1
|
Shukla RP, Tiwari P, Sardar A, Urandur S, Gautam S, Marwaha D, Tripathi AK, Rai N, Trivedi R, Mishra PR. Alendronate-functionalized porous nano-crystalsomes mitigate osteolysis and consequent inhibition of tumor growth in a tibia-induced metastasis model. J Control Release 2024; 372:331-346. [PMID: 38844176 DOI: 10.1016/j.jconrel.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
Bone is one of the most prevalent sites of metastases in various epithelial malignancies, including breast cancer and this metastasis to bone often leads to severe skeletal complications in women due to its osteolytic nature. To address this, we devised a novel drug delivery approach using an Alendronate (ALN) functionalized self-assembled porous crystalsomes for concurrent targeting of Oleanolic acid (OA) and ALN (ALN + OA@NCs) to bone metastasis. Initially, the conjugation of both PEG-OA and OA-PEG-ALN with ALN and OA was achieved, and this conjugation was then self-assembled into porous crystalsomes (ALN + OA@NCs) by nanoemulsion crystallization. The reconstruction of a 3D single particle using transmission electron microscopy ensured the crystalline porous structure of ALN + OA@NCs, was well aligned with characteristic nanoparticle attributes including size distribution, polydispersity, and zeta potential. Further, ALN + OA@NCs showed enhanced efficacy in comparison to OA@NCs suggesting the cytotoxic roles of ALN towards cancer cells, followed by augmentation ROS generation (40.81%), mitochondrial membrane depolarization (57.20%), and induction of apoptosis (40.43%). We found that ALN + OA@NCs facilitated inhibiting osteoclastogenesis and bone resorption followed by inhibited osteolysis. In vivo activity of ALN + OA@NCs in the 4 T1 cell-induced tibia model rendered a reduced bone loss in the treated mice followed by restoring bone morphometric markers which were further corroborated bone-targeting effects of ALN + OA@NCs to reduce RANKL-stimulated osteoclastogenesis. Further, In vivo intravenous pharmacokinetics showed the improved therapeutic profile of the ALN + OA@NCs in comparison to the free drug, prolonging the levels of the drug in the systemic compartment by reducing the clearance culminating the higher accumulation at the tumor site. Our finding proposed that ALN + OA@NCs can effectively target and treat breast cancer metastasis to bone and its associated complications.
Collapse
Affiliation(s)
- Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirban Sardar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Tiwari P, Yadav K, Shukla RP, Bakshi AK, Panwar D, Das S, Mishra PR. Extracellular vesicles-powered immunotherapy: Unleashing the potential for safer and more effective cancer treatment. Arch Biochem Biophys 2024; 756:110022. [PMID: 38697343 DOI: 10.1016/j.abb.2024.110022] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Cancer treatment has seen significant advancements with the introduction of Onco-immunotherapies (OIMTs). Although some of these therapies have received approval for use, others are either undergoing testing or are still in the early stages of development. Challenges persist in making immunotherapy widely applicable to cancer treatment. To maximize the benefits of immunotherapy and minimize potential side effects, it's essential to improve response rates across different immunotherapy methods. A promising development in this area is the use of extracellular vesicles (EVs) as novel delivery systems. These small vesicles can effectively deliver immunotherapies, enhancing their effectiveness and reducing harmful side effects. This article discusses the importance of integrating nanomedicines into OIMTs, highlighting the challenges with current anti-OIMT methods. It also explores key considerations for designing nanomedicines tailored for OIMTs, aiming to improve upon existing immunotherapy techniques. Additionally, the article looks into innovative approaches like biomimicry and the use of natural biomaterial-based nanocarriers (NCs). These advancements have the potential to transform the delivery of immunotherapy. Lastly, the article addresses the challenges of moving OIMTs from theory to clinical practice, providing insights into the future of using advanced nanotechnology in cancer treatment.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Sweety Das
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
3
|
Tiwari P, Shukla RP, Yadav K, Panwar D, Agarwal N, Kumar A, Singh N, Bakshi AK, Marwaha D, Gautam S, Rai N, Mishra PR. Exploring nanocarriers as innovative materials for advanced drug delivery strategies in onco-immunotherapies. J Mol Graph Model 2024; 128:108702. [PMID: 38219505 DOI: 10.1016/j.jmgm.2024.108702] [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: 09/12/2023] [Revised: 12/16/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
In recent years, Onco-immunotherapies (OIMTs) have been shown to be a potential therapy option for cancer. Several immunotherapies have received regulatory approval, while many others are now undergoing clinical testing or are in the early stages of development. Despite this progress, a large number of challenges to the broad use of immunotherapies to treat cancer persists. To make immunotherapy more useful as a treatment while reducing its potentially harmful side effects, we need to know more about how to improve response rates to different types of immunotherapies. Nanocarriers (NCs) have the potential to harness immunotherapies efficiently, enhance the efficiency of these treatments, and reduce the severe adverse reactions that are associated with them. This article discusses the necessity to incorporate nanomedicines in OIMTs and the challenges we confront with current anti-OIMT approaches. In addition, it examines the most important considerations for building nanomedicines for OIMT, which may improve upon current immunotherapy methods. Finally, it highlights the applications and future scenarios of using nanotechnology.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
4
|
Tiwari P, Shukla RP, Yadav K, Singh N, Marwaha D, Gautam S, Bakshi AK, Rai N, Kumar A, Sharma D, Mishra PR. Dacarbazine-primed carbon quantum dots coated with breast cancer cell-derived exosomes for improved breast cancer therapy. J Control Release 2024; 365:43-59. [PMID: 37935257 DOI: 10.1016/j.jconrel.2023.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Imprecise targeting of chemotherapeutic drugs often leads to severe toxicity during breast cancer therapy. To address this issue, we have devised a strategy to load dacarbazine (DC) into fucose-based carbon quantum dots (CQDs), which are subsequently coated with exosomes (Ex-DC@CQDs) derived from breast cancer cells. Nanoparticle tracking analysis and western blotting revealed that Ex-DC@CQDs retained the structural and functional characteristics of exosomes. We found that exosomes facilitated the transport of DC@CQDs to cancer cells via heparan sulfate proteoglycan (HSPG) receptors, followed by an augmented depolarization of the mitochondrial membrane potential, ROS generation, and induction of apoptosis leading to cell death. In vivo imaging and pharmacokinetic studies demonstrated enhanced antitumor targeting and efficacy compared to free DC which we attribute to an improved pharmacokinetic profile, a greater tumor accumulation via exosome-mediated- HSPG receptor-driven cell uptake, and sustained release of the Ex-DC@CQDs. Our findings may pave the way for the further development of biologically sourced nanocarriers for breast cancer targeting.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Deepak Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
5
|
Singh N, Marwaha D, Gautam S, Rai N, Tiwari P, Sharma M, Shukla RP, Mugale MN, Kumar A, Mishra PR. Surface-Modified Lyotropic Crystalline Nanoconstructs Bearing Doxorubicin and Buparvaquone Target Sigma Receptors through pH-Sensitive Charge Conversion to Improve Breast Cancer Therapy. Biomacromolecules 2023; 24:5780-5796. [PMID: 38006339 DOI: 10.1021/acs.biomac.3c00795] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
In the current study, we aimed to develop lyotropic crystalline nanoconstructs (LCNs) based on poly(l-glutamic acid) (PLG) with a two-tier strategy. The first objective was to confer pH-responsive charge conversion properties to facilitate the delivery of both doxorubicin (DOX) and buparvaquone (BPQ) in combination (B + D@LCNs) to harness their synergistic effects. The second goal was to achieve targeted delivery to sigma receptors within the tumor tissues. To achieve this, we designed a pH-responsive charge conversion system using a polymer consisting of poly(ethylenimine), poly(l-lysine), and poly(l-glutamic acid) (PLG), which was then covalently coupled with methoxybenzamide (MBA) for potential sigma receptor targeting. The resulting B + D@LCNs were further modified by surface functionalization with PLG-MBA to confer both sigma receptor targeting and pH-responsive charge conversion properties. Our observations indicated that at physiological pH 7.4, P/B + D-MBA@LCNs exhibited a negative charge, while under acidic conditions (pH 5.5, characteristic of the tumor microenvironment), they acquired a positive charge. The particle size of P/B + D-MBA@LCNs was determined to be 168.23 ± 2.66 nm at pH 7.4 and 201.23 ± 1.46 nm at pH 5.5. The crystalline structure of the LCNs was confirmed through small-angle X-ray scattering (SAXS) diffraction patterns. Receptor-mediated endocytosis, facilitated by P/B + D-MBA@LCNs, was confirmed using confocal laser scanning microscopy and flow cytometry. The P/B + D-MBA@LCNs formulation demonstrated a higher rate of G2/M phase arrest (55.20%) compared to free B + D (37.50%) and induced mitochondrial depolarization (59.39%) to a greater extent than P/B + D@LCNs (45.66%). Pharmacokinetic analysis revealed significantly improved area under the curve (AUC) values for both DOX and BPQ when administered as P/B + D-MBA@LCNs, along with enhanced tumor localization. Tumor regression studies exhibited a substantial reduction in tumor size, with P/B + D-MBA@LCNs leading to 3.2- and 1.27-fold reductions compared to B + D and nontargeted P/B + D@LCNs groups, respectively. In summary, this two-tier strategy demonstrates substantial promise for the delivery of a drug combination through the prototype formulation. It offers a potential chemotherapeutic option by minimizing toxic effects on healthy cells while maximizing therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | | | - Akhilesh Kumar
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| |
Collapse
|
6
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
7
|
Xia Q, Shen J, Ding H, Liu S, Li F, Li F, Feng N. Intravenous nanocrystals: fabrication, solidification, in vivo fate, and applications for cancer therapy. Expert Opin Drug Deliv 2023; 20:1467-1488. [PMID: 37814582 DOI: 10.1080/17425247.2023.2268512] [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: 03/13/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023]
Abstract
INTRODUCTION Intravenous nanocrystals (INCs) have shown intrinsic advantages in antitumor applications, particularly their properties of high drug loading, low toxicity, and controllable size. Therefore, it has a very bright application prospect as a drug delivery system. AREAS COVERED The ideal formulation design principles, fabrication, solidification, in vivo fate of INCs, the applications in drug delivery system (DDS) and the novel applications are covered in this review. EXPERT OPINION It is vital to select a suitable formulation and fabrication method to produce a stable and sterile INCs. Besides, the type of stabilizers and physical characteristics can also influence the in vivo fate of INCs, which is worthy of further studying. Based on wide researches about applications of INCs in cancer, biomimetic INCs are concerned increasingly for its favorable compatibility. The output of these studies suggested that INCs-based drug delivery could be a novel strategy for addressing the delivery of the drug that faces solubility, bioavailability, and toxicity problems.
Collapse
Affiliation(s)
- Qing Xia
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaqi Shen
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huining Ding
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Siyi Liu
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Li
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai, China
| | - Fengqian Li
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai, China
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Yadav PK, Saklani R, Tiwari AK, Verma S, Rana R, Chauhan D, Yadav P, Mishra K, Kedar AS, Kalleti N, Gayen JR, Wahajuddin M, Rath SK, Mugale MN, Mitra K, Sharma D, Chourasia MK. Enhanced apoptosis and mitochondrial cell death by paclitaxel-loaded TPP-TPGS 1000-functionalized nanoemulsion. Nanomedicine (Lond) 2023; 18:343-366. [PMID: 37140535 DOI: 10.2217/nnm-2022-0268] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Background: The present research was designed to develop a nanoemulsion (NE) of triphenylphosphine-D-α-tocopheryl-polyethylene glycol succinate (TPP-TPGS1000) and paclitaxel (PTX) to effectively deliver PTX to improve breast cancer therapy. Materials & methods: A quality-by-design approach was applied for optimization and in vitro and in vivo characterization were performed. Results: The TPP-TPGS1000-PTX-NE enhanced cellular uptake, mitochondrial membrane depolarization and G2M cell cycle arrest compared with free-PTX treatment. In addition, pharmacokinetics, biodistribution and in vivo live imaging studies in tumor-bearing mice showed that TPP-TPGS1000-PTX-NE had superior performance compared with free-PTX treatment. Histological and survival investigations ascertained the nontoxicity of the nanoformulation, suggesting new opportunities and potential to treat breast cancer. Conclusion: TPP-TPGS1000-PTX-NE improved the efficacy of breast cancer treatment by enhancing its effectiveness and decreasing drug toxicity.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ravi Saklani
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Saurabh Verma
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rafquat Rana
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Divya Chauhan
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pooja Yadav
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Keerti Mishra
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Ashwini S Kedar
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Navodayam Kalleti
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Jiaur R Gayen
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Muhammad Wahajuddin
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Srikanta K Rath
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhav N Mugale
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Kalyan Mitra
- Electron Microscopy Division, Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Deepak Sharma
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Manish K Chourasia
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
9
|
Gautam S, Marwaha D, Singh N, Rai N, Sharma M, Tiwari P, Urandur S, Shukla RP, Banala VT, Mishra PR. Self-Assembled Redox-Sensitive Polymeric Nanostructures Facilitate the Intracellular Delivery of Paclitaxel for Improved Breast Cancer Therapy. Mol Pharm 2023; 20:1914-1932. [PMID: 36848489 DOI: 10.1021/acs.molpharmaceut.2c00673] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
A two-tier approach has been proposed for targeted and synergistic combination therapy against metastatic breast cancer. First, it comprises the development of a paclitaxel (PX)-loaded redox-sensitive self-assembled micellar system using betulinic acid-disulfide-d-α-tocopheryl poly(ethylene glycol) succinate (BA-Cys-T) through carbonyl diimidazole (CDI) coupling chemistry. Second, hyaluronic acid is anchored to TPGS (HA-Cys-T) chemically through a cystamine spacer to achieve CD44 receptor-mediated targeting. We have established that there is significant synergy between PX and BA with a combination index of 0.27 at a molar ratio of 1:5. An integrated system comprising both BA-Cys-T and HA-Cys-T (PX/BA-Cys-T-HA) exhibited significantly higher uptake than PX/BA-Cys-T, indicating preferential CD44-mediated uptake along with the rapid release of drugs in response to higher glutathione concentrations. Significantly higher apoptosis (42.89%) was observed with PX/BA-Cys-T-HA than those with BA-Cys-T (12.78%) and PX/BA-Cys-T (33.38%). In addition, PX/BA-Cys-T-HA showed remarkable enhancement in the cell cycle arrest, improved depolarization of the mitochondrial membrane potential, and induced excessive generation of ROS when tested in the MDA-MB-231 cell line. An in vivo administration of targeted micelles showed improved pharmacokinetic parameters and significant tumor growth inhibition in 4T1-induced tumor-bearing BALB/c mice. Overall, the study indicates a potential role of PX/BA-Cys-T-HA in achieving both temporal and spatial targeting against metastatic breast cancer.
Collapse
Affiliation(s)
- Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India.,Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, UP, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Venkatesh Teja Banala
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Preclinical South PCS 002/011, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, UP, India.,Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, UP, India
| |
Collapse
|
10
|
Xiang H, Xu S, Li J, Li Y, Xue X, Liu Y, Li J, Miao X. Functional drug nanocrystals for cancer-target delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
11
|
HPLC method for simultaneous estimation of paclitaxel and baicalein: pharmaceutical and pharmacokinetic applications. Bioanalysis 2022; 14:1005-1020. [PMID: 36066029 DOI: 10.4155/bio-2022-0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: A novel HPLC method was developed and validated for the simultaneous estimation of paclitaxel (PTX) and baicalein (BAC). Materials & methods: The analytes were resolved in a C18 column using the aqueous solution of formic acid (0.10% v/v) and MeOH (30:70 v/v). Results: The developed method was found to be linear over the concentration ranges 0.039-10 μg/ml and 0.019-10 μg/ml for PTX and BAC, respectively. The lower limits of quantification obtained were 0.042 μg/ml and 0.361 μg/ml for PTX and BAC, respectively. Conclusion: The developed method was found to be precise and accurate as per the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use guidelines, for simultaneous estimation of PTX and BAC, having an application in formulation development and bioanalytical studies.
Collapse
|
12
|
Lv Y, Wu W, Corpstein CD, Li T, Lu Y. Biological and Intracellular Fates of Drug Nanocrystals through Different Delivery Routes: Recent Development Enabled by Bioimaging and PK Modeling. Adv Drug Deliv Rev 2022; 188:114466. [PMID: 35905948 DOI: 10.1016/j.addr.2022.114466] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/07/2022] [Accepted: 07/22/2022] [Indexed: 12/25/2022]
Abstract
Nanocrystals have contributed to exciting improvements in the delivery of poorly water-soluble drugs. The biological and intracellular fates of nanocrystals are currently under debate. Due to the remarkable commercial success in enhancing oral bioavailability, nanocrystals have originally been regarded as a simple formulation approach to enhance dissolution. However, the latest findings from novel bioimaging tools lead to an expanded view. Intact nanocrystals may offer long-term durability in the body and offer drug delivery capabilities like those of other nano-carriers. This review renews the understanding of the biological fates of nanocrystals administered via oral, intravenous, and parenteral (e.g., dermal, ocular, and pulmonary) routes. The intracellular pathways and dissolution kinetics of nanocrystals are explored. Additionally, the future trends for in vitro and in vivo quantification of nanocrystals, as well as factors impacting the biological and intracellular fates of nanocrystals are discussed. In conclusion, nanocrystals present a promising and underexplored therapeutic opportunity with immense potential.
Collapse
Affiliation(s)
- Yongjiu Lv
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Fudan Zhangjiang Institute, Shanghai 201203, China
| | - Clairissa D Corpstein
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Tonglei Li
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Fudan Zhangjiang Institute, Shanghai 201203, China.
| |
Collapse
|
13
|
Nižić Nodilo L, Perkušić M, Ugrina I, Špoljarić D, Jakobušić Brala C, Amidžić Klarić D, Lovrić J, Saršon V, Safundžić Kučuk M, Zadravec D, Kalogjera L, Pepić I, Hafner A. In situ gelling nanosuspension as an advanced platform for fluticasone propionate nasal delivery. Eur J Pharm Biopharm 2022; 175:27-42. [PMID: 35489667 DOI: 10.1016/j.ejpb.2022.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/23/2022] [Accepted: 04/24/2022] [Indexed: 11/28/2022]
Abstract
In this work we present the development of in situ gelling nanosuspension as advanced form for fluticasone propionate nasal delivery. Drug nanocrystals were prepared by wet milling technique. Incorporation of drug nanocrystals into polymeric in situ gelling system with pectin and sodium hyaluronate as constitutive polymers was fine-tuned attaining appropriate formulation surface tension, viscosity and gelling ability. Drug nanonisation improved the release profile and enhanced formulation mucoadhesive properties. QbD approach combining formulation and administration parameters resulted in optimised nasal deposition profile, with 51.8% of the dose deposited in the middle meatus, the critical region in the treatment of rhinosinusitis and nasal polyposis. Results obtained in biocompatibility and physico-chemical stability studies confirmed the leading formulation potential for safe and efficient nasal corticosteroid delivery.
Collapse
Affiliation(s)
- Laura Nižić Nodilo
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Mirna Perkušić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Ivo Ugrina
- University of Split, Faculty of Science, Split, Croatia
| | | | | | | | - Jasmina Lovrić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Vesna Saršon
- Jadran-galenski laboratorij d.d, Rijeka, Croatia
| | | | - Dijana Zadravec
- Department of Diagnostic and Interventional Radiology, Sestre milosrdnice University Hospital Center, University of Zagreb, Zagreb, Croatia
| | - Livije Kalogjera
- ENT Department, Zagreb School of Medicine; University Hospital Center "Sestre milosrdnice", Zagreb, Croatia
| | - Ivan Pepić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia.
| | - Anita Hafner
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia.
| |
Collapse
|
14
|
Paclitaxel Drug Delivery Systems: Focus on Nanocrystals' Surface Modifications. Polymers (Basel) 2022; 14:polym14040658. [PMID: 35215570 PMCID: PMC8875890 DOI: 10.3390/polym14040658] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
Paclitaxel (PTX) is a chemotherapeutic agent that belongs to the taxane family and which was approved to treat various kinds of cancers including breast cancer, ovarian cancer, advanced non-small-cell lung cancer, and acquired immunodeficiency syndrome (AIDS)-related Kaposi’s sarcoma. Several delivery systems for PTX have been developed to enhance its solubility and pharmacological properties involving liposomes, nanoparticles, microparticles, micelles, cosolvent methods, and the complexation with cyclodextrins and other materials that are summarized in this article. Specifically, this review discusses deeply the developed paclitaxel nanocrystal formulations. As PTX is a hydrophobic drug with inferior water solubility properties, which are improved a lot by nanocrystal formulation. Based on that, many studies employed nano-crystallization techniques not only to improve the oral delivery of PTX, but IV, intraperitoneal (IP), and local and intertumoral delivery systems were also developed. Additionally, superior and interesting properties of PTX NCs were achieved by performing additional modifications to the NCs, such as stabilization with surfactants and coating with polymers. This review summarizes these delivery systems by shedding light on their route of administration, the methods used in the preparation and modifications, the in vitro or in vivo models used, and the advantages obtained based on the developed formulations.
Collapse
|
15
|
Nanocrystal-loaded liposome for targeted delivery of poorly water-soluble antitumor drugs with high drug loading and stability towards efficient cancer therapy. Int J Pharm 2021; 599:120418. [PMID: 33647414 DOI: 10.1016/j.ijpharm.2021.120418] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/25/2021] [Accepted: 02/19/2021] [Indexed: 01/11/2023]
Abstract
Nanocrystals (NCs) enable the delivery of poorly water-soluble drugs with improved dissolution and bioavailability. However, their uncontrolled release and instability make targeted delivery challenging. Herein, a nano-in-nano delivery system composed of a drug nanocrystal core and liposome shell (NC@Lipo) is presented, which merges the advantages of drug nanocrystals (high drug loading) and liposomes (easy surface functionalization and high stability) for targeted delivery of hydrophobic drugs to tumors. CHMFL-ABL-053 (053), a hydrophobic drug candidate discovered by our group, was employed as a model drug to demonstrate the performance of NC@Lipo delivery system. Surface PEGylated (053-NC@PEG-Lipo) and folic acid-functionalized (053-NC@FA-Lipo) formulations were fabricated by wet ball milling combined with probe sonication. 053-NC@Lipo enabled high drug loading (up to 19.51%), considerably better colloidal stability, and longer circulation in vivo than 053-NC. Compared with free 053, 053-NC@PEG-Lipo and 053-NC@FA-Lipo exhibited higher tumor accumulation and considerably better in vivo antitumor efficacy in K562 xenograft mice with tumor growth inhibition rate (TGI) of up to 98%. Additionally, more effective tumor cell targeting in vitro and higher TGI in vivo were achieved with 053-NC@FA-Lipo. The NC@Lipo strategy may contribute to the targeted delivery of poorly water-soluble drugs with high drug loading, high stability, and tailorable surface, and has potential for the development of more efficient nanocrystal- and liposome-based formulations for commercial and clinical applications. It may also provide new opportunities for potential clinical application of candidate 053.
Collapse
|
16
|
Zhang Y, Sun C, Zhang Q, Deng Y, Hu X, Chen P. Intranasal delivery of Paclitaxel encapsulated nanoparticles for brain injury due to Glioblastoma. J Appl Biomater Funct Mater 2020; 18:2280800020977170. [PMID: 33307944 DOI: 10.1177/2280800020977170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Brain injury is a common cause for physical and emotional effects to the large number of populations. Moreover, glioblastoma is the tumor in brain with no possible treatment leading to death. The blood-brain barrier's makes the treatment more difficult by preventing the drugs to reach central nervous system. Paclitaxel (PTX) encapsulated Poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), PTX-PLGA-NPs were developed using emulsification method. The PTX-PLGA-NPs were characterized using Malvern Zetasizer and Scanning Electron Microscopy and were evaluated for their cytotoxicity in U87MG cells. PTX-PLGA-NPs were prepared using single emulsion method having size of 154 ± 22.19 nm with zeta potential of -23.7 mV. The PTX-PLGA-NPs were spherical in shape and have dose dependent cytotoxicity on U87MG cells. The PTX was released from the particles with initial burst release followed by sustained release pattern. The biodistribution was studied in mice with glioblastoma model using 125I radiolabeled PTX-PLGA-NPs and anti-glioblastoma was studied with PTX-PLGA-NPs. The biodistribution studies revealed PTX-PLGA-NPs after intranasal administration resulted in higher in vivo uptake with high anti-glioblastoma efficacy. The results suggest that PTX-PLGA-NPs administered through intranasal route have potential in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Chao Sun
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Qingtao Zhang
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Yongbing Deng
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Xi Hu
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Peng Chen
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| |
Collapse
|
17
|
Chai Z, Teng C, Yang L, Ren L, Yuan Z, Xu S, Cheng M, Wang Y, Yan Z, Qin C, Han X, Yin L. Doxorubicin delivered by redox-responsive Hyaluronic Acid–Ibuprofen prodrug micelles for treatment of metastatic breast cancer. Carbohydr Polym 2020; 245:116527. [DOI: 10.1016/j.carbpol.2020.116527] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 12/17/2022]
|
18
|
Patel D, Zode SS, Bansal AK. Formulation aspects of intravenous nanosuspensions. Int J Pharm 2020; 586:119555. [PMID: 32562654 DOI: 10.1016/j.ijpharm.2020.119555] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/23/2020] [Accepted: 06/14/2020] [Indexed: 01/04/2023]
Abstract
Intravenous (IV) route is preferred for rapid onset of action, avoiding first pass metabolism and achieving site specific delivery. Development of IV formulations for poorly water soluble drugs poses significant challenges. Formulation approaches like salt formation, co-solvents, surfactants and inclusion complexation using cyclodextrins are used for solubilisation. However, these approaches are not applicable universally and have limitations in extent of solubilisation, hypersensitivity, toxicity and application to only specific type of molecules. IV nanosuspension have been attracting attention as a viable strategy for development of IV formulations of poorly water-soluble drugs. Nanosuspension consists of nanocrystals of poorly water soluble drug suspended in aqueous media and stabilized using minimal concentration of stabilizers. Recent years have witnessed their potential in formulations for toxicological studies and clinical trials. However various challenges are associated with the translational development of IV nanosuspensions. Therefore, the objective of the current review is to provide a holistic view of formulation development and desired properties of IV nanosuspensions. It will also focus on advancements in characterization tools, manufacturing techniques and post-production processing. Challenges associated with translational development and regulatory aspects of IV nanosuspension will be addressed. Additionally, their role in preclinical evaluation and special applications like targeting will also be discussed with the help of case studies. The applications of IV nanosuspensions shall expand as their applications move from preclinical phase to commercialization.
Collapse
Affiliation(s)
- Dipeekakumari Patel
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Sandeep S Zode
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Arvind K Bansal
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
19
|
Alshweiat A, Csóka II, Tömösi F, Janáky T, Kovács A, Gáspár R, Sztojkov-Ivanov A, Ducza E, Márki Á, Szabó-Révész P, Ambrus R. Nasal delivery of nanosuspension-based mucoadhesive formulation with improved bioavailability of loratadine: Preparation, characterization, and in vivo evaluation. Int J Pharm 2020; 579:119166. [PMID: 32084574 DOI: 10.1016/j.ijpharm.2020.119166] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/11/2020] [Accepted: 02/18/2020] [Indexed: 12/23/2022]
Abstract
The unique requirements of poorly water-soluble drug delivery have driven a great deal of research into new formulations and routes of administration. This study investigates the use of nanosuspensions for solubility enhancement and drug delivery. Simple methods were used to prepare nasal formulations of loratadine based on nanosuspension pre-dispersion with sodium hyaluronate as a mucoadhesive agent. The nanosuspension was prepared by antisolvent precipitation method followed by ultrasonication and characterized for particle size, polydispersity index, zeta potential, morphology, and structure. Moreover, the nasal formulations were characterized for drug loading, pH, particle size, viscosity, bioadhesive viscosity parameter, and were evaluated for in vitro dissolution and diffusion, in addition to in vivo studies in a rat model. Loratadine nanosuspension displayed a particle size of 311 nm, a polydispersity index of 0.16, and zeta potential of -22.05 mV. The nanosuspension preserved the crystalline status of the raw drug. The addition of sodium hyaluronate exhibited an increase in the mean particle size and zeta potential of the nanoparticles. The nasal formulations showed enhanced bioadhesive properties compared to the unprocessed loratadine in the reference samples. The nanosuspension based-formulation that contained 5 mg mL-1 sodium hyaluronate and 2.5 mg mL-1 loratadine (NF4) showed a significant enhancement of flux and permeability coefficient through a synthetic membrane. NF4 exhibited 24.73 µg cm-2 h-1 and 0.082 cm h-1, while the reference sample showed 1.49 µg cm-2 h-1 and 0.017 cm h-1, for the flux and the permeability coefficient, respectively. Nasal administration of NF4 showed a bioavailability of 5.54-fold relative to the oral administration. The results obtained in this study indicate the potential of the nasal route and the nanosuspension for loratadine delivery. The relative bioavailability of NF4 was 1.84-fold compared to unprocessed loratadine in the reference sample. Therefore, the nanosized loratadine could be suggested as a practical and simple nanosystem for the intranasal delivery with improved bioavailability.
Collapse
Affiliation(s)
- Areen Alshweiat
- Faculty of Pharmacy, Interdisciplinary Excellence Centre, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary; Faculty of Pharmaceutical Sciences, The Hashemite University, 13133 Zarqa, Jordan.
| | - IIdikó Csóka
- Faculty of Pharmacy, Interdisciplinary Excellence Centre, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary
| | - Ferenc Tömösi
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Tamás Janáky
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Anita Kovács
- Faculty of Pharmacy, Interdisciplinary Excellence Centre, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary
| | - Róbert Gáspár
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary.
| | - Anita Sztojkov-Ivanov
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Eszter Ducza
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Árpád Márki
- Department of Medical Physics and Informatics, University of Szeged, Korányi fasor 9, H-6720 Szeged, Hungary
| | - Piroska Szabó-Révész
- Faculty of Pharmacy, Interdisciplinary Excellence Centre, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary
| | - Rita Ambrus
- Faculty of Pharmacy, Interdisciplinary Excellence Centre, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary.
| |
Collapse
|
20
|
Ahire E, Thakkar S, Darshanwad M, Misra M. Parenteral nanosuspensions: a brief review from solubility enhancement to more novel and specific applications. Acta Pharm Sin B 2018; 8:733-755. [PMID: 30245962 PMCID: PMC6146387 DOI: 10.1016/j.apsb.2018.07.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/20/2018] [Accepted: 06/26/2018] [Indexed: 02/01/2023] Open
Abstract
Advancements in in silico techniques of lead molecule selection have resulted in the failure of around 70% of new chemical entities (NCEs). Some of these molecules are getting rejected at final developmental stage resulting in wastage of money and resources. Unfavourable physicochemical properties affect ADME profile of any efficacious and potent molecule, which may ultimately lead to killing of NCE at final stage. Numerous techniques are being explored including nanocrystals for solubility enhancement purposes. Nanocrystals are the most successful and the ones which had a shorter gap between invention and subsequent commercialization of the first marketed product. Several nanocrystal-based products are commercially available and there is a paradigm shift in using approach from simply being solubility enhancement technique to more novel and specific applications. Some other aspects in relation to parenteral nanosuspensions are concentrations of surfactant to be used, scalability and in vivo fate. At present, there exists a wide gap due to poor understanding of these critical factors, which we have tried to address in this review. This review will focus on parenteral nanosuspensions, covering varied aspects especially stabilizers used, GRAS (Generally Recognized as Safe) status of stabilizers, scalability challenges, issues of physical and chemical stability, solidification techniques to combat stability problems and in vivo fate.
Collapse
Key Words
- ADME, absorption distribution metabolism elimination
- ASEs, aerosols solvent extractions
- AUC, area under curve
- BBB, blood–brain barrier
- BCS, Biopharmaceutical Classification System
- BDP, beclomethasone dipropionate
- CFC, critical flocculation concentration
- CLSM, confocal laser scanning microscopy
- CMC, critical micelle concentration
- DMSO, dimethyl sulfoxide
- EDI, estimated daily intake
- EHDA, electrohydrodynamic atomization
- EPAS, evaporative precipitation in aqueous solution
- EPR, enhanced permeability and retention
- FITC, fluorescein isothiocyanate
- GRAS, Generally Recognized as Safe
- HEC, hydroxyethylcellulose
- HFBII, class II hydrophobin
- HP-PTX/NC, hyaluronic acid-paclitaxel/nanocrystal
- HPC, hydroxypropyl cellulose
- HPH, high-pressure homogenization
- HPMC, hydroxypropyl methylcellulose
- IM, intramuscular
- IP, intraperitoneal
- IV, intravenous
- IVIVC, in vivo–in vitro correlation
- In vivo fate
- LD50, median lethal dose (50%)
- MDR, multidrug resistance effect
- NCE, new chemical entities
- Nanosuspension
- P-gp, permeation glycoprotein
- PEG, polyethylene glycol
- PTX, paclitaxel
- PVA, polyvinyl alcohol
- Parenteral
- QbD, quality by design
- SC, subcutaneous
- SEDS, solution enhanced dispersion by supercritical fluids
- SEM, scanning electron microscopy
- SFL, spray freezing into liquids
- Scalability
- Solidification
- Stabilizer
- TBA, tert-butanol
- TEM, transmission electron microscopy
- US FDA, United States Food and Drug Administration
- Vitamin E TPGS, d-α-tocopheryl polyethylene glycol 1000 succinate
Collapse
Affiliation(s)
| | | | | | - Manju Misra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 380054, India
| |
Collapse
|
21
|
Nanocrystals of Poorly Soluble Drugs: Drug Bioavailability and Physicochemical Stability. Pharmaceutics 2018; 10:pharmaceutics10030134. [PMID: 30134537 PMCID: PMC6161002 DOI: 10.3390/pharmaceutics10030134] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/13/2018] [Accepted: 08/18/2018] [Indexed: 11/16/2022] Open
Abstract
Many approaches have been developed over time to overcome the bioavailability limitations of poorly soluble drugs. With the advances in nanotechnology in recent decades, science and industry have been approaching this issue through the formulation of drugs as nanocrystals, which consist of “pure drugs and a minimum of surface active agents required for stabilization”. They are defined as “carrier-free submicron colloidal drug delivery systems with a mean particle size in the nanometer range, typically between 10–800 nm”. The primary importance of these nanoparticles was the reduction of particle size to nanoscale dimensions, with an increase in the particle surface area in contact with the dissolution medium, and thus in bioavailability. This approach has been proven successful, as demonstrated by the number of such drug products on the market. Nonetheless, despite the definition that indicates nanocrystals as a “carrier-free” system, surface active agents are necessary to prevent colloidal particles aggregation and thus improve stability. In addition, in more recent years, nanocrystal properties and technologies have attracted the interest of researchers as a means to obtain colloidal particles with modified biological properties, and thus their interest is now also addressed to modify the drug delivery and targeting. The present work provides an overview of the achievements in improving the bioavailability of poorly soluble drugs according to their administration route, describes the methods developed to overcome physicochemical and stability-related problems, and in particular reviews different stabilizers and surface agents that are able to modify the drug delivery and targeting.
Collapse
|
22
|
Urandur S, Banala VT, Shukla RP, Mittapelly N, Pandey G, Kalleti N, Mitra K, Rath SK, Trivedi R, Ramarao P, Mishra PR. Anisamide-Anchored Lyotropic Nano-Liquid Crystalline Particles with AIE Effect: A Smart Optical Beacon for Tumor Imaging and Therapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:12960-12974. [PMID: 29577719 DOI: 10.1021/acsami.7b19109] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The prospective design of nanocarriers for personalized oncotherapy should be an ensemble of targeting, imaging, and noninvasive therapeutic capabilities. Herein, we report the development of the inverse hexagonal nano-liquid crystalline (NLC) particles that are able to host formononetin (FMN), a phytoestrogen with known anticancer activity, and tetraphenylethene (TPE), an iconic optical beacon with aggregation-induced emission (AIE) signature, simultaneously. Ordered three-dimensional mesoporous internal structure and high-lipid-volume fraction of NLC nanoparticles (NLC NPs) frame the outer compartment for the better settlement of payloads. Embellishment of these nanoparticles by anisamide (AA), a novel sigma receptor targeting ligand using carbodiimide coupling chemistry ensured NLC's as an outstanding vehicle for possible utility in surveillance of tumor location as well as the FMN delivery through active AIE imaging. The size and structural integrity of nanoparticles were evaluated by quasi-elastic light scattering, cryo field emission scanning electron microscopy small-angle X-ray scattering. The existence of AIE effect in the nanoparticles was evidenced through the photophysical studies that advocate the application of NLC NPs in fluorescence-based bioimaging. Moreover, confocal microscopy illustrated the single living cell imaging ability endowed by the NLC NPs. In vitro and in vivo studies supported the enhanced efficacy of targeted nanoparticles (AA-NLC-TF) in comparison to nontargeted nanoparticles (NLC-TF) and free drug. Apparently, this critically designed multimodal NLC NPs may establish a promising platform for targeted and image-guided chemotherapy for breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Pratibha Ramarao
- Soft Condensed Matter Lab , Raman Research Institute , Bangalore 560080 , India
| | | |
Collapse
|
23
|
Agrawal S, Dwivedi M, Ahmad H, Chadchan SB, Arya A, Sikandar R, Kaushik S, Mitra K, Jha RK, Dwivedi AK. CD44 targeting hyaluronic acid coated lapatinib nanocrystals foster the efficacy against triple-negative breast cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:327-337. [PMID: 29129754 DOI: 10.1016/j.nano.2017.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 08/30/2017] [Accepted: 10/27/2017] [Indexed: 01/19/2023]
Abstract
Lapatinib (LPT) is an orally administered drug for the treatment of metastatic breast cancer. For expanding its therapeutic horizon, we have prepared its nanocrystals (LPT-NCs) that were subsequently coated with hyaluronic acid (HA) to produce LPT-HA-NCs. The detailed in-vitro and in-vivo investigation of LPT-HA-NCs showed the superior anticancer activity due to active targeting to CD44 receptors than the counterparts LPT-NCs and free LPT. In the triple negative 4T1 cells induced breast tumor bearing female Balb/C mice; LPT-HA-NCs treatment caused significant retardation of tumor growth and overall increase in animal survival probability because of their higher tumor localization, increased residence time. Our findings clearly suggest that HA coated LPT-NCs formulation enhances the activity of LPT against triple negative breast cancer. It exhibited magnificent therapeutic outcome at low dose thus presenting a strategy to reduce dose administrations and minimize dose related toxicity.
Collapse
Affiliation(s)
- Satish Agrawal
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Chennai, TN, India
| | - Monika Dwivedi
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Hafsa Ahmad
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | | | - Abhishek Arya
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Chennai, TN, India
| | - Roshan Sikandar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, UP, India; National Institute of Pharmaceutical Education & Research, Raebareli, UP, India
| | - Shweta Kaushik
- Academy of Scientific and Innovative Research (AcSIR), Chennai, TN, India; Division of Biochemisrty, CSIR-Drug Research Institute, Lucknow, UP, India
| | - Kalyan Mitra
- Electron Microscopy Unit, CSIR-Drug Research Institute, Lucknow, UP, India
| | - Rajesh Kumar Jha
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Anil Kumar Dwivedi
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, UP, India.
| |
Collapse
|
24
|
Albumin and Hyaluronic Acid-Coated Superparamagnetic Iron Oxide Nanoparticles Loaded with Paclitaxel for Biomedical Applications. Molecules 2017. [PMID: 28640222 PMCID: PMC6152103 DOI: 10.3390/molecules22071030] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Super paramagnetic iron oxide nanoparticles (SPION) were augmented by both hyaluronic acid (HA) and bovine serum albumin (BSA), each covalently conjugated to dopamine (DA) enabling their anchoring to the SPION. HA and BSA were found to simultaneously serve as stabilizing polymers of Fe3O4·DA-BSA/HA in water. Fe3O4·DA-BSA/HA efficiently entrapped and released the hydrophobic cytotoxic drug paclitaxel (PTX). The relative amount of HA and BSA modulates not only the total solubility but also the paramagnetic relaxation properties of the preparation. The entrapping of PTX did not influence the paramagnetic relaxation properties of Fe3O4·DA-BSA. Thus, by tuning the surface structure and loading, we can tune the theranostic properties of the system.
Collapse
|
25
|
Zhang H, Li W, Guo X, Kong F, Wang Z, Zhu C, Luo L, Li Q, Yang J, Du Y, You J. Specifically Increased Paclitaxel Release in Tumor and Synergetic Therapy by a Hyaluronic Acid-Tocopherol Nanomicelle. ACS APPLIED MATERIALS & INTERFACES 2017; 9:20385-20398. [PMID: 28540720 DOI: 10.1021/acsami.7b02606] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Recently, interest in tumor-targeted and site-specific drug release from nanoparticles as a means of drug delivery has increased. In this study, we report a smart nanosized micelle formed by hyaluronic acid (HA) conjugated with d-α-tocopherol succinate (TOS) using a disulfide bond as the linker (HA-SS-TOS, HSST). HSST micelles can specifically bind to the CD44 receptors that are overexpressed by cancer cells. The high levels of glutathione (GSH) in tumor cells selectively break the disulfide bond linker. This effect results in the synchronous release of the payload and a TOS fragment. These two components subsequently demonstrate synergetic anticancer activity. First, we demonstrate that drug release from HSST occurs rapidly in physiological high redox conditions and inside cancer cells. Significant GSH-triggered drug release was also observed in vivo. Furthermore, an in vivo biodistribution study indicated that the HSST micelles efficiently accumulated at the tumor sites, primarily due to an enhanced permeability and retention effect and the efficient binding to the cancer cells that overexpressed the CD44 receptor. Interestingly, the synchronous release of paclitaxel (PTX) and the TOS fragment from the PTX-loaded HSST caused synergetic tumor cell killing and tumor growth inhibition. Our work presents a useful candidate for a drug delivery system that can specifically accumulate at tumor tissue, selectively release its payload and a TOS fragment, and thus display a synergetic anticancer effect.
Collapse
Affiliation(s)
- Hanbo Zhang
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Wei Li
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xiaomeng Guo
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Fenfen Kong
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Zuhua Wang
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Chunqi Zhu
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Qingpo Li
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jie Yang
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yongzhong Du
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| |
Collapse
|
26
|
Singh J, Singh R, Gupta P, Rai S, Ganesher A, Badrinarayan P, Sastry GN, Konwar R, Panda G. Targeting progesterone metabolism in breast cancer with l-proline derived new 14-azasteroids. Bioorg Med Chem 2017; 25:4452-4463. [PMID: 28693914 DOI: 10.1016/j.bmc.2017.06.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/05/2017] [Accepted: 06/17/2017] [Indexed: 02/08/2023]
Abstract
Breast cancer cell proliferation is promoted by a variety of mitogenic signals. Classically estrogen is considered as most predominant mitogenic signal in hormone-dependent breast cancer and progesterone is primarily considered to have protective effect. However, it is suggested that some progesterone metabolite may promote breast cancer and progesterone metabolites like 5α-pregnane and 4-pregnene could serve as regulators of estrogen-responsiveness of breast cancer cells. Here, we estimated the potential of alternate targeting of breast cancer via progesterone signalling. l-Proline derived novel 14-azasteroid compounds were screened against MCF-7 and MDA-MB-231 cell lines using MTT assay. In silico studies, cell cycle, Annexin-V-FITC/PI, JC-1 mitochondrial assay, ROS analysis were performed to analyse the impact of hit compound 3b on breast cancer cells. Further, we analysed the impact of hit 3b on the progesterone, its metabolites and enzymes responsible for the conversion of progesterone and its metabolites using ELISA. Data suggests that compound 3b binds and down regulates of 5α-reductase by specifically inhibiting production of progesterone metabolites that are capable of promoting breast cancer proliferation, epithelial mesenchymal transition and migration. This study establishes the proof of concept and generation of new leads for additional targeting of breast cancer.
Collapse
Affiliation(s)
- Jyotsana Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritesh Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Preeti Gupta
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Smita Rai
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Asha Ganesher
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Preethi Badrinarayan
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - G Narahari Sastry
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Rituraj Konwar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Chennai 600 113, India.
| | - Gautam Panda
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Chennai 600 113, India.
| |
Collapse
|
27
|
Gao L, Gao L, Fan M, Li Q, Jin J, Wang J, Lu W, Yu L, Yan Z, Wang Y. Hydrotropic polymer-based paclitaxel-loaded self-assembled nanoparticles: preparation and biological evaluation. RSC Adv 2017. [DOI: 10.1039/c7ra04563h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hydrotropic polymer-based paclitaxel-loaded self-assembled nanoparticles: preparation and biological evaluation.
Collapse
|
28
|
Lazzari G, Couvreur P, Mura S. Multicellular tumor spheroids: a relevant 3D model for the in vitro preclinical investigation of polymer nanomedicines. Polym Chem 2017. [DOI: 10.1039/c7py00559h] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Application of 3D multicellular tumor spheroids to the investigation of polymer nanomedicines.
Collapse
Affiliation(s)
- Gianpiero Lazzari
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ Paris-Sud
- Université Paris-Saclay
| | - Patrick Couvreur
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ Paris-Sud
- Université Paris-Saclay
| | - Simona Mura
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ Paris-Sud
- Université Paris-Saclay
| |
Collapse
|