1
|
Kumar KP, Madhusoodanan M, Pangath M, Menon D. Innovative landscapes in intraperitoneal therapy of ovarian cancer. Drug Deliv Transl Res 2025; 15:1877-1906. [PMID: 39888579 DOI: 10.1007/s13346-024-01765-w] [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] [Accepted: 12/09/2024] [Indexed: 02/01/2025]
Abstract
Epithelial ovarian cancer is the most prevalent gynecological malignancy, characterized by high mortality rates due to its late-stage diagnosis and frequent recurrence. The current standard of care for ovarian cancer is a combination of debulking surgery followed by the conventional mode of chemotherapy. Despite significant advances in therapeutic modalities, the overall survival rate of EOC continues to be poor, mainly because low concentrations of the chemotherapeutics reach the peritoneum, which is the primary site of ovarian cancer, leading to disease relapse. Here, intraperitoneal chemotherapy gains advantage due to its ability to deliver the drug molecules directly to the peritoneal cavity and provide localized and sustained effects. This is facilitated by the use of diverse kinds of nano or micron sized delivery systems, which help in transporting drugs, vaccines, antibodies and genes appropriately to the peritoneum for its desired function. This review article delves on how intraperitoneal delivery impacts the therapy of epithelial ovarian cancer spanning the conventional therapeutic modes to the recent nanoinnovations in chemotherapy, immunotherapy and gene therapy.
Collapse
Affiliation(s)
- Krishna Pradeep Kumar
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesha Madhusoodanan
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Meghna Pangath
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Deepthy Menon
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
2
|
Sedrati F, Bouzahouane H, Menaa M, Khaldi F, Bouarroudj T, Gzara L, Bensalem M, Laouar O, Sleimi N, Nasri H, Silva CO, Ouali K. Histological and biochemical evidence of Cr 2O 3 and Al 2O 3 nanoparticles toxicity in the marine gastropod Stramonita haemastoma: A preliminary application of integrated biomarker response (IBR). Comp Biochem Physiol C Toxicol Pharmacol 2025; 292:110159. [PMID: 40010532 DOI: 10.1016/j.cbpc.2025.110159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Nanoparticles (NPs) have actively contributed to nanotechnologies advancement over the last years, due to the unique properties they possess compared to their pristine counterparts. Consequently, NPs found wide applications in various fields such as the medical, biomedical, chemical, agro-food industries, and cosmetology. NP's extensive uses could lead to their release into the environment, especially in the marine ecosystems, considered as NPs sink, resulting in harmful effects on organisms. Concerns regarding NPs' toxicity in aquatic organisms have emerged, however, several points remain unexplored. In the present study, the toxicity of chromium oxide (Cr2O3 = 42 nm) and aluminum oxide (Al2O3 = 38 nm) NPs (1 mg/L, 2.5 mg/L, and 5 mg/L) in the gills of the marine gastropod Stramonita haemastoma was assessed through time (7, 14, and 28 days) by a multi-biomarker, Integrated biomarkers response (IBR), and Histological analysis. Both NPs induced varied changes in the antioxidant system, suggesting the onset of oxidative stress marked by superoxide dismutase (SOD), catalase (CAT), acetylcholinesterase (AChE), metallothionein (MT), and malondialdehyde (MDA) levels imbalance. Varied histological alterations in the gills of S. haemastoma were also observed including inflammation, hypertrophy, and lamellar fusion, IBR proved to be a promising tool for assessing NPs toxicity in gastropods. In this study results indicated the co-response of reduced glutathione (GSH), glutathione S-transferase (GST), glutathione peroxidase (GPx), CAT, SOD, and MT after 28 days of exposure. S. haemastoma showed sensitivity to all exposure concentrations of NPs thus validating this species as a suitable indicator of NPs contamination and toxicity.
Collapse
Affiliation(s)
- Fateh Sedrati
- Laboratory of Sciences and Technology of Water and Environment. Mohamed Cherif Messaadia University, BP 1553, 41000 Souk Ahras, Algeria. https://x.com/fateh_sedr34395
| | - Hana Bouzahouane
- Department of Biology, Faculty of Nature and Life Sciences, Mohamed Cherif Messaadia University, Souk Ahras 41000, Algeria; Laboratory of Environmental Biosurveillance, Department of Biology, Faculty of Sciences, Badji Mokhtar University, BP 12, El Hadjar, Annaba 23000, Algeria.
| | - Mohcen Menaa
- Department of Biology, Faculty of Nature and Life Sciences, Mohamed Cherif Messaadia University, Souk Ahras 41000, Algeria
| | - Fadila Khaldi
- Laboratory of Sciences and Technology of Water and Environment. Mohamed Cherif Messaadia University, BP 1553, 41000 Souk Ahras, Algeria; Department of Biology, Faculty of Nature and Life Sciences, Mohamed Cherif Messaadia University, Souk Ahras 41000, Algeria
| | - Tayeb Bouarroudj
- Center for Scientific and Technical Research in Physico-Chemical Analyzes (CRAPC), BP384, Bou-Ismail, Tipaza, RP 42004, Algeria
| | - Lassaad Gzara
- Center of Excellence in Desalination Technology, King Abdulaziz University, P.O. Box: 80200, Jeddah 21589, Saudi Arabia
| | - Mounira Bensalem
- University August 20, 1955, Skikda, Bp26, El Hadaik, Skikda, Algeria
| | - Omar Laouar
- Central laboratory of pathology and molecular biology, CHU, Annaba, Algeria; Faculty of Medicine, Badji Mokhtar University, BP 12, El Hadjar, Annaba 23000, Algeria
| | - Noomene Sleimi
- RME-Laboratory of Resources, Materials, and Ecosystems, Faculty of Sciences of Bizerte, University of Carthage, Bizerte 7021, Tunisia
| | - Hichem Nasri
- Laboratory of Biodiversity and Ecosystems Pollution, Faculty of Life and Nature Sciences, University of Chadli Bendjedid, El Taref, Algeria
| | - Carla O Silva
- MARE - Marine and Environmental Sciences Centre, ARNET - Aquatic Research Network Associate Laboratory, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Kheireddine Ouali
- Laboratory of Environmental Biosurveillance, Department of Biology, Faculty of Sciences, Badji Mokhtar University, BP 12, El Hadjar, Annaba 23000, Algeria
| |
Collapse
|
3
|
Jia Y, Kong X, Li R, Wang H, Li C, Cheng S, Duan W, Xiao Y, Mai Y, Deng W, Liu Y. Enhanced nasal-to-brain drug delivery by multivalent bioadhesive nanoparticle clusters for cerebral ischemic reperfusion injury protection. Acta Biomater 2025; 194:411-427. [PMID: 39870153 DOI: 10.1016/j.actbio.2025.01.036] [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: 08/02/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Following cerebral ischemia, reperfusion injury can worsen ischemia-induced functional, metabolic disturbances, and pathological damage upon blood flow restoration, potentially leading to irreversible harm. Yet, there's a dearth of advanced, localized drug delivery systems ensuring active pharmaceutical ingredient (API) efficacy in cerebral protection during ischemia-reperfusion. This study introduces a multivalent bioadhesive nanoparticle-cluster, merging bioadhesive nanoparticles (BNPs) with dendritic polyamidoamine (PAMAM), enhancing nose-to-brain delivery and brain protection efficacy against cerebral ischemia-reperfusion injuries (CIRI). The BNPs-PAMAM cluster exhibits superior adhesion within the rat nasal cavity, prolonged retention, enabling sustained drug release, cerebral transportation, and accumulation, resulting in enhanced intracerebral pharmacokinetic profile. Intranasal administration circumvents systemic delivery challenges, ensuring CIRI protection drugs reach ischemic areas pre-reperfusion, overcoming thrombus-related delays. Administering BNPs-PAMAM loaded with dexmedetomidine (DEX) pre-reperfusion effectively prevents neuron apoptosis by α2-adrenoceptor activation, modulating the ischemic microenvironment, exerting triple neuroprotective effects against cerebral reperfusion injury. Importantly, only therapeutic DEX releases and accumulates in the nasal cavity, averting brain nanomaterial toxicity, promising for repeat administrations. This study presents a translational platform for nasal-to-brain drug delivery in CNS disease treatment. STATEMENT OF SIGNIFICANCE: Innovative Drug Delivery System: This study introduces a multivalent bioadhesive nanoparticle-cluster (BNPs-PAMAM) to enhance nasal-to-brain drug delivery for cerebral ischemia-reperfusion injury (CIRI) treatment. Enhanced Retention and Efficacy: The BNPs-PAMAM system significantly improves drug retention in the nasal cavity and ensures sustained release, thereby enhancing the therapeutic efficacy of the neuroprotective agent dexmedetomidine (DEX). Blood-Brain Barrier Circumvention: By leveraging intranasal administration, the system bypasses the blood-brain barrier, delivering DEX directly to ischemic brain regions before reperfusion and minimizing systemic side effects. Triple Neuroprotective Effects for CIRI protection: DEX delivered via BNPs-PAMAM effectively reduces oxidative stress and inflammation while enhancing mitochondrial autophagy, providing comprehensive protection against neuronal damage.
Collapse
Affiliation(s)
- Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaohan Kong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Rui Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Han Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chujie Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shihong Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Wei Duan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yan Xiao
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
4
|
Chang J, Shin K, Lewis JM, Suh HW, Lee J, Damsky W, Xu S, Bosenberg M, Saltzman WM, Girardi M. Enhanced Intratumoral Delivery of Immunomodulator Monophosphoryl Lipid A through Hyperbranched Polyglycerol-Coated Biodegradable Nanoparticles. J Invest Dermatol 2025; 145:593-604. [PMID: 39122142 DOI: 10.1016/j.jid.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Immunomodulatory agents have significant potential to enhance cancer treatment but have demonstrated limited efficacy beyond the preclinical setting owing to poor pharmacokinetics and toxicity associated with systemic administration. Conversely, when locally delivered, immunomodulatory agents require repeated administration to optimize immune stimulation. To overcome these challenges, we encapsulated the toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) within hyperbranched polyglycerol-coated biodegradable nanoparticles (NPs) engineered for gradual drug release from the NP core, resulting in a more persistent stimulation of antitumor immune responses while minimizing systemic side effects. In a model of malignant melanoma, we demonstrate that hyperbranched polyglycerol-NP encapsulation significantly improves the antitumor efficacy of MPLA by enhancing its ability to remodel the tumor microenvironment. Relative to free MPLA, hyperbranched polyglycerol-coated NP-encapsulated MPLA significantly increased the NK cell- and cytotoxic T-cell-mediated antitumor immune response and tuned the tumor-draining lymph nodes toward a T helper 1 response. Furthermore, when combined with local delivery of a chemotherapeutic agent, hyperbranched polyglycerol-NP-MPLA induces the conversion of an immunosuppressive tumor microenvironment to immunogenic tumor microenvironment and significantly improves survival.
Collapse
Affiliation(s)
- Jungsoo Chang
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Julia M Lewis
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hee Won Suh
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Joohyung Lee
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Suzanne Xu
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - W Mark Saltzman
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Department of Chemical & Environmental Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Girardi
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
5
|
Almuqbil RM, Aldhubiab B. Bioadhesive Nanoparticles in Topical Drug Delivery: Advances, Applications, and Potential for Skin Disorder Treatments. Pharmaceutics 2025; 17:229. [PMID: 40006596 PMCID: PMC11860006 DOI: 10.3390/pharmaceutics17020229] [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: 01/15/2025] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Skin disorders are the fourth most common cause of all diseases, which affect nearly one-third of the world's population. Topical drug delivery can be effective in treating a range of skin disorders, including microbial infections, skin cancer, dermatitis, burn injury, wounds, and psoriasis. Bioadhesive nanoparticles (BNPs) can serve as an efficient topical drug delivery system as they can serve dual purposes as bioadhesives and nanocarriers, which can mediate targeted drug delivery, prolong retention time, and deepen drug penetration through skin layers. There is an increasing demand for BNP-based applications in medicine because of their various advantages, including biodegradability, flexibility, biocompatibility, and enhanced adhesive strength. A number of BNPs have already been developed and evaluated as potential topical drug delivery systems. In addition, a range of studies have already been carried out to evaluate the potential of BNPs in the treatment of various skin disorders, including atopic dermatitis, irritant contact dermatitis, skin cancer, psoriasis, microbial infections, wounds, and severe burn injuries. This review article is timely and unique, because it provides an extensive and unique summary of the recent advances of BNPs in the treatment of wide-ranging skin disorders. Moreover, this review also provides a useful discussion on the bioadhesion mechanism and various biopolymers that can be used to prepare BNPs.
Collapse
Affiliation(s)
- Rashed M. Almuqbil
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | | |
Collapse
|
6
|
Pan S, Yuan H, Zhai Q, Zhang Y, He H, Yin T, Tang X, Gou J. The journey of nanoparticles in the abdominal cavity: Exploring their in vivo fate and impact factors. J Control Release 2024; 376:266-285. [PMID: 39396710 DOI: 10.1016/j.jconrel.2024.10.011] [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: 07/24/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Peritoneal carcinomatosis (PC) is caused by metastasis of primary tumor cells from intra-abdominal organs to the peritoneal surface. Intraperitoneal (IP) chemotherapy allows close contact of high concentrations of therapeutic agents with cancer cells in the peritoneal cavity to prolong patient survival. However, conventional IP chemotherapy is prone to rapid elimination from the peritoneal cavity and lacks specificity towards cancer cells. To address these challenges, there is an imperative demand for exploiting novel drug delivery systems to enhance drug retention in the peritoneal cavity and target PC cells. Therefore, in this review, we first recapitulate the physiological structures and barriers associated with IP drug delivery, highlighting the in vivo fate of nanoparticles (NPs) after IP administration. Furthermore, the influence of physicochemical properties (particle size, charge, surface modification, and carrier composition) on the in vivo fate of NPs is discussed. Perspectives on the rational design of NPs for IP therapy and recent clinical progress are also provided.
Collapse
Affiliation(s)
- Shu Pan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Qiyao Zhai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
7
|
Saravanan J, Nair A, Krishna SS, Viswanad V. Nanomaterials in biology and medicine: a new perspective on its toxicity and applications. Drug Chem Toxicol 2024; 47:767-784. [PMID: 38682270 DOI: 10.1080/01480545.2024.2340002] [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: 07/07/2023] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024]
Abstract
Nanotechnology offers excellent prospects for application in biology and medicine. It is used for detecting biological molecules, imaging, and as therapeutic agents. Due to nano-size (1-100 nm) and high surface-to-volume ratio, nanomaterials possess highly specific and distinct characteristics in the biological environment. Recently, the use of nanomaterials as sensors, theranostic, and drug delivery agents has become popular. The safety of these materials is being questioned because of their biological toxicity, such as inflammatory responses, cardiotoxicity, cytotoxicity, inhalation problems, etc., which can have a negative impact on the environment. This review paper focuses primarily on the toxicological effects of nanomaterials along with the mechanisms involved in cell interactions and the generation of reactive oxygen species by nanoparticles, which is the fundamental source of nanotoxicity. We also emphasize the greener synthesis of nanomaterials in biomedicine, as it is non-hazardous, feasible, and economical. The review articles shed light on the complexities of nanotoxicology in biosystems and the environment.
Collapse
Affiliation(s)
- Janani Saravanan
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Ayushi Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Sivadas Swathi Krishna
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Vidya Viswanad
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
8
|
Liang S, Xiao L, Chen T, Roa P, Cocco E, Peng Z, Yu L, Wu M, Liu J, Zhao X, Deng W, Wang X, Zhao C, Deng Y, Mai Y. Injectable Nanocomposite Hydrogels Improve Intraperitoneal Co-delivery of Chemotherapeutics and Immune Checkpoint Inhibitors for Enhanced Peritoneal Metastasis Therapy. ACS NANO 2024; 18:18963-18979. [PMID: 39004822 DOI: 10.1021/acsnano.4c02312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Intraperitoneal co-delivery of chemotherapeutic drugs (CDs) and immune checkpoint inhibitors (ICIs) brings hope to improve treatment outcomes in patients with peritoneal metastasis from ovarian cancer (OC). However, current intraperitoneal drug delivery systems face issues such as rapid drug clearance from lymphatic drainage, heterogeneous drug distribution, and uncontrolled release of therapeutic agents into the peritoneal cavity. Herein, we developed an injectable nanohydrogel by combining carboxymethyl chitosan (CMCS) with bioadhesive nanoparticles (BNPs) based on polylactic acid-hyperbranched polyglycerol. This system enables the codelivery of CD and ICI into the intraperitoneal space to extend drug retention. The nanohydrogel is formed by cross-linking of aldehyde groups on BNPs with amine groups on CMCS via reversible Schiff base bonds, with CD and ICI loaded separately into BNPs and CMCS network. BNP/CMCS nanohydrogel maintained the activity of the biomolecules and released drugs in a sustained manner over a 7 day period. The adhesive property, through the formation of Schiff bases with peritoneal tissues, confers BNPs with an extended residence time in the peritoneal cavity after being released from the nanohydrogel. In a mouse model, BNP/CMCS nanohydrogel loaded with paclitaxel (PTX) and anti-PD-1 antibodies (αPD-1) significantly suppressed peritoneal metastasis of OC compared to all other tested groups. In addition, no systemic toxicity of nanohydrogel-loaded PTX and αPD-1 was observed during the treatment, which supports potential translational applications of this delivery system.
Collapse
Affiliation(s)
- Shu Liang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen 518020, China
| | - Tian Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Paola Roa
- Department of Biochemistry and Molecular Biology/Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine, Miami, Florida 33136, United States
| | - Emiliano Cocco
- Department of Biochemistry and Molecular Biology/Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine, Miami, Florida 33136, United States
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Jie Liu
- ISCTE Business School, BRU-IUL, University Institute of Lisbon, Avenida das Armadas, Lisbon 1649-026, Portugal
| | - Xizhe Zhao
- Department of Chemistry, College of Staten Island, City University of New York, New York, New York 10314, United States
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Xiongjun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Chao Zhao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama 35487, United States
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| |
Collapse
|
9
|
Santhanes D, Zhang H, Wilkins A, John Aitken R, Gannon AL, Liang M. Engineering pH-sensitive dissolution of lipid-polymer nanoparticles by Eudragit integration impacts plasmid DNA (pDNA) transfection. Eur J Pharm Biopharm 2024; 199:114299. [PMID: 38643953 DOI: 10.1016/j.ejpb.2024.114299] [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: 11/25/2023] [Revised: 04/06/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Lipid-polymer nanoparticles offer a promising strategy for improving gene nanomedicines by combining the benefits of biocompatibility and stability associated with the individual systems. However, research to date has focused on poly-lactic-co-glycolic acid (PLGA) and resulted in inefficient transfection. In this study, biocompatible Eudragit constructs E100 and RS100 were formulated as lipid-polymer nanoparticles loaded with pDNA expressing red fluorescent protein (RFP) as a model therapeutic. Using a facile nanoprecipitation technique, a core-shell structure stabilised by lipid-polyethylene glycol (PEG) surfactant was produced and displayed resistance to ultracentrifugation. Both cationic polymers E100 (pH-sensitive dissolution at 5) and RS100 (pH-insensitive dissolution) produced 150-200 nm sized particles with a small positive surface charge (+3-5 mV) and high pDNA encapsulation efficiencies (EE) of 75-90%. The dissolution properties of the Eudragit polymers significantly impacted the biological performance in human embryonic kidney cells (HEK293T). Nanoparticles composed of polymer RS100 resulted in consistently high cell viability (80-100%), whereas polymer E100 demonstrated dose-dependent behaviour (20-90% cell viability). The low dissolution of polymer RS100 over the full pH range and the resulting nanoparticles failed to induce RFP expression in HEK293T cells. In contrast, polymer E100-constructed nanoparticles resulted in reproducible and gradually increasing RFP expression of 26-42% at 48-72 h. Intraperitoneal (IP) injection of the polymer E100-based nanoparticles in C57BL/6 mice resulted in targeted RFP expression in mouse testes with favourable biocompatibility one-week post-administration. These findings predicate Eudragit based lipid-polymer nanoparticles as a novel and effective carrier for nucleic acids, which could facilitate pre-clinical evaluation and translation of gene nanomedicines.
Collapse
Affiliation(s)
- Diviya Santhanes
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Huiming Zhang
- Electron Microscopy and X-ray Unit, Research and Innovation Division, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Alex Wilkins
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Robert John Aitken
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Anne-Louise Gannon
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
10
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
11
|
Liu Y, Ouyang Y, Yu L, Wang P, Peng Z, Liu H, Zhao S, Wang H, Zhou Z, Deng Y, Liu Y, Xie J. Novel approach for enhancing skin allograft survival by bioadhesive nanoparticles loaded with rapamycin. Int J Pharm 2024; 651:123742. [PMID: 38151102 DOI: 10.1016/j.ijpharm.2023.123742] [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: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
Skin graft rejection is a significant challenge in skin allografts for skin defects, particularly in extensive burn injury patients when autografts are insufficient. Enhancing the survival duration of allogeneic skin grafts can improve the success rate of subsequent autologous skin grafting, thereby promoting the therapeutic efficacy for wound healing. Rapamycin (Rapa), a potent immunosuppressant with favorable efficacy in organ transplantation, is limited by its systemic administration-associated toxicity and side effects. Therefore, addressing the short survival time of allogeneic skin grafts and minimizing the toxicity related to systemic application of immunosuppressive agents is an urgent requirement. Here, we present a topical formulation based on bioadhesive poly (lactic acid)-hyperbranched polyglycerol nanoparticles (BNPs) with surface-modified encapsulation of Rapamycin (Rapa/BNPs), applied for local immunosuppression in a murine model of allogeneic skin grafts. Our Rapa/BNPs significantly prolong nanoparticle retention, reduce infiltration of T lymphocytes and macrophages, decrease the level of pro-inflammatory cytokines and ultimately extend skin allograft survival with little systemic toxicity compared to free Rapa or Rapamycin-loaded non-bioadhesive nanoparticles (Rapa/NNPs) administration. In conclusion, Rapa/BNPs effectively deliver local immunosuppression and demonstrate potential for enhancing skin allograft survival while minimizing localized inflammation, thus potentially increasing patient survival rates for various types of skin defects.
Collapse
Affiliation(s)
- Yiling Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yaqi Ouyang
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Liu Yu
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Peng Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zhangwen Peng
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Hengdeng Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shixin Zhao
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Hanwen Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ziheng Zhou
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yang Deng
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China.
| | - Yang Liu
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China; Department of School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China.
| | - Julin Xie
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
12
|
Liang S, Xiao L, Fang Y, Chen T, Xie Y, Peng Z, Wu M, Liu Y, Xie J, Nie Y, Zhao X, Deng Y, Zhao C, Mai Y. A nanocomposite hydrogel for co-delivery of multiple anti-biofilm therapeutics to enhance the treatment of bacterial biofilm-related infections. Int J Pharm 2024; 649:123638. [PMID: 38008233 DOI: 10.1016/j.ijpharm.2023.123638] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
The characteristics of biofilms have exacerbated the issue of clinical antibiotic resistance, rendering it a pressing challenge in need of resolution. The combination of biofilm-dispersing agents and antibiotics can eliminate biofilms and promote healing synergistically in infected wounds. In this study, we developed a novel nanocomposite hydrogel (NC gel) comprised of the poly(lactic acid)-hyperbranched polyglycerol (PLA-HPG) based bioadhesive nanoparticles (BNPs) and a hydrophilic carboxymethyl chitosan (CS) network. The NC gel was designed to co-deliver two biofilm-dispersing agents (an NO-donor SNO, and an α-amylase Am) and an antibiotic, cefepime (Cef), utilizing a synergistic anti-biofilm mechanism in which Am loosens the matrix structure and NO promotes the release of biofilm bacteria via quorum sensing, and Cef kills bacteria. The drug-loaded NC gel (SNO/BNP/CS@Am-Cef) demonstrated sustained drug release, minimal cytotoxicity, and increased drug-bacterial interactions at the site of infection. When applied to mice infected with methicillin-resistant Staphylococcus aureus (MRSA) biofilms in vivo, SNO/BNP/CS@Am-Cef enhanced biofilm elimination and promoted wound healing compared to traditional antibiotic treatments. Our work demonstrates the feasibility of the co-delivery of biofilm-dispersing agents and antibiotics using the NC gel and presents a promising approach for the polytherapy of bacterial biofilm-related infections.
Collapse
Affiliation(s)
- Shu Liang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Yixuan Fang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Tian Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuan Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Julin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-sen University Foshan Hospital, Foshan 528000, China
| | - Xizhe Zhao
- Department of Chemistry, College of Staten Island, City University of New York, NY 10314, USA
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Chao Zhao
- Department of Chemical and Biological Engineering, Center for Convergent Biosciences and Medicine, Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL 35487, USA.
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
13
|
Khang M, Lee JH, Lee T, Suh HW, Lee S, Cavaliere A, Rushing A, Geraldo LH, Belitzky E, Rossano S, de Feyter HM, Shin K, Huttner A, Roussel MF, Thomas JL, Carson RE, Marquez-Nostra B, Bindra RS, Saltzman WM. Intrathecal delivery of nanoparticle PARP inhibitor to the cerebrospinal fluid for the treatment of metastatic medulloblastoma. Sci Transl Med 2023; 15:eadi1617. [PMID: 37910601 PMCID: PMC11078331 DOI: 10.1126/scitranslmed.adi1617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
The morbidity associated with pediatric medulloblastoma, in particular in patients who develop leptomeningeal metastases, remains high in the absence of effective therapies. Administration of substances directly into the cerebrospinal fluid (CSF) is one approach to circumvent the blood-brain barrier and focus delivery of drugs to the site of tumor. However, high rates of CSF turnover prevent adequate drug accumulation and lead to rapid systemic clearance and toxicity. Here, we show that PLA-HPG nanoparticles, made with a single-emulsion, solvent evaporation process, can encapsulate talazoparib, a PARP inhibitor (BMN-673). These degradable polymer nanoparticles improve the therapeutic index when delivered intrathecally and lead to sustained drug retention in the tumor as measured with PET imaging and fluorescence microscopy. We demonstrate that administration of these particles into the CSF, alone or in combination with systemically administered temozolomide, is a highly effective therapy for tumor regression and prevention of leptomeningeal spread in xenograft mouse models of medulloblastoma. These results provide a rationale for harnessing nanoparticles for the delivery of drugs limited by brain penetration and therapeutic index and demonstrate important advantages in tolerability and efficacy for encapsulated drugs delivered locoregionally.
Collapse
Affiliation(s)
- Minsoo Khang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Ju Hyun Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Teresa Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Supum Lee
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Alessandra Cavaliere
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Amy Rushing
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Luiz H. Geraldo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT 06510, USA
| | - Erika Belitzky
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Samantha Rossano
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Henk M. de Feyter
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Martine F. Roussel
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 38103, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| | - Richard E. Carson
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | | | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, CT 06520, USA
| | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT 06510, USA
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT 06511, USA
- Department of Dermatology, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
14
|
Han H, Chen L, Liang S, Lü J, Wu Y, Wang X, Xu F, Ge L, Xiao L. PLA-HPG based coating enhanced anti-biofilm and wound healing of Shikonin in MRSA-infected burn wound. Front Bioeng Biotechnol 2023; 11:1243525. [PMID: 37635995 PMCID: PMC10448828 DOI: 10.3389/fbioe.2023.1243525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Burn wounds are susceptible to bacterial infections, including Methicillin-resistant Staphylococcus aureus (MRSA), which typically form biofilms and exhibit drug resistance. They also have specific feature of abundant exudate, necessitating frequent drug administration. Shikonin (SKN) has been reported to reverse MRSA drug resistance and possesses anti-biofilm and wound healing properties, however, it suffers from drawbacks of low solubility and instability. In this study, we developed PLA-HPG based bioadhesive nanoparticles SKN/BNP, which demonstrated a drug loading capacity of about 3.6%, and exhibited sustained-release behavior of SKN. The aldehyde groups present on the surface of BNP improved the local adhesion of SKN/BNP both in vitro and in vivo, thereby reducing the frequency of drug dosing in exudate-rich burn wounds. BNP alone enhanced proliferation and migration of the fibroblast, while SKN/BNP promoted fibroblast proliferation and migration as well as angiogenesis. Due to its bioadhesive property, BNP directly interacted with biofilm and enhanced the efficacy of SKN against MRSA biofilm in vitro. In a mouse model of MRSA-infected burn wounds, SKN/BNP demonstrated improved anti-biofilm and wound healing efficiency. Overall, our findings suggest that SKN/BNP holds great promise as a novel and effective treatment option for clinical applications in MRSA-infected burn wounds.
Collapse
Affiliation(s)
- Huiyu Han
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Lianheng Chen
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Shu Liang
- Center Lab of Longhua Branch, Department of Infectious Disease, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Southern University of Science and Technology, Shenzhen, Guangdong, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Jiawei Lü
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Yashi Wu
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Xiongjun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Fei Xu
- Department of Plastic Surgery, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Lanlan Ge
- Center Lab of Longhua Branch, Department of Infectious Disease, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lingyun Xiao
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
- Center Lab of Longhua Branch, Department of Infectious Disease, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Xie Y, Ye J, Ouyang Y, Gong J, Li C, Deng Y, Mai Y, Liu Y, Deng W. Microneedle-Assisted Topical Delivery of Idebenone-Loaded Bioadhesive Nanoparticles Protect against UV-Induced Skin Damage. Biomedicines 2023; 11:1649. [PMID: 37371744 DOI: 10.3390/biomedicines11061649] [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: 04/30/2023] [Revised: 05/27/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Ultraviolet (UV) radiation can penetrate the basal layer of the skin and induce profound alterations in the underlying dermal tissues, including skin pigmentation, oxidative stress, photoaging, glycation, and skin cancer. Idebenone (IDB), an effective antioxidant that suppresses melanin biosynthesis and glycation, can protect the skin from UV-induced damage, accounting for its use in commercial anti-aging formulations. Ideally, IDB formulations should retain IDB inside the skin for a sufficient period, despite disturbances such as sweating or swimming. Herein, we present an IDB topical formulation based on Tris (tris(hydroxymethyl)-aminomethane)-modified bioadhesive nanoparticles (Tris-BNPs) and microneedle-assisted delivery. We found that Tris-BNPs loaded with IDB (IDB/Tris-BNPs) effectively reached the basal layer of the skin and were retained for at least 4 days with a slow and continuous drug release profile, unlike non-bioadhesive nanoparticles (NNPs) and bioadhesive nanoparticles (BNPs) of similar sizes (ranging from 120-142 nm) and zeta-potentials (above -20 mV), which experienced a significant reduction in concentration within 24 h. Notably, IDB/Tris-BNPs showed superior performance against UV-induced damage relative to IDB/NNPs and IDB/BNPs. This effect was demonstrated by lower levels of reactive oxygen species and advanced glycation end-products in skin tissues, as well as suppressed melanogenesis. Therefore, the proposed IDB delivery strategy provided long-term protective effects against UV-induced skin damage.
Collapse
Affiliation(s)
- Yuan Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Jingping Ye
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Jianing Gong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Chujie Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
16
|
Breusa S, Zilio S, Catania G, Bakrin N, Kryza D, Lollo G. Localized chemotherapy approaches and advanced drug delivery strategies: a step forward in the treatment of peritoneal carcinomatosis from ovarian cancer. Front Oncol 2023; 13:1125868. [PMID: 37287910 PMCID: PMC10242058 DOI: 10.3389/fonc.2023.1125868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Peritoneal carcinomatosis (PC) is a common outcome of epithelial ovarian carcinoma and is the leading cause of death for these patients. Tumor location, extent, peculiarities of the microenvironment, and the development of drug resistance are the main challenges that need to be addressed to improve therapeutic outcome. The development of new procedures such as HIPEC (Hyperthermic Intraperitoneal Chemotherapy) and PIPAC (Pressurized Intraperitoneal Aerosol Chemotherapy) have enabled locoregional delivery of chemotherapeutics, while the increasingly efficient design and development of advanced drug delivery micro and nanosystems are helping to promote tumor targeting and penetration and to reduce the side effects associated with systemic chemotherapy administration. The possibility of combining drug-loaded carriers with delivery via HIPEC and PIPAC represents a powerful tool to improve treatment efficacy, and this possibility has recently begun to be explored. This review will discuss the latest advances in the treatment of PC derived from ovarian cancer, with a focus on the potential of PIPAC and nanoparticles in terms of their application to develop new therapeutic strategies and future prospects.
Collapse
Affiliation(s)
- Silvia Breusa
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Institut PLAsCAN, Centre de Recherche en Cancérologie de Lyon, Institut national de santé et de la recherche médicale (INSERM) U1052-Centre National de la Recherche Scientifique - Unité Mixte de Recherche (CNRS UMR)5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Serena Zilio
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Sociétés d'Accélération du Transfert de Technologies (SATT) Ouest Valorisation, Rennes, France
| | - Giuseppina Catania
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| | - Naoual Bakrin
- Department of Surgical Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
- Centre pour l'Innovation en Cancérologie de Lyon (CICLY), Claude Bernard University Lyon 1, Lyon, France
| | - David Kryza
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Imthernat Plateform, Hospices Civils de Lyon, Lyon, France
| | - Giovanna Lollo
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| |
Collapse
|
17
|
Yu L, Yu M, Chen W, Sun S, Huang W, Wang T, Peng Z, Luo Z, Fang Y, Li Y, Deng Y, Wu M, Tao W. In Situ Separable Nanovaccines with Stealthy Bioadhesive Capability for Durable Cancer Immunotherapy. J Am Chem Soc 2023. [PMID: 36930579 DOI: 10.1021/jacs.2c12986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Because of tumor heterogeneity and the immunosuppressive tumor microenvironment, most cancer vaccines typically do not elicit robust antitumor immunological responses in clinical trials. In this paper, we report findings about a bioadhesive nanoparticle (BNP)-based separable cancer vaccine, FeSHK@B-ovalbumin (OVA), to target multi-epitope antigens and exert effective cancer immunotherapy. After the FeSHK@B-OVA "nanorocket" initiates the "satellite-rocket separation" procedure in the acidic tumor microenvironment, the FeSHK@B "launch vehicle" can amplify intracellular oxidative stress persistently. This procedure allows for bioadhesiveness-mediated prolonged drug retention within the tumor tissue and triggers the immunogenic death of tumor cells that transforms the primary tumors into antigen depots, which acts synergistically with the OVA "satellite" to trigger robust antigen-specific antitumor immunity. The cooperation of these two immunostimulants not only efficiently inhibits the primary tumor growth and provokes durable antigen-specific immune activation in vivo but also activates a long-term and robust immune memory effect to resist tumor rechallenge and metastasis. These results highlight the enormous potential of FeSHK@B-OVA to serve as an excellent therapeutic and prophylactic cancer nanovaccine. By leveraging the antigen depots in situ and the synergistic effect among multi-epitope antigens, such a nanovaccine strategy with stealthy bioadhesion may offer a straightforward and efficient approach to developing various cancer vaccines for different types of tumors.
Collapse
Affiliation(s)
- Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Wenxin Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Tianqi Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Zewen Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yixuan Fang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
18
|
Cheng X, Xie Q, Sun Y. Advances in nanomaterial-based targeted drug delivery systems. Front Bioeng Biotechnol 2023; 11:1177151. [PMID: 37122851 PMCID: PMC10133513 DOI: 10.3389/fbioe.2023.1177151] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Nanomaterial-based drug delivery systems (NBDDS) are widely used to improve the safety and therapeutic efficacy of encapsulated drugs due to their unique physicochemical and biological properties. By combining therapeutic drugs with nanoparticles using rational targeting pathways, nano-targeted delivery systems were created to overcome the main drawbacks of conventional drug treatment, including insufficient stability and solubility, lack of transmembrane transport, short circulation time, and undesirable toxic effects. Herein, we reviewed the recent developments in different targeting design strategies and therapeutic approaches employing various nanomaterial-based systems. We also discussed the challenges and perspectives of smart systems in precisely targeting different intravascular and extravascular diseases.
Collapse
|
19
|
Shin K, Suh HW, Grundler J, Lynn AY, Pothupitiya JU, Moscato ZM, Reschke M, Bracaglia LG, Piotrowski-Daspit AS, Saltzman WM. Polyglycerol and Poly(ethylene glycol) exhibit different effects on pharmacokinetics and antibody generation when grafted to nanoparticle surfaces. Biomaterials 2022; 287:121676. [PMID: 35849999 DOI: 10.1016/j.biomaterials.2022.121676] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/11/2022] [Accepted: 07/08/2022] [Indexed: 11/02/2022]
Abstract
Poly(ethylene glycol) (PEG) is widely employed for passivating nanoparticle (NP) surfaces to prolong blood circulation and enhance localization of NPs to target tissue. However, the immune response of PEGylated NPs-including anti-PEG antibody generation, accelerated blood clearance (ABC), and loss of delivery efficacy-is of some concern, especially for treatments that require repeat administrations. Although polyglycerol (PG), which has the same ethylene oxide backbone as PEG, has received attention as an alternative to PEG for NP coatings, the pharmacokinetic and immunogenic impact of PG has not been studied systematically. Here, linear PG, hyperbranched PG (hPG), and PEG-coated polylactide (PLA) NPs with varying surface densities were studied in parallel to determine the pharmacokinetics and immunogenicity of PG and hPG grafting, in comparison with PEG. We found that linear PG imparted the NPs a stealth property comparable to PEG, while hPG-grafted NPs needed a higher surface density to achieve the same pharmacokinetic impact. While linear PG-grafted NPs induced anti-PEG antibody production in mice, they exhibited minimal accelerated blood clearance (ABC) effects due to the poor interaction with anti-PEG immunoglobulin M (IgM). Further, we observed no anti-polymer IgM responses or ABC effects for hPG-grafted NPs.
Collapse
Affiliation(s)
- Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA.
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Julian Grundler
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Anna Y Lynn
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Jinal U Pothupitiya
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Zoe M Moscato
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Melanie Reschke
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
| | - Laura G Bracaglia
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | | | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA; Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
20
|
Mai Y, Ouyang Y, Yu M, Qin Y, Girardi M, Saltzman WM, Cocco E, Zhao C, Yu L, Jia Y, Xiao L, Dou L, Deng W, Liu Y, Xie J, Deng Y. Topical formulation based on disease-specific nanoparticles for single-dose cure of psoriasis. J Control Release 2022; 349:354-366. [PMID: 35817278 DOI: 10.1016/j.jconrel.2022.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/09/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
First-line treatments for mild to moderate psoriasis are typically topical formulations containing corticosteroids, however, the therapeutic efficacy of these formulations is compromised by limited penetration and skin retention. Even more challenging, off-target corticosteroids are known to adversely affect healthy skin, including induction of epidermal and dermal atrophy. Here, we report a nanoparticle-based topical formulation that cures psoriasis in a single dose, but leaves healthy skin intact. Specifically, we developed tris(hydroxymethyl)aminomethane-modified bioadhesive nanoparticles (Tris-BNPs) that exploit the high permeability characteristic of psoriasis to penetrate only psoriatic skin but not the healthy skin. Furthermore, as Tris-BNPs diffuse and penetrate into the epidermis, the Tris molecules slowly diffuse away, exposing the aldehyde groups of BNPs, which can bind to amine groups present within lesional skin, leading to long local retention of BNPs in lesions of psoriatic skin. The accumulated BNPs within lesions release corticosteroids over a ~ 3 day period to maintain local drug concentration above the therapeutic level. In addition to deeper penetration and longer retention compared with commercial psoriasis treatments, the topical applied Tris-BNPs were not affected by sweating, humidity, or active wiping due to their preferential accumulation between the stratum corneum and the basal cells of the epidermis. Overall, Tris-BNP as a topical formulation hold promise to overcome the limitations of current psoriasis treatment.
Collapse
Affiliation(s)
- Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yujia Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Michael Girardi
- Department of Dermatology, Yale University, 333 Cedar Street, New Haven, CT 06520, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, 55 Prospect Street, New Haven, CT 06511, USA
| | - Emiliano Cocco
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Liu Dou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China.
| | - Julin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
21
|
Mai Y, Ouyang Y, Qin Y, Jia C, McCoubrey LE, Basit AW, Nie Y, Jia Y, Yu L, Dou L, Deng W, Deng Y, Liu Y. Poly(lactic acid)-hyperbranched polyglycerol nanoparticles enhance bioadhesive treatment of esophageal disease and reduce systemic drug exposure. NANOSCALE 2022; 14:8418-8428. [PMID: 35639565 DOI: 10.1039/d2nr01846b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The effective treatment of esophageal disease represents a significant unmet clinical need, as existing treatments often lead to unnecessary systemic drug exposure and suboptimal concentrations at the disease site. Here, surface-modified bioadhesive poly(lactic acid)-hyperbranched polyglycerol nanoparticles (BNPs), with an average 100-200 nm diameter, were developed for local and sustained esophageal drug delivery. BNPs showed significantly higher adhesion and permeation into ex vivo human and rat esophageal tissue than non-adhesive nanoparticles (NNPs) and had longer residence times within the rat esophagus in vivo. Incubation with human esophagus (Het-1A) cells confirmed BNPs' biocompatibility at clinically relevant concentrations. In a rat model of achalasia, nifedipine-loaded BNPs significantly enhanced esophageal drug exposure, increased therapeutic efficacy, and reduced systemic drug exposure compared to NNPs and free drug. The safety of BNPs was demonstrated by an absence of intestinal, hepatic, and splenic toxicity following administration. This study is the first to demonstrate the efficacy of BNPs for esophageal drug delivery and highlight their potential for improving the lives of patients suffering with esophageal conditions.
Collapse
Affiliation(s)
- Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yujia Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Changchang Jia
- Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
| | - Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-sen University Foshan Hospital, Foshan, 528000, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Liu Dou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
22
|
Lin H, Jia Y, Kong X, Wang S, Liu X, Liu Y, Deng Y. In Vivo Evaluation of Cefuroxime Axetil-Loaded Bioadhesive Nanoparticles to Treat Haemophilus influenzae-Induced Otitis Media. Front Bioeng Biotechnol 2022; 10:884797. [PMID: 35573224 PMCID: PMC9099258 DOI: 10.3389/fbioe.2022.884797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
Otitis media (OM) is a common disease in children. One of the most common pathogens causing OM is non-typeable Haemophilus influenzae (NTHi). NTHi in the middle ear can be successfully eradicated by a regimen of oral antibiotics sustained for 7–10 days (e.g., cefuroxime axetil 250 mg/day for patients aged 3 months to 2 years and 500 mg/day for patients ages ≥2 years). However, lack of compliance is relevant to treatment failure or early relapse. In order to overcome these challenges, we have developed antibiotics-loaded bioadhesive nanoparticles (BNPs) that can adhere to the epidermis of the middle ear after local administration and significantly prolong the release time of antibiotics in the middle ear. Compared with oral administration of CA, local delivery of free antibiotic cefuroxime axetil (CA), and CA loaded non-bioadhesive nanoparticles (CA/NNPs), BNPs loaded with cefuroxime axetil (CA/BNPs) showed significantly longer retention time in the middle ear, resulting in continuous release of the drug and higher therapeutic efficacy against OM with only a single dosage. CA concentrations were maintained above the minimum inhibitory concentration (MIC) for NTHi throughout 7 days’ treatment. NTHi OM in a mouse model was successfully eradicated without causing tissue toxicity. CA/BNPs minimize systemic drug exposure through local administration, as demonstrated by undetectable levels in the blood.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Liu
- *Correspondence: Yang Liu, ; Yang Deng,
| | - Yang Deng
- *Correspondence: Yang Liu, ; Yang Deng,
| |
Collapse
|
23
|
Bioadhesive Nanoparticles for Local Drug Delivery. Int J Mol Sci 2022; 23:ijms23042370. [PMID: 35216484 PMCID: PMC8874699 DOI: 10.3390/ijms23042370] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 12/18/2022] Open
Abstract
Local drug delivery is an effective strategy for achieving direct and instant therapeutic effects. Current clinical treatments have fallen short and are limited by traditional technologies. Bioadhesive nanoparticles (NPs), however, may be a promising carrier for optimized local drug delivery, offering prolonged drug retention time and steadily maintained therapeutic concentrations. In addition, the possibility of clinical applications of this platform are abundant, as most polymers used for bioadhesion are both biodegradable and biocompatible. This review highlights the major advances in the investigations of polymer-based bioadhesive nanoparticles and their innumerable applications in local drug delivery.
Collapse
|
24
|
Targeting nanoparticles to malignant tumors. Biochim Biophys Acta Rev Cancer 2022; 1877:188703. [DOI: 10.1016/j.bbcan.2022.188703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/01/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022]
|
25
|
Yan X, Huang Z, Wu Y, Yu Z, Yang K, Chen Z, Wang W, Hu H, Wang Z. Sequential loading of inclusion complex/nanoparticles improves the gastric retention of Vladimiriae Radix essential oil to promote the protection of acute gastric mucosal injury. Int J Pharm 2021; 610:121234. [PMID: 34718092 DOI: 10.1016/j.ijpharm.2021.121234] [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: 06/16/2021] [Revised: 10/03/2021] [Accepted: 10/23/2021] [Indexed: 12/22/2022]
Abstract
The essential oil from Vladimiriae Radix (VEO) is a medicinal natural product with anti-ulcer activity. A novel gastroretentive drug delivery system was developed by preparing the hydroxypropyl-β-cyclodextrin (HP-β-CD) inclusion complex incorporated into chitosan nanoparticles (V-CD/NPs), to improve the bioavailability of VEO and its protective effect on gastric mucosa. The optimum preparation process of V-CD/NPs was obtained by Plackett-Burman and Box-Behnken response surface methodology. The resulting V-CD/NPs gained a suitable positive potential and small particle size, and showed stability in simulated gastric fluid, whose morphology and in vitro drug release profiles had a pH-sensitivity. Besides, V-CD/NPs was proved to strongly bind with mucin, and in vivo imaging revealed that it could be retained in the stomach for more than 8 h. The results of drug concentration in gastric tissues showed that the sequential loading of inclusion complex/nanoparticles promoted the local absorption of VEO in gastric tissues, which was favorable to reach the effective therapeutic concentration in the lesioned mucosa area. In comparison to VEO and V-CD, the callback effect of V-CD/NPs on 1L-1β, 1L-6, TNF-α, NF-κB, MDA and SOD was comparable to cimetidine, and V-CD/NPs outperformed in gastric mucosal protection. Therefore, the gastroretentive drug delivery system developed in our study effectively enhanced the anti-ulcer activity of VEO, which could be a promising strategy for the prevention and treatment of the acute gastric mucosal injury.
Collapse
Affiliation(s)
- Xiaomin Yan
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Zecheng Huang
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Yuyi Wu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ziwei Yu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ke Yang
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ziqiang Chen
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Wenjun Wang
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Huiling Hu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China.
| | - Zhanguo Wang
- Holistic Integrative Medicine Industry Collaborative Innovation Research Center, Qiang Medicine Standard Research Promotion Base and Collaborative Innovation Research Center, School of Preclinical Medicine, Chengdu University, Sichuan, Chengdu 610106, China.
| |
Collapse
|
26
|
Grundler J, Shin K, Suh HW, Zhong M, Saltzman WM. Surface Topography of Polyethylene Glycol Shell Nanoparticles Formed from Bottlebrush Block Copolymers Controls Interactions with Proteins and Cells. ACS NANO 2021; 15:16118-16129. [PMID: 34633171 PMCID: PMC8919421 DOI: 10.1021/acsnano.1c04835] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Although poly(ethylene glycol) (PEG) is commonly used in nanoparticle design, the impact of surface topography on nanoparticle performance in biomedical applications has received little attention, despite showing significant promise in the study of inorganic nanoparticles. Control of the surface topography of polymeric nanoparticles is a formidable challenge due to the limited conformational control of linear polymers that form the nanoparticle surface. In this work, we establish a straightforward method to precisely tailor the surface topography of PEGylated polymeric nanoparticles based on tuning the architecture of shape-persistent amphiphilic bottlebrush block copolymer (BBCP) building blocks. We demonstrate that nanoparticle formation and surface topography can be controlled by systematically changing the structural parameters of BBCP architecture. Furthermore, we reveal that the surface topography of PEGylated nanoparticles significantly affects their performance. In particular, the adsorption of a model protein and the uptake into HeLa cells were closely correlated to surface roughness and BBCP terminal PEG block brush width. Overall, our work elucidates the importance of surface topography in nanoparticle research as well as provides an approach to improve the performance of PEGylated nanoparticles.
Collapse
Affiliation(s)
- Julian Grundler
- Department of Chemistry, Yale University, New Haven, CT 06511 (USA)
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511 (USA)
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511 (USA)
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511 (USA)
| | - Mingjiang Zhong
- Department of Chemistry, Yale University, New Haven, CT 06511 (USA)
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06511 (USA)
| | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511 (USA)
| |
Collapse
|
27
|
Seo CH, Cui HS, Kim JB. Altered K Ca3.1 expression following burn injury and the therapeutic potential of TRAM-34 in post-burn hypertrophic scar formation. Transl Res 2021; 236:133-146. [PMID: 33905948 DOI: 10.1016/j.trsl.2021.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/18/2021] [Indexed: 01/16/2023]
Abstract
Hypertrophic scars are the most common post-burn complications characterized by fibroblast proliferation and excessive extracellular matrix deposition. The intermediate-conductance Ca2+-activated K+ channel (KCa3.1) mediates fibroblast activation, resulting in several fibrotic diseases; however, this channel's role in the formation of post-burn hypertrophic skin scars remains unknown. Herein, we investigated the role of KCa3.1 and the therapeutic potential of TRAM-34, a selective inhibitor of KCa3.1, in hypertrophic skin scar formation following burn injury. Cytosolic Ca2+ levels, the expression of KCa3.1 and hypertrophic markers, and the proliferation of skin fibroblasts obtained directly from patients with third-degree burns who consequently developed post-burn hypertrophic scars were assessed. The anti-fibrotic effect of KCa3.1 inhibition by TRAM-34 was evaluated in vitro (fibroblasts) and in vivo (mouse burn models). Fibroblasts from burn wounds exhibited remarkably higher levels of cytosolic Ca2+ than normal cells. KCa3.1 expression was markedly higher in the membrane fraction but lower in the cytosolic fraction of burn wound fibroblasts than in normal cells. Selective inhibition of KCa3.1 by TRAM-34 markedly reduced not only the proliferation of burn wound fibroblasts but also the expression of hypertrophic markers in these cells. Anti-scarring molecular, histological, and visual effects of TRAM-34 were confirmed in murine burn models. Altered subcellular expression of KCa3.1 is a novel mechanism underlying the cellular response to burn injury. Our results suggest that selective inhibition of KCa3.1 by TRAM-34 has therapeutic potential against post-burn hypertrophic scar formation.
Collapse
Affiliation(s)
- Cheong Hoon Seo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Hui Song Cui
- Burn Institute, Hangang Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - June-Bum Kim
- Department of Pediatrics, Hangang Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
28
|
βIII-tubulin overexpression in cancer: Causes, consequences, and potential therapies. Biochim Biophys Acta Rev Cancer 2021; 1876:188607. [PMID: 34364992 DOI: 10.1016/j.bbcan.2021.188607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/21/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022]
Abstract
Class III β-tubulin (βIII-tubulin) is frequently overexpressed in human tumors and is associated with resistance to microtubule-targeting agents, tumor aggressiveness, and poor patient outcome. Understanding the mechanisms regulating βIII-tubulin expression and the varied functions βIII-tubulin may have in different cancers is vital to assess the prognostic value of this protein and to develop strategies to enhance therapeutic benefits in βIII-tubulin overexpressing tumors. Here we gather all the available evidence regarding the clinical implications of βIII-tubulin overexpression in cancer, describe factors that regulate βIII-tubulin expression, and discuss current understanding of the mechanisms underlying βIII-tubulin-mediated resistance to microtubule-targeting agents and tumor aggressiveness. Finally, we provide an overview of emerging therapeutic strategies to target tumors that overexpress βIII-tubulin.
Collapse
|
29
|
Santezi C, Reina BD, de Annunzio SR, Calixto G, Chorilli M, Dovigo LN. Photodynamic potential of curcumin in bioadhesive formulations: Optical characteristics and antimicrobial effect against biofilms. Photodiagnosis Photodyn Ther 2021; 35:102416. [PMID: 34197966 DOI: 10.1016/j.pdpdt.2021.102416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Although Curcumin (CUR) has great potential as a photosensitizer, the low solubility in water impairs its clinical performance in photodynamic inactivation (PDI). This study sought to establish an effective antimicrobial protocol for PDI using CUR in three different bioadhesive formulations. METHODS A CUR-loaded chitosan hydrogel with a poloxamer (CUR-CHIH), a CUR-loaded liquid crystal precursor system (CUR-LCP), a CUR-loaded microemulsion (CUR-ME), and CUR in dimethylsulfoxide (DMSO) solution (CUR-S; control formulation) were tested against in vitro and in situ oral biofilms. The optical properties of each formulation were evaluated. RESULTS All of the formulations exhibited lower absorbance than CUR-S; however, the CUR-LCP curve bore the highest resemblance. The CUR present in all formulations was completely degraded after 15 min of illumination. In vitro experiments showed that CUR-S was the only formulation able to significantly reduce biofilm viability of Candida albicans and Lactobacillus casei when compared to the negative control (no PDI); the amount of reduction obtained was 1.8 and 3.7 log (CFU/mL) for C. albicans and L. casei, respectively. There was a significant reduction on the viability of Streptococcus mutans biofilms when CUR-S and CUR-LCP were applied (approximately 3.5 and 1.6 log [CFU/mL], respectively). In situ testing showed antimicrobial efficacy against S. mutans and general microorganisms. CONCLUSIONS Although the evaluated protocols has not been effective to all of the evaluated microorganisms, PDI showed potential against dental biofilms and evidence that the phototoxic effects of CUR have a high relation with the type of formulation in which it is loaded.
Collapse
Affiliation(s)
- Carolina Santezi
- Independent researcher at the moment of the submission (unaffiliated researcher)
| | - Bárbara Donadon Reina
- Department of Social Dentistry, School of Dentistry, UNESP - São Paulo State University, Araraquara, São Paulo, Brazil
| | - Sarah Raquel de Annunzio
- Department of Clinical Analysis, School of Pharmacy, UNESP - São Paulo State University, Araraquara, São Paulo, Brazil
| | - Giovana Calixto
- Department of Biosciences, Piracicaba Dental School, UNICAMP, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmacy, UNESP - São Paulo State University, Araraquara, São Paulo, Brazil
| | - Lívia Nordi Dovigo
- Department of Social Dentistry, School of Dentistry, UNESP - São Paulo State University, Araraquara, São Paulo, Brazil.
| |
Collapse
|
30
|
Wang F, Multhoff G. Repurposing Cannabidiol as a Potential Drug Candidate for Anti-Tumor Therapies. Biomolecules 2021; 11:biom11040582. [PMID: 33921049 PMCID: PMC8071421 DOI: 10.3390/biom11040582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 01/09/2023] Open
Abstract
In recent years, evidence has accumulated that cannabinoids-especially the non-psychoactive compound, cannabidiol (CBD)-possess promising medical and pharmacological activities that might qualify them as potential anti-tumor drugs. This review is based on multiple studies summarizing different mechanisms for how CBD can target tumor cells including cannabinoid receptors or other constituents of the endocannabinoid system, and their complex activation of biological systems that results in the inhibition of tumor growth. CBD also participates in anti-inflammatory activities which are related to tumor progression, as demonstrated in preclinical models. Although the numbers of clinical trials and tested tumor entities are limited, there is clear evidence that CBD has anti-tumor efficacy and is well tolerated in human cancer patients. In summary, it appears that CBD has potential as a neoadjuvant and/or adjuvant drug in therapy for cancer.
Collapse
Affiliation(s)
- Fei Wang
- Radiation-Immuno Oncology Group, TranslaTUM—Central Institute for Translational Cancer Research, Klinikum rechts der Isar, TU München, Einsteinstr. 25, 81675 Munich, Germany;
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
- Correspondence: ; Tel.: +49-89-4140-4514; Fax: +49-89-4140-4299
| | - Gabriele Multhoff
- Radiation-Immuno Oncology Group, TranslaTUM—Central Institute for Translational Cancer Research, Klinikum rechts der Isar, TU München, Einsteinstr. 25, 81675 Munich, Germany;
- Department of Radiation Oncology, Klinikum rechts der Isar, TU München, 81675 Munich, Germany
| |
Collapse
|
31
|
Wang P, Qu X, Che X, Luo Q, Tang X, Liu Y. Pharmaceutical strategies in improving anti-tumour efficacy and safety of intraperitoneal therapy for peritoneal metastasis. Expert Opin Drug Deliv 2021; 18:1193-1210. [PMID: 33682562 DOI: 10.1080/17425247.2021.1896493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: In selected patients with limited peritoneal metastasis (PM), favorable tumor biology, and a good clinical condition, there is an indication for combination of cytoreductive surgery (CRS) and subsequent intravenous (IV) or intraperitoneal (IP) chemotherapy. Compared with IV injection, IP therapy can achieve a high drug concentration within the peritoneal cavity with low systemic toxicity, however, the clinical application of IP chemotherapy is limited by the related abdominal pain, infection, and intolerance.Areas covered:To improve the anti-tumor efficacy and safety of IP therapy, various pharmaceutical strategies have been developed and show promising potential. This review discusses the specialized modification of traditional drug delivery systems and demonstrates the preparation of customized drug carriers for IP therapy, including chemotherapy and gene therapy. IP therapy has important clinical significance in the treatment of PM using novel anti-tumor agents as well as conventional drugs in new applications.Expert opinion: Although IP therapy exhibits good performance both in mouse models and in patients with PM in clinical trials, its clinical application remains limited due to the serious side effects and low acceptability. Further investigations, including pharmaceutical strategies, are needed to develop potential IP therapy, focusing on the efficacy and safety thereof.
Collapse
Affiliation(s)
- Puxiu Wang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, China.,Liaoning Province Clinical Research Center for Cancer, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, China.,Liaoning Province Clinical Research Center for Cancer, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xing Tang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, China.,Liaoning Province Clinical Research Center for Cancer, China
| |
Collapse
|
32
|
Kuper CF, Pieters RHH, van Bilsen JHM. Nanomaterials and the Serosal Immune System in the Thoracic and Peritoneal Cavities. Int J Mol Sci 2021; 22:ijms22052610. [PMID: 33807632 PMCID: PMC7961545 DOI: 10.3390/ijms22052610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/23/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
The thoracic and peritoneal cavities are lined by serous membranes and are home of the serosal immune system. This immune system fuses innate and adaptive immunity, to maintain local homeostasis and repair local tissue damage, and to cooperate closely with the mucosal immune system. Innate lymphoid cells (ILCs) are found abundantly in the thoracic and peritoneal cavities, and they are crucial in first defense against pathogenic viruses and bacteria. Nanomaterials (NMs) can enter the cavities intentionally for medical purposes, or unintentionally following environmental exposure; subsequent serosal inflammation and cancer (mesothelioma) has gained significant interest. However, reports on adverse effects of NM on ILCs and other components of the serosal immune system are scarce or even lacking. As ILCs are crucial in the first defense against pathogenic viruses and bacteria, it is possible that serosal exposure to NM may lead to a reduced resistance against pathogens. Additionally, affected serosal lymphoid tissues and cells may disturb adipose tissue homeostasis. This review aims to provide insight into key effects of NM on the serosal immune system.
Collapse
Affiliation(s)
- C. Frieke Kuper
- Consultant, Haagstraat 13, 3581 SW Utrecht, The Netherlands
- Correspondence: (C.F.K.); (J.H.M.v.B.)
| | - Raymond H. H. Pieters
- Immunotoxicology, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands;
- Innovative Testing in Life Sciences & Chemistry, Research Centre for Healthy and Sustainable Living, University of Applied Sciences Utrecht, Padualaan 97, 3584 CH Utrecht, The Netherlands
| | - Jolanda H. M. van Bilsen
- Department for Risk Analysis for Products in Development, Netherlands Organization for Applied Scientific Research (TNO), Princetonlaan 6, 3584 CB Utrecht, The Netherlands
- Correspondence: (C.F.K.); (J.H.M.v.B.)
| |
Collapse
|
33
|
Nonsurgical treatment of skin cancer with local delivery of bioadhesive nanoparticles. Proc Natl Acad Sci U S A 2021; 118:2020575118. [PMID: 33526595 DOI: 10.1073/pnas.2020575118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Keratinocyte-derived carcinomas, including squamous cell carcinoma (SCC), comprise the most common malignancies. Surgical excision is the therapeutic standard but is not always clinically feasible, and currently available alternatives are limited to superficial tumors. To address the need for a nonsurgical treatment for nodular skin cancers like SCC, we developed a bioadhesive nanoparticle (BNP) drug delivery system composed of biodegradable polymer, poly(lactic acid)-hyperbranched polyglycerol (PLA-HPG), encapsulating camptothecin (CPT). Nanoparticles (NPs) of PLA-HPG are nonadhesive NPs (NNPs), which are stealthy in their native state, but we have previously shown that conversion of the vicinal diols of HPG to aldehydes conferred NPs the ability to form strong covalent bonds with amine-rich surfaces. Herein, we show that these BNPs have significantly enhanced binding to SCC tumor cell surfaces and matrix proteins, thereby significantly enhancing the therapeutic efficacy of intratumoral drug delivery. Tumor injection of BNP-CPT resulted in tumor retention of CPT at ∼50% at 10 d postinjection, while CPT was undetectable in NNP-CPT or free (intralipid) CPT-injected tumors at that time. BNP-CPT also significantly reduced tumor burden, with a portion (∼20%) of BNP-CPT-treated established tumors showing histologic cure. Larger, more fully established PDV SCC tumors treated with a combination of BNP-CPT and immunostimulating CpG oligodeoxynucleotides exhibited enhanced survival relative to controls, revealing the potential for BNP delivery to be used along with local tumor immunotherapy. Taken together, these results indicate that percutaneous delivery of a chemotherapeutic agent via BNPs, with or without adjuvant immunostimulation, represents a viable, nonsurgical alternative for treating cutaneous malignancy.
Collapse
|
34
|
Bortot B, Mongiat M, Valencic E, Dal Monego S, Licastro D, Crosera M, Adami G, Rampazzo E, Ricci G, Romano F, Severini GM, Biffi S. Nanotechnology-Based Cisplatin Intracellular Delivery to Enhance Chemo-Sensitivity of Ovarian Cancer. Int J Nanomedicine 2020; 15:4793-4810. [PMID: 32764921 PMCID: PMC7368240 DOI: 10.2147/ijn.s247114] [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] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Background Platinum resistance is a major challenge in the management of ovarian cancer. Even low levels of acquired resistance at the cellular level lead to impaired response to cisplatin. In ovarian cancer intraperitoneal therapy, nanoparticle formulation can improve the cisplatin’s pharmacokinetics and safety profile. Purpose This work aimed to investigate the chemo-sensitivity of ovarian cancer SKOV3 cells upon short-term (72h) single treatment of cisplatin and cisplatin-loaded biodegradable nanoparticles (Cis-NP). The aim was then to determine the therapeutic properties of Cis-NP in vivo using a SKOV3-luc cells’ xenograft model in mice. Methods Cell cytotoxicity was assessed after the exposure of the cell culture to cisplatin or Cis-NP. The effect of treatments on EMT and CSC-like phenotype was studied by analyzing a panel of markers by flow cytometry. Intracellular platinum concentration was determined by inductively coupled plasma mass spectrometry (ICS-MS), and gene expression was evaluated by RNAseq analysis. The efficacy of intraperitoneal chemotherapy was evaluated in a SKOV3-luc cells’ xenograft model in mice, through a combination of bioluminescence imaging, histological, and immunohistochemical analyses. Results We observed in vitro that short-term treatment of cisplatin has a critical role in determining the potential induction of chemoresistance, and a nanotechnology-based drug delivery system can modulate it. The RNAseq analysis underlines a protective effect of nanoparticle system according to their ability to down-regulate several genes involved in chemoresistance, cell proliferation, and apoptosis. The highest intracellular platinum concentration obtained with Cis-NP treatment significantly improved the efficacy. Consistent with in vitro results, we found that Cis-NP treatment in vivo can significantly reduce tumor burden and aggressiveness compared to the free drug. Conclusion Nanoparticle-mediated cisplatin delivery may serve as an intracellular depot impacting the cisplatin pharmacodynamic performance at cellular levels. These features may contribute to improving the drawbacks of conventional intraperitoneal therapy, and therefore will require further investigations in vivo.
Collapse
Affiliation(s)
- Barbara Bortot
- Department of Medical Genetics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro Di Riferimento Oncologico Di Aviano (CRO) IRCCS, Aviano, Italy
| | - Erica Valencic
- Department of Pediatrics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Simeone Dal Monego
- ARGO Open Lab Platform for Genome Sequencing, AREA Science Park, Trieste, Italy
| | - Danilo Licastro
- ARGO Open Lab Platform for Genome Sequencing, AREA Science Park, Trieste, Italy
| | - Matteo Crosera
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Gianpiero Adami
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Enrico Rampazzo
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Giuseppe Ricci
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Federico Romano
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giovanni Maria Severini
- Department of Medical Genetics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Stefania Biffi
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
35
|
Alavi S, Haeri A, Mahlooji I, Dadashzadeh S. Tuning the Physicochemical Characteristics of Particle-Based Carriers for Intraperitoneal Local Chemotherapy. Pharm Res 2020; 37:119. [DOI: 10.1007/s11095-020-02818-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
|
36
|
Fraguas-Sánchez AI, Torres-Suárez AI, Cohen M, Delie F, Bastida-Ruiz D, Yart L, Martin-Sabroso C, Fernández-Carballido A. PLGA Nanoparticles for the Intraperitoneal Administration of CBD in the Treatment of Ovarian Cancer: In Vitro and In Ovo Assessment. Pharmaceutics 2020; 12:pharmaceutics12050439. [PMID: 32397428 PMCID: PMC7285054 DOI: 10.3390/pharmaceutics12050439] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
The intraperitoneal administration of chemotherapeutics has emerged as a potential route in ovarian cancer treatment. Nanoparticles as carriers for these agents could be interesting by increasing the retention of chemotherapeutics within the peritoneal cavity. Moreover, nanoparticles could be internalised by cancer cells and let the drug release near the biological target, which could increase the anticancer efficacy. Cannabidiol (CBD), the main nonpsychotropic cannabinoid, appears as a potential anticancer drug. The aim of this work was to develop polymer nanoparticles as CBD carriers capable of being internalised by ovarian cancer cells. The drug-loaded nanoparticles (CBD-NPs) exhibited a spherical shape, a particle size around 240 nm and a negative zeta potential (-16.6 ± 1.2 mV). The encapsulation efficiency was high, with values above 95%. A controlled CBD release for 96 h was achieved. Nanoparticle internalisation in SKOV-3 epithelial ovarian cancer cells mainly occurred between 2 and 4 h of incubation. CBD antiproliferative activity in ovarian cancer cells was preserved after encapsulation. In fact, CBD-NPs showed a lower IC50 values than CBD in solution. Both CBD in solution and CBD-NPs induced the expression of PARP, indicating the onset of apoptosis. In SKOV-3-derived tumours formed in the chick embryo model, a slightly higher-although not statistically significant-tumour growth inhibition was observed with CBD-NPs compared to CBD in solution. To sum up, poly-lactic-co-glycolic acid (PLGA) nanoparticles could be a good strategy to deliver CBD intraperitoneally for ovarian cancer treatment.
Collapse
Affiliation(s)
- Ana I. Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., 28040 Madrid, Spain; (A.I.F.-S.); (A.I.T.-S.); (C.M.-S.)
| | - Ana I. Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., 28040 Madrid, Spain; (A.I.F.-S.); (A.I.T.-S.); (C.M.-S.)
- Institute of Industrial Pharmacy, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Marie Cohen
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; (M.C.); (D.B.-R.); (L.Y.)
| | - Florence Delie
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland;
| | - Daniel Bastida-Ruiz
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; (M.C.); (D.B.-R.); (L.Y.)
| | - Lucile Yart
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; (M.C.); (D.B.-R.); (L.Y.)
| | - Cristina Martin-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., 28040 Madrid, Spain; (A.I.F.-S.); (A.I.T.-S.); (C.M.-S.)
- Institute of Industrial Pharmacy, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ana Fernández-Carballido
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., 28040 Madrid, Spain; (A.I.F.-S.); (A.I.T.-S.); (C.M.-S.)
- Institute of Industrial Pharmacy, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramón y Cajal s/n., Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-913941741
| |
Collapse
|
37
|
Correa S, Boehnke N, Barberio AE, Deiss-Yehiely E, Shi A, Oberlton B, Smith SG, Zervantonakis I, Dreaden EC, Hammond PT. Tuning Nanoparticle Interactions with Ovarian Cancer through Layer-by-Layer Modification of Surface Chemistry. ACS NANO 2020; 14:2224-2237. [PMID: 31971772 PMCID: PMC7062411 DOI: 10.1021/acsnano.9b09213] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanoparticle surface chemistry is a fundamental engineering parameter that governs tumor-targeting activity. Electrostatic assembly generates controlled polyelectrolyte complexes through the process of adsorption and charge overcompensation utilizing synthetic polyions and natural biomacromolecules; it can yield films with distinctive hydration, charge, and presentation of functional groups. Here, we used electrostatic layer-by-layer (LbL) assembly to screen 10 different surface chemistries for their ability to preferentially target human ovarian cancer in vitro. Our screen identified that poly-l-aspartate, poly-l-glutamate, and hyaluronate-coated LbL nanoparticles have striking specificity for ovarian cancer, while sulfated poly(β-cyclodextrin) nanoparticles target noncancerous stromal cells. We validated top candidates for tumor-homing ability with a murine model of metastatic disease and with patient-derived ovarian cancer spheroids. Nanoparticle surface chemistry also influenced subcellular trafficking, indicating strategies to target the cell membrane, caveolae, and perinuclear vesicles. Our results confirm LbL is a powerful tool to systematically engineer nanoparticles and achieve specific targeting.
Collapse
Affiliation(s)
- Santiago Correa
- Department of Biological Engineering , Massachusetts Institute of Technology , 21 Ames Street , Cambridge , Massachusetts 02142 , United States
| | - Natalie Boehnke
- Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02142 , United States
| | - Antonio E Barberio
- Department of Chemical Engineering , Massachusetts Institute of Technology , 25 Ames Street , Cambridge , Massachusetts 02142 , United States
| | - Elad Deiss-Yehiely
- Department of Materials Science and Engineering , Massachusetts Institute of Technology , 183 Memorial Drive , Cambridge , Massachusetts 02142 , United States
| | - Aria Shi
- Department of Biological Engineering , Massachusetts Institute of Technology , 21 Ames Street , Cambridge , Massachusetts 02142 , United States
| | - Benjamin Oberlton
- Department of Biological Engineering , Massachusetts Institute of Technology , 21 Ames Street , Cambridge , Massachusetts 02142 , United States
| | - Sean G Smith
- Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02142 , United States
- Department of Chemical Engineering , Massachusetts Institute of Technology , 25 Ames Street , Cambridge , Massachusetts 02142 , United States
| | - Ioannis Zervantonakis
- Department of Cell Biology, Ludwig Center at Harvard , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Erik C Dreaden
- Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02142 , United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02142 , United States
- Department of Chemical Engineering , Massachusetts Institute of Technology , 25 Ames Street , Cambridge , Massachusetts 02142 , United States
- Institute for Soldier Nanotechnologies , Massachusetts Institute of Technology , 500 Technology Square , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
38
|
Liu Z, Zhu X, Zhu T, Tang R. Evaluation of a Biocomposite Mesh Modified with Decellularized Human Amniotic Membrane for Intraperitoneal Onlay Mesh Repair. ACS OMEGA 2020; 5:3550-3562. [PMID: 32118170 PMCID: PMC7045508 DOI: 10.1021/acsomega.9b03866] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/31/2020] [Indexed: 06/10/2023]
Abstract
Various materials and approaches have been used to optimize the biocompatibility of mesh to reduce the implant-induced host response in intraperitoneal onlay mesh (IPOM) repair. Ineffective host integration, limited resistance to contamination, and untargeted administration hinder the wider application of the currently available clinical options. In this study, human amniotic membrane (HAM) was decellularized, fully characterized, and compared with porcine small intestinal submucosa (SIS) in terms of its structure, components, and bioactivity. In an in vivo study, HAM was reinforced with silk fibroin (SF) membrane, which was fabricated as a biodegradable submicroscale template by electrospinning, to construct a bilayer composite mesh. The independent SF membrane, associated with HAM and SIS, was evaluated for tissue remodeling in vitro. The HAM-SF and SIS meshes were then characterized morphologically and implanted intraperitoneally into Sprague-Dawley rats for 28 days for macroscopic investigation of their integration into the host via interactions of regulatory factors. After decellularization, HAM formed a bioagent-rich collagen-based acellular structure. HAM was superior to SIS in concurrently suppressing the expression of transforming growth factor β1 (TGF-β1) and proangiogenic proliferation. When HAM, SF, and SIS were used as regenerative scaffolds, they showed qualified biocompatibility, cell infiltration, and degradation in vitro. Comparatively, macroscopic observation after implantation indicated that HAM-SF induced less-intensive intraperitoneal adhesion and weaker inflammatory responses at the interface but greater angiogenesis in the explant than SIS. Analysis of the expression of regulatory factors showed a greater quantity of hepatocyte growth factor (HGF) in HAM, which partly inhibited the expression of TGF-β1 and promoted vascular endothelial growth factor (VEGF)-induced angiogenesis. This bioactive interaction appeared to be responsible for the better host integration, making HAM more biocompatible than SIS in IPOM repair. When combined with SF, HAM displayed similar mechanical properties to SIS. In conclusion, HAM displayed better bioactivity and biocompatibility than SIS. After its reinforcement with SF, HAM-SF is a promising biocomposite mesh for IPOM repair.
Collapse
Affiliation(s)
- Zhengni Liu
- Department
of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, Tongji University, 150 Ji Mo Road, Shanghai 200120, P. R. China
| | - Xiaoqiang Zhu
- Department
of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, Tongji University, 150 Ji Mo Road, Shanghai 200120, P. R. China
| | - Tonghe Zhu
- Department
of Sports Medicine, Medicine and Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s
Hospital, No. 600 Yishan Road, Shanghai 200233, P. R. China
| | - Rui Tang
- Department
of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, Tongji University, 150 Ji Mo Road, Shanghai 200120, P. R. China
| |
Collapse
|
39
|
Xie Y, Hang Y, Wang Y, Sleightholm R, Prajapati DR, Bader J, Yu A, Tang W, Jaramillo L, Li J, Singh RK, Oupický D. Stromal Modulation and Treatment of Metastatic Pancreatic Cancer with Local Intraperitoneal Triple miRNA/siRNA Nanotherapy. ACS NANO 2020; 14:255-271. [PMID: 31927946 PMCID: PMC7041410 DOI: 10.1021/acsnano.9b03978] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Nanomedicines achieve tumor-targeted delivery mainly through enhanced permeability and retention (EPR) effect following intravenous (IV) administration. Unfortunately, the EPR effect is severely compromised in pancreatic cancer due to hypovascularity and dense desmoplastic stroma. Intraperitoneal (IP) administration may be an effective EPR-independent local delivery approach to target peritoneal tumors. Besides improved delivery, effective combination delivery strategies are needed to improve pancreatic cancer therapy by targeting both cancer cells and cellular interactions within the tumor stroma. Here, we described simple cholesterol-modified polymeric CXCR4 antagonist (PCX) nanoparticles (to block cancer-stroma interactions) for codelivery of anti-miR-210 (to inactivate stroma-producing pancreatic stellate cells (PSCs)) and siKRASG12D (to kill pancreatic cancer cells). IP administration delivered the nanoparticles to an orthotopic syngeneic pancreatic tumors as a result of preferential localization to the tumors and metastases with disrupted mesothelium and effective tumor penetration. The local IP delivery resulted in nearly 15-fold higher tumor accumulation than delivery by IV injection. Through antagonism of CXCR4 and downregulation of miR-210/KRASG12D, the triple-action nanoparticles favorably modulated desmoplastic tumor microenvironment via inactivating PSCs and promoting the infiltration of cytotoxic T cells. The combined therapy displayed improved therapeutic effect when compared with individual therapies as documented by the delayed tumor growth, depletion of stroma, reduction of immunosuppression, inhibition of metastasis, and prolonged survival. Overall, we present data that a local IP delivery of a miRNA/siRNA combination holds the potential to improve pancreatic cancer therapy.
Collapse
Affiliation(s)
- Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Yazhe Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Richard Sleightholm
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Dipakkumar R Prajapati
- Department of Pathology and Microbiology , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Johannes Bader
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy , Ludwig-Maximilians-Universität München , 81337 Munich , Germany
| | - Ao Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Lee Jaramillo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
- Bohemica Pharmaceuticals, LLC , La Vista , Nebraska 68128 , United States
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Rakesh K Singh
- Department of Pathology and Microbiology , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| |
Collapse
|
40
|
Cao F, Zhang L, Wang H, You Y, Wang Y, Gao N, Ren J, Qu X. Defect-Rich Adhesive Nanozymes as Efficient Antibiotics for Enhanced Bacterial Inhibition. Angew Chem Int Ed Engl 2019; 58:16236-16242. [PMID: 31456332 DOI: 10.1002/anie.201908289] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/18/2019] [Indexed: 12/31/2022]
Abstract
Nanozymes have emerged as a new generation of antibiotics with exciting broad-spectrum antimicrobial properties and negligible biotoxicities. However, their antibacterial efficacies are unsatisfactory due to their inability to trap bacteria and their low catalytic activity. Herein, we report nanozymes with rough surfaces and defect-rich active edges. The rough surface increases bacterial adhesion and the defect-rich edges exhibit higher intrinsic peroxidase-like activity compared to pristine nanozymes due to their lower adsorption energies of H2 O2 and desorption energy of OH*, as well as the larger exothermic process for the whole reaction. This was demonstrated using drug-resistant Gram-negative Escherichia coli and Gram-positive Staphylococcus aureus in vitro and in vivo. This strategy can be used to engineer nanozymes with enhanced antibacterial function and will pave a new way for the development of alternative antibiotics.
Collapse
Affiliation(s)
- Fangfang Cao
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Lu Zhang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Yawen You
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Ying Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China
| | - Nan Gao
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin, 130022, P. R. China
| |
Collapse
|
41
|
Cao F, Zhang L, Wang H, You Y, Wang Y, Gao N, Ren J, Qu X. Defect‐Rich Adhesive Nanozymes as Efficient Antibiotics for Enhanced Bacterial Inhibition. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201908289] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Fangfang Cao
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230029 P. R. China
| | - Lu Zhang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230029 P. R. China
| | - Yawen You
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230029 P. R. China
| | - Ying Wang
- State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
| | - Nan Gao
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of Science Changchun Jilin 130022 P. R. China
| |
Collapse
|
42
|
Zhao X, Liu X, Zhang P, Liu Y, Ran W, Cai Y, Wang J, Zhai Y, Wang G, Ding Y, Li Y. Injectable peptide hydrogel as intraperitoneal triptolide depot for the treatment of orthotopic hepatocellular carcinoma. Acta Pharm Sin B 2019; 9:1050-1060. [PMID: 31649853 PMCID: PMC6804453 DOI: 10.1016/j.apsb.2019.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy is among the limited choices approved for the treatment of hepatocellular carcinoma (HCC) at intermediate and advanced stages. Preferential and prolonged drug exposure in diseased sites is required to maximize the therapeutic index of the drug. Here, we report an injectable supramolecular peptide hydrogel as an intraperitoneal depot for localized and sustained release of triptolide for the treatment of orthotopic HCC. We chose peptide amphiphile C16-GNNQQNYKD-OH-based nanofibers as gelators and carriers for triptolide. Sustained triptolide release from the hydrogel was achieved over 14 days in vitro, with higher accumulation in and cytotoxicity against human HCC Bel-7402 in comparison with L-02 fetal hepatocytes. After intraperitoneal injection, the hydrogel showed prolonged retention over 13 days and preferential accumulation in the liver, realizing HCC growth inhibition by 99.7 ± 0.1% and animal median survival extension from 19 to 43 days, without causing noticeable pathological changes in the major organs. These results demonstrate that injectable peptide hydrogel can be a potential carrier for localized chemotherapy of HCC.
Collapse
Key Words
- ANOVA, analysis of variance
- AST, aspartate transaminase
- ATL, alanine transaminase
- AUC0–13, areas under the curve
- AURKA, aurora A kinase
- Akt, protein kinase B
- BUN, blood urea nitrogen
- Bel-7402/Luc, luciferase transfected human HCC cell line Bel-7402
- C16-N, C16-GNNQQNYKD-OH
- C16-N/DiI, DiI-labeled C16-N
- C16-N/DiR, DiR-labeled C16-N hydrogel
- C16-N/T, triptolide-loaded peptide amphiphile-based hydrogel
- CAS, Chinese Academy of Sciences
- CD, circular dichroism
- CKS2, cyclin kinase subunit-2
- CRE, creatinine
- DL, drug loading
- DSPE-PEG, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino (polyethylene glycol)-2000]
- DSPE-PEG/DiI, DiI-labeled DSPE-PEG
- DSPE-PEG/DiR, DiR-labeled DSPE-PEG micelle
- DSPE-PEG/T, drug-loaded DSPE-PEG micelles
- EE, encapsulation efficiency
- FBS, fetal bovine serum
- FI range, fluorescence intensity range
- FI, fluorescence intensity
- GEMOX, gemcitabine and oxaliplatin
- H&E, hematoxylin and eosin
- HFIP, 1,1,1,3,3,3-hexafluoro-2-propanol
- HPLC, high-performance liquid chromatography
- Hepatocellular carcinoma
- Hydrogel
- LC–MS, liquid chromatography–mass spectrometry
- OB glue, EPIGLUs
- Peptide amphiphile
- RFI, relative fluorescence intensity
- Self-assembly
- TACE, transarterial chemoembolization
- TEM, transmission electron microscopy
- TIR, tumor inhibition rate
- Tmax, time to reach highest fluorescence intensity
- Triptolide
- d-Luciferin, (S)-4,5-dihydro-2-(6-hydroxy-2-benzothiazolyl)-4-thiazolecarboxylic acid potassium
Collapse
Affiliation(s)
- Xiyue Zhao
- Department of Chemistry, Shanghai University, Shanghai 200444, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoyu Liu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| | - Yiran Liu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Nano Science and Technology Institute, University of Science and Technology of China, Suzhou 215123, China
| | - Wei Ran
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junyang Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Jilin University, Changchun 130012, China
| | - Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guanru Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Ding
- Department of Chemistry, Shanghai University, Shanghai 200444, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Yantai University, Yantai 264005, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| |
Collapse
|
43
|
Roloff A, Nirmalananthan-Budau N, Rühle B, Borcherding H, Thiele T, Schedler U, Resch-Genger U. Quantification of Aldehydes on Polymeric Microbead Surfaces via Catch and Release of Reporter Chromophores. Anal Chem 2019; 91:8827-8834. [PMID: 31188569 DOI: 10.1021/acs.analchem.8b05515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aldehyde moieties on 2D-supports or micro- and nanoparticles can function as anchor groups for the attachment of biomolecules or as reversible binding sites for proteins on cell surfaces. The use of aldehyde-based materials in bioanalytical and medical settings calls for reliable methods to detect and quantify this functionality. We report here on a versatile concept to quantify the accessible aldehyde moieties on particle surfaces through the specific binding and subsequent release of small reporter molecules such as fluorescent dyes and nonfluorescent chromophores utilizing acylhydrazone formation as a reversible covalent labeling strategy. This is representatively demonstrated for a set of polymer microparticles with different aldehyde labeling densities. Excess reporter molecules can be easily removed by washing, eliminating inaccuracies caused by unspecific adsorption to hydrophobic surfaces. Cleavage of hydrazones at acidic pH assisted by a carbonyl trap releases the fluorescent reporters rapidly and quasi-quantitatively and allows for their fluorometric detection at low concentration. Importantly, this strategy separates the signal-generating molecules from the bead surface. This circumvents common issues associated with light scattering and signal distortions that are caused by binding-induced changes in reporter fluorescence as well as quenching dye-dye interactions on crowded particle surfaces. In addition, we demonstrate that the release of a nonfluorescent chromophore via disulfide cleavage and subsequent quantification by absorption spectroscopy gives comparable results, verifying that both assays are capable of rapid and sensitive quantification of aldehydes on microbead surfaces. These strategies enable a quantitative comparison of bead batches with different functionalization densities, and a qualitative prediction of their coupling efficiencies in bioconjugations, as demonstrated in reductive amination reactions with Streptavidin.
Collapse
Affiliation(s)
- Alexander Roloff
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany
| | - Nithiya Nirmalananthan-Budau
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany.,Institut für Chemie und Biochemie , Freie Universität Berlin , Takustrasse 3 , D-14195 Berlin , Germany
| | - Bastian Rühle
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany
| | | | - Thomas Thiele
- PolyAn GmbH , Rudolf-Baschant-Straße 2 , D-13086 Berlin , Germany
| | - Uwe Schedler
- PolyAn GmbH , Rudolf-Baschant-Straße 2 , D-13086 Berlin , Germany
| | - Ute Resch-Genger
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany
| |
Collapse
|
44
|
Mao X, Liu L, Cheng L, Cheng R, Zhang L, Deng L, Sun X, Zhang Y, Sarmento B, Cui W. Adhesive nanoparticles with inflammation regulation for promoting skin flap regeneration. J Control Release 2019; 297:91-101. [DOI: 10.1016/j.jconrel.2019.01.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/02/2019] [Accepted: 01/19/2019] [Indexed: 12/14/2022]
|
45
|
Chen CC, Li JJ, Guo NH, Chang DY, Wang CY, Chen JT, Lin WJ, Chi KH, Lee YJ, Liu RS, Chen CL, Wang HE. Evaluation of the Biological Behavior of a Gold Nanocore-Encapsulated Human Serum Albumin Nanoparticle (Au@HSANP) in a CT-26 Tumor/Ascites Mouse Model after Intravenous/Intraperitoneal Administration. Int J Mol Sci 2019; 20:ijms20010217. [PMID: 30626093 PMCID: PMC6337091 DOI: 10.3390/ijms20010217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/24/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer is one of the major causes of cancer-related death in Taiwan and worldwide. Patients with peritoneal metastasis from colorectal cancer have reduced overall survival and poor prognosis. Hybrid protein-inorganic nanoparticle systems have displayed multifunctional applications in solid cancer theranostics. In this study, a gold nanocore-encapsulated human serum albumin nanoparticle (Au@HSANP), which is a hybrid protein-inorganic nanoparticle, and its radioactive surrogate 111In-labeled Au@HSANP (111In-Au@HSANP), were developed and their biological behaviors were investigated in a tumor/ascites mouse model. 111In-Au@HSANP was injected either intravenously (iv) or intraperitoneally (ip) in CT-26 tumor/ascites-bearing mice. After ip injection, a remarkable and sustained radioactivity retention in the abdomen was noticed, based on microSPECT images. After iv injection, however, most of the radioactivity was accumulated in the mononuclear phagocyte system. The results of biodistribution indicated that ip administration was significantly more effective in increasing intraperitoneal concentration and tumor accumulation than iv administration. The ratios of area under the curve (AUC) of the ascites and tumors in the ip-injected group to those in the iv-injected group was 93 and 20, respectively. This study demonstrated that the ip injection route would be a better approach than iv injections for applying gold-albumin nanoparticle in peritoneal metastasis treatment.
Collapse
Affiliation(s)
- Chao-Cheng Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Jia-Je Li
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Nai-Hua Guo
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Deng-Yuan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Chung-Yih Wang
- Radiotherapy, Department of Medical Imaging, Cheng Hsin General Hospital, Taipei 112, Taiwan.
| | | | - Wuu-Jyh Lin
- Institute of Nuclear Energy Research, Taoyuan 325, Taiwan.
| | - Kwan-Hwa Chi
- Shin Kong Wu Ho-Su memorial hospital, Taipei 111, Taiwan.
| | - Yi-Jang Lee
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Ren-Shyan Liu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 115, Taiwan.
- Department of Nuclear Medicine and National PET/Cyclotron Center, Taipei Veterans General Hospital, Taipei 112, Taiwan.
| | - Chuan-Lin Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
46
|
Suh H, Lewis J, Fong L, Ramseier JY, Carlson K, Peng Z, Yin ES, Saltzman WM, Girardi M. Biodegradable bioadhesive nanoparticle incorporation of broad-spectrum organic sunscreen agents. Bioeng Transl Med 2019; 4:129-140. [PMID: 30680324 PMCID: PMC6336670 DOI: 10.1002/btm2.10092] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 11/17/2022] Open
Abstract
Conventional emulsion-based sunscreen formulations are limited by postapplication epicutaneous penetration that increases the risk of allergic dermatitis, cellular damage, and filter photodegradation upon ultraviolet radiation (UVR) exposure. Encapsulation of the UVB filter padimate O within bioadhesive biodegradable nanoparticles (BNPs) composed of poly(d,l-lactic acid)-hyperbranched polyglycerol was previously shown to enhance UVR protection while preventing skin absorption. Herein, we assess the capacity of BNP co-incorporation of avobenzone and octocrylene to provide broad-spectrum UVR protection. The ratio of UV filters within nanoparticles (NPs) was optimized for filter-filter stabilization upon UV irradiation and maximum drug loading. In vitro water-resistance test showed significant particle retention at 85% over 3 hr. In a pilot clinical study, protection against UVR-induced erythema of BNPs was found to be comparable to the FDA standard P2. Thus, sunscreen formulations utilizing BNP incorporation of a combination of organic filters may offer key safety and performance advantages.
Collapse
Affiliation(s)
- Hee‐Won Suh
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Julia Lewis
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - Linda Fong
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Julie Ying Ramseier
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - Kacie Carlson
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - Zheng‐Hong Peng
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Emily Sara Yin
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| | - W. Mark Saltzman
- Dept. of Biomedical Engineering, Yale School of Engineering & Applied Science55 Prospect Street, New HavenCT06520
| | - Michael Girardi
- Dept. of Dermatology, Yale School of Medicine333 Cedar Street, New HavenCT06520
| |
Collapse
|
47
|
Jin X, Asghar S, Zhang M, Chen Z, Huang L, Ping Q, Xiao Y. N-acetylcysteine modified hyaluronic acid-paclitaxel conjugate for efficient oral chemotherapy through mucosal bioadhesion ability. Colloids Surf B Biointerfaces 2018; 172:655-664. [DOI: 10.1016/j.colsurfb.2018.09.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022]
|
48
|
Baldwin P, Ohman AW, Tangutoori S, Dinulescu DM, Sridhar S. Intraperitoneal delivery of NanoOlaparib for disseminated late-stage cancer treatment. Int J Nanomedicine 2018; 13:8063-8074. [PMID: 30555227 PMCID: PMC6278886 DOI: 10.2147/ijn.s186881] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background PARP inhibitors, such as Olaparib, have advanced the treatment of ovarian cancer by providing patients with an effective and molecularly-targeted maintenance therapy. However, all orally-administered drugs, including Olaparib, must undergo first-pass metabolism. In contrast, a nanoparticle delivery system has the advantage of administering Olaparib directly into the peritoneal cavity for local treatment. Consequently, we sought to optimize the sustained-release formulation NanoOlaparib, previously deemed effective as an intravenous solid tumor treatment, for the local treatment of disseminated disease via intraperitoneal (i.p.) therapy. Methods The tumor cell line 404, which was derived from a Brca2−/−, Tp53−/−, Pten−/− genetically engineered mouse model, exhibited high sensitivity to Olaparib in vitro. It was chosen for use in developing an i.p. spread xenograft for testing nanotherapy efficacy in vivo. NanoOlaparib as a monotherapy or in combination with cisplatin was compared to oral Olaparib alone or in combination using two different dose schedules. A pilot biodistribution study was performed to determine drug accumulation in various organs following i.p. administration. Results Daily administration of NanoOlaparib reduced tumor growth and decreased the variability of the treatment response observed with daily oral Olaparib administration. However, systemic toxicity was observed in both the NanoOlaparib and vehicle (empty nanoparticle) treated groups. Scaling back the administration to twice weekly was well tolerated up to 100 mg/kg but reduced the effect on tumor growth. Biodistribution profiles indicated that NanoOlaparib began accumulating in tissues within an hour of administration and persisted for at least 72 hours after a single dose, exiting the peritoneal cavity faster than expected. Conclusion NanoOlaparib must be modified for use against disseminated disease. Future avenues to develop NanoOlaparib as an i.p. therapy include a modified surface-coating to retain it in the peritoneal cavity and prevent entry into systemic circulation, in addition to targeting moieties for localization in tumor cells.
Collapse
Affiliation(s)
- Paige Baldwin
- Department of Bioengineering, Northeastern University, Boston, MA, USA,
| | - Anders W Ohman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA,
| | | | - Daniela M Dinulescu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA,
| | - Srinivas Sridhar
- Department of Bioengineering, Northeastern University, Boston, MA, USA, .,Department of Physics, Northeastern University, Boston, MA, USA, .,Division of Radiation Oncology, Harvard Medical School, Boston, MA, USA,
| |
Collapse
|
49
|
Di Lorenzo G, Ricci G, Severini GM, Romano F, Biffi S. Imaging and therapy of ovarian cancer: clinical application of nanoparticles and future perspectives. Theranostics 2018; 8:4279-4294. [PMID: 30214620 PMCID: PMC6134923 DOI: 10.7150/thno.26345] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/08/2018] [Indexed: 12/16/2022] Open
Abstract
Despite significant advances in cancer diagnostics and treatment, ovarian cancers (OC) continue to kill more than 150,000 women every year worldwide. Due to the relatively asymptomatic nature and the advanced stage of the disease at the time of diagnosis, OC is the most lethal gynecologic malignancy. The current treatment for advanced OC relies on the synergistic effect of combining surgical cytoreduction and chemotherapy; however, beside the fact that chemotherapy resistance is a major challenge in OC management, new imaging strategies are needed to target microscopic lesions and improve both cytoreductive surgery and patient outcomes. In this context, nanostructured probes are emerging as a new class of medical tool that can simultaneously provide imaging contrast, target tumor cells, and carry a wide range of medicines resulting in better diagnosis and therapeutic precision. Herein we summarize several exemplary efforts in nanomedicine for addressing unmet clinical needs.
Collapse
Affiliation(s)
| | | | | | | | - Stefania Biffi
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
50
|
Brenner JS, Pan DC, Myerson JW, Marcos-Contreras OA, Villa CH, Patel P, Hekierski H, Chatterjee S, Tao JQ, Parhiz H, Bhamidipati K, Uhler TG, Hood ED, Kiseleva RY, Shuvaev VS, Shuvaeva T, Khoshnejad M, Johnston I, Gregory JV, Lahann J, Wang T, Cantu E, Armstead WM, Mitragotri S, Muzykantov V. Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude. Nat Commun 2018; 9:2684. [PMID: 29992966 PMCID: PMC6041332 DOI: 10.1038/s41467-018-05079-7] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Drug delivery by nanocarriers (NCs) has long been stymied by dominant liver uptake and limited target organ deposition, even when NCs are targeted using affinity moieties. Here we report a universal solution: red blood cell (RBC)-hitchhiking (RH), in which NCs adsorbed onto the RBCs transfer from RBCs to the first organ downstream of the intravascular injection. RH improves delivery for a wide range of NCs and even viral vectors. For example, RH injected intravenously increases liposome uptake in the first downstream organ, lungs, by ~40-fold compared with free NCs. Intra-carotid artery injection of RH NCs delivers >10% of the injected NC dose to the brain, ~10× higher than that achieved with affinity moieties. Further, RH works in mice, pigs, and ex vivo human lungs without causing RBC or end-organ toxicities. Thus, RH is a clinically translatable platform technology poised to augment drug delivery in acute lung disease, stroke, and several other diseases.
Collapse
Affiliation(s)
- Jacob S Brenner
- Pulmonary, Allergy, & Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Daniel C Pan
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Transfusion Medicine and Therapeutic Pathology, Department of Pathology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Priyal Patel
- Pulmonary, Allergy, & Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hugh Hekierski
- Department of Anesthesiology & Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shampa Chatterjee
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jian-Qin Tao
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hamideh Parhiz
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kartik Bhamidipati
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Thomas G Uhler
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Raisa Yu Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vladimir S Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Makan Khoshnejad
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ian Johnston
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason V Gregory
- Department of Chemical Engineering and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joerg Lahann
- Department of Chemical Engineering and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tao Wang
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edward Cantu
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William M Armstead
- Department of Anesthesiology & Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Samir Mitragotri
- School of Engineering & Applied Sciences, Harvard University, Wyss Institute, Cambridge, MA, 02138, USA
| | - Vladimir Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|