1
|
Xia L, Guo X, Lu D, Jiang Y, Liang X, Shen Y, Lin J, Zhang L, Chen H, Jin J, Luan X, Zhang W. S100A13-driven interaction between pancreatic adenocarcinoma cells and cancer-associated fibroblasts promotes tumor progression through calcium signaling. Cell Commun Signal 2025; 23:51. [PMID: 39871271 PMCID: PMC11773924 DOI: 10.1186/s12964-025-02049-7] [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: 12/02/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are key components of the pancreatic adenocarcinoma (PAAD) tumor microenvironment (TME), where they promote tumor progression and metastasis through immunosuppressive functions. Although significant progress has been made in understanding the crosstalk between cancer cells and CAFs, many underlying mechanisms remain unclear. Recent studies have highlighted the importance of calcium signaling in enhancing interactions between tumor cells and the surrounding stroma, with the S100 family of proteins serving as important regulators. While the roles of some S100 proteins have been extensively studied, others, such as S100A13, remain less well understood. METHODS Bioinformatic analysis was employed to predict the pathogenic potential of CAFs and S100A13. Stable S100A13 knockdown CAFs were generated using a short hairpin RNA system. Cellular viability and apoptosis rates were evaluated through CCK-8 and flow cytometry tests, respectively. Additionally, the wound healing and migration assays were conducted to assess the invasive and metastatic capabilities. Transcriptome analysis was conducted to identify differential gene expression and associated signaling pathways in PAAD cells derived from an indirect culture system. Furthermore, the protumoral role of S100A13 in PAAD was further verified using both 3D bioprinting and cell line-based xenograft tumor models. RESULTS In this study, we identified a strong association between S100A13, a calcium-binding protein, and CAFs in PAAD. Gene expression analysis revealed that S100A13 was highly expressed in CAFs and correlated with poor prognosis. Knockdown of S100A13 in CAFs reduced the metastatic potential of PAAD cells. In addition, S100A13 depletion impaired cell motility and calcium signaling pathways within the TME. Furthermore, silencing S100A13 in CAFs markedly slowed PAAD progression in both tumor spheroids and Balb/c nude mice. CONCLUSIONS Together, our findings underscore the critical role of CAFs-derived S100A13 in PAAD progression and suggest that targeting S100A13 may offer a promising therapeutic strategy for PAAD.
Collapse
Affiliation(s)
- Liuyuan Xia
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Xin Guo
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yixin Jiang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaohui Liang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiwen Shen
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Weidong Zhang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100700, China.
| |
Collapse
|
2
|
Wang B, Shen J, Zhou C, Wang X, Wang S, Hou R. Enhanced Pharmacokinetics of Celastrol via Long-Circulating Liposomal Delivery for Intravenous Administration. Int J Nanomedicine 2024; 19:5707-5718. [PMID: 38882540 PMCID: PMC11179669 DOI: 10.2147/ijn.s461624] [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: 03/08/2024] [Accepted: 05/25/2024] [Indexed: 06/18/2024] Open
Abstract
Background Rheumatoid Arthritis (RA) involves prolonged inflammation of the synovium, damaging joints and causing stiffness and deformity. Celastrol (Cel), derived from the Chinese herbal medicine Tripterygium wilfordii Hook F, offers immunosuppressive effects for RA treatment but is limited by poor solubility and bioavailability. Purpose In this study, long-circulating Cel-loaded liposomes (Cel-LPs) were used to increase the pharmacokinetics of Cel, thereby improving drug delivery and efficacy for the treatment of RA. Methods Cel-LPs were prepared and administered orally and intravenously to compare the elimination half-life of drugs and bioavailability of Cel. Cel-LPs were prepared using the lipid thin-layer-hydration-extrusion method. Human rheumatoid arthritis synovial (MH7A) cells were used to investigate the compatibility of Cel-LPs. The pharmacokinetic studies were performed on male Sprague-Dawley (SD) rats. Results The Cel-LPs had an average size of 72.20 ± 27.99 nm, a PDI of 0.267, a zeta potential of -31.60 ± 6.81 mV, 78.77 ± 5.69% drug entrapment efficiency and sustained release (5.83 ± 0.42% drug loading). The cytotoxicity test showed that liposomes had excellent biocompatibility and the fluorescence microscope diagram indicated that liposome entrapment increased intracellular accumulation of Rhodamine B by MH7A cells. Furthermore, the results exhibited that Cel-LPs improved the pharmacokinetics of Cel by increasing the elimination half-life (t1/2) to 11.71 hr, mean residence time (MRT(0-∞)) to 7.98 hr and apparent volume of distribution (Vz/F) to 44.63 L/kg in rats, compared to the Cel solution. Conclusion In this study, liposomes were demonstrated to be effective in optimizing the delivery of Cel, enabling the formulation of Cel-LPs with prolonged blood circulation and sustained release characteristics. This formulation enhanced the intravenous solubility and bioavailability of Cel, developing a foundation for its clinical application in RA and providing insights on poorly soluble drug management.
Collapse
Affiliation(s)
- Bo Wang
- Department of Orthopaedics, Suzhou Ruihua Orthopedic Hospital Affiliated Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China
- Department of Orthopaedics, the Sixth Affiliated Hospital of Wenzhou Medical University, the People's Hospital of Lishui, Lishui, Zhejiang, 323000, People's Republic of China
| | - Jiquan Shen
- Department of Orthopaedics, the Sixth Affiliated Hospital of Wenzhou Medical University, the People's Hospital of Lishui, Lishui, Zhejiang, 323000, People's Republic of China
| | - Changjian Zhou
- Department of Orthopaedics, the Sixth Affiliated Hospital of Wenzhou Medical University, the People's Hospital of Lishui, Lishui, Zhejiang, 323000, People's Republic of China
| | - Xinggao Wang
- Department of Orthopaedics, the Sixth Affiliated Hospital of Wenzhou Medical University, the People's Hospital of Lishui, Lishui, Zhejiang, 323000, People's Republic of China
| | - Shuanghu Wang
- Central Laboratory of the Sixth Affiliated Hospital of Wenzhou Medical University, the People's Hospital of Lishui, Lishui, Zhejiang, 323000, People's Republic of China
| | - Ruixing Hou
- Department of Orthopaedics, Suzhou Ruihua Orthopedic Hospital Affiliated Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China
| |
Collapse
|
3
|
Chintamaneni PK, Pindiprolu SKSS, Swain SS, Karri VVSR, Nesamony J, Chelliah S, Bhaskaran M. Conquering chemoresistance in pancreatic cancer: Exploring novel drug therapies and delivery approaches amidst desmoplasia and hypoxia. Cancer Lett 2024; 588:216782. [PMID: 38453046 DOI: 10.1016/j.canlet.2024.216782] [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: 09/28/2023] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Pancreatic cancer poses a significant challenge within the field of oncology due to its aggressive behaviour, limited treatment choices, and unfavourable outlook. With a mere 10% survival rate at the 5-year mark, finding effective interventions becomes even more pressing. The intricate relationship between desmoplasia and hypoxia in the tumor microenvironment further complicates matters by promoting resistance to chemotherapy and impeding treatment efficacy. The dense extracellular matrix and cancer-associated fibroblasts characteristic of desmoplasia create a physical and biochemical barrier that impedes drug penetration and fosters an immunosuppressive milieu. Concurrently, hypoxia nurtures aggressive tumor behaviour and resistance to conventional therapies. a comprehensive exploration of emerging medications and innovative drug delivery approaches. Notably, advancements in nanoparticle-based delivery systems, local drug delivery implants, and oxygen-carrying strategies are highlighted for their potential to enhance drug accessibility and therapeutic outcomes. The integration of these strategies with traditional chemotherapies and targeted agents reveals the potential for synergistic effects that amplify treatment responses. These emerging interventions can mitigate desmoplasia and hypoxia-induced barriers, leading to improved drug delivery, treatment efficacy, and patient outcomes in pancreatic cancer. This review article delves into the dynamic landscape of emerging anticancer medications and innovative drug delivery strategies poised to overcome the challenges imposed by desmoplasia and hypoxia in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Pavan Kumar Chintamaneni
- Department of Pharmaceutics, GITAM School of Pharmacy, GITAM (Deemed to be University), Rudraram, 502329 Telangana, India.
| | | | - Swati Swagatika Swain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | | | - Jerry Nesamony
- College of Pharmacy and Pharmaceutical Sciences, The University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Selvam Chelliah
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX-77004, USA
| | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, The University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA.
| |
Collapse
|
4
|
Aliabadi A, Haghshenas MR, Kiani R, Panjehshahin MR, Erfani N. Promising anticancer activity of cromolyn in colon cancer: in vitro and in vivo analysis. J Cancer Res Clin Oncol 2024; 150:207. [PMID: 38647571 PMCID: PMC11035410 DOI: 10.1007/s00432-024-05741-2] [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: 01/29/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Colon cancer is a prevalent cancer globally, representing approximately 10% of all cancer cases and accounting for 10% of all cancer-related deaths. Therefore, finding new therapeutic methods with high efficiency will be very valuable. Cromolyn (C), a common anti-allergic and mast cell membrane stabilizing drug, has recently shown valuable anti-cancer effects in several studies. This study was designed to investigate the anti-cancer activity of cromolyn on colon cancer in vitro and in vivo and to determine values such as selectivity index and survival effect. METHODS HT-29 (colon cancer) and MCF-10 (normal epithelial) cell lines were treated with C and Doxorubicin (DOX; Positive control). IC50 values and the effects of C and DOX on apoptosis were explored using methyl thiazole diphenyl-tetrazolium bromide (MTT) assay and Annexin V/PI Apoptosis Assay Kit. To investigate in an animal study, colon cancer was subcutaneously induced by CT26 cells (mouse colon cancer) in bulb/c mice. Mice were treated with 0.05 LD50 intraperitoneal every other day for 35 days. After the death of mice, tumor volume, tumor weight, and survival rate were evaluated. RESULTS C selectively and significantly suppressed the proliferation of cancer cells in a dose-dependent manner. The IC50 values for the MCF-10 and HT29 cell lines were 7.33 ± 0.78 μM and 2.33 ± 0.6 μM, respectively. Notably, the selective index (SI) highlighted that C displayed greater selectivity in inhibiting cancer cell growth compared to DOX, with SI values of 3.15 and 2.60, respectively. C exhibited higher effectiveness and selectivity in inducing apoptosis in cancer cells compared to DOX, with a significant p-value (61% vs. 52%, P-value ≤ 0.0001). Also, in mice bearing colon cancer, C reduced the tumor volume (6317 ± 1685mm3) and tumor weight (9.8 ± 1.6 g) compared to the negative control group (weight 12.45 ± 0.9 g; volume 7346 ± 1077) but these values were not statistically significant (P ≤ 0.05). CONCLUSION Our study showed that cromolyn is a selective and strong drug in inhibiting the proliferation of colon cancer cells. Based on our results, the efficacy of C in vitro analysis (MTT assays and apoptosis), as well as animal studies is competitive with the FDA-approved drug doxorubicin. C is very promising as a low-complication and good-efficacy drug for cancer drug repositioning. This requires clinical research study designs to comprehensively evaluate its anti-cancer effects.
Collapse
Affiliation(s)
- Amin Aliabadi
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razie Kiani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Nasrollah Erfani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Wang M, Xue W, Yuan H, Wang Z, Yu L. Nano-Drug Delivery Systems Targeting CAFs: A Promising Treatment for Pancreatic Cancer. Int J Nanomedicine 2024; 19:2823-2849. [PMID: 38525013 PMCID: PMC10959015 DOI: 10.2147/ijn.s451151] [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: 12/04/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
Currently, pancreatic cancer (PC) is one of the most lethal malignant tumors. PC is typically diagnosed at a late stage, exhibits a poor response to conventional treatment, and has a bleak prognosis. Unfortunately, PC's survival rate has not significantly improved since the 1960s. Cancer-associated fibroblasts (CAFs) are a key component of the pancreatic tumor microenvironment (TME). They play a vital role in maintaining the extracellular matrix and facilitating the intricate communication between cancer cells and infiltrated immune cells. Exploring therapeutic approaches targeting CAFs may reverse the current landscape of PC therapy. In recent years, nano-drug delivery systems have evolved rapidly and have been able to accurately target and precisely release drugs with little or no toxicity to the whole body. In this review, we will comprehensively discuss the origin, heterogeneity, potential targets, and recent advances in the nano-drug delivery system of CAFs in PC. We will also propose a novel integrated treatment regimen that utilizes a nano-drug delivery system to target CAFs in PC, combined with radiotherapy and immunotherapy. Additionally, we will address the challenges that this regimen currently faces.
Collapse
Affiliation(s)
- Mingjie Wang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Wenxiang Xue
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Hanghang Yuan
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Lei Yu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
6
|
Silli EK, Li M, Shao Y, Zhang Y, Hou G, Du J, Liang J, Wang Y. Liposomal nanostructures for Gemcitabine and Paclitaxel delivery in pancreatic cancer. Eur J Pharm Biopharm 2023; 192:13-24. [PMID: 37758121 DOI: 10.1016/j.ejpb.2023.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/01/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Pancreatic cancer (PC) is an incurable disease with a high death rate in the world nowadays. Gemcitabine (GEM) and Paclitaxel (PTX) are considered as references of chemotherapeutic treatments and are commonly used in clinical applications. Factors related to the tumor microenvironment such as insufficient tumor penetration, toxicity, and drug resistance can limit the effectiveness of these therapeutic anticancer drugs. The use of different liposomal nanostructures is a way that can optimize the drug's effectiveness and reduce toxicity. Given the development of PC therapy, this review focuses on advances in Nano-formulation, characterization, and delivery systems of loaded GEM and PTX liposomes using chemotherapy, nucleic acid delivery, and stroma remodeling therapy. As a result, the review covers the literature dealing with the applications of liposomes in PC therapy.
Collapse
Affiliation(s)
- Epiphane K Silli
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Mengfei Li
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Yuting Shao
- College of Engineering, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Yiran Zhang
- College of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Guilin Hou
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jiaqian Du
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jingdan Liang
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ying Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
7
|
Gajbhiye KR, Salve R, Narwade M, Sheikh A, Kesharwani P, Gajbhiye V. Lipid polymer hybrid nanoparticles: a custom-tailored next-generation approach for cancer therapeutics. Mol Cancer 2023; 22:160. [PMID: 37784179 PMCID: PMC10546754 DOI: 10.1186/s12943-023-01849-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/23/2023] [Indexed: 10/04/2023] Open
Abstract
Lipid-based polymeric nanoparticles are the highly popular carrier systems for cancer drug therapy. But presently, detailed investigations have revealed their flaws as drug delivery carriers. Lipid polymer hybrid nanoparticles (LPHNPs) are advanced core-shell nanoconstructs with a polymeric core region enclosed by a lipidic layer, presumed to be derived from both liposomes and polymeric nanounits. This unique concept is of utmost importance as a combinable drug delivery platform in oncology due to its dual structured character. To add advantage and restrict one's limitation by other, LPHNPs have been designed so to gain number of advantages such as stability, high loading of cargo, increased biocompatibility, rate-limiting controlled release, and elevated drug half-lives as well as therapeutic effectiveness while minimizing their drawbacks. The outer shell, in particular, can be functionalized in a variety of ways with stimuli-responsive moieties and ligands to provide intelligent holding and for active targeting of antineoplastic medicines, transport of genes, and theragnostic. This review comprehensively provides insight into recent substantial advancements in developing strategies for treating various cancer using LPHNPs. The bioactivity assessment factors have also been highlighted with a discussion of LPHNPs future clinical prospects.
Collapse
Affiliation(s)
- Kavita R Gajbhiye
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth, Erandwane, Pune, 411038, India
| | - Rajesh Salve
- Nanobioscience, Agharkar Research Institute, Pune, 411038, India
- Savitribai Phule Pune University, Pune, 411007, India
| | - Mahavir Narwade
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth, Erandwane, Pune, 411038, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Virendra Gajbhiye
- Nanobioscience, Agharkar Research Institute, Pune, 411038, India.
- Savitribai Phule Pune University, Pune, 411007, India.
| |
Collapse
|
8
|
Korucu Aktas P, Baysal I, Yabanoglu-Ciftci S, Arica B. Development and In Vitro Evaluation of Crizotinib-Loaded Lipid-Polymer Hybrid Nanoparticles Using Box-Behnken Design in Non-small Cell Lung Cancer. AAPS PharmSciTech 2023; 24:178. [PMID: 37658977 DOI: 10.1208/s12249-023-02634-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
The goal of the study was to produce, optimize, characterize, and compare crizotinib-loaded lipid-polymer hybrid nanoparticles (CL-LPHNPs), representing a novel contribution to the existing literature, and to determine their anticancer activity in non-small cell lung cancer cells (NSCLC). Box-Behnken design was used to investigate the effect of three independent variables: polymer amount (X1), soy phosphatidylcholine (X2), and DSPE-PEG (X3), on three responses: particle size (Y1), polydispersity index (Y2), and zeta potential (Y3). Different parameters were evaluated on the optimized LPHNP formulations such as encapsulation efficiency, drug release study, transmission electron microscopy (TEM) image analysis, and in vitro cell evaluations. The mean particle size of the optimized formulation is between 120 and 220 nm with a PDI< 0.2 and a zeta potential of -10 to -15 mV. The encapsulation efficiency values of crizotinib-loaded PLGA-LPHNPs (CL-PLGA-LPHNPs) and crizotinib-loaded PCL-LPHNPs (CL-PCL-LPHNPs) were 79.25±0.07% and 70.93±1.81%, respectively. Drug release study of CL-PLGA-LPHNPs and CL-PCL-LPHNPs showed a controlled and sustained release pattern as a result of core-shell type. Additionally, after 48 h, CL-PLGA-LPHNPs and CL-PCL-LPHNPs significantly reduced the viability of NCI-H2228 cells compared to free crizotinib. Moreover, CL-PLGA-LPHNPs and CL-PCL-LPHNPs exhibited a significant decrease in RAS, RAF, MEK, and ERK gene/protein expression levels after 48-h incubation. In conclusion, this pioneering study introduces lipid-polymer hybrid nanoparticles containing crizotinib as a novel treatment approach, uniting the advantages of a polymeric core and a lipid shell. The successful formulation optimization using Box-Behnken design yielded nanoparticles with adjustable size, remarkable stability, high drug loading, and a customizable drug release profile. Extensive investigations of key parameters, including particle size, PDI, ZP, TEM analysis, drug release, EE%, and in vitro evaluations, validate the potential of these nanoparticles. Moreover, the examination of two different polymers, PLGA and PCL, highlights their distinct impacts on nanoparticle performance. This research opens up new prospects for advanced therapeutic interventions with lipid-polymer hybrid nanoparticles.
Collapse
Affiliation(s)
- Pelinsu Korucu Aktas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara, Turkey
| | | | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
9
|
Mandarino A, Thiyagarajan S, Martins ACF, Gomes RDS, Vetter SW, Leclerc E. S100s and HMGB1 Crosstalk in Pancreatic Cancer Tumors. Biomolecules 2023; 13:1175. [PMID: 37627239 PMCID: PMC10452588 DOI: 10.3390/biom13081175] [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: 04/13/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic cancer remains a disease that is very difficult to treat. S100 proteins are small calcium binding proteins with diverse intra- and extracellular functions that modulate different aspects of tumorigenesis, including tumor growth and metastasis. High mobility group box 1 (HMGB1) protein is a multifaceted protein that also actively influences the development and progression of tumors. In this study, we investigate the possible correlations, at the transcript level, between S100s and HMGB1 in pancreatic cancer. For this purpose, we calculated Pearson's correlations between the transcript levels of 13 cancer-related S100 genes and HMGB1 in a cDNA array containing 19 pancreatic cancer tumor samples, and in 8 human pancreatic cancer cell lines. Statistically significant positive correlations were found in 5.5% (5 out of 91) and 37.4% (34 of 91) of the possible S100/S100 or S100/HMGB1 pairs in cells and tumors, respectively. Our data suggest that many S100 proteins crosstalk in pancreatic tumors either with other members of the S100 family, or with HMGB1. These newly observed interdependencies may be used to further the characterization of pancreatic tumors based on S100 and HMGB1 transcription profiles.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
10
|
Jin Y, Tomeh MA, Zhang P, Su M, Zhao X, Cai Z. Microfluidic fabrication of photo-responsive Ansamitocin P-3 loaded liposomes for the treatment of breast cancer. NANOSCALE 2023; 15:3780-3795. [PMID: 36723377 DOI: 10.1039/d2nr06215a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Ansamitocin P-3 (AP-3) is a promising anticancer agent. However, its low solubility has limited its biomedical applications. The preparation of liposomal formulations for the delivery of low solubility drugs using the microfluidic platform has attracted increasing attention in the pharmaceutical industry. In addition, photodynamic therapy (PDT) is a non-invasive and efficient strategy for the treatment of cancers, making photodynamic liposomes one of the most promising drug delivery systems (DDSs). In this study, a recently developed microfluidic device (swirl mixer) was used for the manufacturing of temperature-sensitive liposomes (TSL) that can be activated by near-infrared stimulation for the treatment of breast cancer. Changing the processing parameters of the microfluidic system allowed for optimizing the properties of the produced liposomes (e.g., particle size and size distribution). For the first time, the anticancer drug AP-3 and the photosensitizer indocyanine green (ICG) were encapsulated into TSL (AP-3/ICG@TSL) during microfluidic processing. The results show that AP-3/ICG@TSL are biocompatible and can significantly reduce the toxicity of AP-3 to normal tissues. After infrared laser irradiation, the heat generated from ICG not only resulted in the cancer cell toxicity, but also facilitated the release of AP-3 in tumor cells. AP-3/ICG@TSL with infrared laser irradiation was found to be able to significantly inhibit the growth of MCF-7 multicellular tumor spheroids (MCTSs) in vitro and MCF-7 tumors subcutaneously inoculated in nude mice as an in vivo model. In addition, it also showed no signs of damage to other organs. The current results demonstrated that the AP-3/ICG@TSL fabricated using the microfluidic swirl mixer is a promising DDS for breast cancer therapy.
Collapse
Affiliation(s)
- Yi Jin
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213000, China
| | - Mhd Anas Tomeh
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Peng Zhang
- School of Materials Science and Engineering, Changzhou University, Changzhou 213164, China
| | - Mingzhu Su
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213000, China
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Zhiqiang Cai
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
11
|
Ndemazie NB, Bulusu R, Zhu XY, Frimpong EK, Inkoom A, Okoro J, Ebesoh D, Rogers S, Han B, Agyare E. Evaluation of Anticancer Activity of Zhubech, a New 5-FU Analog Liposomal Formulation, against Pancreatic Cancer. Int J Mol Sci 2023; 24:4288. [PMID: 36901721 PMCID: PMC10002367 DOI: 10.3390/ijms24054288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/10/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Pancreatic cancer is projected to be the second leading cause of cancer-related death by 2030 in the US. The benefits of the most common systemic therapy for various pancreatic cancers have been masked by high drug toxicities, adverse reactions, and resistance. The use of nanocarriers such as liposomes to overcome these unwanted effects has become very popular. This study aims to formulate 1,3-bistertrahydrofuran-2yl-5FU (MFU)-loaded liposomal nanoparticles (Zhubech) and to evaluate itsstability, release kinetics, in vitro and in vivo anticancer activities, and biodistribution in different tissues. Particle size and zeta potential were determined using a particle size analyzer, while cellular uptake of rhodamine-entrapped liposomal nanoparticles (Rho-LnPs) was determined by confocal microscopy. Gadolinium hexanoate (Gd-Hex) was synthesized and entrapped into the liposomal nanoparticle (LnP) (Gd-Hex-LnP), as a model contrast agent, to evaluate gadolinium biodistribution and accumulation by LnPs in vivo using inductively coupled plasma mass spectrometry (ICP-MS). The mean hydrodynamic diameters of blank LnPs and Zhubech were 90.0 ± 0.65 nm and 124.9 ± 3.2 nm, respectively. The hydrodynamic diameter of Zhubech was found to be highly stable at 4 °C and 25 °C for 30 days in solution. In vitro drug release of MFU from Zhubech formulation exhibited the Higuchi model (R2 value = 0.95). Both Miapaca-2 and Panc-1 treated with Zhubech showed reduced viability, two- or four-fold lower than that of MFU-treated cells in 3D spheroid (IC50Zhubech = 3.4 ± 1.0 μM vs. IC50MFU = 6.8 ± 1.1 μM) and organoid (IC50Zhubech = 9.8 ± 1.4 μM vs. IC50MFU = 42.3 ± 1.0 μM) culture models. Confocal imaging confirmed a high uptake of rhodamine-entrapped LnP by Panc-1 cells in a time-dependent manner. Tumor-efficacy studies in a PDX bearing mouse model revealed a more than 9-fold decrease in mean tumor volumes in Zhubech-treated (108 ± 13.5 mm3) compared to 5-FU-treated (1107 ± 116.2 mm3) animals, respectively. This study demonstrates that Zhubech may be a potential candidate for delivering drugs for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Nkafu Bechem Ndemazie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Xue You Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Esther Kesewaah Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Andriana Inkoom
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Dexter Ebesoh
- Faculty of Health Sciences, University of Buea, Buea P.O. Box 63, Cameroon
| | - Sherise Rogers
- Department of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Bo Han
- Department of Surgery, Keck School of Medicine University of South California, Los Angeles, CA 90033, USA
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
12
|
Raza F, Evans L, Motallebi M, Zafar H, Pereira-Silva M, Saleem K, Peixoto D, Rahdar A, Sharifi E, Veiga F, Hoskins C, Paiva-Santos AC. Liposome-based diagnostic and therapeutic applications for pancreatic cancer. Acta Biomater 2023; 157:1-23. [PMID: 36521673 DOI: 10.1016/j.actbio.2022.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the harshest and most challenging cancers to treat, often labeled as incurable. Chemotherapy continues to be the most popular treatment yet yields a very poor prognosis. The main barriers such as inefficient drug penetration and drug resistance, have led to the development of drug carrier systems. The benefits, ease of fabrication and modification of liposomes render them as ideal future drug delivery systems. This review delves into the versatility of liposomes to achieve various mechanisms of treatment for pancreatic cancer. Not only are there benefits of loading chemotherapy drugs and targeting agents onto liposomes, as well as mRNA combined therapy, but liposomes have also been exploited for immunotherapy and can be programmed to respond to photothermal therapy. Multifunctional liposomal formulations have demonstrated significant pre-clinical success. Functionalising drug-encapsulated liposomes has resulted in triggered drug release, specific targeting, and remodeling of the tumor environment. Suppressing tumor progression has been achieved, due to their ability to more efficiently and precisely deliver chemotherapy. Currently, no multifunctional surface-modified liposomes are clinically approved for pancreatic cancer thus we aim to shed light on the trials and tribulations and progress so far, with the hope for liposomal therapy in the future and improved patient outcomes. STATEMENT OF SIGNIFICANCE: Considering that conventional treatments for pancreatic cancer are highly associated with sub-optimal performance and systemic toxicity, the development of novel therapeutic strategies holds outmost relevance for pancreatic cancer management. Liposomes are being increasingly considered as promising nanocarriers for providing not only an early diagnosis but also effective, highly specific, and safer treatment, improving overall patient outcome. This manuscript is the first in the last 10 years that revises the advances in the application of liposome-based formulations in bioimaging, chemotherapy, phototherapy, immunotherapy, combination therapies, and emergent therapies for pancreatic cancer management. Prospective insights are provided regarding several advantages resulting from the use of liposome technology in precision strategies, fostering new ideas for next-generation diagnosis and targeted therapies of pancreatic cancer.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Lauren Evans
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Mahzad Motallebi
- Immunology Board for Transplantation And Cell-based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 7616911319, Iran; Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Kalsoom Saleem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 45320, Pakistan
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Clare Hoskins
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
13
|
Park T, Amatya R, Min KA, Shin MC. Liposomal Iron Oxide Nanoparticles Loaded with Doxorubicin for Combined Chemo-Photothermal Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15010292. [PMID: 36678921 PMCID: PMC9860715 DOI: 10.3390/pharmaceutics15010292] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Iron oxide nanoparticle (IONP) possesses unique advantages over other nanoparticles in the use of cancer imaging and therapy. Specifically, it has drawn great attention in the emerging research field of photothermal cancer therapy. Herein, we developed doxorubicin (DOX)-loaded liposomal IONP (Lipo-IONP/DOX) and evaluated in vitro and in vivo their applicability for combined chemo-photothermal cancer therapy. The Lipo-IONP was synthesized by the thin-film evaporation method. The prepared Lipo-IONP was observed as about a 240 nm-sized agglomerate of globular-shaped nanoparticles. The TEM and FT-IR data evidenced the successful formation of liposomal IONP. The superparamagnetic property of the Lipo-IONP was confirmed by the SQUID analysis. The DSC data showed a transition temperature of about 47-48 °C for the mixed lipids composing the Lipo IONP, and the DOX release studies revealed the feasibility of induced burst release of DOX by laser irradiation. The Lipo-IONP/DOX possessed a plasma half-life of 42 min, which could ensure sufficient circulation time for magnetic tumor targeting. The in vivo magnetic targeting enabled a significant increase (6.3-fold) in the tumor accumulation of Lipo-IONP/DOX, leading to greater photothermal effects. Finally, the preliminary efficacy study evidenced the applicability as well as the safety of the Lipo-IONP/DOX for use in combined chemo-photothermal cancer therapy. Overall, the study results demonstrated that the Lipo-IONP/DOX might serve as an effective and safe agent for combined chemo-photothermal cancer therapy.
Collapse
Affiliation(s)
- Taehoon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Republic of Korea
| | - Reeju Amatya
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Republic of Korea
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Republic of Korea
- Correspondence: (K.A.M.); (M.C.S.); Tel.: +82-55-320-3459 (K.A.M.); +82-55-772-2421 (M.C.S.)
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Republic of Korea
- Correspondence: (K.A.M.); (M.C.S.); Tel.: +82-55-320-3459 (K.A.M.); +82-55-772-2421 (M.C.S.)
| |
Collapse
|
14
|
Motawi TK, El-Maraghy SA, Sabry D, Nady OM, Senousy MA. Cromolyn chitosan nanoparticles reverse the DNA methylation of RASSF1A and p16 genes and mitigate DNMT1 and METTL3 expression in breast cancer cell line and tumor xenograft model in mice. Chem Biol Interact 2022; 365:110094. [PMID: 35961540 DOI: 10.1016/j.cbi.2022.110094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Developing epigenetic drugs for breast cancer (BC) remains a novel therapeutic approach. Cromolyn is a mast cell stabilizer emerging as an anticancer drug; its encapsulation in chitosan nanoparticles (CSNPs) improves its effect and bioavailability. However, its effect on DNA and RNA methylation machineries has not been previously tackled. METHODS The possible anticancer effect of cromolyn CSNPs and its potential as an epigenetic drug was investigated in vitro using MCF-7 human BC cell line and in vivo using Ehrlich ascites carcinoma-xenograft model in mice symbolizing murine mammary adenocarcinoma. Mice were injected with a single dose of Ehrlich ascites carcinoma cells subcutaneously for the induction of tumor mass, and then randomized into three groups: control, cromolyn CSNPs (equivalent to 5 mg cromolyn/kg, i.p.) and plain CSNPs twice/week for 2 weeks. RESULTS Cromolyn CSNPs showed prominent anticancer effect in MCF-7 cells by reducing the cell viability percent and enhancing DNA damage in the comet assay demonstrating its apoptotic actions. Mechanistically, cromolyn CSNPs influenced potential epigenetic processes through mitigating DNA methyltransferase 1 (DNMT1) expression, reversing the hypermethylation pattern of the tumor suppressor RASSF1A and p16 genes and attenuating the expression of the RNA N6-methyladenosine writer, methyltransferase-like 3 (METTL3). Cromolyn CSNPs diminished ERK1/2 phosphorylation, a possible arm influencing DNMT1 expression. In vivo, cromolyn CSNPs lessened the tumor volume and halted DNMT1 and METTL3 expression in Ehrlich carcinoma mice. CONCLUSIONS Cromolyn CSNPs have the premise as an epigenetic drug through inhibiting ERK1/2 phosphorylation/DNMT1/DNA methylation and possibly impacting the RNA methylation machinery via mitigating METTL3 expression.
Collapse
Affiliation(s)
- Tarek K Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Shohda A El-Maraghy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Dina Sabry
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Omina M Nady
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud A Senousy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
15
|
Kesserwan S, Mao L, Sharafieh R, Kreutzer DL, Klueh U. A pharmacological approach assessing the role of mast cells in insulin infusion site inflammation. Drug Deliv Transl Res 2022; 12:1711-1718. [PMID: 34561836 PMCID: PMC9639590 DOI: 10.1007/s13346-021-01070-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2021] [Indexed: 01/13/2023]
Abstract
Background Extending the lifespan of subcutaneous insulin administration sets and infusion pumps requires overcoming unreliable insulin delivery induced by dermal reactions. All commercially available insulin formulations contain insulin phenolic preservatives (IPP), which stabilize the insulin molecule but result in unwanted cell and tissue toxicity. Mast cells, which are the first line of defense once the epithelium is breached, are particularly abundant beneath the skin surface. Thus, we hypothesize a sequence of events initiated by device insertion that activates skin mast cells (MC) that subsequently trigger neutrophil and monocyte/macrophage recruitment. The ensuing inflammatory response compromises effective insulin infusion therapy. Methods We employed a non-genetic, pharmacological approach to MC membrane stabilization using Cromolyn sodium (CS), which inhibits MC degranulation. These studies were conducted in our modified air pouch mouse model using non-diabetic and streptozotocin induced diabetic mice. We evaluated the impact of systemic CS through intraperitoneal injections, as well as the impact of local CS through co-infusion, on infusion catheter insertion and IPP-induced inflammation. Results CS at a concentration of 50 mg/kg minimized inflammation triggered in response to insulin phenolic preservatives present in standard insulin formulations. The resultant degree of tissue inflammation was comparable to that observed with saline injections. Conclusion Targeting MC has the potential to extend the longevity of insulin infusion sets by mitigating the inflammatory response. Future studies should be directed at employing other MC models, such as newer Cre/loxP mouse strains, to confirm the sentinel role of MC in insulin infusion therapy.
Collapse
Affiliation(s)
- Shereen Kesserwan
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, MI
| | - Li Mao
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, MI
| | - Roshanak Sharafieh
- Department of Surgery, School of Medicine, University of Connecticut, Farmington, CT
| | - Donald L. Kreutzer
- Department of Surgery, School of Medicine, University of Connecticut, Farmington, CT
| | - Ulrike Klueh
- Department of Biomedical Engineering, Integrative Biosciences Center, Wayne State University, Detroit, MI,Corresponding Author: Ulrike Klueh Ph. D., Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202,
| |
Collapse
|
16
|
Hani U, Osmani RAM, Siddiqua A, Wahab S, Batool S, Ather H, Sheraba N, Alqahtani A. A systematic study of novel drug delivery mechanisms and treatment strategies for pancreatic cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Design, characterization and evaluation of cucurbitacin B-loaded core–shell-type hybrid nano-sized particles using DoE approach. Polym Bull (Berl) 2020. [DOI: 10.1007/s00289-020-03256-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Elmowafy M, Alruwaili NK, Shalaby K, Alharbi KS, Altowayan WM, Ahmad N, Zafar A, Elkomy M. Long-Acting Paliperidone Parenteral Formulations Based on Polycaprolactone Nanoparticles; the Influence of Stabilizer and Chitosan on In Vitro Release, Protein Adsorption, and Cytotoxicity. Pharmaceutics 2020; 12:E160. [PMID: 32079093 PMCID: PMC7076490 DOI: 10.3390/pharmaceutics12020160] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Long-acting preparations containing the antipsychotic paliperidone for intramuscular injection has drawn considerable attention to achieve harmless long-term treatment. This study aimed to develop paliperidone loaded polycaprolactone (PCL) nanoparticles and investigate the influence of PCL/drug ratio, stabilizer type, and chitosan coating on physicochemical properties, protein adsorption, and cellular toxicity. Results showed that chitosan coating produced enlarged particle sizes, shifted the surface charges from negative into positive and did not influence encapsulation efficiencies. Chitosan coating relatively sustained the drug release especially in pluronic stabilized formulations. Pluronic F127 based formulations exhibited the least protein adsorption (384.3 μg/mL). Chitosan coating of Tween 80 and polyvinyl alcohol stabilized formulations significantly (p < 0.05) increased protein adsorption. Cellular viability was concentration-dependent and negatively affected by stabilizers. All formulations did not show cellular death at 1.56 μg/mL. Inflammatory responses and oxidative stress were less affected by Tween 80 compared with other stabilizers. Chitosan minimized all aspects of cellular toxicity. Collectively, stabilizer type and chitosan coating play critical roles in developing safe and effective long-acting PCL nanoparticles intended for parenteral drug delivery. The coated formulations containing Tween 80 and Pluronic F127 as stabilizers are warranted a future in vivo study to delineate its safety and efficacy profiles.
Collapse
Affiliation(s)
- Mohammed Elmowafy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia; (N.K.A.); (K.S.); (N.A.); (A.Z.); (M.E.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy (Boys), Al-Azhar University, 11751 Nasr City, Cairo, Egypt
| | - Nabil K. Alruwaili
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia; (N.K.A.); (K.S.); (N.A.); (A.Z.); (M.E.)
| | - Khaled Shalaby
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia; (N.K.A.); (K.S.); (N.A.); (A.Z.); (M.E.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy (Boys), Al-Azhar University, 11751 Nasr City, Cairo, Egypt
| | - Khalid S. Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia;
| | - Waleed M. Altowayan
- Pharmacy Practice Department, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia;
| | - Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia; (N.K.A.); (K.S.); (N.A.); (A.Z.); (M.E.)
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia; (N.K.A.); (K.S.); (N.A.); (A.Z.); (M.E.)
| | - Mohammed Elkomy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah P.O. Box 2014, Saudi Arabia; (N.K.A.); (K.S.); (N.A.); (A.Z.); (M.E.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, 62521 Beni-Suef, Egypt
| |
Collapse
|
19
|
Han B, Yang Y, Chen J, Tang H, Sun Y, Zhang Z, Wang Z, Li Y, Li Y, Luan X, Li Q, Ren Z, Zhou X, Cong D, Liu Z, Meng Q, Sun F, Pei J. Preparation, Characterization, and Pharmacokinetic Study of a Novel Long-Acting Targeted Paclitaxel Liposome with Antitumor Activity. Int J Nanomedicine 2020; 15:553-571. [PMID: 32158208 PMCID: PMC6986409 DOI: 10.2147/ijn.s228715] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/15/2019] [Indexed: 12/16/2022] Open
Abstract
Background Breast cancer is the leading cause of cancer death in women. Chemotherapy to inhibit the proliferation of cancer cells is considered to be the most important therapeutic strategy. The development of long-circulating PEG and targeting liposomes is a major advance in drug delivery. However, the techniques used in liposome preparation mainly involve conventional liposomes, which have a short half-life, high concentrations in the liver and spleen reticuloendothelial system, and no active targeting. Methods Four kinds of paclitaxel liposomes were prepared and characterized by various analytical techniques. The long-term targeting effect of liposomes was verified by fluorescence detection methods in vivo and in vitro. Pharmacokinetic and acute toxicity tests were conducted in ICR mice to evaluate the safety of different paclitaxel preparations. The antitumor activity of ES-SSL-PTX was investigated in detail using in vitro and in vivo human breast cancer MCF-7 cell models. Results ER-targeting liposomes had a particle size of 137.93±1.22 nm and an acceptable encapsulation efficiency of 88.07±1.25%. The liposome preparation is best stored at 4°C, and is stable for up to 48 hrs. Cytotoxicity test on MCF-7 cells demonstrated the stronger cytotoxic activity of liposomes in comparison to free paclitaxel. We used the near-infrared fluorescence imaging technique to confirm that ES-SSL-PTX was effectively targeted and could quickly and specifically identify the tumor site. Pharmacokinetics and acute toxicity in vivo experiments were carried out. The results showed that ES-SSL-PTX could significantly prolong the half-life of the drug, increase its circulation time in vivo, improve its bioavailability and reduce its toxicity and side effects. ES-SSL-PTX can significantly improve the pharmacokinetic properties of paclitaxel, avoid allergic reaction of the original solvent, increase antitumor efficacy and reduce drug toxicity and side effects. Conclusion ES-SSL-PTX has great potential for improving the treatment of breast cancer, thereby improving patient prognosis and quality of life.
Collapse
Affiliation(s)
- Bing Han
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yue Yang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China.,Department of Pharmacy, Ministry of Health Service, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jinglin Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Huan Tang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yuxin Sun
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Zheng Zhang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Zeng Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yan Li
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yao Li
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Xue Luan
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Qianwen Li
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Zhihui Ren
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Xiaowei Zhou
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Dengli Cong
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Zhiyi Liu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Qin Meng
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Fei Sun
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| |
Collapse
|
20
|
Non-small cell lung cancer-targeted, redox-sensitive lipid-polymer hybrid nanoparticles for the delivery of a second-generation irreversible epidermal growth factor inhibitor-Afatinib: In vitro and in vivo evaluation. Biomed Pharmacother 2019; 120:109493. [PMID: 31586902 DOI: 10.1016/j.biopha.2019.109493] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 01/01/2023] Open
Abstract
Afatinib (Afa), a second-generation irreversible epidermal growth factor inhibitor for the development of non-small cell lung cancer, has low bioavailability and adverse reactions. Nanoscaled drug delivery systems offer promising alternatives to address these defects and improve therapeutic outcomes. In the present study, a Tf contained, redox-sensitive ligand was synthesized and used for the preparation of afatinib loaded, Tf modified redox-sensitive lipid-polymer hybrid nanoparticles (Tf-SS-Afa-LPNs). Subsequently, studies of biological experiments in vitro and in vivo were performed to investigate the therapeutic effect of the system in lung cancer. The results showed that Tf-SS-Afa-LPNs has particle size of 103.5 ± 4.1 nm and zeta potential of -21.2 ± 2.4 mV. Significantly higher drug release was observed in the presence of glutathione (GSH). The area under the plasma concentration - time curve (AUC), peak concentration (Cmax) and terminal half life (T1/2) of Tf-SS-Afa-LPNs were 866.56 mg/L.h, 25.62 ± 3.21 L/kg/h, and 43.25 ± 2.31 h. Tf-SS-Afa-LPNs exhibited the most remarkable in vivo anti-tumor efficiency efficacy, which inhibited the tumor volume from 919 mm3 to 212 mm3. Tf-SS-Afa-LPNs is a promising platform for the lung cancer treatment.
Collapse
|
21
|
Shao Y, Luo W, Guo Q, Li X, Zhang Q, Li J. In vitro and in vivo effect of hyaluronic acid modified, doxorubicin and gallic acid co-delivered lipid-polymeric hybrid nano-system for leukemia therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2043-2055. [PMID: 31388296 PMCID: PMC6607984 DOI: 10.2147/dddt.s202818] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/16/2019] [Indexed: 12/19/2022]
Abstract
Objective: To investigate the hyaluronic acid (HA) modified, doxorubicin (DOX) and gallic acid (GA) co-delivered lipid-polymeric hybrid nano-system for leukemia therapy. Methods: We produced a kind of lipid-polymer hybrid nanoparticle (LPHN) with a core-shell structure in which DOX and GA were co-loaded. In vitro and in vivo leukemia therapeutic effects of the HA modified, DOX and GA co-delivered LPHNs (HA-DOX/GA-LPHNs) were evaluated in DOX resistant human HL-60 promyelocytic leukemia cells (HL-60/ADR cells), DOX resistant human K562 chronic myeloid leukemia cells (K562/ADR cells), and HL-60/ADR cells bearing mouse model. Results: The sizes and zeta potentials of HA modified LPHNs were about 160 nm and −40 mV. HA-DOX/GA-LPHNs showed the most prominent cytotoxicity and the best synergistic effect was obtained when DOX/GA ratio was 2/1. In vivo studies revealed that HA-DOX/GA-LPHNs inhibited tumor growth from 956 mm3 to 213 mm3, with an inhibition rate of 77.7%. Conclusion: In summary, the study showed that HA-DOX/GA-LPHNs can be applied as a promising leukemia therapy system.
Collapse
Affiliation(s)
- Yanping Shao
- Department of Hematology-Oncology, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, People's Republic of China
| | - Wenda Luo
- Department of Hematology-Oncology, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, People's Republic of China
| | - Qunyi Guo
- Department of Hematology-Oncology, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, People's Republic of China
| | - Xiaohong Li
- Department of Hematology, Hebei Province Hospital of Chinese Medicine, Shijiazhuang, Hebei 050011, People's Republic of China
| | - Qianqian Zhang
- Department of Hematology, Hebei Province Hospital of Chinese Medicine, Shijiazhuang, Hebei 050011, People's Republic of China
| | - Jing Li
- Department of Hematology, Hebei Province Hospital of Chinese Medicine, Shijiazhuang, Hebei 050011, People's Republic of China
| |
Collapse
|
22
|
Reduction responsive liposomes based on paclitaxel-ss-lysophospholipid with high drug loading for intracellular delivery. Int J Pharm 2019; 564:244-255. [DOI: 10.1016/j.ijpharm.2019.04.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/02/2019] [Accepted: 04/20/2019] [Indexed: 11/20/2022]
|
23
|
Bresnick AR. S100 proteins as therapeutic targets. Biophys Rev 2018; 10:1617-1629. [PMID: 30382555 PMCID: PMC6297089 DOI: 10.1007/s12551-018-0471-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/21/2018] [Indexed: 12/13/2022] Open
Abstract
The human genome codes for 21 S100 protein family members, which exhibit cell- and tissue-specific expression patterns. Despite sharing a high degree of sequence and structural similarity, the S100 proteins bind a diverse range of protein targets and contribute to a broad array of intracellular and extracellular functions. Consequently, the S100 proteins regulate multiple cellular processes such as proliferation, migration and/or invasion, and differentiation, and play important roles in a variety of cancers, autoimmune diseases, and chronic inflammatory disorders. This review focuses on the development of S100 neutralizing antibodies and small molecule inhibitors and their potential therapeutic use in controlling disease progression and severity.
Collapse
Affiliation(s)
- Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
24
|
Kim CH, Sung SW, Lee ES, Kang TH, Yoon HY, Goo YT, Cho HR, Kim DY, Kang MJ, Choi YS, Lee S, Choi YW. Sterically Stabilized RIPL Peptide-Conjugated Nanostructured Lipid Carriers: Characterization, Cellular Uptake, Cytotoxicity, and Biodistribution. Pharmaceutics 2018; 10:E199. [PMID: 30360549 PMCID: PMC6321264 DOI: 10.3390/pharmaceutics10040199] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/25/2022] Open
Abstract
As a platform for hepsin-specific drug delivery, we previously prepared IPLVVPLRRRRRRRRC peptide (RIPL)-conjugated nanostructured lipid carriers (RIPL-NLCs) composed of Labrafil® M 1944 CS (liquid oil) and Precirol® ATO 5 (solid lipid). In this study, to prevent the recognition by the mononuclear phagocyte system, polyethylene glycol (PEG)-modified RIPL-NLCs (PEG-RIPL-NLCs) were prepared using PEG3000 at different grafting ratios (1, 5, and 10 mole %). All prepared NLCs showed a homogeneous dispersion (130⁻280 nm), with zeta potentials varying from -18 to 10 mV. Docetaxel (DTX) was successfully encapsulated in NLCs: encapsulation efficiency (93⁻95%); drug-loading capacity (102⁻109 µg/mg). PEG-RIPL-NLCs with a grafting ratio of 5% PEG or higher showed significantly reduced protein adsorption and macrophage phagocytosis. The uptake of PEG(5%)-RIPL-NLCs by cancer cell lines was somewhat lower than that of RIPL-NLCs because of the PEG-induced steric hindrance; however, the uptake level of PEG-RIPL-NLCs was still greater than that of plain NLCs. In vivo biodistribution was evaluated after tail vein injection of NLCs to normal mice. Compared to RIPL-NLCs, PEG(5%)-RIPL-NLCs showed lower accumulation in the liver, spleen, and lung. In conclusion, we found that PEG(5%)-RIPL-NLCs could be a promising nanocarrier for selective drug targeting with a high payload of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Chang Hyun Kim
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| | - Si Woo Sung
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| | - Eun Seok Lee
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| | - Tae Hoon Kang
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| | - Ho Yub Yoon
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| | - Yoon Tae Goo
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| | - Ha Ra Cho
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Korea.
| | - Dong Yoon Kim
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Korea.
| | - Myung Joo Kang
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Korea.
| | - Yong Seok Choi
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 330-714, Korea.
| | - Sangkil Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Daegu 704-701, Korea.
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong, Dongjak-gu, Seoul 156-756, Korea.
| |
Collapse
|
25
|
Jin H, Zhao Y, Yang J, Zhang X, Ma S. Hyperthermia enhances the sensitivity of pancreatic cancer SW1990 cells to gemcitabine through ROS/JNK signaling. Oncol Lett 2018; 16:6742-6748. [PMID: 30405817 DOI: 10.3892/ol.2018.9455] [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: 08/10/2017] [Accepted: 05/25/2018] [Indexed: 01/10/2023] Open
Abstract
Pancreatic cancer (PC) is a highly aggressive type of cancer. Gemcitabine (GEM) is a standard chemotherapeutic treatment of advanced PC; however, it requires improvement, and more effective therapeutic methods must be further explored. In the present study, hyperthermia combined with GEM was used on the PC cell line SW1990. The results revealed that mild hyperthermia (at 42°C) effectively increased the inhibitory effect of GEM on cell viability, as determined using an MTT assay, and increased the effect of GEM-induced apoptosis, as determined using an Annexin V-fluorescein isothiocyanate/propidium iodide assay, in PC SW1990 cells. Additionally, it resulted in increased S-phase arrest, downregulated the expression of the anti-apoptosis protein B-cell lymphoma 2 and upregulated the expression of the pro-apoptosis protein Bcl-2-associated X protein, cleaved caspase-3 and cleaved caspase-9, as determined using a reverse transcription-quantitative polymerase chain reaction and western blot analysis. Furthermore, it was revealed that hyperthermia resulted in the rapid generation of reactive oxygen species (ROS) and substantial activation of c-Jun-N-terminal kinase (JNK). The introduction of ROS and JNK inhibitors suppressed hyperthermia-induced apoptosis in GEM-treated cells, suggesting that hyperthermia increased GEM cytotoxicity in PC SW1990 cells by inducing apoptosis via the ROS/JNK signaling pathway.
Collapse
Affiliation(s)
- Hangbin Jin
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Oncology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yanyan Zhao
- Department of Oncology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Jianfeng Yang
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Shenglin Ma
- Department of Oncology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
26
|
Eriksen AZ, Eliasen R, Oswald J, Kempen PJ, Melander F, Andresen TL, Young M, Baranov P, Urquhart AJ. Multifarious Biologic Loaded Liposomes that Stimulate the Mammalian Target of Rapamycin Signaling Pathway Show Retina Neuroprotection after Retina Damage. ACS NANO 2018; 12:7497-7508. [PMID: 30004669 PMCID: PMC6117751 DOI: 10.1021/acsnano.8b00596] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/13/2018] [Indexed: 05/08/2023]
Abstract
A common event in optic neuropathies is the loss of axons and death of retinal ganglion cells (RGCs) resulting in irreversible blindness. Mammalian target of rapamycin (mTOR) signaling pathway agonists have been shown to foster axon regeneration and RGC survival in animal models of optic nerve damage. However, many challenges remain in developing therapies that exploit cell growth and tissue remodeling including (i) activating/inhibiting cell pathways synergistically, (ii) avoiding tumorigenesis, and (iii) ensuring appropriate physiological tissue function. These challenges are further exacerbated by the need to overcome ocular physiological barriers and clearance mechanisms. Here we present liposomes loaded with multiple mTOR pathway stimulating biologics designed to enhance neuroprotection after retina damage. Liposomes were loaded with ciliary neurotrophic factor, insulin-like growth factor 1, a lipopeptide N-fragment osteopontin mimic, and lipopeptide phosphatase tension homologue inhibitors for either the ATP domain or the c-terminal tail. In a mouse model of N-methyl-d-aspartic acid induced RGC death, a single intravitreal administration of liposomes reduced both RGC death and loss of retina electrophysiological function. Furthermore, combining liposomes with transplantation of induced pluripotent stem cell derived RGCs led to an improved electrophysiological outcome in mice. The results presented here show that liposomes carrying multiple signaling pathway modulators can facilitate neuroprotection and transplant electrophysiological outcome.
Collapse
Affiliation(s)
- Anne Z. Eriksen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Rasmus Eliasen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Julia Oswald
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Paul J. Kempen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Fredrik Melander
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thomas L. Andresen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Michael Young
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Petr Baranov
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Andrew J. Urquhart
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
27
|
Palanissami G, Paul SFD. RAGE and Its Ligands: Molecular Interplay Between Glycation, Inflammation, and Hallmarks of Cancer—a Review. Discov Oncol 2018; 9:295-325. [DOI: 10.1007/s12672-018-0342-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
|
28
|
Development and characterization of gemcitabine hydrochloride loaded lipid polymer hybrid nanoparticles (LPHNs) using central composite design. Int J Pharm 2018; 548:255-262. [PMID: 29969712 DOI: 10.1016/j.ijpharm.2018.06.063] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/31/2022]
Abstract
Lipid polymer hybrid nanoparticles (LPHNs) combine the characteristics and beneficial properties of both polymeric nanoparticles and liposomes. The objective of this study was to design and optimize gemcitabine hydrochloride loaded LPHNs based on the central composite design approach. PLGA 50:50/PLGA 65:35 mass ratio (w/w), soya phosphatidylcholine (SPC)/polymer mass ratio (%, w/w) and amount of DSPE-PEG were chosen as the investigated independent variables. The LPHNs were prepared with modified double emulsion solvent evaporation method and characterized by testing their particle size, encapsulation efficiency, and cumulative release. The composition of optimal formulation was determined as 1,5 (w/w) PLGA 50:50/PLGA 65:35 mass ratio, 30% (w/w) SPC/polymer mass ratio and 15 mg DSPE-PEG. The results showed that the optimal formulation gemcitabine hydrochloride loaded LPHNs had encapsulation efficiency of 45,2%, particle size of 237 nm and cumulative release of 62,3% at the end of 24 h. The morphology of LPHNs was found to be spherical by transmission electron microscopy (TEM) observation. Stability studies showed that LPHNs were physically stable until 12 months at 4 °C and 9 months at 25 °C/60% RH. The results suggest that the LPHNs can be an effective drug delivery system for hydrophilic active pharmaceutical ingredient.
Collapse
|
29
|
Jin H, Zhao Y, Zhang S, Yang J, Zhang X, Ma S. Hyperthermia inhibits the motility of gemcitabine-resistant pancreatic cancer PANC-1 cells through the inhibition of epithelial-mesenchymal transition. Mol Med Rep 2018; 17:7274-7280. [PMID: 29568909 DOI: 10.3892/mmr.2018.8763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/20/2017] [Indexed: 11/05/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most common types of malignant tumor and the leading cause of cancer‑associated mortality worldwide. The chemotherapeutic drug gemcitabine (GEM) is used as a first‑line chemotherapeutic agent for advanced PC. However, the acquisition of drug resistance is a major limitation of the clinical effect of GEM and commonly leads to increased metastasis. The occurrence of epithelial‑mesenchymal transition (EMT) has been demonstrated to be the underlying mechanism of acquired resistance. It has been reported that heat treatment is able to inhibit EMT in pancreatic adenocarcinoma cells. In the present study the effect of hyperthermia on the sensitivity of GEM‑resistant PC cells was investigated. First a GEM‑resistant PC cell line PANC‑1 (PAN/GEM) was developed and it was demonstrated that drug resistant PAN/GEM cells exhibited significantly increased migratory and invasive abilities compared with control PANC‑1 cells using a Transwell assay. EMT was induced in resistant PAN/GEM cells, followed by reduced epithelial marker epithelial (E)‑cadherin expression and increased mesenchymal marker Vimentin expression compared with control PANC‑1 cells. Next, the Transwell assay demonstrated that the hyperthermia at 42˚C for 1 h combined with GEM significantly attenuated migration and invasion in drug resistant PAN/GEM cells, while GEM alone treatment did not significantly affect the migration and invasion. Additionally, EMT in PAN/GEM cells was reversed by hyperthermia, as demonstrated by the restoration of E‑cadherin and downregulation of mesenchymal markers Vimentin, matrix metalloproteinase (MMP)2 and MMP9. Furthermore, an MMP2 inhibitor tissue inhibitor of metalloproteinases (TIMP)2 and MMP9 inhibitor TIMP1 were used to treat PAN/GEM cells and it was demonstrated that both inhibitors increased the inhibition of hyperthermia treatment combined with GEM on cell invasion, suggesting an association between cell invasion and MMP2, and MMP9. Additionally, proliferation of PAN/GEM cells following hyperthermia was assessed using an MTT assay. The results demonstrated that proliferation in PAN/GEM cells treated with hyperthermia was significantly inhibited by GEM compared with GEM alone treated cells, indicating that hyperthermia enhanced the inhibition of GEM on cell growth and resensitized the drug‑resistant cells to GEM. Overall, the results of the present study suggested that hyperthermia is able to resensitize GEM‑resistant PANC‑1 cells to GEM by reversing EMT via the regulation of EMT‑associated factors, therefore inhibiting cell migration and invasion.
Collapse
Affiliation(s)
- Hangbin Jin
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yanyan Zhao
- Department of Oncology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Shirong Zhang
- Department of Oncology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Jianfeng Yang
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Shenglin Ma
- Department of Oncology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
30
|
Qiu L, Dong C, Kan X. Lymphoma-targeted treatment using a folic acid-decorated vincristine-loaded drug delivery system. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:863-872. [PMID: 29713144 PMCID: PMC5909786 DOI: 10.2147/dddt.s152420] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose B-cell lymphoma is the most frequently diagnosed lymphoid tumor. Folic acid (FA)-decorated systems were found to be preferentially internalized on the B-cell lymphoma cell line which is reported to express the folate receptor. This study was designed to develop an FA-decorated vincristine (VCR)-loaded system for targeted lymphoma treatment. Methods FA-decorated lipid was synthesized. VCR-loaded lipid-polymer hybrid nanoparticles (LPNs) were fabricated. In vitro cell lines and an in vivo lymphoma animal model was used to evaluate the anti B-cell lymphoma effect. Results FA-decorated, VCR-loaded LPNs (FA-VCR/LPNs) have shown a targeted effect in delivery to B-cell lymphoma cells. FA-VCR/LPNs also showed the highest anti-tumor effect in murine-bearing lymphoma xenografts. Conclusion FA-VCR/LPNs can achieve targeted delivery of VCR, bring about an outstanding therapeutic effect to treat lymphoma, and also reduce the systemic toxicity. FA-VCR/LPNs could be an excellent system for lymphoma therapy.
Collapse
Affiliation(s)
- Lei Qiu
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji'nan, Shandong Province, People's Republic of China
| | - Chao Dong
- Department of Oncology, 105 Hospital of People's Liberation Army, Heifei, Anhui Province, People's Republic of China
| | - Xuan Kan
- Department of Oncology, Hospital of Traditional Chinese Medicine of Laiwu City, Laiwu, Shandong Province, People's Republic of China
| |
Collapse
|
31
|
Tan S, Wang G. Redox-responsive and pH-sensitive nanoparticles enhanced stability and anticancer ability of erlotinib to treat lung cancer in vivo. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3519-3529. [PMID: 29263650 PMCID: PMC5726363 DOI: 10.2147/dddt.s151422] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Purpose Erlotinib (ETB) is a well-established therapeutic for non-small-cell lung cancer (NSCLC). To overcome drug resistance and severe toxicities in the clinical application, redox-responsive and pH-sensitive nanoparticle drug delivery systems were designed for the encapsulation of ETB. Methods Poly(acrylic acid)-cystamine-oleic acid (PAA-ss-OA) was synthesized. PAA-ss-OA-modified ETB-loaded lipid nanoparticles (PAA-ETB-NPs) were prepared using the emulsification and solvent evaporation method. The tumor inhibition efficacy of PAA-ETB-NPs was compared with that of ETB-loaded lipid nanoparticles (ETB-NPs) and free ETB anticancer drugs in tumor-bearing mice. Results PAA-ETB-NPs had a size of 170 nm, with a zeta potential of −32 mV. The encapsulation efficiency and drug loading capacity of PAA-ETB-NPs were over 85% and 2.6%, respectively. In vitro cytotoxicity of ETB-NPs were higher than that of ETB solution. The cytotoxicity of PAA-ETB-NPs was the highest. The in vivo tumor growth inhibition by PAA-ETB-NP treatment was significantly higher than that by ETB-NPs and ETB solution. No obvious weight loss was observed in any of the treatment groups, indicating that all the treatments were well tolerated. Conclusion PAA-ETB-NPs could enhance the stability and anti-cancer ability of ETB to treat lung cancer and are a promising drug delivery system for lung cancer treatment.
Collapse
Affiliation(s)
- Sheng Tan
- Department of Thoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Guoxiang Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| |
Collapse
|
32
|
Roy A, Libard S, Weishaupt H, Gustavsson I, Uhrbom L, Hesselager G, Swartling FJ, Pontén F, Alafuzoff I, Tchougounova E. Mast Cell Infiltration in Human Brain Metastases Modulates the Microenvironment and Contributes to the Metastatic Potential. Front Oncol 2017. [PMID: 28626727 PMCID: PMC5454042 DOI: 10.3389/fonc.2017.00115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Metastatic brain tumors continue to be a clinical problem, despite new therapeutic advances in cancer treatment. Brain metastases (BMs) are among the most common mass lesions in the brain that are resistant to chemotherapies, have a very poor prognosis, and currently lack any efficient diagnostic tests. Predictions estimate that about 40% of lung and breast cancer patients will develop BM. Despite this, very little is known about the immunological and genetic aberrations that drive tumorigenesis in BM. In this study, we demonstrate the infiltration of mast cells (MCs) in a large cohort of human BM samples with different tissues of origin for primary cancer. We applied patient-derived BM cell models to the study of BM cell-MC interactions. BM cells when cocultured with MCs demonstrate enhanced growth and self-renewal capacity. Gene set enrichment analyses indicate increased expression of signal transduction and transmembrane proteins related genes in the cocultured BM cells. MCs exert their effect by release of mediators such as IL-8, IL-10, matrix metalloprotease 2, and vascular endothelial growth factor, thereby permitting metastasis. In conclusion, we provide evidence for a role of MCs in BM. Our findings indicate MCs' capability of modulating gene expression in BM cells and suggest that MCs can serve as a new target for drug development against metastases in the brain.
Collapse
Affiliation(s)
- Ananya Roy
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden.,Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sylwia Libard
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Holger Weishaupt
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Ida Gustavsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Lene Uhrbom
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Göran Hesselager
- Department of Neurosurgery, Uppsala University, University Hospital, Uppsala, Sweden
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden.,Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Elena Tchougounova
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| |
Collapse
|
33
|
Goto A, Yen HC, Anraku Y, Fukushima S, Lai PS, Kato M, Kishimura A, Kataoka K. Facile Preparation of Delivery Platform of Water-Soluble Low-Molecular-Weight Drugs Based on Polyion Complex Vesicle (PICsome) Encapsulating Mesoporous Silica Nanoparticle. ACS Biomater Sci Eng 2017; 3:807-815. [DOI: 10.1021/acsbiomaterials.6b00562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Akinori Goto
- Watarase
Research Center, Kyorin Pharmaceutical CO., LTD., 1848, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | | | | | | | - Ping-Shan Lai
- Department
of Chemistry, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung 402, Taiwan
| | - Masaru Kato
- Graduate
School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033 Japan
| | | | - Kazunori Kataoka
- Innovation
Center of NanoMedicne, Kawasaki Institute of Industry Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| |
Collapse
|
34
|
Cimpean AM, Raica M. The Hidden Side of Disodium Cromolyn: from Mast Cell Stabilizer to an Angiogenic Factor and Antitumor Agent. Arch Immunol Ther Exp (Warsz) 2016; 64:515-522. [DOI: 10.1007/s00005-016-0408-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 03/09/2016] [Indexed: 12/16/2022]
|
35
|
Dubey RD, Saneja A, Gupta PK, Gupta PN. Recent advances in drug delivery strategies for improved therapeutic efficacy of gemcitabine. Eur J Pharm Sci 2016; 93:147-62. [PMID: 27531553 DOI: 10.1016/j.ejps.2016.08.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 02/07/2023]
Abstract
Gemcitabine (2',2'-difluoro-2'-deoxycytidine; dFdC) is an efficacious anticancer agent acting against a wide range of solid tumors, including pancreatic, non-small cell lung, bladder, breast, ovarian, thyroid and multiple myelomas. However, short plasma half-life due to metabolism by cytidine deaminase necessitates administration of high dose, which limits its medical applicability. Further, due to its hydrophilic nature, it cannot traverse cell membranes by passive diffusion and, therefore, enters via nucleoside transporters that may lead to drug resistance. To circumvent these limitations, macromolecular prodrugs and nanocarrier-based formulations of Gemcitabine are gaining wide recognition. The nanoformulations based approaches by virtue of their controlled release and targeted delivery have proved to improve bioavailability, increase therapeutic efficacy and reduce adverse effects of the drug. Furthermore, the combination of Gemcitabine with other anticancer agents as well as siRNAs using nanocarriers has also been investigated in order to enhance its therapeutic potential. This review deals with challenges and recent advances in the delivery of Gemcitabine with particular emphasis on macromolecular prodrugs and nanomedicines.
Collapse
Affiliation(s)
- Ravindra Dhar Dubey
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India
| | - Ankit Saneja
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India
| | - Prasoon K Gupta
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India.
| | - Prem N Gupta
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India.
| |
Collapse
|
36
|
Han NK, Shin DH, Kim JS, Weon KY, Jang CY, Kim JS. Hyaluronan-conjugated liposomes encapsulating gemcitabine for breast cancer stem cells. Int J Nanomedicine 2016; 11:1413-25. [PMID: 27103799 PMCID: PMC4827594 DOI: 10.2147/ijn.s95850] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Investigation of potential therapeutics for targeting breast cancer stem cells (BCSCs) is important because these cells are regarded as culprit of breast cancer relapse. Accomplishing this kind of strategy requires a specific drug-delivery system using the distinct features of liposomes. Studies on targeted liposomal delivery systems have indicated the conjugation of hyaluronan (HA), a primary ligand for CD44 surface markers, as an appropriate method for targeting BCSCs. For this study, enriched BCSCs were obtained by culturing MCF-7 breast cancer cells in nonadherent conditions. The enriched BCSCs were challenged with HA-conjugated liposomes encapsulating gemcitabine (2, 2-difluoro-2-deoxycytidine, GEM). In vitro study showed that the HA-conjugated liposomes significantly enhanced the cytotoxicity, anti-migration, and anti-colony formation abilities of GEM through targeting of CD44 expressed on BCSCs. In pharmacokinetic study, area under the drug concentration vs time curve (AUC) of the immunoliposomal GEM was 3.5 times higher than that of free GEM, indicating that the HA-conjugated liposomes enhanced the stability of GEM in the bloodstream and therefore prolonged its half-life time. The antitumor effect of the immunoliposomal GEM was 3.3 times higher than that of free GEM in a xenograft mouse model, probably reflecting the unique targeting of the CD44 receptor by HA and the increased cytotoxicity and stability through the liposomal formulation. Furthermore, marginal change in body weight demonstrated that the use of liposomes considerably reduced the systemic toxicity of GEM on normal healthy cells. Taken together, this study demonstrates that HA-conjugated liposomes encapsulating GEM show promise for the therapy of breast cancer in vitro and in a xenograft model by targeting the BCSCs.
Collapse
Affiliation(s)
- Na-Kyung Han
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Dae Hwan Shin
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Jung Seok Kim
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Kwon Yeon Weon
- College of Pharmacy, Catholic University of Daegu, Gyeongbuk, Korea
| | - Chang-Young Jang
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Jin-Seok Kim
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| |
Collapse
|
37
|
Leclerc E, Vetter SW. The role of S100 proteins and their receptor RAGE in pancreatic cancer. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2706-11. [PMID: 26435083 DOI: 10.1016/j.bbadis.2015.09.022] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with low survival rates. Current therapeutic treatments have very poor response rates due to the high inherent chemoresistance of the pancreatic-cancer cells. Recent studies have suggested that the receptor for advanced glycation end products (RAGE) and its S100 protein ligands play important roles in the progression of PDAC. We will discuss the potential role of S100 proteins and their receptor, RAGE, in the development and progression of pancreatic cancer.
Collapse
Affiliation(s)
- Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, PO Box 6050, Department 2665, Fargo, ND 58108-6050, USA.
| | - Stefan W Vetter
- Department of Pharmaceutical Sciences, North Dakota State University, PO Box 6050, Department 2665, Fargo, ND 58108-6050, USA
| |
Collapse
|
38
|
Jiang L, Li L, He X, Yi Q, He B, Cao J, Pan W, Gu Z. Overcoming drug-resistant lung cancer by paclitaxel loaded dual-functional liposomes with mitochondria targeting and pH-response. Biomaterials 2015; 52:126-39. [DOI: 10.1016/j.biomaterials.2015.02.004] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 01/22/2015] [Accepted: 02/01/2015] [Indexed: 02/06/2023]
|
39
|
Kirui DK, Celia C, Molinaro R, Bansal SS, Cosco D, Fresta M, Shen H, Ferrari M. Mild hyperthermia enhances transport of liposomal gemcitabine and improves in vivo therapeutic response. Adv Healthc Mater 2015; 4:1092-103. [PMID: 25721343 PMCID: PMC4433418 DOI: 10.1002/adhm.201400738] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 01/17/2015] [Indexed: 12/11/2022]
Abstract
Obstructive biological barriers limit the transport and efficacy of cancer nanotherapeutics. Creative manipulation of tumor microenvironment provides promising avenues towards improving chemotherapeutic response. Such strategies include the use of mechanical stimuli to overcome barriers, and increase drug delivery and therapeutic efficacy. The rational use of gold nanorod-mediated mild hyperthermia treatment (MHT) alters tumor transport properties, increases liposomal gemcitabine (Gem Lip) delivery, and antitumor efficacy in pancreatic cancer CAPAN-1 tumor model. MHT treatment leads to a threefold increase in accumulation of 80-nm liposomes and enhances spatial interstitial distribution. I.v. injection of Gem Lip and MHT treatment lead to a threefold increase in intratumor gemcitabine concentration compared to chemotherapeutic infusion alone. Furthermore, combination of MHT treatment with infusion of 12 mg kg(-1) Gem Lip leads to a twofold increase in therapeutic efficacy and inhibition of CAPAN-1 tumor growth when compared to equimolar chemotherapeutic treatment alone. Enhanced therapeutic effect is confirmed by reduction in tumor size and increase in apoptotic index where MHT treatment combined with 12 mg kg(-1) Gem Lip achieves similar therapeutic efficacy as the use of 60 mg kg(-1) free gemcitabine. In conclusion, improvements in vivo efficacy are demonstrated resulting from MHT treatment that overcome transport barriers, promote delivery, improve efficacy of nanomedicines.
Collapse
Affiliation(s)
- Dickson K Kirui
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
| | - Christian Celia
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Pharmacy, University of Chieti – Pescara “G. d’Annunzio”, Chieti, 66013, Italy
| | - Roberto Molinaro
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Germaneto – Catanzaro, 88100, Italy
| | - Shyam S. Bansal
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
| | - Donato Cosco
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Germaneto – Catanzaro, 88100, Italy
| | - Massimo Fresta
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Germaneto – Catanzaro, 88100, Italy
| | - Haifa Shen
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Cell and Development Biology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Mauro Ferrari
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Biomedical Engineering in Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| |
Collapse
|
40
|
de Souza PC, Ranjan A, Towner RA. Nanoformulations for therapy of pancreatic and liver cancers. Nanomedicine (Lond) 2015; 10:1515-34. [DOI: 10.2217/nnm.14.231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pancreatic and liver cancers often have poor prognoses. Clinically, pancreatic and liver cancer requires early diagnosis, and surgery is often associated with tumor recurrence. Currently, chemotherapies are limited in their ability to accurately target the tumors, and are associated with significant toxicity in patients. Targeting of chemotherapy can be improved by encapsulation in nanocarriers. A variety of preclinical studies indicate relatively superior therapeutic outcomes compared with drug alone therapy. Targeted nanoparticle imaging agents may also additionally facilitate better diagnosis and improve patient outcomes. This review discusses the nanoformulations that are under investigation (mainly preclinical studies, but also with some current clinical trial examples) against pancreatic and liver cancers, understands the challenges and provides future perspectives.
Collapse
Affiliation(s)
- Patricia Coutinho de Souza
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74074, USA
- Advanced Magnetic Resonance Center, MS 60, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Ashish Ranjan
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74074, USA
| | - Rheal A Towner
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74074, USA
- Advanced Magnetic Resonance Center, MS 60, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
41
|
Tila D, Ghasemi S, Yazdani-Arazi SN, Ghanbarzadeh S. Functional liposomes in the cancer-targeted drug delivery. J Biomater Appl 2015; 30:3-16. [PMID: 25823898 DOI: 10.1177/0885328215578111] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer is considered as one of the most severe health problems and is currently the third most common cause of death in the world after heart and infectious diseases. Novel therapies are constantly being discovered, developed and trialed. Many of the current anticancer agents exhibit non-ideal pharmaceutical and pharmacological properties and are distributed non-specifically throughout the body. This results in death of the both normal healthy and malignant cells and substantially leads to accruing a variety of serious toxic side effects. Therefore, the efficient systemic therapy of cancer is almost impossible due to harmful side effects of anticancer agents to the healthy organs and tissues. Furthermore, several problems such as low bioavailability of the drugs, low drug concentrations at the site of action, lack of drug specificity and drug-resistance also cause many restrictions on clinical applications of these drugs in the tumor therapy. Different types of the liposomal formulations have been used in medicine due to their distinctive advantages associated with their structural flexibility in the encapsulation of various agents with different physicochemical properties. They can also mediate delivery of the cargo to the appropriate cell type and subcellular compartment, reducing the effective dosage and possible side effects which are related to high systemic concentrations. Therefore, these novel systems were found very promising and encouraging dosage forms for the treatment of different types of cancer by increasing efficiency and reducing the systemic toxicity due to the specific drug delivery and targeting.
Collapse
Affiliation(s)
- Dena Tila
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Ghasemi
- Department of Medicinal Chemistry, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Saeed Ghanbarzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Galli SJ, Tsai M, Marichal T, Tchougounova E, Reber LL, Pejler G. Approaches for analyzing the roles of mast cells and their proteases in vivo. Adv Immunol 2015; 126:45-127. [PMID: 25727288 DOI: 10.1016/bs.ai.2014.11.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The roles of mast cells in health and disease remain incompletely understood. While the evidence that mast cells are critical effector cells in IgE-dependent anaphylaxis and other acute IgE-mediated allergic reactions seems unassailable, studies employing various mice deficient in mast cells or mast cell-associated proteases have yielded divergent conclusions about the roles of mast cells or their proteases in certain other immunological responses. Such "controversial" results call into question the relative utility of various older versus newer approaches to ascertain the roles of mast cells and mast cell proteases in vivo. This review discusses how both older and more recent mouse models have been used to investigate the functions of mast cells and their proteases in health and disease. We particularly focus on settings in which divergent conclusions about the importance of mast cells and their proteases have been supported by studies that employed different models of mast cell or mast cell protease deficiency. We think that two major conclusions can be drawn from such findings: (1) no matter which models of mast cell or mast cell protease deficiency one employs, the conclusions drawn from the experiments always should take into account the potential limitations of the models (particularly abnormalities affecting cell types other than mast cells) and (2) even when analyzing a biological response using a single model of mast cell or mast cell protease deficiency, details of experimental design are critical in efforts to define those conditions under which important contributions of mast cells or their proteases can be identified.
Collapse
Affiliation(s)
- Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA; Microbiology & Immunology, Stanford University School of Medicine, Stanford, California, USA.
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Thomas Marichal
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA; GIGA-Research and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Elena Tchougounova
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
43
|
Abstract
In humans, the S100 protein family is composed of 21 members that exhibit a high degree of structural similarity, but are not functionally interchangeable. This family of proteins modulates cellular responses by functioning both as intracellular Ca(2+) sensors and as extracellular factors. Dysregulated expression of multiple members of the S100 family is a common feature of human cancers, with each type of cancer showing a unique S100 protein profile or signature. Emerging in vivo evidence indicates that the biology of most S100 proteins is complex and multifactorial, and that these proteins actively contribute to tumorigenic processes such as cell proliferation, metastasis, angiogenesis and immune evasion. Drug discovery efforts have identified leads for inhibiting several S100 family members, and two of the identified inhibitors have progressed to clinical trials in patients with cancer. This Review highlights new findings regarding the role of S100 family members in cancer diagnosis and treatment, the contribution of S100 signalling to tumour biology, and the discovery and development of S100 inhibitors for treating cancer.
Collapse
Affiliation(s)
- Anne R. Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - David J. Weber
- Center for Biomolecular Therapeutics and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, Maryland 20102, USA
| | - Danna B. Zimmer
- Center for Biomolecular Therapeutics and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, Maryland 20102, USA
| |
Collapse
|
44
|
Kennedy LL, Hargrove LA, Graf AB, Francis TC, Hodges KM, Nguyen QP, Ueno Y, Greene JF, Meng F, Huynh VD, Francis HL. Inhibition of mast cell-derived histamine secretion by cromolyn sodium treatment decreases biliary hyperplasia in cholestatic rodents. J Transl Med 2014; 94:1406-18. [PMID: 25365204 DOI: 10.1038/labinvest.2014.129] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/07/2014] [Indexed: 02/07/2023] Open
Abstract
Cholangiopathies are characterized by dysregulation of the balance between biliary growth and loss. We have shown that histamine (HA) stimulates biliary growth via autocrine mechanisms. To evaluate the paracrine effects of mast cell (MC) stabilization on biliary proliferation, sham or BDL rats were treated by IP-implanted osmotic pumps filled with saline or cromolyn sodium (24 mg/kg BW/day (inhibits MC histamine release)) for 1 week. Serum, liver blocks and cholangiocytes were collected. Histidine decarboxylase (HDC) expression was measured using real-time PCR in cholangiocytes. Intrahepatic bile duct mass (IBDM) was evaluated by IHC for CK-19. MC number was determined using toluidine blue staining and correlated to IBDM. Proliferation was evaluated by PCNA expression in liver sections and purified cholangiocytes. We assessed apoptosis using real-time PCR and IHC for BAX. Expression of MC stem factor receptor, c-kit, and the proteases chymase and tryptase were measured by real-time PCR. HA levels were measured in serum by EIA. In vitro, MCs and cholangiocytes were treated with 0.1% BSA (basal) or cromolyn (25 μM) for up to 48 h prior to assessing HDC expression, HA levels and chymase and tryptase expression. Supernatants from MCs treated with or without cromolyn were added to cholangiocytes before measuring (i) proliferation by MTT assays, (ii) HDC gene expression by real-time PCR and (iii) HA release by EIA. In vivo, cromolyn treatment decreased BDL-induced: (i) IBDM, MC number, and biliary proliferation; (ii) HDC and MC marker expression; and (iii) HA levels. Cromolyn treatment increased cholangiocyte apoptosis. In vitro, cromolyn decreased HA release and chymase and tryptase expression in MCs but not in cholangiocytes. Cromolyn-treated MC supernatants decreased biliary proliferation and HA release. These studies provide evidence that MC histamine is key to biliary proliferation and may be a therapeutic target for the treatment of cholangiopathies.
Collapse
Affiliation(s)
- Lindsey L Kennedy
- Digestive Disease Research Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Laura A Hargrove
- Scott and White Digestive Disease Research Center, Scott and White, Temple, TX, USA
| | - Allyson B Graf
- Digestive Disease Research Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Taylor C Francis
- Department of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Kyle M Hodges
- Scott and White Digestive Disease Research Center, Scott and White, Temple, TX, USA
| | - Quy P Nguyen
- Digestive Disease Research Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Yoshi Ueno
- CREST, Japan Science and Technology Corporation, Tokyo, Japan
| | - John F Greene
- Scott and White Digestive Disease Research Center, Scott and White, Temple, TX, USA
| | - Fanyin Meng
- 1] Digestive Disease Research Center, Central Texas Veterans Health Care System, Temple, TX, USA [2] Scott and White Digestive Disease Research Center, Scott and White, Temple, TX, USA [3] Department of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Victoria D Huynh
- Department of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Heather L Francis
- 1] Digestive Disease Research Center, Central Texas Veterans Health Care System, Temple, TX, USA [2] Scott and White Digestive Disease Research Center, Scott and White, Temple, TX, USA [3] Department of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
45
|
Reber LL, Frossard N. Targeting mast cells in inflammatory diseases. Pharmacol Ther 2014; 142:416-35. [PMID: 24486828 DOI: 10.1016/j.pharmthera.2014.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 12/24/2022]
Abstract
Although mast cells have long been known to play a critical role in anaphylaxis and other allergic diseases, they also participate in some innate immune responses and may even have some protective functions. Data from the study of mast cell-deficient mice have facilitated our understanding of some of the molecular mechanisms driving mast cell functions during both innate and adaptive immune responses. This review presents an overview of the biology of mast cells and their potential involvement in various inflammatory diseases. We then discuss some of the current pharmacological approaches used to target mast cells and their products in several diseases associated with mast cell activation.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, Faculté de Pharmacie, France
| |
Collapse
|
46
|
Elmowafy M, Viitala T, Ibrahim HM, Abu-Elyazid SK, Samy A, Kassem A, Yliperttula M. Silymarin loaded liposomes for hepatic targeting: in vitro evaluation and HepG2 drug uptake. Eur J Pharm Sci 2013; 50:161-71. [PMID: 23851081 DOI: 10.1016/j.ejps.2013.06.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 06/04/2013] [Accepted: 06/20/2013] [Indexed: 11/18/2022]
Abstract
Silymarin has hepatoprotective properties and is used in treatment of various liver diseases, but its bioavailability from oral products is very poor. In order to overcome its poor oral bioavailability we have prepared silymarin loaded hepatic targeting liposomes suitable for parenteral administration. The liposomal formulations were composed of hydrogenated soy phosphatidylcholine and cholesterol with or without distearoylphosphoethanolamine-(polyethyleneglycol)-2000 and various amounts of β-sitosterol β-D-glucoside (Sito-G) as the hepatic targeting moiety. Increasing the amount of Sito-G in the liposomes gradually decreased drug encapsulation efficiencies from ∼70% to ∼60%; still showing promising drug encapsulation efficiencies. Addition of Sito-G to non-PEGylated liposomes clearly affected their drug release profiles and plasma protein interactions, whereas no effect on these was seen for the PEGylated liposomes. Non-PEGylated liposomes with 0.17 M ratio of Sito-G exhibited the highest cellular drug uptake of 37.5% for all of the studied liposome formulations. The highest cellular drug uptake in the case of PEGylated liposomes was 18%, which was achieved with 0.17 and 0.33 M ratio of added Sito-G. The liposome formulations with the highest drug delivery efficacy in this study showed hemolytic activities around 12.7% and were stable for at least 2 months upon storage in 20 mM HEPES buffer (pH 7.4) containing 1.5% Polysorbate 80 at 4 °C and room temperature. These results suggest that the Sito-G containing liposomes prepared in this work have hepatic targeting capability and that they are promising candidates for delivering silymarin to the liver.
Collapse
Affiliation(s)
- Mohammed Elmowafy
- Division of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 000014 Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
47
|
Arumugam T, Ramachandran V, Sun D, Peng Z, Pal A, Maxwell DS, Bornmann WG, Logsdon CD. Designing and developing S100P inhibitor 5-methyl cromolyn for pancreatic cancer therapy. Mol Cancer Ther 2013; 12:654-62. [PMID: 23303403 DOI: 10.1158/1535-7163.mct-12-0771] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have previously shown that the antiallergic drug cromolyn blocks S100P interaction with its receptor receptor for advanced glycation end product (RAGE) and improves gemcitabine effectiveness in pancreatic ductal adenocarcinoma (PDAC). However, the concentration required to achieve its effectiveness was high (100 μmol/L). In this study, we designed and synthesized analogs of cromolyn and analyzed their effectiveness compared with the parent molecule. An ELISA was used to confirm the binding of S100P with RAGE and to test the effectiveness of the different analogs. Analog 5-methyl cromolyn (C5OH) blocked S100P binding as well as the increases in NF-κB activity, cell growth, and apoptosis normally caused by S100P. In vivo C5OH systemic delivery reduced NF-κB activity to a greater extent than cromolyn and at 10 times lesser dose (50 mg vs. 5 mg). Treatment of mice-bearing syngeneic PDAC tumors showed that C5OH treatment reduced both tumor growth and metastasis. C5OH treatment of nude mice bearing orthotopic highly aggressive pancreatic Mpanc96 cells increased the overall animal survival. Therefore, the cromolyn analog, C5OH, was found to be more efficient and potent than cromolyn as a therapeutic for PDAC.
Collapse
|
48
|
Martínez A, Muñiz E, Iglesias I, Teijón J, Blanco M. Enhanced preclinical efficacy of tamoxifen developed as alginate–cysteine/disulfide bond reduced albumin nanoparticles. Int J Pharm 2012; 436:574-81. [DOI: 10.1016/j.ijpharm.2012.07.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 07/18/2012] [Accepted: 07/20/2012] [Indexed: 12/11/2022]
|