1
|
Adriaenssens N, Wuyts SCM, Steurbaut S, De Sutter PJ, Vermeulen A, de Haar-Holleman A, Beckwée D, Provyn S, Vande Casteele S, Zhou J, Lanckmans K, Van Bocxlaer J, De Nys L. Synergy of Body Composition, Exercise Oncology, and Pharmacokinetics: A Narrative Review of Personalizing Paclitaxel Treatment for Breast Cancer. Cancers (Basel) 2025; 17:1271. [PMID: 40282447 PMCID: PMC12025660 DOI: 10.3390/cancers17081271] [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: 03/03/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Paclitaxel is a type of small molecule chemotherapy widely used for breast cancer, but its clinical efficacy is often hindered by dose-limiting toxicities such as chemotherapy-induced peripheral neuropathy and neutropenia. Traditional dosing based on body surface area does not account for variations in body composition, which may influence paclitaxel metabolism, toxicity, and treatment outcomes. This review explores the interplay between body composition, physical activity, and paclitaxel pharmacokinetics, emphasizing the potential for personalized dosing strategies. METHODS A comprehensive narrative review was conducted by analyzing the literature on body composition, small molecule chemotherapy-related toxicities, pharmacokinetics, and exercise oncology. Studies examining the role of skeletal muscle mass, adipose tissue, and physical activity in modulating paclitaxel metabolism and side effects were included. RESULTS Evidence suggests that patients with low skeletal muscle mass are at a higher risk of paclitaxel-induced toxicities due to altered drug distribution and clearance. Sarcopenic obesity, characterized by low muscle and high-fat levels, further exacerbates these risks. Exercise, particularly resistance and aerobic training, has been shown to improve muscle mass, mitigate toxicities, and enhance chemotherapy tolerance. However, the precise mechanisms by which exercise influences paclitaxel pharmacokinetics remain underexplored. CONCLUSIONS Personalized chemotherapy dosing, considering body composition and physical activity, may optimize paclitaxel treatment outcomes. Future research should focus on integrating exercise interventions into oncology care and refining dosing models that account for interindividual differences in drug metabolism. These advancements could improve treatment efficacy while minimizing toxicities in breast cancer patients.
Collapse
Affiliation(s)
- Nele Adriaenssens
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Stephanie C. M. Wuyts
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium (S.S.)
- Research Centre for Digital Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Stephane Steurbaut
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium (S.S.)
- Vitality Research Group, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Pieter-Jan De Sutter
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - Amy de Haar-Holleman
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Translational Oncology Research Center, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - David Beckwée
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
| | - Steven Provyn
- Human Physiology and Sports Physiotherapy, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium
| | - Sofie Vande Casteele
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - Jinyu Zhou
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
| | - Katrien Lanckmans
- Clinical Biology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium;
| | - Jan Van Bocxlaer
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - Len De Nys
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
2
|
Xu LB, Smith ER, Koutouratsas V, Chen ZS, Xu XX. The Persistent Power of the Taxane/Platin Chemotherapy. Cancers (Basel) 2025; 17:1208. [PMID: 40227809 PMCID: PMC11987835 DOI: 10.3390/cancers17071208] [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: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
The cancer chemotherapy regimen of a taxane and platinum combination was developed more than forty years ago, yet remains the cornerstone of treatment for several major cancer types today. Although many new agents targeting cancer genes and pathways have been developed and evaluated, none have been sufficient to replace the long-established taxane/platinum combination. This leads us to ponder why, after four decades of colossal efforts, multiple discoveries, and tremendous advances in understanding gene mutations and biology, the development of conceptually superior targeted therapies has not yet achieved overwhelming success in replacing cytotoxic chemotherapy. The concept of targeted therapy is based on the idea that blocking the altered pathway(s) crucial for cancer development (and maintenance), the disturbance in cellular signaling, metabolism, and functions will make the targeted cancer cells unfit and trigger programmed cell death in cancer cells, but without the significant side effects that limit chemotherapy. We propose that the lack of anticipated triumphs of targeted therapy stems from the desensitization of programmed cell death pathways during neoplastic transformation and malignant progression of cancer cells. This renders the targeting drugs largely ineffective at killing cancer cells and mostly insufficient in clinical implements. Recent advances in understanding suggest that, in contrast to targeted therapies, taxanes and platinum agents kill cancer cells by physical rupturing nuclear membranes rather than triggering apoptosis, making their effect independent of the intrinsic cellular programmed cell death mechanism. This new recognition of the non-programmed cell death mechanism in the success of chemotherapeutic agents, such as taxanes and platinum, may inspire oncologists and cancer researchers to focus their efforts more productively on developing effective non-programmed cell death cancer therapies to replace or significantly improve the application of the current standard taxane/platinum regimens.
Collapse
Affiliation(s)
- Lucy B. Xu
- Department of Biology, University of Miami, Miami, FL 33136, USA;
| | - Elizabeth R. Smith
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Vasili Koutouratsas
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (V.K.)
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (V.K.)
| | - Xiang-Xi Xu
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
3
|
Lal S, Snape TJ. Tubulin targeting agents and their implications in non-cancer disease management. Drug Discov Today 2025; 30:104338. [PMID: 40118444 DOI: 10.1016/j.drudis.2025.104338] [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: 07/26/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Microtubules act as molecular 'tracks' for the intracellular transport of accessory proteins, enabling them to assemble into various larger structures, such as spindle fibres formed during the cell cycle. Microtubules provide an organisational framework for the healthy functioning of various cellular processes that work through the process of dynamic instability, driven by the hydrolysis of GTP. In this role, tubulin proteins undergo various modifications, and in doing so modulate various healthy or pathogenic physiological processes within cells. In this review, we provide a detailed update of small molecule chemical agents that interact with tubulin, along with their implications, specifically in non-cancer disease management.
Collapse
Affiliation(s)
- Samridhi Lal
- Amity Institute of Pharmacy, Amity University, Gurugram 122413 Haryana, India.
| | - Timothy J Snape
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| |
Collapse
|
4
|
Li Y, Wang S, Guan X, Zhang S, Yang R, Li M, Liu T, Zhao J, Lin L, Liu X, Song Y, Deng Y. Sialic acid-modified nanomedicines modulate neutrophils for dual therapy of cancer and sepsis: addressing neglected sepsis comorbidities during cancer treatment. Int J Pharm 2025; 671:125211. [PMID: 39800002 DOI: 10.1016/j.ijpharm.2025.125211] [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: 10/12/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Advanced cancer patients face a high risk of sepsis due to immune suppression and infection susceptibility. To tackle this challenge, we developed an innovative animal model that simulates the clinical scenario of late-stage cancer complicated by sepsis and designed a sialic acid (SA)-modified paclitaxel (PTX) liposome (PTX-SAL). This formulation specifically targets overactivated peripheral blood neutrophils (PBNs) by binding to L-selectin on their surface. It effectively eliminates hyperactive neutrophils and blocks their migration, thus reducing infiltration into tumor and inflammation sites. In sepsis and melanoma mouse models, PTX-SAL demonstrated superior therapeutic efficacy and a favorable safety profile. Notably, in the late-stage tumor model with sepsis, PTX-SAL significantly improved survival rates, with a 72-hour survival rate of 66.7%. In stark contrast, the PTX solution (PTX-S) group exhibited accelerated mortality, with all animals succumbing within 24 h, highlighting the detrimental effects of PTX-S's non-selective cytotoxicity on immune cells. These findings underscore the superior long-term safety and therapeutic advantage of nanomedicines like PTX-SAL over conventional drug formulations. In summary, SA-modified nanomedicines offer a dual benefit by targeting and eliminating inflammatory neutrophils, addressing both tumor progression and sepsis, and significantly reducing mortality in preclinical models. This innovative strategy fills a critical gap in the treatment of advanced cancer complicated by sepsis.
Collapse
Affiliation(s)
- Yantong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xinying Guan
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Senyu Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Rui Yang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Mingze Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tingyan Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jingyi Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Lin Lin
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
5
|
Perużyńska M, Birger R, Kłos P, Kwiecień H, Struk Ł, Sośnicki JG, Lafanechère L, Droździk M. The Co-Administration of Paclitaxel with Novel Pyridine and Benzofuran Derivatives that Inhibit Tubulin Polymerisation: A Promising Anticancer Strategy. Pharmaceutics 2025; 17:223. [PMID: 40006590 PMCID: PMC11859455 DOI: 10.3390/pharmaceutics17020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Paclitaxel (PTX), a crucial microtubule-stabilising agent in cancer treatment, is limited by its adverse effects and hydrophobic nature, which necessitate the use of toxic solvents. This study proposes a novel approach combining PTX with new microtubule-destabilising compounds at low, safe doses that are ineffective when used individually. Objective: The aim was to evaluate the therapeutic efficacy of combining PTX with previously described pyridine (S1, S22) and benzofuran derivatives (13b, 14), which have demonstrated promising anticancer properties by inhibiting microtubule polymerisation. Methods: The PrestoBlue assay was used to determine the optimal concentrations of each compound, enabling synergistic interactions with a low dose of PTX in HeLa cervical cancer cells. The combined effects of the compounds and PTX on apoptosis, cell cycle distribution, and mitotic spindle formation were then evaluated. Results: The results showed that compounds 13b (1 µM), 14 (0.1 µM), S1 (2 µM), and S22 (2 µM) enhanced the proapoptotic and antimitotic effects of 1 nM PTX, which was ineffective alone. Notably, live-cell imaging revealed that the concurrent use of S1 and PTX produced effects similar to those of a higher PTX concentration (5 nM). Conclusions: These findings suggest that these compounds enhance the anticancer efficacy of low-dose PTX, potentially paving the way for more effective and safer cancer therapies.
Collapse
Affiliation(s)
- Magdalena Perużyńska
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (R.B.); (M.D.)
| | - Radosław Birger
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (R.B.); (M.D.)
| | - Patrycja Kłos
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Halina Kwiecień
- Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów 42, 71-065 Szczecin, Poland;
| | - Łukasz Struk
- Department of Organic and Physical Chemistry, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów 42, 71-065 Szczecin, Poland; (Ł.S.); (J.G.S.)
| | - Jacek G. Sośnicki
- Department of Organic and Physical Chemistry, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów 42, 71-065 Szczecin, Poland; (Ł.S.); (J.G.S.)
| | - Laurence Lafanechère
- Institute for Advanced Biosciences, University Grenoble Alpes, CNRS UMR 5309, INSERM U1209, F-38700 Grenoble, France;
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (R.B.); (M.D.)
| |
Collapse
|
6
|
Yuan M, Han N, Shu L, Yan L, Tang H. Case report: Multi-organ injuries induced by tislelizumab. Front Immunol 2025; 16:1508293. [PMID: 39967654 PMCID: PMC11832704 DOI: 10.3389/fimmu.2025.1508293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) often develops immune-related adverse events (irAEs). However, irAEs-induced multi-organ injuries remain a rare event. We herein report a case of multi-organ injuries induced by tislelizumab in a lung squamous cell carcinoma (LUSC) patient. A 68-year-old man had undergone neoadjuvant chemotherapy with paclitaxel, carboplatin, and tislelizumab. He presented with a 1-month history of nausea and poor appetite after the second dose of therapy. During investigations, rhabdomyolysis, liver, kidney, and thyroid damage were detected. After multi-disciplinary consultation, multi-organ injuries related to ICIs (striated muscle, liver, kidney, and thyroid) were considered to result from cumulated irAEs induced by tislelizumab. The patient was treated with levothyroxine, methylprednisolone, intravenous immunoglobulins, and continuous renal replacement therapy. After treatment, the patient recovered and was discharged from the hospital. The patient presented with multiple organ damage, not single immunity treatment adverse reactions, relatively rare. In clinical work, irAEs are likely not a single-system organ disorder and many kinds of attention need to be combined with the risk of multi-system damage.
Collapse
Affiliation(s)
- Man Yuan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Ning Han
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Li Shu
- Department of Infectious Diseases, Suining Central Hospital, Suining, China
| | - Libo Yan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Ayhan İ, Turkmen NB, Taslidere A, Aydin M, Ciftci O. Protective Effect of Nerolidol on Paclitaxel-Induced Reproductive Toxicity in Rats: Oxidative Stress and Inflammation. Basic Clin Pharmacol Toxicol 2025; 136:e14126. [PMID: 39815991 DOI: 10.1111/bcpt.14126] [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: 10/22/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025]
Abstract
Paclitaxel (PAC), derived from Taxus brevifolia, is used to treat solid tumours but causes reproductive toxicity due to oxidative stress, affecting sperm quality and testicular tissue. Nerolidol (NRL), an antioxidant sesquiterpene alcohol, has not been studied for its potential to reduce PAC-induced reproductive damage. This study investigates NRL's ability to mitigate PAC-induced reproductive toxicity in rats. Forty healthy adult male Spraque Dawley rats were randomly divided into four equal groups (Control, PAC, NRL, PAC + NRL). PAC was given intraperitoneally at a dose of 2 mg/kg once a week for 4 weeks. NRL was given orally at a dose of 100 mg/kg/day for 4 weeks. Control group received PAC and NRL vehicles. After 4 weeks, testis tissue samples were collected, and parameters, including oxidants, antioxidants, sperm motility, density, abnormal spermatozoon ratios and cytokines, were measured. PAC administration increased oxidant levels and decreased antioxidant enzyme activities. Nerolidol mitigated these alterations significantly. Similarly, PAC elevated IL-1, IL-6, TNF-α levels and lowered IL-10 levels, these effects attenuated by nerolidol in the PAC + NRL group. In conclusion, it was determined that PAC induces reproductive toxicity through oxidative stress, and NRL demonstrates potential in ameliorating these effects through its antioxidant activity.
Collapse
Affiliation(s)
- İdris Ayhan
- Department of Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Nese Basak Turkmen
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Asli Taslidere
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Muhterem Aydin
- Department of Obstetric and Gynecology, Faculty of Veterinary Medicine, Firat University, Elazıg, Turkey
| | - Osman Ciftci
- Department of Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
8
|
Nie L, Ma J, Yu Y, Tao Y, Song Z, Li J. Exosomes as carriers to stimulate an anti-cancer immune response in immunotherapy and as predictive markers. Biochem Pharmacol 2025; 232:116699. [PMID: 39647605 DOI: 10.1016/j.bcp.2024.116699] [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: 09/29/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
During this era of rapid advancements in cancer immunotherapy, the application of cell-released small vesicles that activate the immune system is of considerable interest. Exosomes are cell-derived nanovesicles that show great promise for the immunological treatment of cancer because of their immunogenicity and molecular transfer capacity. Recent technological advancements have enabled the identification of functional functions that exosome cargoes perform in controlling immune responses. Exosomes are originated specifically from immune cells and tumor cells and they show unique composition patterns directly related to the immunotherapy against cancer. Exosomes can also deliver their cargo to particular cells, which can affect the phenotypic and immune-regulatory functions of those cells. Exosomes can influence the course of cancer and have therapeutic benefits by taking part in several cellular processes; as a result, they have the dual properties of activating and restraining cancer. Exosomes have tremendous potential for cancer immunotherapy; they may develop into the most powerful cancer vaccines and carriers of targeted antigens and drugs. Comprehending the potential applications of exosomes in immune therapy is significant for regulating cancer progression. This review offers an analysis of the function of exosomes in immunotherapy, specifically as carriers that function as diagnostic indicators for immunological activation and trigger an anti-cancer immune response. Moreover, it summarizes the fundamental mechanism and possible therapeutic applications of exosome-based immunotherapy for human cancer.
Collapse
Affiliation(s)
- Lili Nie
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, China
| | - Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
9
|
Zhong W, Xu Y, Wang Z, Wang X, Li Y, Liu J, Zhao C, Shi X, He Z, Sun B, Tian C. Dual role of triglyceride structures facilitates anti-tumor drug delivery: Both as a self-assembling module and a responsive module. J Colloid Interface Sci 2025; 678:24-34. [PMID: 39277950 DOI: 10.1016/j.jcis.2024.09.096] [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: 04/22/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Small molecule prodrugs self-assembled nano-delivery systems with tumor responsive linkages are emerging as an effective platform. However, the heterogeneity of tumor microenvironment may limit the anti-tumor effect of prodrug nanomedicines with a single response module. Here, we chose disulfide bond as the response module and branched chain alcohol as the self-assembly modification module to construct a single-responsive prodrug. We also constructed a double-responsive paclitaxel prodrug combining triglyceride and disulfide bond, taking into account of the highly expressed lipase and glutathione levels in tumor cells. The results showed that the anti-tumor effect of single-responsive branched chain alcohol modified prodrug nanoparticles was inferior to triglyceride prodrug nanoparticles with dual response modules. The triglyceride structure can not only serve as a self-assembly modification module, but also serve as a response module for intelligent drug release in tumor. Such dual roles will facilitate the efficient delivery of small molecule self-assembled prodrugs to tumor sites.
Collapse
Affiliation(s)
- Wenxin Zhong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China
| | - Yalin Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zixuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiyan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yaqi Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jinrui Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Can Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China.
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China.
| |
Collapse
|
10
|
Naseri B, Farsad-Akhtar N, Mardi A, Baghbani E, Bornedeli S, Asadi M, Shanehbandi D. lncRNA PVT1 silencing inhibits gastric cancer cells' progression via enhancing chemosensitivity to paclitaxel. Gene 2025; 932:148900. [PMID: 39209180 DOI: 10.1016/j.gene.2024.148900] [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: 05/11/2024] [Revised: 08/18/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide because of its high morbidity and the absence of effective therapies. Even though paclitaxel is a powerful anticancer chemotherapy drug, recent studies have indicated its ineffectiveness against GC cells. Long non-coding RNA (lncRNA) PVT1 has a high expression in GC cells and increases the progression of tumors via inducing drug resistance. In the present study, the effects of the siRNA-mediated lncRNA PVT1 gene silencing along with paclitaxel treatment on the rate of apoptosis, growth, and migration of AGS GC cells were investigated. AGS cells were cultured and then transfected with siRNA PVT1 using electroporation. The MTT test was used to examine the effect of treatments on the viability of cultured cells. Furthermore, the flow cytometry method was used to evaluate the impact of treatments on the cell cycle process and apoptosis induction in GC cells. Finally, the mRNA expression of target genes was assessed using the qRT-PCR method. The results showed that lncRNA PVT1 gene suppression, along with paclitaxel treatment, reduces the viability of cancer cells and significantly increases the apoptosis rate of cancer cells and the number of cells arrested in the G2/M phase compared to the control group. Based on the results of qRT-PCR, combined treatment significantly decreased the expression of MMP3, MMP9, MDR1, MRP1, Bcl-2, k-Ras, and c-Myc genes and increased the expression of the Bax gene compared to the control group. The results of our study showed that lncRNA PVT1 gene targeting, together with paclitaxel treatment, induces apoptosis, inhibits growth, alleviates drug resistance, and reduces the migratory capability of GC cells. Therefore, there is a need for further investigations to evaluate the feasibility and effectiveness of this approach in vivo in animal models.
Collapse
Affiliation(s)
- Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Nader Farsad-Akhtar
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soghra Bornedeli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Alum EU, Tufail T, Uti DE, Aja PM, Offor CE, Ibiam UA, Ukaidi CUA, Alum BN. Utilizing Indigenous Flora in East Africa for Breast Cancer Treatment: An Overview. Anticancer Agents Med Chem 2025; 25:99-113. [PMID: 39297456 DOI: 10.2174/0118715206338557240909081833] [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: 07/16/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 02/18/2025]
Abstract
BACKGROUND Breast cancer is a significant global health challenge, contributing substantially to cancer- related deaths. Conventional treatment methods, including hormone therapy, chemotherapy, surgical interventions, and radiation, have long been utilized. However, these traditional treatments are often associated with serious side effects and drug resistance, limiting their efficacy. AIM This review aims to explore the potential of medicinal plants used in breast cancer management in East Africa, focusing on their bioactive compounds and anticancer properties. METHODS A comprehensive literature search was conducted to examine the effectiveness of medicinal plants in treating breast cancer across Kenya, Ethiopia, Uganda, Tanzania, and Rwanda. Relevant studies published between 2003 and 2023 were identified using keywords related to breast cancer and medicinal plants. The search was performed across multiple databases, including Google Scholar, PubMed, Scopus, Web of Science Core Collection, and Science Direct. RESULTS Numerous natural compounds found in East African medicinal plants including Cymbopogon citratus (Lemongrass,) Tabebuia avellanedae, Prunus africana (African Cherry), Euclea divinorum, Berberis holstii, Withania somnifera (Ashwagandha, Curcuma longa (Turmeric), Garcinia mangostana (Mangosteen, Vitis vinifera (Grapevine), Eugenia jambolana (Java Plum), Moringa oleifera (Drumstick Tree), Camellia sinensis (Tea), Glycine max (Soybean), Catharanthus roseus, Madagascar Periwinkle), Rhus vulgaris (Wild Currant) exhibit significant anticancer properties. These compounds have demonstrated the ability to reduce breast cancer aggressiveness, inhibit cancer cell proliferation, and modulate cancer-related pathways. Current research focuses on these natural and dietary compounds to develop more effective strategies for treating breast cancer. CONCLUSION The findings suggested that East African medicinal plants hold promise as complementary treatments for breast cancer, offering potential benefits such as affordability, cultural appropriateness, and sustainability. Further research into these plants and their bioactive compounds could revolutionize breast cancer treatment, improving survival rates and addressing the rising incidence of breast cancer-related fatalities. Other: The review underscores the importance of continued research, conservation, and the integration of ancient healing methods to fully harness the potential of East African flora in breast cancer management.
Collapse
Affiliation(s)
- Esther Ugo Alum
- Department of Research and Publications, Kampala International University, Kampala, P. O. Box 20000, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria
| | - Tabussam Tufail
- School of Food and Biological, Engineering Jiangsu, University Zhenjiang Kampala, China
- University Institute of Diet and Nutritional Sciences, The University of Lahore, Lahore, Pakistan
| | - Daniel Ejim Uti
- Department of Research and Publications, Kampala International University, Kampala, P. O. Box 20000, Uganda
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State, Nigeria
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria
- Department of Biochemistry, Kampala International University, Western Campus, Kampala, Uganda
| | - Christian Emeka Offor
- Department of Biochemistry, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria
| | - Udu Ama Ibiam
- Department of Biochemistry, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria
- Department of Biochemistry, College of Science, Evangel University Akaeze, Abakaliki, Ebonyi State, Nigeria
| | - Chris U A Ukaidi
- College of Economics and Management, Kampala International University, Kampala, Uganda
| | - Benedict Nnachi Alum
- Department of Research and Publications, Kampala International University, Kampala, P. O. Box 20000, Uganda
| |
Collapse
|
12
|
De Nys L, Barzegar-Fallah A, Lanckmans K, Steurbaut S, Beckwée D, de Haar-Holleman A, Provyn S, Gasthuys E, Vande Casteele S, De Sutter PJ, Vermeulen A, Van Bocxlaer J, Wuyts SCM, Adriaenssens N. Dose-Limiting Toxicities of Paclitaxel in Breast Cancer Patients: Studying Interactions Between Pharmacokinetics, Physical Activity, and Body Composition-A Protocol for an Observational Cohort Study. Cancers (Basel) 2024; 17:50. [PMID: 39796679 PMCID: PMC11719000 DOI: 10.3390/cancers17010050] [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: 11/30/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Paclitaxel (PTX), a commonly used chemotherapy for breast cancer (BC), is associated with dose-limiting toxicities (DLTs) such as peripheral neuropathy and neutropenia. These toxicities frequently lead to dose reductions, treatment delays, or therapy discontinuation, negatively affecting patients' quality of life and clinical outcomes. Current dosing strategies based on body surface area (BSA) fail to account for individual variations in body composition (skeletal muscle mass (SMM) and adipose tissue (AT) mass) and physical activity (PA), which can influence drug metabolism and toxicity. This study aims to explore the relationships between PTX pharmacokinetics, body composition, and PA to predict DLTs. Methods: This single-group observational cohort study will recruit 40 female BC patients undergoing PTX treatment. Data collection will include plasma PTX concentrations, body composition assessments (using dual X-ray absorptiometry and bioelectrical impedance analysis), PA measurements (via accelerometers), and questionnaires to assess BC-related health-related quality of life, chemotherapy-induced peripheral neuropathy, and neutropenia during the PTX schedule using validated questionnaires. Dose-limiting toxicities will be graded according to the Common Terminology Criteria for Adverse Events v5.0 (grade 3 or higher). This protocol is designed to develop a population-based PK-PD model that predicts the occurrence of chemotherapy-induced peripheral neuropathy and neutropenia in women with stage II or III BC undergoing PTX therapy, focusing on explanatory outcomes related to SMM, AT mass, and PA.
Collapse
Affiliation(s)
- Len De Nys
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Jette, Belgium; (L.D.N.)
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Jette, Belgium
| | - Anita Barzegar-Fallah
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Jette, Belgium; (L.D.N.)
| | - Katrien Lanckmans
- Clinical Biology Department, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Jette, Belgium
| | - Stephane Steurbaut
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Jette, Belgium; (S.S.)
- Vitality Research Group, Vrije Universiteit Brussel (VUB), 1090 Jette, Belgium
| | - David Beckwée
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Jette, Belgium; (L.D.N.)
| | - Amy de Haar-Holleman
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Jette, Belgium
- Translational Oncology Research Center, Vrije Universiteit Brusse (VUB), 1050 Brussels, Belgium
| | - Steven Provyn
- Human Physiology and Sports Physiotherapy (MFYS), Vrije Universiteit Brussel (VUB), 1090 Jette, Belgium
| | - Elke Gasthuys
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Gent, Belgium; (E.G.)
| | - Sofie Vande Casteele
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Gent, Belgium; (E.G.)
| | - Pieter-Jan De Sutter
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Gent, Belgium; (E.G.)
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Gent, Belgium; (E.G.)
| | - Jan Van Bocxlaer
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Gent, Belgium; (E.G.)
| | - Stephanie C. M. Wuyts
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Jette, Belgium; (S.S.)
- Research Centre for Digital Medicine, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Nele Adriaenssens
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Jette, Belgium; (L.D.N.)
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Jette, Belgium
| |
Collapse
|
13
|
Dai Y, Ruan T, Yang W, Liu S, Chen J, Fang Y, Li Q. Efficacy and Safety of Paclitaxel-Based PD-1/PD-L1 Immunotherapies for Triple-Negative Breast Cancer: A Systematic Review and Network Meta-Analysis. Clin Med Insights Oncol 2024; 18:11795549241308072. [PMID: 39734512 PMCID: PMC11672372 DOI: 10.1177/11795549241308072] [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: 08/01/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Background Triple negative breast cancer (TNBC) is a deadly subtype of breast cancer with limited treatment options. Currently, programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors have become the first choice for breast cancer immunotherapies. Despite paclitaxel being considered a cornerstone drug in breast cancer treatment, the effectiveness, safety, and optimal drug selection for its combination with PD-1/PD-L1 inhibitors remain uncertain. Methods We conducted a systematic review and network meta-analysis, performing a comprehensive literature search across PubMed, Embase, and the Cochrane Library from the inception of each database through May 18, 2024. Selected trials were those that assessed the efficacy and safety of paclitaxel-based PD-1/PD-L1 therapies for the treatment of TNBC. The primary endpoint assessed was overall survival (OS), while secondary outcomes included progression-free survival (PFS), adverse events (AEs), overall response rate (ORR), and Pathological complete response (pCR). This study is registered in PROSPERO under registration number CRD42023429651. Results A total of 8 RCTs meeting our eligibility criteria were included, involving 4626 patients who received either Paclitaxel (Paclitaxel-placebo/chemotherapy) or a combination of durvalumab, pembrolizumab, atezolizumab, toripalimab with paclitaxel. The pooled results demonstrated that Durvalumab combined with Paclitaxel significantly reduced the hazard ratio for OS (surface under the cumulative ranking [SUCRA]: 91.05%) and PFS compared with Paclitaxel alone (SUCRA: 83.52%). Additionally, Durvalumab plus Paclitaxel significantly improved the ORR compared with Paclitaxel (odds ratio [OR]: 2.30; 95% credible interval [CrI]: 1.10-5.20). For safety outcomes, Atezolizumab plus Paclitaxel showed a favorable profile in AEs, with no significant differences observed between groups. In the pCR study, Pembrolizumab plus Paclitaxel was the most effective treatment option (SUCRA: 81.85%). Conclusions When combined with paclitaxel, PD-1/PD-L1 inhibitors exhibit a favorable survival benefit. The combination of Durvalumab and paclitaxel represents the optimal treatment option. In the future, attention should be paid to the TNBC subtypes and drug dosage, as these factors may help to design personalized TNBC treatment programs.
Collapse
Affiliation(s)
| | - Tianyin Ruan
- Institute of Hepatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenhui Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shan Liu
- Center of Clinical Evaluation and Analysis, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jiahao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingying Fang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiushuang Li
- Center of Clinical Evaluation and Analysis, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
14
|
Bauri S, Yadav PK, Choudhury AM, Maiti P. L-Cystine-Based Polyurethane as a Drug-Delivery Vehicle in Targeted Cancer Therapy and Biomedical Applications. ACS APPLIED BIO MATERIALS 2024; 7:8671-8684. [PMID: 39591387 DOI: 10.1021/acsabm.4c01479] [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: 11/28/2024]
Abstract
Controlled and sustained drug release is a critical aspect of drug-delivery systems (DDSs) that can be used in chemotherapy while ensuring therapy effectiveness and biosafety. Hence, polyurethane (PU) is modified using a biomolecule Cystine (CYS) for protracted drug release, aiming to enhance cancer treatment efficacy while minimizing adverse side effects in tumor patients. To confirm the formation of a polymer structure, characterization techniques such as NMR and FTIR are used, and the morphology is determined using SEM. Biocompatibility of the synthesized polymers is evaluated through cellular assessments, including MTT assay, cell adhesion, and antibacterial assay along with drug release using an anticancer drug, Paclitaxel (PTX). Notably, the incorporation of PTX in the polymer matrix results in minimal mortality (85% viable cells) rates in healthy cells (3T3), in contrast to a 56% mortality rate observed with the pure drug. While PTX shows a burst release and kills cancer cells only for the first 24 h, PU loaded with the drug shows sustained release and kills the cancer cells for 3 days. This vehicle selectively kills 59% of SiHA cells after a consecutive study of 3 days, which highlights the potential of this newly designed vehicle for effective drug delivery, particularly in anticancer treatments. Moreover, cystine's antibacterial property adds up with PU; hence, PU shows antibacterial activity against Staphylococcus aureus (MIC, 20 μg/mL) and also acts as a reductive oxygen species scavenger. Therefore, modifying PU with CYS has shown sustained release of PTX along with a selective effect on cells, underscoring its significance as a superior delivery agent and supported by a shred of convincing evidence.
Collapse
Affiliation(s)
- Sudepta Bauri
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Pravesh Kumar Yadav
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Avishek Mallick Choudhury
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Pralay Maiti
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| |
Collapse
|
15
|
Mikesell AR, Isaeva E, Schulte ML, Menzel AD, Sriram A, Prahl MM, Shin SM, Sadler KE, Yu H, Stucky CL. Increased keratinocyte activity and PIEZO1 signaling contribute to paclitaxel-induced mechanical hypersensitivity. Sci Transl Med 2024; 16:eadn5629. [PMID: 39661703 DOI: 10.1126/scitranslmed.adn5629] [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: 12/15/2023] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Recent work demonstrates that epidermal keratinocytes are critical for normal touch sensation. However, it is unknown whether keratinocytes contribute to touch-evoked pain and hypersensitivity after tissue injury. Here, we used a mouse model of paclitaxel treatment to determine the extent to which keratinocyte activity contributes to the severe neuropathic pain that accompanies chemotherapy. We found that keratinocyte inhibition by either optogenetic or chemogenetic methods largely alleviated paclitaxel-induced mechanical hypersensitivity across acute and persistent time points from 2 days through 3 weeks. Furthermore, we found that paclitaxel exposure sensitized mouse and human keratinocytes to mechanical stimulation and enhanced currents of PIEZO1, a mechanosensitive channel highly expressed in keratinocytes. Deletion of PIEZO1 from keratinocytes alleviated paclitaxel-induced mechanical hypersensitivity in mice. These findings suggest that nonneuronal cutaneous cells contribute substantially to neuropathic pain and pave the way for the development of new pain relief strategies that target epidermal keratinocytes and PIEZO1.
Collapse
Affiliation(s)
- Alexander R Mikesell
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elena Isaeva
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Anthony D Menzel
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anvitha Sriram
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Megan M Prahl
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Katelyn E Sadler
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
16
|
Orleanska J, Bik E, Baranska M, Majzner K. Mechanisms of mitotic inhibition in human aorta endothelial cells: Molecular and morphological in vitro spectroscopic studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 322:124623. [PMID: 39002470 DOI: 10.1016/j.saa.2024.124623] [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: 01/12/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/15/2024]
Abstract
Mitotic inhibitors are drugs commonly used in chemotherapy, but their nonspecific and indiscriminate distribution throughout the body after intravenous administration can lead to serious side effects, particularly on the cardiovascular system. In this context, our investigation into the mechanism of the cytotoxic effects on endothelial cells of mitotic inhibitors widely used in cancer treatment, such as paclitaxel (also known as Taxol) and Vinca alkaloids, holds significant practical implications. Understanding these mechanisms can lead to more targeted and less harmful cancer treatments. Human aorta endothelial cells (HAECs) were incubated with selected mitotic inhibitors in a wide range of concentrations close to those in human plasma during anticancer therapy. The analysis of single cells imaged by Raman spectroscopy allowed for visualization of the nuclear, cytoplasmic, and perinuclear areas to assess biochemical changes induced by the drug's action. The results showed significant changes in the morphology and molecular composition of the nucleus. Moreover, an effect of a given drug on the cytoplasm was observed, which can be related to its mechanism of action (MoA). Raman data supported by fluorescence microscopy measurements identified unique changes in DNA form and proteins and revealed drug-induced inflammation of endothelial cells. The primary goal of mitotic inhibitors is based on the impairment of tubulin formation and the inhibition of the mitosis process. While all three drugs affect microtubules and disrupt cell division, they do so through different MoA, i.e., Vinca alkaloids inhibit microtubule formation, whereas paclitaxel stabilizes microtubules. To sum up, the work shows how a specific drug can interact with endothelial cells.
Collapse
Affiliation(s)
- Jagoda Orleanska
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland; Jagiellonian University, Doctoral School of Exact and Natural Sciences, Lojasiewicza 11, 30-348 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Ewelina Bik
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland; Academic Centre for Materials and Nanotechnology, AGH University of Krakow, Mickiewicza Av. 30, 30-059 Krakow, Poland
| | - Malgorzata Baranska
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Katarzyna Majzner
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
17
|
Bhullar AS, Jin K, Shi H, Jones A, Hironaka D, Xiong G, Xu R, Guo P, Binzel DW, Shu D. Engineered extracellular vesicles for combinatorial TNBC therapy: SR-SIM-guided design achieves substantial drug dosage reduction. Mol Ther 2024; 32:4467-4481. [PMID: 39369270 PMCID: PMC11638871 DOI: 10.1016/j.ymthe.2024.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/22/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that has no therapeutic targets, relies on chemotherapeutics for treatment, and is in dire need of novel therapeutic approaches for improved patient outcomes. Extracellular vesicles (EVs) serve as intercellular communicators and have been proposed as ideal drug delivery vehicles. Here, EVs were engineered with RNA nanotechnology to develop TNBC tumor inhibitors. Using super resolved-structured illumination microscopy, EVs were optimized for precise Survivin small interfering RNA (siRNA) conjugated to chemotherapeutics loading and CD44 aptamer ligand decoration, thereby enhancing specificity toward TNBC cells. Conventional treatments typically employ chemotherapy drugs gemcitabine (GEM) and paclitaxel (PTX) at dosages on the order of mg/kg respectively, per injection (intravenous) in mice. In contrast, engineered EVs encapsulating these drugs saw functional tumor growth inhibition at significantly reduced concentrations: 2.2 μg/kg for GEM or 5.6 μg/kg for PTX, in combination with 21.5 μg/kg survivin-siRNA in mice. The result is a substantial decrease in the chemotherapeutic dose required, by orders of magnitude, compared with standard regimens. In vivo and in vitro evaluations in a TNBC orthotopic xenograft mouse model demonstrated the efficacy of this decreased dosage strategy, indicating the potential for decreased chemotherapy-associated toxicity.
Collapse
Affiliation(s)
- Abhjeet S Bhullar
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy and Comprehensive Cancer Center. The Ohio State University, Columbus, OH 43210, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Kai Jin
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy and Comprehensive Cancer Center. The Ohio State University, Columbus, OH 43210, USA
| | - Haizhu Shi
- Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Austen Jones
- Department of Veterinary Biosciences, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Dalton Hironaka
- Department of Veterinary Biosciences, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Gaofeng Xiong
- Department of Veterinary Biosciences, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Ren Xu
- Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy and Comprehensive Cancer Center. The Ohio State University, Columbus, OH 43210, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel W Binzel
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy and Comprehensive Cancer Center. The Ohio State University, Columbus, OH 43210, USA.
| | - Dan Shu
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy and Comprehensive Cancer Center. The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
18
|
Pfeifer LM, Sensbach J, Pipp F, Werkmann D, Hewitt P. Increasing sustainability and reproducibility of in vitro toxicology applications: serum-free cultivation of HepG2 cells. FRONTIERS IN TOXICOLOGY 2024; 6:1439031. [PMID: 39650261 PMCID: PMC11621109 DOI: 10.3389/ftox.2024.1439031] [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: 05/27/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Fetal Bovine Serum (FBS) is an important ingredient in cell culture media and the current standard for most cells in vitro. However, the use of FBS is controversial for several reasons, including ethical concerns, political, and societal pressure, as well as scientific problems due to the undefined and variable nature of FBS. Nevertheless, scientists hesitate to change the paradigm without solid data de-risking the switch of their assays to alternatives. In this study, HepG2 cells, a human hepatoblastoma cell line commonly used to study drug hepatotoxicity, were adapted to serum-free conditions by using different commercially available media and FBS replacements. After transition to these new culture conditions, the success of adaptation was determined based on cell morphology and growth characteristics. Long-term culturing capacity for each medium was defined as the number of passages HepG2 cells could be cultured without any alterations in morphology or growth behavior. Two media (Advanced DMEM/F12 from ThermoFisher and TCM® Serum Replacement from MP Biomedicals) showed a long-term cultivation capacity comparable to media containing FBS and were selected for further analysis. Both media can be characterized as serum-free, however still contain animal-derived components: bovine serum albumin (both media) and bovine transferrin (only TCM® serum replacement). To assess the functionality of the cells cultivated in either of the two media, HepG2 cells were treated with reference compounds, specifically selected for their known hepatotoxicity characteristics in man. Different toxicological assays focusing on viability, mitochondrial toxicity, oxidative stress, and intracellular drug response were performed. Throughout the different assays, response to reference compounds was comparable, with a slightly higher sensitivity of serum-free cultivated HepG2 cells when assessing viability/cell death and a lower sensitivity towards oxidative stress. Taken together, the two selected media were shown to support growth, morphology, and function of serum-free cultivated HepG2 cells in the early preclinical safety space. Therefore, these results can serve as a starting point to further optimize culture conditions with the goal to remove any remaining animal-derived components.
Collapse
Affiliation(s)
| | - Janike Sensbach
- Early Investigative Toxicology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Frederic Pipp
- Corporate Animal Affairs, Merck KGaA, Darmstadt, Germany
| | - Daniela Werkmann
- Cell Design Lab, Molecular Biology, Merck KGaA, Darmstadt, Germany
| | - Philip Hewitt
- Early Investigative Toxicology, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
19
|
Hwu JR, Panja A, Tsay SC, Huang WC, Lin SY, Yeh CS, Su WC, Yang LX, Shieh DB. β-d-Ribofuranose as a Core with a Phosphodiester Moiety as the Enzyme Recognition Site for Codrug Development. Org Lett 2024; 26:9865-9870. [PMID: 39540692 PMCID: PMC11590094 DOI: 10.1021/acs.orglett.4c03662] [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: 09/30/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
An ideal codrug design should be able to control drug release, offer selectivity during drug delivery, and break down into non-toxic fragments after biodegradation. Our design incorporates d-ribofuranose as the core, with carbamate and carbonate groups as linking joints, a phosphodiester moiety as an enzyme recognition site, and lenalidomide and paclitaxel as the constituent drugs. The codrug synthesis involves seven steps with a 33% overall yield. The target codrug increases its water solubility 685 times versus paclitaxel.
Collapse
Affiliation(s)
- Jih Ru Hwu
- Department
of Chemistry & Frontier Research Center on Fundamental and Applied
Sciences of Matters, National Tsing Hua
University, Hsinchu 300, Taiwan
| | - Avijit Panja
- Department
of Chemistry & Frontier Research Center on Fundamental and Applied
Sciences of Matters, National Tsing Hua
University, Hsinchu 300, Taiwan
| | - Shwu-Chen Tsay
- Department
of Chemistry & Frontier Research Center on Fundamental and Applied
Sciences of Matters, National Tsing Hua
University, Hsinchu 300, Taiwan
| | - Wen-Chieh Huang
- Department
of Chemistry & Frontier Research Center on Fundamental and Applied
Sciences of Matters, National Tsing Hua
University, Hsinchu 300, Taiwan
- Institute
of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 350401, Taiwan
| | - Shu-Yu Lin
- Institute
of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 350401, Taiwan
| | - Chen-Sheng Yeh
- Department
of Chemistry, National Cheng Kung University, Tainan 701, Taiwan
| | - Wu-Chou Su
- Department
of Internal Medicine, National Cheng Kung
University, Tainan 701, Taiwan
| | - Li-Xing Yang
- Department
of Dentistry and Institute of Oral Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Dar-Bin Shieh
- Department
of Dentistry and Institute of Oral Medicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
20
|
Xie Q, Jia Y, Tao J, Bu T, Wang Q, Shen N, Zhang X, Xiao Y, Ye L, Chen Z, Huang H, Li Q, Tang Z. Chemical constituents and biological activities of endophytic fungi from Fagopyrum dibotrys. PeerJ 2024; 12:e18529. [PMID: 39575167 PMCID: PMC11580677 DOI: 10.7717/peerj.18529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024] Open
Abstract
Background Fagopyrum dibotrys is an important wild food and feed germplasm resource. It has high nutritional and medicinal value and is rich in natural products, including flavonoids, phenolic acids, coumarins, and alkaloids. Endophytic fungi in F. dibotrys have emerged as valuable sources of natural products. However, studies on the biological activity and chemical composition of these endophytic fungi remain limited. Methods In this paper, a new method to obtain natural active ingredients by fermentation of endophytic fungi from medicinal plants was proposed. Then the antioxidant and pathogenic activities of the endophytic fungi extracts were determined in vitro. In addition, secondary metabolites produced by endophytic fungi with medicinal activity were analyzed by high performance liquid chromatography-tandem mass spectrometry (LC-MS). Results Among the 95 endophytic fungal strains in F. dibotrys, four strains with high phenol yields were selected by reaction: Alternaria alstroemeriae (J2), Fusarium oxysporum (J15), Colletotrichum karsti (J74), and Colletotrichum boninense (J61). Compared with those of various extracts, the ethyl acetate fractions of A. alstroemeriae (J2), F. oxysporum (J15), and C. boninense (J61) exhibited superior antioxidant and antibacterial properties. The results indicated that the fungal extract was an excellent natural antioxidant and might be a potential antibacterial agent. The DPPH free radical clearance of A. alstroemeriae was 94.96 ± 0.004%. These findings indicated that A. alstroemeriae had strong antioxidant activity. In addition, the extract of A. alstroemeriae had good antibacterial activity against Escherichia coli and Staphylococcus aureus, with MICs of 0.5 and 0.05 mg/mL, respectively. The chemical constituents of the ethyl acetate extract from A. alstroemeriae were further analyzed by liquid chromatography-mass spectrometry (LC-MS). We noted that A. alstroemeriae can create a variety of medicinal substances that have high value in medicine, such as caffeic acid (884.75 ng/mL), 3-phenyllactic acid (240.72 ng/mL) and norlichexanthone (74.36 ng/mL). Discussion In summary, many valuable active substances and medicinal substances can be obtained through the study of endophytic fungi of F. dibotrys.
Collapse
Affiliation(s)
- Qiqi Xie
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Yujie Jia
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Jiwen Tao
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Tongliang Bu
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Qing Wang
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Nayu Shen
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Xinyu Zhang
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Yirong Xiao
- Sichuan Agricultural University Hospital, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Lin Ye
- College of Animal Science and Technology, Sichuan Agricultural University, Cheng’du, Sichuan, China
| | - Zhao Chen
- Ya’an People’s Hospital, Ya’an People’s Hospital, Ya’an, Sichuan, China
| | - Huahai Huang
- Da’zhu Institute of Scientific and Technical Information, Unaffiliated, Da’zhu, Sichuan, China
| | - Qingfeng Li
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Zizhong Tang
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| |
Collapse
|
21
|
Xia Y, Chen Q, Liu HN, Chi Y, Zhu Y, Shan LS, Dai B, Wu L, Shi X. Synthetic routes and clinical application of new drugs approved by EMA during 2023. Eur J Med Chem 2024; 277:116762. [PMID: 39151275 DOI: 10.1016/j.ejmech.2024.116762] [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: 07/06/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
In 2023, the European Medicines Agency (EMA) granted approval to 77 new molecular entities (NMEs), consisting of 45 new chemical entities (NCEs) and 32 new biological entities (NBEs). These pharmacological agents encompass a broad spectrum of therapeutic domains, including oncology, cardiology, dermatology, diagnostic medicine, endocrinology, gastroenterology and hepatology, metabolic disorders, and neurology. Among the 77 approved pharmaceuticals, three received accelerated review status, and 17 (22 %) were granted orphan drug designation for the treatment of rare diseases. This review provides an overview of the clinical applications and synthetic routes of 42 newly approved NCEs by the EMA in 2023. The objective is to offer a comprehensive understanding of the synthetic approaches used in the development of these drug molecules, thereby inspiring the creation of novel, efficient, and applicable synthetic methodologies.
Collapse
Affiliation(s)
- Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Nan Liu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Lin Wu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xiaobao Shi
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
22
|
Dominguez AA, Perz MT, Xu Y, Cedillo LG, Huang OD, McIntyre CA, Vudatha V, Trevino JG, Liu J, Wang P. Unveiling the Promise: Navigating Clinical Trials 1978-2024 for PDAC. Cancers (Basel) 2024; 16:3564. [PMID: 39518005 PMCID: PMC11544830 DOI: 10.3390/cancers16213564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Despite many decades of research, pancreatic ductal adenocarcinoma (PDAC) remains one of the most difficult cancers to diagnose and treat effectively. Although there have been improvements in the 5-year overall survival rate, it is still very low at 12.5%. The limited efficacy of current therapies, even when PDAC is detected early, underscores the aggressive nature of the disease and the urgent need for more effective treatments. Clinical management of PDAC still relies heavily on a limited repertoire of therapeutic interventions, highlighting a significant gap between research efforts and available treatments. Over 4300 clinical trials have been or are currently investigating different treatment modalities and diagnostic strategies for PDAC, including targeted therapies, immunotherapies, and precision medicine approaches. These trials aim to develop more effective treatments and improve early detection methods through advanced imaging techniques and blood-based biomarkers. This review seeks to categorize and analyze PDAC-related clinical trials across various dimensions to understand why so few chemotherapeutic options are available to patients despite the numerous trials being conducted. This review aims to provide a comprehensive and nuanced understanding of the landscape of PDAC-related clinical trials, with the overarching goal of identifying opportunities to accelerate progress in drug development and improve patient outcomes in the fight against this devastating disease.
Collapse
Affiliation(s)
- Angel A. Dominguez
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Matthew T. Perz
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Yi Xu
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Leonor G. Cedillo
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Orry D. Huang
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Caitlin A. McIntyre
- Division of Surgical Oncology and Endocrine Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (V.V.); (J.G.T.)
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (V.V.); (J.G.T.)
| | - Jun Liu
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Pei Wang
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| |
Collapse
|
23
|
Noor L, Hafeez A, Rahman MA, Vishwakarma KK, Kapoor A, Ara N, Aqeel R. Demystifying the Potential of Embelin-Loaded Nanoformulations: a Comprehensive Review. AAPS PharmSciTech 2024; 25:249. [PMID: 39433611 DOI: 10.1208/s12249-024-02968-7] [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: 07/05/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Phytoconstituent based therapies have the potential to reduce the adverse effects and enhance overall patient compliance for different diseased conditions. Embelin (EMB) is a natural compound extracted from Embelia ribes that has demonstrated high therapeutic potential, particularly as anti-inflammatory and anticancer therapeutic applications. However, its poor water solubility and low oral bioavailability limitations make it challenging to use in biomedical applications. Nanostructure-based novel formulations have shown the potential to improve physicochemical and biological characteristics of active pharmaceutical ingredients obtained from plants. Different nanoformulations that have been utilized to encapsulate/entrap EMB for various therapeutic applications are nanoliposomes, nanostructured lipid carriers, niosomes, polymeric nanoparticles, nanosuspensions, phytosomes, self nanoemulsifying drug delivery system, silver nanoparticles, microparticles, solid lipid nanoparticle, gold nanoparticles and nanomicelles. The common methods reported for the preparation of EMB nanoformulations are thin film hydration, nanoprecipitation, ethanol injection, emulsification followed by sonication. The size of nanoformulations ranged in between 50 and 345 nm. In this review, the mentioned EMB loaded nanocarriers are methodically discussed for size, shape, drug entrapment, zeta potential, in vitro release & permeation and in vivo studies. Potential of EMB with other drugs (dual drug approach) incorporated in nanocarriers are also discussed (physicochemical and preclinical characteristics). Patents related to EMB nanoformulations are also presented which showed the clinical translation of this bioactive for future utilization in different indications.
Collapse
Affiliation(s)
- Layba Noor
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Abdul Hafeez
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Md Azizur Rahman
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | | | - Archita Kapoor
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Nargis Ara
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Rabia Aqeel
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
24
|
Ling J, Wu J, Cao Y, Zhang T, Cao X, Ge X, Liu Y, Wang M, Ren B, Lu J. Advances in nano-preparations for improving tetrandrine solubility and bioavailability. Arch Pharm (Weinheim) 2024; 357:e2400274. [PMID: 39031554 DOI: 10.1002/ardp.202400274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 07/22/2024]
Abstract
Tetrandrine (TET) is a natural bis-benzylisoquinoline alkaloid isolated from Stephania species with a wide range of biological and pharmacologic activities; it mainly serves as an anti-inflammatory agent or antitumor adjuvant in clinical applications. However, limitations such as prominent hydrophobicity, severe off-target toxicity, and low absorption result in suboptimal therapeutic outcomes preventing its widespread adoption. Nanoparticles have proven to be efficient devices for targeted drug delivery since drug-carrying nanoparticles can be passively transported to the tumor site by the enhanced permeability and retention (EPR) effects, thus securing a niche in cancer therapies. Great progress has been made in nanocarrier construction for TET delivery due to their outstanding advantages such as increased water-solubility, improved biodistribution and blood circulation, reduced off-target irritation, and combinational therapy. Herein, we systematically reviewed the latest advancements in TET-loaded nanoparticles and their respective features with the expectation of providing perspective and guidelines for future research and potential applications of TET.
Collapse
Affiliation(s)
- Jie Ling
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingping Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuening Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tingting Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiujun Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Ge
- School of Marxism, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yilan Liu
- Hematology Department, The General Hospital of the Western Theater Command PLA, Chengdu, China
| | - Maolin Wang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Bo Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
25
|
Aslan C, Maralbashi S, Shekari N, Javadian M, Shomali N, Kazemi T. Differential effects of docosahexaenoic acid (DHA) and linoleic acid (LA) on miR-101 and miR-342 tumor suppressor microRNAs in Taxol-treated HER2-positive breast cancer cells. Clin Nutr ESPEN 2024; 63:502-507. [PMID: 39047867 DOI: 10.1016/j.clnesp.2024.07.007] [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: 07/30/2023] [Revised: 05/14/2024] [Accepted: 07/06/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND & AIMS Docosahexaenoic acid (DHA) and linoleic acid (LA) have been shown to exhibit anti-proliferative effects against breast cancer cells. However, the mechanisms underlying these effects are not yet fully understood. One potential mechanism is through the regulation of microRNAs (miRs), which are known to play a crucial role in breast cancer development and progression. This study aimed to investigate the expression of miR-342 and miR-101 as tumor-suppressor miRs in the human HER-2 positive breast cancer cell line BT-474 after treatment with DHA, LA, alone or in combination with Taxol, a standard chemotherapy agent. METHODS The human breast cancer cell line BT-474 was cultured, and the IC50 for Taxol was determined using the MTT assay. Cells were then cultured and treated for 24 h with 100 μM DHA and 50 μM LA, alone or in combination with the respective IC50 of Taxol. Cells were harvested, and miRNA extraction and cDNA synthesis were performed using standard methods. Expression levels of miRs were analyzed using quantitative real-time PCR (qRT-PCR), and results were normalized against U6 snRNA expression levels. RESULTS The Taxol IC50 for BT-474 cells was 19 nM. According to the data obtained from our study, it was observed that Taxol treatment resulted in the down-regulation of both miR-101 and miR-342 (3.69 (p < 0.0001) and 1.88 fold, (p < 0.0001) respectively). In addition, DHA, LA and DHA + LA caused up-regulation of miR-101 (0.11, 0.05, 0.03 fold (p < 0.0001) respectively) but not miR-342 (decreased by 1.93 (p < 0.0001), 2.89 (p < 0.0001) and 1.19 fold (p = 0.0029) respectively). Notably, treatment with DHA, LA and DHA + LA was able to restore the down-regulated expression of miR-101 (0.25 (p < 0.0001), 0.05 (p = 0.0012) and 0.06 fold (p < 0.0001) respectively) during Taxol treatment. CONCLUSION Our study demonstrates that DHA and LA can effectively compensate for the reduced expression of miR-101 during Taxol treatment. These findings suggest that dietary fatty acids may play a critical role in modulating the anti-cancer effects of chemotherapy agents. Future studies are needed to investigate the functional aspects of dietary fatty acids on breast cancer development and progression.
Collapse
Affiliation(s)
- Cynthia Aslan
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Maralbashi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Najibeh Shekari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Javadian
- Department of Immunology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Gupta A, Niveria K, Chandpa HH, Singh M, Kumar V, Panda AK, Meena J. Stimuli-responsive magnetic silica-poly-lactic-co-glycolic acid hybrid nanoparticles for targeted cancer chemo-immunotherapy. Drug Deliv Transl Res 2024; 14:2712-2726. [PMID: 38347431 DOI: 10.1007/s13346-024-01521-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2024] [Indexed: 09/10/2024]
Abstract
Chemotherapy and immunotherapy are two important modalities in cancer management. However, due to multiple reasons, a monotherapy is only partially effective. Hence, if used concurrently in targeted and stimuli-responsive manner, it could have been superior therapeutically. To facilitate co-delivery of chemotherapeutic and immunotherapeutic agent to the target cancer cells, engineered nanoparticles, i.e., a pH-responsive polymer PLGA-coated magnetic silica nanoparticles (Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs) encapsulating paclitaxel (PTX) and siRNA against programmed cell death ligand-1 (PD-L1) are synthesized and characterized. Developed nanoparticles demonstrated pH-sensitive sustained drug release up to 10 days. In vitro 4T1 cell line studies showed efficient cellular uptake, PD-L1 gene downregulation, and apoptosis. Further, in vivo efficacy studies carried out in the mice model demonstrated a significant reduction of tumor growth following treatment with dual-Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs as compared with monotherapy with Fe3O4-SiO2-PLGA-PDA-PTX NPs. The high therapeutic efficacy observed with dual-Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs was mainly due to the cytotoxic effect of PTX combined with targeted silencing of the gene of interest, i.e., PD-L1, which in turn improve CD8+ T cell-mediated cancer cell death as evident with increased proliferation of CD8+ T cells in co-culture experiments. Thereby, dual-Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs may have a promising anti-cancer treatment potential against breast cancer; however, the beneficial effects of dual loading of PTX + PD-L1 siRNA may be corroborated against other cancer models such as lung and colorectal cancer models as well as in clinical trials.
Collapse
Affiliation(s)
- Anuradha Gupta
- School of Material Science and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi, 221005, India
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Karishma Niveria
- Nanobiotech Lab, Department of Zoology, Kirori Mal College, University of Delhi, Delhi, 110007, India
| | - Hitesh Harsukhbhai Chandpa
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi, 221005, India
| | - Mamta Singh
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Vikas Kumar
- Cell Biology Lab, Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, 110067, India
| | - Amulya Kumar Panda
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Jairam Meena
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi, 221005, India.
| |
Collapse
|
27
|
Jia M, Pei Y, Li N, Zhang Y, Song J, Niu JB, Yang H, Zhang S, Sun M. Synthesis and biological evaluation of 4-phenyl-5-quinolinyl substituted isoxazole analogues as potent cytotoxic and tubulin polymerization inhibitors against ESCC. Eur J Med Chem 2024; 275:116611. [PMID: 38901104 DOI: 10.1016/j.ejmech.2024.116611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
The identification of chemically different inhibitors that target the colchicine site of tubulin is still of great value for cancer treatment. Combretastatin A-4(CA-4), a naturally occurring colchicine-site binder characterized by its structural simplicity and biological activity, has served as a structural blueprint for the development of novel analogues with improved safety and therapeutic efficacy. In this study, a library of forty-eight 4-phenyl-5-quinolinyl substituted triazole, pyrazole or isoxazole analouges of CA-4, were synthesized and evaluated for their cytotoxicity against Esophageal Squamous Cell Carcinoma (ESCC) cell lines. Compound C11, which features a 2-methyl substitution at the quinoline and carries an isoxazole ring, emerged as the most promising, with 48 h IC50s of less than 20 nmol/L against two ESCC cell lines. The findings from EBI competitive assay, CETA, and in vitro tubulin polymerization assay of C11 are consistent with those of the positive control colchicine, demonstrating the clear affinity of compound C11 to the colchicine binding site. The subsequent cellular-based mechanism studies revealed that C11 significantly inhibited ESCC cell proliferation, arrested cell cycle at the M phase, induced apoptosis, and impeded migration. Experiments conducted in vivo further confirmed that C11 effectively suppressed the growth of ESCC without showing any toxicity towards the selected animal species. Overall, our research suggests that the tubulin polymerization inhibitor incorporating quinoline and the isoxazole ring may deserve consideration for cancer therapy.
Collapse
Affiliation(s)
- Meiqi Jia
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yuanyuan Pei
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Na Li
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Jian Song
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, China
| | - Hua Yang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Saiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
28
|
Malmberg R, van Doorn L, Cox JM, Daloul A, Ettafahi H, Oomen-de Hoop E, Zietse M, Bos MEMM, Koch BCP, van Leeuwen RWF. Effect of Switching the Histamine-1 Receptor Antagonist Clemastine to Cetirizine in Paclitaxel Premedication Regimens: The H1-Switch Study. JCO Oncol Pract 2024; 20:1243-1251. [PMID: 38848522 DOI: 10.1200/op.24.00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/19/2024] [Accepted: 04/15/2024] [Indexed: 06/09/2024] Open
Abstract
PURPOSE Premedication, including a histamine-1 receptor (H1) antagonist, is recommended to all patients treated with paclitaxel chemotherapy to reduce the incidence of hypersensitivity reactions (HSRs). However, the scientific basis for this premedication is not robust, which provides opportunities for optimization. Substitution of intravenously administered first-generation H1 antagonist for orally administered second-generation H1 antagonist could reduce side effects, and improve efficiency and sustainability. This study investigates the efficacy and safety of substituting intravenous clemastine for oral cetirizine as prophylaxis for paclitaxel-induced HSRs. METHODS This single-center, prospective, noninferiority study compares a historic cohort receiving a premedication regimen with intravenous clemastine to a prospective cohort receiving oral cetirizine. Primary end point of the study is HSR grade ≥3. The difference in incidence was calculated together with the 90% CI. We determined that the two-sided 90% CI of HSR grade ≥3 incidence in the oral cetirizine cohort should not be more than 4% higher (ie, the noninferiority margin) compared with the intravenous clemastine cohort. RESULTS Two hundred and twelve patients were included in the oral cetirizine cohort (June 2022 and May 2023) and 183 in the intravenous clemastine cohort. HSR grade ≥3 incidence was 1.6% (n = 3) in the intravenous clemastine cohort and 0.5% (n = 1) in the oral cetirizine cohort, resulting in a difference of -1.2% (90% CI, -3.4 to 1.1). CONCLUSION Premedication containing oral cetirizine is as safe as premedication containing intravenous clemastine in preventing paclitaxel-induced HSR grade ≥3. These findings could contribute to optimization of care for patients and improve efficiency and sustainability.
Collapse
Affiliation(s)
- Ruben Malmberg
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Leni van Doorn
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Juul M Cox
- Department of Clinical Pharmacy and Maasstad Lab, Maasstad Hospital, Rotterdam, the Netherlands
| | - Alaa Daloul
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Halima Ettafahi
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Michiel Zietse
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Monique E M M Bos
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roelof W F van Leeuwen
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
29
|
Gao X, Guo Y, Chen K, Wang H, Xie W. Study on the Chemical Constituents, Pharmacological Activities, and Clinical Application of Taxus. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1329-1357. [PMID: 39212496 DOI: 10.1142/s0192415x24500538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Taxus, a rare and protected genus predominantly distributed across the Northern Hemisphere's temperate regions, has garnered global attention due to its significant potential in medical research and pharmaceutical development, bolstered by advancements in cultivation techniques and medical technology. This review primarily focuses on the chemical constituents and pharmacological activities of Taxus, underscoring the progress and potential of these components in clinical applications. Recent studies have revealed that Taxus contains not only taxane active components but also flavonoids and polysaccharides with distinct activities. These compounds from Taxus exhibit potent antitumor, anti-inflammatory, immunomodulatory, antibacterial, and antidiabetic properties with evident mechanisms of action. Notably, the representative compound, paclitaxel, has demonstrated significant efficacy in treating various cancers, such as ovarian, breast, and lung cancer. This paper also reviews the basic situation of Taxus drug formulations, with extracts primarily administered orally and monomeric taxanes typically via injection, reflecting a mature development stage with ongoing research into oral formulations. Finally, this review summarizes the pharmacokinetic characteristics of crucial compounds in Taxus, including their absorption, distribution, metabolism, and excretion patterns in the human body. These pharmacokinetic profiles provide crucial guidance for evaluating the overall dosing regimen of Taxus and its components. The paper concludes with a forward-looking analysis of the potential applications of these compounds in disease treatment, envisioning their role in the future of medical and pharmaceutical advancements.
Collapse
Affiliation(s)
- Xinyu Gao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Yifan Guo
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Kuilin Chen
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Hanyi Wang
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Weidong Xie
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| |
Collapse
|
30
|
Si C, Gao J, Ma X. Natural killer cell-derived exosome-based cancer therapy: from biological roles to clinical significance and implications. Mol Cancer 2024; 23:134. [PMID: 38951879 PMCID: PMC11218398 DOI: 10.1186/s12943-024-02045-4] [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: 02/18/2024] [Accepted: 06/15/2024] [Indexed: 07/03/2024] Open
Abstract
Natural killer (NK) cells are important immune cells in the organism and are the third major type of lymphocytes besides T cells and B cells, which play an important function in cancer therapy. In addition to retaining the tumor cell killing function of natural killer cells, natural killer cell-derived exosomes cells also have the characteristics of high safety, wide source, easy to preserve and transport. At the same time, natural killer cell-derived exosomes are easy to modify, and the engineered exosomes can be used in combination with a variety of current cancer therapies, which not only enhances the therapeutic efficacy, but also significantly reduces the side effects. Therefore, this review summarizes the source, isolation and modification strategies of natural killer cell-derived exosomes and the combined application of natural killer cell-derived engineered exosomes with other antitumor therapies, which is expected to accelerate the clinical translation process of natural killer cell-derived engineered exosomes in cancer therapy.
Collapse
Affiliation(s)
- Chaohua Si
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100000, China
| | - Jianen Gao
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100000, China.
| | - Xu Ma
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100000, China.
| |
Collapse
|
31
|
Sun Y, Cheng Y, Hertz DL. Using maximum plasma concentration (C max) to personalize taxane treatment and reduce toxicity. Cancer Chemother Pharmacol 2024; 93:525-539. [PMID: 38734836 DOI: 10.1007/s00280-024-04677-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Taxanes are a widely used class of anticancer agents that play a vital role in the treatment of a variety of cancers. However, toxicity remains a major concern of using taxane drugs as some toxicities are highly prevalent, they can not only adversely affect patient prognosis but also compromise the overall treatment plan. Among all kinds of factors that associated with taxane toxicity, taxane exposure has been extensively studied, with different pharmacokinetic (PK) parameters being used as toxicity predictors. Compared to other widely used predictors such as the area under the drug plasma concentration curve versus time (AUC) and time above threshold plasma drug concentration, maximum plasma concentration (Cmax) is easier to collect and shows promise for use in clinical practice. In this article, we review the previous research on using Cmax to predict taxane treatment outcomes. While Cmax and toxicity have been extensively studied, research on the relationship between Cmax and efficacy is lacking. Most of the articles find a positive relationship between Cmax and toxicity but several articles have contradictory findings. Future clinical trials are needed to validate the relationship between Cmax and treatment outcome and determine whether Cmax can serve as a useful surrogate endpoint of taxane treatment efficacy.
Collapse
Affiliation(s)
- Yuchen Sun
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yue Cheng
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
Zhou J, Cai X, Lu Z, Xiong B, Peng C. Short-Term Safety Evaluation of Albumin-Bound Paclitaxel in Intraoperative and Postoperative Hyperthermic Intraperitoneal Chemotherapy for Gastric Cancer. J Gastrointest Cancer 2024; 55:877-887. [PMID: 38367177 DOI: 10.1007/s12029-024-01031-9] [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: 02/12/2024] [Indexed: 02/19/2024]
Abstract
PURPOSE To evaluate the short-term safety of albumin-bound paclitaxel in hyperthermic intraperitoneal chemotherapy (HIPEC) during and after gastric cancer (GC) surgery. METHODS A retrospective analysis of clinical data was conducted for GC surgery patients at Zhongnan Hospital of Wuhan University, from January 2020 to September 2022. The study group (n = 120) received HIPEC and the control group (n = 268) did not receive albumin-bound paclitaxel. Short-term safety indicators including intraoperative complications, hematological toxicity, liver and kidney function, and gastrointestinal function recovery were compared between the two groups. RESULTS There were no statistically significant differences between the two groups regarding intraoperative complications, hematological toxicity, liver and kidney function, and gastrointestinal function recovery time (P > 0.05 for all). In the study group, patients were further divided into subgroups based on dose and timing. Subgroup analysis revealed no significant differences among the different dose subgroups. However, when focusing on timing subgroups, the postoperative subgroup exhibited significantly higher white blood cell counts and bilirubin levels compared to the intraoperative subgroup, while the intraoperative subgroup had significantly higher bilirubin levels compared to both postoperative and intraoperative plus postoperative subgroups. CONCLUSION Albumin-bound paclitaxel demonstrates good safety and tolerability in HIPEC during and after GC surgery, without increasing the risk of intraoperative complications.
Collapse
Affiliation(s)
- Jingxiang Zhou
- Second Clinical College, Wuhan University, Wuhan , Hubei Province, 430070, China
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Xiaopeng Cai
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Zhao Lu
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Chunwei Peng
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China.
| |
Collapse
|
33
|
Wu Y, Jiang P, Chen Z, Li W, Dong B, Zhang Y. Efficacy and safety of different chemotherapy regimens concurrent with radiotherapy in the treatment of locally advanced cervical cancer. BMC Cancer 2024; 24:589. [PMID: 38745137 PMCID: PMC11094845 DOI: 10.1186/s12885-024-12358-8] [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: 11/17/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Evaluate the efficacy and safety of different chemotherapy regimens concurrent with radiotherapy in treating locally advanced cervical cancer (LACC). METHODS Retrospective data was collected from LACC patients who were treated at our institution. These patients were categorized into three groups: the single-agent cisplatin (DDP) chemoradiotherapy group, the paclitaxel plus cisplatin (TP) chemoradiotherapy group, and the nanoparticle albumin-bound (nab-) paclitaxel combined with cisplatin (nPP) chemoradiotherapy group. The primary endpoints were overall survival (OS) and progression-free survival (PFS) and the secondary endpoints were objective response rate (ORR) and incidence of adverse events (AEs). RESULTS A total of 124 patients were enrolled (32 in the DDP group, 41 in the TP group, and 51 in the nPP group). There were differences in OS (P = 0.041, HR 0.527, 95% CI 0.314-0.884) and PFS (P = 0.003, HR 0.517, 95% CI 0.343-0.779) between the three groups. Notably, the 2-year OS rate was significantly higher in the nPP group compared to the DDP group (92.2% vs. 85.4%, P = 0.012). The 2-year PFS rates showed a marked increase in the TP group (78.0% vs. 59.4%, P = 0.048) and the nPP group (88.2% vs. 59.4%, P = 0.001) relative to the DPP group, with multiple comparisons indicating that the 2-year PFS rate was significantly superior in the nPP group versus the DDP group (88.2% vs. 59.4%, P = 0.001). Moreover, the ORR was also significantly higher in the nPP group than in the DDP group (P = 0.013); and no statistically significant differences were found in the incidence of AEs among the groups (P > 0.05). CONCLUSIONS In LACC treatment, the two cisplatin-based doublet chemotherapy regimens are associated with better outcomes, with the nab-paclitaxel plus cisplatin regimen showing better efficacy than the paclitaxel plus cisplatin regimen. Furthermore, the AEs associated with these regimens were deemed tolerable. These findings could provide a reference for the clinical treatment of LACC. However, further prospective studies are needed to verify it.
Collapse
Affiliation(s)
- Yaping Wu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Peng Jiang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Zhiying Chen
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Wei Li
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Bin Dong
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yongchun Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
34
|
Chen R, Liu E, Fang Y, Gao N, Zhang M, Zhang X, Chen W, Liang C, Zhang Y, Huang Y. Naturally sourced amphiphilic peptides as paclitaxel vehicles for breast cancer treatment. BIOMATERIALS ADVANCES 2024; 159:213824. [PMID: 38490019 DOI: 10.1016/j.bioadv.2024.213824] [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: 10/13/2023] [Revised: 01/09/2024] [Accepted: 03/03/2024] [Indexed: 03/17/2024]
Abstract
The marketed paclitaxel (PTX) formulation Taxol relies on the application of Cremophor EL as a solubilizer. The major drawback of Taxol is its hypersensitivity reactions and a pretreatment of anti-allergic drugs is a necessity. Therefore, developing an efficient and safe delivery vehicle is a solution to increase PTX treatment outcomes with minimal adverse effects. In this work, we prepared the amphiphilic peptides (termed AmP) from soybean proteins using a facile two-step method. AmP could efficiently solubilize PTX by self-assembling into mixed micelles with D-α-tocopherol polyethylene glycol succinate (TPGS), a common pharmaceutical expedient (PTX@TPGS-AmP). The intravenously administrated PTX@TPGS-AmP exhibited a slow clearance (0.24 mL·(min·kg)-1) and an enhanced AUC (41.4 μg.h/mL), manifesting a 3.6-fold increase compared to Taxol. In a murine 4T1 tumor model, PTX@TPGS-AmP displayed a superior antitumor effect over Taxol. Importantly, safety assessment showed a high biocompatibility of AmP and an i.v. dose up to 2500 mg/kg led to no observable abnormalities in the mice. In summary, the AmP presents a new green and easily-prepared amphiphilic biomaterial, with promising potential as a pharmaceutical excipient for drug delivery.
Collapse
Affiliation(s)
- Rongli Chen
- Shenyang Pharmaceutical University, Shenyang 110016, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Yuefei Fang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Nan Gao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaoru Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Wanying Chen
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Chuxin Liang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yu Zhang
- Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| |
Collapse
|
35
|
Bhat AA, Afzal M, Goyal A, Gupta G, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Shahwan M, Paudel KR, Ali H, Sahu D, Prasher P, Singh SK, Dua K. The impact of formaldehyde exposure on lung inflammatory disorders: Insights into asthma, bronchitis, and pulmonary fibrosis. Chem Biol Interact 2024; 394:111002. [PMID: 38604395 DOI: 10.1016/j.cbi.2024.111002] [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: 02/16/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
Lung inflammatory disorders are a major global health burden, impacting millions of people and raising rates of morbidity and death across many demographic groups. An industrial chemical and common environmental contaminant, formaldehyde (FA) presents serious health concerns to the respiratory system, including the onset and aggravation of lung inflammatory disorders. Epidemiological studies have shown significant associations between FA exposure levels and the incidence and severity of several respiratory diseases. FA causes inflammation in the respiratory tract via immunological activation, oxidative stress, and airway remodelling, aggravating pre-existing pulmonary inflammation and compromising lung function. Additionally, FA functions as a respiratory sensitizer, causing allergic responses and hypersensitivity pneumonitis in sensitive people. Understanding the complicated processes behind formaldehyde-induced lung inflammation is critical for directing targeted strategies aimed at minimizing environmental exposures and alleviating the burden of formaldehyde-related lung illnesses on global respiratory health. This abstract explores the intricate relationship between FA exposure and lung inflammatory diseases, including asthma, bronchitis, allergic inflammation, lung injury and pulmonary fibrosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Mahal Road, Jaipur, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2050, Australia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Dipak Sahu
- Department of Pharmacology, Amity University, Raipur, Chhattisgarh, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
36
|
Agnihotram R, Dhar R, Dhar D, Purushothaman K, Narasimhan AK, Devi A. Fusion of Exosomes and Nanotechnology: Cutting-Edge Cancer Theranostics. ACS APPLIED NANO MATERIALS 2024; 7:8489-8506. [DOI: 10.1021/acsanm.4c01033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Rohan Agnihotram
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Debolina Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Kaavya Purushothaman
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Ashwin Kumar Narasimhan
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| |
Collapse
|
37
|
Szulc A, Woźniak M. Targeting Pivotal Hallmarks of Cancer for Enhanced Therapeutic Strategies in Triple-Negative Breast Cancer Treatment-In Vitro, In Vivo and Clinical Trials Literature Review. Cancers (Basel) 2024; 16:1483. [PMID: 38672570 PMCID: PMC11047913 DOI: 10.3390/cancers16081483] [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: 03/03/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
This literature review provides a comprehensive overview of triple-negative breast cancer (TNBC) and explores innovative targeted therapies focused on specific hallmarks of cancer cells, aiming to revolutionize breast cancer treatment. TNBC, characterized by its lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), presents distinct features, categorizing these invasive breast tumors into various phenotypes delineated by key elements in molecular assays. This article delves into the latest advancements in therapeutic strategies targeting components of the tumor microenvironment and pivotal hallmarks of cancer: deregulating cellular metabolism and the Warburg effect, acidosis and hypoxia, the ability to metastasize and evade the immune system, aiming to enhance treatment efficacy while mitigating systemic toxicity. Insights from in vitro and in vivo studies and clinical trials underscore the promising effectiveness and elucidate the mechanisms of action of these novel therapeutic interventions for TNBC, particularly in cases refractory to conventional treatments. The integration of targeted therapies tailored to the molecular characteristics of TNBC holds significant potential for optimizing clinical outcomes and addressing the pressing need for more effective treatment options for this aggressive subtype of breast cancer.
Collapse
Affiliation(s)
| | - Marta Woźniak
- Department of Clinical and Experimental Pathology, Division of General and Experimental Pathology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| |
Collapse
|
38
|
Li Y, Zhao Q, Zhu X, Zhou L, Song P, Liu B, Tian D, Chen Q, Zhou J, Deng G. Self-Assembled nanoparticles of natural bioactive molecules enhance the delivery and efficacy of paclitaxel in glioblastoma. CNS Neurosci Ther 2024; 30:e14528. [PMID: 38044793 PMCID: PMC11017454 DOI: 10.1111/cns.14528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary malignant tumor in the central nervous system. Paclitaxel (PTX) is a well-established and highly effective anti-cancer drug for peripheral solid tumors. However, the application of PTX in GBM is hindered by several limitations, including poor water solubility, restricted entry across the blood-brain barrier (BBB), and enhanced excretion by efflux transporters. P-glycoprotein (P-gp) is a crucial efflux transporter that is abundantly present in cerebral vascular endothelial cells and GBM cells. It plays a significant role in the exocytosis of PTX within tumor tissues. METHODS Recently, we have developed a novel technique for creating self-assembled nanoparticles utilizing a range of natural bioactive molecules. These nanoparticles can encapsulate insoluble drugs and effectively cross the BBB. In additional, we revealed that certain nanoparticles have the potential to act as P-gp inhibitors, thereby reducing the excretion of PTX. In this study, we conducted a screening of bioactive molecular nanoparticles to identify those that effectively inhibit the function of P-gp transporters. RESULTS Among the candidates, we identified ursolic acid nanoparticles (UA NPs) as the P-gp inhibitors. Furthermore, we prepared co-assembled UA NPs embedded with paclitaxel, referred to as UA-PTX NPs. Our results demonstrate that UA-PTX NPs can enhance the blood concentration of PTX, facilitate its entry into the BBB, and inhibit the function of P-gp, resulting in a decrease in the excretion of PTX. This discovery effectively addressed the above three issues associated with the use of PTX in glioma treatment. CONCLUSIONS UA-PTX NPs demonstrate strong anti-tumor effects and show great potential for treating GBM.
Collapse
Affiliation(s)
- Yong Li
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Qingyu Zhao
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Xinyi Zhu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Long Zhou
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Ping Song
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Baohui Liu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Daofeng Tian
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Qianxue Chen
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Jiangbing Zhou
- Department of NeurosurgeryYale UniversityNew HavenConnecticutUSA
| | - Gang Deng
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| |
Collapse
|
39
|
Hu J, Zhu BY, Niu ZX. Catalysts of Healing: A Symphony of Synthesis and Clinical Artistry in Small-Molecule Agents for Breast Cancer Alleviation. Molecules 2024; 29:1166. [PMID: 38474678 DOI: 10.3390/molecules29051166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Breast cancer, characterized by its molecular intricacy, has witnessed a surge in targeted therapeutics owing to the rise of small-molecule drugs. These entities, derived from cutting-edge synthetic routes, often encompassing multistage reactions and chiral synthesis, target a spectrum of oncogenic pathways. Their mechanisms of action range from modulating hormone receptor signaling and inhibiting kinase activity, to impeding DNA damage repair mechanisms. Clinical applications of these drugs have resulted in enhanced patient survival rates, reduction in disease recurrence, and improved overall therapeutic indices. Notably, certain molecules have showcased efficacy in drug-resistant breast cancer phenotypes, highlighting their potential in addressing treatment challenges. The evolution and approval of small-molecule drugs have ushered in a new era for breast cancer therapeutics. Their tailored synthetic pathways and defined mechanisms of action have augmented the precision and efficacy of treatment regimens, paving the way for improved patient outcomes in the face of this pervasive malignancy. The present review embarks on a detailed exploration of small-molecule drugs that have secured regulatory approval for breast cancer treatment, emphasizing their clinical applications, synthetic pathways, and distinct mechanisms of action.
Collapse
Affiliation(s)
- Jing Hu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Bi-Yue Zhu
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Zhen-Xi Niu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| |
Collapse
|
40
|
Gliech CR, Yeow ZY, Tapias-Gomez D, Yang Y, Huang Z, Tijhuis AE, Spierings DC, Foijer F, Chung G, Tamayo N, Bahrami-Nejad Z, Collins P, Nguyen TT, Plata Stapper A, Hughes PE, Payton M, Holland AJ. Weakened APC/C activity at mitotic exit drives cancer vulnerability to KIF18A inhibition. EMBO J 2024; 43:666-694. [PMID: 38279026 PMCID: PMC10907621 DOI: 10.1038/s44318-024-00031-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024] Open
Abstract
The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Colin R Gliech
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Daniel Tapias-Gomez
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yuchen Yang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zhaoyu Huang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Andréa E Tijhuis
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, AV, 9713, The Netherlands
| | - Diana Cj Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, AV, 9713, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, AV, 9713, The Netherlands
| | - Grace Chung
- Oncology Research, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - Nuria Tamayo
- Medicinal Chemistry, Amgen Research, Thousand Oaks, CA, 91320, USA
| | | | - Patrick Collins
- Genome Analysis Unit, Amgen Research, South San Francisco, CA, 94084, USA
| | - Thong T Nguyen
- Genome Analysis Unit, Amgen Research, South San Francisco, CA, 94084, USA
| | - Andres Plata Stapper
- Center for Research Acceleration by Digital Innovation, Amgen Research, South San Francisco, CA, 94084, USA
| | - Paul E Hughes
- Oncology Research, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - Marc Payton
- Oncology Research, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
41
|
Ivone R, Karabots A, Meenach SA. Development of Aerosol Dry Powder Chemotherapeutic-Loaded Microparticles for the Treatment of Lung Cancer. AAPS PharmSciTech 2024; 25:42. [PMID: 38366056 DOI: 10.1208/s12249-024-02751-8] [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: 09/27/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, resulting in the highest mortality rates among both men and women with respect to all other types of cancer. Difficulties in treating lung cancer arise from late-stage diagnoses and tumor heterogeneity and current treatment involves a combination of chemotherapeutics, surgery, and radiation. Chemotherapeutics administered systemically can lead to undesirable side effects and severe off-site toxicity. For example, chronic administration of the chemotherapeutic doxorubicin (DOX) leads to cardiotoxicity, thereby limiting its long-term use. Systemic administration of the highly lipophilic molecule paclitaxel (PTX) is hindered by its water solubility, necessitating the use of solubilizing agents, which can induce side effects. Thus, in this investigation, formulations consisting of spray-dried microparticles (MP) containing DOX and PTX were produced to be administered as dry powder aerosols directly to the lungs. Acetalated dextran (Ac-Dex) was used as the polymer in these formulations, as it is a biocompatible and biodegradable polymer that exhibits pH-responsive degradation. Solid-state characterization revealed that DOX and PTX remained in solubility favoring amorphous states in the MP formulations and that both drugs remained thermally stable throughout the spray drying process. In vitro release studies demonstrated the pH sensitivity of the formulations due to the use of Ac-Dex, as well as the release of both therapeutics over the course of at least 48 h. In vitro aerosol dispersion studies demonstrated that both formulations exhibited suitable aerosol dispersion properties for deep lung delivery.
Collapse
Affiliation(s)
- Ryan Ivone
- Department of Chemical Engineering, University of Rhode Island, 360 Fascitelli Center for Advanced Engineering, 2 Upper College Road, Kingston, Rhode Island, 02881, USA
| | - Ana Karabots
- Department of Chemical Engineering, University of Rhode Island, 360 Fascitelli Center for Advanced Engineering, 2 Upper College Road, Kingston, Rhode Island, 02881, USA
| | - Samantha A Meenach
- Department of Chemical Engineering, University of Rhode Island, 360 Fascitelli Center for Advanced Engineering, 2 Upper College Road, Kingston, Rhode Island, 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, 02881, USA.
| |
Collapse
|
42
|
Abhang A, Katari O, Ghadi R, Chaudhari D, Jain S. Exploring the synergistic behavior of paclitaxel and vorinostat upon co-loading in albumin nanoparticles for breast cancer management. Drug Deliv Transl Res 2024; 14:510-523. [PMID: 37605040 DOI: 10.1007/s13346-023-01415-7] [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: 08/07/2023] [Indexed: 08/23/2023]
Abstract
Breast cancer is challenging to treat accompanied with poor clinical outcomes. Paclitaxel (PTX) is a first-line chemotherapeutic agent, but possesses limitations due to side effects, high dose, non-specific tissue distribution, and drug resistance. An epigenetic modulator, vorinostat (VOR) is known to enhance PTX efficacy and therefore to resolve the issues of conventional PTX formulations, we designed PTX- and VOR-bound albumin nanoparticles (PTX-VOR-BSA-NPs) using antisolvent precipitation technique where albumin is used as a carrier and a targeting agent. The PTX-VOR-BSA-NPs were of 140 nm size, polydispersity index around 0.18, and about 78% and 68% of entrapment efficiency for PTX and VOR, respectively. A bi-pattern release of both PTX and VOR was observed from PTX-VOR-BSA-NPs with a burst release for 2 h succeeded by sustained release until 24 h. A significantly lower %cell viability was observed in MCF-7 cell lines, while efficient cellular drug uptake was found in MDA-MB-231 cells. Furthermore, a greater apoptotic index was found compared to free PTX and VOR because of the synergistic activity of these drugs. The PTX-VOR-BSA-NPs also showcased superior pharmacokinetic profile and noteworthy reduction in the tumor volume compared to Intaxel in 4T1 cell line-induced breast tumor model. Further, the NPs showed similar levels of toxicity biomarkers as that of control. Overall, the developed PTX-VOR-BSA-NPs were found to have less toxicity and more effectiveness compared to the marketed formulation, thus affirming the generation of a potent as well as and safe product.
Collapse
Affiliation(s)
- Ashwin Abhang
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India
| | - Oly Katari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India
| | - Dasharath Chaudhari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| |
Collapse
|
43
|
Danziger M, Noble H, Roque DM, Xu F, Rao GG, Santin AD. Microtubule-Targeting Agents: Disruption of the Cellular Cytoskeleton as a Backbone of Ovarian Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1452:1-19. [PMID: 38805122 DOI: 10.1007/978-3-031-58311-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Microtubules are dynamic polymers composed of α- and β-tubulin heterodimers. Microtubules are universally conserved among eukaryotes and participate in nearly every cellular process, including intracellular trafficking, replication, polarity, cytoskeletal shape, and motility. Due to their fundamental role in mitosis, they represent a classic target of anti-cancer therapy. Microtubule-stabilizing agents currently constitute a component of the most effective regimens for ovarian cancer therapy in both primary and recurrent settings. Unfortunately, the development of resistance continues to present a therapeutic challenge. An understanding of the underlying mechanisms of resistance to microtubule-active agents may facilitate the development of novel and improved approaches to this disease.
Collapse
Affiliation(s)
- Michael Danziger
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Helen Noble
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dana M Roque
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fuhua Xu
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gautam G Rao
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
44
|
Yan D, Arakelyan J, Wan T, Raina R, Chan TK, Ahn D, Kushnarev V, Cheung TK, Chan HC, Choi I, Ho PY, Hu F, Kim Y, Lau HL, Law YL, Leung CS, Tong CY, Wong KK, Yim WL, Karnaukhov NS, Kong RY, Babak MV, Matsuda Y. Genomics-driven derivatization of the bioactive fungal sesterterpenoid variecolin: Creation of an unnatural analogue with improved anticancer properties. Acta Pharm Sin B 2024; 14:421-432. [PMID: 38261827 PMCID: PMC10793096 DOI: 10.1016/j.apsb.2023.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/25/2023] [Accepted: 08/24/2023] [Indexed: 01/25/2024] Open
Abstract
A biosynthetic gene cluster for the bioactive fungal sesterterpenoids variecolin (1) and variecolactone (2) was identified in Aspergillus aculeatus ATCC 16872. Heterologous production of 1 and 2 was achieved in Aspergillus oryzae by expressing the sesterterpene synthase VrcA and the cytochrome P450 VrcB. Intriguingly, the replacement of VrcB with homologous P450s from other fungal terpenoid pathways yielded three new variecolin analogues (5-7). Analysis of the compounds' anticancer activity in vitro and in vivo revealed that although 5 and 1 had comparable activities, 5 was associated with significantly reduced toxic side effects in cancer-bearing mice, indicating its potentially broader therapeutic window. Our study describes the first tests of variecolin and its analogues in animals and demonstrates the utility of synthetic biology for creating molecules with improved biological activities.
Collapse
Affiliation(s)
- Dexiu Yan
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Jemma Arakelyan
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Teng Wan
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Ritvik Raina
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Tsz Ki Chan
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Dohyun Ahn
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Vladimir Kushnarev
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Tsz Kiu Cheung
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Ho Ching Chan
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Inseo Choi
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Pui Yi Ho
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Feijun Hu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Yujeong Kim
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Hill Lam Lau
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Ying Lo Law
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Chi Seng Leung
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Chun Yin Tong
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Kai Kap Wong
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Wing Lam Yim
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Nikolay S. Karnaukhov
- Moscow Clinical Research Center Named After A.S. Loginov, Moscow 111123, Russian Federation
| | - Richard Y.C. Kong
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Maria V. Babak
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| | - Yudai Matsuda
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
- iGEM Team “VarieCure”, City University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
45
|
Passos JS, Apolinario AC, Ishida K, Martins TS, Lopes LB. Nanostructured lipid carriers loaded into in situ gels for breast cancer local treatment. Eur J Pharm Sci 2024; 192:106638. [PMID: 37967657 DOI: 10.1016/j.ejps.2023.106638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/18/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
In this study, nanostructured lipid carriers (NLC) were developed and employed to obtain in situ thermosensitive formulations for the ductal administration and prolonged retention of drugs as a new strategy for breast cancer local treatment. NLC size was influenced by the type and concentration of the oil phase, surfactants, and drug incorporation, ranging from 221.6 to 467.5 nm. The type of liquid lipid influenced paclitaxel and 5-fluorouracil cytotoxicity, with tributyrin-containing NLC reducing IC50 values by 2.0-7.0-fold compared to tricaprylin NLC in MCF-7, T-47D and MDA-MB-231 cells. In spheroids, the NLCs reduced IC50 compared to either drug solution (3.2-6.2-fold). Although a significant reduction (1.26 points, p < 0.001) on the health index of Galleria mellonella larvae was observed 5 days after NLC administration, survival was not significantly reduced. To produce thermosensitive gels, the NLCs were incorporated in a poloxamer (11 %, w/w) dispersion, which gained viscosity (2-fold) at 37 °C. After 24 h, ∼53 % of paclitaxel and 83 % of 5-fluorouracil were released from the NLC; incorporation in the poloxamer gel further prolonged release. Intraductal administration of NLC-loaded gel increased the permanence of hydrophilic (2.2-3.0-fold) and lipophilic (2.1-2.3-fold) fluorescent markers in the mammary tissue compared to the NLC (as dispersion) and the markers solutions. In conclusion, these results contribute to improving our understanding of nanocarrier design with increased cytotoxicity and prolonged retention for the intraductal route. Tributyrin incorporation increased the cytotoxicity of paclitaxel and 5-fluorouracil in monolayer and spheroids, while NLC incorporation in thermosensitive gels prolonged tissue retention of both hydrophilic and hydrophobic compounds.
Collapse
Affiliation(s)
- Julia S Passos
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Av. Prof. Lineu Prestes, Sao Paulo SP 05508-000, Brazil
| | - Alexsandra C Apolinario
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Av. Prof. Lineu Prestes, Sao Paulo SP 05508-000, Brazil
| | - Kelly Ishida
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Tereza S Martins
- Department of Chemistry, Federal University of Sao Paulo (UNIFESP), Diadema, São Paulo, Brazil
| | - Luciana B Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Av. Prof. Lineu Prestes, Sao Paulo SP 05508-000, Brazil.
| |
Collapse
|
46
|
Fateh ST, Fateh ST, Salehi-Najafabadi A, Aref AR. Commercial and regulatory challenges in cancer nanomedicine. FUNCTIONALIZED NANOMATERIALS FOR CANCER RESEARCH 2024:579-601. [DOI: 10.1016/b978-0-443-15518-5.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
Liu C, Xia C, Xia C. Biology and function of exosomes in tumor immunotherapy. Biomed Pharmacother 2023; 169:115853. [PMID: 37951023 DOI: 10.1016/j.biopha.2023.115853] [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: 08/03/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023] Open
Abstract
Exosomes are nano-scale extracellular vesicles that are found widely in various biological fluids. As messengers, exosomes deliver characteristic biological information from donor cells, facilitating their accumulation and subsequent transfer of information to tumor immune cells. Immunotherapy is a cutting-edge strategy for cancer therapy, but it has not yet reached its full potential owing to severe side effects and limited efficacy. Exosomes possess antigens and immunostimulatory molecules and can serve as cell-free vaccines to induce antitumor immunity. In addition, given their stability, low immunogenicity, and targeting ability, exosomes represent ideal drug delivery systems in tumor immunotherapy by delivering cargoes, including non-coding ribonucleic acids (RNAs), membrane proteins, chemotherapeutic agents, and immune cell death inducers. Exosomes can also be engineered to precisely target tumor cells. However, as a rising star in tumor immunotherapy, exosomes are also impeded by some challenges, including the lack of uniform technical standards for their isolation and purification, the need to improve exosomal cargo loading for efficient exosome delivery, and the expansion of clinical trials, which are currently in their infancy. Long-term, multi-center, and large-scale clinical trials are needed to evaluate the performance of exosomes in the future. Nonetheless, exosomes have demonstrated encouraging performance in tumor immunotherapy. In this review, we summarize the potential and challenges of exosomes in tumor immunotherapy, with the aim to shed light on exosomes as new-era tumor immunotherapy tools.
Collapse
Affiliation(s)
- Can Liu
- Foshan Maternity and Chlid Healthcare Hospital, Foshan 528000, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 515150, China
| | - Cong Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Chenglai Xia
- Foshan Maternity and Chlid Healthcare Hospital, Foshan 528000, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 515150, China.
| |
Collapse
|
48
|
Tan Y, Hu H, Zhu W, Wang T, Gao T, Wang H, Chen J, Xu J, Xu S, Zhu H. Design, synthesis and biological evaluation of novel dihydroquinolin-4(1H)-one derivatives as novel tubulin polymerization inhibitors. Eur J Med Chem 2023; 262:115881. [PMID: 37883897 DOI: 10.1016/j.ejmech.2023.115881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023]
Abstract
A series of novel dihydroquinolin-4(1H)-one derivatives targeting colchicine binding site on tubulin were designed, synthesized and evaluated as anticancer agents. The most potent compound 6t showed remarkable antiproliferative activities against four cancer cell lines with IC50 values among 0.003-0.024 μM and tubulin polymerization inhibitory activity (IC50 = 3.06 μM). Further mechanism studies revealed that compound 6t could induce K562 cells apoptosis and arrest at the G2/M phase. Meanwhile, 6t significantly inhibited migration and invasion of MDA-MB-231 cells, and disrupted the angiogenesis in human umbilical vein endothelial cells (HUVECs) in vitro. In addition, compound 6t inhibited tumor growth in H22 allograft tumor model with a tumor growth inhibition (TGI) rate of 63.3 % (i.v., 20 mg/kg per day) without obvious toxicity. Collectively, these results indicated that compound 6t was a novel tubulin polymerization inhibitor with potent anticancer properties in vitro and in vivo.
Collapse
Affiliation(s)
- Yuchen Tan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Han Hu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wenjian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Tao Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Tian Gao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Hongqi Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Jian Chen
- Department of Hepatobiliary Surgery, The First People's Hospital of Kunshan, Suzhou, Jiangsu, 215300, PR China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China; Shenzhen Research Institute of China Pharmaceutical University, Nanshan District, Shenzheng, 518052, PR China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China; Department of Hepatobiliary Surgery, The First People's Hospital of Kunshan, Suzhou, Jiangsu, 215300, PR China; Shenzhen Research Institute of China Pharmaceutical University, Nanshan District, Shenzheng, 518052, PR China.
| | - Huajian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China.
| |
Collapse
|
49
|
Mikesell AR, Isaeva E, Schulte ML, Menzel AD, Sriram A, Prahl MM, Shin SM, Sadler KE, Yu H, Stucky CL. Keratinocyte Piezo1 drives paclitaxel-induced mechanical hypersensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571332. [PMID: 38168305 PMCID: PMC10760029 DOI: 10.1101/2023.12.12.571332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Recent work demonstrates that epidermal keratinocytes are critical for normal touch sensation. However, it is unknown if keratinocytes contribute to touch evoked pain and hypersensitivity following tissue injury. Here, we used inhibitory optogenetic and chemogenetic techniques to determine the extent to which keratinocyte activity contributes to the severe neuropathic pain that accompanies chemotherapeutic treatment. We found that keratinocyte inhibition largely alleviates paclitaxel-induced mechanical hypersensitivity. Furthermore, we found that paclitaxel exposure sensitizes mouse and human keratinocytes to mechanical stimulation through the keratinocyte mechanotransducer Piezo1. These findings demonstrate the contribution of non-neuronal cutaneous cells to neuropathic pain and pave the way for the development of new pain-relief strategies that target epidermal keratinocytes and Piezo1.
Collapse
Affiliation(s)
- Alexander R Mikesell
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | - Elena Isaeva
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | | | - Anthony D Menzel
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | - Anvitha Sriram
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | - Megan M Prahl
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | - Katelyn E Sadler
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin; Milwaukee, WI 53226, USA
| |
Collapse
|
50
|
Galos D, Balacescu L, Vidra R, Sur D. Real-World Data on Second-Line Therapy with Ramucirumab for Metastatic Gastric Cancer: A Two-Center Study on Romanian Population. Life (Basel) 2023; 13:2300. [PMID: 38137901 PMCID: PMC10744814 DOI: 10.3390/life13122300] [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: 10/24/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
(1) Background: Following the results of RAINBOW and REGARD trials, ramucirumab was approved as the standard second-line treatment for patients with advanced or metastatic gastric or gastroesophageal junction (GEJ) cancer, alone or in combination with paclitaxel. The present study aimed to evaluate the efficacy and safety of ramucirumab in the Romanian population during every-day clinical practice. (2) Methods: A two-center, retrospective, observational study evaluated patients with metastatic gastric and GEJ cancer treated with ramucirumab monotherapy or associated with paclitaxel. The patients were treated between 2018 and 2022 in two Romanian centers as follows: 18 patients underwent treatment with ramucirumab monotherapy, while 51 received the combined treatment regimen. Study endpoints included median progression-free survival (PFS), median overall survival (OS), and the evaluation of treatment-induced adverse events (AEs). (3) Results: In the study cohort (n = 69), the most frequent treatment-induced AE in the ramucirumab plus paclitaxel arm was hematological toxicity; the most common AE for patients treated with ramucirumab monotherapy was fatigue and headache. Overall, the median PFS was 4.7 months (95% CI: 3.4-5.9 months) and median OS was 18.23 months (95% CI: 15.6-20.7 months). PFS was correlated with the number of treatment cycle administrations, Eastern Cooperative Oncology Group performance status at treatment initiation, and metastatic site (visceral vs. peritoneal). OS was correlated with the number of treatment cycles administered and human epidermal growth factor receptor-2 status. (4) Conclusions: The results support the previously described toxicity profile for ramucirumab monotherapy or associated with paclitaxel and demonstrated a relatively superior median PFS.
Collapse
Affiliation(s)
- Diana Galos
- Department of Medical Oncology, The Oncology Institute Prof. Dr. Ion Chiricuţă, 400015 Cluj-Napoca, Romania;
| | - Loredana Balacescu
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute Prof. Dr. Ion Chiricuţă, 400015 Cluj-Napoca, Romania;
| | - Radu Vidra
- Postgraduate Program for Bio-Behavioral Integrative Medicine, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
- Department of Medical Oncology, Regional Institute of Gastroenterology and Hepatology Prof. Dr. Octavian Fodor, 400162 Cluj-Napoca, Romania
| | - Daniel Sur
- Department of Medical Oncology, The Oncology Institute Prof. Dr. Ion Chiricuţă, 400015 Cluj-Napoca, Romania;
- Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania
| |
Collapse
|