1
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [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: 12/10/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
2
|
Ghosh D, Guin A, Kumar A, Das A, Paul S. Comprehensive insights of etiological drivers of hepatocellular carcinoma: Fostering targeted nano delivery to anti-cancer regimes. Biochim Biophys Acta Rev Cancer 2025; 1880:189318. [PMID: 40222420 DOI: 10.1016/j.bbcan.2025.189318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Hepatocellular carcinoma (HCC) stands as one of the most prevalent and deadliest malignancies on a global scale. Its complex pathogenesis arises from multifactorial etiologies, including viral infections, metabolic syndromes, and environmental carcinogens, all of which drive genetic and molecular aberrations in hepatocytes. This intricate condition is associated with multiple causative factors, resulting in the abnormal activation of various cellular and molecular pathways. Given that HCC frequently manifests within the context of a compromised or cirrhotic liver, coupled with the tendency of late-stage diagnoses, the overall prognosis tends to be unfavorable. Systemic therapy, especially conventional cytotoxic drugs, generally proves ineffective. Despite advancements in therapeutic interventions, conventional treatments such as chemotherapy often exhibit limited efficacy and substantial systemic toxicity. In this context, nanomedicine, particularly lipid-based nanoparticles (LNPs), has emerged as a promising strategy for enhancing drug delivery specificity and reducing adverse effects. This review provides a comprehensive overview of the molecular and metabolic underpinnings of HCC. Furthermore, we explored the role of lipid-based nano-formulations including liposomes, solid lipid nanoparticles, and nanostructured lipid carriers in targeted drug delivery for HCC. We have highlighted recent advances in LNP-based delivery approaches, FDA-approved drugs, and surface modification strategies to improve liver-specific delivery and therapeutic efficacy. It will provide a comprehensive summary of various treatment strategies, recent clinical advances, receptor-targeting strategies and the role of lipid composition in cellular uptake. The review concludes with a critical assessment of existing challenges and future prospects in nanomedicines-driven HCC therapy.
Collapse
Affiliation(s)
- Dipanjan Ghosh
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Aharna Guin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Aryan Kumar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Amlan Das
- Department of Microbiology & Department of Biochemistry, Royal School of Biosciences, The Assam Royal Global University, Guwahati 781035, Assam, India.
| | - Santanu Paul
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India.
| |
Collapse
|
3
|
Sun G, Wu Y, Li J, Yang M, Xu H, Li Y, Tong P, Shao R, Liu Y, Kong X. Quercetin liposomes conjugated with hyaluronidase: An efficient drug delivery system to block pancreatic cancer. J Control Release 2025; 382:113642. [PMID: 40127723 DOI: 10.1016/j.jconrel.2025.113642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/03/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic cancer characterized with intense hydraulic tissue in tumor extracellular matrix (ECM) resists most of chemotherapeutic drugs. Increased levels of hyaluronic acid (HA) represent the primary component of the hydraulic tissue, rendering tumors protective from drug targeting. Quercetin (Que), a natural flavonoid, has the ability to inhibit tumor cell growth in a number of cancers; however, its poor water solubility and low bioavailability largely limit its application in cancer therapy. Hence, we developed an efficient drug delivery system by encapsulation of Que. into liposomes and conjugation with hyaluronidase (HAase) at liposome surface, termed as HQL. In the presence of HAase, HQL were predominantly accumulated at tumor with enhanced permeability and retention effect. Treatment of xenografted tumor mice with HQL gave rise to suppressed tumor growth, while no toxic effects were observed in mice. HQL demonstrated the strong ability to inhibit cell proliferation, promote cell apoptosis, and induce arrest at G2/M cell cycle in pancreatic cancer lines, three-dimensional cultured cell spheroids and pancreatic ductal adenocarcinoma (PDAC)-derived organoids. Mechanistically, HQL downregulated expression of cell cycle-associated protein (CCNB1, CDK1 and PLK1) and cell apoptosis-associated factors PI3K/AKT and Bcl-2. In summary, HQL degraded HA in the tumor microenvironment to enhance nano-particle penetration and inhibited tumor cell growth, eliciting efficacy of anti-tumor therapy. Thereof, HQL may provide a novel efficient drug delivery approach for the adjuvant treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ge Sun
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Ying Wu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Jiekai Li
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Mingjie Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hang Xu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Yiping Li
- Department of Oncology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430065, China
| | - Peilin Tong
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Shao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China; Shanghai Key Laboratory of Biliary Tract Diseases, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of General Surgery, Jiading Branch, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China.
| | - Xianming Kong
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China.
| |
Collapse
|
4
|
Cammarata A, Marino J, Atia MN, Durán H, Glisoni RJ. Novel doxycycline gold nanoparticles via green synthesis using PEO-PPO block copolymers for enhanced radiosensitization of melanoma. Biomater Sci 2025; 13:3223-3241. [PMID: 40261332 DOI: 10.1039/d5bm00253b] [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: 04/24/2025]
Abstract
This study focuses on a green and sustainable nanoplatform for the delivery of therapeutic agents, based on gold nanoparticles (AuNPs) synthesized using PEO-PPO block copolymers (F127, F68, P85, and their F127:P85 combination) as dual-function reducing and stabilizing agents. This eco-friendly approach eliminates the need for toxic chemical reductants, adheres to green chemistry principles, and yields highly stable, biocompatible nanosystems. The resulting polymer-stabilized AuNPs were associated with doxycycline (DOXY), a mitochondrial biogenesis inhibitor with radiosensitizing properties, and characterized using UV-Vis spectroscopy, dynamic light scattering (DLS), transmission electron microscopy (TEM), and X-ray fluorescence (XRF). The nanoparticles exhibited high colloidal stability, with tunable hydrodynamic diameters modulated by the copolymer composition. In vitro studies on A-375 and IIB-MEL-J melanoma cell lines revealed that DOXY-associated AuNPs, combined with gamma radiation (2 Gy, 137Cs), significantly enhanced radiosensitivity, reducing both cell viability and clonogenic survival. The physicochemical features of the nanosystems, particularly particle size and surface composition, influenced cellular uptake and therapeutic response. Notably, AuNPs stabilized with F127:P85 copolymer combination (∼19 nm) outperformed those with F127 (∼30 nm), despite displaying slightly higher polydispersity. Compared to Turkevich AuNPs, our copolymer-coated nanosystems demonstrated superior colloidal stability and cellular internalization. These findings highlight the potential of green-synthesized AuNPs as multifunctional, biocompatible platforms for therapeutic delivery, supporting the development of effective and environmentally responsible multimodal cancer therapies. Moreover, the simplicity, scalability, and cost-effectiveness of the synthesis process support its potential for future translational applications.
Collapse
Affiliation(s)
- Agostina Cammarata
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Biotecnología, Junín 956, C1113AAD Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Nanobiotecnología (NANOBIOTEC), Buenos Aires, Argentina.
| | - Julieta Marino
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Mariel N Atia
- Comisión Nacional de Energía Atómica (CNEA), Gerencia de Investigación y Aplicaciones, Subgerencia de Tecnología y Aplicaciones de Aceleradores, San Martín, Buenos Aires, Argentina
- Instituto de Nanociencia y Nanotecnología (INN-CNEA-CONICET), San Martín, Buenos Aires, Argentina
| | - Hebe Durán
- Comisión Nacional de Energía Atómica (CNEA), Gerencia de Investigación y Aplicaciones, Subgerencia de Tecnología y Aplicaciones de Aceleradores, San Martín, Buenos Aires, Argentina
- Instituto de Nanociencia y Nanotecnología (INN-CNEA-CONICET), San Martín, Buenos Aires, Argentina
- Universidad Nacional de San Martín, Escuela de Ciencia y Tecnología, San Martín, Buenos Aires, Argentina
| | - Romina J Glisoni
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Biotecnología, Junín 956, C1113AAD Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Nanobiotecnología (NANOBIOTEC), Buenos Aires, Argentina.
| |
Collapse
|
5
|
Jain A, Mishra AK, Hurkat P, Shilpi S, Mody N, Jain SK. Navigating liver cancer: Precision targeting for enhanced treatment outcomes. Drug Deliv Transl Res 2025; 15:1935-1961. [PMID: 39847205 DOI: 10.1007/s13346-024-01780-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Cancer treatments such as surgery and chemotherapy have several limitations, including ineffectiveness against large or persistent tumors, high relapse rates, drug toxicity, and non-specificity of therapy. Researchers are exploring advanced strategies for treating this life-threatening disease to address these challenges. One promising approach is targeted drug delivery using prodrugs or surface modification with receptor-specific moieties for active or passive targeting. While various drug delivery systems have shown potential for reaching hepatic cells, nano-carriers offer significant size, distribution, and targetability advantages. Engineered nanocarriers can be customized to achieve effective and safe targeting of tumors by manipulating physical characteristics such as particle size or attaching receptor-specific ligands. This method is particularly advantageous in treating liver cancer by targeting specific hepatocyte receptors and enzymatic pathways for both passive and active therapeutic strategies. It highlights the epidemiology of liver cancer and provides an in-depth analysis of the various targeting approaches, including prodrugs, liposomes, magneto-liposomes, micelles, glycol-dendrimers, magnetic nanoparticles, chylomicron-based emulsion, and quantum dots surface modification with receptor-specific moieties. The insights from this review can be immensely significant for preclinical and clinical researchers working towards developing effective treatments for liver cancer. By utilizing these novel strategies, we can overcome the limitations of conventional therapies and offer better outcomes for liver cancer patients.
Collapse
Affiliation(s)
- Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
- Central Ayurveda Research Institute, Jhansi, Uttar Pradesh, 284003, India
| | - Pooja Hurkat
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | - Satish Shilpi
- School of Pharmaceuticals and Population Health Informatics, FOP, DIT University, Dehradun, Uttarakahnad, India
| | - Nishi Mody
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | | |
Collapse
|
6
|
Chen Z, Sang L, Qixi Z, Li X, Liu Y, Bai Z. Ultrasound-responsive nanoparticles for imaging and therapy of brain tumors. Mater Today Bio 2025; 32:101661. [PMID: 40206140 PMCID: PMC11979416 DOI: 10.1016/j.mtbio.2025.101661] [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: 10/23/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Central nervous system (CNS) cancers, particularly glioblastoma (GBM), are associated with high mortality and disability rates. Despite aggressive surgical resection, radiotherapy, and chemotherapy, patient survival remains poor. The blood-brain barrier (BBB) significantly impedes therapeutic efficacy, making BBB penetration a critical focus of research. Focused ultrasound (FUS) combined with microbubbles (MBs) can transiently open the BBB through mechanisms such as cavitation, modulation of tight junction protein expression, and enhanced vesicular transport in endothelial cells. This review highlights precision delivery and personalized treatment strategies under ultrasound visualization, including precise control of ultrasound parameters and modulation of the immune microenvironment. We discuss the applications of ultrasound-responsive nanoparticles in brain tumor therapy, including enhanced radiotherapy, gene delivery, immunotherapy, and sonodynamic therapy (SDT), with a particular emphasis on piezoelectric catalytic immunotherapy. Finally, we provide insights into the clinical translation potential of ultrasound-responsive nanoparticles for personalized and precision treatment of brain tumors.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Zhai Qixi
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | | | | | | |
Collapse
|
7
|
Al-Baidhani SAS, Pouresmaeil V, Homayouni Tabrizi M. Synthesis of liposomal nanoparticles to load 4-farnesyloxycoumarin and investigating its anti-cancer and anti-metastatic effects. J Liposome Res 2025; 35:125-134. [PMID: 39550704 DOI: 10.1080/08982104.2024.2428168] [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: 06/25/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
The aim of this study was to load 4-farnesyloxycoumarin (4-FLC) in nanoliposomes (4-FLC-LNPs) and evaluate its anti-cancer and anti-metastatic effects. 4-FLC-LNPs were synthesized using a combination of lecithin-cholesterol-polyethylene glycol. The physicochemical properties were evaluated using DLS, FTIR, and microscopy methods. The toxicity against breast cancer (MCF-7), prostate cancer (PS3), pancreatic cancer (PANC), gastric cancer (AGS), and normal cell lines (HUVEC) was evaluated using the MTT assay. Fluorescent staining and flow cytometry were used to assess the occurrence of apoptosis. Molecular analysis methods were used to study the apoptosis and metastasis effects of these nanoliposomes. The antioxidant power of 4-FLC-LNPs was measured using the ABTS and DPPH free radicals methods. 4-FLC-LNPs exhibit a spherical morphology, with an average size of 57.43 nm, a polydispersity index of 0.29, and a zeta potential of -31.4 mV. They demonstrate an encapsulation efficiency of 82.4% for 4-FLC. The IC50 value of 4-FLC-LNPs against the breast cancer cell line was reported as the most sensitive, at approximately 60 μg/mL. ABTS and DPPH results were reported at approximately 30 µg/mL. The inductive effects of nanoliposomes on the apoptosis process were confirmed by an increase in the number of apoptotic cells, as well as the arrest of cells in various phases of cell growth. The increased expression of BAX and decreased expression of Bcl-2, MMP-2, and MMP-9 confirmed the pro-apoptotic and anti-metastatic effects of 4-FLC-LNPs. These finding validate the therapeutic potential of 4-FLC-LNPs, which may be utilized in preclinical studies.
Collapse
Affiliation(s)
| | - Vahid Pouresmaeil
- Department of Biochemistry, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
8
|
Wang X, Li Q, Liu J, Xie C, Zou L, Shi Y, Jiang L, Qin X. Harnessing nano-delivery systems to un-cover the challenges for cervical cancer therapy. Int J Pharm 2025; 677:125657. [PMID: 40306445 DOI: 10.1016/j.ijpharm.2025.125657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Cervical cancer (CC) remains a prevalent malignancy among women, with current therapeutic strategies facing significant challenges in curbing its rising incidence. Nano-delivery systems have emerged as a promising approach to hinder CC progression. This review provides a comprehensive examination of CC pathogenesis and its physiological characteristics while focusing on applying various nano-delivery systems in CC therapy. Specifically, it highlights the potential of both internal (e.g., pH, reactive oxygen species, glutathione) and external (e.g., Photo, magnetism, sound waves, microwaves, electricity) stimuli-responsive nano-delivery platforms to enhance therapeutic efficacy. The challenges of nano-delivery systems in CC therapy, encompassing in vivo stability, biosafety, distribution, and metabolic processes, are addressed, along with potential remedies. Additionally, the review underscores recent preclinical advances in nano-delivery systems for CC therapy. By thoroughly exploring nanomaterial applications, this review provides valuable perspectives for advancing CC treatment and stimulating future research and innovation in this domain.
Collapse
Affiliation(s)
- Xinyu Wang
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Qi Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Jianxin Liu
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Chunbao Xie
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Liang Zou
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yi Shi
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China; Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Xianyan Qin
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
9
|
Filipová M, Tavares MR, Hovorková M, Heine V, Nekvasilová P, Křen V, Etrych T, Chytil P, Bojarová P. Selective Glycopolymer Inhibitors of Galectin-3: Supportive Anti-Cancer Agents Protecting Monocytes and Preserving Interferon-Gamma Function. Int J Nanomedicine 2025; 20:6591-6609. [PMID: 40438186 PMCID: PMC12118576 DOI: 10.2147/ijn.s503381] [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: 11/01/2024] [Accepted: 04/21/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction The immunosuppressive roles of galectin-3 (Gal-3) in carcinogenesis make this lectin an attractive target for pharmacological inhibition in immunotherapy. Although current clinical immunotherapies appear promising in the treatment of solid tumors, their efficacy is significantly weakened by the hostile immunosuppressive tumor microenvironment (TME). Gal-3, a prominent TME modulator, efficiently subverts the elimination of cancer, either directly by inducing apoptosis of immune cells or indirectly by binding essential effector molecules, such as interferon-gamma (IFNγ). Methods N-(2-Hydroxypropyl)methacrylamide (HPMA)-based glycopolymers bearing poly-N-acetyllactosamine-derived tetrasaccharide ligands of Gal-3 were designed, synthesized, and characterized using high-performance liquid chromatography, dynamic light scattering, UV-Vis spectrophotometry, gel permeation chromatography, nuclear magnetic resonance, high-resolution mass spectrometry and CCK-8 assay for evaluation of glycopolymer non-toxicity. Pro-immunogenic effects of purified glycopolymers were tested by apoptotic assay using flow cytometry, competitive ELISA, and in vitro cell-free INFγ-based assay. Results All tested glycopolymers completely inhibited Gal-3-induced apoptosis of monocytes/macrophages, of which the M1 subtype is responsible for eliminating cancer cells during immunotherapy. Moreover, the glycopolymers suppressed Gal-3-induced capture of glycosylated IFNγ by competitive inhibition to Gal-3 carbohydrate recognition domain (CRD), which enables further inherent biological activities of this effector, such as differentiation of monocytes into M1 macrophages and repolarization of M2-macrophages to the M1 state. Conclusion The prepared glycopolymers are promising inhibitors of Gal-3 and may serve as important supportive anti-cancer nanosystems enabling the infiltration of proinflammatory macrophages and the reprogramming of unwanted M2 macrophages into the M1 subtype.
Collapse
Affiliation(s)
- Marcela Filipová
- Department of Biological Models, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marina Rodrigues Tavares
- Department of Biomedical Polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michaela Hovorková
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Viktoria Heine
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pavlína Nekvasilová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Etrych
- Department of Biomedical Polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Chytil
- Department of Biomedical Polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pavla Bojarová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| |
Collapse
|
10
|
Lee YJ, Hong J, Seo BY, Lee BH, Sarangthem V, Park RW. Strategic Optimization of Nanoparticle Characteristics to Enhance Tumor Targeting and Doxorubicin Delivery. Int J Nanomedicine 2025; 20:6357-6378. [PMID: 40416731 PMCID: PMC12103874 DOI: 10.2147/ijn.s513336] [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: 12/19/2024] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Background Doxorubicin (Dox) is a potent anticancer agent; however, its therapeutic efficacy is constrained by a narrow therapeutic index, resulting in nonselective cardiotoxicity and nephrotoxicity. To improve its specificity and therapeutic efficacy, multivalent targeting strategies are being developed. Methods A chimeric polypeptide consisting of an elastin-like polypeptides (ELP) copolymer with a repeating IL-4 receptor-specific targeting peptide, AP-1, and a (GGCGSCGSC)2 sequence encoding 6 cysteine residues (C6) at the carboxyl-terminus for Dox conjugation was designed. Several AP1-ELPs of varying molecular sizes and structures, ranging from unimers to micelle-forming polymers, were characterized to evaluate their influence on Dox delivery and tumor inhibition. Results Conjugating Dox to the C6 via an acid-labile linker induced self-assembly into micelle-like structures at body temperature. The size of these multivalent constructs significantly influenced their tumor penetration and overall therapeutic outcomes. High molecular weight, micelle-forming AP1-ELP constructs demonstrated faster tumor entry and enhanced inhibition compared to lower molecular weight linear AP1-ELPs. Tumor uptake of Dox was five times greater than that of free drug and twice that of low molecular weight, linear AP1-ELPs. Furthermore, systemic administration of these high molecular weight constructs effectively inhibited tumor growth in breast carcinoma xenograft models without inducing specific organ toxicity. Conclusion Outperforming free Dox, high molecular weight micelle-forming AP1-ELP constructs achieve superior tumor targeting and efficacy with minimal toxicity, highlighting their potential as safer and more promising carriers for targeted drug delivery.
Collapse
Affiliation(s)
- Young-Jin Lee
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Jisan Hong
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Bo-Yeon Seo
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Vijaya Sarangthem
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| |
Collapse
|
11
|
Yadav PK, Chauhan D, Yadav P, Tiwari AK, Sultana N, Gupta D, Mishra K, Gayen JR, Wahajuddin M, Chourasia MK. Nanotechnology Assisted Drug Delivery Strategies for Chemotherapy: Recent Advances and Future Prospects. ACS APPLIED BIO MATERIALS 2025; 8:3601-3622. [PMID: 40318022 DOI: 10.1021/acsabm.5c00046] [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] [Indexed: 05/07/2025]
Abstract
In pursuit of the treatment of cancer, nanotechnology engineering has emerged as the simplest and most effective means, with the potential to deliver antitumor chemotherapeutics at the targeted site. Employing nanotechnology for drug delivery provides diverse nanosize particles ranging from one to a thousand nanometers. Reduced size improves drug bioavailability by increasing drug diffusion and decreasing the efflux rate. These nanocarriers offer an enormous scope for modification following the chemical and biological properties of both the drug and its disease. Moreover, these nanoformulations assist in targeting pharmaceutically active drug molecules to the desired site and have gained importance in recent years. Their modern use has revolutionized the antitumor action of many therapeutic agents. Higher drug loading efficiency, thermal stability, easy fabrication, low production cost, and large-scale industrial production draw attention to the application of nanotechnology as a better platform for the delivery of drug molecules. Furthermore, the interaction of nanocarrier technology-assisted agents lowers a drug's toxicity and therapeutic dosage, reduces drug tolerance, and enhances active drug concentration in neoplasm tissue, thus decreasing the concentration in healthy tissue. Nanotechnology-based medications are being widely explored and have depicted effective cancer management in vivo and in vitro systems, leading to many clinical trials with promising results. This review summarizes the innovative impact and application of different nanocarriers developed in recent years in cancer therapy. Subsequently, it also describes the essential findings and methodologies and their effects on cancer treatment. Compared with conventional therapy, nanomedicines can significantly improve the therapeutic effectiveness of antitumor drugs. Thus, the adverse effects associated with healthy tissues are decreased, and adverse effects are scaled back through enhanced permeability and retention effects. Lastly, future insights assisting nanotechnology in active therapeutics delivery and their scope in cancer chemotherapeutics have also been discussed.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Chauhan
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pooja Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nazneen Sultana
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Deepak Gupta
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Keerti Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Jiaur R Gayen
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, United Kingdom
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
12
|
Shi L, Yu Y, Li J, Ma B, Zhang X, Yang P, Chen P, Qu Z, Zhang F, Liu K, Gao S, Cheng H. NIR-responsive Cu 2 - xSe@Fc nanoparticles for photothermal- ferroptosis combination therapy in esophageal cancer. J Nanobiotechnology 2025; 23:356. [PMID: 40380293 PMCID: PMC12084917 DOI: 10.1186/s12951-025-03434-7] [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: 11/14/2024] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Esophageal cancer (EC) represents a highly recurrent and aggressive malignancy within the digestive system. However, conventional therapeutic strategies exhibit notable limitations in their clinical applications. Photothermal therapy (PTT), combined with ferroptosis, has attracted considerable attentions, emerging as a promising novel strategy for EC treatment. Therefore, there is a critical need to develop a drug delivery system capable of effectively integrating these two therapeutic approaches. In this work, we report a novel drug delivery system based on ferrocene (Fc), which is mixed with lauric acid (a phase-change material with a melting point around 44 oC) and then coated on the surface of Cu2 - xSe nanoparticles. The photothermal properties of Cu2 - xSe triggers the melting of lauric acid under near-infrared (NIR) laser irradiation, facilitating controlled release of Fc. Following internalization by tumor cells via endocytosis, the synergistic effect of PTT and ferroptosis, triggered by Cu2 - xSe@Fc, induced immunogenic cell death, which promoted dendritic cell maturation and cytotoxic T lymphocytes recruitment while decreasing the proportion of regulatory T cells, thereby strengthening the antitumor immune surveillance and improving the therapeutic efficacy of Anti-PD-1 blockade. These findings propose that the NIR-responsive Cu2 - xSe@Fc formulation represents a promising and effective strategy with prospecting application for cancer treatment.
Collapse
Affiliation(s)
- Linlin Shi
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Ying Yu
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Jiayi Li
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Beng Ma
- School of Materials Science and Engineering, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiaoman Zhang
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Pingjuan Yang
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Pan Chen
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Zhifeng Qu
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Fengqi Zhang
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Ke Liu
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Shegan Gao
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
- Institute of Organoid on Chip and Drug Translation Research, Henan Academy of Sciences, Zhengzhou, 450003, China.
| | - Haoyan Cheng
- School of Materials Science and Engineering, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471023, China.
| |
Collapse
|
13
|
Wang Y, Zhu N, Liu J, Chen F, Song Y, Ma Y, Yang Z, Wang D. Role of tumor microenvironment in ovarian cancer metastasis and clinical advancements. J Transl Med 2025; 23:539. [PMID: 40369674 PMCID: PMC12079989 DOI: 10.1186/s12967-025-06508-0] [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: 01/13/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy worldwide, characterized by heterogeneity at the molecular, cellular and anatomical levels. Most patients are diagnosed at an advanced stage, characterized by widespread peritoneal metastasis. Despite optimal cytoreductive surgery and platinum-based chemotherapy, peritoneal spread and recurrence of OC are common, resulting in poor prognoses. The overall survival of patients with OC has not substantially improved over the past few decades, highlighting the urgent necessity of new treatment options. Unlike the classical lymphatic and hematogenous metastasis observed in other malignancies, OC primarily metastasizes through widespread peritoneal seeding. Tumor cells (the "seeds") exhibit specific affinities for certain organ microenvironments (the "soil"), and metastatic foci can only form when there is compatibility between the "seeds" and "soil." Recent studies have highlighted the tumor microenvironment (TME) as a critical factor influencing the interactions between the "seeds" and "soil," with ascites and the local peritoneal microenvironment playing pivotal roles in the initiation and progression of OC. Prior to metastasis, the interplay among tumor cells, immunosuppressive cells, and stromal cells leads to the formation of an immunosuppressive pre-metastatic niche in specific sites. This includes characteristic alterations in tumor cells, recruitment and functional anomalies of immune cells, and dysregulation of stromal cell distribution and function. TME-mediated crosstalk between cancer and stromal cells drives tumor progression, therapy resistance, and metastasis. In this review, we summarize the current knowledge on the onset and metastatic progression of OC. We provide a comprehensive discussion of the characteristics and functions of TME related to OC metastasis, as well as its association with peritoneal spread. We also outline ongoing relevant clinical trials, aiming to offer new insights for identifying potential effective biomarkers and therapeutic targets in future clinical practice.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China
| | - Na Zhu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China
| | - Jing Liu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China
| | - Fang Chen
- Department of Gynecology, People's Hospital of Liaoning Province, Shenyang, Liaoning Province, 110016, People's Republic of China
| | - Yang Song
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, People's Republic of China
| | - Yue Ma
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China.
| | - Zhuo Yang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China.
| | - Danbo Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China.
| |
Collapse
|
14
|
Oskouie IM, Amirzargar H, Dezfuli AS, Mashhadi R, Mirzaei A, Shamshirgaran A, Nikoofar P, Aghamir SMK. Reducing the effective dosage of Mitomycin C on a high-grade bladder cancer cell line through combination with selenium nanoparticles: An in vitro study. Med Oncol 2025; 42:207. [PMID: 40348879 DOI: 10.1007/s12032-025-02758-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
This study aimed to assess the effectiveness of combining selenium nanoparticles (SeNPs) with mitomycin C (MMC) in treating the T24 high-grade bladder cancer cell line to decrease MMC dosage and alleviate its side effects. The T24 (EJ138) cell line was exposed to various concentrations of SeNPs and MMC to identify the IC50 values via the MTT assay. The IC50 of MMC was then lowered by 25%, 50%, and 75%, and different SeNPs concentrations were added, to find the new IC50 values of these combinations. Apoptosis rates were measured using Annexin-V/PI staining, while the DNA cell cycle was analyzed using the PI staining method. The scratch-wound assay, colony-forming assay, and Hoechst staining were employed to examine the cell migration, proliferative capacity, and nuclear morphology, respectively. Real-time PCR assessed the expression levels of SNAIL, E-cadherin, and genes related to angiogenesis and proliferation (VEGF-C and HIF-1α), alongside the apoptosis markers (Bcl-2 and BAX). The co-administration of SeNPs and MMC (178.8 µM SeNPs + 14.9 µM MMC) significantly increased the rate of early apoptosis in the T24 cell line compared to MMC alone (29.8 µM, p < 0.0001). Additionally, SeNPs and MMC induced cell cycle arrest at the SubG1/G1 and G2/M phases, respectively. This effect was observed in the combination group at both phases. Similar to MMC alone, the combination group inhibited cell proliferation, colony formation, and migration in T24 cells (p > 0.05). Our findings indicate that the treatment with the combination increased the expression of apoptosis-related genes and decreased angiogenesis and proliferation-related gene expression similar to MMC alone (p > 0.05). The combined administration of MMC and SeNPs enhances the antitumor efficacy on the T24 cell line. It is proposed that the concurrent use of SeNPs and MMC could effectively reduce the required dosage of MMC, thus minimizing its negative side effects.
Collapse
Affiliation(s)
- Iman Menbari Oskouie
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | - Hossein Amirzargar
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | | | - Rahil Mashhadi
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | - Akram Mirzaei
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | - Amirreza Shamshirgaran
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | - Parsa Nikoofar
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
- Section of Tissue Engineering and Stem Cells Therapy, Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Ronash Technology Pars Company (AMINBIC), Tehran University Science and Technology Park, North Campus of Tehran University, Tehran, Iran
| | - Seyed Mohammad Kazem Aghamir
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Urology, Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada.
- Urology Research Center, Sina Hospital, Hassan Abad Sq., Imam Khomeini Ave., Tehran, Iran.
| |
Collapse
|
15
|
Wang X, Stihl A, Höppener C, Vitz J, Schacher FH, Deckert V. Nanoscale Investigation of Elasticity Changes and Augmented Rigidity of Block Copolymer Micelles Induced by Reversible Core-Cross-Linking. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27557-27567. [PMID: 40266783 PMCID: PMC12067378 DOI: 10.1021/acsami.5c04826] [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] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Drug-delivery systems have attracted considerable attention due to their potential to increase the bioavailability of certain drugs and mitigate side effects by enabling targeted drug release. Reversibly core-cross-linked block copolymer micelles providing a hydrophilic and potentially nonimmunogenic shell and a hydrophobic core suitable for the uptake of hydrophobic drugs are frequently considered because of their high stability against environmental changes and dilution. Ultimately, triggering core-de-cross-linking enables the implementation of strategies for targeted drug release, which requests insights into the impact of varying nanomechanical properties on the stability of individual micelles. Here, atomic force microscopy nanoindentation in aqueous media is applied to intact α-allyl-PEG80-b-P(tBGE52-co-FGE12) micelles to quantify changes in their nanomechanical properties induced by dithiobismaleimidoethane (DTME)-mediated Diels-Alder cross-linking of furfuryl moieties and sequential de-cross-linking by reduction of its disulfide bond by tris(2-carboxyethyl)phosphine. As a result of crosslinking by DTME, the apparent Young's modulus of the micelles roughly doubles to 1.18 GPa. Changes to the Young's modulus can be largely reversed by de-cross-linking. Cross-linked and de-cross-linked micelles maintain their structural integrity even in diluted aqueous media below the critical micelle concentration, in contrast to the micelles prior to crosslinking. Understanding the structure-property relationships associated with the observed augmented mechanical stability in native environments is crucial for improving the efficiency of drug encapsulation and introducing refined temporal and spatially controlled drug-release mechanisms.
Collapse
Affiliation(s)
- Xinyue Wang
- Institute
of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller University, D-07743 Jena, Germany
- Leibniz
Institute of Photonic Technology, D-07745 Jena, Germany
| | - Andreas Stihl
- Institute
of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller University, D-07743 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich-Schiller
University, Philosophenweg
7, D-07743 Jena, Germany
| | - Christiane Höppener
- Institute
of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller University, D-07743 Jena, Germany
- Leibniz
Institute of Photonic Technology, D-07745 Jena, Germany
| | - Jürgen Vitz
- Institute
of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller University, D-07743 Jena, Germany
| | - Felix H. Schacher
- Institute
of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller University, D-07743 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich-Schiller
University, Philosophenweg
7, D-07743 Jena, Germany
| | - Volker Deckert
- Institute
of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller University, D-07743 Jena, Germany
- Leibniz
Institute of Photonic Technology, D-07745 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich-Schiller
University, Philosophenweg
7, D-07743 Jena, Germany
| |
Collapse
|
16
|
Kodous AS, Eldin ES, Mohamed HE, Ghobashy MM, El-Maghraby DF. Targeting cell signaling pathway ALKBH5/Beclin1/ULK1 in lung cancer by 5-flurouracil- loaded P (AAm/SA) nanogel in rats. Apoptosis 2025:10.1007/s10495-025-02102-3. [PMID: 40310576 DOI: 10.1007/s10495-025-02102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE Lung cancer is the second most common Cancer in the United States; however, it remains the leading cause of cancer-related death in the United States and worldwide. 5-fluorouracil (5-FU) is among the most administrated chemotherapeutic agents for various neoplasms. This study focused on synthesizing and characterizing P(AAm/SA)/5-Fu nanogels as a potential drug delivery system. METHODS The nanogels were prepared by combining sodium alginate (SA) and acrylamide (AAm) monomers, followed by gamma irradiation-induced polymerization at a dose of 5 kGy. Then, the obtained nanogel was loaded with 500 ppm of 5-Fu. Transmission electron microscopy (TEM) imaging was utilized to characterize the nanogels' morphology and monodispersity with a particle size of (50 nm). Rats were randomly assigned to four groups (six animals per group): Group 1: (Control): normal healthy. Group 2: Cancer-bearing animals (animals injected with diethylnitrosamine (DEN) 20 mg/kg body weight for 3 months. Group 3: Cancer+ 5-fluorouracil (12 mg/kg body weight). Group4: Cancer+ 5-Flurouracil- Loaded P (AAm/SA) Nanogel. RESULTS DEN markedly increased PTGS2, Cox2, PKB, PFKm, and ERK1 levels. Also, observed up-regulation in ALKBH5, Beclin1, ULK1, and P53 gene expressions in the cancer-bearing animal group compared with the control group. 5-fluorouracil nano gel significantly ameliorated the above-mentioned parameters and immunohistochemistry study. 5-fluorouracil nanogel significantly ameliorated the parameters mentioned above, as well as the immunohistochemistry study. CONCLUSION The 5-FU-loaded P(AAm/SA) nanogel could serve as a promising approach for targeting tumor cell proliferation, speeding up autophagic processes, and overcoming chemotherapy resistance in lung carcinoma.
Collapse
Affiliation(s)
- Ahmad S Kodous
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt.
- Department of Molecular Oncology, Cancer Institute (WIA), 38, Sardar Patel Road, P.O. Box 600036, Chennai, Tamilnadu, India.
| | - Eman S Eldin
- Department of Health Radiation Research, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Hebatallah E Mohamed
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Mohamed Mohamady Ghobashy
- Department of of Radiation Research of Polymer Chemistry, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Dina F El-Maghraby
- Department of Health Radiation Research, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| |
Collapse
|
17
|
Chithra P, Bhatia D, Solanki R. Advanced nanomicelles for targeted glioblastoma multiforme therapy. BIOMATERIALS ADVANCES 2025; 170:214221. [PMID: 39922136 DOI: 10.1016/j.bioadv.2025.214221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and malignant primary brain tumor, classified as grade IV by the WHO. Despite standard treatments like surgical resection, radiotherapy and chemotherapy (i.e. temozolomide), GBM's prognosis remains poor due to its heterogeneity, recurrence and the impermeability of the blood-brain barrier (BBB). The exact cause of GBM is unclear with potential factors including genetic predisposition and ionizing radiation. Innovative approaches such as nanomicelles-nanoscale, self-assembled structures made from lipids and amphiphilic polymers show promise for GBM therapy. These nanocarriers enhance drug solubility and stability, enabling targeted delivery of therapeutic agents across the BBB. This review explores the synthesis strategies, characterization and applications of nanomicelles in GBM treatment. Nanomicelles improve the delivery of both hydrophobic and hydrophilic drugs and provide non-invasive delivery options. By offering site-specific targeting, biocompatibility, and stability, nanomicelles can potentially overcome the limitations of current GBM therapies. This review highlights recent advancements in the use of nanomicelles for delivering therapeutic agents and nucleic acids addressing the critical need for advanced treatments to improve GBM patient outcomes.
Collapse
Affiliation(s)
- P Chithra
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
18
|
Roussel T, Cruz-Dubois T, Louis B, Laurini E, Ding L, Balasse L, Nail V, Dignat-George F, Giorgio S, Pricl S, Guillet B, Garrigue P, Peng L. Impact of inner hydrophobicity of dendrimer nanomicelles on biodistribution: a PET imaging study. J Mater Chem B 2025; 13:5041-5050. [PMID: 39699216 DOI: 10.1039/d4tb01266f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Self-assembly is a powerful strategy for building nanosystems for biomedical applications. We have recently developed small amphiphilic dendrimers capable of self-assembling into nanomicelles for tumor imaging. In this context, we studied the impact of increased hydrophobicity of the amphiphilic dendrimer on hydrophilic/hydrophobic balance and consequently on the self-assembly and subsequent biodistribution. Remarkably, despite maintaining the exact same surface chemistry, similar zeta potential, and small size, the altered and enlarged hydrophobic component within the amphiphilic dendrimer led to enhanced stability of the self-assembled nanomicelles, with prolonged circulation time and massive accumulation in the liver. This study reveals that even structural alteration within the interior of nanomicelles can dramatically impact biodistribution profiles. This finding highlights the deeper complexity of rational design for nanomedicine and the need to consider factors other than surface charge and chemistry, as well as size, all of which significantly impact the biodistribution of self-assembling nanosystems.
Collapse
Affiliation(s)
- Tom Roussel
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| | - Twiany Cruz-Dubois
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Beatrice Louis
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory, Department of Engineering and Architectures, University of Trieste, Trieste 34127, Italy
| | - Ling Ding
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| | - Laure Balasse
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Vincent Nail
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | | | - Suzanne Giorgio
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory, Department of Engineering and Architectures, University of Trieste, Trieste 34127, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-136, Poland
| | - Benjamin Guillet
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Philippe Garrigue
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Ling Peng
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| |
Collapse
|
19
|
Chai M, Wang S, Chen Y, Pei X, Zhen X. Targeted and intelligent nano-drug delivery systems for colorectal cancer treatment. Front Bioeng Biotechnol 2025; 13:1582659. [PMID: 40352359 PMCID: PMC12061940 DOI: 10.3389/fbioe.2025.1582659] [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: 02/24/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
Colorectal cancer (CRC) remains a highly heterogeneous malignancy with significant morbidity and mortality worldwide. Despite advancements in surgery, chemotherapy, immunotherapy, and targeted therapy, treatment efficacy is often hampered by drug resistance and systemic toxicity. In recent years, nano-drug delivery systems (NDDS) have emerged as a promising strategy to enhance therapeutic precision, reduce adverse effects, and overcome resistance in CRC treatment. This review discusses the recent advancements in NDDS for CRC treatment, focusing on the optimization of oral drug delivery systems, the development of tumor-specific targeting strategies, and the design of intelligent delivery systems responsive to the tumor microenvironment (TME). Furthermore, we summarize current challenges in NDDS translation and explore future research directions for enhancing their clinical feasibility and therapeutic impact.
Collapse
Affiliation(s)
- Meihong Chai
- Department of Pharmacy, Xi’an Hospital of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Shihua Wang
- School of Medicine, Xi’an Peihua University, Xi’an, Shaanxi, China
| | - Yuxin Chen
- Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xing Pei
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| | - Xueyan Zhen
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Sonkar C, Ranjan R, Mukhopadhyay S. Inorganic nanoparticle-based nanogels and their biomedical applications. Dalton Trans 2025; 54:6346-6360. [PMID: 40019330 DOI: 10.1039/d4dt02986k] [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: 03/01/2025]
Abstract
The advent of nanotechnology has brought tremendous progress in the field of biomedical science and opened avenues for advanced diagnostics and therapeutics applications. Several nanocarriers such as nanoparticles, liposomes, and nanogels have been designed to increase the drug efficiency and targeting ability in patients. Nanoparticles based on gold, silver, and iron are dominantly used for biomedical purposes owing to their biocompatibility properties. Nanoparticles offer an enhanced permeation into tissue vessels; however, their short half-life, toxicity, and off-site accumulations limit their functionality. The above shortcomings could be prevented by employing an integrated system combining nanoparticles with a nanogel-based system. These nanogels are 3D polymeric networks formed by physical and chemical crosslinking and are capable of incorporating nanoparticles, drugs, proteins, and genetic materials. Modification, functionalization, and introduction of inorganic nanoparticles have been shown to enhance the properties of nanogels, such as biocompatibility, stimuli responsiveness, stability, and selectivity. This review paper is focused on the design, synthesis, and biomedical application of inorganic nanoparticle-based nanogels. Current challenges and future perspectives will be briefly discussed to emphasize the versatile role of these multifunctional nanogels for therapeutic and diagnostic purposes.
Collapse
Affiliation(s)
- Chanchal Sonkar
- School of Life Sciences, Devi Ahilya Vishwavidyalaya, Takshila campus, Khandwa road, Indore 452012, India.
| | - Rishi Ranjan
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, Missouri 63103, USA.
| | - Suman Mukhopadhyay
- Department of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453552, India.
| |
Collapse
|
21
|
Mendes M, António M, Daniel-da-Silva AL, Sereno J, Oliveira R, Arnaut LG, Gomes C, Ramos ML, Castelo-Branco M, Sousa J, Pais A, Vitorino C. A switch-on chemo-photothermal nanotherapy impairs glioblastoma. MATERIALS HORIZONS 2025. [PMID: 40202681 DOI: 10.1039/d5mh00351b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Judiciously combined modality approaches have proved highly effective for treating most forms of cancer, including glioblastoma. This study introduces a hybrid nanoparticle-based treatment designed to induce a synergistic effect. It employs repurposed celecoxib-loaded hybrid nanoparticles (HNPs) that are thermally activated by near-infrared laser irradiation to damage glioblastoma cells. The HNPs are constructed by covalently binding organic (ultra-small nanostructured lipid carriers, usNLCs) and inorganic nanoparticles (gold nanorods, AuNRs, with photothermal therapy capability), using c(RGDfK) that serves the dual purpose of a biolinker and a tumor-targeting peptide. The HNPs are further functionalized with transferrin (Tf) as a blood-brain barrier ligand denoted as HNPsTf. Our comprehensive in vitro and in vivo studies have unveiled the remarkable capability of HNPsTf to safely and specifically increase blood-brain barrier permeability through transferrin receptor interactions, facilitating precise nanoparticle accumulation in the tumor region within orthotopic tumor-bearing mice. Furthermore, the orchestrated combination of chemo- and photothermal therapy has exhibited a substantial therapeutic impact on glioblastoma, showcasing a noteworthy 78% inhibition in tumor volume growth and an impressive 98% delay in tumor growth. Notably, this treatment approach has resulted in prolonged survival rates among tumor-bearing mice, accompanied by a favorable side effect profile. Overall, our findings unequivocally demonstrate that celecoxib-loaded HNPsTf offer a game-changing, chemo-photothermal combination, unleashing a synergistic effect that significantly enhances both brain drug delivery and the efficacy of anti-glioblastoma treatments.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Maria António
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana L Daniel-da-Silva
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - José Sereno
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR)/Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Germano de Sousa Pathological Anatomy Center, Quinta de Voimarães, Rua de S. Teotónio, 3000-377 Coimbra, Portugal
| | - Luís G Arnaut
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Célia Gomes
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- Germano de Sousa Pathological Anatomy Center, Quinta de Voimarães, Rua de S. Teotónio, 3000-377 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Luísa Ramos
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| |
Collapse
|
22
|
Jang D, Choi H, Lee J, Chun Y, Heo YH, Lee LP, Ahn DJ, Shin IS, Kim DH, Seo YH, Kim S. Inflamed Tissue-Targeting Polyphenol-Condensed Antioxidant Nanoparticles with Therapeutic Potential. Adv Healthc Mater 2025; 14:e2500495. [PMID: 40033968 DOI: 10.1002/adhm.202500495] [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: 01/28/2025] [Revised: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Inflammation is essential for pathogen eradication and tissue repair; However, chronic inflammation can bring on multi-organ dysfunction due to an overproduction of reactive oxygen species (ROS). Among various anti-inflammatory agents, polyphenol-based nanotherapeutics offer potential advantages, including enhanced stability, targeted delivery, multiple therapeutic functions, and personalized therapy tailored to the severity. Despite these advantages, the development of biocompatible nanomedicines capable of selective accumulation in inflamed tissues and efficient inhibition of ROS-induced inflammatory signaling pathways remains a considerable challenge. In this study, a novel anti-inflammatory nanotherapeutic is engineered through the temperature-dependent condensation of polyphenolic catechin facilitated by hydrothermal reactions. The resulting catechin-condensed nanotherapeutic (CCN150), synthesized at a relatively low temperature, retains physicochemical and functional properties akin to its precursor, catechin, but with a marked enhancement in water solubility. CCN150 protects cells from oxidative stress by eliminating intracellular ROS and augmenting antioxidant enzymes. In vivo studies reveal that intravenously administered CCN150 predominantly accumulates in inflamed tissues, with minimal distribution to healthy regions. Furthermore, CCN150 effectively reduces systemic inflammation in mouse models by disrupting the cycles of ROS instigated by a pro-inflammatory oxidative milieu. Exhibiting negligible toxicity, CCN150 holds substantial promise for extensive therapeutic applications in the treatment of various ROS-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Dohyub Jang
- Department of Biomicrosystem Technology, Korea University, Seoul, 02792, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Honghwan Choi
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Juhyang Lee
- Biosensor Group, Korea Institute of Science and Technology Europe, Campus E7.1, 66123, Saarbrücken, Germany
| | - Yousun Chun
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoon-Ho Heo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Luke P Lee
- Harvard Institute of Medicine, Harvard Medical School Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| | - Dong June Ahn
- Department of Biomicrosystem Technology, Korea University, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ik-Soo Shin
- Department of Chemistry, College of Natural Science, Soongsil University, 3, Seoul, 15674, Republic of Korea
| | - Dong Ha Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
- College of Medicine, Ewha Womans University, 25, Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
- Basic Sciences Research Institute (Priority Research Institute), Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
- Nanobio·Energy Materials Center (National Research Facilities and Equipment Center), Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Young Hun Seo
- Biosensor Group, Korea Institute of Science and Technology Europe, Campus E7.1, 66123, Saarbrücken, Germany
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
23
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
24
|
Bhadouria N, Yadav S, Bukke SPN, Narapureddy BR. Advancements in vaccine delivery: harnessing 3D printing for microneedle patch technology. Ann Med Surg (Lond) 2025; 87:2059-2067. [PMID: 40212146 PMCID: PMC11981410 DOI: 10.1097/ms9.0000000000003060] [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: 09/17/2024] [Accepted: 01/31/2025] [Indexed: 04/13/2025] Open
Abstract
The development of 3D-printed microneedle (MN) technology is a significant step in vaccine delivery, providing a painless, effective, and adaptable substitute for conventional injection-based techniques. Direct transdermal vaccination distribution without the need for needles is made possible by microneedle patches, which employ a variety of tiny needles that dissolve when they penetrate the skin. By using 3D printing to precisely customise microneedles' size, shape, and density to meet particular vaccine requirements, administration control can be improved and vaccine efficiency may even be increased. Furthermore, rapid prototyping made possible by 3D printing speeds up the development process, enabling quicker testing and improvement of vaccines. Additionally, this scalable technology can greatly increase vaccine accessibility, particularly in environments with limited resources. Research indicates that by directly interacting with the skin's immune-rich layers, microneedle patches enhance antigen delivery and elicit a strong immune response. Because MN technology offers a useful, self-administrable vaccination approach with little waste, it has significant potential for use in public health applications, notably during pandemics. This study emphasises how 3D-printed microneedle patches have the potential to revolutionise vaccination procedures and increase vaccine accessibility globally.
Collapse
Affiliation(s)
- Namrata Bhadouria
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Sarad Pawar Naik Bukke
- Department of Pharmaceutics and Pharmaceutical Technology, Kampala International University, Western Campus, Ishaka-Bushenyi, Uganda
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
25
|
Wei S, Cui X, Li T, Ma X, Liu L. Pillar[n]arene-Based Supramolecular Nanodrug Delivery Systems for Cancer Therapy. ChemMedChem 2025; 20:e202400822. [PMID: 39833508 DOI: 10.1002/cmdc.202400822] [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: 10/18/2024] [Revised: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Macrocyclic supramolecular materials play an important role in encapsulating anticancer drugs to improve the anticancer efficiency and reduce the toxicity to normal tissues through host-guest interactions. Among them, pillar[n]arenes, as an emerging class of supramolecular macrocyclic compounds, have attracted increasing attention in drug delivery and drug-controlled release due to their high biocompatibility, excellent host-guest chemistry, and simplicity of modification. In this review, we summarize the research progress of pillar[n]arene-based supramolecular nanodrug delivery systems (SNDs) in recent years in the field of tumor therapy, including drug-controlled release, imaging diagnostics and therapeutic modalities. Furthermore, the opportunities and major limitations of pillar[n]arene-based SNDs for tumor therapy are discussed.
Collapse
Affiliation(s)
- Shubin Wei
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Xinyi Cui
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Tingting Li
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Xin Ma
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Luzhi Liu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
- Guangxi Key Laboratory of Green Chemical Materials and Safety Technology, Guangxi Engineering Research Center for New Chemical Materials and Safety Technology, College of Petroleum and Chemical Engineering, Beibu Gulf University, Qinzhou, Guangxi, 535011, PR China
| |
Collapse
|
26
|
Aldayel AM, Bosaeed M, Almansour S, Alharbi NK, Alenazi M, Aljami HA, Aldibasi O, Aljouie A, Xu H, Cui Z. IgM has a better relative distribution in inflammation sites and tumor tissues than IgG. J Nanobiotechnology 2025; 23:253. [PMID: 40156022 PMCID: PMC11954317 DOI: 10.1186/s12951-025-03213-4] [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: 05/05/2024] [Accepted: 02/10/2025] [Indexed: 04/01/2025] Open
Abstract
Immunoglobulins (Igs) play a crucial role in host's defense and in developing therapies against inflammatory diseases and cancer. Herein, we first studied the relative distribution of IgM and IgG in mouse models with acute or chronic inflammation. We found that IgM showed a more selective distribution towards inflammation sites than IgG. Similarly, in a tumor-bearing mouse model, IgM showed a higher tumor-to-blood or -to healthy organs ratio than IgG. We hypothesized that the difference in the sizes between IgM and IgG may have contributed to the differences in their relative distribution, which was supported by using an IgG nanoparticle system that was similar to IgM in size. To confirm the findings in clinics, we investigated IgM and IgG levels in the blood and bronchoalveolar lavage fluid (BALF) of patients diagnosed with fungal pneumonia and showed that the relative distribution of IgM was significantly higher than IgG in the BALF samples as compared to that in serum. Such an understanding of our immune system at the nano-level may help us develop more effective biotechnological interventions against inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Abdulaziz M Aldayel
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, 78712, USA.
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia.
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia.
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia.
| | - Mohammad Bosaeed
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Sarah Almansour
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Naif Khalaf Alharbi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia
| | - Mohammed Alenazi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Haya A Aljami
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Omar Aldibasi
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Abdulrhman Aljouie
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Haiyue Xu
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Zhengrong Cui
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
27
|
Ow V, Lin Q, Wong JHM, Sim B, Tan YL, Leow Y, Goh R, Loh XJ. Understanding the interplay between pH and charges for theranostic nanomaterials. NANOSCALE 2025; 17:6960-6980. [PMID: 40008569 DOI: 10.1039/d4nr03706e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Nanotechnology has emerged as a highly promising platform for theranostics, offering dual capabilities in targeted imaging and therapy. Interactions between the nanomaterial and biological components determine the in vivo fate of these materials which makes the control of their surface properties of utmost importance. Nanoparticles with neutral or negative surface charge have a longer circulation time while positively charged nanoparticles have higher affinity to cells and better cellular uptake. This trade-off presents a key challenge in optimizing surface charge for theranostic applications. A sophisticated solution is an on-demand switch of surface charge, enabled by leveraging the distinct pH conditions at the target site. In this review, we explore the intricate relationship between pH and charge modulation, summarizing recent advances in pH-induced charge-switchable nanomaterials for theranostics over the past five years. Additionally, we discuss how these innovations enhance targeted drug delivery and imaging contrast and provide perspectives on future directions for this transformative field.
Collapse
Affiliation(s)
- Valerie Ow
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
- Department of Biomedical Engineering, National University of Singapore (NUS), Singapore
| | - Qianyu Lin
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Joey Hui Min Wong
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Belynn Sim
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore
| | - Yee Lin Tan
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Yihao Leow
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore
| | - Rubayn Goh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
28
|
Zhou X, Jiang K, Han Y, Yang S. A preclinical study of a novel dual-modality contrast agent in rodent models. Front Bioeng Biotechnol 2025; 13:1557772. [PMID: 40182986 PMCID: PMC11966174 DOI: 10.3389/fbioe.2025.1557772] [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: 01/09/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Glioblastoma (GBM) represents the most aggressive and prevalent form of primary malignant brain tumor in adults, with surgical intervention being the primary treatment modality. To enhance surgical outcomes and extend patient survival, we have engineered a dual-modality MRI/FI contrast agent known as PL002 to aid in the surgical management of GBM. Methods In this study, an orthotopic glioma model was established in mice via intracranial injection of U-87 MG cells. Subsequently, the model animals were intravenously injected with PL002 and placed in a 7.0T magnetic resonance imaging (MRI) device to evaluate the imaging effects. After the MRI scan, fluorescence imaging techniques were employed to observe the distribution of PL002 at both the brain tissue and cellular levels. Moreover, healthy rat models were utilized to investigate the pharmacokinetic characteristics, tissue distribution, and safety profile of PL002. Results The molecular structure of PL002 contains both gadolinium (Gd3+) and indocyanine green (ICG), demonstrating optimal imaging effects within the dosage range of 10-50 mg/kg, with a half-life of 2.51 to 4.87 hours. Even at relatively low concentrations in the brain, PL002 can provide stable and sustained support for MRI and fluorescence imaging for up to 72 hours. No abnormalities were observed in rats at a dosage of 100 mg/kg. Discussion Compared to Gadavist® and ICG, PL002 provided sustained support for MRI and FI of GBM for 72 h, with a broad therapeutic window. This dual-modality contrast agent holds significant potential and promise for applications in preoperative assessment of resection margins, real-time intraoperative guidance, and postoperative verification of the extent of resection.
Collapse
Affiliation(s)
- Xuelai Zhou
- Zhejiang Poly Pharm. Co., Ltd., Hangzhou, China
| | | | - Yuxin Han
- Zhejiang Poly Pharm. Co., Ltd., Hangzhou, China
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Zhao S, Xuan C, Diao W, Bai R, Wu F, Yu W, Yang F, Wu J, Xu W, Jiang G, Gao Z, Li H. Study on the Construction and Anti-Tumor Effect of aPDL1/aMUC1 Double Antibody Modification of Doxorubicin Liposome. ACS OMEGA 2025; 10:10107-10121. [PMID: 40124041 PMCID: PMC11923672 DOI: 10.1021/acsomega.4c08564] [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: 09/18/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
In recent years, the primary treatments for cancer have included chemotherapy, radiotherapy, and surgery. However, challenges such as poor prognosis, high recurrence rates, low survival rates, and diminished quality of life persist in cancer management. Recently, immunotherapy has emerged as a potent therapeutic approach for treating tumors. To this end, we developed antibodies for mucin 1 (MUC1) and programmed cell death ligand 1 (PD-L1) to functionalize liposomes and incorporate doxorubicin (DOX) (DOX-aMUC1/aPDL1-Lip). This formulation is designed to enhance its targeting capability and antitumor activity against cancer cells. The DOX-aMUC1/aPDL1-Lip formulation demonstrated significant antitumor effects both in vivo and in vitro, effectively inhibiting tumor cell growth. Utilizing antibodies against PD-L1 and MUC1 to modify liposomes represents a novel strategy for cancer treatment.
Collapse
Affiliation(s)
- Shouzhen Zhao
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Cuiling Xuan
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Wenbin Diao
- Clinical
Laboratory, Second People’s Hospital
of Weifang, Weifang 261053, Shandong, China
| | - Ran Bai
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Fei Wu
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Wenjing Yu
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Fan Yang
- Shandong
Kanghua Biotechnology Co., Ltd., Weifang 261053, Shandong, China
| | - Jingliang Wu
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Wei Xu
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Guosheng Jiang
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
- Institute
of Immunology and Biotechnology Transformation, Binzhou Medical University, Yantai 264000, Shandong, China
| | - Zhiqin Gao
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| | - Haimei Li
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, Shandong, China
| |
Collapse
|
30
|
Kubbara EA, Bolad A, Malibary H. Advances in Liposomal Interleukin and Liposomal Interleukin Gene Therapy for Cancer: A Comprehensive Review of Preclinical Studies. Pharmaceutics 2025; 17:383. [PMID: 40143046 PMCID: PMC11945541 DOI: 10.3390/pharmaceutics17030383] [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: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Preclinical studies on liposomal interleukin (IL) therapy demonstrate considerable promise in cancer treatment. This review explores the achievements, challenges, and future potential of liposomal IL encapsulation, focusing on preclinical studies. METHODS A structured search was conducted using the PubMed and Web of Science databases with the following search terms and Boolean operators: ("liposomal interleukin" OR "liposome-encapsulated interleukin") AND ("gene therapy" OR "gene delivery") AND ("cancer" OR "tumor" OR "oncology") AND ("pre-clinical studies" OR "animal models" OR "in vitro studies". RESULTS Liposomal IL-2 formulations are notable for enhancing delivery and retention at tumor sites. Recombinant human interleukin (rhIL-2) adsorbed onto small liposomes (35-50 nm) substantially reduces metastases in murine models. Hepatic metastasis models demonstrate superior efficacy of liposomal IL-2 over free IL-2 by enhancing immune responses, particularly in the liver. Localized delivery strategies, including nebulized liposomal IL-2 in canine pulmonary metastases and intrathoracic administration in murine sarcoma models, reduce systemic toxicity while promoting immune activation and tumor regression. Liposomal IL gene therapy, delivering cytokine genes directly to tumor sites, represents a notable advancement. Combining IL-2 gene therapy with other cytokines, including IL-6 or double-stranded RNA adjuvants, synergistically enhances macrophage and T-cell activation. Liposomal IL-4, IL-6, and IL-21 therapies show potential across various tumor types. Pairing liposomal IL-2 with chemotherapy or immune agents improves remission and survival. Innovative strategies, including PEGylation and ligand-targeted systems, optimize delivery, release, and therapeutic outcomes. CONCLUSIONS Utilizing immune-stimulatory ILs through advanced liposomal delivery and gene therapy establishes a strong foundation for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Eman A. Kubbara
- Clinical Biochemistry Department, Faculty of Medicine, Rabigh Branch, King Abdulaziz University, Rabigh 21911, Saudi Arabia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Ahmed Bolad
- Department of Microbiology and Unit of Immunology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Husam Malibary
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| |
Collapse
|
31
|
Beretta GL, Cassinelli G, Rossi G, Azzariti A, Corbeau I, Tosi D, Perego P. Novel insights into taxane pharmacology: An update on drug resistance mechanisms, immunomodulation and drug delivery strategies. Drug Resist Updat 2025; 81:101223. [PMID: 40086175 DOI: 10.1016/j.drup.2025.101223] [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: 01/29/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
Taxanes are effective in several solid tumors. Paclitaxel, the main clinically available taxane, was approved in the early nineties, for the treatment of ovarian cancer and later on, together with the analogs docetaxel and cabazitaxel, for other malignancies. By interfering with microtubule function and impairing the separation of sister cells at mitosis, taxanes act as antimitotic agents, thereby counteracting the high proliferation rate of cancer cells. The action of taxanes goes beyond their antimitotic function because their main cellular targets, the microtubules, participate in multiple processes such as intracellular transport and cell shape maintenance. The clinical efficacy of taxanes is limited by the development of multiple resistance mechanisms. Among these, extracellular vesicles have emerged as new players. In addition, taxane metronomic schedules shows an impact on the tumor microenvironment reflected by antiangiogenic and immunomodulatory effects, an aspect of growing interest considering their inclusion in treatment regimens with immunotherapeutics. Preclinical studies have paved the bases for synergistic combinations of taxanes both with conventional and targeted agents. A variety of drug delivery strategies have provided novel opportunities to increase the drug activity. The ability of taxanes to orchestrate different cellular effects amenable to modulation suggests novel options to improve cures in lethal malignancies.
Collapse
Affiliation(s)
- Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, Milan 20133, Italy.
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, Milan 20133, Italy.
| | - Giacomina Rossi
- Unit of Neurology 8, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.le O. Flacco, 65, Bari 70124, Italy.
| | - Iléana Corbeau
- Early Clinical Trial Unit, Medical Oncology Department, Institut régional du Cancer de Montpellier, Inserm U1194, Montpellier University, 208, rue de Apothicaires, 34298 Montpellier, France; Fondazione Gianni Bonadonna, via Bertani, 14, Milan 20154, Italy.
| | - Diego Tosi
- Early Clinical Trial Unit, Medical Oncology Department, Institut régional du Cancer de Montpellier, Inserm U1194, Montpellier University, 208, rue de Apothicaires, 34298 Montpellier, France; Fondazione Gianni Bonadonna, via Bertani, 14, Milan 20154, Italy.
| | - Paola Perego
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, Milan 20133, Italy.
| |
Collapse
|
32
|
Wei X, Cao W, Wang S, Zhang Y, Gao Z, Wang S, Yao L, Zhang Z, Li X, Deng W, Xie Y, Li M. Progress in the Application of Novel Nanomaterials in Targeted Therapy for Liver Cancer. Int J Nanomedicine 2025; 20:2623-2643. [PMID: 40061885 PMCID: PMC11887507 DOI: 10.2147/ijn.s509409] [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: 12/16/2024] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
In recent years, nanobiotechnology, widely used in hepatoma, holds great promise for improving targeted hepatocarcinoma therapy. On account of the unique properties of low toxicity, good tolerance, biocompatibility, and biodegradability of new nanomaterials, a targeted drug delivery system (TDDS) has been constructed, which can boost the therapeutic effect of hepatoma-targeted drugs, reduce drug toxicity, and minimize off target reactions by enhancing permeability retention effect (EPR) and active targeting, thus improving existing liver cancer targeted therapy strategies. Different nanoparticles have their own advantages and disadvantages. They can be loaded with multiple drugs on the same nanoparticle and can also be surface modified with each other to achieve synergistic anti-tumor effects. This essay provides a comprehensive overview of the current status of targeted therapy for hepatocarcinoma, nanoparticles' structure, advantages and disadvantages of each nanoparticle, and the application progress of nanoparticles in targeted therapy for liver cancer. We hope to provide a basis for the future clinical targeted therapy of hepatoma using nanotechnology.
Collapse
Affiliation(s)
- Xin Wei
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Weihua Cao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Shiyu Wang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Yaqin Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Zixuan Gao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Shuojie Wang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Linmei Yao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Ziyu Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Xinxin Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Wen Deng
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Yao Xie
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
- Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, Beijing, 100015, People’s Republic of China
| | - Minghui Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
- Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, Beijing, 100015, People’s Republic of China
| |
Collapse
|
33
|
Maeda S, Sato T, Nishiofuku H, Toyoda S, Taiji R, Matsumoto T, Chanoki Y, Tachiiri T, Kunichika H, Sho M, Tanaka T. Intra-arterial Therapy Using Micellar Nanoparticles Incorporating SN-38 in a Rat Pancreatic Tumor Model. Cardiovasc Intervent Radiol 2025; 48:372-378. [PMID: 39984668 DOI: 10.1007/s00270-024-03939-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 12/05/2024] [Indexed: 02/23/2025]
Abstract
PURPOSE To evaluate advantages of micellar nanoparticles encapsulating SN-38, a biologically active metabolite of irinotecan, in intraarterial therapy for pancreatic cancer. MATERIALS AND METHODS Rat pancreatic cancer cells (DSL-6A/C1) were implanted in Lewis rats under laparotomy. This study consists of two parts. Firstly, after confirming tumor formation by ultrasonography, celiac arteriography was performed, and tumor blood supply was visually evaluated by dye injection and CT during arteriography. Secondly, 18 rats were divided into two groups; the Micellar Nanoparticles group and the Irinotecan Infusion group. Micellar nanoparticles or irinotecan was injected via the celiac artery, and SN-38 and irinotecan concentrations in the tumor, duodenum and pancreatic parenchyma, were measured at 5 min, 6 h and 24 h. RESULTS The maximum concentration (Cmax) of SN-38 were shown at 6 h in the Micellar Nanoparticles group, while Cmax of irinotecan was shown at 5 min in the Irinotecan Infusion group. Tumor concentration in the Micellar Nanoparticles group maintained elevated for 24 h without significant decrease (P = 0.068). Conversely, a significant decrease was observed in the regular pancreas parenchyma (P = 0.006) and duodenum (P = 0.028). In the Irinotecan Infusion group, tumor irinotecan concentration significantly decreased at 24 h (P = 0.016). CONCLUSION Micellar nanoparticles may improve arterial infusion chemotherapy for pancreatic cancer. These nanoparticles have the potential to reduce SN-38 accumulation in duodenum, while increasing it in the tumor. Further research is warranted to validate and expand upon these findings.
Collapse
Affiliation(s)
- Shinsaku Maeda
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan.
| | - Takeshi Sato
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Hideyuki Nishiofuku
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Shohei Toyoda
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Ryosuke Taiji
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Takeshi Matsumoto
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Yuto Chanoki
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Tetsuya Tachiiri
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Hideki Kunichika
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| | - Toshihiro Tanaka
- Department of Diagnostic and Interventional Radiology, Nara Medical University, Shijo-Cho 840, Kashihara, 634-8522, Japan
| |
Collapse
|
34
|
Moradi Kashkooli F, Mirala F, H H Tehrani M, Alirahimi M, Souri M, Golzaryan A, Kar S, Soltani M. Mechanical Forces in Tumor Growth and Treatment: Perspectives From Biology, Physics, Engineering, and Mathematical Modeling. WIREs Mech Dis 2025; 17:e70000. [PMID: 40170456 DOI: 10.1002/wsbm.70000] [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: 07/21/2023] [Revised: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025]
Abstract
The progression of tumors is influenced by mechanical forces and biological elements, such as hypoxia and angiogenesis. Mechanical factors, including stress, pressure, interstitial fluid pressure, and cellular traction forces, compromise normal tissue architecture, augmenting stiffness and thus promoting tumor growth and invasion. The selective elimination of specific tumor components can reduce growth-induced mechanical stress, thereby improving therapeutic efficacy. Furthermore, stress-relief drugs have the potential in enhancing chemotherapy outcomes. In this setting, computational modeling functions as an essential tool for quantitatively elucidating the mechanical principles underlying tumor formation. These models can precisely replicate the impact of mechanical pressures on solid tumors, offering insight into the regulation of tumor behavior by these forces. Tumor growth produces mechanical forces, including compression, displacement, and deformation, leading to irregular stress patterns, expedited tumor advancement, and reduced treatment efficacy. This review analyzes the impact of mechanical forces on carcinogenesis and solid tumor proliferation, emphasizing the significance of stress alleviation in regulating tumor growth. Furthermore, we investigate the influence of mechanical forces on tumor dissemination and emphasize the promise of integrating computational modeling with force-targeted cancer therapies to improve treatment efficacy by tackling the fundamental mechanics of tumor proliferation.
Collapse
Affiliation(s)
| | - Fatemeh Mirala
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Masoud H H Tehrani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mahvash Alirahimi
- Department of Obstetrics & Gynecology, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Aryan Golzaryan
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Egaila, Kuwait
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
35
|
Roszkowski S, Durczyńska Z, Szablewska S. Targeted nanodelivery systems for personalized cancer therapy. Rep Pract Oncol Radiother 2025; 29:776-788. [PMID: 40104662 PMCID: PMC11912883 DOI: 10.5603/rpor.103524] [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: 11/27/2023] [Accepted: 11/12/2024] [Indexed: 03/20/2025] Open
Abstract
Conventional cancer therapies such as chemotherapy face challenges such as poor tumor targeting, systemic toxicity, and drug resistance. Nanotechnology offers solutions through advanced drug delivery systems that preferentially accumulate in tumors while avoiding healthy tissues. Recent innovations have enabled the optimization of engineered nanocarriers for extended circulation and tumor localization via both passive and active targeting mechanisms. Passive accumulation exploits the leaky vasculature of tumors, whereas active strategies use ligands to selectively bind cancer cell receptors. Multifunctional nanoparticles also allow the combination of imaging, multiple therapeutic modalities and on-demand drug release within a single platform. Overall, precisely tailored nanotherapeutics that leverage unique pathophysiological traits of malignancies provide opportunities to overcome the limitations of traditional treatment regimens. This emerging field promises more effective and personalized nanomedicine approaches to detect and treat cancer. The key aspects highlighted in this review include the biological barriers associated with nanoparticles, rational design principles to optimize nanocarrier pharmacokinetics and tumor uptake, passive and active targeting strategies, multifunctionality, and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Zofia Durczyńska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Sylwia Szablewska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| |
Collapse
|
36
|
Graham W, Torbett-Dougherty M, Islam A, Soleimani S, Bruce-Tagoe TA, Johnson JA. Magnetic Nanoparticles and Drug Delivery Systems for Anti-Cancer Applications: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:285. [PMID: 39997849 PMCID: PMC11858650 DOI: 10.3390/nano15040285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Cancer continues to be a prominent fatal health issue worldwide, driving the urgent need for more effective treatment strategies. The pressing demand has sparked significant interest in the development of advanced drug delivery systems for chemotherapeutics. The advent of nanotechnology offers a groundbreaking approach, presenting a promising pathway to revolutionize cancer treatment and improve patient outcomes. Nanomedicine-based drug delivery systems have demonstrated the capability of improving the pharmacokinetic properties and accumulation of chemotherapeutic agents in cancer sites while minimizing the adverse side effects. Despite these advantages, most NDDSs exhibit only limited improvement in cancer treatment during clinical trials. The recent development of magnetic nanoparticles (MNPs) for biomedical applications has revealed a potential opportunity to further enhance the performance of NDDSs. The magnetic properties of MNPs can be utilized to increase the targeting capabilities of NDDSs, improve the controlled release of chemotherapeutic agents, and weaken the chemoresistance of tumors with magnetic hyperthermia. In this review, we will explore recent advancements in research for NDDSs for oncology applications, how MNPs and their properties can augment the capabilities of NDDSs when complexed with them and emphasize the challenges and safety concerns of incorporating these systems into cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jacqueline Ann Johnson
- Department of Mechanical, Aerospace, and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (W.G.); (M.T.-D.); (A.I.); (S.S.); (T.A.B.-T.)
| |
Collapse
|
37
|
Einen C, Snipstad S, Wesche HF, Nordlund V, Devold EJ, Amini N, Hansen R, Sulheim E, Davies CDL. Impact of the tumor microenvironment on delivery of nanomedicine in tumors treated with ultrasound and microbubbles. J Control Release 2025; 378:656-670. [PMID: 39701458 DOI: 10.1016/j.jconrel.2024.12.037] [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: 09/04/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
The delivery of nanoparticles to tumors has been shown preclinically to be improved by microbubble-mediated ultrasound. However, the mechanisms and biological effects are not fully understood. In this study, we explored the influence of the tumor microenvironment on nanoparticle uptake and microdistribution both with and without ultrasound and microbubble treatment. Three murine tumor models, KPC (pancreatic ductal adenocarcinoma), 4T1 (triple negative mammary carcinoma) and CT26 (colon carcinoma), were characterized with respect to extracellular matrix composition, tumor stiffness and perfusion. KPC and 4T1 tumors presented higher levels of collagen and hyaluronic acid and were stiffer compared to CT26, whereas all three tumors had similar levels of sulfated glycosaminoglycans. Furthermore, the 4T1 tumors appeared poorly vascularized with a lower cell density compared to KPC and CT26. All three tumors presented similar nanoparticle uptake, but extravasated nanoparticles traveled significantly shorter in KPC tumors compared to 4T1 and CT26. The effect of ultrasound and microbubble treatment on the tumor uptake and penetration of polymer nanoparticles into the extracellular matrix were evaluated using a treatment protocol previously shown to increase nanoparticle delivery to tumors. Interestingly, we found a significant increase in nanoparticle uptake in the soft CT26 tumor, but no effect of the ultrasound treatment in the stiff KPC and 4T1 tumors, suggesting that tumor stiffness is an important parameter for treatment with ultrasound and microbubbles. Ultrasound treatment resulted in a modest but not statistically significant improvement in nanoparticle penetration through the extracellular matrix. In tumors demonstrating increased uptake of nanoparticles following ultrasound treatment, the uptake correlated positively with blood volume. These findings emphasize the importance of taking the tumor microenvironment into consideration when optimizing ultrasound parameters for delivery of nanomedicine.
Collapse
Affiliation(s)
- Caroline Einen
- Porelab and Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Håkon F Wesche
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Veronica Nordlund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ella J Devold
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | | |
Collapse
|
38
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [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: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
39
|
Ke G, Zhang M, Hu P, Zhang J, Naeem A, Wang L, Xu H, Liu Y, Cao M, Zheng Q. Exploratory Study on Nanoparticle Co-Delivery of Temozolomide and Ligustilide for Enhanced Brain Tumor Therapy. Pharmaceutics 2025; 17:191. [PMID: 40006558 PMCID: PMC11858958 DOI: 10.3390/pharmaceutics17020191] [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: 11/20/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Temozolomide (TMZ) is the first-line therapy for glioblastoma (GBM), but its clinical efficacy is limited by its short half-life, poor brain targeting, adverse side effects, and the development of drug resistance. Ligustilide (LIG) has been shown to enhance blood-brain barrier permeability and reduce P-glycoprotein activity, thereby potentiating the synergistic effect of TMZ against GBM. Methods: The dual-drug-loaded nanoparticles encapsulating both TMZ and LIG (TMZ/LIG-NPs) were prepared using Poly (d,l-lactic-co-glycolide)-monomethoxy poly (ethylene glycol) (PLGA-mPEG). The physicochemical properties of the NPs, including particle size and zeta potential, were characterized. Cellular uptake of NPs was evaluated using flow cytometry and fluorescence staining. The pharmacokinetic profile and cytotoxicity of TMZ/LIG-NPs were compared to those of free TMZ and a mixture of TMZ and LIG in rat and glioma cells, respectively. Results: The mean particle size of TMZ/LIG-NPs was 117.6 ± 0.7 nm, with a zeta potential of -26.5 ± 0.4 mV. Cellular uptake of NPs was significantly higher than that of free drug in U251 cells. Encapsulation of TMZ in NPs significantly increased its half-life by 1.62-fold compared to free TMZ and significantly improved its pharmacokinetic profile. Moreover, the storage stability of the TMZ/LIG-NPs solution was extended to one month. The toxicity of TMZ/LIG-NPs to glioma cells C6 and U251 was markedly enhanced compared to the mixture of TMZ and LIG. Conclusions: The development of TMZ/LIG-NPs using PLGA-mPEG effectively enhanced the stability and efficacy of both TMZ and LIG. This dual drug-loaded nanoparticle system represents a promising strategy for glioblastoma therapy.
Collapse
Affiliation(s)
- Gang Ke
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
- Luzhou People’s Hospital, Luzhou 646000, China
| | - Mingxia Zhang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Jing Zhang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Abid Naeem
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China;
| | - Lianfang Wang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Huixin Xu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Yu Liu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Ming Cao
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (G.K.); (M.Z.); (P.H.); (J.Z.); (L.W.); (H.X.); (Y.L.); (M.C.)
| |
Collapse
|
40
|
Yu Y, Tao Y, Ma J, Li J, Song Z. Targeting the tumor microenvironment with mesenchymal stem cells based delivery approach for efficient delivery of anticancer agents: An updated review. Biochem Pharmacol 2025; 232:116725. [PMID: 39746456 DOI: 10.1016/j.bcp.2024.116725] [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: 09/02/2024] [Revised: 11/14/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Drug delivery to cancer cells continues to present a major therapeutic challenge. Mesenchymal stem cells (MSCs) possess an intrinsic ability to migrate specifically to tumor tissues, making them promising candidates for targeted drug delivery. Evidence from preclinical studies indicates that MSCs loaded with therapeutic anti-cancer agents exhibit considerable anti-tumor activity. Moreover, several clinical trials are currently evaluating their effectiveness in cancer patients. The integration of MSCs with synthetic nanoparticles (NPs) enhances their therapeutic potential, particularly through the use of cell membrane-coated NPs, which represent a significant advancement in the field. This review systematically investigates the tumor microenvironment, the sources of MSCs, the tumor homing mechanisms, and the methods of loading and releasing anticancer drugs from MSCs. Furthermore, cutting-edge strategies to improve the efficacy of MSCs based drug delivery systems (DDS) including the innovative use of MSC membrane coated nanoparticles have been discussed. The study concludes with an overview of the therapeutic use of MSCs as drug carriers, including a detailed analysis of the mechanisms by which MSCs deliver therapeutics to cancer cells, enabling targeted drug delivery. It aims to elucidate the current state of this approach, identify key areas for development, and outline potential future directions for advancing MSCs based cancer therapies.
Collapse
Affiliation(s)
- Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
41
|
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
|
42
|
Izadiyan Z, Misran M, Kalantari K, Webster TJ, Kia P, Basrowi NA, Rasouli E, Shameli K. Advancements in Liposomal Nanomedicines: Innovative Formulations, Therapeutic Applications, and Future Directions in Precision Medicine. Int J Nanomedicine 2025; 20:1213-1262. [PMID: 39911259 PMCID: PMC11794392 DOI: 10.2147/ijn.s488961] [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: 07/27/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Liposomal nanomedicines have emerged as a pivotal approach for the treatment of various diseases, notably cancer and infectious diseases. This manuscript provides an in-depth review of recent advancements in liposomal formulations, highlighting their composition, targeted delivery strategies, and mechanisms of action. We explore the evolution of liposomal products currently in clinical trials, emphasizing their potential in addressing diverse medical challenges. The integration of immunotherapeutic agents within liposomes marks a paradigm shift, enabling the design of 'immuno-modulatory hubs' capable of orchestrating precise immune responses while facilitating theranostic applications. The recent COVID-19 pandemic has accelerated research in liposomal-based vaccines and antiviral therapies, underscoring the need for improved delivery mechanisms to overcome challenges like rapid clearance and organ toxicity. Furthermore, we discuss the potential of "smart" liposomes, which can respond to specific disease microenvironments, enhancing treatment efficacy and precision. The integration of artificial intelligence and machine learning in optimizing liposomal designs promises to revolutionize personalized medicine, paving the way for innovative strategies in disease detection and therapeutic interventions. This comprehensive review underscores the significance of ongoing research in liposomal technologies, with implications for future clinical applications and enhanced patient outcomes.
Collapse
Affiliation(s)
- Zahra Izadiyan
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Biomedical Engineering, Hebei University of Technology, Tianjin, People’s Republic of China
- School of Engineering, Saveetha University, Chennai, India
| | - Pooneh Kia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Elisa Rasouli
- Department of Electrical and Electronics Engineering, Nanyang Technological University, Nanyang, Singapore
| | - Kamyar Shameli
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
| |
Collapse
|
43
|
Khalid W, Aslam A, Ahmed N, Sarfraz M, Khan JA, Mohsin S, Rajoka MSR, Nazir I, Amirzada MI. Human Plasma-Derived Exosomes: A Promising Carrier System for the Delivery of Hydroxyurea to Combat Breast Cancer. AAPS PharmSciTech 2025; 26:42. [PMID: 39843767 DOI: 10.1208/s12249-024-03028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
The aim of the present study was to investigate the potential of human plasma derived exosomes for the delivery of hydroxyurea to enhance its therapeutic efficacy in breast cancer. Plasma derived exosomes were isolated using differential centrifugation along with ultrafiltration method. Hydroxyurea was encapsulated in exosomes using a freeze-thaw method. The exosomes and Exo-HU were characterized for their size distribution, drug entrapment efficiency, in-vitro drug release profile, morphological analysis and cytotoxic effects on MCF-7 cell line. The results showed a mean size of 178.8 nm and a zeta potential of -18.3 mV, indicating good stability and 70% encapsulation effectiveness for HU. Exo-HU produced sustained drug release action with a considerable percentage released within 72 h. The morphological analysis indicated that the plasma derived exosomes were spherical, and cup shaped. In cytotoxicity studies on MCF-7 cells, Exo-HU has reduced cell viability compared to HU and blank exosomes. Findings of this study showed that human plasma-derived exosomes have been considered as effective delivery vehicle for hydroxyurea, potentially improving breast cancer treatment outcomes.
Collapse
Affiliation(s)
- Wajeeha Khalid
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Afeefa Aslam
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Nadeem Ahmed
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, 54000, Pakistan
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, 64141, Al Ain, United Arab Emirates
| | | | - Sabeeh Mohsin
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | | | - Imran Nazir
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan.
| | - Muhammad Imran Amirzada
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan.
| |
Collapse
|
44
|
Basingab FS, Alshahrani OA, Alansari IH, Almarghalani NA, Alshelali NH, Alsaiary AH, Alharbi N, Zaher KA. From Pioneering Discoveries to Innovative Therapies: A Journey Through the History and Advancements of Nanoparticles in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:27-51. [PMID: 39867813 PMCID: PMC11761866 DOI: 10.2147/bctt.s501448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
Nanoparticle technology has revolutionized breast cancer treatment by offering innovative solutions addressing the gaps in traditional treatment methods. This paper aimed to comprehensively explore the historical journey and advancements of nanoparticles in breast cancer treatment, highlighting their transformative impact on modern medicine. The discussion traces the evolution of nanoparticle-based therapies from their early conceptualization to their current applications and future potential. We initially explored the historical context of breast cancer treatment, highlighting the limitations of conventional therapies, such as surgery, radiation, and chemotherapy. The advent of nanotechnology has introduced a new era characterized by the development of various nanoparticles, including liposomes, dendrimers, and gold nanoparticles, designed to target cancer cells with remarkable precision. We further described the mechanisms of action for nanoparticles, including passive and active targeting, and reviewed significant breakthroughs and clinical trials that have validated their efficacy. Current applications of nanoparticles in breast cancer treatment have been examined, showcasing clinically approved therapies and comparing their effectiveness with traditional methods. This article also discusses the latest advancements in nanoparticle research, including drug delivery systems and combination therapy innovations, while addressing the current technical, biological, and regulatory challenges. The technical challenges include efficient and targeted delivery to tumor sites without affecting healthy tissue; biological, such as potential toxicity, immune system activation, or resistance mechanisms; economic, involving high production and scaling costs; and regulatory, requiring rigorous testing for safety, efficacy, and long-term effects to meet stringent approval standards. Finally, we have explored emerging trends, the potential for personalized medicine, and the ethical and social implications of this transformative technology. In conclusion, through comprehensive analysis and case studies, this paper underscores the profound impact of nanoparticles on breast cancer treatment and their future potential.
Collapse
Affiliation(s)
- Fatemah S Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Omniah A Alshahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Ibtehal H Alansari
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada A Almarghalani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada H Alshelali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Abeer Hamad Alsaiary
- Biology Department, College of Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Najwa Alharbi
- Department of Biology Science, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Kawther A Zaher
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| |
Collapse
|
45
|
Siva M, Das K, Rana P, Saha A, Mandal D, Barik A, Stewart A, Maity B, Das P. Liposomal Encapsulation of Chlorambucil with a Terpyridine-Based, Glutathione-Targeted Optical Probe Facilitates Cell Entry and Cancer Cell Death. ACS APPLIED BIO MATERIALS 2025; 8:570-581. [PMID: 39686811 DOI: 10.1021/acsabm.4c01448] [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] [Indexed: 12/18/2024]
Abstract
The nitrogen mustard alkylating agent chlorambucil (CBL) is a critical component of chemotherapeutic regimens used in the treatment of chronic lymphocytic leukemia. The cancer cell-killing actions of CBL are limited by glutathione (GSH) conjugation, a process catalyzed by the GSH transferase hGSTA1-1 that triggers CBL efflux from cells. In the cancer cell microenvironment, intracellular GSH levels are elevated to counterbalance oxidative stress generated due to the high glycolytic demand. As many chemotherapeutic drugs trigger cell death through mechanisms that depend on reactive oxygen species (ROS), antioxidant capacity in cancer cells also represents a barrier to anticancer therapies. Here, we demonstrate that a heightened GSH content in cancer cells can also be exploited for cell-selective drug delivery. We successfully synthesized a malononitrile conjugate terpyridine-based derivative L1, which specifically reacts with GSH in the presence of other biologically relevant amino acids including cysteine (Cys) and homocysteine (Hcy). The significant change in the electronic spectra of L1 in the presence of GSH confirmed GSH detection, which was further corroborated by density functional theory calculations. We next encapsulated CBL into L1-containing, anthracene-functionalized, and 10,12-pentacosadiynoic acid (PCDA)- and 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC)-based liposomes (Lip-CBL-L1). We established successful CBL encapsulation and release from L1-containing liposomes in GSH-enriched cancer cells in vitro. Both Lip-CBL-L1 and the L1-lacking Lip-CBL control displayed cell-killing activity. However, human triple-negative breast cancer cells MDAMB231, human lung cancer cells A549, and murine leukemic WEHI cells were more sensitive to the cytotoxic effects of Lip-CBL-L1 compared to the nonmalignant cells (AC16 and HEK293). Indeed, in these cancer cell lines, Lip-CBL-L1 induced greater ROS generation compared to that of Lip-CBL. Together, our results provide initial evidence of the feasibility of exploiting the unique oxidant environment of cancer cells for optimized drug delivery.
Collapse
Affiliation(s)
- Mallayasamy Siva
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Kiran Das
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI) Campus, Raebareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Priya Rana
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Abhijit Saha
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Debasish Mandal
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Atanu Barik
- Radiation and Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai, Maharashtra 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Adele Stewart
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Biswanath Maity
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI) Campus, Raebareli Road, Lucknow, Uttar Pradesh 226014, India
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN80, Sector V, Bidhan Nagar, Kolkata, West Bengal 700091, India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
46
|
Yang M, Kim S, Jeong S, Lee S, Lee S, Jo H, Kim N, Song N, Park SC, Lee D. Harnessing Dual-Responsive Polymeric Micelles for Precision Oxidative Stress Amplification in Targeted Cancer Therapy. Biomacromolecules 2025; 26:437-448. [PMID: 39648938 DOI: 10.1021/acs.biomac.4c01250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Targeting the altered redox balance in cancer cells, this study explores a strategy to induce selective cancer cell death by combining reactive oxygen species (ROS) generation with glutathione (GSH) depletion. We developed oxidative stress-amplifying polymeric (pCB) micelles that function both as therapeutic agents and carriers for GSH-depleting retinoic acid prodrug (BRDP). pCB incorporating ROS-generating cinnamaldehyde and a GSH-depleting quinone methide precursor could self-assemble into micelles encapsulating BRDP, delivering both ROS generators and GSH-depleting drugs. The micelles were surface-functionalized with the tripeptide Arg-Gly-Asp (RGD) for targeted delivery to integrin-overexpressing tumors. In a mouse xenograft model, RGD-decorated BRDP-loaded micelles significantly accumulated in tumor sites, enhancing anticancer efficacy without toxicity to normal tissues. This study marks significant advancement in the field of oxidative stress-amplifying polymeric precursors, presenting a novel and highly effective anticancer therapeutic approach that integrates multiple tumor-specific triggers and ROS-mediated mechanisms.
Collapse
Affiliation(s)
- Manseok Yang
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Sujin Kim
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Seungwon Jeong
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Suyeon Lee
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Seunga Lee
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Hanui Jo
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Nuri Kim
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Nanhee Song
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| | - Seong-Cheol Park
- Department of Polymer Engineering, Suncheon National University, Suncheon, Chonnam 57922, Korea
| | - Dongwon Lee
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
- Department of Polymer-Nano Science and Technology, Jeonbuk National University, Jeonju, Jeonbuk 54896, Korea
| |
Collapse
|
47
|
Wan W, Liu H, Zou J, Xie T, Zhang G, Ying W, Zou X. The optimization and application of photodynamic diagnosis and autofluorescence imaging in tumor diagnosis and guided surgery: current status and future prospects. Front Oncol 2025; 14:1503404. [PMID: 39845324 PMCID: PMC11750647 DOI: 10.3389/fonc.2024.1503404] [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: 09/28/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Photodynamic diagnosis (PDD) and autofluorescence imaging (AFI) are emerging cancer diagnostic technologies that offer significant advantages over traditional white-light endoscopy in detecting precancerous lesions and early-stage cancers; moreover, they hold promising potential in fluorescence-guided surgery (FGS) for tumors. However, their shortcomings have somewhat hindered the clinical application of PDD and AFI. Therefore, it is imperative to enhance the efficacy of PDD and AFI, thereby maximizing their potential for practical clinical use. This article reviews the principles, characteristics, current research status, and advancements of PDD and AFI, focusing on analyzing and discussing the optimization strategies of PDD and AFI in tumor diagnosis and FGS scenarios. Considering the practical and technical feasibility, optimizing PDD and AFI may result in an effective real-time diagnostic tool to guide clinicians in tumor diagnosis and surgical guidance to achieve the best results.
Collapse
Affiliation(s)
- Wei Wan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huiquan Liu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tianpeng Xie
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Guoxi Zhang
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Weihai Ying
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofeng Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
48
|
Kampaengsri S, Yong GY, Aryamueang S, Ouengwanarat B, Pewklang T, Chansaenpak K, Jitrapakdee S, Kue CS, Kamkaew A. Heptamethine cyanine-based polymeric nanoparticles for photothermal therapy in HCT116 human colon cancer model. Sci Rep 2025; 15:884. [PMID: 39762372 PMCID: PMC11704253 DOI: 10.1038/s41598-024-83249-y] [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: 10/14/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
In this work, we synthesize a quinoline-based heptamethine cyanine, QuCy7, with sulfonate groups to enhance water solubility. This dye demonstrates exceptional near-infrared absorption beyond 750 nm, accompanied by photothermal properties but low photostability. Encapsulating QyCy7 with polyethylene glycol to form nanopolymer, QuCy7@mPEG NPs, addresses the issue of its photoinstability. TEM showed that QuCy7@mPEG NPs possess a spherical morphology, featuring a core-shell structure with a size of around 120 nm in diameter. Upon irradiation with an 808 nm laser for 10 min, a significant increase in temperature up to 24 °C can be achieved with a photothermal conversion (PTC) rate of approximately 35%. QuCy7@mPEG NPs exhibit remarkable photothermal stability as compared to QuCy7. The efficiency of QuCy7@mPEG NPs was demonstrated by the in vitro PTT studies. Finally, the nanoparticles' acute toxicity and effectiveness were assessed using the chick embryo model. The results provide compelling evidence that QuCy7@mPEG NPs are safe without inducing hemolysis, inhibit angiogenesis when exposed to light, and exhibit anti-tumor activity with a 76% reduction in tumor size compared to QuCy7 (40%). Thus suggesting the sulfonate groups can enhance water solubility, and its nanopolymer is biocompatible and possesses superior anti-tumor efficacy.
Collapse
Affiliation(s)
- Sastiya Kampaengsri
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Gong Yi Yong
- School of Graduate Studies, Management and Science University, Seksyen 13, Shah Alam, 40100, Selangor, Malaysia
| | - Sirimongkon Aryamueang
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Bongkot Ouengwanarat
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Thitima Pewklang
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Kantapat Chansaenpak
- National Nanotechnology Center, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, 12120, Thailand
| | - Sarawut Jitrapakdee
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Chin-Siang Kue
- Faculty of Health and Life Sciences, Management and Science University, Seksyen 13, 40100, Shah Alam, Selangor, Malaysia.
| | - Anyanee Kamkaew
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand.
| |
Collapse
|
49
|
Elwan AG, Mohamed TM, Beltagy DM, El Gamal DM. The therapeutic role of naringenin nanoparticles on hepatocellular carcinoma. BMC Pharmacol Toxicol 2025; 26:3. [PMID: 39754228 PMCID: PMC11697747 DOI: 10.1186/s40360-024-00823-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Naringenin, a flavonoid compound found in citrus fruits, possesses valuable anticancer properties. However, its potential application in cancer treatment is limited by poor bioavailability and pharmacokinetics at tumor sites. To address this, Naringenin nanoparticles (NARNPs) were prepared using the emulsion diffusion technique and their anticancer effects were investigated in HepG2 cells. METHODS The particle size of NARNPs was determined by transmission electron microscopy and scanning electron microscopy analysis. NARNP is characterized by Fourier transform infrared spectroscopy and X-ray diffraction. Study the cytotoxic effects of various doses of naringenin, NARNPs and DOX on HepG2 and WI38 cell lines after 24 h and 48 h using the MTT assay. Flow cytometric analysis was used to study the apoptotic cells. The study also examined the expression of apoptotic proteins (p53) and autophagy-related genes ATG5, LC3 after treatment with naringenin, NARNPs, doxorubicin, and their combinations in HepG2 cells. RESULTS The particle size of NARNPs was determined by transmission electron microscopy and scanning electron microscopy analysis, showing mean diameters of 54.96 ± 18.6 nm and 31.79 ± 6.8 nm, respectively. Fourier transform infrared spectroscopy confirmed successful conjugation between naringenin and NARNPs. NARNPs were in an amorphous state that was determined by X-ray diffraction. The IC50 values were determined as 22.32 µg/ml for naringenin, 1.6 µg/ml for NARNPs and 0.46 µg/ml for doxorubicin. Flow cytometric analysis showed that NARNPs induced late apoptosis in 56.1% of HepG2 cells and had no cytotoxic effect on WI38 cells with 97% viable cells after 48 h of incubation. NARNPs induced cell cycle arrest in the Go/G1 and G2/M phases in HepG2 cells. The results showed increased expression of ATG5, LC3, and p53 in HepG2 cells treated with IC50 concentrations after 48 h of incubation. NARNPs enhanced the cytotoxic effect of doxorubicin in HepG2 cells but decreased the cytotoxic effect of doxorubicin in WI38 cells. CONCLUSIONS The study demonstrated that NARNPs effectively inhibit cell proliferation and induce apoptosis in human hepatocellular carcinoma cells. Importantly, NARNPs showed no cytotoxic effects on normal cells, indicating their potential as a promising therapy for hepatocarcinogenesis. Combining NARNPs with chemotherapy drugs could present a novel approach for treating human cancers.
Collapse
Affiliation(s)
- Aya G Elwan
- Biochemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Tarek M Mohamed
- Biochemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Doha M Beltagy
- Biochemistry Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Doaa M El Gamal
- Biochemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
50
|
Meher N, Bidkar AP, Wadhwa A, Bobba KN, Dhrona S, Dasari C, Mu C, Fong COY, Cámara JA, Ali U, Basak M, Bulkley D, Steri V, Fontaine SD, Zhu J, Oskowitz A, Aggarwal RR, Sriram R, Chou J, Wilson DM, Seo Y, Santi DV, Ashley GW, VanBrocklin HF, Flavell RR. PET Imaging Using 89Zr-Labeled StarPEG Nanocarriers Reveals Heterogeneous Enhanced Permeability and Retention in Prostate Cancer. Mol Cancer Ther 2025; 24:141-151. [PMID: 39331510 DOI: 10.1158/1535-7163.mct-24-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/05/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
The enhanced permeability and retention (EPR) effect controls passive nanodrug uptake in tumors and may provide a high tumor payload with prolonged retention for cancer treatment. However, EPR-mediated tumor uptake and distribution vary by cancer phenotype. Thus, we hypothesized that a companion PET imaging surrogate may benefit EPR-mediated therapeutic drug delivery. We developed two 89Zr-radiolabeled nanocarriers based on 4-armed starPEG40kDa with or without talazoparib (TLZ), a potent PARP inhibitor, as surrogates for the PEG-TLZ4 therapeutic scaffold. For PET imaging, PEG-DFB4 and PEG-DFB1-TLZ3 were radiolabeled with 89Zr by replacing one or all four copis of TLZ on PEG-TLZ4 with deferoxamine B (DFB). The radiolabeled nanodrugs [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 were tested in vivo in prostate cancer subcutaneous (s.c.) xenografts (22Rv1, LTL-545, and LTL-610) and 22Rv1 metastatic models. Their EPR-mediated tumoral uptake and penetration was compared with CT26, a known EPR-high cell line. MicroPET/CT images, organ biodistribution, and calculated kinetic parameters showed high uptake in CT26 and LTL-545 and moderate to low uptake in LTL-610 and 22Rv1. MicroPET/CT and high-resolution autoradiographic images showed nanocarrier penetration into highly permeable CT26, but heterogeneous peripheral accumulation was observed in LTL-545, LTL-610, and 22Rv1 s.c. xenografts and metastatic tumors. CD31 staining of tumor sections showed homogenous vascular development in CT26 tumors and heterogeneity in other xenografts. Both [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 showed similar accumulation and distribution in s.c. and metastatic tumor models. Both nanocarriers can measure tumor model passive uptake heterogeneity. Although heterogeneous, prostate cancer xenografts had low EPR. These starPEG nanocarriers could be used as PET imaging surrogates to predict drug delivery and efficacy.
Collapse
Affiliation(s)
- Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Chandrashekhar Dasari
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, California
| | - Changhua Mu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Cyril O Y Fong
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Juan A Cámara
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Umama Ali
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Megha Basak
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - David Bulkley
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | - Jun Zhu
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Adam Oskowitz
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, California
| | - Rahul R Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Jonathan Chou
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | | | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| |
Collapse
|