1
|
Venturini J, Chakraborty A, Baysal MA, Tsimberidou AM. Developments in nanotechnology approaches for the treatment of solid tumors. Exp Hematol Oncol 2025; 14:76. [PMID: 40390104 PMCID: PMC12090476 DOI: 10.1186/s40164-025-00656-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/21/2025] Open
Abstract
Nanotechnology has revolutionized cancer therapy by introducing advanced drug delivery systems that enhance therapeutic efficacy while reducing adverse effects. By leveraging various nanoparticle platforms-including liposomes, polymeric nanoparticles, and inorganic nanoparticles-researchers have improved drug solubility, stability, and bioavailability. Additionally, new nanodevices are being engineered to respond to specific physiological conditions like temperature and pH variations, enabling controlled drug release and optimizing therapeutic outcomes. Beyond drug delivery, nanotechnology plays a crucial role in the theranostic field due to the functionalization of specific materials that combine tumor detection and targeted treatment features. This review analyzes the clinical impact of nanotechnology, spanning from early-phase trials to pivotal phase 3 studies that have obtained regulatory approval, while also offering a critical perspective on the preclinical domain and its translational potential for future human applications. Despite significant progress, greater attention must be placed on key challenges, such as biocompatibility barriers and the lack of regulatory standardization, to ensure the successful translation of nanomedicine into routine clinical practice.
Collapse
Affiliation(s)
- Jacopo Venturini
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
- Current Affiliation: Department of Medical Oncology, Careggi University Hospital, Florence, Italy
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
George M, Boukherroub R, Sanyal A, Szunerits S. Treatment of lung diseases via nanoparticles and nanorobots: Are these viable alternatives to overcome current treatments? Mater Today Bio 2025; 31:101616. [PMID: 40124344 PMCID: PMC11930446 DOI: 10.1016/j.mtbio.2025.101616] [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: 12/05/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Challenges Respiratory diseases remain challenging to treat, with current efforts primarily focused on managing symptoms rather than maintaining overall lung health. Traditional treatment methods, such as oral or parenteral administration of antiviral, antibacterial, and anti-inflammatory drugs, face limitations. These include difficulty in delivering therapeutic agents to pathogens residing deep in the airways and the risk of severe side effects due to high systemic drug concentrations. The growing threat of drug-resistant pathogens further complicates infection management. Advancements The lung's large surface area offers an attractive target for inhalation-based drug delivery. Nanoparticles (NP) enable uniform and sustained drug distribution across the alveolar network, overcoming challenges posed by complex lung anatomy. Recent breakthroughs in nanorobots (NR) have demonstrated precise navigation through biological environments, delivering therapies directly to affected lung areas with enhanced accuracy. Nanotechnology has also shown promise in treating lung cancer, with nanoparticles engineered to overcome biological barriers, improve drug solubility, and enable controlled drug release. Future scope This review explores the progress of NP and NR in addressing challenges in pulmonary drug delivery. These innovations allow targeted delivery of nucleic acids, drugs, or peptides to the pulmonary epithelium with unprecedented accuracy, offering significant potential for improving therapeutic effectiveness in respiratory disorders.
Collapse
Affiliation(s)
- Meekha George
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | - Sabine Szunerits
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| |
Collapse
|
3
|
Rong D, Gao L, Chen Y, Gao XZ, Tang M, Tang H, Gao Y, Lu G, Ling ZQ, Shen HM. Suppression of the LKB1-AMPK-SLC7A11-GSH signaling pathway sensitizes NSCLC to albumin-bound paclitaxel via oxidative stress. Redox Biol 2025; 81:103567. [PMID: 40023979 PMCID: PMC11915006 DOI: 10.1016/j.redox.2025.103567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Albumin-bound paclitaxel (nab-PTX) is an important chemotherapeutic drug used for the treatment of advanced and metastatic non-small cell lung cancer (NSCLC). One critical issue in its clinical application is the development of resistance; thus, a deeper understanding of the mechanisms underlying the primary resistance to nab-PTX is expected to help to develop effective therapeutic strategies to overcome resistance. In this study, we made an unexpected discovery that NSCLC with wild-type (WT) Liver kinase B1 (LKB1), an important tumor suppressor and upstream kinase of AMP-activated protein kinase (AMPK), is more resistant to nab-PTX than NSCLC with mutant LKB1. Mechanistically, LKB1 status does not alter the intracellular concentration of nab-PTX or affect its canonical pharmacological action in promoting microtubule polymerization. Instead, we found that LKB1 mediates AMPK activation, leading to increased expression of SLC7A11, a key amino acid transporter and intracellular level of glutathione (GSH), which then attenuates the production of reactive oxygen species (ROS) and apoptotic cell death induced by nab-PTX. On the other hand, genetic or pharmacological inhibition of AMPK in LKB1-WT NSCLC reduces the expression of SLC7A11 and intracellular GSH, increases ROS level, and eventually promotes the apoptotic cell death induced by nab-PTX in vitro. Consistently, the combination of nab-PTX with an AMPK inhibitor exhibits a greater therapeutic efficacy in LKB1-WT NSCLC using xenograft models in vivo. Taken together, our data reveal a novel role of LKB1-AMPK-SLC7A11-GSH signaling pathway in the primary resistance to nab-PTX, and provide a therapeutic strategy for the treatment of LKB1-WT NSCLC by targeting the LKB1-AMPK-SLC7A11-GSH pathway.
Collapse
Affiliation(s)
- Dade Rong
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Yiguan Chen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Xiang-Zheng Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Mingzhu Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Haimei Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China; Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Qiang Ling
- Experimental Research Centre, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Han-Ming Shen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
4
|
Hacker RM, Smith JJ, Platt DC, Brennessel WW, Jones MA, Webb MI. Ruthenium(II)-Arene Complexes with a 2,2'-Bipyridine Ligand as Anti-Aβ Agents. Biomolecules 2025; 15:475. [PMID: 40305171 PMCID: PMC12024814 DOI: 10.3390/biom15040475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Agents that target the amyloid-β (Aβ) peptide associated with Alzheimer's disease have seen renewed interest following the clinical success of antibody therapeutics. Small molecules, specifically metal-based complexes, are excellent candidates for advancement, given their relative ease of preparation and modular scaffold. Herein, several ruthenium-arene complexes containing 2,2-bipyridine (bpy) ligands were prepared and evaluated for their respective ability to modulate the aggregation of Aβ. This was carried out using the three sequential methods of thioflavin T (ThT) fluorescence, dynamic ligand scattering (DLS), and transmission electron microscopy (TEM). Overall, it was observed that RuBA, the complex with a 4,4-diamino-2,2-bipyridine ligand, had the greatest impact on Aβ aggregation. Further evaluation of the complexes was performed to determine their relative affinity for serum albumin and biocompatibility towards two neuronal cell lines. Ultimately, RuBA outperformed the other Ru complexes, where the structure-activity relationship codified the importance of the amino groups on the bpy for anti-Aβ activity.
Collapse
Affiliation(s)
- Ryan M. Hacker
- Department of Chemistry and Biochemistry, SUNY Geneseo, Geneseo, NY 14454, USA; (R.M.H.); (J.J.S.)
| | - Jacob J. Smith
- Department of Chemistry and Biochemistry, SUNY Geneseo, Geneseo, NY 14454, USA; (R.M.H.); (J.J.S.)
| | - David C. Platt
- Department of Chemistry, Illinois State University, Normal, IL 69701, USA; (D.C.P.); (M.A.J.)
| | | | - Marjorie A. Jones
- Department of Chemistry, Illinois State University, Normal, IL 69701, USA; (D.C.P.); (M.A.J.)
| | - Michael I. Webb
- Department of Chemistry and Biochemistry, SUNY Geneseo, Geneseo, NY 14454, USA; (R.M.H.); (J.J.S.)
| |
Collapse
|
5
|
Raj M, Meena A, Seth R, Mathur A, Luqman S. An update on nanoformulations with FDA approved drugs for female reproductive cancer. J Microencapsul 2025:1-34. [PMID: 40114400 DOI: 10.1080/02652048.2025.2474457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Female reproductive cancers, including ovarian, cervical, breast, gestational trophoblastic and endometrial cancer, present significant challenges in therapy and patient prognosis. Conventional chemotherapy often lacks selectivity, leading to systemic toxicity and reduced treatment efficacy. Nanotechnology has emerged as a promising approach to improve drug delivery and therapeutic outcomes. Encapsulation of FDA-approved drugs within nanocarriers such as liposomes, polymeric nanoparticles, and lipid nanoparticles enables controlled drug release, reduces off-target effects, and enhances drug accumulation at tumor sites. This targeted delivery minimizes damage to healthy tissues and improves patient survival rates. Additionally, nanoformulations facilitate combination therapy, overcoming drug resistance and maximizing therapeutic efficacy. Despite promising results, challenges like scalability, reproducibility, and regulatory approvals hinder widespread clinical applications. Developing personalized nanoformulations tailored to individual patient profiles offers potential for precision cancer therapy. This study explores the role of nanoformulations in enhancing the therapeutic potential of FDA-approved drugs for treating female reproductive cancers.
Collapse
Affiliation(s)
- Mahima Raj
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Richa Seth
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anurag Mathur
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Stukan I, Żuk A, Pukacka K, Mierzejewska J, Pawłowski J, Kowalski B, Dąbkowska M. Wolf in Sheep's Clothing: Taming Cancer's Resistance with Human Serum Albumin? Int J Nanomedicine 2025; 20:3493-3525. [PMID: 40125439 PMCID: PMC11930253 DOI: 10.2147/ijn.s500997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Human serum albumin (HSA) has emerged as a promising carrier for nanodrug delivery, offering unique structural properties that can be engineered to overcome key challenges in cancer treatment, especially resistance to chemotherapy. This review focuses on the cellular uptake of albumin-based nanoparticles and the modifications that enhance their ability to bypass resistance mechanisms, particularly multidrug resistance type 1 (MDR1), by improving targeting to cancer cells. In our unique approach, we integrate the chemical properties of albumin, its interactions with cancer cells, and surface modifications of albumin-based delivery systems that enable to bypass resistance mechanisms, particularly those related to MDR1, and precisely target receptors on cancer cells to improve treatment efficacy. We discuss that while well-established albumin receptors such as gp60 and gp18/30 are crucial for cellular uptake and transcytosis, their biology remains underexplored, limiting their translational potential. Additionally, we explore the potential of emerging targets, such as cluster of differentiation 44 (CD44), cluster of differentiation (CD36) and transferrin receptor TfR1, as well as the advantages of using dimeric forms of albumin (dHSA) to further enhance delivery to resistant cancer cells. Drawing from clinical examples, including the success of albumin-bound paclitaxel (Abraxane) and new formulations like Pazenir and Fyarro (for Sirolimus), we identify gaps in current knowledge and propose strategies to optimize albumin-based systems. In conclusion, albumin-based nanoparticles, when tailored with appropriate modifications, have the potential to bypass multidrug resistance and improve the targeting of cancer cells. By enhancing albumin's ability to efficiently deliver therapeutic agents, these carriers represent a promising approach to addressing one of oncology's most persistent challenges, with substantial potential to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Iga Stukan
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Anna Żuk
- Independent Laboratory of Community Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Kamila Pukacka
- Department of Pharmaceutical Technology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Julia Mierzejewska
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jakub Pawłowski
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bogusław Kowalski
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Maria Dąbkowska
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
7
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|
8
|
Shastri D, Raorane CJ, Raj V, Lee S. Human serum albumin-3-amino-1-propanesulfonic acid conjugate inhibits amyloid-β aggregation and mitigates cognitive decline in Alzheimer's disease. J Control Release 2025; 379:390-408. [PMID: 39805463 DOI: 10.1016/j.jconrel.2025.01.019] [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: 08/27/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD) is the most commonly occurring brain disorder, characterized by the accumulation of amyloid-β (Aβ) and tau, subsequently leading to neurocognitive decline. 3-Amino-1-propanesulfonic acid (TPS) and its prodrug, currently under clinical trial III, serve as promising therapeutic agents targeting Aβ pathology by specifically preventing monomer-to-oligomer formation. Inspired by the potency of TPS prodrug, we hypothesized that conjugating TPS with human serum albumin (HSA) could enhance brain delivery and synergistically inhibit Aβ aggregation in mild to moderate AD. Thus, we prepared and extensively characterized HSA-TPS (h-TPS) conjugate using an eco-friendly coupling method. In vitro studies on Aβ aggregation kinetics and AFM imaging revealed significant prevention of Aβ aggregation. Additionally, h-TPS significantly reduced Aβ-induced neurotoxicity and H2O2-mediated reactive oxygen species (ROS) stress in SH-SY5Y cells. Moreover, h-TPS administration improved blood-brain barrier permeability and cellular uptake into neuronal cells as well as showed in vivo uptake inside the brain within 1 h. In vivo studies using an Aβ1-42-induced acute AD rat model exhibited a dose-dependent significant reduction in hippocampal Aβ levels and restoration of declined spatial learning and memory with h-TPS treatment. Overall, findings suggest that h-TPS conjugate might be a promising neuroprotective agent for preventing Aβ aggregation in mild to moderate AD.
Collapse
Affiliation(s)
- Divya Shastri
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea; College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | | | - Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Wang N, Mei X, Cao Y, Wang L, Xie H, Jia J, Xiao Y, Han J, Huang A, Ma H. Nab-Paclitaxel Promotes Radiosensitization by Inducing DNA Damage and Inhibiting Macrophage M2 Polarization in Cholangiocarcinoma. Cancer Biother Radiopharm 2025. [PMID: 40000014 DOI: 10.1089/cbr.2024.0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Background: Nab-paclitaxel effectively inhibits tumor proliferation and modulates macrophage polarization to improve the tumor microenvironment. However, its potential to achieve radiosensitization in cholangiocarcinoma remains to be elucidated. Materials and Methods: The proliferation inhibition and radiosensitizing effects of nab-paclitaxel were assessed using cell counting kit-8 and colony formation assays in NOZ and TFK1 cell lines. Cell apoptosis, cell cycle progression, DNA damage, and macrophage polarization status were analyzed via flow cytometry immunofluorescence, enzyme-linked immunosorbent assay, and qRT-PCR. A tumor-bearing mouse model was established to validate radiosensitization in vivo. Potentially related genes and proteins involved in nab-paclitaxel-induced radiosensitization were identified through RNA transcriptome sequencing and Western blotting. Results: Nab-paclitaxel exhibited significant radiosensitizing effects on cholangiocarcinoma cells. Combined with radiotherapy, nab-paclitaxel increased DNA damage, promoted apoptosis, and effectively inhibited M2 polarization of macrophages in vivo and in vitro. The radiosensitizing effect is involved in the activation of the AMP-dependent protein kinase (AMPK) signaling pathway. Nab-paclitaxel significantly upregulated phosphorylated AMPKα, apoptotic proteins as zinc finger matrin-type 3, and nuclear factor kappa-B levels following radiation exposure. Conclusions: Our study confirmed the radiosensitizing effect of nab-paclitaxel on cholangiocarcinoma cells through a dual effect of antitumor proliferation and inhibition of M2 macrophage polarization, and the underlying mechanism involved activation of the AMPK signaling pathway.
Collapse
Affiliation(s)
- Ningyu Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangping Mei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Cao
- Department of Gastrointestinal Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Lingfang Wang
- Department of Gastrointestinal Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | | | - Jingru Jia
- Department of Oncology, Renmin Hospital of Gucheng, Gucheng, China
| | - Yong Xiao
- Department of Gastrointestinal Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Han
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ai Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Escalera-Anzola S, Rosado M, Yang Y, Parra-Sanchez D, Pedro-Liberal CS, Acedo P. Breakthroughs in nanoparticle-based strategies for pancreatic cancer therapy. Biochem Pharmacol 2025; 232:116685. [PMID: 39613113 DOI: 10.1016/j.bcp.2024.116685] [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: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, mainly due to its high heterogeneity, resistance to therapy and late diagnosis, with a 5-year survival rate of less than 10%. This dismal prognosis has promoted strategies to develop more effective treatments. Nanoparticle-based strategies have emerged, in the last decades, as a great opportunity because they can enhance drug delivery and promote controlled release, presenting lower side effects than conventional therapeutic regimens. Moreover, nanoparticles can often be modified to target specific cells or to achieve a sustained release of the drugs into the tumor. However, very few nanoparticle-based therapies are clinically approved. Concretely for pancreatic cancer treatment only two nanoformulations have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) so far. Clinical translation of nanoparticles remains a challenge for modern medicine, and in particular for pancreatic cancer therapy, because of the complexity of the disease, and a lack of studies been performed in clinically relevant in vitro and in vivo models. In this review, we have summarized the most recent clinical trials using nanoparticle-based formulations in PDAC, giving a small context of the diverse types of nanoparticles employed and the most recent advancements in the field.
Collapse
Affiliation(s)
- Sara Escalera-Anzola
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom; Smart Devices for Nano Medicine Group, Unidad Excelencia Instituto de BioMedicina y Genética Molecular (IBGM) de Valladolid, University of Valladolid and CSIC, Valladolid, Spain
| | - Maria Rosado
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Yuchen Yang
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Daniel Parra-Sanchez
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Carolina San Pedro-Liberal
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom.
| |
Collapse
|
11
|
Lin N, Chen S, Zheng Z, Song X. Cost-effectiveness of first-line sintilimab plus chemotherapy versus chemotherapy for advanced esophageal carcinoma in China. Expert Rev Pharmacoecon Outcomes Res 2025; 25:205-213. [PMID: 39327693 DOI: 10.1080/14737167.2024.2410248] [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: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND To evaluate the cost-effectiveness of first-line sintilimab plus chemotherapy versus chemotherapy for advanced esophageal squamous cell carcinoma (ESCC) from the perspective of the Chinese health service system. METHODS A partitioned survival model was constructed to simulate quality-adjusted life years and incremental cost-effectiveness ratios over a patient's lifetime based on a phase III clinical trial. RESULTS Sintilimab plus chemotherapy increased by 0.316 QALY and 0.285 QALY with the additional cost of $5692 and $5269, which led to the ICER of $18000/QALY and $18519/QALY gained in the overall population and the patients with CPS ≥ 10, respectively. CONCLUSIONS Compared with chemotherapy alone, sintilimab may be a cost-effective first-line treatment choice for locally advanced or metastatic ESCC.
Collapse
Affiliation(s)
- Nanlong Lin
- Department of Thoracic surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shiting Chen
- Department of General Surgery, Quangang General Hospital, The First Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhiwei Zheng
- Department of Pharmacy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaobing Song
- Department of Quality Management, Ganzhou Fifth People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
12
|
Yang M, Kim Y, Youn SY, Jeong H, Shirbhate ME, Uhm C, Kim G, Nam KT, Cha SS, Kim KM, Yoon J. Conversion of albumin into a BODIPY-like photosensitizer by a flick reaction, tumor accumulation and photodynamic therapy. Biomaterials 2025; 313:122792. [PMID: 39226652 DOI: 10.1016/j.biomaterials.2024.122792] [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: 05/31/2024] [Revised: 08/11/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
The accumulation of photosensitizers (PSs) in lesion sites but not in other organs is an important challenge for efficient image guiding in photodynamic therapy. Cancer cells are known to express a significant number of albumin-binding proteins that take up albumin as a nutrient source. Here, we converted albumin to a novel BODIPY-like PS by generating a tetrahedral boron environment via a flick reaction. The formed albumin PS has almost the same 3-dimensional structural feature as free albumin because binding occurs at Sudlow Site 1, which is located in the interior space of albumin. An i.v. injection experiment in tumor-bearing mice demonstrated that the human serum albumin PS effectively accumulated in cancer tissue and, more surprisingly, albumin PS accumulated much more in the cancer tissue than in the liver and kidneys. The albumin PS was effective at killing tumor cells through the generation of reactive oxygen species under light irradiation. The crystal structure of the albumin PS was fully elucidated by X-ray crystallography; thus, further tuning of the structure will lead to novel physicochemical properties of the albumin PS, suggesting its potential in biological and clinical applications.
Collapse
Affiliation(s)
- Mengyao Yang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea
| | - Yujin Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea
| | - So-Yeon Youn
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea; TODD PHARM, Ewha Womans University, Seoul, 03760, South Korea
| | - Haengdueng Jeong
- Department of Biomedical Sciences, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03760, South Korea
| | | | - Chanyang Uhm
- Department of Biomedical Sciences, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03760, South Korea
| | - Gyoungmi Kim
- Research Center for Biomaterials, KYTECBIO, Ewhayeodae-gil 52, Seoul, 03760, South Korea
| | - Ki Taek Nam
- Department of Biomedical Sciences, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, 03760, South Korea.
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea; TODD PHARM, Ewha Womans University, Seoul, 03760, South Korea.
| | - Kwan Mook Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea.
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea.
| |
Collapse
|
13
|
Chen XC, Jiao DC, Qiao JH, Wang CZ, Sun XF, Lu ZD, Li LF, Zhang CJ, Yan M, Wei Y, Chen B, Feng YQ, Deng M, Ma MD, Plichta JK, He YW, Liu ZZ. De-escalated neoadjuvant weekly nab-paclitaxel with trastuzumab and pertuzumab versus docetaxel, carboplatin, trastuzumab, and pertuzumab in patients with HER2-positive early breast cancer (HELEN-006): a multicentre, randomised, phase 3 trial. Lancet Oncol 2025; 26:27-36. [PMID: 39612919 DOI: 10.1016/s1470-2045(24)00581-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND A previous phase 2 trial showed promising outcomes for patients with HER2-positive early-stage breast cancer using neoadjuvant de-escalation chemotherapy with paclitaxel, trastuzumab, and pertuzumab. We aimed to evaluate the efficacy of weekly nab-paclitaxel compared with the standard regimen of docetaxel plus carboplatin, both with trastuzumab and pertuzumab, as neoadjuvant therapies for patients with HER2-positive breast cancer. METHODS HELEN-006 was a multicentre, randomised, phase 3 trial done at six hospitals in China. We enrolled patients aged 18-70 years with untreated, histologically confirmed stage II-III invasive HER2-positive breast cancer and an Eastern Cooperative Oncology Group performance status of 0 or 1. Using an interactive response system, patients were randomly assigned (1:1) under a permuted block randomisation scheme (block size of four), stratified by tumour stage, nodal status, and hormone receptor status. Patients received either intravenous nab-paclitaxel (125 mg/m2 on days 1, 8, and 15) for six 3-week cycles, or intravenous docetaxel (75 mg/m2 on day 1) plus intravenous carboplatin (area under the concentration-time curve 6 mg/mL per min on day 1) for six 3-week cycles. Both groups also received concurrent intravenous trastuzumab, with an initial loading dose of 8 mg/kg and a maintenance dose of 6 mg/kg on day 1, as well as intravenous pertuzumab with a loading dose of 840 mg and a maintenance dose of 420 mg on day 1. This report is the final analysis of the primary endpoint, pathological complete response (ypT0/is ypN0), analysed in all patients who started treatment (modified intention to treat). The trial is registered with ClinicalTrials.gov, NCT04547907, and follow-up of the adjuvant phase is ongoing. FINDINGS Between Sept 20, 2020, and March 1, 2023, 789 patients were screened for eligibility, 689 of whom were randomly assigned (343 to the nab-paclitaxel group and 346 to the docetaxel plus carboplatin group). All 689 patients were Asian women. 669 patients received at least one dose of the study treatment and were included in the full analysis set (332 in the nab-paclitaxel group and 337 in the docetaxel plus carboplatin group). Median age of the patients was 50 years (IQR 43-55). Median follow-up time was 26 months (IQR 19-32). 220 (66·3% [95% CI 61·2-71·4]) patients in the nab-paclitaxel group had a pathological complete response, compared with 194 (57·6% [52·3-62·9]) in the docetaxel plus carboplatin group (combined odds ratio 1·54 [95% CI 1·10-2·14]; stratified p=0·011). 100 (30%) patients in the nab-paclitaxel group and 128 (38%) in the docetaxel plus carboplatin group had grade 3-4 adverse events. The most common grade 3-4 adverse events were nausea (22 [7%] in the nab-paclitaxel group vs 76 [23%] in the docetaxel plus carboplatin group), diarrhoea (25 [8%] vs 55 [16%]), and neuropathy (43 [13%] vs eight [2%]). Serious drug-related adverse events were reported in three (1%) patients in the nab-paclitaxel group and five (2%) in the docetaxel plus carboplatin group. No treatment-related deaths were reported in either group. INTERPRETATION These findings might suggest a potential advantage of nab-paclitaxel combined with trastuzumab and pertuzumab compared with the standard regimen in neoadjuvant therapy for patients with HER2-positive early breast cancer, suggesting that this new combination might establish a new standard for neoadjuvant treatment in this patient population. FUNDING National Natural Science Foundation of China, and Science and Technology Research Projects of Henan Province, China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Xiu-Chun Chen
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - De-Chuang Jiao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiang-Hua Qiao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Cheng-Zheng Wang
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xian-Fu Sun
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Zhen-Duo Lu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lian-Fang Li
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Chong-Jian Zhang
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Min Yan
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Ya Wei
- Department of Breast Surgery, Anyang Tumor Hospital, Anyang, China
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yue-Qing Feng
- Department of Breast Surgery, Xinxiang Central Hospital, Xinxiang, China
| | - Miao Deng
- Department of Breast Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Ming-De Ma
- Department of Thyroid and Breast Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jennifer K Plichta
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Zhen-Zhen Liu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
14
|
Liu C, Xu W, Song X, Tian W, Liu F, Wang F. A Nanotheranostic Agent for Synergistic Antitumor Chemo/Phototherapy Prepared by Paclitaxel-Induced Self-Assembly of PEGylated Human Serum Albumin with Prolonged Circulation. ACS OMEGA 2024; 9:51062-51072. [PMID: 39758671 PMCID: PMC11696392 DOI: 10.1021/acsomega.4c05986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 01/07/2025]
Abstract
The integration of different therapies to enhance the efficacy and minimize adverse reactions has become popular recently. This approach leverages the complementary mechanisms of action of different treatments, which can lead to better therapeutic outcomes and reduced side effects. Human serum albumin (HSA) exhibits excellent drug loading ability and is often used for biomimetic tumor delivery in multidrug nanocarriers. However, albumin nanocarriers are often unstable with a short plasma half-life. Therefore, a nanotheranostic agent for synergistic antitumor chemo/phototherapy was designed to improve HSA's pharmacokinetic properties, including prolonged circulation. Cys34-specifically PEGylated HSA (PEG-cys34HSA) was used as the nanocarrier, hydrophobic paclitaxel (PTX) served as the chemotherapeutic drug and self-assembly inducer of nanoparticles (NPs), and near-infrared dye indocyanine green (ICG) was utilized for phototherapy and fluorescence imaging. PEGylation with 20 kDa polyethylene glycol (PEG20kD) promoted the formation of uniform and regular NPs more effectively than PEG5kD. PEG20kD also enhanced the particle size, drug loading, and encapsulation efficiency. Moreover, PEG20kD significantly enhanced tumor targeting without hindering endocytosis, transport, and release of NPs. PEG20kD-cys34HSA/PTX/ICG-mediated combination therapy exhibited synergistic inhibitory effects on tumor growth both in vitro and in vivo. Thus, PEG20kD-cys34HSA shows potential as an alternative nanocarrier. This study provides the foundation for future investigations into PEG-modified nanocarriers and comprehensive tumor treatment.
Collapse
Affiliation(s)
- Changsong Liu
- Key
Laboratory of Chemical Biology of Natural Products (Ministry of Education),
Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical
Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Wenjia Xu
- Key
Laboratory of Chemical Biology of Natural Products (Ministry of Education),
Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical
Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xinlei Song
- Department
of Pharmacy, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Weilu Tian
- Key
Laboratory of Chemical Biology of Natural Products (Ministry of Education),
Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical
Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Fuyan Liu
- School
of Biological Sciences and Technology, University
of Jinan, Jinan 250022, China
| | - Fengshan Wang
- Key
Laboratory of Chemical Biology of Natural Products (Ministry of Education),
Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical
Sciences, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
15
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [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: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
16
|
Macone A, Cappelletti C, Incocciati A, Piacentini R, Botta S, Boffi A, Bonamore A. Challenges in Exploiting Human H Ferritin Nanoparticles for Drug Delivery: Navigating Physiological Constraints. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2016. [PMID: 39541599 DOI: 10.1002/wnan.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Over the past two decades, ferritin has emerged as a promising nanoparticle for drug delivery, catalyzing the development of numerous prototypes capable of encapsulating a wide array of therapeutic agents. These ferritin-based nanoparticles exhibit selectivity for various molecular targets and are distinguished by their potential biocompatibility, unique symmetrical structure, and highly controlled size. The hollow interior of ferritin nanoparticles allows for efficient encapsulation of diverse therapeutic agents, enhancing their delivery and effectiveness. Despite these promising features, the anticipated clinical advancements have yet to be fully realized. As a physiological protein with a central role in both health and disease, ferritin can exert unexpected effects on physiology when employed as a drug delivery system. Many studies have not thoroughly evaluated the pharmacokinetic properties of the ferritin protein shell when administered in vivo, overlooking crucial aspects such as biodistribution, clearance, cellular trafficking, and immune response. Addressing these challenges is crucial for achieving the desired transition from bench to bedside. Biodistribution studies need to account for ferritin's natural accumulation in specific organs (liver, spleen, and kidneys), which may lead to off-target effects. Moreover, the mechanisms of clearance and cellular trafficking must be elucidated to optimize the delivery and reduce potential toxicity of ferritin nanoparticles. Additionally, understanding the immune response elicited by exogenous ferritin is essential to mitigate adverse reactions and enhance therapeutic efficacy. A comprehensive understanding of these physiological constraints, along with innovative solutions, is essential to fully realize the therapeutic potential of ferritin nanoparticles paving the way for their successful clinical translation.
Collapse
Affiliation(s)
- Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Chiara Cappelletti
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessio Incocciati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Roberta Piacentini
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Sofia Botta
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alberto Boffi
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessandra Bonamore
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
17
|
Berardi AJ, Francisco SD, Chang A, Zelaya JC, Raymond JE, Lahann J. Synthetic Protein Nanoparticles via Photoreactive Electrohydrodynamic Jetting. Macromol Rapid Commun 2024; 45:e2400349. [PMID: 39171381 DOI: 10.1002/marc.202400349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/20/2024] [Indexed: 08/23/2024]
Abstract
Protein nanoparticles are an attractive class of materials for nanomedicine applications due to the intrinsic biocompatibility, biodegradability, and intrinsic functionality of their constituent proteins. Despite the clinical success of select protein nanoparticles, this class of nanocarriers remains understudied and underdeveloped compared to lipid and polymer nanoparticles due to challenges related to formulation optimization, large design space, and their structural complexity. In this work, a modular strategy for protein nanoparticle preparation based on the concept of photoreactive jetting is introduced. The process relies on continuous ultraviolet irradiation during electrohydrodynamic (EHD) jetting of protein solutions that contain a homobifunctional photocrosslinker. Protein nanoparticles exhibit nanogel-like architectures comprised of proteins that are linked via synthetic moieties. Compared to conventional protein nanoparticles, this method reduces nanoparticle processing times to minutes, rather than hours to days. The inclusion of an emissive structural motif as the molecular scaffold of the photocrosslinker is used to study the supramolecular architecture of the stable nanoparticles via time-resolved fluorescence spectroscopy.
Collapse
Affiliation(s)
- Anthony J Berardi
- Macromolecular Science and Engineering Program, Ann Arbor, 48109, USA
- Biointerfaces Institute, Ann Arbor, 48109, USA
| | - Sonja D Francisco
- Biointerfaces Institute, Ann Arbor, 48109, USA
- Department of Chemistry, Ann Arbor, 48109, USA
| | - Albert Chang
- Biointerfaces Institute, Ann Arbor, 48109, USA
- Department of Materials Science and Engineering, Ann Arbor, 48109, USA
| | - Julio C Zelaya
- Macromolecular Science and Engineering Program, Ann Arbor, 48109, USA
- Biointerfaces Institute, Ann Arbor, 48109, USA
| | - Jeffery E Raymond
- Biointerfaces Institute, Ann Arbor, 48109, USA
- Department of Chemical Engineering, Ann Arbor, 48109, USA
- Center for Complex Particle Systems, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joerg Lahann
- Macromolecular Science and Engineering Program, Ann Arbor, 48109, USA
- Biointerfaces Institute, Ann Arbor, 48109, USA
- Department of Materials Science and Engineering, Ann Arbor, 48109, USA
- Department of Chemical Engineering, Ann Arbor, 48109, USA
- Center for Complex Particle Systems, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
18
|
Zhong T, Yang Y, Pang M, Pan Y, Jing S, Qi Y, Huang Y. Human Serum Albumin-Coated 10B Enriched Carbon Dots as Targeted "Pilot Light" for Boron Neutron Capture Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406577. [PMID: 39324650 DOI: 10.1002/advs.202406577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Boron neutron capture therapy (BNCT) is a physiologically focused radiation therapy that relies on nuclear capture and fission processes. BNCT is regarded as one of the most promising treatments due to its excellent accuracy, short duration of therapy, and low side effects. The creation of novel boron medicines with high selectivity, ease of delivery, and high boron-effective load is a current research topic. Herein, boron-containing carbon dots (BCDs) and their human serum albumin (HSA) complexes (BCDs-HSA) are designed and synthesized as boron-containing drugs for BNCT. BCDs (10B: 7.1 wt%) and BCDs-HSA exhibited excitation-independent orange fluorescent emission which supported the use of fluorescence imaging for tracking 10B in vivo. The introduction of HSA enabled BCDs-HSA to exhibit good biocompatibility and increased tumor accumulation. The active and passive targeting abilities of BCDs-HSA are explored in detail. Subcutaneous RM-1 tumors and B16-F10 tumors both significantly decrease with BNCT, which consists of injecting BCDs-HSA and then irradiating the area with neutrons. In short, this study provides a novel strategy for the delivery of boron and may broaden the perspectives for the design of boron-containing carbon dots nanomedicine for BNCT.
Collapse
Affiliation(s)
- Tianyuan Zhong
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Yongjin Yang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, 730000, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, China
| | - Miao Pang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Shiwei Jing
- School of Physics, Northeast Normal University, Changchun, 130024, China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
19
|
Yin L, Jiang W, Liu S, Fu Y, Zhou L, Pei X, Ye S, Shen W, Yang H, Shan B. Efficacy and safety of nanoparticle albumin-bound paclitaxel plus carboplatin as neoadjuvant chemotherapy for stages III-IV, unresectable ovarian cancer: a single-arm, open-label, phase Ib/II study. BMC Med 2024; 22:496. [PMID: 39468597 PMCID: PMC11520812 DOI: 10.1186/s12916-024-03697-1] [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: 05/24/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy may be considered for patients with ovarian cancer (OC) whose tumors are deemed unlikely to be completely cytoreduced to no gross residual disease (R0) or who are poor surgical candidates. This Ib/II study was designed to assess the efficacy and safety of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) plus carboplatin as neoadjuvant chemotherapy for stages III-IV, unresectable OC. METHODS Eligible patients with stage III-IV, unresectable OC were enrolled in this phase Ib/II study. All patients received neoadjuvant nab-paclitaxel (260 mg/m2, day 1, every 3 weeks) plus carboplatin (AUC 5, day 1, every 3 weeks) for 3 cycles before surgery, followed by 3-6 cycles of adjuvant chemotherapy. The phase Ib primary endpoint was safety; the phase II primary endpoint was the R0 resection rate. Secondary endpoints were progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and safety (for all populations). RESULTS Sixty-two patients were enrolled and were given neoadjuvant therapy treated between October 2019 and December 2020, of whom 9 were in the phase Ib portion and 53 in the phase II portion. A total of 53 patients underwent surgery with an R0 resection rate of 73.6% (95% CI, 59.7-84.7%). With a median follow-up of 17.5 (range 0.7-36.7) months, for all patients, the best ORR was 83.9% (95% CI, 71.7-92.4%) with 47 partial responses, the median PFS was 18.6 (95% CI, 13.8-23.3%) months, and median OS was not reached. During the neoadjuvant chemotherapy, treatment-related adverse events (TRAEs) of any grade occurred in 91.9% (57/62) of all patients. The most common hematologic TRAEs were neutropenia (55/62, 88.7%), and non-hematologic toxicity was alopecia (36/62, 58.1%). Forty-nine patients (79.0%) experienced at least one grade 3-4 TRAEs, with the most common was neutropenia (44/62, 71.0%). Besides, delays in neoadjuvant chemotherapy and surgery due to AEs were observed in 9 (1 in phase Ib; 8 in phase II) and 7 (phase II) patients, respectively. CONCLUSIONS The study demonstrated an encouraging efficacy and manageable safety profile of neoadjuvant chemotherapy nab-paclitaxel plus carboplatin in stage III-IV, unresectable OC. In addition, AEs resulting in chemotherapy and surgery delays should be cautiously considered in this clinical setting. TRIAL REGISTRATION ClinicalTrials.gov, ChiCTR1900026893. Registered at 25 October 2019.
Collapse
Affiliation(s)
- Lina Yin
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Wei Jiang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Shuai Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Yi Fu
- Department of Radiology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Lin Zhou
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xuan Pei
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Shuang Ye
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Wenbin Shen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Huijuan Yang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| | - Boer Shan
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
20
|
Dominguez AA, Perz MT, Xu Y, Cedillo LG, Huang OD, McIntyre CA, Vudatha V, Trevino JG, Liu J, Wang P. Unveiling the Promise: Navigating Clinical Trials 1978-2024 for PDAC. Cancers (Basel) 2024; 16:3564. [PMID: 39518005 PMCID: PMC11544830 DOI: 10.3390/cancers16213564] [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: 09/10/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Despite many decades of research, pancreatic ductal adenocarcinoma (PDAC) remains one of the most difficult cancers to diagnose and treat effectively. Although there have been improvements in the 5-year overall survival rate, it is still very low at 12.5%. The limited efficacy of current therapies, even when PDAC is detected early, underscores the aggressive nature of the disease and the urgent need for more effective treatments. Clinical management of PDAC still relies heavily on a limited repertoire of therapeutic interventions, highlighting a significant gap between research efforts and available treatments. Over 4300 clinical trials have been or are currently investigating different treatment modalities and diagnostic strategies for PDAC, including targeted therapies, immunotherapies, and precision medicine approaches. These trials aim to develop more effective treatments and improve early detection methods through advanced imaging techniques and blood-based biomarkers. This review seeks to categorize and analyze PDAC-related clinical trials across various dimensions to understand why so few chemotherapeutic options are available to patients despite the numerous trials being conducted. This review aims to provide a comprehensive and nuanced understanding of the landscape of PDAC-related clinical trials, with the overarching goal of identifying opportunities to accelerate progress in drug development and improve patient outcomes in the fight against this devastating disease.
Collapse
Affiliation(s)
- Angel A. Dominguez
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Matthew T. Perz
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Yi Xu
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Leonor G. Cedillo
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Orry D. Huang
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Caitlin A. McIntyre
- Division of Surgical Oncology and Endocrine Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (V.V.); (J.G.T.)
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (V.V.); (J.G.T.)
| | - Jun Liu
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Pei Wang
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| |
Collapse
|
21
|
Negoro Y, Maeda T, Igarashi H, Shigemori M, Tanaka T, Ito Y, Tanizawa N, Nishikawa S, Ogawa J, Kamitani Y, Watanabe K, Tsukamoto H, Goto N. Dexamethasone is non-inferior to antihistamine plus dexamethasone premedication in preventing ramucirumab plus nab-paclitaxel infusion-related reactions in gastric cancer: a multicenter retrospective study. Support Care Cancer 2024; 32:704. [PMID: 39370483 PMCID: PMC11456548 DOI: 10.1007/s00520-024-08910-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE Ramucirumab (RAM) is recommended as premedication with H1-receptor antagonists (H1RA) to prevent infusion-related reactions (IRRs). However, RAM is a human antibody with a low incidence of IRRs. We evaluated the noninferiority of non-H1RA (dexamethasone [DEX] alone) premedication to H1RA (plus DEX) premedication in terms of IRRs in patients with gastric cancer receiving RAM plus nanoparticle albumin-bound paclitaxel (nab-PTX). METHODS This was a noninferiority, multicenter, retrospective trial conducted in three Japanese centers to assess the incidence of IRRs in patients receiving RAM plus nab-PTX for gastric cancer between 2018 and 2023. Patients with gastric cancer receiving RAM plus nab-PTX were divided into groups with and without H1RA premedication. The incidence of IRRs was compared between the two groups. RESULTS Ninety patients were evaluated, with non-H1RA and H1RA premedications in 43 and 47 cases, respectively. After the first dose of RAM, IRRs were not observed in either group. IRRs during the overall doses were 0% for non-H1RA premedication and 2.1% for H1RA premedication (90% confidence interval (CI): -5.6%-1.3% for each comparison). The upper limit of the 90% CI (1.3%) did not exceed the noninferiority margin (Δ) of + 10% and therefore met the noninferiority criteria. CONCLUSION RAM plus nab-PTX for gastric cancer with DEX premedication may be possible without H1RA premedication.
Collapse
Affiliation(s)
- Yutaka Negoro
- Department of Pharmacy, University of Fukui Hospital, Yoshida-Gun, 23-3 Matsuoka-Shimoaizuki, Eiheiji-Cho, Fukui, Japan.
| | - Taichi Maeda
- Department of Pharmacy, Japanese Red Cross Fukui Hospital, Fukui, Japan
| | | | - Mina Shigemori
- Department of Pharmacy, University of Fukui Hospital, Yoshida-Gun, 23-3 Matsuoka-Shimoaizuki, Eiheiji-Cho, Fukui, Japan
| | - Toshihiro Tanaka
- Department of Pharmacy, University of Fukui Hospital, Yoshida-Gun, 23-3 Matsuoka-Shimoaizuki, Eiheiji-Cho, Fukui, Japan
| | - Yukio Ito
- Department of Pharmacy, Fukui-Ken Saiseikai Hospital, Fukui, Japan
| | - Norihiko Tanizawa
- Department of Pharmacy, Japanese Red Cross Fukui Hospital, Fukui, Japan
| | - Shota Nishikawa
- Department of Pharmacy, Japanese Red Cross Fukui Hospital, Fukui, Japan
| | - Jyunya Ogawa
- Department of Pharmacy, Japanese Red Cross Fukui Hospital, Fukui, Japan
| | - Yukio Kamitani
- Department of Pharmacy, University of Fukui Hospital, Yoshida-Gun, 23-3 Matsuoka-Shimoaizuki, Eiheiji-Cho, Fukui, Japan
| | - Kyohei Watanabe
- Medical Research Support Center, University of Fukui Hospital, Yoshida-Gun, Fukui, Japan
| | - Hitoshi Tsukamoto
- Department of Pharmacy, University of Fukui Hospital, Yoshida-Gun, 23-3 Matsuoka-Shimoaizuki, Eiheiji-Cho, Fukui, Japan
| | - Nobuyuki Goto
- Department of Pharmacy, University of Fukui Hospital, Yoshida-Gun, 23-3 Matsuoka-Shimoaizuki, Eiheiji-Cho, Fukui, Japan
| |
Collapse
|
22
|
Yang D, Xu G, Ding H, Zhong L, Zhu J, Mi X, Xin W, Zhou T, Wang J, Fang L. Population pharmacokinetic and exposure-toxicity analyses of nab-paclitaxel after pegylated recombinant human granulocyte colony-stimulating factor administration in patients with metastatic breast cancer. Cancer Chemother Pharmacol 2024; 94:523-534. [PMID: 39080018 DOI: 10.1007/s00280-024-04702-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/12/2024] [Indexed: 09/29/2024]
Abstract
PURPOSE This study aimed to establish a population pharmacokinetic (PK) model to evaluate the dynamic relationship between the concentrations of total and unbound paclitaxel, and the exposure-response analysis of albumin-bound paclitaxel (nab-paclitaxel) after pegylated recombinant human granulocyte colony-stimulating factor (PEG-G-CSF) administration in patients with metastatic breast cancer. METHODS A total of 653 concentrations corresponding to total paclitaxel and 334 concentrations corresponding to unbound paclitaxel were analyzed in 24 subjects who randomized received a single 260 mg/m2 dose of two nab-paclitaxel formulations with a 21-35-day washout period. PEG-G-CSF was administered to all the patients in each cycle to prevent neutropenia. The exposure-response relationships were evaluated using the exposure to total, albumin-coated, and unbound paclitaxel, as well as the reduction in neutrophil count. The exposure data were analyzed using nonlinear mixed-effect modeling. A linear regression model was used to test the statistical significance of the correlation between percentage of reduction in neutrophil count and exposure. RESULTS The PK characteristics of total paclitaxel were described using a three-compartment model with first-order elimination, and a mechanism-based model incorporating linear release of nab-paclitaxel and the saturated binding of unbound paclitaxel to plasma components was established. The release ratio of paclitaxel from nab-paclitaxel was estimated to be 4.60% and the maximum unbound fraction (2.76%) was reached at the end of the infusion. The study found that a longer duration of total paclitaxel concentration > 0.19 µmol/L was significantly correlated with a reduction in neutrophil count (r2 = 0.23, P = 0.00062). Specifically, a duration of > 8.6 h was a predictor of a decreased neutrophil count. CONCLUSION The decrease in neutrophils induced by nab-paclitaxel was significantly correlated with the duration above a total paclitaxel concentration of 0.19 µmol/L despite the use of PEG-G-CSF.
Collapse
Affiliation(s)
- Dihong Yang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Gaoqi Xu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Haiying Ding
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Like Zhong
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Junfeng Zhu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Xiufang Mi
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Wenxiu Xin
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China
| | - Tianyan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jiaqi Wang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China.
| | - Luo Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 1 East Banshan Road, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
23
|
Fei Y, Li X, Lv Z, Liu Z, Xie Y, Chen J, Li W, Liu X, Guo H, Liu H, Zhang Z, Wang X, Fan J, Hu C, Jin X, Jiang R, Xu N, Xia J, Li Y, Shi D. Promoting chondrogenesis by targeted delivery to the degenerating cartilage in early treatment of osteoarthritis. Bioact Mater 2024; 40:624-633. [PMID: 39247402 PMCID: PMC11377143 DOI: 10.1016/j.bioactmat.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Osteoarthritis (OA) is a highly incident total joint degenerative disease with cartilage degeneration as the primary pathogenesis. The cartilage matrix is mainly composed of collagen, a matrix protein with a hallmark triple-helix structure, which unfolds with collagen degradation on the cartilage surface. A collagen hybridizing peptide (CHP) is a synthetic peptide that binds the denatured collagen triple helix, conferring a potential disease-targeting possibility for early-stage OA. Here, we constructed an albumin nanoparticle (An) conjugated with CHP, loaded with a chondrogenesis-promoting small molecule drug, kartogenin (KGN). The CHP-KGN-An particle exhibited sustained release of KGN in vitro and prolonged in vivo retention selectively within the degenerated cartilage in the knee joints of model mice with early-stage OA. Compared to treatment with KGN alone, CHP-KGN-An robustly attenuated cartilage degradation, synovitis, osteophyte formation, and subchondral bone sclerosis in OA model mice and exhibited a more prominent effect on physical activity improvement and pain alleviation. Our study showcases that targeting the degenerated cartilage by collagen hybridization can remarkably promote the efficacy of small molecule drugs and may provide a novel delivery strategy for early-stage OA therapeutics.
Collapse
Affiliation(s)
- Yuxiang Fei
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiaojing Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Zizheng Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ya Xie
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jiaqi Chen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Weitong Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiyu Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Hu Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Huan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Zhaofeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xunhao Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jingjing Fan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Chunqing Hu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiaoyu Jin
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ruiyang Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Nuo Xu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
| | - Yang Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| |
Collapse
|
24
|
Meng X, Shen Y, Zhao H, Lu X, Wang Z, Zhao Y. Redox-manipulating nanocarriers for anticancer drug delivery: a systematic review. J Nanobiotechnology 2024; 22:587. [PMID: 39342211 PMCID: PMC11438196 DOI: 10.1186/s12951-024-02859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024] Open
Abstract
Spatiotemporally controlled cargo release is a key advantage of nanocarriers in anti-tumor therapy. Various external or internal stimuli-responsive nanomedicines have been reported for their ability to increase drug levels at the diseased site and enhance therapeutic efficacy through a triggered release mechanism. Redox-manipulating nanocarriers, by exploiting the redox imbalances in tumor tissues, can achieve precise drug release, enhancing therapeutic efficacy while minimizing damage to healthy cells. As a typical redox-sensitive bond, the disulfide bond is considered a promising tool for designing tumor-specific, stimulus-responsive drug delivery systems (DDS). The intracellular redox imbalance caused by tumor microenvironment (TME) regulation has emerged as an appealing therapeutic target for cancer treatment. Sustained glutathione (GSH) depletion in the TME by redox-manipulating nanocarriers can exacerbate oxidative stress through the exchange of disulfide-thiol bonds, thereby enhancing the efficacy of ROS-based cancer therapy. Intriguingly, GSH depletion is simultaneously associated with glutathione peroxidase 4 (GPX4) inhibition and dihydrolipoamide S-acetyltransferase (DLAT) oligomerization, triggering mechanisms such as ferroptosis and cuproptosis, which increase the sensitivity of tumor cells. Hence, in this review, we present a comprehensive summary of the advances in disulfide based redox-manipulating nanocarriers for anticancer drug delivery and provide an overview of some representative achievements for combinational therapy and theragnostic. The high concentration of GSH in the TME enables the engineering of redox-responsive nanocarriers for GSH-triggered on-demand drug delivery, which relies on the thiol-disulfide exchange reaction between GSH and disulfide-containing vehicles. Conversely, redox-manipulating nanocarriers can deplete GSH, thereby enhancing the efficacy of ROS-based treatment nanoplatforms. In brief, we summarize the up-to-date developments of the redox-manipulating nanocarriers for cancer therapy based on DDS and provide viewpoints for the establishment of more stringent anti-tumor nanoplatform.
Collapse
Affiliation(s)
- Xuan Meng
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China.
| | - Yongli Shen
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China
| | - Huanyu Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China
| | - Xinlei Lu
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China
| | - Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China.
| |
Collapse
|
25
|
Hassan MS, Johnson C, Ponna S, Scofield D, Awasthi N, von Holzen U. Inhibition of Insulin-like Growth Factor 1 Receptor/Insulin Receptor Signaling by Small-Molecule Inhibitor BMS-754807 Leads to Improved Survival in Experimental Esophageal Adenocarcinoma. Cancers (Basel) 2024; 16:3175. [PMID: 39335147 PMCID: PMC11430189 DOI: 10.3390/cancers16183175] [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: 08/12/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The insulin-like growth factor-1 (IGF-1) and insulin axes are upregulated in obesity and obesity-associated esophageal adenocarcinoma (EAC). Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a contemporary nanotechnology-based paclitaxel (PT) bound to human albumin, ensuring its solubility in water rather than a toxic solvent. Here, we examined the benefits of inhibiting insulin-like growth factor-1 receptor/insulin receptor (IGF-1/IR) signaling and the enhancement of nab-paclitaxel effects by inclusion of the small-molecule inhibitor BMS-754807 using both in vitro and in vivo models of EAC. Using multiple EAC cell lines, BMS-754807 and nab-paclitaxel were evaluated as mono and combination therapies for in vitro effects on cell proliferation, cell death, and cell movement. We then analyzed the in vivo anticancer potency with survival improvement with BMS-754807 and nab-paclitaxel mono and combination therapies. BMS-754807 monotherapy suppressed in vitro cell proliferation and wound healing while increasing apoptosis. BMS-754807, when combined with nab-paclitaxel, enhanced those effects on the inhibition of cell proliferation, increment in cell apoptosis, and inhibition of wound healing. BMS-754807 with nab-paclitaxel produced substantially greater antitumor effects by increasing in vivo apoptosis, leading to increased mice survival compared to those of BMS-754807 or nab-paclitaxel monotherapy. Our outcomes support the use of BMS-754807, alone and in combination with nab-paclitaxel, as an efficient and innovative treatment choice for EAC.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Chloe Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Saisantosh Ponna
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Dimitri Scofield
- Department of Biology, Indiana University, South Bend, IN 47405, USA
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
- Goshen Center for Cancer Care, Goshen, IN 46526, USA
- School of Medicine, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
26
|
Adick A, Hoheisel W, Schneid S, Hester S, Langer K. Development of a screening platform for the formulation of poorly water-soluble drugs as albumin-stabilized nanosuspensions using nab™ technology. Int J Pharm 2024; 662:124491. [PMID: 39032872 DOI: 10.1016/j.ijpharm.2024.124491] [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: 05/06/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
The nanoparticle albumin bound™ (nab™) technology generally offers great potential for the formulation of poorly water-soluble drugs as albumin-stabilized nanosuspensions for intravenous use while avoiding solubilizers and cross-linking agents. The nab™ technology is a three-step process consisting of emulsification, high-pressure homogenization and solvent evaporation. Within this work, a screening approach was developed to predict whether active pharmaceutical ingredients are suitable for nab™ formulations. A design of experiments approach was used to investigate the effects of ultrasonic homogenization on an albumin-stabilized itraconazole nanosuspension. Based on this, a screening platform was developed, and subsequently evaluated and applied to a selection of poorly water-soluble drugs. The screening process to produce albumin-stabilized nanosuspensions consists of two process steps: Ultrasonic treatment, which combined emulsification and homogenization, followed by solvent evaporation. The results of the screening process were fully transferable to the standard three-step process of nab™ technology. In addition, based on drug screening, drug properties were highlighted that are important for the development of nab™ formulations. All in all, the nab™ technology is a promising but not universal formulation platform for poorly water-soluble drugs. Nevertheless, for some poorly soluble drugs it offers a valuable approach for the formulation of nanosuspensions for intravenous use.
Collapse
Affiliation(s)
- Annika Adick
- Institute of Pharmaceutical Technology and Biopharmacy, University Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| | - Werner Hoheisel
- Bayer AG, Process Technologies, Chempark, Building E41, 51368 Leverkusen, Germany.
| | - Stefan Schneid
- Bayer AG, Pharmaceuticals, Drug Product Development, Friedrich-Ebert-Straße 217-333, 42117 Wuppertal, Germany.
| | - Sarah Hester
- Institute of Pharmaceutical Technology and Biopharmacy, University Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| |
Collapse
|
27
|
Piper K, Kumar JI, Domino J, Tuchek C, Vogelbaum MA. Consensus review on strategies to improve delivery across the blood-brain barrier including focused ultrasound. Neuro Oncol 2024; 26:1545-1556. [PMID: 38770775 PMCID: PMC11376463 DOI: 10.1093/neuonc/noae087] [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: 05/21/2024] [Indexed: 05/22/2024] Open
Abstract
Drug delivery to the central nervous system (CNS) has been a major challenge for CNS tumors due to the impermeability of the blood-brain barrier (BBB). There has been a multitude of techniques aimed at overcoming the BBB obstacle aimed at utilizing natural transport mechanisms or bypassing the BBB which we review here. Another approach that has generated recent interest in the recently published literature is to use new technologies (Laser Interstitial Thermal Therapy, LITT; or Low-Intensity Focused Ultrasound, LIFU) to temporarily increase BBB permeability. This review overviews the advantages, disadvantages, and major advances of each method. LIFU has been a major area of research to allow for chemotherapeutics to cross the BBB which has a particular emphasis in this review. While most of the advances remain in animal studies, there are an increasing number of translational clinical trials that will have results in the next few years.
Collapse
Affiliation(s)
- Keaton Piper
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Jay I Kumar
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Joseph Domino
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Chad Tuchek
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Michael A Vogelbaum
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
28
|
Berardi AJ, Raymond JE, Chang A, Mauser AK, Lahann J. Self-Reporting Therapeutic Protein Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43350-43363. [PMID: 39106360 PMCID: PMC11898306 DOI: 10.1021/acsami.4c09114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
We present a modular strategy to synthesize nanoparticle sensors equipped with dithiomaleimide-based, fluorescent molecular reporters capable of discerning minute changes in interparticle chemical environments based on fluorescence lifetime analysis. Three types of nanoparticles were synthesized with the aid of tailor-made molecular reporters, and it was found that protein nanoparticles exhibited greater sensitivity to changes in the core environment than polymer nanogels and block copolymer micelles. Encapsulation of the hydrophobic small-molecule drug paclitaxel (PTX) in self-reporting protein nanoparticles induced characteristic changes in fluorescence lifetime profiles, detected via time-resolved fluorescence spectroscopy. Depending on the mode of drug encapsulation, self-reporting protein nanoparticles revealed pronounced differences in their fluorescence lifetime signatures, which correlated with burst- vs diffusion-controlled release profiles observed in previous reports. Self-reporting nanoparticles, such as the ones developed here, will be critical for unraveling nanoparticle stability and nanoparticle-drug interactions, informing the future development of rationally engineered nanoparticle-based drug carriers.
Collapse
Affiliation(s)
- Anthony J Berardi
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, Michigan 48105, United States
- Biointerfaces Institute, Ann Arbor, Michigan 48105, United States
| | - Jeffery E Raymond
- Biointerfaces Institute, Ann Arbor, Michigan 48105, United States
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
- Center for Complex Particle Systems, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Albert Chang
- Biointerfaces Institute, Ann Arbor, Michigan 48105, United States
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Ava K Mauser
- Biointerfaces Institute, Ann Arbor, Michigan 48105, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Joerg Lahann
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, Michigan 48105, United States
- Biointerfaces Institute, Ann Arbor, Michigan 48105, United States
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
| |
Collapse
|
29
|
Eker F, Duman H, Akdaşçi E, Bolat E, Sarıtaş S, Karav S, Witkowska AM. A Comprehensive Review of Nanoparticles: From Classification to Application and Toxicity. Molecules 2024; 29:3482. [PMID: 39124888 PMCID: PMC11314082 DOI: 10.3390/molecules29153482] [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: 07/03/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Nanoparticles are structures that possess unique properties with high surface area-to-volume ratio. Their small size, up to 100 nm, and potential for surface modifications have enabled their use in a wide range of applications. Various factors influence the properties and applications of NPs, including the synthesis method and physical attributes such as size and shape. Additionally, the materials used in the synthesis of NPs are primary determinants of their application. Based on the chosen material, NPs are generally classified into three categories: organic, inorganic, and carbon-based. These categories include a variety of materials, such as proteins, polymers, metal ions, lipids and derivatives, magnetic minerals, and so on. Each material possesses unique attributes that influence the activity and application of the NPs. Consequently, certain NPs are typically used in particular areas because they possess higher efficiency along with tenable toxicity. Therefore, the classification and the base material in the NP synthesis hold significant importance in both NP research and application. In this paper, we discuss these classifications, exemplify most of the major materials, and categorize them according to their preferred area of application. This review provides an overall review of the materials, including their application, and toxicity.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Ecem Bolat
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Anna Maria Witkowska
- Department of Food Biotechnology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
30
|
Qu N, Song K, Ji Y, Liu M, Chen L, Lee RJ, Teng L. Albumin Nanoparticle-Based Drug Delivery Systems. Int J Nanomedicine 2024; 19:6945-6980. [PMID: 39005962 PMCID: PMC11246635 DOI: 10.2147/ijn.s467876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/28/2024] [Indexed: 07/16/2024] Open
Abstract
Nanoparticle-based systems are extensively investigated for drug delivery. Among others, with superior biocompatibility and enhanced targeting capacity, albumin appears to be a promising carrier for drug delivery. Albumin nanoparticles are highly favored in many disease therapies, as they have the proper chemical groups for modification, cell-binding sites for cell adhesion, and affinity to protein drugs for nanocomplex generation. Herein, this review summarizes the recent fabrication techniques, modification strategies, and application of albumin nanoparticles. We first discuss various albumin nanoparticle fabrication methods, from both pros and cons. Then, we provide a comprehensive introduction to the modification section, including organic albumin nanoparticles, metal albumin nanoparticles, inorganic albumin nanoparticles, and albumin nanoparticle-based hybrids. We finally bring further perspectives on albumin nanoparticles used for various critical diseases.
Collapse
Affiliation(s)
- Na Qu
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Ke Song
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, the Netherlands
| | - Yating Ji
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Mingxia Liu
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Lijiang Chen
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, 130023, People's Republic of China
- College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, 130023, People's Republic of China
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Yantai, 264000, People's Republic of China
| |
Collapse
|
31
|
Kwok L, Mahmood A, Kumar S, Chang A. Case Report: Macula Structural and Functional Assessment in Nab-Paclitaxel-Related Cystoid Macular Edema. Ophthalmic Surg Lasers Imaging Retina 2024; 55:408-411. [PMID: 38531022 DOI: 10.3928/23258160-20240227-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Nab-paclitaxel-related cystoid macular edema is a rare ophthalmic adverse drug reaction. We present a 54-year-old woman with metastatic hormone receptor positive, HER-2 negative breast carcinoma who developed profound bilateral vision loss after the seventh cycle of nab-paclitaxel treatment. Optical coherence tomography and macula microperimetry demonstrated macular edema and decreased threshold sensitivity, respectively. Cessation of nab-paclitaxel improved structural and functional vision after 4 weeks. The introduction of topical dorzolamide 1% in one eye demonstrated further rapid resolution of edema. We demonstrate the objective and subjective visual changes and recovery of nab-paclitaxel-related cystoid macular edema. [Ophthalmic Surg Lasers Imaging Retina 2024;55:408-411.].
Collapse
|
32
|
Sun Y, Cheng Y, Hertz DL. Using maximum plasma concentration (C max) to personalize taxane treatment and reduce toxicity. Cancer Chemother Pharmacol 2024; 93:525-539. [PMID: 38734836 DOI: 10.1007/s00280-024-04677-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Taxanes are a widely used class of anticancer agents that play a vital role in the treatment of a variety of cancers. However, toxicity remains a major concern of using taxane drugs as some toxicities are highly prevalent, they can not only adversely affect patient prognosis but also compromise the overall treatment plan. Among all kinds of factors that associated with taxane toxicity, taxane exposure has been extensively studied, with different pharmacokinetic (PK) parameters being used as toxicity predictors. Compared to other widely used predictors such as the area under the drug plasma concentration curve versus time (AUC) and time above threshold plasma drug concentration, maximum plasma concentration (Cmax) is easier to collect and shows promise for use in clinical practice. In this article, we review the previous research on using Cmax to predict taxane treatment outcomes. While Cmax and toxicity have been extensively studied, research on the relationship between Cmax and efficacy is lacking. Most of the articles find a positive relationship between Cmax and toxicity but several articles have contradictory findings. Future clinical trials are needed to validate the relationship between Cmax and treatment outcome and determine whether Cmax can serve as a useful surrogate endpoint of taxane treatment efficacy.
Collapse
Affiliation(s)
- Yuchen Sun
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yue Cheng
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA.
| |
Collapse
|
33
|
Kida K, Yamada A, Shimada K, Narui K, Sugae S, Shimizu D, Doi T, Oba M, Endo I, Ishikawa T. A prospective comparison study utilizing patient-reported outcomes of taxane-related peripheral neuropathy between nab-paclitaxel and standard paclitaxel in patients with breast cancer. Breast Cancer 2024; 31:409-416. [PMID: 38453739 DOI: 10.1007/s12282-024-01551-z] [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: 09/24/2023] [Accepted: 02/04/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND: Characteristics of taxane-induced peripheral neuropathy (PN) could be different between paclitaxel and nab-paclitaxel. The purpose of this prospective observational multicenter cohort study was to compare tri-weekly nab-paclitaxel to weekly standard paclitaxel regarding the severity, onset and recovery of sensory and motor PN in patients with breast cancer. METHODS Patients with histologically confirmed breast cancer who were scheduled to receive standard weekly paclitaxel (80 mg/m2) or tri-weekly nab-paclitaxel (260 mg/m2) at institutions in our multicenter group were eligible for this study. Sensory and motor PN were evaluated every 3 weeks until PN improved for up to one year using patient-reported outcome. RESULTS Between February 2011 and April 2013, 115 patients were enrolled, including 57 and 58 in the paclitaxel and nab-paclitaxel groups, respectively. The incidence of moderate or severe sensory PN was not significantly different between the two groups (p = 0.40). The incidence of moderate or higher motor PN was more frequent in the nab-paclitaxel group than in the paclitaxel group (p = 0.048). The median period for demonstrating PN were shorter in the nab-paclitaxel group than in the paclitaxel group (sensory, p = 0.003; motor, p = 0.001). The recovery of motor PN was slower in the nab-paclitaxel group than in the paclitaxel group (p = 0.035), while the recovery period of sensory PN was not statistically different. CONCLUSION Nab-paclitaxel induced sensory PN sooner than paclitaxel, and no difference was observed in the severity and recovery duration between the two agents. Motor PN was more severe, started sooner, and improved over a longer period in the nab-paclitaxel-treated patients than in the paclitaxel-treated patients.
Collapse
Affiliation(s)
- Kumiko Kida
- Department of Gastroenterological Surgery and Surgical Oncology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
- Department of Breast Surgical Oncology, St. Luke's International Hospital, Tokyo, Japan
| | - Akimitsu Yamada
- Department of Gastroenterological Surgery and Surgical Oncology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Kazuhiro Shimada
- Department of Breast Surgery, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
- Department of Surgery, Yokohama Saiseikai Nanbu Hospital, Kanagawa, Japan
| | - Kazutaka Narui
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Kanagawa, Japan
| | - Sadatoshi Sugae
- Department of Gastroenterological Surgery and Surgical Oncology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
- Department of Breast Surgery, Fujisawa City Hospital, Kanagawa, Japan
| | - Daisuke Shimizu
- Department of Breast Surgery, Yokohama Minato Red Cross Hospital, Kanagawa, Japan
| | - Takako Doi
- Department of Breast Surgery, Shonan Memorial Hospital, Kanagawa, Japan
| | - Mari Oba
- Department of Medical Statistics, Toho University, Tokyo, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery and Surgical Oncology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
34
|
Vahedi F, Javan B, Sharbatkhari M, Soltani A, Shafiee M, Memarian A, Erfani-Moghadam V. Synergistic anticancer effects of co-delivery of linc-RoR siRNA and curcumin using polyamidoamine dendrimers against breast cancer. Biochem Biophys Res Commun 2024; 705:149729. [PMID: 38452515 DOI: 10.1016/j.bbrc.2024.149729] [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: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Breast cancer resistance to chemotherapy necessitates novel combination therapeutic approaches. Linc-RoR is a long intergenic noncoding RNA that regulates stem cell differentiation and promotes metastasis and invasion in breast cancer. Herein, we report a dual delivery system employing polyamidoamine dendrimers to co-administer the natural compound curcumin and linc-RoR siRNA for breast cancer treatment. Polyamidoamine dendrimers efficiently encapsulated curcumin and formed complexes with linc-RoR siRNA at an optimal N/P ratio. In MCF-7 breast cancer cells, the dendriplexes were effectively internalized and the combination treatment synergistically enhanced cytotoxicity, arresting the cell cycle at the G1 phase and inducing apoptosis. Linc-RoR gene expression was also significantly downregulated. Individual treatments showed lower efficacy, indicating synergism between components. Mechanistic studies are warranted to define the molecular underpinnings of this synergistic interaction. Our findings suggest dual delivery of linc-RoR siRNA and curcumin via dendrimers merits further exploration as a personalized therapeutic approach for overcoming breast cancer resistance.
Collapse
Affiliation(s)
- Farzaneh Vahedi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Bita Javan
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Alireza Soltani
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Shafiee
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran; Stem Cell Research Center, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Memarian
- Stem Cell Research Center, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran; Department of Immunology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Erfani-Moghadam
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran; Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
35
|
Symons R, Heath F, Duggan J, Bui KT, Byun L, Friedlander M, Lee YC. Rates of paclitaxel hypersensitivity reactions using a modified Markman's infusion protocol as primary prophylaxis. Support Care Cancer 2024; 32:292. [PMID: 38632132 PMCID: PMC11023961 DOI: 10.1007/s00520-024-08460-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Markman's desensitisation protocol allows successful retreatment of patients who have had significant paclitaxel hypersensitivity reactions. We aimed to reduce the risk and severity of paclitaxel hypersensitivity reactions by introducing this protocol as primary prophylaxis. METHODS We evaluated all patients with a gynaecological malignancy receiving paclitaxel before (December 2018 to September 2019) and after (October 2019 to July 2020) the implementation of a modified Markman's desensitisation protocol. The pre-implementation group received paclitaxel over a gradually up-titrated rate from 60 to 180 ml/h. The post-implementation group received paclitaxel via 3 fixed-dose infusion bags in the first 2 cycles. Rates and severity of paclitaxel hypersensitivity reactions were compared. RESULTS A total of 426 paclitaxel infusions were administered to 78 patients. The median age was 64 years (range 34-81), and the most common diagnosis was ovarian, fallopian tube and primary peritoneal cancer (67%, n = 52/78). Paclitaxel hypersensitivity reaction rates were similar in the pre-implementation (8%, n = 16/195) and post-implementation groups (9%, n = 20/231; p = 0.87). Most paclitaxel hypersensitivity reactions occurred within 30 min (pre- vs. post-implementation, 88% [n = 14/16] vs. 75% [n = 15/20]; p = 0.45) and were grade 2 in severity (pre- vs. post-implementation, 81% [n = 13/16] vs. 75% [n = 15/20]; p = 0.37). There was one grade 3 paclitaxel hypersensitivity reaction in the pre-implementation group. All patients were successfully rechallenged in the post-implementation group compared to 81% (n = 13/16) in the pre-implementation group (p = 0.43). CONCLUSION The modified Markman's desensitisation protocol as primary prophylaxis did not reduce the rate or severity of paclitaxel hypersensitivity reactions, although all patients could be successfully rechallenged.
Collapse
Affiliation(s)
- Rebecca Symons
- Royal Hospital for Women, Randwick, NSW, Australia
- Prince of Wales Hospital, Randwick, NSW, Australia
| | - Fiona Heath
- Royal Hospital for Women, Randwick, NSW, Australia
| | | | - Kim Tam Bui
- Royal Hospital for Women, Randwick, NSW, Australia
- Prince of Wales Hospital, Randwick, NSW, Australia
| | - Lily Byun
- Royal Hospital for Women, Randwick, NSW, Australia
| | - Michael Friedlander
- Royal Hospital for Women, Randwick, NSW, Australia
- Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Yeh Chen Lee
- Royal Hospital for Women, Randwick, NSW, Australia.
- Prince of Wales Hospital, Randwick, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
36
|
Petersen EF, Larsen BS, Nielsen RB, Pijpers I, Versweyveld D, Holm R, Tho I, Snoeys J, Nielsen CU. Co-release of paclitaxel and encequidar from amorphous solid dispersions increase oral paclitaxel bioavailability in rats. Int J Pharm 2024; 654:123965. [PMID: 38442796 DOI: 10.1016/j.ijpharm.2024.123965] [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: 01/22/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
The oral bioavailability of paclitaxel is limited due to low solubility and high affinity for the P-glycoprotein (P-gp) efflux transporter. Here we hypothesized that maximizing the intestinal paclitaxel levels through apparent solubility enhancement and controlling thesimultaneous release of both paclitaxel and the P-gp inhibitor encequidar from amorphous solid dispersions (ASDs) would increase the oral bioavailability of paclitaxel. ASDs of paclitaxel and encequidar in polyvinylpyrrolidone K30 (PVP-K30), hydroxypropylmethylcellulose 5 (HPMC-5), and hydroxypropylmethylcellulose 4 K (HPMC-4K) were hence prepared by freeze-drying. In vitro dissolution studies showed that both compounds were released fastest from PVP-K30, then from HPMC-5, and slowest from HPMC-4K ASDs. The dissolution of paclitaxel from all polymers resulted in stable concentration levels above the apparent solubility. The pharmacokinetics of paclitaxel after oral administration to male Sprague-Dawley rats was investigated with or without 1 mg/kg encequidar, as amorphous solids or polymer-based ASDs. The bioavailability of paclitaxel increased 3- to 4-fold when administered as polymer-based ASDs relative to solid amorphous paclitaxel. However, when amorphous paclitaxel was co-administered with encequidar, either as an amorphous powder or as a polymer-based ASD, the bioavailability increased 2- to 4-fold, respectively. Interestingly, a noticeable increase in paclitaxel bioavailability of 24-fold was observed when paclitaxel and encequidar were co-administered as HPMC-5-based ASDs. We, therefore, suggest that controlling the dissolution rate of paclitaxel and encequidar in order to obtain simultaneous and timed release from polymer-based ASDs is a strategy to increase oral paclitaxel bioavailability.
Collapse
Affiliation(s)
- Emilie Fynbo Petersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Bjarke Strøm Larsen
- Department of Pharmacy, University of Oslo, Sem Sælands vei 3, NO-0371 Oslo, Norway
| | - Rasmus Blaaholm Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Ils Pijpers
- Bioanalytical Discovery and Development Sciences, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, BE-2340 Beerse, Belgium
| | - Dries Versweyveld
- In vivo Sciences, Preclinical Sciences & Translational Safety (PSTS), Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, BE-2340 Beerse, Belgium
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Ingunn Tho
- Department of Pharmacy, University of Oslo, Sem Sælands vei 3, NO-0371 Oslo, Norway
| | - Jan Snoeys
- Translational Pharmacokinetics Pharmacodynamics and Investigative Toxicology, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, BE-2340 Beerse, Belgium
| | - Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.
| |
Collapse
|
37
|
Liu Q, Xu R, Shen J, Tao Y, Shao J, Ke Y, Liu B. In situ chemoimmunotherapy hydrogel elicits immunogenic cell death and evokes efficient antitumor immune response. J Transl Med 2024; 22:341. [PMID: 38594751 PMCID: PMC11005214 DOI: 10.1186/s12967-024-05102-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/15/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Chemoimmunotherapy has shown promising advantages of eliciting immunogenic cell death and activating anti-tumor immune responses. However, the systemic toxicity of chemotherapy and tumor immunosuppressive microenvironment limit the clinical application. METHODS Here, an injectable sodium alginate hydrogel (ALG) loaded with nanoparticle albumin-bound-paclitaxel (Nab-PTX) and an immunostimulating agent R837 was developed for local administration. Two murine hepatocellular carcinoma and breast cancer models were established. The tumor-bearing mice received the peritumoral injection of R837/Nab-PTX/ALG once a week for two weeks. The antitumor efficacy, the immune response, and the tumor microenvironment were investigated. RESULTS This chemoimmunotherapy hydrogel with sustained-release character was proven to have significant effects on killing tumor cells and inhibiting tumor growth. Peritumoral injection of our hydrogel caused little harm to normal organs and triggered a potent antitumor immune response against both hepatocellular carcinoma and breast cancer. In the tumor microenvironment, enhanced immunogenic cell death induced by the combination of Nab-PTX and R837 resulted in 3.30-fold infiltration of effector memory T cells and upregulation of 20 biological processes related to immune responses. CONCLUSIONS Our strategy provides a novel insight into the combination of chemotherapy and immunotherapy and has the potential for clinical translation.
Collapse
Affiliation(s)
- Qin Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Rui Xu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jingwen Shen
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yaping Tao
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jingyi Shao
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yaohua Ke
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
38
|
Maphutha J, Twilley D, Lall N. The Role of the PTEN Tumor Suppressor Gene and Its Anti-Angiogenic Activity in Melanoma and Other Cancers. Molecules 2024; 29:721. [PMID: 38338464 PMCID: PMC10856229 DOI: 10.3390/molecules29030721] [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: 12/04/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Human malignant melanoma and other solid cancers are largely driven by the inactivation of tumor suppressor genes and angiogenesis. Conventional treatments for cancer (surgery, radiation therapy, and chemotherapy) are employed as first-line treatments for solid cancers but are often ineffective as monotherapies due to resistance and toxicity. Thus, targeted therapies, such as bevacizumab, which targets vascular endothelial growth factor, have been approved by the US Food and Drug Administration (FDA) as angiogenesis inhibitors. The downregulation of the tumor suppressor, phosphatase tensin homolog (PTEN), occurs in 30-40% of human malignant melanomas, thereby elucidating the importance of the upregulation of PTEN activity. Phosphatase tensin homolog (PTEN) is modulated at the transcriptional, translational, and post-translational levels and regulates key signaling pathways such as the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) pathways, which also drive angiogenesis. This review discusses the inhibition of angiogenesis through the upregulation of PTEN and the inhibition of hypoxia-inducible factor 1 alpha (HIF-1-α) in human malignant melanoma, as no targeted therapies have been approved by the FDA for the inhibition of angiogenesis in human malignant melanoma. The emergence of nanocarrier formulations to enhance the pharmacokinetic profile of phytochemicals that upregulate PTEN activity and improve the upregulation of PTEN has also been discussed.
Collapse
Affiliation(s)
- Jacqueline Maphutha
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Danielle Twilley
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
- School of Natural Resources, University of Missouri, Columbia, MO 65211, USA
- College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
| |
Collapse
|
39
|
Chen W, Wang W, Xie Z, Centurion F, Sun B, Paterson DJ, Tsao SCH, Chu D, Shen Y, Mao G, Gu Z. Size-Dependent Penetration of Nanoparticles in Tumor Spheroids: A Multidimensional and Quantitative Study of Transcellular and Paracellular Pathways. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304693. [PMID: 37822153 DOI: 10.1002/smll.202304693] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/16/2023] [Indexed: 10/13/2023]
Abstract
Tumor penetration of nanoparticles is crucial in nanomedicine, but the mechanisms of tumor penetration are poorly understood. This work presents a multidimensional, quantitative approach to investigate the tissue penetration behavior of nanoparticles, with focuses on the particle size effect on penetration pathways, in an MDA-MB-231 tumor spheroid model using a combination of spectrometry, microscopy, and synchrotron beamline techniques. Quasi-spherical gold nanoparticles of different sizes are synthesized and incubated with 2D and 3D MDA-MB-231 cells and spheroids with or without an energy-dependent cell uptake inhibitor. The distribution and penetration pathways of nanoparticles in spheroids are visualized and quantified by inductively coupled plasma mass spectrometry, two-photon microscopy, and synchrotron X-ray fluorescence microscopy. The results reveal that 15 nm nanoparticles penetrate spheroids mainly through an energy-independent transcellular pathway, while 60 nm nanoparticles penetrate primarily through an energy-dependent transcellular pathway. Meanwhile, 22 nm nanoparticles penetrate through both transcellular and paracellular pathways and they demonstrate the greatest penetration ability in comparison to other two sizes. The multidimensional analytical methodology developed through this work offers a generalizable approach to quantitatively study the tissue penetration of nanoparticles, and the results provide important insights into the designs of nanoparticles with high accumulation at a target site.
Collapse
Affiliation(s)
- Wenjing Chen
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Wenqian Wang
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zhouzun Xie
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Franco Centurion
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Bin Sun
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | | | - Simon Chang-Hao Tsao
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
- Department of Surgery, Austin Hospital, University of Melbourne, Melbourne, VIC, 3084, Australia
| | - Dewei Chu
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Yansong Shen
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zi Gu
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW RNA Institute, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
40
|
Scianò F, Terrana F, Pecoraro C, Parrino B, Cascioferro S, Diana P, Giovannetti E, Carbone D. Exploring the therapeutic potential of focal adhesion kinase inhibition in overcoming chemoresistance in pancreatic ductal adenocarcinoma. Future Med Chem 2024; 16:271-289. [PMID: 38269431 DOI: 10.4155/fmc-2023-0234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 01/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related deaths worldwide. Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase often overexpressed in PDAC. FAK has been linked to cell migration, survival, proliferation, angiogenesis and adhesion. This review first highlights the chemoresistant nature of PDAC. Second, the role of FAK in PDAC cancer progression and resistance is carefully described. Additionally, it discusses recent developments of FAK inhibitors as valuable drugs in the treatment of PDAC, with a focus on diamine-substituted-2,4-pyrimidine-based compounds, which represent the most potent class of FAK inhibitors in clinical trials for the treatment of PDAC disease. To conclude, relevant computational studies performed on FAK inhibitors are reported to highlight the key structural features required for interaction with the protein, with the aim of optimizing this novel targeted therapy.
Collapse
Affiliation(s)
- Fabio Scianò
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Francesca Terrana
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Camilla Pecoraro
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Barbara Parrino
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Stella Cascioferro
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Patrizia Diana
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Via Ferruccio Giovannini 13, San Giuliano Terme, Pisa, 56017, Italy
| | - Daniela Carbone
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| |
Collapse
|
41
|
Passos JS, Apolinario AC, Ishida K, Martins TS, Lopes LB. Nanostructured lipid carriers loaded into in situ gels for breast cancer local treatment. Eur J Pharm Sci 2024; 192:106638. [PMID: 37967657 DOI: 10.1016/j.ejps.2023.106638] [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: 07/04/2023] [Revised: 10/18/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
In this study, nanostructured lipid carriers (NLC) were developed and employed to obtain in situ thermosensitive formulations for the ductal administration and prolonged retention of drugs as a new strategy for breast cancer local treatment. NLC size was influenced by the type and concentration of the oil phase, surfactants, and drug incorporation, ranging from 221.6 to 467.5 nm. The type of liquid lipid influenced paclitaxel and 5-fluorouracil cytotoxicity, with tributyrin-containing NLC reducing IC50 values by 2.0-7.0-fold compared to tricaprylin NLC in MCF-7, T-47D and MDA-MB-231 cells. In spheroids, the NLCs reduced IC50 compared to either drug solution (3.2-6.2-fold). Although a significant reduction (1.26 points, p < 0.001) on the health index of Galleria mellonella larvae was observed 5 days after NLC administration, survival was not significantly reduced. To produce thermosensitive gels, the NLCs were incorporated in a poloxamer (11 %, w/w) dispersion, which gained viscosity (2-fold) at 37 °C. After 24 h, ∼53 % of paclitaxel and 83 % of 5-fluorouracil were released from the NLC; incorporation in the poloxamer gel further prolonged release. Intraductal administration of NLC-loaded gel increased the permanence of hydrophilic (2.2-3.0-fold) and lipophilic (2.1-2.3-fold) fluorescent markers in the mammary tissue compared to the NLC (as dispersion) and the markers solutions. In conclusion, these results contribute to improving our understanding of nanocarrier design with increased cytotoxicity and prolonged retention for the intraductal route. Tributyrin incorporation increased the cytotoxicity of paclitaxel and 5-fluorouracil in monolayer and spheroids, while NLC incorporation in thermosensitive gels prolonged tissue retention of both hydrophilic and hydrophobic compounds.
Collapse
Affiliation(s)
- Julia S Passos
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Av. Prof. Lineu Prestes, Sao Paulo SP 05508-000, Brazil
| | - Alexsandra C Apolinario
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Av. Prof. Lineu Prestes, Sao Paulo SP 05508-000, Brazil
| | - Kelly Ishida
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Tereza S Martins
- Department of Chemistry, Federal University of Sao Paulo (UNIFESP), Diadema, São Paulo, Brazil
| | - Luciana B Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Av. Prof. Lineu Prestes, Sao Paulo SP 05508-000, Brazil.
| |
Collapse
|
42
|
Paramasivam G, Sanmugam A, Palem VV, Sevanan M, Sairam AB, Nachiappan N, Youn B, Lee JS, Nallal M, Park KH. Nanomaterials for detection of biomolecules and delivering therapeutic agents in theragnosis: A review. Int J Biol Macromol 2024; 254:127904. [PMID: 37939770 DOI: 10.1016/j.ijbiomac.2023.127904] [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: 06/25/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Nanomaterials are emerging facts used to deliver therapeutic agents in living systems. Nanotechnology is used as a compliment by implementing different kinds of nanotechnological applications such as nano-porous structures, functionalized nanomaterials, quantum dots, carbon nanomaterials, and polymeric nanostructures. The applications are in the initial stage, which led to achieving several diagnoses and therapy in clinical practice. This review conveys the importance of nanomaterials in post-genomic employment, which includes the design of immunosensors, immune assays, and drug delivery. In this view, genomics is a molecular tool containing large databases that are useful in choosing an apt molecular inhibitor such as drug, ligand and antibody target in the drug delivery process. This study identifies the expression of genes and proteins in analysis and classification of diseases. Experimentally, the study analyses the design of a disease model. In particular, drug delivery is a boon area to treat cancer. The identified drugs enter different phase trails (Trails I, II, and III). The genomic information conveys more essential entities to the phase I trials and helps to move further for other trails such as trails-II and III. In such cases, the biomarkers play a crucial role by monitoring the unique pathological process. Genetic engineering with recombinant DNA techniques can be employed to develop genetically engineered disease models. Delivering drugs in a specific area is one of the challenging issues achieved using nanoparticles. Therefore, genomics is considered as a vast molecular tool to identify drugs in personalized medicine for cancer therapy.
Collapse
Affiliation(s)
- Gokul Paramasivam
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha Nagar, Thandalam, Chennai 602105, Tamil Nadu, India.
| | - Anandhavelu Sanmugam
- Department of Applied Chemistry, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur 602117, Tamil Nadu, India
| | - Vishnu Vardhan Palem
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha Nagar, Thandalam, Chennai 602105, Tamil Nadu, India
| | - Murugan Sevanan
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Karunya Nagar, Coimbatore 641114, Tamil Nadu, India
| | - Ananda Babu Sairam
- Department of Applied Chemistry, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur 602117, Tamil Nadu, India
| | - Nachiappan Nachiappan
- Department of Applied Chemistry, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur 602117, Tamil Nadu, India
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Jung Sub Lee
- Department of Orthopaedic Surgery, Biomedical Research Institute, Pusan National University Hospital, Busan 46241, Republic of Korea; School of Medicine, Pusan National University, Busan 46241, Republic of Korea
| | - Muthuchamy Nallal
- Department of Chemistry, Pusan National University, Busan 46241, Republic of Korea.
| | - Kang Hyun Park
- Department of Chemistry, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
43
|
Dylewski JF, Haddad G, Blaine J. Exploiting the neonatal crystallizable fragment receptor to treat kidney disease. Kidney Int 2024; 105:54-64. [PMID: 38707675 PMCID: PMC11068363 DOI: 10.1016/j.kint.2023.09.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 05/07/2024]
Abstract
The neonatal Fc receptor (FcRn) was initially discovered as the receptor that allowed passive immunity in newborns by transporting maternal IgG through the placenta and enterocytes. Since its initial discovery, FcRn has been found to exist throughout all stages of life and in many different cell types. Beyond passive immunity, FcRn is necessary for intrinsic albumin and IgG recycling and is important for antigen processing and presentation. Given its multiple important roles, FcRn has been utilized in many disease treatments including a new class of agents that were developed to inhibit FcRn for treatment of a variety of autoimmune diseases. Certain cell populations within the kidney also express high levels of this receptor. Specifically, podocytes, proximal tubule epithelial cells, and vascular endothelial cells have been found to utilize FcRn. In this review, we summarize what is known about FcRn and its function within the kidney. We also discuss how FcRn has been used for therapeutic benefit, including how newer FcRn inhibiting agents are being used to treat autoimmune diseases. Lastly, we will discuss what renal diseases may respond to FcRn inhibitors and how further work studying FcRn within the kidney may lead to therapies for kidney diseases.
Collapse
Affiliation(s)
- James F. Dylewski
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Nephrology, Denver Health Medical Center, Denver, CO, USA
| | - George Haddad
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Judith Blaine
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
44
|
Sulthana S, Bhatti A, Mathew E, Quazi SH, Gaudreault NN, DeLong R, Aryal S. Synthetic graphene-copper nanocomposites interact with the hACE-2 enzyme and inhibit its biochemical activity. NANOSCALE ADVANCES 2023; 6:188-196. [PMID: 38125590 PMCID: PMC10729868 DOI: 10.1039/d3na00468f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023]
Abstract
This study demonstrates the copper nanocomposite-induced enzymatic inhibition of human angiotensin I-converting enzyme-2 (hACE-2) by complex stabilization through the formation of the enzyme nanocomposite. The immediate application of this work is related to ACE-2 as a mechanism of SARS-CoV-2 entry into cells. Moreover, ACE-2 enzyme regulation is a potential therapeutic strategy in hypertension and cardiovascular disease, diabetes, lung injury, and fibrotic disorders. Thus, inhibition of ACE-2 with nanocomposite therapy, may have pharmacologic application with regard to infectious and non-infectious diseases. Synthesized copper nanocomposites described here alone with a commercially available compound, were tested for their potential to inhibit hACE-2 activities. Following wet chemical synthesis, Cu/CuO nanoparticles and graphene-copper (GO-Cu) complexes were synthesized and characterized for their chemical integrity. Cu/CuO formed well-dispersed clusters of 390 ± 100 nm, that when complexed with the hACE-2 enzyme exhibited larger clusters of 506 ± 56 nm. The formation of the Cu/CuO and hACE-2 enzyme complex was monitored by analyzing the zeta potential, which reflected the surface charge distribution of the complex. A negatively charged Cu/CuO nanocomposite nearly becomes neutral when complexed with hACE-2 further assuring the complex formation. Formation of this complex and its inactivation of hACE-2 was evaluated using a standardized protocal for enzymatic activity. Similarly, carboxylate-functionalized graphene was complexed with copper, and its inhibitory effect was studied. Each step in the GO-Cu composite formation was monitored by characterizing its surface electrical properties, resulting in a decrease in its zeta potential and conductivity when complexed with copper. The interaction of the nanocomposites with hACE-2 was confirmed by 2D-FDS and gel electrophoresis analysis. GO-Cu was a rapid and efficacious inhibitor compared to Cu-CuO, especially at lower concentrations (2 μg ml-1). Considering the environmental friendliness of copper and graphene and their use in industries as surface coating materials, we anticipate that use of these composites once proven effective, may have future antimicrobial application. Utility of nanocomposites as antimicrobials, either as a surface antimicrobial or as an in vivo therapeutic, could be invisioned for use against current unknown and/or emergent pathogens.
Collapse
Affiliation(s)
- Shoukath Sulthana
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler Tyler TX 75799 USA
| | - Abeera Bhatti
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University Manhattan KS 66506 USA
| | - Elza Mathew
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University Manhattan KS 66506 USA
| | - Sohel H Quazi
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler Tyler TX 75799 USA
- Department of Biology, Division of Natural and Computational Sciences, Texas College Tyler TX 75702 USA
| | - Natasha N Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University Manhattan KS 66506 USA
| | - Robert DeLong
- Landmark Bio, Innovation Development Laboratory Watertown MA 02472 USA
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler Tyler TX 75799 USA
| |
Collapse
|
45
|
Adick A, Hoheisel W, Schneid S, Mulac D, Azhdari S, Langer K. Challenges of nanoparticle albumin bound (nab™) technology: Comparative study of Abraxane® with a newly developed albumin-stabilized itraconazole nanosuspension. Eur J Pharm Biopharm 2023; 193:129-143. [PMID: 37918678 DOI: 10.1016/j.ejpb.2023.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Nanoparticle albumin bound™ (nab™) technology is an established delivery platform for development of albumin stabilized nanoparticles as drug delivery systems for poorly water-soluble drugs. By using albumin for particle stabilization, nab™ technology does not require solubilizers or emulsifiers for the formulation of poorly water-soluble drugs for intravenous use. Despite the great potential, however, to date only two products based on nab™ technology have been approved by the Food and Drug Administration: Abraxane® (nab™ paclitaxel) and Fyarro® (nab™ rapamycin). In this study, the commercially available product Abraxane® was characterized in comparison to an albumin stabilized nanosuspension for the poorly water-soluble drug itraconazole. The aim of this study was to identify critical product parameters of the nanosuspensions depending on the manufacturing process in order to assess the transferability of nab™ technology to other drugs. The colloidal properties, stabilizing protein composition and particle disintegration behavior were analyzed. In addition, studies were carried out on the impact of the key process step, the high-pressure homogenization, using a design of experiments (DoE) approach. A nanosuspension comprising spherical, stable drug nanoparticles stabilized by a large fraction of dissolved albumin around the nanoparticles were identified. During the manufacturing process, the drug core was coated with a layer of albumin, which was cross-linked to a certain level. The Abraxane® and itraconazole suspensions differed in the analyzed protein fraction, with stronger cross-linking at the particle surface for Abraxane®. Both active pharmaceutical ingredients were present in the amorphous state as nanoparticles. In vitro disintegration studies performed to mimic a strong dilution during intravenous application showed the disintegration of the nanoparticles. All in all, the analysis underlined the transferability of the nab™ technology to selected other poorly water-soluble drugs with the great advantage of eliminating solubilizers and emulsifiers for intravenous applications.
Collapse
Affiliation(s)
- Annika Adick
- Institute of Pharmaceutical Technology and Biopharmacy, University Muenster, Corrensstraße 48, 48149 Muenster, Germany
| | - Werner Hoheisel
- Invite GmbH, Formulation Technology, Chempark, Building W 32, 51368 Leverkusen, Germany
| | - Stefan Schneid
- Bayer AG, Pharmaceuticals, Drug Product Development, Friedrich-Ebert-Straße 475, 42117 Wuppertal, Germany
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University Muenster, Corrensstraße 48, 48149 Muenster, Germany
| | - Suna Azhdari
- Institute of Physical Chemistry, University Muenster, Corrensstraße 28/30, 48149 Muenster, Germany
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| |
Collapse
|
46
|
Hester S, B Ferenz K, Adick A, Kakalias C, Mulac D, Azhdari S, Langer K. Triglyceride-filled albumin-based nanocapsules: A promising new system to avoid discarding poorly water-soluble drug candidates. Int J Pharm 2023; 646:123454. [PMID: 37776966 DOI: 10.1016/j.ijpharm.2023.123454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Even though current drug discovery provides a variety of potential drug candidates, many of those substances are difficult to formulate due to their poor water-solubility. To overcome this obstacle a technological formulation is crucial. Albumin-based nanocarriers are a possible intravenous delivery system which is already approved and commercially available. However, no universal carrier for poorly water-soluble substances is found yet. In the present study, new preparation processes for nanocapsules consisting of a medium-chain triglyceride (MCT) core and a human serum albumin (HSA) shell were developed. The nanocarrier system exhibits desirable physicochemical properties with a hydrodynamic diameter of 150 nm and a polydispersity index of 0.1. Furthermore, the nanocapsules were stable towards the addition of electrolytes and also in basic to neutral pH range. The nanocapsules were storage stable for at least 7 months at 4 °C and could also be lyophilized to reach an even longer shelf life of at least 21 months. In addition, the nanocapsule system showed no cytotoxicity in cell culture. The developed system represents a suitable carrier for a variety of different poorly water-soluble drug substances (e.g., fenofibrate, naproxen, indomethacin) showing a high potential for a universal formulation platform for further lipophilic active pharmaceutical ingredients (APIs).
Collapse
Affiliation(s)
- Sarah Hester
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany.
| | - Katja B Ferenz
- Institute of Physiology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany.
| | - Annika Adick
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany.
| | - Christos Kakalias
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany.
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany.
| | - Suna Azhdari
- Institute of Physical Chemistry, University of Muenster, Corrensstr. 28/30, 48149 Muenster, Germany.
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany.
| |
Collapse
|
47
|
Li T, Luo R, Su L, Lv F, Mei L, Yu Y. Advanced Materials and Delivery Systems for Enhancement of Chimeric Antigen Receptor Cells. SMALL METHODS 2023; 7:e2300880. [PMID: 37653606 DOI: 10.1002/smtd.202300880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/12/2023] [Indexed: 09/02/2023]
Abstract
Chimeric antigen receptor (CAR) cell therapy is a great success and breakthrough in immunotherapy. However, there are still lots of barriers to its wide use in clinical, including long time consumption, high cost, and failure against solid tumors. For these challenges, researches are deplored to explore CAR cells to more appliable products in clinical. This minireview focuses on the advanced non-viral materials for CAR-T transfection ex vivo with better performance, delivery systems combined with other therapy for enhancement of CAR-T therapy in solid tumors. In addition, the targeted delivery platform for CAR cells in vivo generation as a breakthrough technology as its low cost and convenience. In the end, the prospective direction and future of CAR cell therapy are discussed.
Collapse
Affiliation(s)
- Tingxuan Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Ran Luo
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Lina Su
- Department of Pharmacy, Qujing Medical College, Qujing, Yunnan, 655000, P. R. China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yongkang Yu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
48
|
Kaltbeitzel J, Wich PR. Protein-based Nanoparticles: From Drug Delivery to Imaging, Nanocatalysis and Protein Therapy. Angew Chem Int Ed Engl 2023; 62:e202216097. [PMID: 36917017 DOI: 10.1002/anie.202216097] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/16/2023]
Abstract
Proteins and enzymes are versatile biomaterials for a wide range of medical applications due to their high specificity for receptors and substrates, high degradability, low toxicity, and overall good biocompatibility. Protein nanoparticles are formed by the arrangement of several native or modified proteins into nanometer-sized assemblies. In this review, we will focus on artificial nanoparticle systems, where proteins are the main structural element and not just an encapsulated payload. While under natural conditions, only certain proteins form defined aggregates and nanoparticles, chemical modifications or a change in the physical environment can further extend the pool of available building blocks. This allows the assembly of many globular proteins and even enzymes. These advances in preparation methods led to the emergence of new generations of nanosystems that extend beyond transport vehicles to diverse applications, from multifunctional drug delivery to imaging, nanocatalysis and protein therapy.
Collapse
Affiliation(s)
- Jonas Kaltbeitzel
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
49
|
Colombo N, Van Gorp T, Matulonis UA, Oaknin A, Grisham RN, Fleming GF, Olawaiye AB, Nguyen DD, Greenstein AE, Custodio JM, Pashova HI, Tudor IC, Lorusso D. Relacorilant + Nab-Paclitaxel in Patients With Recurrent, Platinum-Resistant Ovarian Cancer: A Three-Arm, Randomized, Controlled, Open-Label Phase II Study. J Clin Oncol 2023; 41:4779-4789. [PMID: 37364223 PMCID: PMC10602497 DOI: 10.1200/jco.22.02624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/21/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
PURPOSE Despite therapeutic advances, outcomes for patients with platinum-resistant/refractory ovarian cancer remain poor. Selective glucocorticoid receptor modulation with relacorilant may restore chemosensitivity and enhance chemotherapy efficacy. METHODS This three-arm, randomized, controlled, open-label phase II study (ClinicalTrials.gov identifier: NCT03776812) enrolled women with recurrent, platinum-resistant/refractory, high-grade serous or endometrioid epithelial ovarian, primary peritoneal, or fallopian tube cancer, or ovarian carcinosarcoma treated with ≤4 prior chemotherapeutic regimens. Patients were randomly assigned 1:1:1 to (1) nab-paclitaxel (80 mg/m2) + intermittent relacorilant (150 mg the day before, of, and after nab-paclitaxel); (2) nab-paclitaxel (80 mg/m2) + continuous relacorilant (100 mg once daily); or (3) nab-paclitaxel monotherapy (100 mg/m2). Nab-paclitaxel was administered on days 1, 8, and 15 of each 28-day cycle. The primary end point was progression-free survival (PFS) by investigator assessment; objective response rate (ORR), duration of response (DOR), overall survival (OS), and safety were secondary end points. RESULTS A total of 178 women were randomly assigned. Intermittent relacorilant + nab-paclitaxel improved PFS (hazard ratio [HR], 0.66; log-rank test P = .038; median follow-up, 11.1 months) and DOR (HR, 0.36; P = .006) versus nab-paclitaxel monotherapy, while ORR was similar across arms. At the preplanned OS analysis (median follow-up, 22.5 months), the OS HR was 0.67 (P = .066) for the intermittent arm versus nab-paclitaxel monotherapy. Continuous relacorilant + nab-paclitaxel showed numerically improved median PFS but did not result in significant improvement over nab-paclitaxel monotherapy. Adverse events were comparable across study arms, with neutropenia, anemia, peripheral neuropathy, and fatigue/asthenia being the most common grade ≥3 adverse events. CONCLUSION Intermittent relacorilant + nab-paclitaxel improved PFS, DOR, and OS compared with nab-paclitaxel monotherapy. On the basis of protocol-prespecified Hochberg step-up multiplicity adjustment, the primary end point did not reach statistical significance (P < .025). A phase III evaluation of this regimen is underway (ClinicalTrials.gov identifier: NCT05257408).
Collapse
Affiliation(s)
- Nicoletta Colombo
- Gynecologic Oncology Program, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Medicine and Surgery, University Milan-Bicocca, Milan, Italy
| | - Toon Van Gorp
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospital Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | - Ana Oaknin
- Gynaecologic Cancer Programme, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rachel N. Grisham
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| | | | - Alexander B. Olawaiye
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | | | | | | | - Domenica Lorusso
- Fondazione Policlinico Universitario Gemelli IRCCS, Catholic University of Sacred Heart, Rome, Italy
| |
Collapse
|
50
|
Liu J, Zhang Y, Liu C, Jiang Y, Wang Z, Li X. Paclitaxel prodrug-encapsulated polypeptide micelles with redox/pH dual responsiveness for cancer chemotherapy. Int J Pharm 2023; 645:123398. [PMID: 37690658 DOI: 10.1016/j.ijpharm.2023.123398] [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: 07/11/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/12/2023]
Abstract
Polypeptides are a highly promising carrier for delivering hydrophobic drugs, due to their excellent biocompatibility, non-toxicity, and non-immunogenicity. Herein, a redox and pH dual-responsive poly(ethylene glycol)-SS-b-polypeptide micelles encapsulated with disulfide bridged paclitaxel-pentadecanoic acid prodrug was developed for cancer chemotherapy. First of all, disulfide bridged paclitaxel-pentadecanoic acid prodrug (PTX-SS-COOH) and poly(ethylene glycol)-SS-b-polylysine-b-polyphenylalanine (mPEG-SS-b-PLys-b-PPhe, ESLP) were synthesized and confirmed via NMR, MS, FT-IR or GPC. After that, PTX-SS-COOH (PSH) embedded mPEG-SS-b-PLys-b-PPhe (ESLP/PSH) micelles were prepared by mixing method based on electrostatic interactions and hydrophobic forces. For comparison, mPEG-b-PLys-b-PPhe (ELP) was mixed with PTX-SS-COOH to generate another kind of micelles (ELP/PSH). The characterization of ESLP/PSH micelles through dynamic light scattering (DLS) and transmission electron microscopy (TEM) revealed a spherical structure with a diameter of approximately 170 nm. It is noteworthy that ESLP/PSH micelles displayed a high drug-loading rate of 22.84%, and excellent stability, which can be attributed to the specific interactions between the prodrug and copolymer. Drug release analysis demonstrated that the micelles exhibited a substantial release of PTX in the presence of GSH at pH 5.0, indicating a pH and redox dual responsiveness. In vivo pharmacokinetic study revealed the ESLP/PSH micelles had increased bioavailability and an extended circulation time. Ultimately, antitumor efficacy and systemic toxicity evaluation in 4 T1 tumor-bearing mice confirmed that ESLP/PSH micelles achieved the highest level of tumor growth inhibition (ca. 83%) and the lowest systemic toxicity in comparison with ELP/PSH micelles and commercialized Taxol®. Taken together, the dual responsive micelles represent a promising PTX formulation with potential clinical application in cancer chemotherapy.
Collapse
Affiliation(s)
- Jinyu Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yanhao Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Chao Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuhao Jiang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Zihao Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|