1
|
Kapoor DU, Pareek A, Sharma M, Prajapati BG, Suttiruengwong S, Sriamornsak P. Exploring starch-based excipients in pharmaceutical formulations: Versatile applications and future perspectives. Eur J Pharm Biopharm 2025:114727. [PMID: 40286879 DOI: 10.1016/j.ejpb.2025.114727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/14/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Starch, a naturally abundant and biocompatible polysaccharide, serves as a key excipient in pharmaceutical formulations, enhancing drug stability, efficacy, and manufacturability. This review explores the properties, modifications, and diverse applications of starch-based excipients. Native starches from corn, potato, rice, and wheat are commonly used as disintegrants, binders, and fillers. Physical (e.g., pre-gelatinization), chemical (e.g., cross-linking, acetylation), and enzymatic modifications improve their functionality, such as enhanced stability and colon-specific drug delivery. Starch excels as a binder, improving tablet cohesion and strength, and as a disintegrant, promoting rapid drug release. It also supports controlled and sustained-release systems and advanced drug delivery methods, like nanoparticles and microparticles. Compared to other natural and synthetic excipients, starch offers advantages in biodegradability, non-toxicity, and cost-effectiveness, despite challenges like stability and batch variability. Innovations such as starch nanocrystals show promise in boosting drug solubility and bioavailability. Looking ahead, starch-based excipients hold potential for sustainable pharmaceutical development, personalized medicine, and 3D printing.
Collapse
Affiliation(s)
- Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Bardoli, Gujarat 394601, India
| | - Anil Pareek
- Department of Pharmaceutics, Lachoo Memorial College of Science and Technology, Jodhpur, Rajasthan 342001, India
| | - Mayank Sharma
- School of Pharmacy and Technology Management, NMIMS, Shirpur, Dhule, Maharashtra 425405, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, Gujarat, India,; Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| | - Supakij Suttiruengwong
- Sustainable Materials Laboratory, Department of Materials Science and Engineering, Faculty of Engineering and Industrial Technology, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Pornsak Sriamornsak
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Academy of Science, The Royal Society of Thailand, Bangkok 10300, Thailand.
| |
Collapse
|
2
|
Bai X, Xiong J, Li L, Yu C, Sun C. Suppression of hypoxia-induced CAV1 autophagic degradation enhances nanoalbumin-paclitaxel transcytosis and improves therapeutic activity in pancreatic cancer. Eur J Pharmacol 2024; 969:176431. [PMID: 38395374 DOI: 10.1016/j.ejphar.2024.176431] [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: 10/29/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024]
Abstract
Nanoalbumin-paclitaxel (nab-paclitaxel) is a standard chemotherapy for pancreatic cancer but has shown limited efficacy. However, the mechanism through which circulating nab-paclitaxel passes through the tumour vascular endothelium has not been determined. In our study, a new nonradioactive and highly sensitive method for analysing nab-paclitaxel transcytosis was established. Based on these methods, we found that hypoxia significantly enhanced the autophagic degradation of CAV1 and therefore attenuated caveolae-mediated nab-paclitaxel transcytosis across endothelial cells (ECs). In a proof-of-concept experiment, higher levels of CAV1, accompanied by lower levels of LC3B, were observed in the vascular endothelium of pancreatic cancer tissues collected from patients who showed a good response to nab-paclitaxel compared with those from patients who showed a poor response to nab-paclitaxel. Furthermore, both in vivo and in vitro studies confirmed that suppressing the autophagic degradation of CAV1 via EC-specific ATG5 knockdown or hydroxychloroquine sulfate (HCQ) treatment significantly enhanced nab-paclitaxel translocation across the endothelial barrier into pancreatic cancer cells and amplified the inhibitory effect of nab-paclitaxel on pancreatic tumour growth. The stimulation of CAV1 expression by EC-specific overexpression of exogenous CAV1 or administration of gemcitabine hydrochloride (GE) had the same effect. These results demonstrated that suppressing CAV1 autophagic degradation is a novel translatable strategy for enhancing nab-paclitaxel chemotherapeutic activity in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Xiangli Bai
- School of Basic Medicine, Guizhou Medical University, 5500025, Guiyang, Guizhou, China; Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China; Department of Laboratory Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430077, Wuhan, Hubei, China
| | - Jia Xiong
- Department of Cardiovascular Surgery, Jinan University 2nd Clinical Medicine College People's Hospital of Shenzhen, 518020, Shenzhen, Guangdong, China
| | - Lin Li
- School of Basic Medicine, Guizhou Medical University, 5500025, Guiyang, Guizhou, China; Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Chao Yu
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Chengyi Sun
- School of Basic Medicine, Guizhou Medical University, 5500025, Guiyang, Guizhou, China; Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China.
| |
Collapse
|
3
|
Pei J, Yan Y, Jayaraman S, Rajagopal P, Natarajan PM, Umapathy VR, Gopathy S, Roy JR, Sadagopan JC, Thalamati D, Palanisamy CP, Mironescu M. A review on advancements in the application of starch-based nanomaterials in biomedicine: Precision drug delivery and cancer therapy. Int J Biol Macromol 2024; 265:130746. [PMID: 38467219 DOI: 10.1016/j.ijbiomac.2024.130746] [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: 10/07/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
The burgeoning field of starch-based nanomaterials in biomedical applications has perceived notable progressions, with a particular emphasis on their pivotal role in precision drug delivery and the inhibition of tumor growth. The complicated challenges in current biomedical research require innovative approaches for improved therapeutic outcomes, prompting an exploration into the possible of starch-based nanomaterials. The conceptualization of this review emerged from recognizing the need for a comprehensive examination of the structural attributes, versatile properties, and mechanisms underlying the efficiency of starch-based nanomaterials in inhibiting tumor growth and enabling targeted drug delivery. This review delineates the substantial growth in utilizing starch-based nanomaterials, elucidating their small size, high surface-volume ratio, and biocompatibility, predominantly emphasizing their possible to actively recognize cancer cells, deliver anticancer drugs, and combat tumors efficiently. The investigation of these nanomaterials encompasses to improving biocompatibility and targeting specific tissues, thereby contributing to the evolving landscape of precision medicine. The review accomplishes by highlighting the auspicious strategies and modern developments in the field, envisioning a future where starch-based nanomaterials play a transformative role in molecular nanomaterials, evolving biomedical sciences. The translation of these advancements into clinical applications holds the potential to revolutionize targeted drug delivery and expand therapeutic outcomes in the realm of precision medicine.
Collapse
Affiliation(s)
- JinJin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - Yuqiang Yan
- Department of anaesthesia, Xi'an Central Hospital, No. 161, West 5th Road, Xincheng District, Xi'an 710003, China
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Ponnulakshmi Rajagopal
- Central Research Laboratory, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai-600 095, India
| | - Prabhu Manickam Natarajan
- Department of Clinical Sciences, Center of Medical and Bio-allied Health Sciences and Research, College of Dentistry, Ajman University, Ajman, United Arab Emirates
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Thai Moogambigai Dental College and Hospital, Chennai-600107, India
| | - Sridevi Gopathy
- Department of Physiology, SRM Dental College, Ramapuram campus, Chennai 600089, India
| | - Jeane Rebecca Roy
- Department of Anatomy, Bhaarath Medical College and hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai, Tamil Nadu 600 073, India
| | - Janaki Coimbatore Sadagopan
- Department of Anatomy, Bhaarath Medical College and hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai, Tamil Nadu 600 073, India
| | | | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Monica Mironescu
- Faculty of Agricultural Sciences Food Industry and Environmental Protection, Lucian Blaga University of Sibiu, Sibiu 550024, Romania.
| |
Collapse
|
4
|
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
|
5
|
Bietsch J, Baker L, Duffney A, Mao A, Foutz M, Ackermann C, Wang G. Para-Methoxybenzylidene Acetal-Protected D-Glucosamine Derivatives as pH-Responsive Gelators and Their Applications for Drug Delivery. Gels 2023; 9:445. [PMID: 37367116 DOI: 10.3390/gels9060445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Carbohydrate-based low molecular weight gelators (LMWGs) are compounds with the capability to self-assemble into complex molecular networks within a solvent, leading to solvent immobilization. This process of gel formation depends on noncovalent interactions, including Van der Waals, hydrogen bonding, and π-π stacking. Due to their potential applications in environmental remediation, drug delivery, and tissue engineering, these molecules have emerged as an important area of research. In particular, various 4,6-O-benzylidene acetal-protected D-glucosamine derivatives have shown promising gelation abilities. In this study, a series of C-2-carbamate derivatives containing a para-methoxy benzylidene acetal functional group were synthesized and characterized. These compounds exhibited good gelation properties in several organic solvents and aqueous mixtures. Upon removal of the acetal functional group under acidic conditions, a number of deprotected free sugar derivatives were also synthesized. Analysis of these free sugar derivatives revealed two compounds were hydrogelators while their precursors did not form hydrogels. For those protected carbamates that are hydrogelators, removal of the 4,6-protection will result in a more water-soluble compound that produces a transition from gel to solution. Given the ability of these compounds to form gels from solution or solution from gels in situ in response to acidic environments, these compounds may have practical applications as stimuli-responsive gelators in an aqueous medium. In turn, one hydrogelator was studied for the encapsulation and release of naproxen and chloroquine. The hydrogel exhibited sustained drug release over a period of several days, with the release of chloroquine being faster at lower pH due to the acid lability of the gelator molecule. The synthesis, characterization, gelation properties, and studies on drug diffusion are discussed.
Collapse
Affiliation(s)
- Jonathan Bietsch
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| | - Logan Baker
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| | - Anna Duffney
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| | - Alice Mao
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| | - Mary Foutz
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| | - Cheandri Ackermann
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| | - Guijun Wang
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA
| |
Collapse
|
6
|
Novel amphiphilic hydroxyethyl starch-based nanoparticles loading camptothecin exhibit high anticancer activity in HepG2 cells and zebrafish. Colloids Surf B Biointerfaces 2023; 224:113215. [PMID: 36841205 DOI: 10.1016/j.colsurfb.2023.113215] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/01/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
Camptothecin is a naturally occurred anticancer drug but exhibits limitations including poor aqueous solubility, low bioavailability, and high level of adverse drug reactions on normal organs. To overcome these problems, this paper developed a novel amphiphilic Lau-Leu-HES carrier using hydroxyethyl starch, lauric acid, and L-leucine as starting materials. The carrier was successfully applied to prepare Lau-Leu-HES nanoparticles loading camptothecin. The drug loading efficiency and encapsulation efficiency of the nanoparticles were calculated to be 29.04% and 81.85%, respectively. The nanoparticles exhibited high zeta potential (-15.51 mV) and small hydrodynamic diameter (105.4 nm). Camptothecin in nanoparticles could be rapidly released under acidic condition (pH = 4.5), thereby indicating the high sensitivity under cancer microenvironments. Anticancer investigation revealed that the nanoparticles could inhibit the proliferation of HepG2 cells in vitro. Compared with commercial available drug doxorubicin, the nanoparticles could significantly inhibit the expression of krasv12 oncogene in transgenic Tg (EGFP-krasV12) zebrafish. These results indicate that the camptothecin-loaded Lau-Leu-HES nanoparticles are expected to be a potential candidate for cancer therapy.
Collapse
|
7
|
Abbasi YF, Bera H, Cun D, Yang M. Recent advances in pH/enzyme-responsive polysaccharide-small-molecule drug conjugates as nanotherapeutics. Carbohydr Polym 2023; 312:120797. [PMID: 37059536 DOI: 10.1016/j.carbpol.2023.120797] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/26/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Now-a-days, the polysaccharides are extensively employed for the delivery of small-molecule drugs ascribed to their excellent biocompatibility, biodegradability and modifiability. An array of drug molecules is often chemically conjugated with different polysaccharides to augment their bio-performances. As compared to their therapeutic precursors, these conjugates could typically demonstrate an improved intrinsic solubility, stability, bioavailability and pharmacokinetic profiles of the drugs. In current years, various stimuli-responsive particularly pH and enzyme-sensitive linkers or pendants are also exploited to integrate the drug molecules into the polysaccharide backbone. The resulting conjugates could experience a rapid molecular conformational change upon exposure to the microenvironmental pH and enzyme changes of the diseased states, triggering the release of the bioactive cargos at the targeted sites and eventually minimize the systemic side effects. Herein, the recent advances in pH and enzyme -responsive polysaccharide-drug conjugates and their therapeutic benefits are systematically reviewed, following a brief description on the conjugation chemistry of the polysaccharides and drug molecules. The challenges and future perspectives of these conjugates are also precisely discussed.
Collapse
|
8
|
Qian R, Cao G, Su W, Zhang J, Jiang Y, Song H, Jia F, Wang H. Enhanced Sensitivity of Tumor Cells to Autophagy Inhibitors Using Fasting-Mimicking Diet and Targeted Lysosomal Delivery Nanoplatform. NANO LETTERS 2022; 22:9154-9162. [PMID: 36342406 DOI: 10.1021/acs.nanolett.2c03890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Autophagy is one of the key pathways for tumor cell survival and proliferation. Therefore, inhibition of autophagy has been extensively studied for cancer therapy. However, current autophagy inhibitors lack specificity and are ineffective in limiting tumor progression. Herein, we report a nanoplatform for tumor-site-targeted delivery of hydroxychloroquine (HCQ) using insulin-like growth factors 2 receptor (IGF2R)-targeted liposomes (iLipo-H). A fasting-mimicking diet (FMD) is used to increase the autophagy levels in tumor cells, thereby increasing the sensitivity of tumor cells to HCQ. In addition, FMD treatment upregulates the expression of IGF2R in tumor cells, but not normal cells. Consequently, iLipo-H nanoparticles efficiently accumulate at the tumor site under FMD condition. In vivo studies demonstrate that iLipo-H nanoparticles efficiently inhibit 4T1 tumor growth without obvious side effects, especially under FMD condition. This study provides a promising strategy to increase the sensitivity of tumor cells to autophagy inhibitors for effective cancer therapy.
Collapse
Affiliation(s)
- Ruihao Qian
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoliang Cao
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150040, China
| | - Wen Su
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Jie Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Jiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haohao Song
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Fuhao Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| |
Collapse
|
9
|
Sivamaruthi BS, Nallasamy PK, Suganthy N, Kesika P, Chaiyasut C. Pharmaceutical and biomedical applications of starch-based drug delivery system: A review. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
10
|
Fabrication of a magnetic nanocarrier for doxorubicin delivery based on hyperbranched polyglycerol and carboxymethyl cellulose: An investigation on the effect of borax cross-linker on pH-sensitivity. Int J Biol Macromol 2022; 203:80-92. [PMID: 35092736 DOI: 10.1016/j.ijbiomac.2022.01.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 11/22/2022]
Abstract
A new core-shell pH-responsive nanocarrier was prepared based on magnetic nanoparticle (MNP) core. Magnetic nanoparticles were first modified with hyperbranched polyglycerol as the first shell. Then the magnetic core was decorated with doxorubicin anticancer drug (DOX) and covered with PEGylated carboxymethylcellulose as the second shell. Borax was used to partially cross-link organic shells in order to evaluate drug loading content and pH-sensitivity. The structure of nanocarrier, organic shell loadings, magnetic responsibility, morphology, size, dispersibility, and drug loading content were investigated by IR, NMR, TG, VSM, XRD, DLS, HR-TEM and UV-Vis analyses. In vitro release investigations demonstrated that the use of borax as cross-linker between organic shells make the nanocarrier highly sensitive to pH so that more that 70% of DOX is released in acidic pH. A reverse pH-sensitivity was observed for the nanocarrier without borax cross-linker. The MTT assay determined that the nanocarrier exhibited excellent biocompatibility toward normal cells (HEK-293) and high toxicity against cancerous cells (HeLa). The nanocarrier also showed high hemocompatibility. Cellular uptake revealed high ability of nanocarrier toward HeLa cells comparable with free DOX. The results also suggested that low concentration of nanocarrier has a great potential for use as contrast agent in magnetic resonance imaging (MRI).
Collapse
|
11
|
Paskeh MDA, Entezari M, Clark C, Zabolian A, Ranjbar E, Farahani MV, Saleki H, Sharifzadeh SO, Far FB, Ashrafizadeh M, Samarghandian S, Khan H, Ghavami S, Zarrabi A, Łos MJ. Targeted regulation of autophagy using nanoparticles: New insight into cancer therapy. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166326. [DOI: 10.1016/j.bbadis.2021.166326] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/31/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022]
|
12
|
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
|
13
|
Eivazzadeh-Keihan R, Asgharnasl S, Moghim Aliabadi HA, Tahmasebi B, Radinekiyan F, Maleki A, Bahreinizad H, Mahdavi M, Alavijeh MS, Saber R, Lanceros-Méndez S, Shalan AE. Magnetic graphene oxide-lignin nanobiocomposite: a novel, eco-friendly and stable nanostructure suitable for hyperthermia in cancer therapy. RSC Adv 2022; 12:3593-3601. [PMID: 35425373 PMCID: PMC8979318 DOI: 10.1039/d1ra08640e] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023] Open
Abstract
In this research, a novel magnetic nanobiocomposite was designed and synthesized in a mild condition, and its potential in an alternating magnetic field was evaluated for hyperthermia applications. For this purpose, in the first step, graphene oxide was functionalized with a natural lignin polymer using epichlorohydrin as the cross-linking agent. In the second step, the designed magnetic graphene oxide-lignin nanobiocomposite was fabricated by the in situ preparation of magnetic Fe3O4 nanoparticles in the presence of graphene oxide functionalized with lignin. The resultant magnetic nanobiocomposite possessed certain main properties, including stability and homogeneity in aqueous solutions, making it suitable for hyperthermia applications. The chemical and structural properties of the synthesized magnetic graphene oxide-lignin composite were characterized using FT-IR, EDX, FE-SEM, TEM, TG and VSM analyses. The saturation magnetization value of this magnetic nanocomposite was recorded as 17.2 emu g-1. Further, the maximum specific absorption rate was determined to be 121.22 W g-1. Given these results, this newly fabricated magnetic nanobiocomposite may achieve considerable performance under the alternating magnetic field in fluid hyperthermia therapy.
Collapse
Affiliation(s)
- Reza Eivazzadeh-Keihan
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology Tehran 16846-13114 Iran +98-21-73021584 +98-21-73228313
| | - Somayeh Asgharnasl
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology Tehran 16846-13114 Iran +98-21-73021584 +98-21-73228313
| | - Hooman Aghamirza Moghim Aliabadi
- Protein Chemistry Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran Tehran Iran
- Advanced Chemistry Studies Lab, Department of Chemistry, K. N. Toosi University of Technology Tehran Iran
| | - Behnam Tahmasebi
- School of Chemistry, College of Science, University of Tehran Tehran Iran
| | - Fateme Radinekiyan
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology Tehran 16846-13114 Iran +98-21-73021584 +98-21-73228313
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology Tehran 16846-13114 Iran +98-21-73021584 +98-21-73228313
| | - Hossein Bahreinizad
- Mechanical Engineering Department, Sahand University of Technology Tabriz Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| | | | - Reza Saber
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences Tehran Iran
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Senentxu Lanceros-Méndez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, Martina Casiano, UPV/EHU Science Park Barrio Sarriena s/n Leioa 48940 Spain
- IKERBASQUE, Basque Foundation for Science 48009 Bilbao Spain
| | - Ahmed Esmail Shalan
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, Martina Casiano, UPV/EHU Science Park Barrio Sarriena s/n Leioa 48940 Spain
- Central Metallurgical Research and Development Institute (CMRDI) P. O. Box 87, Helwan Cairo 11421 Egypt
| |
Collapse
|
14
|
Mariadoss AVA, Saravanakumar K, Sathiyaseelan A, Karthikkumar V, Wang MH. Smart drug delivery of p-Coumaric acid loaded aptamer conjugated starch nanoparticles for effective triple-negative breast cancer therapy. Int J Biol Macromol 2022; 195:22-29. [PMID: 34861273 DOI: 10.1016/j.ijbiomac.2021.11.170] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/15/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022]
Abstract
The nano-drug delivery system utilizing the ligand functionalized nanoparticles have a tremendous application in cancer therapeutics. The present study was aimed to fabricate the p-Coumaric acid-loaded aptamer (ligand) conjugated starch nanoparticles (Apt-p-CA-AStNPs) for effective treatment of triple-negative breast cancer (MDA-MB-231). The FT-IR spectrum showed the presence of functional groups associated with para-Coumaric acid (p-CA) and amino starch (AS) in p-CA-AStNPs. Further, the conjugation of aptamer in p-CA-AStNPs was confirmed by agarose gel electrophoresis. Transmission electron microscopic analysis revealed that the synthesized Apt-p-CA-AStNPs were less agglomerated. The zeta size analyzer displayed the average particle size of 218.97 ± 3.07 nm with ȥ-potential -29.2 ± 1.35 mV, and PDI 0.299 ± 0.05 for Apt-p-CA-AStNPs. The drug encapsulation and loading efficiencies were 80.30 ± 0.53% and 10.35 ± 0.85% respectively for Apt-p-CA-AStNPs. Apt-p-CA-AStNPs showed a rapid and bursting release in the initial five hours of the experiment in pH 5.4. A significant change was found in their cytotoxic efficacy between the samples: p-CA, p-CA-AStNPs, and Apt-p-CA-AStNPs. Among the tested samples, Apt-p-CA-AStNPs caused higher cytotoxicity in MDA-MB-231 cells through ROS regulation, nuclear damage, mitochondrial membrane potential, and apoptosis-related protein expressions. Overall, these results proved that Apt-p-CA-AStNPs were efficiently inhibited the MDA-MB-231 cells by regulating apoptosis.
Collapse
Affiliation(s)
| | - Kandasamy Saravanakumar
- Department of Bio-Health Convergences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Anbazhagan Sathiyaseelan
- Department of Bio-Health Convergences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Venkatachalam Karthikkumar
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Myeong-Hyeon Wang
- Department of Bio-Health Convergences, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| |
Collapse
|
15
|
Zhen W, Khalid A, Ali P, Rehman H, Siddiqui MK, Ullah H. Topological Study of Some Covid-19 Drugs by Using Temperature Indices. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2025864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Wang Zhen
- School of Computer Engineering, Anhui Wenda University of Information Engineering, Hefei, China
| | - Asma Khalid
- Department of Mathematics, Air University Islamabad, Multan, Pakistan
| | - Parvez Ali
- Department of Mechanical Engineering, College of Engineering, Qassim University, Unaizah, Saudi Arabia
| | - Haider Rehman
- Department of Mathematics, Air University Islamabad, Multan, Pakistan
| | | | - Hameed Ullah
- Department of Mathematics, Comsats University Islamabad, Sahiwal, Pakistan
| |
Collapse
|
16
|
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
|
17
|
Gopinath V, Kamath SM, Priyadarshini S, Chik Z, Alarfaj AA, Hirad AH. Multifunctional applications of natural polysaccharide starch and cellulose: An update on recent advances. Biomed Pharmacother 2021; 146:112492. [PMID: 34906768 DOI: 10.1016/j.biopha.2021.112492] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The emergence of clinical complications and therapeutic challenges for treating various diseases necessitate the discovery of novel restorative functional materials. Polymer-based drug delivery systems have been extensively reported in the last two decades. Recently, there has been an increasing interest in the progression of natural biopolymers based controlled therapeutic strategies, especially in drug delivery and tissue engineering applications. However, the solubility and functionalisation due to their complex network structure and intramolecular bonding seem challenging. This review explores the current advancement and prospects of the most promising natural polymers such as cellulose, starch and their derivatives-based drug delivery vehicles like hydrogels, films and composites, in combating major ailments such as bone infections, microbial infections, and cancers. In addition, selective drug targeting using metal-drug (MD) and MD-based polymeric missiles have been exciting but challenging for its application in cancer therapeutics. Owing to high biocompatibility of starch and cellulose, these materials have been extensively evaluated in biomedical and pharmaceutical applications. This review presents a detailed impression of the current trends for the construction of biopolymer-based tissue engineering, drug/gene/protein delivery vehicles.
Collapse
Affiliation(s)
- V Gopinath
- University of Malaya Centre for Proteomics Research, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - S Manjunath Kamath
- Department of Translational Medicine and Research, SRM Medical College Hospital and Research, SRMIST, Kattankulathur 603203, India.
| | - S Priyadarshini
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Zamri Chik
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Abdurahman H Hirad
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
18
|
Zambuzi GC, Camargos CH, Ferreira MP, Rezende CA, de Freitas O, Francisco KR. Modulating the controlled release of hydroxychloroquine mobilized on pectin films through film-forming pH and incorporation of nanocellulose. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2021. [DOI: 10.1016/j.carpta.2021.100140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
19
|
Kumar N, Fazal S, Miyako E, Matsumura K, Rajan R. Avengers against cancer: A new era of nano-biomaterial-based therapeutics. MATERIALS TODAY 2021; 51:317-349. [DOI: 10.1016/j.mattod.2021.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
20
|
Li J, Chen X, Kang R, Zeh H, Klionsky DJ, Tang D. Regulation and function of autophagy in pancreatic cancer. Autophagy 2021; 17:3275-3296. [PMID: 33161807 PMCID: PMC8632104 DOI: 10.1080/15548627.2020.1847462] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Oncogenic KRAS mutation-driven pancreatic ductal adenocarcinoma is currently the fourth-leading cause of cancer-related deaths in the United States. Macroautophagy (hereafter "autophagy") is one of the lysosome-dependent degradation systems that can remove abnormal proteins, damaged organelles, or invading pathogens by activating dynamic membrane structures (e.g., phagophores, autophagosomes, and autolysosomes). Impaired autophagy (including excessive activation and defects) is a pathological feature of human diseases, including pancreatic cancer. However, dysfunctional autophagy has many types and plays a complex role in pancreatic tumor biology, depending on various factors, such as tumor stage, microenvironment, immunometabolic state, and death signals. As a modulator connecting various cellular events, pharmacological targeting of nonselective autophagy may lead to both good and bad therapeutic effects. In contrast, targeting selective autophagy could reduce potential side effects of the drugs used. In this review, we describe the advances and challenges of autophagy in the development and therapy of pancreatic cancer.Abbreviations: AMPK: AMP-activated protein kinase; CQ: chloroquine; csc: cancer stem cells; DAMP: danger/damage-associated molecular pattern; EMT: epithelial-mesenchymal transition; lncRNA: long noncoding RNA; MIR: microRNA; PanIN: pancreatic intraepithelial neoplasia; PDAC: pancreatic ductal adenocarcinoma; PtdIns3K: phosphatidylinositol 3-kinase; SNARE: soluble NSF attachment protein receptor; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Jingbo Li
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Xin Chen
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
21
|
Yang J, Muhammad MH, Siddiqui MK, Hanif MF, Nasir M, Ali S, Liu JB. Topological Co-indices of Hydroxyethyl Starch Conjugated with Hydroxychloroquine Used for COVID-19 Treatment. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1996407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Jun Yang
- GongQing Institute of Science and Technology, Gong Qing, China
| | - Mehwish Hussain Muhammad
- College of Chemistry, School of Chemical Engineering and Energy, Zhengzhou University, Zhengzhou, China
| | | | | | - Muhammad Nasir
- Department of Mathematics, COMSATS University Islamabad, Lahore Campus, Pakistan
| | - Safdar Ali
- Department of Mathematics, COMSATS University Islamabad, Lahore Campus, Pakistan
| | - Jia-Bao Liu
- School of Mathematics and Physics, Anhui Jianzhu University, Hefei, China
| |
Collapse
|
22
|
Hernandes IS, Da Silva HC, Dos Santos HF, De Almeida WB. Unveiling the Molecular Structure of Antimalarial Drugs Chloroquine and Hydroxychloroquine in Solution through Analysis of 1H NMR Chemical Shifts. J Phys Chem B 2021; 125:3321-3342. [PMID: 33760611 DOI: 10.1021/acs.jpcb.1c00609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) have been standard antimalarial drugs since the early 1950s, and very recently, the possibility of their use for the treatment of COVID-19 patients has been considered. To understand the drug mode of action at the submicroscopic level (atoms and molecules), molecular modeling studies with the aid of computational chemistry methods have been of great help. A fundamental step in such theoretical investigations is the knowledge of the predominant drug molecular structure in solution, which is the real environment for the interaction with biological targets. Our strategy to access this valuable information is to perform density functional theory (DFT) calculations of 1H NMR chemical shifts for several plausible molecular conformers and then find the best match with experimental NMR profile in solution (since it is extremely sensitive to conformational changes). Through this procedure, after optimizing 30 trial distinct molecular structures (ωB97x-D/6-31G(d,p)-PCM level of calculation), which may be considered representative conformations, we concluded that the global minimum (named M24), stabilized by an intramolecular N-H hydrogen bond, is not likely to be observed in water, chloroform, and dimethyl sulfoxide (DMSO) solution. Among fully optimized conformations (named M1 to M30, and MD1 and MD2), we found M12 (having no intramolecular H-bond) as the most probable structure of CQ and HCQ in water solution, which is a good approximate starting geometry in drug-receptor interaction simulations. On the other hand, the preferred CQ and HCQ structure in chloroform (and CQ in DMSO-d6) solution was assigned as M8, showing the solvent effects on conformational preferences. We believe that the analysis of 1H NMR data in solution can establish the connection between the macro level (experimental) and the sub-micro level (theoretical), which is not so apparent to us and appears to be more appropriate than the thermodynamic stability criterion in conformational analysis studies.
Collapse
Affiliation(s)
- Isabel S Hernandes
- Laboratório de Química Computacional e Modelagem Molecular (LQC-MM), Departamento de Química Inorgânica, Instituto de Química, Universidade Federal Fluminense (UFF), Outeiro de São João Batista s/n, Campus do Valonguinho, Centro, Niterói 24020-141, Rio de Janeiro, Brazil
| | - Haroldo C Da Silva
- Laboratório de Química Computacional e Modelagem Molecular (LQC-MM), Departamento de Química Inorgânica, Instituto de Química, Universidade Federal Fluminense (UFF), Outeiro de São João Batista s/n, Campus do Valonguinho, Centro, Niterói 24020-141, Rio de Janeiro, Brazil
| | - Hélio F Dos Santos
- Núcleo de Estudos em Química Computacional (NEQC), Departamento de Química, ICE, Universidade Federal de Juiz de Fora (UFJF), Campus Universitário, Martelos, Juiz de Fora 36036-330, Minas Gerais, Brazil
| | - Wagner B De Almeida
- Laboratório de Química Computacional e Modelagem Molecular (LQC-MM), Departamento de Química Inorgânica, Instituto de Química, Universidade Federal Fluminense (UFF), Outeiro de São João Batista s/n, Campus do Valonguinho, Centro, Niterói 24020-141, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Abstract
Since the last decade, the polymer-drug conjugate (PDC) approach has emerged as one of the most promising drug-delivery technologies owing to several benefits like circumventing premature drug release, offering controlled and targeted drug delivery, improving the stability, safety, and kinetics of conjugated drugs, and so forth. In recent years, PDC technology has advanced with the objective to further enhance the treatment outcomes by integrating nanotechnology and multifunctional characteristics into these systems. One such development is the ability of PDCs to act as theranostic agents, permitting simultaneous diagnosis and treatment options. Theranostic nanocarriers offer the opportunity to track the distribution of PDCs within the body and help to localize the diseased site. This characteristic is of particular interest, especially among those therapeutic approaches where external stimuli are supposed to be applied for abrupt drug release at the target site for localized delivery to avoid systemic side effects (e.g., Visudyne®). Thus, with the help of this review article, we are presenting the most recent updates in the domain of PDCs as nanotheranostic agents. Different methodologies utilized to design PDCs along with imaging characteristics and their applicability in a wide range of diseases, have been summarized in this article.
Collapse
|
24
|
Rauf A, Ishtiaq M, Siddiqui MK. Topological Study of Hydroxychloroquine Conjugated Molecular Structure Used for Novel Coronavirus (COVID-19) Treatment. Polycycl Aromat Compd 2021. [PMCID: PMC7852296 DOI: 10.1080/10406638.2021.1873807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The novel coronavirus disease 2019 (Covid-19) is a mutating and recombining pandemic that potentially spreading through an infected person in droplet-generated forms that have affected more than 200 countries and endanger the entire globe. There is no clear strategy for the care of COVID-19 cases. Moreover, experts across the globe are working actively to develop medicinal or anti-virus drugs. On the basis of recent clinical findings and recommendations, the study examined a variety of new medications that have shown antiviral activity against SARS-CoV-2, among other drugs, antimalarial medications Chloroquine (CQ) and Hydroxychloroquine (HCQ) have gained significant publicity to have promising effects against SARS-CoV-2. Linking a bioactive substance to a biocompatible polymer typically provides various concerns, such as improved drug solubilization, improved modification, precise restriction, and controlled discharge. An enormous number of medical analyses have confirmed that the characteristics of medical drugs have a nearby connection with their atomic structure. Medication properties can be acquired by considering the atomic structure of relating drugs. The calculation of the topological index of a medication structure empowers researchers to have a superior comprehension of the physical science and bio-organic attributes of drugs. Ev-degree and ve-degree based topological indices are two novel degrees based indices as of late defined in graph theory. Ev-degree and ve-degree based topological indices have been defined as corresponding to their relating partners. In this paper, we have computed topological indices based on ev-degree and ve-degree for the Hydroxyethyl Starch and Hydroxychloroquine (HCQ-HEC) bioconjugate molecular structure.
Collapse
Affiliation(s)
- Abdul Rauf
- Department of Computer Science and Engineering, Air University Multan Campus, Multan, Pakistan
| | - Muhammad Ishtiaq
- Department of Computer Science and Engineering, Air University Multan Campus, Multan, Pakistan
| | | |
Collapse
|
25
|
Tan R, Wan Y, Yang X. Hydroxyethyl starch and its derivatives as nanocarriers for delivery of diagnostic and therapeutic agents towards cancers. BIOMATERIALS TRANSLATIONAL 2020; 1:46-57. [PMID: 35837654 PMCID: PMC9255820 DOI: 10.3877/cma.j.issn.2096-112x.2020.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 01/17/2023]
Abstract
Many types of drugs and agents used for cancer diagnosis and therapy often have low bioavailability and insufficient efficacy, as well as causing various side effects due to their nonspecific delivery. Nanocarriers with purposely-designed compositions and structures have shown varying degrees of abilities to deliver these compounds towards cancers in passive or active manners. Despite the availability of a variety of materials for the construction of nanocarriers, natural polymers with good biocompatibility and biodegradability are preferable for such usage because of their high in vivo safety as well as easy removal of degradation products. Among the natural polymers intended for building nanocarriers, hydroxyethyl starch and its derivatives have gained tremendous attention in the field of drug delivery in the form of nanomedicines over the last decade. There is growing optimism that ever more hydroxyethyl starch-based nanomedicines will be a significant addition to the armoury currently used for cancer diagnosis and therapy.
Collapse
Affiliation(s)
| | - Ying Wan
- Corresponding authors: Ying Wan, ; Xiangliang Yang,
| | | |
Collapse
|
26
|
Manzoor S, Siddiqui MK, Ahmad S. On Entropy Measures of Polycyclic Hydroxychloroquine Used for Novel Coronavirus (COVID-19) Treatment. Polycycl Aromat Compd 2020. [DOI: 10.1080/10406638.2020.1852289] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Shazia Manzoor
- Department of Mathematics, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | | | - Sarfraz Ahmad
- Department of Mathematics, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| |
Collapse
|
27
|
Garcia MAVT, Garcia CF, Faraco AAG. Pharmaceutical and Biomedical Applications of Native and Modified Starch: A Review. STARCH-STARKE 2020. [DOI: 10.1002/star.201900270] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Maria Aparecida Vieira Teixeira Garcia
- Departamento de Alimentos, Faculdade de Farmácia/UFMG Av. Presidente Antônio Carlos, 6627 ‐ Campus Pampulha ‐ CEP 31270‐901 Belo Horizonte ‐ MG ‐ Brasil Brazil
| | - Cleverson Fernando Garcia
- Departamento de QuímicaCentro Federal de Educação Tecnológica de Minas Gerais (CEFET‐MG) Av. Amazonas, 5.253, Nova Suiça. CEP 30421‐169. Belo Horizonte ‐ MG ‐ Brasil Brazil
| | - André Augusto Gomes Faraco
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia/UFMGAv. Presidente Antônio Carlos, 6627 ‐ Campus Pampulha ‐ CEP 31270‐901 Belo Horizonte ‐ MG ‐ Brasil Brazil
| |
Collapse
|
28
|
Zheng W, Chen Q, Wang C, Yao D, Zhu L, Pan Y, Zhang J, Bai Y, Shao C. Inhibition of Cathepsin D (CTSD) enhances radiosensitivity of glioblastoma cells by attenuating autophagy. Mol Carcinog 2020; 59:651-660. [PMID: 32253787 DOI: 10.1002/mc.23194] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Postoperative radiotherapy combined with chemotherapy is a commonly used treatment for glioblastoma (GBM) but radiotherapy often fails to achieve the expected results mainly due to tumor radioresistance. In this study, we established a radioresistant subline from human glioma cell line U251 and found that Cathepsin D (CTSD), a gene closely related to the clinical malignancy and prognosis in glioma, had higher expression level in radioresistant clones than that in parental cells, and knocking down CTSD by small interfering RNA (siRNA) or its inhibitor Pepstatin-A increased the radiosensitivity. The level of autophagy was enhanced in the radioresistant GBM cells compared with its parent cells, and silencing autophagy by light chain 3 (LC3) siRNA significantly sensitized GBM cells to ionizing radiation (IR). Moreover, the protein expression level of CTSD was positively correlated with the autophagy marker LC3 II/I and negatively correlated with P62 after IR in radioresistant cells. As expected, through the combination of Western blot and immunofluorescence assays, inhibition of CTSD increased the formation of autophagosomes, while decreased the formation of autolysosomes, which indicating an attenuated autophagy level, leading to radiosensitization ultimately. Our results revealed for the first time that CTSD regulated the radiosensitivity of glioblastoma by affecting the fusion of autophagosomes and lysosomes. In significance, CTSD might be a potential molecular biomarker and a new therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Wang Zheng
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qianping Chen
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Wang
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan Yao
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Zhu
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Pan
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianghong Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Bai
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chunlin Shao
- Department of Radiation Biology, Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Torlopov MA, Drozd NN, Tarabukin DV, Udoratina EV. Synthesis and hemocompatibility of amino (di-)butyldeoxy modified hydroxyethyl starch. Int J Biol Macromol 2020; 145:936-943. [DOI: 10.1016/j.ijbiomac.2019.09.184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 01/22/2023]
|
30
|
Zhang H, Ren Y, Cao F, Chen J, Chen C, Chang J, Hou L, Zhang Z. In Situ Autophagy Disruption Generator for Cancer Theranostics. ACS APPLIED MATERIALS & INTERFACES 2019; 11:29641-29654. [PMID: 31364350 DOI: 10.1021/acsami.9b10578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Cancer remains a serious clinical disease awaiting new effective treatment strategies. Autophagy modulation has emerged as a novel and promising pharmacologic target critical to future drug development and anti-cancer therapy applications. Herein, we constructed an in situ autophagy disruption generator to break the balance of autophagy flow for tumor-targeting therapy. Hollow mesoporous manganese trioxide (Mn2O3) nanoparticles (NPs) were synthesized and conjugated with hyaluronic acid (HA) to form tumor-targeting drug carriers. Then, traditional autophagy inhibitor hydroxychloroquine (HCQ) was loaded into the hollow core of HA-Mn2O3, to form a multifunctional theranostics platform (HA-Mn2O3/HCQ). This nanoplatform displayed specific localization and retention in lysosomes after entering tumor cells. The synchronous release of HCQ and manganese ion (Mn2+) induced lysosomal alkalization and osmotic pressure elevation. Significantly greater lysosomal deacidification and autophagy blockade effect emerged after treatment by this nanoplatform, with in vitro tumor inhibition rate of 92.2%. Imaging experiment proved that it could selectively deliver HCQ to tumor sites and further degrade to realize simultaneous release of Mn2+ and HCQ. Micromorphological and immunofluorescence analysis demonstrated that in situ high concentrations of these two substances would achieve effective autophagy blockade. Pharmacodynamics test showed that this nanogenerator displayed the best therapeutic efficacy with 5.08-fold tumor inhibition ratio compared with the HCQ group. Moreover, the generated Mn2+ can be used as T1 contrast agent for visualizing tumor lesions and monitoring therapeutic effects. Overall, the as-made multifunctional drug-delivery system might provide a promising platform for cancer theranostics upon in situ autophagy disruption.
Collapse
Affiliation(s)
- Huijuan Zhang
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases , Zhengzhou 450001 , Henan Province , China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation , Zhengzhou 450001 , Henan Province , China
| | - Yanping Ren
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , China
| | - Fang Cao
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , China
| | - Jianjiao Chen
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , China
| | - Chengqun Chen
- Department of Pharmacy , The First Affiliated Hospital of Zhengzhou University , Mailing Address: No. 100, Kexue Road , Zhengzhou 450001 , P. R. China
| | - Junbiao Chang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation , Zhengzhou 450001 , Henan Province , China
| | - Lin Hou
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases , Zhengzhou 450001 , Henan Province , China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation , Zhengzhou 450001 , Henan Province , China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases , Zhengzhou 450001 , Henan Province , China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation , Zhengzhou 450001 , Henan Province , China
| |
Collapse
|
31
|
Tang Y, Li Y, Li S, Hu H, Wu Y, Xiao C, Chu Z, Li Z, Yang X. Transformable nanotherapeutics enabled by ICG: towards enhanced tumor penetration under NIR light irradiation. NANOSCALE 2019; 11:6217-6227. [PMID: 30874705 DOI: 10.1039/c9nr01049a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Tumor penetration is the bottleneck for current cancer nanomedicine, limiting the ultimate antitumor efficacy in the clinic. Herein, by exploiting the well-known instability of indocyanine green (ICG), we report the preparation of near infrared (NIR) light responsive nanoparticles (NP) for enhanced tumor penetration. ICG crosslinks hydroxyethyl starch (HES) and doxorubicin (DOX) conjugates (HES-SS-DOX) via noncovalent interactions, facilitating the formation of ICG@HES-SS-DOX NP. The light triggered degradation of ICG leads to the dissociation of such NP, and the resulting HES-SS-DOX has been shown to penetrate deeper in both H22 tumor spheroids and tumor bearing mice, due to the photothermal effect of ICG. Therefore, the disintegrable ICG@HES-SS-DOX NP have better tumor penetration capacity than their counterparts, which originally cannot dissociate under NIR light stimulation. The reported ICG@HES-SS-DOX NP might be potent in treating malignant tumors with dense extracellular matrices, such as liver and pancreatic cancers. This study opens up a novel functionality of FDA-approved ICG for cancer nanotherapeutics.
Collapse
Affiliation(s)
- Yuxiang Tang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Xiao C, Hu H, Yang H, Li S, Zhou H, Ruan J, Zhu Y, Yang X, Li Z. Colloidal hydroxyethyl starch for tumor-targeted platinum delivery. NANOSCALE ADVANCES 2019; 1:1002-1012. [PMID: 36133197 PMCID: PMC9473228 DOI: 10.1039/c8na00271a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/23/2018] [Indexed: 05/06/2023]
Abstract
Cis-platinum has been widely used as a first-line chemotherapy agent in clinics for more than 40 years. Although considerable efforts have been expended for developing platinum-based nano drug delivery systems (NDDS) to resolve the problems of low water solubility, short half-life, and severe side effects of cis-platinum, it remains challenging to apply these nanoplatforms to cancer treatments in clinics on account of the issues related to safety, complex fabrication procedures, and limited cellular uptake. Herein, we constructed a novel cis-platinum delivery system with hydroxyethyl starch (HES), which is a semisynthetic polysaccharide that has been used worldwide as colloidal plasma volume expanders (PVE) in clinics for several decades. By combining TEM, AFM, and DLS, we have found that HES particles are colloidal nanoparticles in solution, with diameters ranging from 15 to 40 nm as a function of molecular weight. We further revealed that HES adopted a hyperbranched colloidal structure with rather compact conformation. These results demonstrate that HES is a promising nanocarrier to deliver drug molecules. Taking advantage of the poly-hydroxyl sites of HES, we constructed a novel HES-based cis-platinum delivery nanoplatform. HES was directly conjugated with cis-platinum prodrug via an ester bond and decorated with an active targeting molecule, lactobionic acid (LA), contributing toward higher in vitro antitumor activity against hepatoma carcinoma cells as compared to cis-platinum. These results have significant implications for the clinically used plasma volume expander-HES and shed light on the clinical translation of HES-based nano drug delivery systems.
Collapse
Affiliation(s)
- Chen Xiao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China +86 27 87792234 +86 27 87792234
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Hang Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China +86 27 87792234 +86 27 87792234
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Hai Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China +86 27 87792234 +86 27 87792234
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Si Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China +86 27 87792234 +86 27 87792234
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Hui Zhou
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Jian Ruan
- Wuhan HUST Life Science & Technology Co., Ltd Wuhan 430223 China
| | - Yuting Zhu
- Wuhan HUST Life Science & Technology Co., Ltd Wuhan 430223 China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China +86 27 87792234 +86 27 87792234
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology Wuhan 430074 China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China +86 27 87792234 +86 27 87792234
- Department of Nanomedicine and Biopharmaceutics, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology Wuhan 430074 China
- Wuhan Institute of Biotechnology High Tech Road 666, East Lake High Tech Zone Wuhan 430040 China
| |
Collapse
|
33
|
Lycium barbarum polysaccharides grafted with doxorubicin: An efficient pH-responsive anticancer drug delivery system. Int J Biol Macromol 2019; 121:964-970. [DOI: 10.1016/j.ijbiomac.2018.10.103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/28/2018] [Accepted: 10/14/2018] [Indexed: 12/19/2022]
|
34
|
Wu H, Hu H, Wan J, Li Y, Wu Y, Tang Y, Xiao C, Xu H, Yang X, Li Z. Hydroxyethyl starch stabilized polydopamine nanoparticles for cancer chemotherapy. CHEMICAL ENGINEERING JOURNAL 2018; 349:129-145. [DOI: 10.1016/j.cej.2018.05.082] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2025]
|
35
|
Autophagy therapeutics: preclinical basis and initial clinical studies. Cancer Chemother Pharmacol 2018; 82:923-934. [PMID: 30225602 DOI: 10.1007/s00280-018-3688-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Autophagy captures and degrades intracellular components such as proteins and organelles to sustain metabolism and homeostasis. Rapidly accumulating attention is being paid to the role of autophagy in the development of cancer, which makes autophagy attractive tools and targets for novel therapeutic approaches. Functional studies have confirmed that autophagy dysregulation is causal in many cases of cancer, with autophagy acting as tumor suppressors or tumor promoters, and autophagy inhibitor or promoter has shown promise in preclinical studies. The autophagy-targeted therapeutics using chloroquine/hydroxychloroquine have reached clinical development for treating cancer, but these drugs are actually not efficient probably because of a reduced penetration within the tumor. In this review, we first discuss the discoveries related to dual function of autophagy in cancer. Then, we provide an overview of preclinical studies and clinical trials involved in the development of autophagy therapeutics and finally discuss the future of such therapies.
Collapse
|
36
|
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
|
37
|
Yu E, Mangunuru HPR, Telang NS, Kong CJ, Verghese J, Gilliland Iii SE, Ahmad S, Dominey RN, Gupton BF. High-yielding continuous-flow synthesis of antimalarial drug hydroxychloroquine. Beilstein J Org Chem 2018; 14:583-592. [PMID: 29623120 PMCID: PMC5852550 DOI: 10.3762/bjoc.14.45] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/14/2018] [Indexed: 11/23/2022] Open
Abstract
Numerous synthetic methods for the continuous preparation of fine chemicals and active pharmaceutical ingredients (API’s) have been reported in recent years resulting in a dramatic improvement in process efficiencies. Herein we report a highly efficient continuous synthesis of the antimalarial drug hydroxychloroquine (HCQ). Key improvements in the new process include the elimination of protecting groups with an overall yield improvement of 52% over the current commercial process. The continuous process employs a combination of packed bed reactors with continuous stirred tank reactors for the direct conversion of the starting materials to the product. This high-yielding, multigram-scale continuous synthesis provides an opportunity to achieve increase global access to hydroxychloroquine for treatment of malaria.
Collapse
Affiliation(s)
- Eric Yu
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Hari P R Mangunuru
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Nakul S Telang
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Caleb J Kong
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Jenson Verghese
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Stanley E Gilliland Iii
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Saeed Ahmad
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - Raymond N Dominey
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| | - B Frank Gupton
- Department of Chemistry and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main St., Richmond, VA 23220, USA
| |
Collapse
|