1
|
G P, Singh M, Gupta PK, Shukla R. Synergy of Microfluidics and Nanomaterials: A Revolutionary Approach for Cancer Management. ACS APPLIED BIO MATERIALS 2025; 8:2716-2734. [PMID: 40100776 DOI: 10.1021/acsabm.5c00123] [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: 03/20/2025]
Abstract
Cancer affects millions of individuals every year and is the second most common cause of death. Various therapeutic strategies are explored for the management of cancer including radiation therapy and chemotherapy with or without surgical procedures. However, the drawbacks like poor cancer cell targeting and higher toxicity for healthy cells need the advancement of the therapeutic strategy. The exploration of nanomedicine achieves targeted distribution, and the adoption of microfluidics technology for the preparation of the nanoparticulate system has enhanced the efficacy and uniformity of the nanocarriers. The overview of the existing designs of the microfluidics device assisted in the preparation of the nanoparticles, and various nanodelivery systems formulated using the microfluidic device including liposomes, lipidic nanocarriers, quantum dots, polymeric nanoparticles, and metallic nanocarriers are discussed in this review. Further, the challenges associated with the fabrication of the microfluidics device and the fabrication of microfluidics device-based nanoparticles are detailed here.
Collapse
Affiliation(s)
- Pramoda G
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Mansi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Piyush Kumar Gupta
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Bio-Science and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310, India
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002, India
| |
Collapse
|
2
|
Wu J, Huang H, Xu W, Cui B, Sun P, Hao X, Jiang S, Hou X, Qi X, Wei Z, Cheng Y, Zheng Y, Liu K, He J. Inflammation-driven biomimetic nano-polyphenol drug delivery system alleviates severe acute pancreatitis by inhibiting macrophage PANoptosis and pancreatic enzymes oversecretion. J Adv Res 2025:S2090-1232(25)00225-5. [PMID: 40210149 DOI: 10.1016/j.jare.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
INTRODUCTION Severe acute pancreatitis (SAP) is a critical inflammatory disease with high morbidity and mortality. Current treatments focused on symptomatic relief but failed to prevent inflammation progression in cellular level. OBJECTIVES In order to develop an SAP-targeting drug delivery system to alleviate SAP in cellular level and illustrate its mechanism, we explored the use of proanthocyanidin (PYD) and pentoxifylline (PTX) loaded into macrophage membrane-coated self-assembly nanoparticles (FePTX@CM NPs) for targeted SAP treatment. The combination application of these two drugs was innovative in SAP aid. METHODS We developed the NPs by self-assembly strategy and cell membrane coating. Its particle size and zeta potential were measured by dynamic light scatter (DLS). The morphology of the NPs was observed by transmission electron microscopy (TEM). And the encapsulation efficiency was evaluated by nano-flow cytometry. The total protein profile was determined via Coomassie brilliant blue. We explore the mechanism of our NPs against SAP in cellular and animal levels. Bioinformatics approaches, TEM, immunofluorescent assay and co-immunoprecipitation were performed to comprehensively explain the specific anti-SAP mechanism of FePTX@CM NPs. RESULTS After inflammation-driven targeting, PYD in the NPs inhibited pancreatic amylase and lipase release by suppressing mitochondrial reactive oxygen species (mtROS)/Golgi stress, while PTX prevented SAP-associated macrophage PANoptosis by inhibiting Zbp1 signal pathway. The protection effect of these biomimetic NPs worked from different aspects to alleviate SAP symptoms and inflammation progression in relative cells. CONCLUSION The FePTX@CM NPs demonstrated effective pancreas targeting, reduced systemic inflammation especially pro-inflammatory cell recruitment and activation, and minimized tissue damage in SAP mouse models, offering a promising therapeutic strategy for clinical SAP management.
Collapse
Affiliation(s)
- Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Hai Huang
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Beibei Cui
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Pengcheng Sun
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - XinYan Hao
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Shihe Jiang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xuyang Hou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaoyan Qi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zuxing Wei
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yimiao Cheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yanwen Zheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Kuijie Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
3
|
Zhou M, Han R, Xu W, Hao X, Peng Y, Tang Y, Sun P, Tang T, Wu J, Xiang D. Biomimetic Atorvastatin Self-Assembled Nanomedicine Inhibits the Cyclooxygenase-2/Prostaglandin E2 Pathway Enhanced Photothermal and Antitumor Immunity. Biomater Res 2025; 29:0149. [PMID: 40040956 PMCID: PMC11876541 DOI: 10.34133/bmr.0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Cancer continues to pose remarkable medical challenges worldwide. While current cancer therapies can lead to initial clinical improvement, they are often followed by recurrence, metastasis, and drug resistance, underscoring the urgent need for innovative treatment strategies. Atorvastatin calcium (AC), a widely used lipid-lowering and anti-inflammation drug in the clinic, has shown antitumor potential. To further improve the antitumor efficacy, we developed self-assembled AC and polydopamine (PDA) nanoparticles whose surface was coated with macrophage membranes (CM) as a biomimetic drug delivery system [AC@PDA@CM (APM)]. APM showed high drug-loading capacity, excellent stability, excellent bioavailability, and tumor-targeting ability, ultimately achieving photothermal synergistic cancer immunotherapy. Our findings indicate that APM efficiently delivers AC to tumor sites while leveraging photothermal therapy (PTT) to enhance local tumor ablation and antitumor immune effect. Notably, APM mitigates tumor immunosuppression triggered by PTT through AC, suppressing the COX-2/PGE2 pathway and immune evasion signal CD47. Furthermore, APM notably reduced nonspecific distribution and side effects, which is conducive to ensuring the safety level of medication. This integrated approach boosts therapeutic efficacy and highlights the potential of APM as a multifunctional agent for cancer therapy, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Ruyue Han
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Xinyan Hao
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Yanjin Peng
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Yucheng Tang
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Pengcheng Sun
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| |
Collapse
|
4
|
Wu J, Hao X, Qi L, Xu W, Yin C, Tang Y, Sun P, Liao D, Hu X, Tang T, Tu C, Xiang D, Li Z. Assembly of a biomimetic copper-based nanocomplex for alleviating hypoxia to enhance cuproptosis against osteosarcoma and lung metastasis. Acta Biomater 2025; 193:348-361. [PMID: 39710219 DOI: 10.1016/j.actbio.2024.12.049] [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: 08/29/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Osteosarcoma tissues demonstrated elevated expression of proteins (FDX1 and DLAT) integral to cuproptosis in our preliminary study, indicating the potential effectiveness of anti-tumor strategies predicated on this process. Nevertheless, the overexpression of copper export proteins and the challenge of copper ion penetration may contribute to insufficient local copper ion concentration for inducing cuproptosis. Herein, we engineered a biomimetic copper-elesclomol-polyphenol network for the efficient delivery of copper ions and the copper ionophore elesclomol. Simultaneously, we integrated catalase (CAT) to alleviate tumor hypoxia, thereby inducing a greater reliance of tumor cells on aerobic respiration and enhancing cuproptosis sensitivity. In vitro analyses revealed that the nanocomplex exhibited potent cytotoxicity and displayed hallmark characteristics of cuproptosis. In vivo trials further validated targeted tumor accumulation, resulting in the suppression of tumor growth and lung metastasis. An augmentation in the proportion of activated immune cells in both tumor and draining lymph nodes was observed. The improvement of immunosuppressive microenvironment facilitated a synergistic antitumor effect with cuproptosis. The therapeutic efficacy was further evidenced in two osteosarcoma models, highlighting the potential as a safe and effective strategy against osteosarcoma and lung metastasis. STATEMENT OF SIGNIFICANCE: Osteosarcoma tissues exhibit a marked increase in the expression of proteins FDX1 and DLAT, which are crucial for cuproptosis. Moreover, cells that depend on mitochondrial respiration are more susceptible to cuproptosis. Here we developed a biomimetic copper-based nanocomplex to trigger cuproptosis against osteosarcoma and lung metastases. The nanocomplex demonstrated excellent biocompatibility and tumor targeting. Catalase incorporating facilitated oxygen generation within tumor microenvironment and alleviated hypoxia, thereby inducing a greater reliance of tumor cells on aerobic respiration and enhancing cuproptosis sensitivity. Simultaneously, the released Cu-elesclomol complexes induced proteotoxic stress responses and efficiently elicited cuproptosis, leading to increased release of proinflammatory factors and triggering anti-tumor immune activation. Our strategy holds promise for osteosarcoma treatment by inducing cuproptosis and achieving potent tumor suppression.
Collapse
Affiliation(s)
- Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China; Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Xinyan Hao
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China
| | - Chi Yin
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Yucheng Tang
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China
| | - Pengcheng Sun
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China
| | - Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University Changsha 410031, China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy, Central South University Changsha 410011, China.
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China.
| |
Collapse
|
5
|
Palaniyandi T, Ravi M, Sivaji A, Baskar G, Viswanathan S, Wahab MRA, Surendran H, Nedunchezhian S, Ahmad I, Veettil VN. Recent advances in microfluidic chip technologies for applications as preclinical testing devices for the diagnosis and treatment of triple-negative breast cancers. Pathol Res Pract 2024; 264:155711. [PMID: 39536540 DOI: 10.1016/j.prp.2024.155711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The leading cause of cancer-related death among female patients is breast cancer. Among all the types of breast cancer, triple-negative breast cancer (TNBC) is the most dangerous molecular subtype of breast cancer characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2) expression. Since there is no particular therapeutic strategy for TNBC that has been shown to worsen the disease prognosis, 3D models are superior to 2D models as a predictive tool for drug discovery because they more accurately reflect the in vivo biological components of humans. Importantly, all 3D models struggle to gather many high-quality tumour cells from clinical tumours. Physicians may not get huge tumour tissues from patients, and clinical tumours may have necrosis, fat, and blood vessel components. Therefore, there is an immediate need to find an efficient method to consistently and quickly produce a large number of homogeneous tumour models for individual treatment without cell wastage. Microfluidic technologies, which are specifically engineered to manipulate small quantities of fluids, have been utilised to produce particles for drug delivery applications. This development is indicative of a recent trend, as it provides the ability to regulate particle size and material composition. This review focuses on the topic of tumor-on-a-chip, microfluidic chip manufacturing, and drug screening for triple-negative breast cancer. Particular emphasis is placed on cancer biomarker diagnostics, 3D preclinical model development, and treatment strategies for triple-negative breast cancer.
Collapse
Affiliation(s)
- Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu 600095, India; ACS-Advanced Medical Research Institute, Dr. M.G.R Educational and Research Institute, Maduravoyal, Chennai 600095, India.
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600 116, India
| | - Asha Sivaji
- Department of Biochemistry, DKM College for Women, Vellore, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu 600095, India
| | - Sandhiya Viswanathan
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu 600095, India
| | - Mugip Rahaman Abdul Wahab
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu 600095, India
| | - Hemapreethi Surendran
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu 600095, India
| | - Sandhya Nedunchezhian
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu 600095, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia; Health and Medical Research Center, King Khalid University, AlQura'a, Abha, Saudi Arabia
| | - Vajid Nettoor Veettil
- Iqraa Centre for Research and Development, IQRAA International Hospital and Research Centre, Kozhikode, Kerala, India
| |
Collapse
|
6
|
Hou X, Chen Q, Fang Y, Zhang L, Huang S, Xu M, Ren Y, Shi Z, Wei Y, Li L. iRGD-Guided Silica/Gold Nanoparticles for Efficient Tumor-Targeting and Enhancing Antitumor Efficacy Against Breast Cancer. Int J Nanomedicine 2024; 19:8237-8251. [PMID: 39157735 PMCID: PMC11329605 DOI: 10.2147/ijn.s474135] [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: 04/19/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024] Open
Abstract
Background Breast cancer presents significant challenges due to the limited effectiveness of available treatments and the high likelihood of recurrence. iRGD possesses both RGD sequence and C-terminal sequence and has dual functions of targeting and membrane penetration. iRGD-modified nanocarriers can enhance drug targeting of tumor vascular endothelial cells and penetration of new microvessels, increasing drug concentration in tumor tissues. Methods The amidation reaction was carried out between SiO2/AuNCs and iRGD/PTX, yielding a conjugated drug delivery system (SiO2/AuNCs-iRGD/PTX, SAIP@NPs). The assessment encompassed the characterization of the morphology, particle size distribution, physicochemical properties, in vitro release profile, cytotoxicity, and cellular uptake of SAIP@NPs. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed using a small animal in vivo imaging system and a tumor-bearing nude mice model, respectively. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed utilizing a small animal in vivo imaging system and an in situ nude mice breast cancer xenograft model, respectively. Results The prepared SAIP@NPs exhibited decent stability and a certain slow-release effect in phosphate buffer (PBS, pH 7.4). In vitro studies had shown that, due to the dual functions of transmembrane and targeting of iRGD peptide, SAIP@NPs exhibited strong binding to integrin αvβ3, which was highly expressed on the membrane of MDA-MB-231 cells, improving the uptake capacity of tumor cells, inhibiting the rapid growth of tumor cells, and promoting tumor cell apoptosis. The results of animal experiments further proved that SAIP@NPs had longer residence time in tumor sites, stronger anti-tumor effect, and no obvious toxicity to major organs of experimental animals. Conclusion The engineered SAIP@NPs exhibited superior functionalities including efficient membrane permeability, precise tumor targeting, and imaging, thereby significantly augmenting the therapeutic efficacy against breast cancer with a favorable safety profile.
Collapse
Affiliation(s)
- Xuefeng Hou
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Qi Chen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Ying Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Li Zhang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Shuoheng Huang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Minjie Xu
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Yaning Ren
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Zhansen Shi
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Yan Wei
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Lihua Li
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
- Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| |
Collapse
|
7
|
Pan W, Gu F, Yan X, Huang J, Liao H, Niu F. Biomacromolecular carriers based hydrophobic natural products for potential cancer therapy. Int J Biol Macromol 2024; 269:132274. [PMID: 38734357 DOI: 10.1016/j.ijbiomac.2024.132274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Cancer is the second leading cause of death worldwide. It was estimated that 90 % of cancer-related deaths were attributable to the development of multi-drug resistance (MDR) during chemotherapy, which results in ineffective chemotherapy. Hydrophobic natural products plays a pivotal role in the field of cancer therapy, with the potential to reverse MDR in tumor cells, thereby enhancing the efficacy of tumor therapy. However, their targeted delivery is considered a major hurdle in their application. The advent of numerous approaches for encapsulating bioactive ingredients in the nanodelivery systems has improved the stability and targeted delivery of these biomolecules. The manuscript comprehensively analyses the nanodelivery systems of bioactive compounds with potential cancer therapy applications, including liposomes, emulsions, solid lipid nanoparticles (NPs), and polymeric NPs. Then, the advantages and disadvantages of various nanoagents in the treatment of various cancer types are critically discussed. Further, the application of multiple-compbine delivery methods to overcome the limitations of single-delivery have need critically analyzed, which thus could help in the designing nanodrug delivery systems for bioactive compounds in clinical settings. Therefore, the review is timely and important for development of efficient nanodelivery systems involving hydrophobic natural products to improve pharmacokinetic properties for effective cancer treatment.
Collapse
Affiliation(s)
- Weichun Pan
- Food Safety Key Lab of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Feina Gu
- Food Safety Key Lab of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Xinyu Yan
- College of Materials and Metallurgy, Guizhou University, Guiyang 550025, China
| | - Jianghui Huang
- Food Safety Key Lab of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Huabin Liao
- Food Safety Key Lab of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Fuge Niu
- Food Safety Key Lab of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| |
Collapse
|
8
|
Dasari N, Guntuku GS, Pindiprolu SKSS. Targeting triple negative breast cancer stem cells using nanocarriers. DISCOVER NANO 2024; 19:41. [PMID: 38453756 PMCID: PMC10920615 DOI: 10.1186/s11671-024-03985-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
Breast cancer is a complex and heterogeneous disease, encompassing various subtypes characterized by distinct molecular features, clinical behaviors, and treatment responses. Categorization of subtypes is based on the presence or absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), leading to subtypes such as luminal A, luminal B, HER2-positive, and triple-negative breast cancer (TNBC). TNBC, comprising around 20% of all breast cancers, lacks expression of ER, PR, and HER2 receptors, rendering it unresponsive to targeted therapies and presenting significant challenges in treatment. TNBC is associated with aggressive behavior, high rates of recurrence, and resistance to chemotherapy. Tumor initiation, progression, and treatment resistance in TNBC are attributed to breast cancer stem cells (BCSCs), which possess self-renewal, differentiation, and tumorigenic potential. Surface markers, self-renewal pathways (Notch, Wnt, Hedgehog signaling), apoptotic protein (Bcl-2), angiogenesis inhibition (VEGF inhibitors), and immune modulation (cytokines, immune checkpoint inhibitors) are among the key targets discussed in this review. However, targeting the BCSC subpopulation in TNBC presents challenges, including off-target effects, low solubility, and bioavailability of anti-BCSC agents. Nanoparticle-based therapies offer a promising approach to target various molecular pathways and cellular processes implicated in survival of BSCS in TNBC. In this review, we explore various nanocarrier-based approaches for targeting BCSCs in TNBC, aiming to overcome these challenges and improve treatment outcomes for TNBC patients. These nanoparticle-based therapeutic strategies hold promise for addressing the therapeutic gap in TNBC treatment by delivering targeted therapies to BCSCs while minimizing systemic toxicity and enhancing treatment efficacy.
Collapse
Affiliation(s)
- Nagasen Dasari
- Andhra University College of Pharmaceutical Sciences, Andhra University, Vishakhapatnam, Andhra Pradesh, India.
- Aditya Pharmacy College, Surampalem, Andhra Pradesh, India.
- Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India.
| | - Girija Sankar Guntuku
- Andhra University College of Pharmaceutical Sciences, Andhra University, Vishakhapatnam, Andhra Pradesh, India
| | - Sai Kiran S S Pindiprolu
- Aditya Pharmacy College, Surampalem, Andhra Pradesh, India
- Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India
| |
Collapse
|
9
|
Naghib SM, Mohammad-Jafari K. Microfluidics-mediated Liposomal Nanoparticles for Cancer Therapy: Recent Developments on Advanced Devices and Technologies. Curr Top Med Chem 2024; 24:1185-1211. [PMID: 38424436 DOI: 10.2174/0115680266286460240220073334] [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: 12/09/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Liposomes, spherical particles with phospholipid double layers, have been extensively studied over the years as a means of drug administration. Conventional manufacturing techniques like thin-film hydration and extrusion have limitations in controlling liposome size and distribution. Microfluidics enables superior tuning of parameters during the self-assembly of liposomes, producing uniform populations. This review summarizes microfluidic methods for engineering liposomes, including hydrodynamic flow focusing, jetting, micro mixing, and double emulsions. The precise control over size and lamellarity afforded by microfluidics has advantages for cancer therapy. Liposomes created through microfluidics and designed to encapsulate chemotherapy drugs have exhibited several advantageous properties in cancer treatment. They showcase enhanced permeability and retention effects, allowing them to accumulate specifically in tumor tissues passively. This passive targeting of tumors results in improved drug delivery and efficacy while reducing systemic toxicity. Promising results have been observed in pancreatic, lung, breast, and ovarian cancer models, making them a potential breakthrough in cancer therapy. Surface-modified liposomes, like antibodies or carbohydrates, also achieve active targeting. Overall, microfluidic fabrication improves reproducibility and scalability compared to traditional methods while maintaining drug loading and biological efficacy. Microfluidics-engineered liposomal formulations hold significant potential to overcome challenges in nanomedicine-based cancer treatment.
Collapse
Affiliation(s)
- Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology, P.O. Box 16846-13114, Tehran, Iran
| | - Kave Mohammad-Jafari
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology, P.O. Box 16846-13114, Tehran, Iran
| |
Collapse
|