1
|
Balaji A, Bell CA, Houston ZH, Bridle KR, Genz B, Fletcher NL, Ramm GA, Thurecht KJ. Exploring the impact of severity in hepatic fibrosis disease on the intrahepatic distribution of novel biodegradable nanoparticles targeted towards different disease biomarkers. Biomaterials 2023; 302:122318. [PMID: 37708659 DOI: 10.1016/j.biomaterials.2023.122318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
Nanoparticle-based drug delivery systems (DDS) have shown promising results in reversing hepatic fibrosis, a common pathological basis of chronic liver diseases (CLDs), in preclinical animal models. However, none of these nanoparticle formulations has transitioned to clinical usage and there are currently no FDA-approved drugs available for liver fibrosis. This highlights the need for a better understanding of the challenges faced by nanoparticles in this complex disease setting. Here, we have systematically studied the impact of targeting strategy, the degree of macrophage infiltration during fibrosis, and the severity of fibrosis, on the liver uptake and intrahepatic distribution of nanocarriers. When tested in mice with advanced liver fibrosis, we demonstrated that the targeting ligand density plays a significant role in determining the uptake and retention of the nanoparticles in the fibrotic liver whilst the type of targeting ligand modulates the trafficking of these nanoparticles into the cell population of interest - activated hepatic stellate cells (aHSCs). Engineering the targeting strategy indeed reduced the uptake of nanoparticles in typical mononuclear phagocyte (MPS) cell populations, but not the infiltrated macrophages. Meanwhile, additional functionalization may be required to enhance the efficacy of DDS in end-stage fibrosis/cirrhosis compared to early stages.
Collapse
Affiliation(s)
- Arunpandian Balaji
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Australia
| | - Craig A Bell
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Australia; Australian Research Council Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zachary H Houston
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Australia
| | - Kim R Bridle
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia; Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland 4120, Australia
| | - Berit Genz
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland 4102, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Nicholas L Fletcher
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Australia; Australian Research Council Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Grant A Ramm
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Australia; Australian Research Council Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
2
|
Zhang YW, Hou LS, Xing JH, Zhang TR, Zhou SY, Zhang BL. Two-Membrane Hybrid Nanobiomimetic Delivery System for Targeted Autophagy Inhibition of Activated Hepatic Stellate Cells To Synergistically Treat Liver Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37899504 DOI: 10.1021/acsami.3c11046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Liver fibrosis is one of the most common and highly prevalent chronic liver diseases caused by multiple pathogenic factors, and there is still no effective therapeutic drugs up to now. The activated hepatic stellate cells (aHSCs) are the main executor in liver fibrosis, and the autophagy plays a key role in the proliferation and differentiation of aHSCs, which promotes the development of liver fibrosis. However, autophagy has the opposite effect on the different kinds of liver cells in the development of liver fibrosis, and the clinical treatment has been limited by the poor selectivity and inefficient drug delivery to aHSCs. Therefore, in this study, a liposome (Lip) and exosome (Exo) two-membrane hybrid nanobiomimetic delivery system HCQ@VA-Lip-Exo was designed, which was modified by vitamin A (VA) to target the aHSCs and carried the autophagy inhibitor hydroxychloroquine (HCQ). The experimental results in vitro and in vivo revealed that the constructed aHSC-targeted hybrid delivery system HCQ@VA-Lip-Exo combined with the benefits of HCQ and exosomes derived from bone marrow mesenchymal stem cells. HCQ@VA-Lip-Exo had good aHSC-targeted delivery ability, effective autophagy inhibition, and synergistical anti-liver fibrosis performance, thus reducing the production and deposition of the extracellular matrix to inhibit the liver fibrosis. This combined strategy provided a potential idea for the construction and clinical application of a two-membrane hybrid delivery system as an effective targeted therapy of liver fibrosis.
Collapse
Affiliation(s)
- Yao-Wen Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Li-Shuang Hou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Jie-Hua Xing
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Tang-Rui Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Si-Yuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| |
Collapse
|
3
|
Lipophilic poly(glycolide) blocks in morpholin-2-one-based CARTs for plasmid DNA delivery: Polymer regioregularity, sequence of lipophilic/polyamine blocks, and nanoparticle stability as factors of transfection efficiency. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
4
|
Luo F, Yu Y, Li M, Chen Y, Zhang P, Xiao C, Lv G. Polymeric nanomedicines for the treatment of hepatic diseases. J Nanobiotechnology 2022; 20:488. [PMCID: PMC9675156 DOI: 10.1186/s12951-022-01708-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022] Open
Abstract
The liver is an important organ in the human body and performs many functions, such as digestion, detoxification, metabolism, immune responses, and vitamin and mineral storage. Therefore, disorders of liver functions triggered by various hepatic diseases, including hepatitis B virus infection, nonalcoholic steatohepatitis, hepatic fibrosis, hepatocellular carcinoma, and transplant rejection, significantly threaten human health worldwide. Polymer-based nanomedicines, which can be easily engineered with ideal physicochemical characteristics and functions, have considerable merits, including contributions to improved therapeutic outcomes and reduced adverse effects of drugs, in the treatment of hepatic diseases compared to traditional therapeutic agents. This review describes liver anatomy and function, and liver targeting strategies, hepatic disease treatment applications and intrahepatic fates of polymeric nanomedicines. The challenges and outlooks of hepatic disease treatment with polymeric nanomedicines are also discussed.
Collapse
Affiliation(s)
- Feixiang Luo
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Ying Yu
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Mingqian Li
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Yuguo Chen
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Peng Zhang
- grid.9227.e0000000119573309Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 People’s Republic of China
| | - Chunsheng Xiao
- grid.9227.e0000000119573309Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 People’s Republic of China
| | - Guoyue Lv
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| |
Collapse
|
5
|
Vukmanovic Nosrat I, Palacios JL, Kezian S, Luong G, Tran A, Vu K, Henson BS, Nosrat P, Lutfy K, Nosrat CA. Brain-derived neurotrophic factor overexpression in taste buds diminishes chemotherapy induced taste loss. Eur J Neurosci 2022; 56:4967-4982. [PMID: 35986485 PMCID: PMC9804163 DOI: 10.1111/ejn.15799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/05/2023]
Abstract
Vismodegib is used in patients suffering from advanced basal cell carcinoma (BCC), but 100% of the patients taking it report dysgeusia and 50% discontinue the treatment. Treatment with neurotrophic factors can stimulate neuronal survival and functional improvement in injured organs. Here, we analysed novel transgenic mouse lines in which brain-derived neurotrophic factor (BDNF) is overexpressed in taste buds, to examine whether higher levels of BDNF would reduce or prevent negative side effects of vismodegib in the taste system. BDNF plays crucial roles for development, target innervation, and survival of gustatory neurons and taste buds. The behavioural test in this study showed that vehicle-treated wild-type mice prefered 10 mM sucrose over water, whereas vismodegib treatment in wild-type mice caused total taste loss. Gustducin-BDNF mice had a significantly increased preference for low concentration of sucrose solution over water compared to wild-type mice, and most importantly the transgenic mice were able to detect low concentrations of sucrose following vismodegib treatment. We evaluated taste cell morphology, identity, innervation and proliferation using immunohistochemistry. All drug-treated mice exhibited deficits, but because of a possible functional upcycled priming of the peripheral gustatory system, GB mice demonstrated better morphological preservation of the peripheral gustatory system. Our study indicates that overexpression of BDNF in taste buds plays a role in preventing degeneration of taste buds. Counteracting the negative side effects of vismodegib treatment might improve compliance and achieve better outcome in patients suffering from advanced BCC.
Collapse
Affiliation(s)
| | - Jerry L. Palacios
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCaliforniaUSA
| | - Steven Kezian
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCaliforniaUSA
| | - Gloria Luong
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCaliforniaUSA
| | - Andrew Tran
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCaliforniaUSA
| | - Kim Vu
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCaliforniaUSA
| | - Bradley S. Henson
- College of Dental MedicineWestern University of Health SciencesPomonaCaliforniaUSA
| | - Philip Nosrat
- College of Dental MedicineWestern University of Health SciencesPomonaCaliforniaUSA
| | - Kabirullah Lutfy
- Department of Pharmaceutical Sciences, College of PharmacyWestern University of Health SciencesPomonaCaliforniaUSA
| | | |
Collapse
|
6
|
Ma J, Kumar V, Mahato RI. Nanoparticle Delivery of Novel PDE4B Inhibitor for the Treatment of Alcoholic Liver Disease. Pharmaceutics 2022; 14:pharmaceutics14091894. [PMID: 36145643 PMCID: PMC9501368 DOI: 10.3390/pharmaceutics14091894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/24/2022] [Accepted: 09/04/2022] [Indexed: 11/30/2022] Open
Abstract
The incidence of alcoholic liver disease (ALD) is increasing worldwide while no effective treatment has been approved. The progression of ALD has proven to be related to the upregulation of phosphodiesterase 4 (PDE4) expression, and PDE4 inhibitors showed potential to improve ALD. However, the application of PDE4 inhibitors is limited by the gastrointestinal side effects due to PDE4D inhibition. Therefore, we used a novel PDE4B inhibitor KVA-D88 as the therapeutic for ALD treatment. KVA-D88 inhibited inflammatory response, promoted β-oxidation, increased the level of antioxidants in the hepatocytes, and suppressed hepatic stellate cell (HSC) activation in vitro. To improve the solubility and availability in vivo, KVA-D88 was encapsulated into mPEG-b-P(CB-co-LA) nanoparticles (NPs) by solvent evaporation, with a mean particle size of 135 nm and drug loading of 4.2%. We fed the male C57BL/6 mice with a Lieber–DeCarli liquid diet containing 5% (v/v) ethanol for 6 weeks to induce ALD. Systemic administration of KVA-D88 free drug and KVA-D88-loaded NPs at 5 mg/kg significantly improved the ALD in mice. KVA-D88 significantly ameliorated alcohol-induced hepatic injury and inflammation. KVA-D88 also markedly reduced steatosis by promoting fatty acid β-oxidation. Liver fibrosis and reactive oxygen species (ROS)-caused cellular damage was observed to be alleviated by KVA-D88. KVA-D88-loaded NPs proved better efficacy than free drug in the animal study. In conclusion, the novel PDE4B inhibitor KVA-D88-loaded NPs have the potential to treat ALD in mice
Collapse
Affiliation(s)
| | - Virender Kumar
- Correspondence: (V.K.); (R.I.M.); Tel.: +1-(402)-559-6422 (R.I.M.); Fax: +1-(402)-559-9543 (R.I.M.)
| | - Ram I. Mahato
- Correspondence: (V.K.); (R.I.M.); Tel.: +1-(402)-559-6422 (R.I.M.); Fax: +1-(402)-559-9543 (R.I.M.)
| |
Collapse
|
7
|
Kumar V, Xin X, Ma J, Tan C, Osna N, Mahato RI. Therapeutic targets, novel drugs, and delivery systems for diabetes associated NAFLD and liver fibrosis. Adv Drug Deliv Rev 2021; 176:113888. [PMID: 34314787 PMCID: PMC8440458 DOI: 10.1016/j.addr.2021.113888] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/12/2021] [Accepted: 07/18/2021] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) associated non-alcoholic fatty liver disease (NAFLD) is the fourth-leading cause of death. Hyperglycemia induces various complications, including nephropathy, cirrhosis and eventually hepatocellular carcinoma (HCC). There are several etiological factors leading to liver disease development, which involve insulin resistance and oxidative stress. Free fatty acid (FFA) accumulation in the liver exerts oxidative and endoplasmic reticulum (ER) stresses. Hepatocyte injury induces release of inflammatory cytokines from Kupffer cells (KCs), which are responsible for activating hepatic stellate cells (HSCs). In this review, we will discuss various molecular targets for treating chronic liver diseases, including homeostasis of FFA, lipid metabolism, and decrease in hepatocyte apoptosis, role of growth factors, and regulation of epithelial-to-mesenchymal transition (EMT) and HSC activation. This review will also critically assess different strategies to enhance drug delivery to different cell types. Targeting nanocarriers to specific liver cell types have the potential to increase efficacy and suppress off-target effects.
Collapse
Affiliation(s)
- Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Xiaofei Xin
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jingyi Ma
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, University, MS 38677, USA
| | - Natalia Osna
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
8
|
Kumar V, Rahman M, Gahtori P, Al-Abbasi F, Anwar F, Kim HS. Current status and future directions of hepatocellular carcinoma-targeted nanoparticles and nanomedicine. Expert Opin Drug Deliv 2020; 18:673-694. [PMID: 33295218 DOI: 10.1080/17425247.2021.1860939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a major health problem worldwide. Conventional therapies covering either chemotherapy or combination therapy still have sub-optimal responses with significant adverse effects and toxicity. Moreover, tumor cells usually acquire resistance quickly for traditional approaches, limiting their use in HCC. Interest in nanomedicine due to minimal systemic toxicity and a high degree of target-specific drug-delivery have pulled the attention of health scientists in this area of therapeutics. AREA COVERED The review covers the incidence and epidemiology of HCC, proposed molecular drug targets, mechanistic approach and emergence of nanomedicines including nanoparticles, lipidic nanoparticles, vesicular-based nanocarrier, virus-like particles with momentous therapeutic aspects including biocompatibility, and toxicity of nanocarriers along with conclusions and future perspective, with an efficient approach to safely cross physiological barriers to reach the target site for treating liver cancer. EXPERT OPINION Remarkable outcomes have recently been observed for the therapeutic efficacy of nanocarriers with respect to a specific drug target against the treatment of HCC by existing under trial drugs.
Collapse
Affiliation(s)
- Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Mahfoozur Rahman
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Prashant Gahtori
- School of Pharmacy, Graphic Era Hill University Dehradun 248002, Uttarakhand, India
| | - Fahad Al-Abbasi
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Firoz Anwar
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Jangan-gu, Suwon 16419, 2066, Seobu-ro, Korea
| |
Collapse
|
9
|
Wang M, Zhang M, Fu L, Lin J, Zhou X, Zhou P, Huang P, Hu H, Han Y. Liver-targeted delivery of TSG-6 by calcium phosphate nanoparticles for the management of liver fibrosis. Am J Cancer Res 2020; 10:36-49. [PMID: 31903104 PMCID: PMC6929629 DOI: 10.7150/thno.37301] [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: 06/04/2019] [Accepted: 09/15/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) transplantation is a promising antifibrotic strategy but facing clinical controversies. Inspired by advances in nanomedicine, we aimed to bypass these clinical barriers of MSCs by identifying the key antifibrotic molecule of MSCs and developing a specific liver-targeting nanocarrier. Methods: Cytokines secreted by MSCs were examined with serum stimulation of cirrhotic patients. Immunohistochemistry, microarray, immunoblotting, and quantitative real-time PCR (qRT-PCR) were applied to identify the critical antifibrotic cytokine and to discover its role in modulating antifibrotic effects. Biomineralization method was used to prepare calcium phosphate nanoparticles (NPs). The targeting and therapeutic efficiency of NPs were evaluated by in vivo imaging and biochemical studies on fibrotic mice induced by CCl4. Results: The stimulated MSCs exhibited high-level expression of Tumor necrosis factor (TNF)-stimulated gene 6 (TSG-6). On animal study, exogenous administration of TSG-6 alone can ameliorate liver fibrosis while TSG-6 knocked MSCs (Lv-TSG-6 MSCs) lost antifibrotic effects. Further studies verified the importance of TSG-6 and identified its antifibrotic mechanism by modulating M2 macrophages and increasing matrix metalloproteinase 12 (MMP12) expression. Additionally, we found a feedback loop between TSG-6, MMP12 and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), which may improve our understanding of the aggravating process of cirrhosis and antifibrotic mechanisms of TSG-6 and MSCs. Based on these findings, we developed calcium phosphate nanoparticles (CaP@BSA NPs) by biomineralization method using bovine serum albumin (BSA) as the biotemplate. Imaging tracking and drug loading studies showed specific liver targeting and high TSG-6 loading efficacy of as-prepared CaP@BSA NPs. In vivo therapeutic study further demonstrated the improved therapeutic effects of TSG-6 loaded CaP@BSA. Conclusions: TSG-6 was a major antifibrotic cytokine of MSCs, TSG-6 loaded CaP@BSA NPs showed specific liver accumulation and improved therapeutic effects, which indicated translational potentials of CaP@BSA as a promising drug carrier for the liver disease management.
Collapse
|
10
|
Kumar V, Dong Y, Kumar V, Almawash S, Mahato RI. The use of micelles to deliver potential hedgehog pathway inhibitor for the treatment of liver fibrosis. Am J Cancer Res 2019; 9:7537-7555. [PMID: 31695785 PMCID: PMC6831471 DOI: 10.7150/thno.38913] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/28/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: Hedgehog (Hh) pathway plays an essential role in liver fibrosis by promoting the proliferation of hepatic stellate cells (HSCs) by enhancing their metabolism via yes-associated protein 1 (YAP1). Despite the presence of several inhibitors, Hh signaling cannot be controlled exclusively due to their poor efficacy and the lack of a suitable delivery system to the injury site. Therefore, it is rationale to develop new potent Hh inhibitors and suitable delivery carriers. Methods: Based on the structure and activity of Hh inhibitor GDC-0449, we replaced its sulfonamide group with two methylpyridine-2yl at amide nitrogen to synthesize MDB5. We compared the Hh pathway inhibition and anti-fibrotic effect of MDB5 with GDC-0449 in vitro. Next, we developed MDB5 loaded micelles using our methoxy poly(ethylene glycol)-blockpoly(2-methyl-2-carboxyl-propylene carbonate-graft-dodecanol (PEG-PCC-g-DC) copolymer and characterized for physicochemical properties. We evaluated the therapeutic efficacy of MDB5 loaded micelles in common bile duct ligation (CBDL) induced liver fibrosis, mouse model. We also determined the intrahepatic distribution of fluorescently labeled micelles after MDB5 treatment. Results: Our results show that MDB5 was more potent in inhibiting Hh pathway components and HSC proliferation in vitro. We successfully developed MDB5 loaded micelles with particle size of 40 ± 10 nm and drug loading up to 10% w/w. MDB5 loaded micelles at the dose of 10 mg/kg were well tolerated by mice, without visible sign of toxicity. The serum enzyme activities elevated by CBDL was significantly decreased by MDB5 loaded micelles compared to GDC-0449 loaded micelles. MDB5 loaded micelles further decreased collagen deposition, HSC activation, and Hh activity and its target genes in the liver. MDB5 loaded micelles also prevented liver sinusoidal endothelial capillarization (LSEC) and therefore restored perfusion between blood and liver cells. Conclusions: Our study provides evidence that MDB5 was more potent in inhibiting Hh pathway in HSC-T6 cells and showed better hepatoprotection in CBDL mice compared to GDC-0449.
Collapse
|
11
|
Morsy MA, Nair AB. Prevention of rat liver fibrosis by selective targeting of hepatic stellate cells using hesperidin carriers. Int J Pharm 2018; 552:241-250. [PMID: 30291958 DOI: 10.1016/j.ijpharm.2018.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/29/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022]
|
12
|
Bariwal J, Kumar V, Dong Y, Mahato RI. Design of Hedgehog pathway inhibitors for cancer treatment. Med Res Rev 2018; 39:1137-1204. [PMID: 30484872 DOI: 10.1002/med.21555] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022]
Abstract
Hedgehog (Hh) signaling is involved in the initiation and progression of various cancers and is essential for embryonic and postnatal development. This pathway remains in the quiescent state in adult tissues but gets activated upon inflammation and injuries. Inhibition of Hh signaling pathway using natural and synthetic compounds has provided an attractive approach for treating cancer and inflammatory diseases. While the majority of Hh pathway inhibitors target the transmembrane protein Smoothened (SMO), some small molecules that target the signaling cascade downstream of SMO are of particular interest. Substantial efforts are being made to develop new molecules targeting various components of the Hh signaling pathway. Here, we have discussed the discovery of small molecules as Hh inhibitors from the diverse chemical background. Also, some of the recently identified natural products have been included as a separate section. Extensive structure-activity relationship (SAR) of each chemical class is the focus of this review. Also, clinically advanced molecules are discussed from the last 5 to 7 years. Nanomedicine-based delivery approaches for Hh pathway inhibitors are also discussed concisely.
Collapse
Affiliation(s)
- Jitender Bariwal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
13
|
Design, Synthesis and Biological Evaluation of novel Hedgehog Inhibitors for treating Pancreatic Cancer. Sci Rep 2017; 7:1665. [PMID: 28490735 PMCID: PMC5431907 DOI: 10.1038/s41598-017-01942-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
Hedgehog (Hh) pathway is involved in epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) maintenance resulting in tumor progression. GDC-0449, an inhibitor of Hh pathway component smoothened (Smo) has shown promise in the treatment of various cancers including pancreatic cancer. However, the emergence of resistance during GDC-0449 treatment with numerous side effects limits its use. Therefore, here we report the design, synthesis and evaluation of novel GDC-0449 analogs using N-[3-(2-pyridinyl) phenyl] benzamide scaffold. Cell-based screening followed by molecular simulation revealed 2-chloro-N1-[4-chloro-3-(2-pyridinyl)phenyl]-N4,N4-bis(2-pyridinylmethyl)-1,4-benzenedicarboxamide (MDB5) as most potent analog, binding with an extra interactions in seven-transmembrane (7-TM) domain of Smo due to an additional 2-pyridylmethyl group than GDC-0449. Moreover, MDB5 was more efficient in inhibiting Hh pathway components as measured by Gli-1 and Shh at transcriptional and translational levels. Additionally, a significant reduction of ALDH1, CD44 and Oct-3/4, key markers of pancreatic CSC was observed when MIA PaCa-2 cells were treated with MDB5 compared to GDC-0449. In a pancreatic tumor mouse model, MDB5 containing nanoparticles treated group showed significant inhibition of tumor growth without loss in body weight. These evidence highlight the enhanced Hh pathway inhibition and anticancer properties of MDB5 leaving a platform for mono and/or combination therapy.
Collapse
|