so far, which usually form spheres upon drug loading. The drugs co-loading in the micelles result in a slowed-down release, improved pharmacokinetics, and increased tumor distribution of both drugs. A superior antitumor activity of co-loaded EP/PE drug micelles compared to single drug micelles or their combination as well as free drug combination was demonstrated using several animal models of SCLC and non-small cell lung cancer.
Collapse
Affiliation(s)
- Xiaomeng Wan
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Herdis Bludau
- Chair of Macromolecular Chemistry, Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstr. 4, 01069 Dresden, Germany
| | - Andrew Keith
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Sergei S. Sheiko
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rainer Jordan
- Chair of Macromolecular Chemistry, Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstr. 4, 01069 Dresden, Germany
| | - Andrew Z. Wang
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Chair of Macromolecular Chemistry, Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstr. 4, 01069 Dresden, Germany
- Department of Radiation Oncology, Xuzhou Medical College, Xuzhou 221100, China
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Corresponding Authors: .
| | - Alexander V. Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
- Corresponding Authors: .
| |
Collapse
|
42
|
Co-delivery of doxorubicin and imatinib by pH sensitive cleavable PEGylated nanoliposomes with folate-mediated targeting to overcome multidrug resistance. Int J Pharm 2018; 542:266-279. [PMID: 29551747 DOI: 10.1016/j.ijpharm.2018.03.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 01/23/2023]
Abstract
Multidrug resistance to chemotherapeutic drugs is a major obstacle to breast cancer treatment. In this study, doxorubicin (DOX) and imatinib (IM) were co-loaded into folate receptor targeted (FR-targeted) pH-sensitive liposomes (denoted as FPL-DOX/IM) to fulfill intracellular acid-sensitive release and reverse drug resistance. FPL-DOX/IM could maintain stability in blood circulation with approximate diameters of 100 nm and rapidly release encapsulated drugs in tumor acidic microenvironment. Moreover, the IM in combination therapy could overcome chemoresistance associated with DOX effectively by inhibiting ABC transporter function and improving chemotherapy sensitivity. The designed liposomes co-loaded with DOX and IM significantly enhanced anti-tumor effects both in vitro and in vivo. These findings suggest that FPL-DOX/IM provides a novel strategy to improve chemotherapeutic efficacy against MDR tumors.
Collapse
|
43
|
Zhang M, Liu E, Cui Y, Huang Y. Nanotechnology-based combination therapy for overcoming multidrug-resistant cancer. Cancer Biol Med 2017; 14:212-227. [PMID: 28884039 PMCID: PMC5570599 DOI: 10.20892/j.issn.2095-3941.2017.0054] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/03/2017] [Indexed: 12/28/2022] Open
Abstract
Multidrug resistance (MDR) is a major obstacle to successful cancer treatment and is crucial to cancer metastasis and relapse. Combination therapy is an effective strategy for overcoming MDR. However, the different pharmacokinetic (PK) profiles of combined drugs often undermine the combination effect in vivo, especially when greatly different physicochemical properties (e.g., those of macromolecules and small drugs) combine. To address this issue, nanotechnology-based codelivery techniques have been actively explored. They possess great advantages for tumor targeting, controlled drug release, and identical drug PK profiles. Thus, a powerful tool for combination therapy is provided, and the translation from in vitro to in vivo is facilitated. In this review, we present a summary of various combination strategies for overcoming MDR and the nanotechnology-based combination therapy.
Collapse
Affiliation(s)
- Meng Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ergang Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanna Cui
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
44
|
Zhu Z, Li Y, Yang X, Pan W, Pan H. The reversion of anti-cancer drug antagonism of tamoxifen and docetaxel by the hyaluronic acid-decorated polymeric nanoparticles. Pharmacol Res 2017; 126:84-96. [PMID: 28734999 DOI: 10.1016/j.phrs.2017.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/24/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022]
Abstract
Docetaxel (DTX) and tamoxifen (TMX) are first-line drugs used to treat breast cancer. However when used in combination, they produce antagonism because of their differential metabolic pathways. In order to prevent this antagonism, an amphiphilic copolymer, cholesterol modified hyaruronic acid (HA-CHOL), was synthesized for investigating the co-delivery of TMX and DTX. In vitro drug release experiment of the Co-encapsulated (encapsulated DTX+TMX) nanoparticles (Co-NPs) revealed that NPs with unique release mechanism can markedly reduce the release of these drugs in the circulatory system. However, when reaching in cell, TMX can release rapidly to prevent DTX from coming into contact with metabolizing enzymes. In vitro cytotoxicity experiment revealed that the Co-NPs exhibited a significant synergistic effect for inhibiting the proliferation of the cancer cell lines A549, MCF7 and S180. NPs carrying Coumarin-6(Cou6) exhibited increased cellular uptake compared with Cou6 solution at similar drug concentrations. As an in vivo treatment of xenograft tumors involving 180 cells, the Co-NPs displayed a clear tumor-inhibiting effect. This led us to conclude that the reversion of drug antagonism by NPs was attributed to the increased stability of the nanoparticles in the blood circulation, the efficient cellular uptake, the hierarchical drug metabolism in the tumor and the good and orderly delivery of the drugs to the tumor tissue.
Collapse
Affiliation(s)
- Zhihong Zhu
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Yuenan Li
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Xinggang Yang
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Weisan Pan
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China.
| | - Hao Pan
- College of Pharmacy, Liaoning University, Shenyang, 110036, China, China.
| |
Collapse
|
45
|
Dai W, Wang X, Song G, Liu T, He B, Zhang H, Wang X, Zhang Q. Combination antitumor therapy with targeted dual-nanomedicines. Adv Drug Deliv Rev 2017; 115:23-45. [PMID: 28285944 DOI: 10.1016/j.addr.2017.03.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
Combination therapy is one of the important treatment strategies for cancer at present. However, the outcome of current combination therapy based on the co-administration of conventional dosage forms is suboptimal, due to the short half-lives of chemodrugs, their deficient tumor selectivity and so forth. Nanotechnology-based targeted delivery systems show great promise in addressing the associated problems and providing superior therapeutic benefits. In this review, we focus on the combination of therapeutic strategies between different nanomedicines or drug-loaded nanocarriers, rather than the co-delivery of different drugs via a single nanocarrier. We introduce the general concept of various targeting strategies of nanomedicines, present the principles of combination antitumor therapy with dual-nanomedicines, analyze their advantages and limitations compared with co-delivery strategies, and overview the recent advances of combination therapy based on targeted nanomedicines. Finally, we reviewed the challenges and future perspectives regarding the selection of therapeutic agents, targeting efficiency and the gap between the preclinical and clinical outcome.
Collapse
Affiliation(s)
- Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyou Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Tongzhou Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China.
| |
Collapse
|
46
|
Parvanian S, Mostafavi SM, Aghashiri M. Multifunctional nanoparticle developments in cancer diagnosis and treatment. SENSING AND BIO-SENSING RESEARCH 2017. [DOI: 10.1016/j.sbsr.2016.08.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
47
|
Liang Y, Tian B, Zhang J, Li K, Wang L, Han J, Wu Z. Tumor-targeted polymeric nanostructured lipid carriers with precise ratiometric control over dual-drug loading for combination therapy in non-small-cell lung cancer. Int J Nanomedicine 2017; 12:1699-1715. [PMID: 28280336 PMCID: PMC5340246 DOI: 10.2147/ijn.s121262] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gemcitabine (GEM) and paclitaxel (PTX) are effective combination anticancer agents against non-small-cell lung cancer (NSCLC). At the present time, a main challenge of combination treatment is the precision of control that will maximize the combined effects. Here, we report a novel method to load GEM (hydrophilic) and PTX (hydrophobic) into simplex tumor-targeted nanostructured lipid carriers (NLCs) for accurate control of the ratio of the two drugs. We covalently preconjugated the dual drugs through a hydrolyzable ester linker to form drug conjugates. N-acetyl-d-glucosamine (NAG) is a glucose receptor-targeting ligand. We added NAG to the formation of NAG-NLCs. In general, synthesis of poly(6-O-methacryloyl-d-galactopyranose)–GEM/PTX (PMAGP-GEM/PTX) conjugates was demonstrated, and NAG-NLCs were prepared using emulsification and solvent evaporation. NAG-NLCs displayed sphericity with an average diameter of 120.3±1.3 nm, a low polydispersity index of 0.233±0.04, and accurate ratiometric control over the two drugs. A cytotoxicity assay showed that the NAG-NLCs had better antitumor activity on NSCLC cells than normal cells. There was an optimal ratio of the two drugs, exhibiting the best cytotoxicity and combinatorial effects among all the formulations we tested. In comparison with both the free-drug combinations and separately nanopackaged drug conjugates, PMAGP-GEM/PTX NAG-NLCs (3:1) exhibited superior synergism. Flow cytometry and confocal laser scanning microscopy showed that NAG-NLCs exhibited higher uptake efficiency in A549 cells via glucose receptor-mediated endocytosis. This combinatorial delivery system settles problems with ratiometric coloading of hydrophilic and hydrophobic drugs for tumor-targeted combination therapy to achieve maximal anticancer efficacy in NSCLC.
Collapse
Affiliation(s)
- Yan Liang
- School of Pharmacy, Binzhou Medical University
| | | | - Jing Zhang
- School of Pharmacy, Binzhou Medical University
| | - Keke Li
- School of Pharmacy, Binzhou Medical University
| | - Lele Wang
- School of Pharmacy, Binzhou Medical University
| | | | - Zimei Wu
- School of Pharmacy, Yantai University, Yantai, China
| |
Collapse
|
48
|
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: 213] [Impact Index Per Article: 26.6] [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]
|
49
|
Abstract
The intrinsic limits of conventional cancer therapies prompted the development and application of various nanotechnologies for more effective and safer cancer treatment, herein referred to as cancer nanomedicine. Considerable technological success has been achieved in this field, but the main obstacles to nanomedicine becoming a new paradigm in cancer therapy stem from the complexities and heterogeneity of tumour biology, an incomplete understanding of nano-bio interactions and the challenges regarding chemistry, manufacturing and controls required for clinical translation and commercialization. This Review highlights the progress, challenges and opportunities in cancer nanomedicine and discusses novel engineering approaches that capitalize on our growing understanding of tumour biology and nano-bio interactions to develop more effective nanotherapeutics for cancer patients.
Collapse
Affiliation(s)
- Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
50
|
Poon C, Duan X, Chan C, Han W, Lin W. Nanoscale Coordination Polymers Codeliver Carboplatin and Gemcitabine for Highly Effective Treatment of Platinum-Resistant Ovarian Cancer. Mol Pharm 2016; 13:3665-3675. [PMID: 27712076 PMCID: PMC5673481 DOI: 10.1021/acs.molpharmaceut.6b00466] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Due to the ability of ovarian cancer (OCa) to acquire drug resistance, it has been difficult to develop efficient and safe chemotherapy for OCa. Here, we examined the therapeutic use of a new self-assembled core-shell nanoscale coordination polymer nanoparticle (NCP-Carbo/GMP) that delivers high loadings of carboplatin (28.0 ± 2.6 wt %) and gemcitabine monophosphate (8.6 ± 1.5 wt %). A strong synergistic effect was observed between carboplatin and gemcitabine against platinum-resistant OCa cells, SKOV-3 and A2780/CDPP, in vitro. The coadministration of carboplatin and gemcitabine in the NCP led to prolonged blood circulation half-life (11.8 ± 4.8 h) and improved tumor uptake of the drugs (10.2 ± 4.4% ID/g at 24 h), resulting in 71% regression and 80% growth inhibition of SKOV-3 and A2780/CDDP tumors, respectively. Our findings demonstrate that NCP particles provide great potential for the codelivery of multiple chemotherapeutics for treating drug-resistant cancer.
Collapse
Affiliation(s)
| | | | - Christina Chan
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Wenbo Han
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| |
Collapse
|