1
|
Panja S, Kapoor E, Siddhanta K, Jogdeo CM, Sil D, Khan RI, Kumari N, Ding L, Gendelman HE, Singh AB, Oupický D. Bioactive polymers as stimulus-responsive anti-metastatic combination agents to treat pancreatic cancer. Biomaterials 2025; 320:123255. [PMID: 40107179 DOI: 10.1016/j.biomaterials.2025.123255] [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: 09/25/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
The intractable and devastating nature of pancreatic ductal adenocarcinoma (PDAC) necessitates an urgent need for novel therapies. This study presents the development of a novel polymer prodrug system for the combination treatment of PDAC, based on an optimized pharmacologically active anti-metastatic macromolecular carrier, PCQ, conjugated with gemcitabine (GEM). Structure-activity relationship evaluations showed that random PCQ copolymers exhibited superior anti-migratory activity compared to the gradient PCQ analogs. GEM was incorporated into the random PCQ copolymers using disulfide linker to prepare a reduction-responsive prodrug, PCQ(r)6-SS-GEM12. The resultant therapeutic system presents a pharmacologically active delivery strategy that targets both the proliferative and the metastatic phenotype in PDAC. The PCQ(r)6-SS-GEM12 prodrug demonstrated a selective release of GEM under the reductive tumor environment leading to a significant inhibition of tumor growth with pronounced anti-metastatic effect. Collectively, our data show that the combination of anti-metastatic PCQ and cytotoxic GEM-based reduction-responsive prodrug polymer offers an innovative strategy to treat PDAC.
Collapse
Affiliation(s)
- Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA; Department of Pharmacology and Experimental Neuroscience, USA
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Diptesh Sil
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Rubayat I Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | | | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA.
| |
Collapse
|
2
|
Kreofsky NW, Roy P, Reineke TM. pH-Responsive Micelles Containing Quinine Functionalities Enhance Intracellular Gene Delivery and Expression. Bioconjug Chem 2024; 35:1762-1778. [PMID: 39467734 DOI: 10.1021/acs.bioconjchem.4c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Quinine is a promising building block for creating polymer carriers for intracellular nucleic acid delivery. This is due to its ability to bind to genetic material through intercalation and electrostatic interactions and the balance of hydrophobicity and hydrophilicity dependent on the pH/charge state. Yet, studies utilizing cinchona alkaloid natural products in gene delivery are limited. Herein, we present the incorporation of a quinine functionalized monomer (Q) into block polymer architectures to form self-assembled micelles for highly efficient gene delivery. Q was incorporated into the core and/or the shell of the micelles to introduce the unique advantages of quinine to the system. We found that incorporation of Q into the core of the micelle resulted in acid-induced disassembly of the micelle and a boost in transfection efficiency by promoting endosomal escape. This effect was especially evident in the cancerous cell line, A549, which has a more acidic intracellular environment. Incorporation of Q into the shell of the micelles resulted in intercalative binding to the genetic payload as well as larger micelle-DNA complexes (micelleplexes) from the hydrophobicity of Q in the shell. These factors enable the micelleplexes to be more resistant to serum and have more persistent protein expression post-transfection. Overall, this study is the first to demonstrate the benefits of including quinine functionalities into self-assembled micelles for highly efficient gene delivery and presents a platform for inclusion of other natural products with similar properties into micellar systems.
Collapse
Affiliation(s)
- Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
3
|
Lo JH, Gbur EF, Francini N, Ma J, Sorets AG, Fletcher RB, Yu F, D'Arcy R, Oltman CG, Uddin MJ, Duvall CL. Synthesis and characterization of chloroquine-modified albumin-binding siRNA-lipid conjugates for improved intracellular delivery and gene silencing in cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618042. [PMID: 39464033 PMCID: PMC11507671 DOI: 10.1101/2024.10.14.618042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
siRNA therapeutics have considerable potential as molecularly-targeted therapeutics in malignant disease, but identification of effective delivery strategies that mediate rapid intracellular delivery while minimizing toxicity has been challenging. Our group recently developed and optimized an siRNA conjugate platform termed "siRNA-L 2 ," which harnesses non-covalent association with endogenous circulating albumin to extend circulation half-life and achieve tumor-selective delivery without the use of traditional cationic lipids or polymers for transfection. To improve intracellular delivery and particularly the endosomal escape properties of siRNA-L 2 towards more efficient gene silencing, we report synthesis of siRNA-CQ-L 2 conjugates, in which chloroquine (CQ), an endosomolytic quinoline alkaloid, is covalently incorporated into the branching lipid tail structure. We accomplished this via synthesis of a novel CQ phosphoramidite, which can be incorporated into a modular siRNA-L 2 backbone using on-column solid-phase synthesis through use of asymmetric branchers with levulinyl-protected hydroxide groups that allow covalent addition of pendant CQ repeats. We demonstrate that siRNA-CQ-L 2 maintains the ability to non-covalently bind albumin, and with multiple copies of CQ, siRNA-CQ-L 2 mediates higher endosomal disruption, cellular uptake/retention, and reporter gene knockdown in cancer cells. Further, in mice, the addition of CQ did not significantly affect circulation kinetics nor organ biodistribution and did not produce hematologic or organ-level toxicity. Thus, controlled, multivalent conjugation of albumin-binding siRNA-L 2 to endosomolytic small molecule compounds holds promise in improving siRNA-L 2 knockdown potency while maintaining albumin-binding properties and overall safety.
Collapse
|
4
|
Zhou M, Liu C, Li B, Li J, Zhang P, Huang Y, Li L. Cell surface patching via CXCR4-targeted nanothreads for cancer metastasis inhibition. Nat Commun 2024; 15:2763. [PMID: 38553476 PMCID: PMC10980815 DOI: 10.1038/s41467-024-47111-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
The binding of therapeutic antagonists to their receptors often fail to translate into adequate manipulation of downstream pathways. To fix this 'bug', here we report a strategy that stitches cell surface 'patches' to promote receptor clustering, thereby synchronizing subsequent mechano-transduction. The "patches" are sewn with two interactable nanothreads. In sequence, Nanothread-1 strings together adjacent receptors while presenting decoy receptors. Nanothread-2 then targets these decoys multivalently, intertwining with Nanothread-1 into a coiled-coil supramolecular network. This stepwise actuation clusters an extensive vicinity of receptors, integrating mechano-transduction to disrupt signal transmission. When applied to antagonize chemokine receptors CXCR4 expressed in metastatic breast cancer of female mice, this strategy elicits and consolidates multiple events, including interception of metastatic cascade, reversal of immunosuppression, and potentiation of photodynamic immunotherapy, reducing the metastatic burden. Collectively, our work provides a generalizable tool to spatially rearrange cell-surface receptors to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Minglu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Chendong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Bo Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Junlin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Ping Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Kreofsky NW, Roy P, Brown ME, Perez U, Leighton RE, Frontiera RR, Reineke TM. Cinchona Alkaloid Polymers Demonstrate Highly Efficient Gene Delivery Dependent on Stereochemistry, Methoxy Substitution, and Length. Biomacromolecules 2024; 25:486-501. [PMID: 38150323 DOI: 10.1021/acs.biomac.3c01099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Nucleic acid delivery with cationic polymers is a promising alternative to expensive viral-based methods; however, it often suffers from a lower performance. Herein, we present a highly efficient delivery system based on cinchona alkaloid natural products copolymerized with 2-hydroxyethyl acrylate. Cinchona alkaloids are an attractive monomer class for gene delivery applications, given their ability to bind to DNA via both electrostatics and intercalation. To uncover the structure-activity profile of the system, four structurally similar cinchona alkaloids were incorporated into polymers: quinine, quinidine, cinchonine, and cinchonidine. These polymers differed in the chain length, the presence or absence of a pendant methoxy group, and stereochemistry, all of which were found to alter gene delivery performance and the ways in which the polymers overcome biological barriers to transfection. Longer polymers that contained the methoxy-bearing cinchona alkaloids (i.e., quinine and quinidine) were found to have the best performance. These polymers exhibited the tightest DNA binding, largest and most abundant DNA-polymer complexes, and best endosomal escape thanks to their increased buffering capacity and closest nuclear proximity of the payload. Overall, this work highlights the remarkable efficiency of polymer systems that incorporate cinchona alkaloid natural products while demonstrating the profound impact that small structural changes can have on overcoming biological hurdles associated with gene delivery.
Collapse
Affiliation(s)
- Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary E Brown
- University Imaging Centers, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ulises Perez
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ryan E Leighton
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Renee R Frontiera
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
6
|
Low LE, Kong CK, Yap WH, Siva SP, Gan SH, Siew WS, Ming LC, Lai-Foenander AS, Chang SK, Lee WL, Wu Y, Khaw KY, Ong YS, Tey BT, Singh SK, Dua K, Chellappan DK, Goh BH. Hydroxychloroquine: Key therapeutic advances and emerging nanotechnological landscape for cancer mitigation. Chem Biol Interact 2023; 386:110750. [PMID: 37839513 DOI: 10.1016/j.cbi.2023.110750] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 09/12/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Hydroxychloroquine (HCQ) is a unique class of medications that has been widely utilized for the treatment of cancer. HCQ plays a dichotomous role by inhibiting autophagy induced by the tumor microenvironment (TME). Preclinical studies support the use of HCQ for anti-cancer therapy, especially in combination with conventional anti-cancer treatments since they sensitize tumor cells to drugs, potentiating the therapeutic activity. However, clinical evidence has suggested poor outcomes for HCQ due to various obstacles, including non-specific distribution, low aqueous solubility and low bioavailability at target sites, transport across tissue barriers, and retinal toxicity. These issues are addressable via the integration of HCQ with nanotechnology to produce HCQ-conjugated nanomedicines. This review aims to discuss the pharmacodynamic, pharmacokinetic and antitumor properties of HCQ. Furthermore, the antitumor performance of the nanoformulated HCQ is also reviewed thoroughly, aiming to serve as a guide for the HCQ-based enhanced treatment of cancers. The nanoencapsulation or nanoconjugation of HCQ with nanoassemblies appears to be a promising method for reducing the toxicity and improving the antitumor efficacy of HCQ.
Collapse
Affiliation(s)
- Liang Ee Low
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia; Monash-Industry Plant Oils Research Laboratory (MIPO), Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Chee Kei Kong
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia; Department of Primary Care Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Wei-Hsum Yap
- School of Biosciences, Taylor's University, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia; Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Sangeetaprivya P Siva
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Siew Hua Gan
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Wei Sheng Siew
- School of Biosciences, Taylor's University, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Long Chiau Ming
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Sunway City, Selangor, Malaysia.
| | - Ashley Sean Lai-Foenander
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Sui Kiat Chang
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar, 31900, Perak, Malaysia.
| | - Wai-Leng Lee
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Yongjiang Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Kooi-Yeong Khaw
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Yong Sze Ong
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Beng Ti Tey
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, Uttarakhand, India.
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), 57000 Bukit Jalil, Kuala Lumpur, Malaysia.
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway City, Selangor, Malaysia.
| |
Collapse
|
7
|
Khan R, Panja S, Ding L, Tang S, Tang W, Kapoor E, Bennett RG, Oupický D. Polymeric Chloroquine as an Effective Antimigration Agent in the Treatment of Pancreatic Cancer. Mol Pharm 2022; 19:4631-4643. [PMID: 36346968 DOI: 10.1021/acs.molpharmaceut.2c00596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hydroxychloroquine (HCQ) has been the subject of multiple recent preclinical and clinical studies for its beneficial use in the combination treatments of different types of cancers. Polymeric HCQ (PCQ), a macromolecular multivalent version of HCQ, has been shown to be effective in various cancer models both in vitro and in vivo as an inhibitor of cancer cell migration and experimental lung metastasis. Here, we present detailed in vitro studies that show that low concentrations of PCQ can efficiently inhibit cancer cell migration and colony formation orders of magnitude more effectively compared to HCQ. After intraperitoneal administration of PCQ in vivo, high levels of tumor accumulation and penetration are observed, combined with strong antimetastatic activity in an orthotopic pancreatic cancer model. These studies support the idea that PCQ may be effectively used at low doses as an adjuvant in the therapy of pancreatic cancer. In conjunction with previously published literature, these studies further undergird the potential of PCQ as an anticancer agent.
Collapse
Affiliation(s)
- Rubayat Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Robert G Bennett
- Department of Internal Medicine and Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68105, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
8
|
Niemann B, Puleo A, Stout C, Markel J, Boone BA. Biologic Functions of Hydroxychloroquine in Disease: From COVID-19 to Cancer. Pharmaceutics 2022; 14:pharmaceutics14122551. [PMID: 36559044 PMCID: PMC9787624 DOI: 10.3390/pharmaceutics14122551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Abstract
Chloroquine (CQ) and Hydroxychloroquine (HCQ), initially utilized in the treatment of malaria, have now developed a long list of applications. Despite their clinical relevance, their mechanisms of action are not clearly defined. Major pathways by which these agents are proposed to function include alkalinization of lysosomes and endosomes, downregulation of C-X-C chemokine receptor type 4 (CXCR4) expression, high-mobility group box 1 protein (HMGB1) inhibition, alteration of intracellular calcium, and prevention of thrombus formation. However, there is conflicting data present in the literature. This is likely the result of the complex overlapping pathways between these mechanisms of action that have not previously been highlighted. In fact, prior research has focused on very specific portions of particular pathways without describing these in the context of the extensive CQ/HCQ literature. This review summarizes the detailed data regarding CQ/HCQ's mechanisms of action while also providing insight into the overarching themes. Furthermore, this review provides clinical context to the application of these diverse drugs including their role in malaria, autoimmune disorders, cardiovascular disease, thrombus formation, malignancies, and viral infections.
Collapse
Affiliation(s)
- Britney Niemann
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
- Correspondence: ; Tel.: +1-304-293-1254
| | - Amanda Puleo
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Conley Stout
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Justin Markel
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
9
|
Kucharski DJ, Jaszczak MK, Boratyński PJ. A Review of Modifications of Quinoline Antimalarials: Mefloquine and (hydroxy)Chloroquine. Molecules 2022; 27:1003. [PMID: 35164267 PMCID: PMC8838516 DOI: 10.3390/molecules27031003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
Late-stage modification of drug molecules is a fast method to introduce diversity into the already biologically active scaffold. A notable number of analogs of mefloquine, chloroquine, and hydroxychloroquine have been synthesized, starting from the readily available active pharmaceutical ingredient (API). In the current review, all the modifications sites and reactivity types are summarized and provide insight into the chemistry of these molecules. The approaches include the introduction of simple groups and functionalities. Coupling to other drugs, polymers, or carriers afforded hybrid compounds or conjugates with either easily hydrolyzable or more chemically inert bonds. The utility of some of the compounds was tested in antiprotozoal, antibacterial, and antiproliferative assays, as well as in enantiodifferentiation experiments.
Collapse
Affiliation(s)
| | | | - Przemysław J. Boratyński
- Department of Organic and Medicinal Chemistry, Wrocław University of Technology, Wyspiańskiego 27, 50-370 Wrocław, Poland; (D.J.K.); (M.K.J.)
| |
Collapse
|
10
|
Dong J, Zhu C, Zhang F, Zhou Z, Sun M. "Attractive/adhesion force" dual-regulatory nanogels capable of CXCR4 antagonism and autophagy inhibition for the treatment of metastatic breast cancer. J Control Release 2021; 341:892-903. [PMID: 34953982 DOI: 10.1016/j.jconrel.2021.12.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/11/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023]
Abstract
Metastasis is refractory systemic disease resulting in low survival rate of breast cancer patients, especially in the late stage. The processes of metastasis are mainly initiated by strong "attractive force" from distant organs and deteriorated by weak "adhesion force" in primary tumor. Here, we reported "attractive/adhesion force" dual-regulatory nanogels (CQ-HF/PTX) for the precise treatment of both primary and metastasis of metastatic breast cancer. Hydroxychloroquine (HCQ) and hydrophobic Fmoc were grafted on hydrophilic hydroxyethyl starch (HES) to obtain amphiphilic CQ-HF polymer, which was assembly with chemotherapy drug paclitaxel (PTX) to form the nanogels for anti-primary tumor. Meanwhile, CQ-HF/PTX nanogels play two roles in anti-metastasis: i) For reducing the "attractive force", it could block the CXCR4/SDF-1 pathway, preventing tumor cells metastasis to the lung; ii) For reinforcing "adhesion force", it could inhibit the excessive autophagy for hindering the degradation of paxillin and enhancing the cell adhesion. As a result, dual-regulatory CQ-HF/PTX nanogels dramatically inhibited tumor and the lung metastasis of mouse breast cancer. Therefore, the fabricating of synergetic dual-regulatory nanogels uncovered the explicit mechanism and provided an efficient strategy for combating malignant metastatic tumors.
Collapse
Affiliation(s)
- Jingwen Dong
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural, Department of Pharmaceutics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Chenfei Zhu
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural, Department of Pharmaceutics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Feiran Zhang
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural, Department of Pharmaceutics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Zhanwei Zhou
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural, Department of Pharmaceutics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Minjie Sun
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural, Department of Pharmaceutics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
11
|
Wang J, Fan Q, Yu T, Zhang Y. Identifying the hub genes and immune cell infiltration in synovial tissue between osteoarthritic and rheumatoid arthritic patients by bioinformatic approach. Curr Pharm Des 2021; 28:497-509. [PMID: 34736376 DOI: 10.2174/1381612827666211104154459] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Osteoarthritis (OA) and rheumatoid arthritis (RA) are two common diseases that result in limb disability and a decrease in quality of life. The major symptoms of OA and RA are pain, swelling, stiffness, and malformation of joints, and each disease also has unique characteristics. OBJECTIVE To compare the pathological mechanisms of OA and RA via weighted correlation network analysis (WGCNA) and immune infiltration analysis and find potential diagnostic and pharmaceutical targets for the treatment of OA and RA. METHODS The gene expression profiles of ten OA and ten RA synovial tissue samples were downloaded from the Gene Expression Omnibus (GEO) database (GSE55235). After obtaining differentially expressed genes (DEGs) via GEO2R, WGCNA was conducted using an R package, and modules and genes that were highly correlated with OA and RA were identified. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and protein-protein interaction (PPI) network analyses were also conducted. Hub genes were identified using the Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape software. Immune infiltration analysis was conducted using the Perl program and CIBERSORT software. RESULTS Two hundred ninety-nine DEGs, 24 modules, 16 GO enrichment terms, 6 KEGG pathway enrichment terms, 10 hub genes (CXCL9, CXCL10, CXCR4, CD27, CD69, CD3D, IL7R, STAT1, RGS1, and ISG20), and 8 kinds of different infiltrating immune cells (plasma cells, CD8 T cells, activated memory CD4 T cells, T helper follicular cells, M1 macrophages, Tregs, resting mast cells, and neutrophils) were found to be involved in the different pathological mechanisms of OA and RA. CONCLUSION Inflammation-associated genes were the top differentially expressed hub genes between OA and RA, and their expression was downregulated in OA. Genes associated with lipid metabolism may have upregulated expression in OA. In addition, immune cells that participate in the adaptive immune response play an important role in RA. OA mainly involves immune cells that are associated with the innate immune response.
Collapse
Affiliation(s)
- Junjie Wang
- Qingdao University, Qingdao, Shandong 266000. China
| | - Qin Fan
- Qingdao University, Qingdao, Shandong 266000. China
| | - Tengbo Yu
- Qingdao University, Qingdao, Shandong 266000. China
| | - Yingze Zhang
- Qingdao University, Qingdao, Shandong 266000. China
| |
Collapse
|
12
|
Liang T, Qiang T, Ren L, Wang B, Hu W. An ultrasensitive polarity-specific two-photon probe for revealing autophagy in live cells during scrap leather-induced neuroinflammation process. Analyst 2021; 146:4659-4665. [PMID: 34190222 DOI: 10.1039/d1an00667c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A polarity-sensitive fluorescence probe AMN was developed to demonstrate the role of autophagy inhibitory drugs in the process of leather residue-induced neuroinflammation, promoting the knowledge of the relationship between autophagy and neuroinflammation. AMN showed a turn-on fluorescent signal in the process of autophagy inhibition via two-photon confocal imaging, which is different from the current popular autophagy probes. Therefore, AMN can offer high-sensitive imaging analysis of the autophagy inhibition process to better understand the role of autophagy in the process of neuroinflammation. The model of scrap leather-induced neuroinflammation using PC12 cells demonstrated that neuroinflammation can induce autophagy by releasing reactive oxygen species (ROS), and autophagy can alleviate neuroinflammation significantly via ROS scavenging.
Collapse
Affiliation(s)
- Tianyu Liang
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Taotao Qiang
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China. and Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Longfang Ren
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Baoshuai Wang
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Wei Hu
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China. and Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| |
Collapse
|
13
|
Zhou M, Luo C, Zhou Z, Li L, Huang Y. Improving anti-PD-L1 therapy in triple negative breast cancer by polymer-enhanced immunogenic cell death and CXCR4 blockade. J Control Release 2021; 334:248-262. [PMID: 33915224 DOI: 10.1016/j.jconrel.2021.04.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/18/2022]
Abstract
Triple negative breast cancer (TNBC) with highly metastatic features generally does not respond to anti-programmed cell death 1 ligand 1 (PD-L1) therapy due to multiple immunosuppressive mechanisms to exclude and disable T cells. Here, we develop a polymer-based combinatory approach consisting of both immunogenic cell death (ICD)-inducing and CXCR4-inhibiting function to prime tumor microenvironment and improve anti-PD-L1 therapy in TNBC. Our findings revealed that the combination therapy was able to spur the T cell response in primary tumors by increasing the tumor immunogenicity to recruit T cells, removing the physiological barriers of intratumoral fibrosis and collagen to increase T cell infiltration, and reducing the immunosuppressive cells to revive T cells. Meanwhile, such approach efficiently inhibited the formation of pre-metastatic niche in abscopal lung. Because of the significant promotion of anti-tumor and anti-metastasis immunity, the non-responding TNBC gained robust responsiveness to anti-PD-L1 therapy which resulted in complete eradication of orthotopic tumors, inhibition of pulmonary metastasis, and durable memory effects against tumor recurrence. Our work provided a generalizable approach of simultaneous ICD induction and CXCR4 blockade to apply anti-PD-L1 therapy in TNBC.
Collapse
Affiliation(s)
- Minglu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chaohui Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhou Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Liu W, Jing X, Xu Z, Teng C. PEGDA/HA mineralized hydrogel loaded with Exendin4 promotes bone regeneration in rat models with bone defects by inducing osteogenesis. J Biomater Appl 2021; 35:1337-1346. [PMID: 33467965 DOI: 10.1177/0885328220987046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Scaffolds with osteogenic differentiation function play an important role in the healing process of bone defects. Here, we designed a high strength Poly(ethyleneglycol) diacrylate/Hydroxyapatite (PEGDA/HA) mineralized hydrogel loaded with Exendin4 for inducing osteogenic differentiation. In this study, PEGDA hydrogel was prepared by photo initiating method. PEGDA/HA mineralized hydrogel was prepared by in-situ precipitation method, and Exendin4 was loaded by gel adsorption. The effects of different calcium and phosphorus concentrations on the strength and Exendin4 release of PEGDA/HA hydrogels were investigated. Rat models of bone defect were made and randomly divided into 5 groups. The experimental group was implanted with PEGDA hydrogel, Exendin4-PEGDA hydrogel, PEGDA/HA mineralized hydrogel, Exendin4-PEGDA/HA mineralized hydrogel, and no materials were implanted in the blank control group. Computed tomography (CT) and histology were observed 4 and 8 weeks after operation. Our results revealed that the PEGDA/HA mineralized hydrogel had porous structure, high mechanical strength and good biocompatibility. In vitro release test showed that the mineralized hydrogel exhibited good sustained release profile within 20 d. The animal experiments showed that the mineralized hydrogel accelerated the formation of new bone after 4 and 8 weeks, and formed a seamless union on the defected bone area after 8 weeks. In conclusions, The Exendin4-PEGDA/HA mineralized hydrogel can effectively repair bone defects in rats, and it is expected to be used as a biomaterial for human bone tissue repair.
Collapse
Affiliation(s)
- Wei Liu
- The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Xiaowei Jing
- The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Zhiwen Xu
- The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Chong Teng
- The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
15
|
Van Bruggen C, Punihaole D, Keith AR, Schmitz AJ, Tolar J, Frontiera RR, Reineke TM. Quinine copolymer reporters promote efficient intracellular DNA delivery and illuminate a protein-induced unpackaging mechanism. Proc Natl Acad Sci U S A 2020; 117:32919-32928. [PMID: 33318196 PMCID: PMC7777095 DOI: 10.1073/pnas.2016860117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Polymeric vehicles that efficiently package and controllably release nucleic acids enable the development of safer and more efficacious strategies in genetic and polynucleotide therapies. Developing delivery platforms that endogenously monitor the molecular interactions, which facilitate binding and release of nucleic acids in cells, would aid in the rational design of more effective vectors for clinical applications. Here, we report the facile synthesis of a copolymer containing quinine and 2-hydroxyethyl acrylate that effectively compacts plasmid DNA (pDNA) through electrostatic binding and intercalation. This polymer system poly(quinine-co-HEA) packages pDNA and shows exceptional cellular internalization, transgene expression, and low cytotoxicity compared to commercial controls for several human cell lines, including HeLa, HEK 293T, K562, and keratinocytes (N/TERTs). Using quinine as an endogenous reporter for pDNA intercalation, Raman imaging revealed that proteins inside cells facilitate the unpackaging of polymer-DNA complexes (polyplexes) and the release of their cargo. Our work showcases the ability of this quinine copolymer reporter to not only facilitate effective gene delivery but also enable diagnostic monitoring of polymer-pDNA binding interactions on the molecular scale via Raman imaging. The use of Raman chemical imaging in the field of gene delivery yields unprecedented insight into the unpackaging behavior of polyplexes in cells and provides a methodology to assess and design more efficient delivery vehicles for gene-based therapies.
Collapse
Affiliation(s)
- Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455
| | - David Punihaole
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455
| | - Allison R Keith
- Department of Pediatrics, Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Andrew J Schmitz
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455
| | - Jakub Tolar
- Department of Pediatrics, Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Renee R Frontiera
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455;
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455;
| |
Collapse
|
16
|
Peng L, Zhu N, Mao J, Huang L, Yang Y, Zhou Z, Wang L, Wu B. Expression levels of CXCR4 and CXCL12 in patients with rheumatoid arthritis and its correlation with disease activity. Exp Ther Med 2020; 20:1925-1934. [PMID: 32782501 PMCID: PMC7401245 DOI: 10.3892/etm.2020.8950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to investigate the expression levels of C-X-C motif chemokine receptor 4 (CXCR4) and CXC ligand 12 (CXCL12) in patients with rheumatoid arthritis (RA) and the correlation with disease activity. In total, 60 patients with RA were selected as the study group, comprising of 28 patients in active-stage and 32 patients in remission-stage. In addition, 60 patients with osteoarthritis were selected as the control group. Western blotting and ELISA were used to detect the expression of CXCR4 and CXCL12, respectively. The Spearman's correlation test was used to analyze correlations between CXCR4 and CXCL12, and erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), disease activity score 28 (DAS28) scores and rheumatoid factor (RF). The present results suggested that CXCR4 and CXCL12 expression levels in the serum and joint synovial fluid of the study group were significantly higher compared with the control group (P<0.05). Moreover, CXCR4 and CXCL12 expression levels in the RA-active group were higher compared with the remission (P<0.05) and control groups (P<0.01). The Pearson test results suggested that the expression levels of CXCR4 and CXCL12 in the serum and joint synovial fluid of patients with RA had a positive correlation with the ESR, CRP, RF and DAS28 scores (P<0.05). CXCL12 and CXCR4 were highly expressed in the serum and joint synovial fluid of patients with RA, and these expression levels were positively correlated with ESR, CRP, RF and DAS28 scores. Therefore, these clinical parameters may be used as indicators to evaluate the disease activity of patients with RA.
Collapse
Affiliation(s)
- Liping Peng
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Ning Zhu
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jing Mao
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Li Huang
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Yameng Yang
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Zhengju Zhou
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Li Wang
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Bin Wu
- Department of Rheumatology and Immunology, First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
17
|
Jiao F, Sun H, Yang Q, Sun H, Wang Z, Liu M, Chen J. Association of CXCL13 and Immune Cell Infiltration Signature in Clear Cell Renal Cell Carcinoma. Int J Med Sci 2020; 17:1610-1624. [PMID: 32669964 PMCID: PMC7359384 DOI: 10.7150/ijms.46874] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/02/2020] [Indexed: 01/05/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most commonly diagnosed kidney tumors and is often accompanied by immune cell infiltration. In this study, we attempted to identify microenvironment-associated genes and explore the correlation between CXCL13 and tumor-infiltrating immune cells (TIICs). Gene expression profiles and their corresponding clinical information were downloaded from The Cancer Genome Atlas (TCGA) database. The ESTIMATE (Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data) algorithm was used to calculate immune cell and stromal cell scores, according to which patients were divided into high- and low-score groups, allowing differentially expressed genes (DEGs) to be identified. Functional enrichment and PPI network analysis were used to identify the functions of the DEGs. CIBERSORT algorithm and TIMER analysis were used to evaluate the immune score. Oncomine and TCGA database were used to explore CXCL13 mRNA expression level in ccRCC. High ESTIMATE score was significantly associated with prognosis. Functional enrichment analysis clarified that DEGs were associated with T cell activation, immune response-regulating cell surface receptor signaling pathway, and positive regulation of cytokine production. PPI network was used to identify CXCL13 as a hub gene. And CIBERSORT algorithm and TIMER analysis showed that strong correlation between CXCL13 expression level and TIICs. Oncomine database was used to validate high CXCL13 expression level in ccRCC tissue, compared to normal tissues. In conclusion, we obtained a list of tumor microenvironment-related genes and identified CXCL13 as an immune response biomarker in patients with ccRCC, GSEA analysis, wound healing and transwell assays showed CXCL13 played a role in tumor migration.
Collapse
Affiliation(s)
- Fangdong Jiao
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Hao Sun
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Qingya Yang
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Hui Sun
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Zehua Wang
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Ming Liu
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Jun Chen
- Department of Urology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, China
| |
Collapse
|
18
|
Yu F, Wang Y, Hang Y, Tang W, Zhao Z, Oupický D. Synthesis and biological characterization of clicked chloroquine copolymers as macromolecular inhibitors of cancer cell migration. ACTA ACUST UNITED AC 2019. [DOI: 10.1002/pola.29512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha Nebraska 68198
| | - Yazhe Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha Nebraska 68198
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha Nebraska 68198
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha Nebraska 68198
| | - Zhifeng Zhao
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha Nebraska 68198
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha Nebraska 68198
| |
Collapse
|
19
|
Chen Y, Tang W, Yu F, Xie Y, Jaramillo L, Jang HS, Li J, Padanilam BJ, Oupický D. Determinants of preferential renal accumulation of synthetic polymers in acute kidney injury. Int J Pharm 2019; 568:118555. [PMID: 31344445 PMCID: PMC6708481 DOI: 10.1016/j.ijpharm.2019.118555] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/17/2019] [Accepted: 07/21/2019] [Indexed: 12/17/2022]
Abstract
Acute kidney injury (AKI) is a major kidney disease associated with high mortality and morbidity. AKI may lead to chronic kidney disease and end-stage renal disease. Currently, the management of AKI is mainly focused on supportive treatments. Previous studies showed macromolecular delivery systems as a promising method to target AKI, but little is known about how physicochemical properties affect the renal accumulation of polymers in ischemia-reperfusion AKI. In this study, a panel of fluorescently labeled polymers with a range of molecular weights and net charge was synthesized by living radical polymerization. By testing biodistribution of the polymers in unilateral ischemia-reperfusion mouse model of AKI, the results showed that negatively charged and neutral polymers had the greatest potential for selectively accumulating in I/R kidneys. The polymers passed through glomerulus and were retained in proximal tubular cells for up to 24 h after injection. The results obtained in the unilateral model were validated in a bilateral ischemic-reperfusion model. This study demonstrates for the first time that polymers with specific physicochemical characteristics exhibit promising ability to accumulate in the injured AKI kidney, providing initial insights on their use as polymeric drug delivery systems in AKI.
Collapse
Affiliation(s)
- Yi Chen
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lee Jaramillo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
20
|
Bu HL, Xia YZ, Liu PM, Guo HM, Yuan C, Fan XC, Huang C, Wen YY, Kong CL, Wang T, Ma LT, Li XX, Zhang HW, Zhang LR, Ma MY, Ai YQ, Zhang W. The Roles of Chemokine CXCL13 in the Development of Bone Cancer Pain and the Regulation of Morphine Analgesia in Rats. Neuroscience 2019; 406:62-72. [PMID: 30826523 DOI: 10.1016/j.neuroscience.2019.02.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/04/2019] [Accepted: 02/18/2019] [Indexed: 11/16/2022]
Abstract
Chemokines are important regulators of immune, inflammatory, and neuronal responses in peripheral and central pain pathway. The aim of this study was to investigate whether chemokine (C-X-C motif) ligand 13 (CXCL13) and its receptor (C-X-C chemokine receptor type 5, CXCR5) involve in the development of bone cancer pain (BCP) and the regulation of morphine analgesia in rats. The change of pain behaviors in BCP rats were measured by testing paw withdrawal threshold (PWT). The levels of CXCL13, CXCR5 and signal pathway proteins (p-p38, p-ERK and p-AKT etc.) in the spinal cord were measured via western blots. The expression of CXCL13 and CXCR5 in spinal cord was increased in BCP rats. The BCP rats showed decrease of PWTs, which was relieved by CXCR5i. Intrathecally injection of murine recombinant CXCL13 (mrCXCL13) decreased the PWTs of BCP rats and opposed morphine-induced analgesia in BCP rats. In BCP rats, the signal pathway proteins (p38, ERK and AKT) in the spinal cord were activated. CXCL13 and morphine had contrary effect on the phosphorylation of these proteins. MrCXCL13 directly increased the levels of p-p38, p-ERK and p-AKT in BCP rats. However, morphine decreased the levels of these proteins in BCP rats. While blocking the activation of p-p38, p-ERK and p-AKT, morphine analgesia was enhanced. These results suggest CXCL13 participated in bone cancer pain and opposed morphine analgesia via p38, ERK and AKT pathways. It may be a target to enhance pain management in cancer pain patients.
Collapse
Affiliation(s)
- Hui-Lian Bu
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yu-Zhong Xia
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Pan-Mei Liu
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hai-Ming Guo
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chang Yuan
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiao-Chong Fan
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chen Huang
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuan-Yuan Wen
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Cun-Long Kong
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tao Wang
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Le-Tian Ma
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin-Xin Li
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hong-Wei Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Li-Rong Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Min-Yu Ma
- Department of Pain management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Yan-Qiu Ai
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Wei Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
21
|
Yin S, Xia C, Wang Y, Wan D, Rao J, Tang X, Wei J, Wang X, Li M, Zhang Z, Liu J, He Q. Dual receptor recognizing liposomes containing paclitaxel and hydroxychloroquine for primary and metastatic melanoma treatment via autophagy-dependent and independent pathways. J Control Release 2018; 288:148-160. [PMID: 30099017 DOI: 10.1016/j.jconrel.2018.08.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 12/23/2022]
Abstract
Autophagy acts as a cytoprotective mechanism for malignant tumors, thus maintaining the survival and promoting proliferation and metastasis of malignant tumors. Recent studies have showed that autophagy inhibitors can enhance the chemotherapeutic efficacy of anti-tumor growth. However, the antimetastasis effects and the possible mechanisms of chemotherapeutics combined with autophagy inhibitors have not been thoroughly explored. Here, we prepared R8-dGR peptide modified paclitaxel (PTX) and hydroxychloroquine (HCQ) co-loaded liposomes (PTX/HCQ-R8-dGR-Lip) for enhanced delivery by recognizing integrin αvβ3 receptors and neuropilin-1 receptors on B16F10 melanoma cells. Our results showed that R8-dGR modified liposomes (R8-dGR-Lip) enhanced tumor-targeting delivery in vitro and in vivo. Besides, PTX/HCQ-R8-dGR-Lip exhibited the optimum inhibitory effects on migration, invasion and anoikis resistance of B16F10 cells in vitro, and showed enhanced efficiency on inhibiting primary tumor growth and reducing lung metastasis in vivo. Meanwhile, the antimetastasis mechanism studies confirmed that the combination of the chemotherapeutic PTX and the autophagy inhibitor HCQ further suppressed the degradation of paxillin, the expression of MMP9 and MMP2. Moreover, HCQ disturbed the CXCR4/CXCL12 axis which could induce invasion and metastasis of malignant melanoma in an autophagy-independent way.
Collapse
Affiliation(s)
- Sheng Yin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Chunyu Xia
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yashi Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Dandan Wan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Jingdong Rao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Xian Tang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Jiaojie Wei
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Xuhui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Ji Liu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China.
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China.
| |
Collapse
|
22
|
Xie Y, Wang Y, Li J, Hang Y, Oupický D. Promise of chemokine network-targeted nanoparticles in combination nucleic acid therapies of metastatic cancer. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1528. [PMID: 29700990 DOI: 10.1002/wnan.1528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/28/2018] [Accepted: 04/01/2018] [Indexed: 01/10/2023]
Abstract
Chemokines and chemokine receptors play key roles in cancer progression and metastasis. Although multiple chemokines and chemokine receptors have been investigated, inhibition of CXCR4 emerged as one of the most promising approaches in combination cancer therapy, especially when focused on the metastatic disease. Small RNA molecules, such as small interfering RNA (siRNA) and microRNA (miRNA), represent new class of therapeutics for cancer treatment through RNA interference-mediated gene silencing. However, the clinical applicability of siRNA and miRNA is severely limited by the lack of effective delivery systems. There is a significant therapeutic potential for CXCR4-targeted nanomedicines in combination with the delivery of siRNA and miRNA in cancer. Recently developed CXCR4-targeted polymeric drugs and nanomedicines, including cyclam- and chloroquine-based polymeric CXCR4 antagonists are introduced here and their ability to deliver functional siRNA and miRNA is discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Ying Xie
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yazhe Wang
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jing Li
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yu Hang
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - David Oupický
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
23
|
Xie Y, Yu F, Tang W, Alade B, Peng ZH, Wang Y, Li J, Oupický D. Synthesis and Evaluation of Chloroquine-Containing DMAEMA Copolymers as Efficient Anti-miRNA Delivery Vectors with Improved Endosomal Escape and Antimigratory Activity in Cancer Cells. Macromol Biosci 2018; 18:10.1002/mabi.201700194. [PMID: 28776937 PMCID: PMC5997184 DOI: 10.1002/mabi.201700194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/11/2017] [Indexed: 12/19/2022]
Abstract
Chloroquine-containing 2-(dimethylamino)ethyl methacrylate copolymers (PDCs) are synthesized by reversible addition-fragmentation chain-transfer polymerization. Systematic evaluation is performed to test the hypothesis that presence of chloroquine (CQ) in the PDC structure will improve miRNA delivery due to enhanced endosomal escape while simultaneously contribute to anticancer activity of PDC/miRNA polyplexes through inhibition of cancer cell migration. The results show that miRNA delivery efficiency is dependent both on the molecular weight and CQ. The best performing PDC/miRNA polyplexes show effective endosomal escape of miRNA. PDC polyplexes with therapeutic miR-210 show promising anticancer activity in human breast cancer cells. PDC/miRNA polyplexes show excellent ability to inhibit migration of cancer cells. Overall, this study supports the use of PDC as a promising polymeric drug platform for use in combination anti-metastatic and anticancer miRNA therapeutic strategies.
Collapse
Affiliation(s)
- Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bolutito Alade
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zheng-Hong Peng
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yazhe Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
24
|
Sleightholm R, Yang B, Yu F, Xie Y, Oupický D. Chloroquine-Modified Hydroxyethyl Starch as a Polymeric Drug for Cancer Therapy. Biomacromolecules 2017; 18:2247-2257. [PMID: 28708385 PMCID: PMC5996760 DOI: 10.1021/acs.biomac.7b00023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hydroxyethyl starch (HES) is a clinically used polysaccharide colloidal plasma volume expander. The goal of this study was to synthesize HES modified with hydroxychloroquine (HCQ) as a novel polymeric drug with the ability to inhibit the invasive character of pancreatic cancer (PC) cells. HES was conjugated with HCQ using a simple carbonyldiimidazole coupling to prepare Chloroquine-modified HES (CQ-HES). CQ-HES with various degrees of HCQ substitution were synthesized and characterized. Atomic force microscopy was used to demonstrate a pH-dependent assembly of CQ-HES into well-defined nanoparticles. In vitro studies in multiple PC cell lines showed CQ-HES to have a similar toxicity profile as HCQ. Confocal microscopy revealed the propensity of CQ-HES to localize to lysosomes and mechanistic studies confirmed the ability of CQ-HES to inhibit autophagy in PC cells. Further studies demonstrated a greatly enhanced ability of CQ-HES to inhibit the migration and invasion of PC cells when compared with HCQ. The enhanced inhibitory actions of CQ-HES compared to HCQ appeared to arise in part from the increased inhibition of ERK and Akt phosphorylation. We found no significant HCQ release from CQ-HES, which confirmed that the observed activity was due to the action of CQ-HES as a polymeric drug. Due to its promising ability to block cancer cell invasion and the ability to form nanoparticles, CQ-HES has the potential as a drug delivery platform suitable for future development with chemotherapeutics to establish novel antimetastatic treatments.
Collapse
Affiliation(s)
- Richard Sleightholm
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Bin Yang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
25
|
Yu F, Li J, Xie Y, Sleightholm RL, Oupický D. Polymeric chloroquine as an inhibitor of cancer cell migration and experimental lung metastasis. J Control Release 2016; 244:347-356. [PMID: 27473763 DOI: 10.1016/j.jconrel.2016.07.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 01/16/2023]
Abstract
Chloroquine (CQ) is a widely used antimalarial drug with emerging potential in anticancer therapies due to its apparent inhibitory effects on CXCR4 chemokine receptor, autophagy, and cholesterol metabolism. This study reports on polymeric CQ (pCQ) as a macromolecular drug with antimetastatic activity. The pCQ polymers were synthesized by copolymerization of methacryloylated hydroxy-CQ (HCQ) and N-(2-hydroxypropyl)methacrylamide (HPMA). The results show that pCQ is significantly more effective in inhibiting cancer cell migration and invasion when compared with the parent HCQ. The proposed mechanism of action at least partially relies on the ability of pCQ to inhibit cell migration mediated by the CXCR4/CXCL12 pathway. The pCQ also demonstrates superior inhibitory activity over HCQ when tested in a mouse model of experimental lung metastasis. Lastly, pCQ shows the ability to efficiently translocate to the cytoplasm while exhibiting lower cytotoxicity than HCQ. Overall, this study supports pCQ as a promising polymeric drug platform suitable for use in combination antimetastatic strategies and potential use in cytoplasmic drug delivery.
Collapse
Affiliation(s)
- Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Richard L Sleightholm
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|