1
|
Iizuka A, Kanda M, Sato Y, Shimizu D, Umeda S, Tanaka H, Hattori N, Hayashi M, Tanaka C, Kodera Y. Association of NR0B1 with Malignant Phenotypes in Esophageal Squamous Cell Carcinoma Through Modulation of p53-Independent Cell-Cycle Regulation. Ann Surg Oncol 2025; 32:4464-4475. [PMID: 40080367 DOI: 10.1245/s10434-025-17109-y] [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: 10/01/2024] [Accepted: 02/17/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Transcriptome analysis of primary tumor tissues from esophageal squamous cell carcinoma (ESCC) patients with early postoperative distant metastasis identified nuclear receptor subfamily 0, group B, member 1 (NR0B1) as a novel gene associated with the malignant phenotypes of ESCC. This study aimed to elucidate the oncological functions of NR0B1 in ESCC and assess the significance of its tissue expression. METHODS We investigated the effects of NR0B1 knockdown on the proliferation, migration, and adhesion capacities, in vivo tumor growth, and intracellular signaling pathways of ESCC cell lines. The correlation between tissue NR0B1 expression at both the mRNA and protein levels and postoperative prognosis was analyzed by using two independent cohorts. RESULTS Silencing NR0B1 significantly inhibited the proliferation, migration, and adhesion capacities of ESCC cell lines and decreased tumor growth in mouse cell line derived xenograft models. Knockdown of NR0B1 results in the upregulation of cell cycle regulators p21 and p27, alongside the downregulation of TK1, cyclin E1, CDK2 and CDT1, in a manner independent of p53. Although elevated tissue NR0B1 expression did not show a significant association with TNM stage, it was identified as an independent prognostic factor at both the mRNA and protein levels across two distinct patient cohorts. CONCLUSIONS NR0B1 plays a critical role in the malignant phenotype of ESCC by modulating cell cycle regulators. Tissue NR0B1 expression may serve as a valuable biomarker for assessing prognostic risk in ESCC patients.
Collapse
Affiliation(s)
- Akimitsu Iizuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruyoshi Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
2
|
Song S, Wang J, Ouyang X, Huang R, Wang F, Xie J, Chen Q, Hu D. Therapeutic connections between pyroptosis and paclitaxel in anti-tumor effects: an updated review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04036-8. [PMID: 40257490 DOI: 10.1007/s00210-025-04036-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/06/2025] [Indexed: 04/22/2025]
Abstract
As a form of inflammation-associated cell death, pyroptosis has gained widespread attention in recent years. Accumulating evidence indicates that pyroptosis regulates tumor growth and is associated with autoimmune disorders and inflammatory response. Paclitaxel, a traditional Chinese medicine, usually induces death of cancer cells as a chemotherapeutic agent. Previous studies have revealed that paclitaxel can exert an anti-tumor effect through a variety of cell death mechanisms, of which pyroptosis plays a pivotal role in inhibiting tumor growth and enhancing anti-tumor immunity. In this review, we summarize the current advances in therapeutic connections between pyroptosis and paclitaxel in anti-tumor effects.
Collapse
Affiliation(s)
- Shuxin Song
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Renyin Huang
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Wang
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junke Xie
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qianyun Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Wang Y, Sun X, Ren M, Ma F, Zhao R, Zhu X, Xu Y, Cao N, Chen Y, Pan Y, Zhao A. Integrative network pharmacology, transcriptomics, and proteomics reveal the material basis and mechanism of the Shen Qing Weichang Formula against gastric cancer. Chin Med 2025; 20:42. [PMID: 40155922 PMCID: PMC11954191 DOI: 10.1186/s13020-025-01091-4] [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: 11/19/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common malignancy with poor prognosis and lack of efficient therapeutic methods. Shen Qing Weichang Formula (SQWCF) is a patented traditional herbal prescription for GC, but its efficacy and underlying mechanism remains to be clarified. PURPOSE To explore the efficacy and potential mechanism of SQWCF in treating GC. METHODS A subcutaneous transplantation tumor model of human GC was established for assessing SQWCF's efficacy and safety. A comprehensive strategy integrating mass spectrometry, network pharmacology, omics analysis, and bioinformatic methods was adopted to explore the core components, key targets, and potential mechanism of SQWCF in treating GC. Molecular docking, immunohistochemistry, quantitative real-time PCR, and western blot were applied to validation. RESULTS In the mouse model of GC, SQWCF effectively suppressed the GC growth without evident toxicity and enhanced the therapeutic efficacy of paclitaxel. Network pharmacology and molecular docking based on mass spectrometry showed that key targets (CASP3, TP53, Bcl-2, and AKT1) and core active components (Calycosin, Glycitein, Liquiritigenin, Hesperetin, and Eriodictyol) involved in the anti-GC effect of SQWCF had stable binding affinity, of which AKT1 ranked the top in the affinity. Validation based on network pharmacology and omics analysis confirmed that PI3K-AKT and MAPK signaling pathways, as well as downstream apoptosis pathway, explained the therapeutic effects of SQWCF on GC. In addition, family with sequence similarity 81 member A (FAM81A) was identified as a novel biomarker of GC that was aberrantly highly expressed in GC and associated with poor prognosis by bioinformatic analysis, and was an effector target of SQWCF at both mRNA and protein levels. CONCLUSION This study uncovers a synergistic multi-component, multi-target, and multi-pathway regulatory mechanism of SQWCF in treating GC comprehensively, emphasizing its potential for therapeutic use and providing new insights into GC treatment.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Xiaoyu Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Mingming Ren
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Fangqi Ma
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Ruohan Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Xiaohong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yuanyuan Chen
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yongfu Pan
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China.
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China.
| |
Collapse
|
4
|
Kong D, Duan J, Chen S, Wang Z, Ren J, Lu J, Chen T, Song Z, Wu D, Chang Y, Yin Z, Shen Z, Zheng H. Transplant oncology and anti-cancer immunosuppressants. Front Immunol 2025; 15:1520083. [PMID: 39840041 PMCID: PMC11747528 DOI: 10.3389/fimmu.2024.1520083] [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: 10/30/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Organ transplantation is a life-saving intervention that enhances the quality of life for patients with end-stage organ failure. However, long-term immunosuppressive therapy is required to prevent allogeneic graft rejection, which inadvertently elevates the risk of post-transplant malignancies, especially for liver transplant recipients with a prior history of liver cancer. In response, the emerging field of transplant oncology integrates principles from oncology and immunology to improve outcomes for patients at high risk of tumor occurrence or recurrence following transplantation. Therefore, it is of substantial clinical significance to develop immunosuppressants that possess both immunosuppressive and anti-tumor properties. For instance, mTOR inhibitors demonstrate anti-tumor effects among antimetabolite immunosuppressive drugs, and recent studies indicate that capecitabine, an antimetabolite chemotherapeutic, may also exhibit immunosuppressive activity in the clinic for liver transplants suffering from hepatocellular carcinoma. This review systematically explores potential immunosuppressants with dual anti-tumor and immunosuppressive effects to support the management of transplant patients at elevated risk of tumor occurrence or recurrence.
Collapse
Affiliation(s)
- Dejun Kong
- Nankai University School of Medicine, Tianjin, China
| | - Jinliang Duan
- Nankai University School of Medicine, Tianjin, China
| | - Shaofeng Chen
- Nankai University School of Medicine, Tianjin, China
| | - Zhenglu Wang
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Jiashu Ren
- Tianjin First Central Clinical College, Tianjin, China
| | - Jianing Lu
- Tianjin First Central Clinical College, Tianjin, China
| | - Tao Chen
- Nankai University School of Medicine, Tianjin, China
| | - Zhuolun Song
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
| | - Di Wu
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
| | - Yuan Chang
- Nankai University School of Medicine, Tianjin, China
| | - Zhongqian Yin
- Tianjin First Central Clinical College, Tianjin, China
| | - Zhongyang Shen
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| | - Hong Zheng
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| |
Collapse
|
5
|
Qiao JX, Guo DY, Tian H, Wang ZP, Fan QQ, Tian Y, Sun J, Zhang XF, Zou JB, Cheng JX, Luan F, Zhai BT. Research progress of paclitaxel nanodrug delivery system in the treatment of triple-negative breast cancer. Mater Today Bio 2024; 29:101358. [PMID: 39677523 PMCID: PMC11638641 DOI: 10.1016/j.mtbio.2024.101358] [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: 07/16/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by the loss or low expression of estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR). Due to the lack of clear therapeutic targets, paclitaxel (PTX) is often used as a first-line standard chemotherapy drug for the treatment of high-risk and locally advanced TNBC. PTX is a diterpenoid alkaloid extracted and purified from Taxus plants, functioning as an anticancer agent by inducing and promoting tubulin polymerization, inhibiting spindle formation in cancer cells, and preventing mitosis. However, its clinical application is limited by low solubility and high toxicity. Nanodrug delivery system (NDDS) is one of the feasible methods to improve the water solubility of PTX and reduce side effects. In this review, we summarize the latest advancements in PTX-targeted NDDS, as well as its combination with other codelivery therapies for TNBC treatment. NDDS includes passive targeting, active targeting, stimuli-responsive, codelivery, and multimode strategies. These systems have good prospects in improving the bioavailability of PTX, enhancing tumor targeting, reducing toxicity, controlling drug release, and reverse tumor multidrug resistance (MDR). This review provides valuable insights into the clinical development and application of PTX-targeted NDDS in the treatment of TNBC.
Collapse
Affiliation(s)
- Jia-xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Department of Pharmacy, National Old Pharmacist Inheritance Studio, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Zhan-peng Wang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qiang-qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| |
Collapse
|
6
|
Li X, Jin K, Liao YC, Lee WJ, Chen LC, Cheng TC, Ho YS, Guo P. RNA Nanotechnology for Codelivering High-Payload Nucleoside Analogs to Cancer with a Synergetic Effect. Mol Pharm 2024; 21:5690-5702. [PMID: 39388598 PMCID: PMC12012820 DOI: 10.1021/acs.molpharmaceut.4c00674] [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] [Indexed: 10/12/2024]
Abstract
Nucleoside analogs are potent inhibitors for cancer treatment, but the main obstacles to their application in humans are their toxicity, nonspecificity, and lack of targeted delivery tools. Here, we report the use of RNA four-way junctions (4WJs) to deliver two nucleoside analogs, floxuridine (FUDR) and gemcitabine (GEM), with high payloads through routine and simple solid-state RNA synthesis and nanoparticle assembly. The design of RNA nanotechnology for the co-delivery of nucleoside analogs and the chemotherapeutic drug paclitaxel (PTX) resulted in synergistic effects and high efficacy in the treatment of Triple-Negative Breast Cancer (TNBC). The 4WJ-drug complexes were confirmed to have efficient tumor spontaneous targeting and no toxicity because the motility of RNA nanoparticles has been previously shown to enable these RNA-drug complexes to spontaneously accumulate in tumor blood vessels. The negative charge of RNA enables those RNA complexes that are not targeted to tumor vasculature to circulate in the blood and enter the urine through the kidney glomerulus, without accumulating in organs, therefore being nontoxic. Drug incorporation into RNA 4WJ can be precisely controlled with a defined loading amount, location, and ratio. The incorporation of nucleoside analogs into 4WJ only requires one step using nucleoside analogue phosphoramidites during solid-phase RNA synthesis, without the need for additional conjugation and purification processes.
Collapse
Affiliation(s)
- Xin Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Kai Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University; Taipei, Taiwan
| | - Wen-Jui Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Li-Ching Chen
- Department of Biological Science & Technology, China Medical University, Taichung, Taiwan
| | - Tzu-chun Cheng
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan
| | - Peixuan Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
7
|
Butera F, Sero JE, Dent LG, Bakal C. Actin networks modulate heterogeneous NF-κB dynamics in response to TNFα. eLife 2024; 13:e86042. [PMID: 39110005 PMCID: PMC11524587 DOI: 10.7554/elife.86042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 08/05/2024] [Indexed: 11/01/2024] Open
Abstract
The canonical NF-κB transcription factor RELA is a master regulator of immune and stress responses and is upregulated in pancreatic ductal adenocardinoma (PDAC) tumours. In this study, we characterised previously unexplored endogenous RELA-GFP dynamics in PDAC cell lines through live single-cell imaging. Our observations revealed that TNFα stimulation induces rapid, sustained, and non-oscillatory nuclear translocation of RELA. Through Bayesian analysis of single-cell datasets with variation in nuclear RELA, we predicted that RELA heterogeneity in PDAC cell lines is dependent on F-actin dynamics. RNA-seq analysis identified distinct clusters of RELA-regulated gene expression in PDAC cells, including TNFα-induced RELA upregulation of the actin regulators NUAK2 and ARHGAP31. Further, siRNA-mediated depletion of ARHGAP31 and NUAK2 altered TNFα-stimulated nuclear RELA dynamics in PDAC cells, establishing a novel negative feedback loop that regulates RELA activation by TNFα. Additionally, we characterised the NF-κB pathway in PDAC cells, identifying how NF-κB/IκB proteins genetically and physically interact with RELA in the absence or presence of TNFα. Taken together, we provide computational and experimental support for interdependence between the F-actin network and the NF-κB pathway with RELA translocation dynamics in PDAC.
Collapse
Affiliation(s)
- Francesca Butera
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondonUnited Kingdom
| | - Julia E Sero
- Department of Life Sciences, University of BathBathUnited Kingdom
| | - Lucas G Dent
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondonUnited Kingdom
| | - Chris Bakal
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondonUnited Kingdom
| |
Collapse
|
8
|
Pan L, Schneider F, Ottenbruch M, Wiechert R, List T, Schoch P, Mertes B, Gaich T. A general strategy for the synthesis of taxane diterpenes. Nature 2024; 632:543-549. [PMID: 38862025 DOI: 10.1038/s41586-024-07675-8] [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: 03/06/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
The carbon skeleton of any organic molecule serves as the foundation for its three-dimensional structure, playing a pivotal role in determining its physical and biological properties1. As such, taxane diterpenes are one of the most well-known natural product families, primarily owing to the success of their most prominent compound, paclitaxel, an effective anticancer therapeutic for more than 25 years2-6. In contrast to classical taxanes, the bioactivity of cyclotaxanes (also referred to as complex taxanes) remains significantly underexplored. The carbon skeletons of these two groups of taxanes differ significantly, and so would typically their own distinct synthetic approaches. Here we report a versatile synthetic strategy based on the interconversion of complex molecular frameworks, providing general access to the wider taxane diterpene family. A range of classical and cyclotaxane frameworks was prepared including, among others, the total syntheses of taxinine K (2), canataxapropellane (5) and dipropellane C from a single advanced intermediate. The synthetic approach deliberately eschews biomimicry, emphasizing instead the power of stereoelectronic control in orchestrating the interconversion of polycyclic frameworks.
Collapse
Affiliation(s)
- Lu Pan
- University of Konstanz, Department of Chemistry, Konstanz, Germany.
| | - Fabian Schneider
- University of Konstanz, Department of Chemistry, Konstanz, Germany
- Scripps Research, La Jolla, CA, USA
| | | | - Rainer Wiechert
- University of Konstanz, Department of Chemistry, Konstanz, Germany
- Department of Chemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Tatjana List
- University of Konstanz, Department of Chemistry, Konstanz, Germany
| | - Philipp Schoch
- University of Konstanz, Department of Chemistry, Konstanz, Germany
| | - Bastian Mertes
- University of Konstanz, Department of Chemistry, Konstanz, Germany
| | - Tanja Gaich
- University of Konstanz, Department of Chemistry, Konstanz, Germany.
| |
Collapse
|
9
|
Zhang C, Deng J, Li K, Lai G, Liu H, Zhang Y, Xie B, Zhong X. Mononuclear phagocyte system-related multi-omics features yield head and neck squamous cell carcinoma subtypes with distinct overall survival, drug, and immunotherapy responses. J Cancer Res Clin Oncol 2024; 150:37. [PMID: 38279056 PMCID: PMC10817853 DOI: 10.1007/s00432-023-05512-5] [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: 08/08/2023] [Accepted: 11/10/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Recent research reported that mononuclear phagocyte system (MPS) can contribute to immune defense but the classification of head and neck squamous cell carcinoma (HNSCC) patients based on MPS-related multi-omics features using machine learning lacked. METHODS In this study, we obtain marker genes for MPS through differential analysis at the single-cell level and utilize "similarity network fusion" and "MoCluster" algorithms to cluster patients' multi-omics features. Subsequently, based on the corresponding clinical information, we investigate the prognosis, drugs, immunotherapy, and biological differences between the subtypes. A total of 848 patients have been included in this study, and the results obtained from the training set can be verified by two independent validation sets using "the nearest template prediction". RESULTS We identified two subtypes of HNSCC based on MPS-related multi-omics features, with CS2 exhibiting better predictive prognosis and drug response. CS2 represented better xenobiotic metabolism and higher levels of T and B cell infiltration, while the biological functions of CS1 were mainly enriched in coagulation function, extracellular matrix, and the JAK-STAT signaling pathway. Furthermore, we established a novel and stable classifier called "getMPsub" to classify HNSCC patients, demonstrating good consistency in the same training set. External validation sets classified by "getMPsub" also illustrated similar differences between the two subtypes. CONCLUSIONS Our study identified two HNSCC subtypes by machine learning and explored their biological difference. Notably, we constructed a robust classifier that presented an excellent classifying prediction, providing new insight into the precision medicine of HNSCC.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
| | - Jielian Deng
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
| | - Kangjie Li
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
| | - Hui Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
| | - Yuan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
| | - Biao Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
| | - Xiaoni Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
| |
Collapse
|
10
|
Ali MU, Chaudhary BN, Panja S, Gendelman HE. Theranostic Diagnostics. Results Probl Cell Differ 2024; 73:551-578. [PMID: 39242393 DOI: 10.1007/978-3-031-62036-2_22] [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: 09/09/2024]
Abstract
Diagnosing and then treating disease defines theranostics. The approach holds promise by facilitating targeted disease outcomes. The simultaneous analysis of finding the presence of disease pathophysiology while providing a parallel in treatment is a novel and effective strategy for seeking improved medical care. We discuss how theranostics improves disease outcomes is discussed. The chapter reviews the delivery of targeted therapies. Bioimaging techniques are highlighted as early detection and tracking systems for microbial infections, degenerative diseases, and cancers.
Collapse
Affiliation(s)
- Mohammad Uzair Ali
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bharat N Chaudhary
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sudipta Panja
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
11
|
Yun D, Liu D, Liu J, Feng Y, Chen H, Chen S, Xie Q. In Vitro/In Vivo Preparation and Evaluation of cRGDyK Peptide-Modified Polydopamine-Bridged Paclitaxel-Loaded Nanoparticles. Pharmaceutics 2023; 15:2644. [PMID: 38004622 PMCID: PMC10674738 DOI: 10.3390/pharmaceutics15112644] [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: 10/02/2023] [Revised: 11/05/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer remains a disease with one of the highest mortality rates worldwide. The poor water solubility and tissue selectivity of commonly used chemotherapeutic agents contribute to their poor efficacy and serious adverse effects. This study proposes an alternative to the traditional physicochemically combined modifications used to develop targeted drug delivery systems, involving a simpler surface modification strategy. cRGDyK peptide (RGD)-modified PLGA nanoparticles (NPs) loaded with paclitaxel were constructed by coating the NP surfaces with polydopamine (PD). The average particle size of the produced NPs was 137.6 ± 2.9 nm, with an encapsulation rate of over 80%. In vitro release tests showed that the NPs had pH-responsive drug release properties. Cellular uptake experiments showed that the uptake of modified NPs by tumor cells was significantly better than that of unmodified NPs. A tumor cytotoxicity assay demonstrated that the modified NPs had a lower IC50 and greater cytotoxicity than those of unmodified NPs and commercially available paclitaxel formulations. An in vitro cytotoxicity study indicated good biosafety. A tumor model in female BALB/c rats was established using murine-derived breast cancer 4T1 cells. RGD-modified NPs had the highest tumor-weight suppression rate, which was higher than that of the commercially available formulation. PTX-PD-RGD-NPs can overcome the limitations of antitumor drugs, reduce drug toxicity, and increase efficacy, showing promising potential in cancer therapy.
Collapse
Affiliation(s)
- Dan Yun
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dengyuan Liu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jinlin Liu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yanyi Feng
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hongyu Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Simiao Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qingchun Xie
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
12
|
Alsadooni JFK, Haghi M, Barzegar A, Feizi MAH. The effect of chitosan hydrogel containing gold nanoparticle complex with paclitaxel on colon cancer cell line. Int J Biol Macromol 2023; 247:125612. [PMID: 37390995 DOI: 10.1016/j.ijbiomac.2023.125612] [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: 02/13/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Colon cancer is a significant global health issue, and its primary treatment, chemotherapy, is limited by toxicity and drug resistance. This has led researchers to explore alternative therapeutic approaches. One such approach is the use of chitosan, a natural biopolymer with anti-cancer properties, and paclitaxel, a potent chemotherapeutic agent with promising activity against many types of cancer. In this study, the effectiveness of a chitosan hydrogel that contains a complex of gold nanoparticles with paclitaxel in treating LS174T colon cancer cell line was investigated. The synthesized chitosan hydrogel was characterized and used to treat the colon cancer cells in cell culture. MTT assay and apoptotic gene expression analysis were conducted to evaluate the complex's effectiveness. The results showed that the chitosan hydrogel-loaded gold nanoparticle-paclitaxel complex exhibited a potent cytotoxic effect against the cancer cells. Moreover, the treatment resulted in a significant increase in pro-apoptotic BAX and BAD expression and a decrease in anti-apoptotic BCL2 expression, indicating a pro-apoptotic effect. These findings suggest that utilizing a chitosan hydrogel that contains a complex of gold nanoparticles with paclitaxel shows promise as a viable treatment option for colon cancer. Further research is needed to determine the potential efficacy and safety of this treatment approach in clinical settings.
Collapse
Affiliation(s)
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Abolfazl Barzegar
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
13
|
Gialelis TL, Wang Z, Homer JA, Yang WH, Chung T, Hu Q, Smedley CJ, Pawar NJ, Upadhyay NS, Tuveson DA, Lyons SK, Lukey MJ, Moses JE. Inhibition of mitochondrial metabolism by (-)-jerantinine A: synthesis and biological studies in triple-negative breast cancer cells. RSC Med Chem 2023; 14:710-714. [PMID: 37122543 PMCID: PMC10131581 DOI: 10.1039/d3md00049d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
A concise semi-synthesis of the Aspidosperma alkaloids, (-)-jerantinine A and (-)-melodinine P, and derivatives thereof, is reported. The novel compounds were shown to have potent activity against MDA-MB-231 triple-negative breast cancer cells. Furthermore, unbiased metabolomics and live cell reporter assays reveal (-)-jerantinine A alters cellular redox metabolism and induces oxidative stress that coincides with cell cycle arrest.
Collapse
Affiliation(s)
- Timothy L Gialelis
- La Trobe Institute for Molecular Science, La Trobe University Melbourne VIC 3086 Australia
| | - Zifei Wang
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Joshua A Homer
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Wen-Hsuan Yang
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Taemoon Chung
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Qingting Hu
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
- Graduate Program in Genetics, Stony Brook University Stony Brook NY 11794 USA
| | - Christopher J Smedley
- La Trobe Institute for Molecular Science, La Trobe University Melbourne VIC 3086 Australia
| | - Nitin J Pawar
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Nitinkumar S Upadhyay
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - David A Tuveson
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Scott K Lyons
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - Michael J Lukey
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| | - John E Moses
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Road Cold Spring Harbor NY 11724 USA
| |
Collapse
|
14
|
Minerva, Bhat A, Verma S, Chander G, Jamwal RS, Sharma B, Bhat A, Katyal T, Kumar R, Shah R. Cisplatin-based combination therapy for cancer. J Cancer Res Ther 2023; 19:530-536. [PMID: 37470570 DOI: 10.4103/jcrt.jcrt_792_22] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Cisplatin, that is, cis-diamminedichloroplatinum is a coordinate compound that is mainly preferred as prior treatment against several solid tumors and malignancies like ovaries, head and neck, testicular, and lung cancers because of its anticancer activity. Cisplatin binds at the N7 position of purine and forms adducts, leading to altered activity of DNA that triggers apoptosis. DNA damage is followed by several signaling pathways like induced oxidative stress, upregulated p53, mitogen-activated protein kinase (MAPK), and Jun N-terminal kinases (JNK) or Akt pathways along with induced apoptosis. Additionally, cisplatin treatment comes with few disadvantages such as toxic effects, that is, hepatotoxicity, cardiotoxicity, neurotoxicity, etc., and drug resistance. Furthermore, to overcome cisplatin resistance and toxicological effects, combination drug therapy has been considered. The aim of the review is to focus on the molecular mechanism of action of cisplatin and combination drug therapy to reduce the side effects in cancer therapy.
Collapse
Affiliation(s)
- Minerva
- ICMR-CAR, School of Biotechnology, SMVDU, Katra, Jammu and Kashmir, India
| | - Amrita Bhat
- ICMR-CAR, School of Biotechnology, SMVDU, Katra, Jammu and Kashmir, India
| | - Sonali Verma
- ICMR-CAR, School of Biotechnology, SMVDU, Katra, Jammu and Kashmir, India
| | - Gresh Chander
- ICMR-CAR, School of Biotechnology, SMVDU, Katra, Jammu and Kashmir, India
| | | | - Bhawani Sharma
- ICMR-CAR, School of Biotechnology, SMVDU, Katra, Jammu and Kashmir, India
| | - Audesh Bhat
- Department of Molecular Biology, Central University of Jammu, Jammu and Kashmir, India
| | - Taruna Katyal
- Reproductive Biology Maternal, Child Health and Nutrition Division, ICMR, New Delhi, India
| | - Rakesh Kumar
- ICMR-CAR, School of Biotechnology, SMVDU, Katra, Jammu and Kashmir, India
| | - Ruchi Shah
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
15
|
Huang SQ, Zhang HM, Zhang YC, Wang LY, Zhang ZR, Zhang L. Comparison of two methods for tumour-targeting peptide modification of liposomes. Acta Pharmacol Sin 2023; 44:832-840. [PMID: 36271156 PMCID: PMC10043035 DOI: 10.1038/s41401-022-01011-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/27/2022] [Indexed: 11/09/2022]
Abstract
Liposomes decorated with tumour-targeting cell-penetrating peptides can enhance specific drug delivery at the tumour site. The TR peptide, c(RGDfK)-AGYLLGHINLHHLAHL(Aib)HHIL, is pH-sensitive and actively targets tumour cells that overexpress integrin receptor αvβ3, such as B16F10 melanoma cells. Liposomes can be modified with the TR peptide by two different methods: utilization of the cysteine residue on TR to link DSPE-PEG2000-Mal contained in the liposome formula (LIPTR) or decoration of TR with a C18 stearyl chain (C18-TR) for direct insertion into the liposomal phospholipid bilayer through electrostatic and hydrophobic interactions (LIPC18-TR). We found that both TR and C18-TR effectively reversed the surface charge of the liposomes when the systems encountered the low pH of the tumour microenvironment, but LIPC18-TR exhibited a greater increase in the charge, which led to higher cellular uptake efficiency. Correspondingly, the IC50 values of PTX-LIPTR and PTX-LIPC18-TR in B16F10 cells in vitro were 2.1-fold and 2.5-fold lower than that of the unmodified PTX-loaded liposomes (PTX-LIP), respectively, in an acidic microenvironment (pH 6.3). In B16F10 tumour-bearing mice, intravenous administration of PTX-LIPTR and PTX-LIPC18-TR (8 mg/kg PTX every other day for a total of 4 injections) caused tumour reduction ratios of 39.4% and 56.1%, respectively, compared to 20.8% after PTX-LIP administration. Thus, we demonstrated that TR peptide modification could improve the antitumour efficiency of liposomal delivery systems, with C18-TR presenting significantly better results. After investigating different modification methods, our data show that selecting an adequate method is vital even when the same molecule is used for decoration.
Collapse
Affiliation(s)
- Shi-Qi Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy; College of Polymer Science and Engineering; Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Han-Ming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy; College of Polymer Science and Engineering; Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Yi-Cong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy; College of Polymer Science and Engineering; Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Lu-Yao Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy; College of Polymer Science and Engineering; Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Zhi-Rong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy; College of Polymer Science and Engineering; Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy; College of Polymer Science and Engineering; Med-X Center for Materials, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
16
|
Response of Patients with Taxane-Refractory Advanced Urothelial Cancer to Enfortumab Vedotin, a Microtubule-Disrupting Agent. Case Rep Urol 2023; 2023:1024239. [PMID: 36691441 PMCID: PMC9867573 DOI: 10.1155/2023/1024239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Enfortumab vedotin (EV), a nectin-4-directed antibody conjugated to monomethyl auristatin E (MMAE), has been approved for patients with advanced urothelial carcinoma (aUC) previously treated with platinum-based chemotherapy and immune inhibitors. Taxane agents and MMAE share antitumor mechanisms through microtubule disruption, thus raising a notable concern regarding cross-resistance between these drugs. This case report describes two patients with taxane-based chemotherapy-refractory aUC who responded well to EV. A 71-year-old man (case 1) with pT3N0M0 renal pelvic UC showed a partial response to EV in metastatic lesions of the bilateral lungs and right pelvic lymph nodes after three cycles of paclitaxel plus gemcitabine chemotherapy. A 53-year-old man (case 2) with cT3bN2M0 bladder UC underwent platinum-based neoadjuvant chemotherapy and the following radial cystectomy (ypTis ypN0). He developed bilateral lung metastases and showed a complete response to EV in the metastatic lesions after 20 cycles of paclitaxel plus nedaplatin chemotherapy. Our experience of two cases demonstrated that tumor response to EV can be expected in patients with taxane-refractory aUC.
Collapse
|
17
|
Swed S, Shaheen N, Hafez W, Elsayed Talat N, Rozan SS, Diab R, Nashwan AJ, Motawea KR, Alibrahim H, Albuni MK, Battikh E, Sawaf B, Shoib S. Pembrolizumab versus paclitaxel for previously treated, advanced gastro-esophageal junction cancer: A systematic review and meta-analysis of randomized clinical trials. Medicine (Baltimore) 2022; 101:e31940. [PMID: 36482610 PMCID: PMC9726310 DOI: 10.1097/md.0000000000031940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This paper aims to compare the effectiveness and safety of pembrolizumab and paclitaxel as a second line for patients with locally advanced gastroesophageal cancer. METHODS By searching PubMed, Scopus, Web of Science, and Ovid, any randomized clinical study comparing the effectiveness of paclitaxel and pembrolizumab as second-line therapy for advanced gastroesophageal cancer met the inclusion criteria. Only 3 of the 23 eligible studies that were fully reviewed were eligible for meta-analysis. RESULTS The total number of patients included in the meta-analysis was 635 in the pembrolizumab group and 596 in the paclitaxel group. In terms of objective response rate, there was no statistically significant difference between pembrolizumab and paclitaxel (relative risk = 1.10, 95% CI = 0.80-1.50, P = .57). Furthermore, Pembrolizumab and paclitaxel did not differ in terms of the rate of partial response statistically significantly from one another, according to the overall analysis (relative risk = 0.93, 95% CI = 0.57-1.52, P-value = .78). CONCLUSION There is no difference between pembrolizumab and paclitaxel in objective response rate. The objective response rate shows that doctors may consider either treatment for patients with advanced gastroesophageal cancer, given the time to response is comparable across therapies.
Collapse
Affiliation(s)
- Sarya Swed
- Faculty of Medicine, Aleppo University, Aleppo, Syria
| | - Nour Shaheen
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Wael Hafez
- NMC Royal Hospital, 16th Street, Khalifa City, Abu Dhabi, UAE
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo, Egypt
| | | | - Samah S. Rozan
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rehab Diab
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | | | | | | | | | - Elias Battikh
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Bisher Sawaf
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Sheikh Shoib
- Department of Psychiatry, Jawahar Lal Nehru Memorial Hospital, Srinagar, Kashmir, India
| |
Collapse
|
18
|
Ye J, Li L, Yin J, Wang H, Li R, Yang Y, Guan Y, Xia X, Liu Y. Tumor-targeting intravenous lipid emulsion of paclitaxel: Characteristics, stability, toxicity, and toxicokinetics. J Pharm Anal 2022; 12:901-912. [PMID: 36605580 PMCID: PMC9805944 DOI: 10.1016/j.jpha.2022.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022] Open
Abstract
Lipid nanoemulsions are promising nanodrug delivery carriers that can improve the efficacy and safety of paclitaxel (PTX). However, no intravenous lipid emulsion of PTX has been approved for clinical treatment, and systemic safety profiles have not yet been reported. Here we outline the development of a PTX-loaded tumor-targeting intravenous lipid emulsion (PTX Emul) and describe its characteristics, colloidal stability, and systemic safety profiles in terms of acute toxicity, long-term toxicity, and toxicokinetics. We also compare PTX Emul with conventional PTX injection. Results showed that PTX Emul exhibited an ideal average particle size (approximately 160 nm) with narrow size distribution and robust colloidal stability under different conditions. Hypersensitivity reaction and hemolysis tests revealed that PTX Emul did not induce hypersensitivity reactions and had no hemolytic potential. In addition, where the alleviated systemic toxicity of PTX Emul may be attributed to the altered toxicokinetic characteristics in beagle dogs, including the decreased AUC and increased plasma clearance and volume of distribution, PTX Emul alleviated acute and long-term toxicity as evidenced by the enhanced the median lethal dose and approximate lethal dose, moderate body weight change, decreased bone marrow suppression and organ toxicity compared with those under PTX injection at the same dose. A fundamental understanding of the systemic safety profiles, high tumor-targeting efficiency, and superior antitumor activity in vivo of PTX Emul can provide powerful evidence of its therapeutic potential as a future treatment for breast cancer.
Collapse
Affiliation(s)
- Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiye Yin
- National Beijing Center for Drug Safety Evaluation and Research, Beijing Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Renjie Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yongbiao Guan
- National Beijing Center for Drug Safety Evaluation and Research, Beijing Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China,Corresponding author.
| | - Xuejun Xia
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Corresponding author.
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Corresponding author.
| |
Collapse
|
19
|
Li D, Zhang Q, Zhou Y, Zhu H, Li T, Du F. A novel nitidine chloride nanoparticle overcomes the stemness of CD133 +EPCAM + Huh7 hepatocellular carcinoma cells for liver cancer therapy. BMC Pharmacol Toxicol 2022; 23:48. [PMID: 35820920 PMCID: PMC9277916 DOI: 10.1186/s40360-022-00589-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/04/2022] [Indexed: 01/15/2023] Open
Abstract
Background Stemness of CD133+EPCAM+ hepatocellular carcinoma cells ensures cancer resistance to apoptosis,which is a challenge to current liver cancer treatments. In this study, we evaluated the tumorcidal activity of a novel nanoparticle of nitidine chloride (TPGS-FA/NC, TPGS-FA: folic acid modified D-α-tocopheryl polyethylene glycol 1000 succinate, NC: nitidine chloride), against human hepatocellular carcinoma (HCC) cell line Huh7 growth in vitro and in vivo. Methods Huh7 cells were treated with TPGS-FA/NC. Cell proliferation was assessed using MTT and colony assays. The expression of cell markers and signaling proteins was detected using western blot analyses. A sphere culture technique was used to enrich cancer stem cells (CSC) in Huh7 cells. TPGS-FA/NC (7.5, 15, 30, 60, 120 μg/mL) dose-dependently inhibited the proliferation of HCC cells, which associated with a reduction in AQP3 and STAT3 expression. Importantly,TPGS-FA/NC (10, 20, and 40 μg/mL) significantly reduced the EpCAM+/CD133+cell numbers, suppressed the sphere formation. The in vivo antitumor efficacy of TPGS-FA/NC was proved in Huh7 cell xenograft model in BALB/c nude mice, which were administered TPGS-FA/NC(4 mg· kg − 1· d − 1, ig) for 2 weeks. Results TPGS-FA/NC dose-dependently suppressed the AQP3/STAT3/CD133 axis in Huh7 cells. In Huh7 xenograft bearing nude mice, TPGS-FA/NC administration markedly inhibited Huh7 xenograft tumor growth . Conclusions TPGS-FA/NC inhibit HCC tumor growth through multiple mechanisms, and it may be a promising candidate drug for the clinical therapy of hepatocellular carcinoma. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00589-z.
Collapse
Affiliation(s)
- Danni Li
- School of Chemistry and Chemical Eengineering, Guangxi Minzu University, No.158, Da Xue Xi street, Xixiangtang District, Nanning, 530006, Guangxi Province, China.
| | - Qiying Zhang
- School of Chemistry and Chemical Eengineering, Guangxi Minzu University, No.158, Da Xue Xi street, Xixiangtang District, Nanning, 530006, Guangxi Province, China
| | - Yuzhu Zhou
- School of Chemistry and Chemical Eengineering, Guangxi Minzu University, No.158, Da Xue Xi street, Xixiangtang District, Nanning, 530006, Guangxi Province, China
| | - Hua Zhu
- College of Pharmacy, Guangxi University for Chinese Medicine, No.13 , Wu He street, Qingxiu District, Nanning, 530200, Guangxi Province, China
| | - Tong Li
- College of Pharmacy, Guangxi University for Chinese Medicine, No.13 , Wu He street, Qingxiu District, Nanning, 530200, Guangxi Province, China
| | - Fangkai Du
- School of Chemistry and Chemical Eengineering, Guangxi Minzu University, No.158, Da Xue Xi street, Xixiangtang District, Nanning, 530006, Guangxi Province, China
| |
Collapse
|
20
|
Ion transporters: emerging agents for anticancer therapy. Sci China Chem 2022. [DOI: 10.1007/s11426-022-1258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
21
|
Lang J, Wang W, Zhou Y, Guo D, Shi R, Zhou N. Electrochemical Behavior and Direct Quantitative Determination of Paclitaxel. Front Chem 2022; 10:834154. [PMID: 35559219 PMCID: PMC9090486 DOI: 10.3389/fchem.2022.834154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
The electrochemical behavior and direct quantitative determination of paclitaxel, a poorly soluble drug made into microemulsion, were researched by cyclic voltammetry in acetate buffer solutions (pH = 4.0) at a glassy carbon electrode. The results show that the oxidation process is irreversible and controlled by diffusion. Moreover, the effects of anodic peak current (Ipa), anodic peak potential, scan rate, pH, and the electrochemical redox mechanism have been studied. The anodic peak current varied linearly with paclitaxel concentration in the range of 5 × 10-5 mol/L to 5 × 10-4 mol/L, and the detection limit was 9.15 × 10-8 mol/L. The results of RSD (0.90%) and recovery (99.22%-101.69%) were obtained. Additionally, it has been proved that one electron and one proton are involved in the electrochemical redox process. The present research has been successfully used to determine paclitaxel in pure and real samples, which further supported the electrochemical behavior investigation of paclitaxel and direct determination of micro-emulsion.
Collapse
Affiliation(s)
- Jiaqi Lang
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area’s Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Wenting Wang
- Wanzhou Institute for Food and Drug Control, Chongqing, China
| | - You Zhou
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area’s Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Dongqin Guo
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area’s Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Rujie Shi
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area’s Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Nong Zhou
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area’s Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| |
Collapse
|
22
|
Kaur M, Kaur M, Bandopadhyay T, Sharma A, Priya A, Singh A, Banerjee B. Naturally occurring, natural product inspired and synthetic heterocyclic anti-cancer drugs. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2022-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract
This chapter describes the importance and activity of a huge number of commercially available naturally occurring, natural product derived or synthetic heterocyclic anti-cancer drugs.
Collapse
Affiliation(s)
- Manmeet Kaur
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Mandeep Kaur
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Tania Bandopadhyay
- Completed MBBS from North Bengal Medical College and Hospital , Darjeeling , West Bengal , Pin-734432 , India
| | - Aditi Sharma
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Anu Priya
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Arvind Singh
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Bubun Banerjee
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| |
Collapse
|
23
|
Lohan-Codeço M, Barambo-Wagner ML, Nasciutti LE, Ribeiro Pinto LF, Meireles Da Costa N, Palumbo A. Molecular mechanisms associated with chemoresistance in esophageal cancer. Cell Mol Life Sci 2022; 79:116. [PMID: 35113247 PMCID: PMC11073146 DOI: 10.1007/s00018-022-04131-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
Abstract
Esophageal cancer (EC) is one of the most incident and lethal tumors worldwide. Although surgical resection is an important approach in EC treatment, late diagnosis, metastasis and recurrence after surgery have led to the management of adjuvant and neoadjuvant therapies over the past few decades. In this scenario, 5-fluorouracil (5-FU) and cisplatin (CISP), and more recently paclitaxel (PTX) and carboplatin (CBP), have been traditionally used in EC treatment. However, chemoresistance to these agents along EC therapeutic management represents the main obstacle to successfully treat this malignancy. In this sense, despite the fact that most of chemotherapy drugs were discovered several decades ago, in many cases, including EC, they still represent the most affordable and widely employed treatment approach for these tumors. Therefore, this review summarizes the main mechanisms through which the response to the most widely chemotherapeutic agents used in EC treatment is impaired, such as drug metabolism, apoptosis resistance, cancer stem cells (CSCs), cell cycle, autophagy, energetic metabolism deregulation, tumor microenvironment and epigenetic modifications.
Collapse
Affiliation(s)
- Matheus Lohan-Codeço
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Prédio do Centro de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, Rua César Pernetta, 1766 (LS.3.01), Rio de Janeiro, RJ, Brasil
| | - Maria Luísa Barambo-Wagner
- Programa de Carcinogênese Molecular Coordenação de Pesquisa, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6ºandar-Centro, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Luiz Eurico Nasciutti
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Prédio do Centro de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, Rua César Pernetta, 1766 (LS.3.01), Rio de Janeiro, RJ, Brasil
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular Coordenação de Pesquisa, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6ºandar-Centro, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Nathalia Meireles Da Costa
- Programa de Carcinogênese Molecular Coordenação de Pesquisa, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6ºandar-Centro, Rio de Janeiro, RJ, 20231-050, Brazil.
| | - Antonio Palumbo
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Prédio do Centro de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, Rua César Pernetta, 1766 (LS.3.01), Rio de Janeiro, RJ, Brasil.
| |
Collapse
|
24
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy-Induced Sinus Bradycardia: A Narrative Review of a Forgotten Adverse Effect of Cardiotoxicity. Drug Saf 2022; 45:101-126. [PMID: 35025085 DOI: 10.1007/s40264-021-01132-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/20/2022]
Abstract
Cardiotoxicity is a common adverse effect of anticancer drugs (ACDs), including the so-called targeted drugs, and increases morbidity and mortality in patients with cancer. Attention has focused mainly on ACD-induced heart failure, myocardial ischemia, hypertension, thromboembolism, QT prolongation, and tachyarrhythmias. Yet, although an increasing number of ACDs can produce sinus bradycardia (SB), this proarrhythmic effect remains an underappreciated complication, probably because of its low incidence and severity since most patients are asymptomatic. However, SB merits our interest because its incidence increases with the aging of the population and cancer is an age-related disease and because SB represents a risk factor for QT prolongation. Indeed, several ACDs that produce SB also prolong the QT interval. We reviewed published reports on ACD-induced SB from January 1971 to November 2020 using the PubMed and EMBASE databases. Published reports from clinical trials, case reports, and recent reviews were considered. This review describes the associations between ACDs and SB, their clinical relevance, risk factors, and possible mechanisms of onset and treatment.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain.
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| |
Collapse
|
25
|
Cabeza L, El-Hammadi MM, Ortiz R, Cayero-Otero MD, Jiménez-López J, Perazzoli G, Martin-Banderas L, Baeyens JM, Melguizo C, Prados J. Evaluation of poly (lactic-co-glycolic acid) nanoparticles to improve the therapeutic efficacy of paclitaxel in breast cancer. BIOIMPACTS : BI 2022; 12:515-531. [PMID: 36644541 PMCID: PMC9809141 DOI: 10.34172/bi.2022.23433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/10/2021] [Accepted: 02/20/2021] [Indexed: 01/18/2023]
Abstract
Introduction: Paclitaxel (PTX) is a cornerstone in the treatment of breast cancer, the most common type of cancer in women. However, this drug has serious limitations, including lack of tissue-specificity, poor water solubility, and the development of drug resistance. The transport of PTX in a polymeric nanoformulation could overcome these limitations. Methods: In this study, PLGA-PTX nanoparticles (NPs) were assayed in breast cancer cell lines, breast cancer stem cells (CSCs) and multicellular tumor spheroids (MTSs) analyzing cell cycle, cell uptake (Nile Red-NR-) and α-tubulin expression. In addition, PLGA-PTX NPs were tested in vivo using C57BL/6 mice, including a biodistribution assay. Results: PTX-PLGA NPs induced a significant decrease in the PTX IC50 of cancer cell lines (1.31 and 3.03-fold reduction in MDA-MB-231 and E0771 cells, respectively) and CSCs. In addition, MTSs treated with PTX-PLGA exhibited a more disorganized surface and significantly higher cell death rates compared to free PTX (27.9% and 16.3% less in MTSs from MCF-7 and E0771, respectively). PTX-PLGA nanoformulation preserved PTX's mechanism of action and increased its cell internalization. Interestingly, PTX-PLGA NPs not only reduced the tumor volume of treated mice but also increased the antineoplastic drug accumulation in their lungs, liver, and spleen. In addition, mice treated with PTX-loaded NPs showed blood parameters similar to the control mice, in contrast with free PTX. Conclusion: These results suggest that our PTX-PLGA NPs could be a suitable strategy for breast cancer therapy, improving antitumor drug efficiency and reducing systemic toxicity without altering its mechanism of action.
Collapse
Affiliation(s)
- Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Mazen M. El-Hammadi
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Seville, 41012 Sevilla, Spain
| | - Raul Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Maria D. Cayero-Otero
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Seville, 41012 Sevilla, Spain
| | - Julia Jiménez-López
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Lucia Martin-Banderas
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Seville, 41012 Sevilla, Spain
| | - Jose M. Baeyens
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
,Corresponding author: Consolación Melguizo,
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| |
Collapse
|
26
|
Calvo E, Sessa C, Harada G, de Miguel M, Kahatt C, Luepke-Estefan XE, Siguero M, Fernandez-Teruel C, Cullell-Young M, Stathis A, Drilon A. Phase I study of lurbinectedin in combination with weekly paclitaxel with or without bevacizumab in patients with advanced solid tumors. Invest New Drugs 2022; 40:1263-1273. [PMID: 35947247 PMCID: PMC9652263 DOI: 10.1007/s10637-022-01281-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/03/2022] [Indexed: 01/12/2023]
Abstract
Lurbinectedin and paclitaxel showed synergism in preclinical studies and have non-completely overlapping toxicity profiles. This phase I trial evaluated a combination of paclitaxel and lurbinectedin with/without bevacizumab in advanced tumors. This trial was divided into Group A, which evaluated weekly paclitaxel (60 or 80 mg) plus lurbinectedin (3.0-5.0 mg flat dose [FD] or 2.2 mg/m2) every 3 weeks in advanced solid tumors; and Group B, which evaluated bevacizumab (BEV, 15 mg/kg) added to the recommended dose (RD) defined in Group A in advanced epithelial ovarian or non-small cell lung cancer (NSCLC). 67 patients (A, n = 55; B, n = 12) were treated. The RD was paclitaxel 80 mg/m2 on Day (D)1,D8 plus lurbinectedin 2.2 mg/m2 on D1. At this RD, myelotoxicity was reversible and manageable, and most non-hematological toxicities were mild/moderate. Adding BEV did not notably change tolerability. Twenty-five confirmed responses were observed: 20/51 evaluable patients in Group A (overall response rate [ORR] = 39% at all dose levels and at the RD), and 5/10 evaluable patients in Group B (ORR = 50%). Most responders had breast (n = 7/12 patients), small cell lung (SCLC) (n = 5/7), epithelial ovarian (n = 3/9) and endometrial cancer (n = 3/11) in Group A, and epithelial ovarian (n = 3/4) and NSCLC (n = 2/6) in Group B. Clinical benefit rate was 61% in Group A (58% at the RD), and 90% in Group B. No major pharmacokinetic drug-drug interactions were observed. Paclitaxel/lurbinectedin and paclitaxel/lurbinectedin/BEV are feasible combinations. Further development is warranted of paclitaxel/lurbinectedin in SCLC, breast, and endometrial cancer, and of paclitaxel/lurbinectedin/BEV in epithelial ovarian cancer.
Collapse
Affiliation(s)
- Emiliano Calvo
- START Madrid - HM CIOCC, Hospital Madrid Norte Sanchinarro, Madrid, Spain
| | - Cristiana Sessa
- Oncology Institute of Southern Switzerland, EOC, Ospedale San Giovanni, Bellinzona, Switzerland
| | - Guilherme Harada
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY USA
| | - Maria de Miguel
- START Madrid - HM CIOCC, Hospital Madrid Norte Sanchinarro, Madrid, Spain
| | | | | | | | | | | | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Ospedale San Giovanni, Bellinzona, Switzerland
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
27
|
Determination of Paclitaxel Solubility and Stability in the Presence of Injectable Excipients. Pharm Chem J 2021. [DOI: 10.1007/s11094-021-02526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
28
|
Kang JH, Turabee MH, Lee DS, Kwon YJ, Ko YT. Temperature and pH-responsive in situ hydrogels of gelatin derivatives to prevent the reoccurrence of brain tumor. Biomed Pharmacother 2021; 143:112144. [PMID: 34509823 DOI: 10.1016/j.biopha.2021.112144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV malignant brain tumor with a median survival time of approximately 12-16 months. Because of its highly aggressive and heterogeneous nature it is very difficult to remove by surgical resection. Herein we have reported dual stimuli-responsive and biodegradable in situ hydrogels of oligosulfamethazine-grafted gelatin and loaded with anticancer drug paclitaxel (PTX) for preventing the progress of Glioblastoma. The oligosulfamethazine (OSM) introduced to the gelatin backbone for the formation of definite and stable in situ hydrogel. The hydrogels transformed from a sol to a gel state upon changes in stimuli. pH and temperature and retained a distinct shape after subcutaneous administration in BALB/c mice. The viscosity of the sol state hydrogels was tuned by varying the feed molar ratio between gelatin and OSM. The porosity of the hydrogels was confirmed to be lower in higher degree OSM by SEM. Sustained release of PTX from hydrogels in physiological environments (pH 7.4) was further retarded up to 63% in 9th days in tumor environments (pH 6.5). While the empty hydrogels were non-toxic in cultured cells, the hydrogels loaded with PTX showed antitumor efficacy in orthotopic-GBM xenograft mice. Collectively, the gelatin-OSM formed porous hydrogels and released the cargo in a sustained manner in tumor environments efficiently suppressing the progress of GBM. Thus, gelatin-OSM hydrogels are a potential candidate for the direct delivery of therapeutics to the local areas in brain diseases.
Collapse
Affiliation(s)
- Ji Hee Kang
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, 21936 Incheon, South Korea
| | - Md Hasan Turabee
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, 21936 Incheon, South Korea
| | - Doo Sung Lee
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, 16419 Suwon, South Korea
| | - Young Jik Kwon
- Department of Chemical Engineering and Materials Science, University of California, 92697 Irvine, CA, United States; Department of Molecular Biology and Biochemistry, University of California, 92697 Irvine, CA, United States; Department of Pharmaceutical Sciences, University of California, 92697 Irvine, CA, United States; Department of Biomedical Engineering, University of California, 92697 Irvine, CA, United States
| | - Young Tag Ko
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, 21936 Incheon, South Korea.
| |
Collapse
|
29
|
Qi H, Wang Y, Wang X, Su L, Wang Y, Wang S. The different interactions of two anticancer drugs with bovine serum albumin based on multi-spectrum method combined with molecular dynamics simulations. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 259:119809. [PMID: 33965887 DOI: 10.1016/j.saa.2021.119809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 06/12/2023]
Abstract
Paclitaxel is the best natural anticancer drug and artemisinin also has anticancer effect. In this study, the interactions between BSA and these two drugs were determined in PBS (pH 7.40) by multi-spectroscopic method and molecular dynamics (MD) simulations. The results showed that paclitaxel and artemisinin could statically quench the BSA fluorescence when the complexes were formed and the stoichiometric ratio of BSA-drugs was 1:1. Particularly, the BSA-paclitaxel complex was more stable than BSA-artemisinin complex. During the binding, the surroundings around Trp residue site was largely affected than Tyr site, especially Trp 214 to a more hydrophobic environment. In addition, the binding processes were mainly spontaneous through electrostatic force interaction. In summary, we concluded that the free drug of paclitaxel in blood was low and duration time of artemisinin was shorter.
Collapse
Affiliation(s)
- Haiyan Qi
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42, Wenhua Street, Qiqihar, PR China; Heilongjiang Industrial Hemp Processing Technology Innovation Center, Qiqihar University, No. 42, Wenhua Street, Qiqihar, PR China.
| | - Yan Wang
- Key Laboratory of Theoretical Chemistry of Environment Ministry of Education, School of Chemistry, South China Normal University, No. 378 Waihuan West Road, Higher Education Mega Center, Panyu District, Guangzhou 510006, PR China
| | - Xiuwen Wang
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42, Wenhua Street, Qiqihar, PR China
| | - Liqiang Su
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42, Wenhua Street, Qiqihar, PR China
| | - Ying Wang
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42, Wenhua Street, Qiqihar, PR China
| | - Shu Wang
- College of Chemistry and Chemical Engineering, Qiqihar University, No. 42, Wenhua Street, Qiqihar, PR China
| |
Collapse
|
30
|
Single-cell adhesion strength and contact density drops in the M phase of cancer cells. Sci Rep 2021; 11:18500. [PMID: 34531409 PMCID: PMC8445979 DOI: 10.1038/s41598-021-97734-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023] Open
Abstract
The high throughput, cost effective and sensitive quantification of cell adhesion strength at the single-cell level is still a challenging task. The adhesion force between tissue cells and their environment is crucial in all multicellular organisms. Integrins transmit force between the intracellular cytoskeleton and the extracellular matrix. This force is not only a mechanical interaction but a way of signal transduction as well. For instance, adhesion-dependent cells switch to an apoptotic mode in the lack of adhesion forces. Adhesion of tumor cells is a potential therapeutic target, as it is actively modulated during tissue invasion and cell release to the bloodstream resulting in metastasis. We investigated the integrin-mediated adhesion between cancer cells and their RGD (Arg-Gly-Asp) motif displaying biomimetic substratum using the HeLa cell line transfected by the Fucci fluorescent cell cycle reporter construct. We employed a computer-controlled micropipette and a high spatial resolution label-free resonant waveguide grating-based optical sensor calibrated to adhesion force and energy at the single-cell level. We found that the overall adhesion strength of single cancer cells is approximately constant in all phases except the mitotic (M) phase with a significantly lower adhesion. Single-cell evanescent field based biosensor measurements revealed that at the mitotic phase the cell material mass per unit area inside the cell-substratum contact zone is significantly less, too. Importantly, the weaker mitotic adhesion is not simply a direct consequence of the measured smaller contact area. Our results highlight these differences in the mitotic reticular adhesions and confirm that cell adhesion is a promising target of selective cancer drugs as the vast majority of normal, differentiated tissue cells do not enter the M phase and do not divide.
Collapse
|
31
|
Nguyen LD, Fischer TT, Ehrlich BE. Pharmacological rescue of cognitive function in a mouse model of chemobrain. Mol Neurodegener 2021; 16:41. [PMID: 34174909 PMCID: PMC8235868 DOI: 10.1186/s13024-021-00463-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND After chemotherapy, many cancer survivors suffer from long-lasting cognitive impairment, colloquially known as "chemobrain." However, the trajectories of cognitive changes and the underlying mechanisms remain unclear. We previously established paclitaxel-induced inositol trisphosphate receptor (InsP3R)-dependent calcium oscillations as a mechanism for peripheral neuropathy, which was prevented by lithium pretreatment. Here, we investigated if a similar mechanism also underlay paclitaxel-induced chemobrain. METHOD Mice were injected with 4 doses of 20 mg/kg paclitaxel every other day to induced cognitive impairment. Memory acquisition was assessed with the displaced object recognition test. The morphology of neurons in the prefrontal cortex and the hippocampus was analyzed using Golgi-Cox staining, followed by Sholl analyses. Changes in protein expression were measured by Western blot. RESULTS Mice receiving paclitaxel showed impaired short-term spatial memory acquisition both acutely 5 days post injection and chronically 23 days post injection. Dendritic length and complexity were reduced in the hippocampus and the prefrontal cortex after paclitaxel injection. Concurrently, the expression of protein kinase C α (PKCα), an effector in the InsP3R pathway, was increased. Treatment with lithium before or shortly after paclitaxel injection rescued the behavioral, cellular, and molecular deficits observed. Similarly, memory and morphological deficits could be rescued by pretreatment with chelerythrine, a PKC inhibitor. CONCLUSION We establish the InsP3R calcium pathway and impaired neuronal morphology as mechanisms for paclitaxel-induced cognitive impairment. Our findings suggest lithium and PKC inhibitors as candidate agents for preventing chemotherapy-induced cognitive impairment.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA.,Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06520, USA.,Present Address: Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Tom T Fischer
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA.,Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA. .,Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
32
|
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is a frequently occurring type of cancer leading loss of huge number of lives. Folic acid (FA) conjugated solid lipid nanoparticle (SLN) loaded paclitaxel (PTX) and ascorbic acid (AA) has been used as a novel approach in this study. METHODS The FA conjugated SLN were prepared by following high speed homogenization and ultrasonication methods. FA conjugated SLN were used alone and in combination to evaluate their efficacy against OSCC induced animal model. FA conjugated PTX and FA conjugated AA loaded SLN were further subjected to pharmacokinetic and biodistribution. RESULTS The FA conjugated SLN showed a biphasic drug release behavior both in in vitro as well as in vivo system. FA conjugated PTX loaded SLN and FA conjugated AA loaded SLN shows high efficiency when used in combination as compared to when used individually in vivo. FA conjugated SLN shows a better therapeutic efficacy as compared to normal drug as depicted by the observation of pharmacokinetic and biodistribution studies. CONCLUSION The in vitro and in vivo evaluation of the FA conjugated SLN concluded with a remark that, these SLN can be effectively used in the treatment of OSCC.
Collapse
Affiliation(s)
- Rituraj Bharadwaj
- Department of Bioengineering and Technology, Laboratory of Molecular Virology and Oncology, Gauhati University, Guwahati, India
| | - Subhash Medhi
- Department of Bioengineering and Technology, Laboratory of Molecular Virology and Oncology, Gauhati University, Guwahati, India
| |
Collapse
|
33
|
Belkadi A, Kenouche S, Melkemi N, Daoud I, Djebaili R. K-means clustering analysis, ADME/pharmacokinetic prediction, MEP, and molecular docking studies of potential cytotoxic agents. Struct Chem 2021. [DOI: 10.1007/s11224-021-01796-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
34
|
Schneider F, Pan L, Ottenbruch M, List T, Gaich T. The Chemistry of Nonclassical Taxane Diterpene. Acc Chem Res 2021; 54:2347-2360. [PMID: 33942612 DOI: 10.1021/acs.accounts.0c00873] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The taxane diterpenes are a pharmaceutically vital family of natural products, consisting of more than 550 congeners. All taxane diterpenes are isolated from slow growing evergreen shrubs (genus Taxus) commonly known as "yews" and have a history of over 50 years as potent anticancer compounds. The most prominent congener, taxol (paclitaxel = PTX), has been used in clinics for more than 25 years and is one of the top-selling anticancer drugs worldwide, with annual sales reaching 1.5 billion USD in 1999. Within the taxane diterpene family 11 different scaffolds originating from rearrangements, fragmentations, or transannular C-C bond formations of the "classical taxane core" are known. Among them, five different scaffolds alone belong to the so-called complex or cyclotaxane subfamily, their signature structural feature bearing different types and numbers of transannular C-C bonds across the classical taxane backbone. For synthetic chemists, these five scaffolds represent by far the most challenging of all and have thus evaded total synthesis as well as detailed pharmaceutical evaluation-the latter due to extremely poor sourcing from natural producers. The cousinship of complex taxanes to taxol renders them potentially interesting compounds for drug research in the fight against cancer.This Account specifically summarizes the work on nonclassical taxanes from a biosynthetic, as well as a synthetic, point and provides a synthetic perspective on complex taxanes. Special attention is given to the biosynthetic relationship of complex taxanes and their biological emergence from classical taxanes. The transannular C-C bond forming events in the biosynthesis leading to the five individual scaffolds within this subfamily are structured on the basis of the exact type and number of these specific C-C bond formations. Since functionalization of the classical taxane core in the "oxidase phase" of the biosynthesis precedes the formation of complex taxanes, and is in part prerequisite for these transannular cyclization events, a detailed discussion of these oxidations of the classical taxane backbone is provided. Synthetic efforts toward nonclassical taxanes are scarce in literature and are thus presented in a comprehensive manner for abeotaxanes and complex taxanes. The last part of this Account deals with a synthetic perspective on the synthesis of complex taxanes (cyclotaxanes) and how these most intricate scaffolds can potentially be obtained via a deconvolution strategy. This discussion involves in part unpublished results by our group and is based upon synthetic studies in the literature. The deconvolution strategy we advocate aims for selective fragmentations of the signature transannular C-C bonds of the most intricate scaffold represented by the natural product canataxpropellane, which has recently been synthesized by our group. This strategy represents the converse process of the biosynthesis of complex taxanes (e.g., transannular cyclizations) and is enabled and feasible due to our approach to the canataxpropellane scaffold. We show that, by following this deconvolution strategy, all five scaffolds of complex taxanes can thereby be accessed.
Collapse
Affiliation(s)
- Fabian Schneider
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| | - Lu Pan
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| | - Moritz Ottenbruch
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| | - Tatjana List
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| | - Tanja Gaich
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
35
|
Nawara HM, Afify SM, Hassan G, Zahra MH, Seno A, Seno M. Paclitaxel-Based Chemotherapy Targeting Cancer Stem Cells from Mono- to Combination Therapy. Biomedicines 2021; 9:biomedicines9050500. [PMID: 34063205 PMCID: PMC8147479 DOI: 10.3390/biomedicines9050500] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Paclitaxel (PTX) is a chemotherapeutical agent commonly used to treat several kinds of cancer. PTX is known as a microtubule-targeting agent with a primary molecular mechanism that disrupts the dynamics of microtubules and induces mitotic arrest and cell death. Simultaneously, other mechanisms have been evaluated in many studies. Since the anticancer activity of PTX was discovered, it has been used to treat many cancer patients and has become one of the most extensively used anticancer drugs. Regrettably, the resistance of cancer to PTX is considered an extensive obstacle in clinical applications and is one of the major causes of death correlated with treatment failure. Therefore, the combination of PTX with other drugs could lead to efficient therapeutic strategies. Here, we summarize the mechanisms of PTX, and the current studies focusing on PTX and review promising combinations.
Collapse
Affiliation(s)
- Hend M. Nawara
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
| | - Said M. Afify
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
- Division of Biochemistry, Chemistry Department, Faculty of Science, Menoufia University, Menoufia 32511, Egypt
| | - Ghmkin Hassan
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
- Department of Microbiology and Biochemistry, Faculty of Pharmacy, Damascus University, Damascus 10769, Syria
| | - Maram H. Zahra
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
| | - Akimasa Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
| | - Masaharu Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
- Correspondence: ; Tel.: +81-86-251-8216
| |
Collapse
|
36
|
Schneider PA, Varcoe RL, Secemsky E, Schermerhorn M, Holden A. Update on paclitaxel for femoral-popliteal occlusive disease in the 15 months following a summary level meta-analysis demonstrated increased risk of late mortality and dose response to paclitaxel. J Vasc Surg 2021; 73:311-322. [PMID: 32890719 PMCID: PMC8076887 DOI: 10.1016/j.jvs.2020.07.093] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/20/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Peripheral vascular devices (stents and balloons) coated with paclitaxel were developed to address suboptimal outcomes associated with percutaneous revascularization procedures of the femoral-popliteal arteries. In randomized controlled trials (RCT), paclitaxel-coated devices (PCD) provided increased long-term patency and a decreased need for repeat revascularization procedures compared with uncoated devices. This finding resulted in the adoption of their use for endovascular lower extremity revascularization procedures. However, in late 2018 a study-level meta-analysis showed increased all-cause mortality at 2 years or more after the procedure in patients treated with PCDs. This review examines the subsequent data evaluation following the publication of the meta-analysis. METHODS We review the published responses of physicians, regulatory agencies, and patient advocates during 15-month period after the meta-analysis. We present the additional data gathered from RCTs that comprised the meta-analysis and safety outcomes from large insurance databases in both the United States and Europe. RESULTS Immediately after the publication of the meta-analysis, concern for patient safety resulted in less PCD use, the suspension of large RCTs evaluating their use, and the publication of a letter from the U.S. Food and Drug Administration informing physicians that there was uncertainty in the benefit-risk profile of these devices for indicated patients and that the potential risk should be assessed before the use of PCDs. Review of the meta-analysis found that a mortality signal was present, but criticisms included that the evaluation was performed on study-level, not patient-level data, and the studies in the analysis were heterogenous in device type, paclitaxel doses, and patient characteristics. Further, the studies were not designed to be pooled nor were they powered for evaluating long-term safety. Clinical characteristics associated with a drug effect or causal relationship were also absent. Specifically, there was no dose response, no clustering of causes of death, and a lack of signal consistency across geographic regions. As more long-term data became available in the RCTs the strength of the mortality signal diminished and analysis of real-world use in large insurance databases, showed that there was no significant increase in all-cause mortality associated with PCD use. CONCLUSIONS The available data do not provide definitive proof for increased mortality with PCD use. A key observation is that trial design improvements will be necessary to better evaluate the risk-benefit profile of PCDs.
Collapse
Affiliation(s)
- Peter A Schneider
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, Calif.
| | - Ramon L Varcoe
- The Vascular Institute, Prince of Wales Hospital and University of New South Wales, Sydney, Australia
| | - Eric Secemsky
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Mass
| | - Marc Schermerhorn
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, Mass
| | - Andrew Holden
- Department of Interventional Radiology, Auckland Hospital and University of Auckland School of Medicine, Auckland, New Zealand
| |
Collapse
|
37
|
Li B, Xia X, Chen J, Xia D, Xu R, Zou X, Wang H, Liang C. Paclitaxel-loaded lignin particle encapsulated into electrospun PVA/PVP composite nanofiber for effective cervical cancer cell inhibition. NANOTECHNOLOGY 2021; 32:015101. [PMID: 33043894 DOI: 10.1088/1361-6528/abb55a] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Electrospun composite nanofibrous scaffolds have been regarded as a potential carrier for local drug delivery to prevent tumor recurrence. Herein, a model drug (paclitaxel) was creatively loaded into lignin nanoparticles (PLNPs) and then encapsulated into the polymer of poly (vinyl alcohol)/polyvinyl pyrrolidone which has been fabricated into a composite nanofibrous membrane (PVA/PVP-PLNPs) for use as a drug carrier using the electrospinning technique. The fabricated PVA/PVP-PLNPs membranes exhibited good particle distribution, mechanical properties, thermal stability and biocompatibility. In vitro experiments showed that combining lignin nanoparticles by electrospinning not only improved the drug release profile, but also enhanced the hydrophilicity of nanofibrous membranes which was beneficial to cell adhesion and proliferation. Cellular experiments demonstrated that PVA/PVP-2%PLNPs membrane showed good cell inhibition ability, and the cell survival rate was only 21% at day 7. It indicates that the as-prepared PVA/PVP-PLNPs composite nanofibers are promising candidates for local anticancer therapy.
Collapse
Affiliation(s)
- Baoe Li
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| | - Xiaomei Xia
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| | - Jiatian Chen
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| | - Dan Xia
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| | - Ruodan Xu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Xianrui Zou
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| | - Hongshui Wang
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| | - Chunyong Liang
- School of Materials Science and Engineering, Hebei University of Technology; Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, Tianjin 300130, People's Republic of China
| |
Collapse
|
38
|
Abstract
The tropone sesquiterpene phaeocaulisin D, isolated from the rhizomes of Curcuma phaeocaulis, has previously been shown to inhibit nitric oxide production in macrophages. A total synthesis of phaeocaulisin D was accomplished by using an intramolecular cyclization-dearomatization as a key step. The highlights of the synthesis are effective formation of the 5-7 fused tropone system, and selective methylation of a late-stage intermediate.
Collapse
Affiliation(s)
- Nameer Ezzat
- Department of Chemistry, University of Central Florida, 4111 Libra Drive, Orlando, Florida 32816, USA
- Department of Chemistry, College of Education, University of Mosul, Mosul 41002, Iraq
| | - Katelyn Bobek
- Department of Chemistry, University of Central Florida, 4111 Libra Drive, Orlando, Florida 32816, USA
| | - Yu Yuan
- Department of Chemistry, University of Central Florida, 4111 Libra Drive, Orlando, Florida 32816, USA
| |
Collapse
|
39
|
Tsolou A, Angelou E, Didaskalou S, Bikiaris D, Avgoustakis K, Agianian B, Koffa MD. Folate and Pegylated Aliphatic Polyester Nanoparticles for Targeted Anticancer Drug Delivery. Int J Nanomedicine 2020; 15:4899-4918. [PMID: 32764924 PMCID: PMC7369311 DOI: 10.2147/ijn.s244712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/09/2020] [Indexed: 01/05/2023] Open
Abstract
Purpose The use of chemotherapeutic agents to combat cancer is accompanied by high toxicity due to their inability to discriminate between cancer and normal cells. Therefore, cancer therapy research has focused on the targeted delivery of drugs to cancer cells. Here, we report an in vitro study of folate-poly(ethylene glycol)-poly(propylene succinate) nanoparticles (FA-PPSu-PEG-NPs) as a vehicle for targeted delivery of the anticancer drug paclitaxel in breast and cervical cancer cell lines. Methods Paclitaxel-loaded-FA-PPSu-PEG-NPs characterization was performed by in vitro drug release studies and cytotoxicity assays. The NPs cellular uptake and internalization mechanism were monitored by live-cell imaging in different cancer cell lines. Expression of folate receptor-α (FOLR1) was examined in these cell lines, and specific FOLR1-mediated entry of the FA-PPSu-PEG-NPs was investigated by free folic acid competition. Using inhibitors for other endocytic pathways, alternative, non-FOLR1 dependent routes for NPs uptake were also examined. Results Drug release experiments of Paclitaxel-loaded PPSu-PEG-NPs indicated a prolonged release of Paclitaxel over several days. Cytotoxicity of Paclitaxel-loaded PPSu-PEG-NPs was similar to free drug, as monitored in cancer cell lines. Live imaging of cells treated with either free Paclitaxel or Paclitaxel-loaded PPSu-PEG-NPs demonstrated tubulin-specific cell cycle arrest, with similar kinetics. Folate-conjugated NPs (FA-PPSu-PEG-NPs) targeted the FOLR1 receptor, as shown by free folic acid competition of the FA-PPSu-PEG-NPs cellular uptake in some of the cell lines tested. However, due to the differential expression of FOLR1 in the cancer cell lines, as well as the intrinsic differences between the different endocytic pathways utilized by different cell types, other mechanisms of nanoparticle cellular entry were also used, revealing that dynamin-dependent endocytosis and macropinocytosis pathways mediate, at least partially, cellular entry of the FA-PPSu-PEG NPs. Conclusion Our data provide evidence that Paclitaxel-loaded-FA-PPSu-PEG-NPs can be used for targeted delivery of the drug, FA-PPSu-PEG-NPs can be used as vehicles for other anticancer drugs and their cellular uptake is mediated through a combination of FOLR1 receptor-specific endocytosis, and macropinocytosis. The exploration of the different cellular uptake mechanisms could improve treatment efficacy or allow a decrease in dosage of anticancer drugs.
Collapse
Affiliation(s)
- Avgi Tsolou
- Laboratory of Molecular Cell Biology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Eftychia Angelou
- Biomolecular Structure and Function Group, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Stylianos Didaskalou
- Laboratory of Molecular Cell Biology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Dimitrios Bikiaris
- Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Macedonia, Greece
| | | | - Bogos Agianian
- Biomolecular Structure and Function Group, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Maria D Koffa
- Laboratory of Molecular Cell Biology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| |
Collapse
|
40
|
Bai H, Wang Z, Li M, Sun P, Wei S, Wang W, Wang Z, Xing Y, Li J, Dardik A. Inhibition of programmed death‐1 decreases neointimal hyperplasia after patch angioplasty. J Biomed Mater Res B Appl Biomater 2020; 109:269-278. [PMID: 32770622 DOI: 10.1002/jbm.b.34698] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/19/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Hualong Bai
- Department of Vascular and Endovascular Surgery First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City Zhengzhou Henan China
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Peng Sun
- Department of Vascular and Endovascular Surgery First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Shunbo Wei
- Department of Vascular and Endovascular Surgery First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Wang Wang
- Department of Physiology Medical school of Zhengzhou University Zhengzhou Henan China
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City Zhengzhou Henan China
| | - Zhiju Wang
- Department of Physiology Medical school of Zhengzhou University Zhengzhou Henan China
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City Zhengzhou Henan China
| | - Ying Xing
- Department of Physiology Medical school of Zhengzhou University Zhengzhou Henan China
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City Zhengzhou Henan China
| | - Jingan Li
- School of Material Science and Engineering and Henan Key Laboratory of Advanced Magnesium Alloy and Key Laboratory of materials processing and mold technology (Ministry of Education) Zhengzhou University Zhengzhou Henan China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program Yale School of Medicine New Haven Connecticut USA
- Departments of Surgery and of Cellular and Molecular Physiology Yale School of Medicine New Haven Connecticut USA
| |
Collapse
|
41
|
Medhi H, Khumukcham SS, Manavathi B, Paik P. Effective in vitro delivery of paclitaxel by nanocargo of mesoporous polycaprolactone against triple negative breast cancer cells by minimalizing drug dose. RSC Adv 2020; 10:24095-24107. [PMID: 35517325 PMCID: PMC9055105 DOI: 10.1039/d0ra04505e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/13/2020] [Indexed: 12/27/2022] Open
Abstract
Among the breast cancers, triple negative breast cancer (TNBC) has relatively poor outcomes with a lower survival rate and personalised chemotherapy is the only option available for treatment. Currently in the biomedical domain, nanomaterials with porous morphology have revealed their tremendous possibilities to be used as a nanocarrier in treating cancer by offering void space to encapsulate/entrap biological agents. However, the development of nanocarrier-based targeted therapy with high therapeutic efficacy and fewer side effects to normal cells is always a challenge. Here, we have developed nanocargos based on biodegradable mesoporous PCL (polycaprolactone) of approx. diameter of 75 nm by template removal synthesis techniques. Succeeding the comparative analysis of the nanocarriers, the efficiencies of core shell PCL-mZnO (PZ) and mesoporous PCL (HPZ) to deliver paclitaxel (Taxol/T) into breast cancer cells, is investigated. We found that HPZ nanocapsules have less cytotoxicity and drug loading efficiency of about 600 μg mg-1. The Taxol-loaded nanoparticles (T-HPZ) have exhibited more cytotoxicity than Taxol alone treated cancer cells. Furthermore, T-HPZ treated MDA-MB231 cells are accumulated at G2/M phase of the cell cycle and eventually undergo apoptosis. In support of this, anchorage independent growth of MDA-MB231 cells are significantly inhibited by T-HPZ treatment. Together, our findings suggest that T-HPZ-based paclitaxel (Taxol/T) loaded nanoparticles provide a novel therapeutic option in the treatment of TNBC.
Collapse
Affiliation(s)
- Himadri Medhi
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad 500046 India
| | | | - Bramanandam Manavathi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad Hyderabad 500046 India
| | - Pradip Paik
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad 500046 India
- School of Biomedical Engineering, Indian Institute of Technology, BHU Varanasi 221 005 India
| |
Collapse
|
42
|
Banskota S, Saha S, Bhattacharya J, Kirmani N, Yousefpour P, Dzuricky M, Zakharov N, Li X, Spasojevic I, Young K, Chilkoti A. Genetically Encoded Stealth Nanoparticles of a Zwitterionic Polypeptide-Paclitaxel Conjugate Have a Wider Therapeutic Window than Abraxane in Multiple Tumor Models. NANO LETTERS 2020; 20:2396-2409. [PMID: 32125864 DOI: 10.1021/acs.nanolett.9b05094] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Small-molecule therapeutics demonstrate suboptimal pharmacokinetics and bioavailability due to their hydrophobicity and size. One way to overcome these limitations-and improve their efficacy-is to use "stealth" macromolecular carriers that evade uptake by the reticuloendothelial system. Although unstructured polypeptides are of increasing interest as macromolecular drug carriers, current recombinant polypeptides in the clinical pipeline typically lack stealth properties. We address this challenge by developing new unstructured polypeptides, called zwitterionic polypeptides (ZIPPs), that exhibit "stealth" behavior in vivo. We show that conjugating paclitaxel to a ZIPP imparts amphiphilicity to the polypeptide chain that is sufficient to drive its self-assembly into micelles. This in turn increases the half-life of paclitaxel by 17-fold compared to free paclitaxel, and by 1.6-fold compared to the nonstealth control, i.e., ELP-paclitaxel. Treatment of mice bearing highly aggressive prostate or colon cancer with a single dose of ZIPP-paclitaxel nanoparticles leads to near-complete eradication of the tumor, and these nanoparticles have a wider therapeutic window than Abraxane, an FDA-approved taxane nanoformulation.
Collapse
Affiliation(s)
- Samagya Banskota
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Soumen Saha
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Jayanta Bhattacharya
- Center for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Nadia Kirmani
- Department of Biology, Trinity College of Arts and Sciences, Duke University, Durham, North Carolina 27708, United States
| | - Parisa Yousefpour
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Michael Dzuricky
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Nikita Zakharov
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Xinghai Li
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Ivan Spasojevic
- Department of Medicine, Pharmaceutical Research PK/PD Core Laboratory, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Kenneth Young
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
43
|
Ultra-thermostable RNA nanoparticles for solubilizing and high-yield loading of paclitaxel for breast cancer therapy. Nat Commun 2020; 11:972. [PMID: 32080195 PMCID: PMC7033104 DOI: 10.1038/s41467-020-14780-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 01/24/2020] [Indexed: 11/17/2022] Open
Abstract
Paclitaxel is widely used in cancer treatments, but poor water-solubility and toxicity raise serious concerns. Here we report an RNA four-way junction nanoparticle with ultra-thermodynamic stability to solubilize and load paclitaxel for targeted cancer therapy. Each RNA nanoparticle covalently loads twenty-four paclitaxel molecules as a prodrug. The RNA-paclitaxel complex is structurally rigid and stable, demonstrated by the sub-nanometer resolution imaging of cryo-EM. Using RNA nanoparticles as carriers increases the water-solubility of paclitaxel by 32,000-fold. Intravenous injections of RNA-paclitaxel nanoparticles with specific cancer-targeting ligand dramatically inhibit breast cancer growth, with nearly undetectable toxicity and immune responses in mice. No fatalities are observed at a paclitaxel dose equal to the reported LD50. The use of ultra-thermostable RNA nanoparticles to deliver chemical prodrugs addresses issues with RNA unfolding and nanoparticle dissociation after high-density drug loading. This finding provides a stable nano-platform for chemo-drug delivery as well as an efficient method to solubilize hydrophobic drugs. Although paclitaxel is widely used as a chemotherapy, it suffers from poor solubility and toxicity issues. Here, the authors develop thermostable RNA nanoparticles and report the RNA-paclitaxel complex to display improved stability, drug loading capacity and solubility for improved targeted cancer therapy and reduced immune responses.
Collapse
|
44
|
Lau MT, Ghazanfar S, Parkin A, Chou A, Rouaen JR, Littleboy JB, Nessem D, Khuong TM, Nevoltris D, Schofield P, Langley D, Christ D, Yang J, Pajic M, Neely GG. Systematic functional identification of cancer multi-drug resistance genes. Genome Biol 2020; 21:27. [PMID: 32028983 PMCID: PMC7006212 DOI: 10.1186/s13059-020-1940-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Drug resistance is a major obstacle in cancer therapy. To elucidate the genetic factors that regulate sensitivity to anti-cancer drugs, we performed CRISPR-Cas9 knockout screens for resistance to a spectrum of drugs. RESULTS In addition to known drug targets and resistance mechanisms, this study revealed novel insights into drug mechanisms of action, including cellular transporters, drug target effectors, and genes involved in target-relevant pathways. Importantly, we identified ten multi-drug resistance genes, including an uncharacterized gene C1orf115, which we named Required for Drug-induced Death 1 (RDD1). Loss of RDD1 resulted in resistance to five anti-cancer drugs. Finally, targeting RDD1 leads to chemotherapy resistance in mice and low RDD1 expression is associated with poor prognosis in multiple cancers. CONCLUSIONS Together, we provide a functional landscape of resistance mechanisms to a broad range of chemotherapeutic drugs and highlight RDD1 as a new factor controlling multi-drug resistance. This information can guide personalized therapies or instruct rational drug combinations to minimize acquisition of resistance.
Collapse
Affiliation(s)
- Man-Tat Lau
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Genome Editing Initiative, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
- The Judith and David Coffey Life Lab, Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Ashleigh Parkin
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Angela Chou
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jourdin R Rouaen
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jamie B Littleboy
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Danielle Nessem
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Thang M Khuong
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Damien Nevoltris
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Peter Schofield
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, Sydney, NSW, 2010, Australia
| | - David Langley
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, Sydney, NSW, 2010, Australia
| | - Jean Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, Sydney, NSW, 2010, Australia.
| | - G Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
- Genome Editing Initiative, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
45
|
Schneider F, Samarin K, Zanella S, Gaich T. Total synthesis of the complex taxane diterpene canataxpropellane. Science 2020; 367:676-681. [DOI: 10.1126/science.aay9173] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/14/2020] [Indexed: 11/02/2022]
Abstract
Canataxpropellane belongs to the medicinally important taxane diterpene family. The most prominent congener, Taxol, is one of the most commonly used anticancer agent in clinics today. Canataxpropellane exhibits a taxane skeleton with three additional transannular C–C bonds, resulting in a total of six contiguous quaternary carbons, of which four are located on a cyclobutane ring. Unfortunately, isolation of canataxpropellane from natural sources is inefficient. Here, we report a total synthesis of (–)-canataxpropellane in 26 steps and 0.5% overall yield from a known intermediate corresponding to 29 steps from commercial material. The core structure of the (–)-canataxpropellane (2) was assembled in two steps using a Diels–Alder/ortho-alkene-arene photocycloaddition sequence. Enantioselectivity was introduced by designing chiral siloxanes to serve as auxiliaries in the Diels–Alder reaction.
Collapse
Affiliation(s)
- Fabian Schneider
- Department of Chemistry, University of Konstanz, 78467 Konstanz, Germany
| | - Konstantin Samarin
- Department of Chemistry, University of Konstanz, 78467 Konstanz, Germany
| | - Simone Zanella
- Department of Chemistry, University of Konstanz, 78467 Konstanz, Germany
| | - Tanja Gaich
- Department of Chemistry, University of Konstanz, 78467 Konstanz, Germany
| |
Collapse
|
46
|
Paclitaxel-loaded micro or nano transfersome formulation into novel tablets for pulmonary drug delivery via nebulization. Int J Pharm 2020; 575:118919. [DOI: 10.1016/j.ijpharm.2019.118919] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 01/16/2023]
|
47
|
Abstract
Mass spectrometry (MS) is an indispensable analytical technique for bioanalysis. Based on the measurement of mass/charge ratios (m/z) of ions, MS can be used for sensitive detection and accurate identification of species of interest. In traditional studies, MS is utilized to measure analytes in prepared solutions or gas-phase samples. Benefited from recent development of sampling and ionization approaches, MS has been extensively applied to the analysis of broad ranges of biological samples. We have developed a new device, the Single-probe, that can be used for in situ, real-time MS analysis of metabolites inside individual living cells. The Single-probe is a miniaturized multifunctional sampling and ionization device that is directly coupled to the mass spectrometer. With a sampling tip size smaller than 10 μm, we can insert the Single-probe tip into single cells to extract intracellular compounds, which are analyzed using MS in real-time. We have successfully used the Single-probe MS technique to detect a variety of endogenous and exogenous cellular metabolites in individual eukaryotic cells. Single cell mass spectrometry (SCMS) is a new scientific technology that has the potential to reshape approaches in biological and pharmaceutical bioanalytical research.
Collapse
Affiliation(s)
- Ning Pan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
| | - Wei Rao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA.
| |
Collapse
|
48
|
Oliverius M, Flasarova D, Mohelnikova-Duchonova B, Ehrlichova M, Hlavac V, Kocik M, Strouhal O, Dvorak P, Ojima I, Soucek P. KRAS pathway expression changes in pancreatic cancer models by conventional and experimental taxanes. Mutagenesis 2019; 34:403-411. [PMID: 31375828 PMCID: PMC6923165 DOI: 10.1093/mutage/gez021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/06/2019] [Indexed: 12/30/2022] Open
Abstract
The KRAS signalling pathway is pivotal for pancreatic ductal adenocarcinoma (PDAC) development. After the failure of most conventional cytotoxic and targeted therapeutics tested so far, the combination of taxane nab-paclitaxel (Abraxane) with gemcitabine recently demonstrated promising improvements in the survival of PDAC patients. This study aimed to explore interactions of conventional paclitaxel and experimental taxane SB-T-1216 with the KRAS signalling pathway expression in in vivo and in vitro PDAC models in order to decipher potential predictive biomarkers or targets for future individualised therapy. Mouse PDAC PaCa-44 xenograft model was used for evaluation of changes in transcript and protein levels of the KRAS signalling pathway caused by administration of experimental taxane SB-T-1216 in vivo. Subsequently, KRAS wild-type (BxPc-3) and mutated (MiaPaCa-2 and PaCa-44) cell line models were treated with paclitaxel to verify dysregulation of the KRAS signalling pathway gene expression profile in vitro and investigate the role of KRAS mutation status. By comparing the gene expression profiles, this study observed for the first time that in vitro cell models differ in the basal transcriptional profile of the KRAS signalling pathway, but there were no differences between KRAS mutated and wild-type cells in sensitivity to taxanes. Generally, the taxane administration caused a downregulation of the KRAS signalling pathway both in vitro and in vivo, but this effect was not dependent on the KRAS mutation status. In conclusion, putative biomarkers for prediction of taxane activity or targets for stimulation of taxane anticancer effects were not discovered by the KRAS signalling pathway profiling in various PDAC models.
Collapse
Affiliation(s)
- M Oliverius
- Department of Surgery, Faculty Hospital Kralovske Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Transplantation Center, Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - D Flasarova
- Department of Oncology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - B Mohelnikova-Duchonova
- Department of Oncology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- Department of Toxicogenomics, National Institute of Public Health, Prague, Czech Republic
| | - M Ehrlichova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - V Hlavac
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Kocik
- Transplantation Center, Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - O Strouhal
- Department of Oncology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - P Dvorak
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - I Ojima
- Institute of Chemical Biology and Drug Discovery, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - P Soucek
- Department of Toxicogenomics, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
49
|
Fu Q, Sun X, Lustburg MB, Sparreboom A, Hu S. Predicting Paclitaxel Disposition in Humans With Whole-Body Physiologically-Based Pharmacokinetic Modeling. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:931-939. [PMID: 31671477 PMCID: PMC6930855 DOI: 10.1002/psp4.12472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022]
Abstract
Paclitaxel is a commonly used drug in the treatment of multiple solid tumors, including cancers of the breast, lung, and ovaries. Despite the established exposure–pharmacodynamic relationships for paclitaxel, treatment is associated with wide interindividual pharmacokinetic variability that leads to unpredictability of the agent's clinical activity and toxicity. We hypothesized that physiologically‐based modeling approaches could be employed to predict the human pharmacokinetics of paclitaxel following administration of the approved Cremophor‐based formulation (Taxol). The model was developed from tissue distribution studies performed in mice and applied to plasma concentration‐time data obtained in adult cancer patients receiving Taxol at the approved dose and schedule (175 mg/m2 by a 3‐hour intravenous infusion), taking into account interspecies differences in physiological parameters. The final model adequately captured the observed concentrations in patients and allowed prediction of paclitaxel distribution profiles in multiple target organs and can be applied to further refine the chemotherapeutic treatment with a clinically important agent.
Collapse
Affiliation(s)
- Qiang Fu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Xinxin Sun
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Maryam B Lustburg
- Department of Medical Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
50
|
Jadala C, Sathish M, Anchi P, Tokala R, Lakshmi UJ, Reddy VG, Shankaraiah N, Godugu C, Kamal A. Synthesis of Combretastatin‐A4 Carboxamidest that Mimic Sulfonyl Piperazines by a Molecular Hybridization Approach:
in vitro
Cytotoxicity Evaluation and Inhibition of Tubulin Polymerization. ChemMedChem 2019; 14:2052-2060. [DOI: 10.1002/cmdc.201900541] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/15/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Chetna Jadala
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Manda Sathish
- Medicinal Chemistry and PharmacologyCSIR-Indian Institute of Chemical Technology Hyderabad 500007 India
| | - Pratibha Anchi
- Department of Regulatory ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Ramya Tokala
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Uppu Jaya Lakshmi
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Velma Ganga Reddy
- Medicinal Chemistry and PharmacologyCSIR-Indian Institute of Chemical Technology Hyderabad 500007 India
| | - Nagula Shankaraiah
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Chandraiah Godugu
- Department of Regulatory ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Ahmed Kamal
- Medicinal Chemistry and PharmacologyCSIR-Indian Institute of Chemical Technology Hyderabad 500007 India
- School of Pharmaceutical Education and Research (SPER) Jamia Hamdard New Delhi 110062 India
| |
Collapse
|