1
|
Yan Z, Bu X, Chen H, Ren C, Li J, Wu Y, Xing J. The Stem Cell Transcription Factor OCT4 Silences Target DNA Methyltransferase 1 to Strengthen DNA Damage Response in Cisplatin-Treated Gastric Cancer Cells. Biotechnol Appl Biochem 2025. [PMID: 40433841 DOI: 10.1002/bab.2777] [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: 12/05/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025]
Abstract
OCT4 plays a crucial role in the DNA damage response(DDR) mechanism, whereas cisplatin (CDDP) acts as an anti-tumor agent by inducing DDR. This study aimed to investigate the role of OCT4 in regulating DNA methyltransferase 1 (DNMT1) in CDDP-treated gastric cancer (GC). A dual-luciferase reporter assay was performed to detect the relationship between DNMT1 and OCT4. Human GC cell lines HGC-27 and MGC-803 were transfected with siRNA-OCT4 or ov-DNMT1 to construct interfering cell lines; CDDP of 0, 2.5, 5, 10, and 20 µM was used to treat GC cell lines, respectively. As follows, γ-H2AX immunofluorescence was used to detect DDR. The protein expressions of OCT4 and DNMT1 were detected by Western blot (WB), and the effects of CDDP treatment on cell apoptosis and proliferation were assessed using CCK8, cell cloning, and flow cytometry. The IC50 of CDDP-treated GC cells was reduced by OCT4 silence but enhanced by DNMT1 overexpression. A targeted regulatory relationship exists between OCT4 and DNMT1. The expression of OCT4 and DNMT1 was increased in CDDP-treated cells, and DNMT1 was decreased in the siRNA-OCT4 group. In the CDDP-treated GC cells, DNMT1 overexpression significantly reversed the siRNA-OCT4-induced cell apoptosis, γ-H2AX upregulation, and proliferation decrease. OCT4 silence may target DNMT1 to induce DDR in GC cells to strengthen the CDDP-induced cell apoptosis and proliferation inhibition.
Collapse
Affiliation(s)
- Zhengzheng Yan
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| | - Xiaoqian Bu
- The Department of Tumor Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| | - Haixia Chen
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| | - Chongren Ren
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| | - Ji Li
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| | - Yongjie Wu
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| | - Jun Xing
- Department of Breast Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, No. 99 LongCheng Street, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Catania G, Guerriero G, Bakrin N, Pourchez J, Kaouane G, Leclerc L, Augeul L, Haegebaert R, Remaut K, Kryza D, Lollo G. Generation of continuous production of polymeric nanoparticles via microfluidics for aerosolised localised drug delivery. Int J Pharm 2025; 675:125532. [PMID: 40154816 DOI: 10.1016/j.ijpharm.2025.125532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Transferring the production of nanoparticles from laboratory batches to large-scale production for preclinical and clinical applications represents a challenge due to difficulties in scaling up formulations and lack of suitable preclinical models for testing. Here, we transpose the production of hyaluronic acid and polyarginine-based nanoparticles encapsulating the platinum-derivative dichloro(1,2 diaminocyclohexane)platinum(II), from conventional bulk method to continuous production using microfluidics. The microfluidic-based drug delivery system is then tested in a customised preclinical setup to assess its suitability for pressurised intraperitoneal aerosol chemotherapy (PIPAC), a locoregional chemotherapy used to treat peritoneal carcinomatosis. PIPAC consists of the aerosolization of drugs under pressure using laparoscopy. In our preclinical setup, two clinical aerosol devices, CapnoPen® and TOPOL®, are used in conjunction with syringe pump to achieve the clinically optimal aerosol droplet size range (25-50 μm). Aerosol droplet sizes of 38 and 64 μm are obtained at upstream pressures of 14.7 and 7.4 bar and flow rates of 0.4 and 1.1 mL/s, for CapnoPen® and TOPOL®, respectively. To study the spatial distribution of the aerosol, our preclinical setup is then coupled to an ex-vivo model (inverted porcine urinary bladder) that mimics the physiological peritoneal cavity environment. The smaller droplet size obtained with CapnoPen® provided more homogeneous aerosol distribution in the bladder cavity, crucial for maximising treatment coverage within the peritoneal cavity. Furthermore, stability studies reveal that nanoparticles maintained their physicochemical properties and anticancer activity post-aerosolization. Overall, this study provides a scalable approach for the production of platinum-derivative-loaded polymeric nanoparticles and demonstrates the suitability of this DDS for PIPAC.
Collapse
Affiliation(s)
| | - Giulia Guerriero
- University of Lyon 1, CNRS, LAGEPP UMR 5007, Villeurbanne, France
| | - Naoual Bakrin
- Department of Surgical Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
| | - Jérémie Pourchez
- École Nationale Supérieure des Mines de Saint-Etienne, Mines Saint-Etienne, INSERM, U1059 Sainbiose, Centre CIS, Université de Lyon, Université Jean Monnet, 158 Cours Fauriel, CS 62362, 42023, Saint-Etienne Cedex 2, France
| | - Ghalia Kaouane
- École Nationale Supérieure des Mines de Saint-Etienne, Mines Saint-Etienne, INSERM, U1059 Sainbiose, Centre CIS, Université de Lyon, Université Jean Monnet, 158 Cours Fauriel, CS 62362, 42023, Saint-Etienne Cedex 2, France
| | - Lara Leclerc
- École Nationale Supérieure des Mines de Saint-Etienne, Mines Saint-Etienne, INSERM, U1059 Sainbiose, Centre CIS, Université de Lyon, Université Jean Monnet, 158 Cours Fauriel, CS 62362, 42023, Saint-Etienne Cedex 2, France
| | - Lionel Augeul
- Laboratoire CarMeN-IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, University of Lyon, Bron, France
| | - Ragna Haegebaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - David Kryza
- University of Lyon 1, CNRS, LAGEPP UMR 5007, Villeurbanne, France; Imthernat Plateform, Centre Léon Bérard, Lyon, France
| | - Giovanna Lollo
- University of Lyon 1, CNRS, LAGEPP UMR 5007, Villeurbanne, France; Institut universitaire de FranceFrance (IUF), Paris, France.
| |
Collapse
|
3
|
He L, Liu B, Wang Z, Han Q, Chen H. Evolving Landscape of HER2-Targeted Therapies for Gastric Cancer Patients. Curr Treat Options Oncol 2025; 26:260-277. [PMID: 40056280 DOI: 10.1007/s11864-025-01300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/10/2025]
Abstract
OPINION STATEMENT Gastric cancer (GC) is a deadly disease worldwide, and trastuzumab in combination with chemotherapy has been the standard first-line treatment for HER2-positive GC following the TOGA trial. Besides adjuvant therapy, HER2-directed therapy is widely used as neoadjuvant or translational therapy, and survival benefit even surgical opportunities is seen in these patients. However, resistance is not rare in recent years, and the second-line treatment for trastuzumab beyond progression has received widespread attention in GC. Moreover, current evidence cannot recommend trastuzumab for patients with IHC1+ HER2 low expression GC yet. Researchers are currently investigating whether GC patients with low HER2 expression could also benefit from HER2-directed therapies. In addition to using HER2 as a target for targeted therapy, HER2-mediated targeted delivery of cytotoxic drugs and targeted immunity have made important contributions to overcoming trastuzumab resistance in recent trials. HER2/neu-derived peptide epitopes vaccination and HER2-specific chimeric antigen receptor (CAR) therapy focus on reestablishing anti-tumor immunity in different ways and show significant anti-tumor activity. Other antibodies that target different regions of the HER2 receptor or block key downstream pathways such as AKT or PI3K also offer potential anti-tumor activity against HER2. HER2 use in GC will not be hampered by resistance or low expression and will play a bigger role. We review the current efforts to enable GC patients with trastuzumab-resistant and HER2 low-expressing accessible to HER2 targeted therapy and present our consideration for future HER2 in GC.
Collapse
Affiliation(s)
- Lijuan He
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Ben Liu
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zhuanfang Wang
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qinying Han
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou, 730030, China.
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China.
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, 730030, China.
- Humanized animal model laboratory, Lanzhou University Second Hospital, Lanzhou, 730030, China.
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| |
Collapse
|
4
|
Zhang Y, Yang Y, Zhou J, Yu Q, Chen L, Zhao L, Meng Y, Wang J, Yan L, Huang Z, Song S, Bai W, Sun R, Yang X. Patient-derived tumor xenograft animal model of gastric cancer in precision chemotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03903-8. [PMID: 39969603 DOI: 10.1007/s00210-025-03903-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025]
Abstract
This study aims to establish a patient-derived tumor xenograft (PDX) model for gastric cancer and apply it in pharmacodynamic studies for chemotherapeutic agents. So as to provide guidance for personalized treatment in gastric cancer patients. Fresh gastric cancer tissues were collected from surgical patients and transplanted into NOG mice to establish PDX model. Human source markers were identified by immunofluorescence and immunohistochemistry methods. Exon sequencing was used to verify the retention of humanized features in the PDX model. The efficacy of six chemotherapeutic drugs (oxaliplatin, cisplatin, paclitaxel, fluorouracil, Tegafur, capecitabine) and optimal dose of capecitabine were evaluated by the PDX model. The study has successfully established the PDX model of gastric cancer with transplantation success rates of P0 to P1 (45%), P1 to P2 (88.89%), and P2 to P3 (87.5%). Importantly, original tumor features were effectively preserved till P3. Drug screening results revealed notable antitumor effect of capecitabine in the PDX model of gastric cancer. The study has successfully established a gastric cancer PDX model, highlighting its potential for chemotherapy drug screening and personalized treatment. The transplantation success rates and preservation of original tumor features underscore the model's reliability and relevance for future studies. Findings from drug screening, especially capecitabine's effectiveness, suggest a promising avenue for precision treatment strategies in gastric cancer patients.
Collapse
Affiliation(s)
- Yiyin Zhang
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Yongming Yang
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Jiayi Zhou
- Arts and Science Faculty, Master of Media Practice, University of Sydney, Sydney, NSW, Australia
| | - Qianqian Yu
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Lixia Chen
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Lili Zhao
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Yongsheng Meng
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Jing Wang
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Lei Yan
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Ziyang Huang
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Shuchen Song
- Rehabilitation Therapeutics, The First Clinical School of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wenqi Bai
- Department of Colorectal Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China.
| | - Ruifang Sun
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China.
| | - Xihua Yang
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China.
| |
Collapse
|
5
|
Zhou X, Han J, Zuo A, Ba Y, Liu S, Xu H, Zhang Y, Weng S, Zhou Z, Liu L, Luo P, Cheng Q, Zhang C, Chen Y, Shan D, Liu B, Yang S, Han X, Deng J, Liu Z. THBS2 + cancer-associated fibroblasts promote EMT leading to oxaliplatin resistance via COL8A1-mediated PI3K/AKT activation in colorectal cancer. Mol Cancer 2024; 23:282. [PMID: 39732719 DOI: 10.1186/s12943-024-02180-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/20/2024] [Indexed: 12/30/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) exert multiple tumor-promoting functions and are key contributors to drug resistance. The mechanisms by which specific subsets of CAFs facilitate oxaliplatin resistance in colorectal cancer (CRC) have not been fully explored. This study found that THBS2 is positively associated with CAF activation, epithelial-mesenchymal transition (EMT), and chemoresistance at the pan-cancer level. Together with single-cell RNA sequencing and spatial transcriptomics analyses, we identified THBS2 specifically derived from subsets of CAFs, termed THBS2 + CAFs, which could promote oxaliplatin resistance by interacting with malignant cells via the collagen pathway in CRC. Mechanistically, COL8A1 specifically secreted from THBS2 + CAFs directly interacts with the ITGB1 receptor on resistant malignant cells, activating the PI3K-AKT signaling pathway and promoting EMT, ultimately leading to oxaliplatin resistance in CRC. Moreover, elevated COL8A1 promotes EMT and contributes to CRC oxaliplatin resistance, which can be mitigated by ITGB1 knockdown or AKT inhibitor. Collectively, these results highlight the crucial role of THBS2 + CAFs in promoting oxaliplatin resistance of CRC by activating EMT and provide a rationale for a novel strategy to overcome oxaliplatin resistance in CRC.
Collapse
Affiliation(s)
- Xing Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiashu Han
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, 100020, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Long Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chuhan Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yukang Chen
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Dan Shan
- Clinical Science Institute, University Hospital Galway, Galway, Ireland
| | - Benyu Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer Research, Comprehensive Cancer Centre, Kings College London, London, UK.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
6
|
Jaeger S, Lanquaye H, Dwivedi SK, Arachchige DL, Xia J, Waters M, Bigari BL, Olowolagba AM, Agyemang P, Zhang Y, Zhang Y, Ata A, Kathuria I, Luck RL, Werner T, Liu H. Near-Infrared Visualization of NAD(P)H Dynamics in Live Cells and Drosophila melanogaster Larvae Using a Coumarin-Based Pyridinium Fluorescent Probe. ACS APPLIED BIO MATERIALS 2024; 7:8465-8478. [PMID: 39562316 PMCID: PMC11792162 DOI: 10.1021/acsabm.4c01294] [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: 11/21/2024]
Abstract
A near-infrared fluorescent probe, A, was designed by substituting the carbonyl group of the coumarin dye's lactone with a 4-cyano-1-methylpyridinium methylene group and then attaching an electron-withdrawing NADH-sensing methylquinolinium acceptor via a vinyl bond linkage to the coumarin dye at the 4-position. The probe exhibits primary absorption maxima at 603, 428, and 361 nm, and fluoresces weakly at 703 nm. The addition of NAD(P)H results in a significant blue shift in the fluorescence peak from 703 to 670 nm, accompanied by a substantial increase in fluorescence intensity. This spectral shift is attributed to the transformation from an A-π-A-π-D configuration to a D-π-A-π-D pyridinium platform in probe AH, owing to the addition of a hydride from NADH to the electron-accepting quinolinium acceptor producing the electron-contributing 1-methyl-1,4-dihydroquinoline donor in probe AH. This conclusion is supported by theoretical calculations. The probe was utilized to investigate NAD(P)H dynamics under various conditions. In HeLa cells, treatment with glucose or maltose resulted in a substantial elevation in near-infrared emission intensity, suggesting increased NAD(P)H levels. Chemotherapeutic agents including cisplatin and fludarabine at concentrations of 5, 10, and 20 μM brought about a dose-dependent increase in emission intensity, reflecting heightened NAD(P)H levels due to drug-induced stress and cellular damage. In vivo experiments with hatched, starved Drosophila melanogaster larvae were also conducted. The results showed a clear relationship between emission intensity and the levels of NADH, glucose, and oxaliplatin, confirming that the probe can detect variations in NAD(P)H levels in a living organism. Our investigation also demonstrates that NAD(P)H levels are significantly elevated in the cystic kidneys of ADPKD mouse models and human patients, indicating substantial metabolic alterations associated with the disease. This near-infrared emissive probe offers a highly sensitive and specific method for monitoring NAD(P)H levels across cellular, tissue and whole-organism systems. The ability to detect NAD(P)H variations in reaction to varying stimuli, including nutrient availability and chemotherapeutic stress, underscores its potential as a valuable resource for biomedical research and therapeutic monitoring.
Collapse
Affiliation(s)
- Sophia Jaeger
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Henry Lanquaye
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Sushil K Dwivedi
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Dilka Liyana Arachchige
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - James Xia
- Woodbury high school, 2665 Woodlane Drive, Woodbury, Minnesota 55125, United States
| | - May Waters
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Bella Lyn Bigari
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Adenike Mary Olowolagba
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Peter Agyemang
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Yang Zhang
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Yan Zhang
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Athar Ata
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Ishana Kathuria
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Rudy L Luck
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Thomas Werner
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Haiying Liu
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
7
|
Pigg HC, Alley KR, Griffin CR, Moon CH, Kraske SJ, DeRose VJ. The unique Pt(II)-induced nucleolar stress response and its deviation from DNA damage response pathways. J Biol Chem 2024; 300:107858. [PMID: 39374783 PMCID: PMC11612370 DOI: 10.1016/j.jbc.2024.107858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 10/09/2024] Open
Abstract
The mechanisms of action for the platinum compounds cisplatin and oxaliplatin have yet to be fully elucidated, despite the worldwide use of these drugs. Recent studies suggest that the two compounds may be working through different mechanisms, with cisplatin inducing cell death via the DNA damage response (DDR) and oxaliplatin utilizing a nucleolar stress-based cell death pathway. While cisplatin-induced DDR has been subject to much research, the mechanisms for oxaliplatin's influence on the nucleolus are not well understood. Prior work has outlined structural parameters for Pt(II) derivatives capable of nucleolar stress induction. In this work, we gain insight into the nucleolar stress response induced by these Pt(II) derivatives by investigating potential correlations between this unique pathway and DDR. Key findings from this study indicate that Pt(II)-induced nucleolar stress occurs when DDR is inhibited and works independently of the ATM/ATR-dependent DDR pathway. We also determine that Pt(II)-induced stress may be linked to the G1 cell cycle phase, as cisplatin can induce nucleolar stress when cell cycle inhibition occurs at the G1/S checkpoint. Finally, we compare Pt(II)-induced nucleolar stress with other small-molecule nucleolar stress-inducing compounds Actinomycin D, BMH-21, and CX-5461 and find that Pt(II) compounds cause irreversible nucleolar stress, whereas the reversibility of nucleolar stress induced by small-molecules varies. Taken together, these findings contribute to a better understanding of Pt(II)-induced nucleolar stress, its deviation from ATM/ATR-dependent DDR, and the possible influence of cell cycle on the ability of Pt(II) compounds to cause nucleolar stress.
Collapse
Affiliation(s)
- Hannah C Pigg
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Katelyn R Alley
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | | | - Caleb H Moon
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Sarah J Kraske
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Victoria J DeRose
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA.
| |
Collapse
|
8
|
Wang P, Fares M, Eladwy RA, Bhuyan DJ, Wu X, Lewis W, Loeb SJ, Macreadie LK, Gale PA. Platinum-based metal complexes as chloride transporters that trigger apoptosis. Chem Sci 2024; 15:11584-11593. [PMID: 39055016 PMCID: PMC11268493 DOI: 10.1039/d4sc02115k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
In this paper we demonstrate that Pt(ii) complexes can function as efficient transmembrane chloride transporters. A series of Pt(ii) metal complexes with urea-appended isoquinoline ligands were synthesised and operate via classical hydrogen bonding interactions rather than ligand exchange. A number of the complexes exhibited potent transmembrane chloride activity in vesicle studies, while also showing strong antiproliferative activity in cisplatin-resistant cell lines via induction of apoptosis and inhibition of intracellular ROS.
Collapse
Affiliation(s)
- Patrick Wang
- School of Chemistry, The University of Sydney NSW 2006 Australia
| | - Mohamed Fares
- School of Pharmacy, The University of Sydney NSW 2006 Australia
| | - Radwa A Eladwy
- NICM, Research Health Institute, Western Sydney University NSW 2751 Australia
| | - Deep J Bhuyan
- NICM, Research Health Institute, Western Sydney University NSW 2751 Australia
| | - Xin Wu
- School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 Fujian China
| | - William Lewis
- School of Chemistry, The University of Sydney NSW 2006 Australia
| | - Stephen J Loeb
- Department of Chemistry and Biochemistry, University of Windsor Ontario N9B 3P4 Canada
| | | | - Philip A Gale
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney Ultimo NSW 2007 Australia
| |
Collapse
|
9
|
Arias-Martinez A, Martínez de Castro E, Gallego J, Arrazubi V, Custodio A, Fernández Montes A, Diez M, Hernandez R, Limón ML, Cano JM, Vidal-Tocino R, Macias I, Visa L, Martin Richard M, Sauri T, Hierro C, Gil M, Cerda P, Martínez Moreno E, Martínez Lago N, Mérida-García AJ, Gómez González L, García Navalón FJ, Ruiz Martín M, Marín G, López-López F, Ruperez Blanco AB, Fernández AF, Jimenez-Fonseca P, Carmona-Bayonas A, Alvarez-Manceñido F. Is there a preferred platinum and fluoropyrimidine regimen for advanced HER2-negative esophagogastric adenocarcinoma? Insights from 1293 patients in AGAMENON-SEOM registry. Clin Transl Oncol 2024; 26:1674-1686. [PMID: 38361134 PMCID: PMC11178610 DOI: 10.1007/s12094-024-03388-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/06/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND The optimal chemotherapy backbone for HER2-negative advanced esophagogastric cancer, either in combination with targeted therapies or as a comparator in clinical trials, is uncertain. The subtle yet crucial differences in platinum-based regimens' safety and synergy with combination treatments need consideration. METHODS We analyzed cases from the AGAMENON-SEOM Spanish registry of HER2-negative advanced esophagogastric adenocarcinoma treated with platinum and fluoropyrimidine from 2008 to 2021. This study focused exclusively on patients receiving one of the four regimens: FOLFOX (5-FU and oxaliplatin), CAPOX (capecitabine and oxaliplatin), CP (capecitabine and cisplatin) and FP (5-FU and cisplatin). The aim was to determine the most effective and tolerable platinum and fluoropyrimidine-based chemotherapy regimen and to identify any prognostic factors. RESULTS Among 1293 patients, 36% received either FOLFOX (n = 468) or CAPOX (n = 466), 20% CP (n = 252), and 8% FP (n = 107). FOLFOX significantly increased PFS (progression free survival) compared to CP, with a hazard ratio of 0.73 (95% CI 0.58-0.92, p = 0.009). The duration of treatment was similar across all groups. Survival outcomes among regimens were similar, but analysis revealed worse ECOG-PS (Eastern Cooperative Oncology Group-Performance Status), > 2 metastatic sites, bone metastases, hypoalbuminemia, higher NLR (neutrophil-to-lymphocyte ratio), and CP regimen as predictors of poor PFS. Fatigue was common in all treatments, with the highest incidence in FOLFOX (77%), followed by FP (72%), CAPOX (68%), and CP (60%). Other notable toxicities included neuropathy (FOLFOX 69%, CAPOX 62%), neutropenia (FOLFOX 52%, FP 55%), hand-foot syndrome in CP (46%), and thromboembolic events (FP 12%, CP 11%). CONCLUSIONS FOLFOX shown better PFS than CP. Adverse effects varied: neuropathy was more common with oxaliplatin, while thromboembolism was more frequent with cisplatin.
Collapse
Affiliation(s)
- Aranzazu Arias-Martinez
- Doctoral Program in Pharmacy, Universidad de Granada, Barrio Verxeles n°13 2°, CP 27850, Granada, Viveiro, Spain.
| | - Eva Martínez de Castro
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Javier Gallego
- Medical Oncology Department, Hospital General Universitario de Elche, Elche, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Ana Custodio
- Medical Oncology Department, Hospital Universitario La Paz, CIBERONC, CB16/12/00398, Madrid, Spain
| | - Ana Fernández Montes
- Medical Oncology Department, Complejo Hospitalario Universitario de Orense, Orense, Spain
| | - Marc Diez
- Medical Oncology Department, Hospital Universitario Vall d'Hebron, VHIO, Barcelona, Spain
| | - Raquel Hernandez
- Medical Oncology Department, Hospital Universitario de Canarias, Tenerife, Spain
| | - María Luisa Limón
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Juana María Cano
- Medical Oncology Department, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Rosario Vidal-Tocino
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca - IBSAL, Salamanca, Spain
| | - Ismael Macias
- Medical Oncology Department, Hospital Universitario Parc Tauli, Sabadell, Spain
| | - Laura Visa
- Medical Oncology Department, Hospital Universitario El Mar, Barcelona, Spain
| | - Marta Martin Richard
- Medical Oncology Department, Instituto Catalán de Oncología (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Tamara Sauri
- Medical Oncology Department, Hospital Clinic, Barcelona, Spain
| | - Cinta Hierro
- Medical Oncology Department, Instituto Catalán de Oncología (ICO)-Badalona, Barcelona; Badalona-Applied Research Group in Oncology (B-ARGO), Badalona, Spain
| | - Mireia Gil
- Medical Oncology Department, Hospital General Universitario de Valencia-Ciberonc CB16/12/0035, Valencia, Spain
| | - Paula Cerda
- Medical Oncology Department, Hospital Universitario Santa Creu y Sant Pau, Barcelona, Spain
| | - Elia Martínez Moreno
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Nieves Martínez Lago
- Medical Oncology Department, Complejo Hospitalario Universitario de Ferrol, Ferrol, Spain
| | | | - Lucía Gómez González
- Medical Oncology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | | | - Maribel Ruiz Martín
- Medical Oncology Department, Complejo Asistencial Universitario de Palencia, Palencia, Spain
| | - Gema Marín
- Medical Oncology Department, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Flora López-López
- Medical Oncology Department, Hospital Universitario del Sureste, Madrid, Spain
| | | | | | - Paula Jimenez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Alberto Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, University of Murcia, IMIB, Murcia, Spain
| | | |
Collapse
|
10
|
Yu Z, Deng P, Chen Y, Lin D, Liu S, Hong J, Guan P, Chen J, Zhong ME, Chen J, Chen X, Sun Y, Wang Y, Wang P, Cai Z, Chan JY, Huang Y, Xiao R, Guo Y, Zeng X, Wang W, Zou Y, Yu Q, Lan P, Teh BT, Wu X, Tan J. Pharmacological modulation of RB1 activity mitigates resistance to neoadjuvant chemotherapy in locally advanced rectal cancer. Proc Natl Acad Sci U S A 2024; 121:e2304619121. [PMID: 38289962 PMCID: PMC10861914 DOI: 10.1073/pnas.2304619121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2024] Open
Abstract
Resistance to neoadjuvant chemotherapy leads to poor prognosis of locally advanced rectal cancer (LARC), representing an unmet clinical need that demands further exploration of therapeutic strategies to improve clinical outcomes. Here, we identified a noncanonical role of RB1 for modulating chromatin activity that contributes to oxaliplatin resistance in colorectal cancer (CRC). We demonstrate that oxaliplatin induces RB1 phosphorylation, which is associated with the resistance to neoadjuvant oxaliplatin-based chemotherapy in LARC. Inhibition of RB1 phosphorylation by CDK4/6 inhibitor results in vulnerability to oxaliplatin in both intrinsic and acquired chemoresistant CRC. Mechanistically, we show that RB1 modulates chromatin activity through the TEAD4/HDAC1 complex to epigenetically suppress the expression of DNA repair genes. Antagonizing RB1 phosphorylation through CDK4/6 inhibition enforces RB1/TEAD4/HDAC1 repressor activity, leading to DNA repair defects, thus sensitizing oxaliplatin treatment in LARC. Our study identifies a RB1 function in regulating chromatin activity through TEAD4/HDAC1. It also provides the combination of CDK4/6 inhibitor with oxaliplatin as a potential synthetic lethality strategy to mitigate oxaliplatin resistance in LARC, whereby phosphorylated RB1/TEAD4 can serve as potential biomarkers to guide the patient stratification.
Collapse
Affiliation(s)
- Zhaoliang Yu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Peng Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yufeng Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Dezheng Lin
- Department of Endoscopic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510060, People’s Republic of China
| | - Shini Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Jinghan Hong
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
| | - Peiyong Guan
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore138672, Singapore
| | - Jianfeng Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Min-er Zhong
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jinghong Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Xiaochuan Chen
- Department of Obstetrics and Gynecology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Yichen Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yali Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Peili Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Zerong Cai
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jason Yongsheng Chan
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| | - Yulin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Rong Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yaoyu Guo
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Xian Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Wenyu Wang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Yifeng Zou
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Qiang Yu
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore138672, Singapore
| | - Ping Lan
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Bin Tean Teh
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| | - Xiaojian Wu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jing Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| |
Collapse
|
11
|
Arachchige DL, Dwivedi SK, Waters M, Jaeger S, Peters J, Tucker DR, Geborkoff M, Werner T, Luck RL, Godugu B, Liu H. Sensitive monitoring of NAD(P)H levels within cancer cells using mitochondria-targeted near-infrared cyanine dyes with optimized electron-withdrawing acceptors. J Mater Chem B 2024; 12:448-465. [PMID: 38063074 PMCID: PMC10918806 DOI: 10.1039/d3tb02124f] [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: 01/05/2024]
Abstract
A series of near-infrared fluorescent probes, labeled A to E, were developed by combining electron-rich thiophene and 3,4-ethylenedioxythiophene bridges with 3-quinolinium and various electron deficient groups, enabling the sensing of NAD(P)H. Probes A and B exhibit absorptions and emissions in the near-infrared range, offering advantages such as minimal interference from autofluorescence, negligible photo impairment in cells and tissues, and exceptional tissue penetration. These probes show negligible fluorescence when NADH is not present, and their absorption maxima are at 438 nm and 470 nm, respectively. In contrast, probes C-E feature absorption maxima at 450, 334 and 581 nm, respectively. Added NADH triggers the transformation of the electron-deficient 3-quinolinium units into electron-rich 1,4-dihydroquinoline units resulting in fluorescence responses which were established at 748, 730, 575, 625 and 661 for probes AH-EH, respectively, at detection limits of 0.15 μM and 0.07 μM for probes A and B, respectively. Optimized geometries based on theoretical calculations reveal non-planar geometries for probes A-E due to twisting of the 3-quinolinium and benzothiazolium units bonded to the central thiophene group, which all attain planarity upon addition of hydride resulting in absorption and fluorescence in the near-IR region for probes AH and BH in contrast to probes CH-EH which depict fluorescence in the visible range. Probe A has been successfully employed to monitor NAD(P)H levels in glycolysis and specific mitochondrial targeting. Furthermore, it has been used to assess the influence of lactate and pyruvate on the levels of NAD(P)H, to explore how hypoxia in cancer cells can elevate levels of NAD(P)H, and to visualize changes in levels of NAD(P)H under hypoxic conditions with CoCl2 treatment. Additionally, probe A has facilitated the examination of the potential impact of chemotherapy drugs, namely gemcitabine, camptothecin, and cisplatin, on metabolic processes and energy generation within cancer cells by affecting NAD(P)H levels. Treatment of A549 cancer cells with these drugs has been shown to increase NAD(P)H levels, which may contribute to their anticancer effects ultimately leading to programmed cell death or apoptosis. Moreover, probe A has been successfully employed in monitoring NAD(P)H level changes in D. melanogaster larvae treated with cisplatin.
Collapse
Affiliation(s)
- Dilka Liyana Arachchige
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Sushil K Dwivedi
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - May Waters
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Sophia Jaeger
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Joe Peters
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Daniel R Tucker
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
| | - Micaela Geborkoff
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA
| | - Thomas Werner
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA
| | - Rudy L Luck
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
| | - Bhaskar Godugu
- Department of Chemistry, University of Pittsburgh, Chevron Science Center, 219 Parkman Avenue, Pittsburgh, PA 15260, USA
| | - Haiying Liu
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
12
|
Ali MD, Al-Matouq JA, Ahmad A, Patel M, Alshnbari AS, Alhussini SH, Almozien SF, Alowaywi GA, Kannan LS. Survival after Trastuzumab Therapy in Patients with Locally Advanced or Metastatic HER2-Positive Gastric or Gastroesophageal Junction Cancer: A Meta-Analysis of Randomized Controlled Trials. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:24-30. [PMID: 38694962 PMCID: PMC11060624 DOI: 10.4103/jpbs.jpbs_563_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 05/04/2024] Open
Abstract
Background In the Trastuzumab for Gastric Cancer study, it was found that trastuzumab combined with doublet chemotherapy (fluoropyrimidine and platinum) was the gold-standard treatment for gastroesophageal adenocarcinoma (GEA) that was locally advanced, unresectable, or metastatic (HER2+). Materials and Methods We performed a meta-analysis of randomized phase II/III studies testing trastuzumab in combination or alone. Results This meta-analysis's findings involved 2048 patients in total. The treatment arm and hormone receptor status were used to stratify the combined HR. Overall, the PFS (Random model) HR [0.80] and 95% confidence intervals (CI) [0.68-0.95] were significantly higher for regimens containing trastuzumab, fluoropyrimidine, and platinum compared to regimens containing fluoropyrimidine and platinum. Conclusions The results of this meta-analysis provide additional support for trastuzumab's use in treating HER2-positive GEA, particularly in cases where the disease lacks a HER2+ receptor.
Collapse
Affiliation(s)
- Mohammad D. Ali
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Jenan A. Al-Matouq
- Department of Clinical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Ayaz Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Munfis Patel
- Department of Clinical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Afnan S. Alshnbari
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Sara H. Alhussini
- Department of Respiratory Care, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Sara F. Almozien
- Department of Respiratory Care, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Ghadeer A. Alowaywi
- Department of Respiratory Care, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| | - Latha S. Kannan
- Department of Nursing, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam, Saudi Arabia
| |
Collapse
|
13
|
Rasouli N, Shahbazi-Gahrouei D, Hematti S, Baradaran B, Salehi R, Varshosaz J, Jafarizad A. Assessment of Oxaliplatin-Loaded Iodine Nanoparticles for Chemoradiotherapy of Human Colorectal Cancer (HT-29) Cells. Polymers (Basel) 2022; 14:4131. [PMID: 36236079 PMCID: PMC9572447 DOI: 10.3390/polym14194131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022] Open
Abstract
Colorectal cancer is highly prevalent worldwide and has significant morbidity and mortality in humans. High-atomic-number nanoparticles such as iodine can act as X-rays absorbers to increase the local dose. The synthesis and fabrication of oxaliplatin-loaded iodine nanoparticles, their characterization, cell toxicity, radiosensitivity, cell apoptosis, and cell cycle assay in human colorectal cancer (HT-29) cells are investigated. Results show that the synthesis of a new iodine nanoparticle, polymerized triiodobenzene coated with chitosan and combined with oxaliplatin as a chemotherapeutic drug, performed well in vitro in an intracellular radiosensitizer as chemoradiotherapy agent in HT-29 cell lines. Findings also show that the INPs alone have no impact on cell cycle development and apoptosis. In contrast, oxaliplatin-loaded INPs along with 2 and 6 MV radiation doses produced more apoptosis. The interaction of INPs with mega-voltage photon energies is the cause of a major radiosensitization enhancement in comparison to radiation alone. Furthermore, results show that INPs may work as radiosensitization nanoprobe agents in the treatment of HT-29 cells due to their effect on increasing radiation dose absorption. Overall, iodine nanoparticles may be used in the treatment of colorectal cancers in clinical studies.
Collapse
Affiliation(s)
- Naser Rasouli
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Daryoush Shahbazi-Gahrouei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Simin Hematti
- Department of Radiooncology, School of Medicine, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran
| | - Roya Salehi
- Drug Applied Research Center, Department of Medical Nanotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Abbas Jafarizad
- Department of Chemical Engineering, Sahand University of Technology, Tabriz 5165665931, Iran
| |
Collapse
|
14
|
Oxaliplatin Induces Immunogenic Cell Death in Human and Murine Laryngeal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:3760766. [PMID: 36131787 PMCID: PMC9484908 DOI: 10.1155/2022/3760766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022]
Abstract
Background Cisplatin resistance is observed in patients with laryngeal cancer. The present study was designed to explore the efficacy of oxaliplatin on laryngeal cancer and elucidate the underlying mechanisms. Methods Cell viability was determined by using MTT assays. Cell apoptosis was determined by using annexin V and propidium iodide (PI) staining. Flow cytometry and immunofluorescence were applied to determine the levels of calreticulin (CALR) and DiD (1,1-dioctadecyl-3,3,3,3-tetramethylindodicarbocyanine). Flow cytometry was applied to analyze the levels of CD83, CD86, IFN-γ-producing CD8+ T cells, and CD4+CD25+FoxP3+ Tregs. The levels of adenosine triphosphate (ATP) were determined by using a chemiluminescent ATP kit and cytokines were determined by using specific enzyme-linked immunosorbent assays (ELISAs). The levels of HMGB1 were determined by using Western blot and ELISA, respectively. The xenograft animal model was constructed to evaluate the antitumor effects of oxaliplatin. Results Oxaliplatin inhibited cell growth, promoted cell apoptosis, and induced the levels of CALR, ATP, and high mobility group box protein 1 (HMGB1) in Hep-2 cells. Oxaliplatin-treated Hep-2 cells increased the intensity of DiD and the levels of CD83 and CD86 in dendritic cells (DCs), as well as induced the supernatant IL-6 and TNF-α. Oxaliplatin-treated primary laryngeal cancer cell-pulsed DCs increased the IFN-γ-producing CD8+ T cells and suppressed CD4+CD25+FoxP3+ Tregs. In vivo data showed that oxaliplatin suppressed tumor growth and increased the populations of CD86+CD80+ and CD8+CD45+ cells in the tumor tissues. Conclusion Treatment with oxaliplatin inhibited laryngeal cancer cells by inducing immunogenic cell death.
Collapse
|
15
|
Liu P, Chen J, Zhao L, Hollebecque A, Kepp O, Zitvogel L, Kroemer G. PD-1 blockade synergizes with oxaliplatin-based, but not cisplatin-based, chemotherapy of gastric cancer. Oncoimmunology 2022; 11:2093518. [PMID: 35769948 PMCID: PMC9235886 DOI: 10.1080/2162402x.2022.2093518] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Preclinical experimentation revealed that established cancers treated with the immunogenic cell death (ICD) inducer oxaliplatin are sensitized to immune checkpoint inhibitors targeting PD-1. In contrast, no such sensitizing effect is observed when cisplatin, a non-immunogenic cell death inducer is used. Two randomized phase III clinical trials targeting unresectable gastric and gastro-esophageal junction carcinomas apparently validate this observation. Thus, oxaliplatin-based chemotherapy (together with capecitabine or 5-fluorouracil plus leucovorin) favorably interacted with nivolumab, yielding improved outcome. In contrast, the outcome of cisplatin-based chemotherapy (together with capecitabine or 5-fluorouracil) failed to be improved by concomitant treatment with pembrolizumab. These clinical findings underscore the importance of choosing appropriate ICD-inducing cytotoxicants for the development of chemoimmunotherapeutic regimens. Unfortunately, the FDA and EMA have approved PD-1 blockade in combination with “platinum-based chemotherapy” without specifying the precise nature of the platinum-containing drug. This is a non sequitur. Based on the available clinical data, such approvals should be restricted to the use of oxaliplatin.
Collapse
Affiliation(s)
- Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Jianzhou Chen
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Antoine Hollebecque
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.,Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy, ClinicObiome, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, APHP, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
16
|
Slagter AE, Caspers IA, van Grieken NCT, Walraven I, Lind P, Meershoek-Klein Kranenbarg E, Grootscholten C, Nordsmark M, van Sandick JW, Sikorska K, van de Velde CJH, Jansen EPM, Verheij M, van Laarhoven HWM, Cats A. Triplet Chemotherapy with Cisplatin versus Oxaliplatin in the CRITICS Trial: Treatment Compliance, Toxicity, Outcomes and Quality of Life in Patients with Resectable Gastric Cancer. Cancers (Basel) 2022; 14:2963. [PMID: 35740628 PMCID: PMC9221508 DOI: 10.3390/cancers14122963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Perioperative chemotherapy is the current standard treatment for patients with resectable gastric cancer. Based on studies in patients with metastatic gastric cancer, oxaliplatin has replaced cisplatin in the curative setting as well. However, evidence to prefer oxaliplatin over cisplatin in the curative setting is limited. (2) Methods: We compared patient-related and tumor-related outcomes for cisplatin versus oxaliplatin in patients with resectable gastric cancer treated with perioperative chemotherapy in the CRITICS trial. (3) Results: Preoperatively, 632 patients received cisplatin and 149 patients received oxaliplatin. Preoperative severe toxicity was encountered in 422 (67%) patients who received cisplatin versus 89 (60%) patients who received oxaliplatin (p = 0.105). Severe neuropathy was observed in 5 (1%) versus 6 (4%; p = 0.009) patients, respectively. Postoperative severe toxicity occurred in 109 (60%) versus 26 (51%) (p = 0.266) patients; severe neuropathy in 2 (1%) versus 2 (4%; p = 0.209) for patients who received cisplatin or oxaliplatin, respectively. Diarrhea impacted the quality of life more frequently in patients who received oxaliplatin compared to cisplatin. Complete or near-complete pathological response was achieved in 94 (21%) versus 16 (15%; p = 0.126) patients who received cisplatin or oxaliplatin, respectively. Overall survival was not significantly different in both groups (p = 0.300). (4) Conclusions: Both cisplatin and oxaliplatin are legitimate options as part of systemic treatment in patients with resectable gastric cancer.
Collapse
Affiliation(s)
- Astrid E. Slagter
- Department of Radiation Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.E.S.); (I.W.); (E.P.M.J.); (M.V.)
| | - Irene A. Caspers
- Department of Gastrointestinal Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (I.A.C.); (C.G.)
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands;
| | - Nicole C. T. van Grieken
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands;
| | - Iris Walraven
- Department of Radiation Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.E.S.); (I.W.); (E.P.M.J.); (M.V.)
- Department of Epidemiology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Pehr Lind
- Department of Oncology, Stockholm Söder Hospital, 118 83 Stockholm, Sweden;
- Karolinska Institutet, Research Oncology, 171 77 Stockholm, Sweden
| | | | - Cecile Grootscholten
- Department of Gastrointestinal Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (I.A.C.); (C.G.)
| | | | - Johanna W. van Sandick
- Department of Surgery, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | - Karolina Sikorska
- Department of Biometrics, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | - Cornelis J. H. van de Velde
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (E.M.-K.K.); (C.J.H.v.d.V.)
| | - Edwin P. M. Jansen
- Department of Radiation Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.E.S.); (I.W.); (E.P.M.J.); (M.V.)
| | - Marcel Verheij
- Department of Radiation Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.E.S.); (I.W.); (E.P.M.J.); (M.V.)
- Department of Radiation Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Hanneke W. M. van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Annemieke Cats
- Department of Gastrointestinal Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (I.A.C.); (C.G.)
| |
Collapse
|
17
|
Rich MM, Housley SN, Nardelli P, Powers RK, Cope TC. Imbalanced Subthreshold Currents Following Sepsis and Chemotherapy: A Shared Mechanism Offering a New Therapeutic Target? Neuroscientist 2022; 28:103-120. [PMID: 33345706 PMCID: PMC8215085 DOI: 10.1177/1073858420981866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Both sepsis and treatment of cancer with chemotherapy are known to cause neurologic dysfunction. The primary defects seen in both groups of patients are neuropathy and encephalopathy; the underlying mechanisms are poorly understood. Analysis of preclinical models of these disparate conditions reveal similar defects in ion channel function contributing to peripheral neuropathy. The defects in ion channel function extend to the central nervous system where lower motoneurons are affected. In motoneurons the defect involves ion channels responsible for subthreshold currents that convert steady depolarization into repetitive firing. The inability to correctly translate depolarization into steady, repetitive firing has profound effects on motor function, and could be an important contributor to weakness and fatigue experienced by both groups of patients. The possibility that disruption of function, either instead of, or in addition to neurodegeneration, may underlie weakness and fatigue leads to a novel approach to therapy. Activation of serotonin (5HT) receptors in a rat model of sepsis restores the normal balance of subthreshold currents and normal motoneuron firing. If an imbalance of subthreshold currents also occurs in other central nervous system neurons, it could contribute to encephalopathy. We hypothesize that pharmacologically restoring the proper balance of subthreshold currents might provide effective therapy for both neuropathy and encephalopathy in patients recovering from sepsis or treatment with chemotherapy.
Collapse
Affiliation(s)
- Mark M. Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Stephen N. Housley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA,Integrated Cancer Research Center, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Paul Nardelli
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Randall K. Powers
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Timothy C. Cope
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA,Integrated Cancer Research Center, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA,Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
18
|
Jiang Z, Zhou A, Sun Y, Zhang W. Biweekly oxaliplatin plus S1 for Chinese elderly patients with advanced gastric or gastroesophageal junction cancer as the first-line therapy: a single-arm, phase 2 study. BMC Cancer 2022; 22:253. [PMID: 35264150 PMCID: PMC8908595 DOI: 10.1186/s12885-022-09332-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/23/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND SOX (oxaliplatin and S1, every 3 weeks) is one of the most common first-line chemotherapy for advanced or metastatic G/GEJ (gastric or gastroesophageal junction) cancer in Asia, but it has noticeable hematological and neurological toxicity. In China, the majority of gastric cancer patients are middle-aged and elderly with poor tolerance to 3-weekly chemotherapy. Therefore, we aimed to assess efficacy and safety of biweekly SOX for Chinese advanced G/GEJ cancer patients aged ≥ 60 years as the first-line treatment in a single arm phase 2 study. METHODS Oxaliplatin was administered intravenously on day 1 at 85 mg/m2. S-1 was given at 80, 100 or 120 mg/day, depending on the body surface area (< 1.25 m2, 1.25 to < 1.5 m2, or ≥ 1.5 m2), twice daily, on day 1-10, every 2 weeks. The primary endpoint was objective response rate (ORR), and the secondary endpoints included progression free survival (PFS), overall survival (OS), disease control rate (DCR), duration of response (DOR) and safety. RESULTS Between May 2016 and Sep 2018, 42 patients were enrolled. The median follow-up was 43.6 months. The ORR and DCR were 52.4% and 85.7%, respectively. The median PFS was 4.6 months (95%CI 2.486-6.714), and the median OS was 11.1 months (95%CI 8.001-14.199). The most common treatment-related adverse events (TRAEs) of any grade included thrombocytopenia (59.5%), neutropenia (57.1%), appetite loss (57.1%) and nausea (54.8%). Only two patients suffered from grade 3 TRAEs (4.8%), including neutropenia (1 patient, [2.4%]) and diarrhea (1 patient, [2.4%]). No ≥ grade 4 TRAEs occurred. CONCLUSIONS Biweekly SOX seemed to have favorable tolerance without compromising the efficacy as the first-line therapy in Chinese elderly patients aged ≥ 60 years with advanced G/GEJ cancer. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT04694404 (5/1/2021). This study was approved by the Ethical Committee of National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, (17-048/1303).
Collapse
Affiliation(s)
- Zhichao Jiang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. No, 17, Panjiayuannanli Street, Chaoyang District, Beijing, 100021, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. No, 17, Panjiayuannanli Street, Chaoyang District, Beijing, 100021, China
| | - Yongkun Sun
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. No, 17, Panjiayuannanli Street, Chaoyang District, Beijing, 100021, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. No, 17, Panjiayuannanli Street, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
19
|
Gronau F, Feldbruegge L, Oberwittler F, Gonzalez-Moreno S, Villeneuve L, Eveno C, Glehen O, Kusamura S, Rau B. HIPEC in Peritoneal Metastasis of Gastric Origin: A Systematic Review of Regimens and Techniques. J Clin Med 2022; 11:jcm11051456. [PMID: 35268546 PMCID: PMC8911234 DOI: 10.3390/jcm11051456] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Peritoneal metastasis in gastric cancer is associated with a poor prognosis. Complete cytoreductive surgery including gastrectomy and complete removal of all peritoneal lesions followed by hyperthermic intraperitoneal chemotherapy (HIPEC) achieves promising results. There exists an immersive variety of approaches for HIPEC that makes it difficult to weigh different results obtained in the literature. In order to enable standardization and development of HIPEC, we here present a systematic review of different drug regimens and technical approaches. (2) Methods: PubMed, Embase, and the Cochrane Library were systematically searched on 26 May 2021 using the mesh terms “intraperitoneal chemotherapy AND gastric cancer”. Under consideration of systematic review guidelines, articles reporting on HIPEC in combination with CRS were selected. Data on duration, drugs, dosage, and other application parameters as well as morbidity and long term survival data were extracted for subsequent statistical analysis, tabulation, and descriptive synthesis. We assessed the risk of bias due to inhomogeneity of the patient cohort and incompleteness of report of HIPEC parameters. (3) Results: Out of 1421 screened publications, 42 publications presenting data from 1325 patients met the criteria. Most of the publications were single institutional retrospective cohort studies. The most common HIPEC regimen is performed after gastrointestinal anastomosis and consists of 50–200 mg/m2 cisplatinum and 30–40 mg/m2 mytomycin C at 42–43 °C for 60–90 min in a closed abdomen HIPEC system with three tubes. Almost every study reported incompletely on HIPEC parameters. Lower rates of anastomotic leakage were reported in studies that performed HIPEC after gastrointestinal anastomosis. Studies that performed open HIPEC and integrated a two-drug regimen indicated better overall survival rates. (4) Discussion: This is an exhaustive overview of the use of drug regimens and techniques for HIPEC after CRS for gastric cancer peritoneal metastasis. Other indications and application modes of intraperitoneal chemotherapy such as prophylactic or palliative HIPEC apart from CRS were not addressed. (5) Conclusion: Complete report of HIPEC parameters should be included in every publication. A consensus for dose expression either per BSA or as flat dose is desirable for comparison of the drug regimens. Despite numerous variations, we identified the most common regimens and techniques and their advantages and disadvantages according to the data in the literature. More phase I/II studies are needed to identify the best approach for HIPEC. (6) Other: This review was not supported by third parties.
Collapse
Affiliation(s)
- Felix Gronau
- Department of Surgery, Chirurgische Klinik Campus Charité Mitte|Campus Virchow Klinikum, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (F.G.); (L.F.); (F.O.)
| | - Linda Feldbruegge
- Department of Surgery, Chirurgische Klinik Campus Charité Mitte|Campus Virchow Klinikum, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (F.G.); (L.F.); (F.O.)
| | - Frauke Oberwittler
- Department of Surgery, Chirurgische Klinik Campus Charité Mitte|Campus Virchow Klinikum, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (F.G.); (L.F.); (F.O.)
| | | | - Laurent Villeneuve
- Réseau National de Prise en Charge des Tumeurs Rares du Péritoine, French National Registry of Rare Peritoneal Surface Malignancies, 69002 Lyon, France;
| | - Clarisse Eveno
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, 69495 Pierre-Bénite, France; (C.E.); (O.G.)
| | - Olivier Glehen
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, 69495 Pierre-Bénite, France; (C.E.); (O.G.)
| | - Shigeki Kusamura
- Peritoneal Surface Malignancies Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori dei Tumori di Milano, 20133 Milano, Italy;
| | - Beate Rau
- Department of Surgery, Chirurgische Klinik Campus Charité Mitte|Campus Virchow Klinikum, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (F.G.); (L.F.); (F.O.)
- Correspondence: ; Tel.: +49-30-450-622-214
| |
Collapse
|
20
|
Catenacci DVT, Chung HC, Shen L, Moehler M, Yoon HH, Rosales MK, Kang YK. Safety and efficacy of HER2 blockade by trastuzumab-based chemotherapy-containing combination strategies in HER2+ gastroesophageal adenocarcinoma. ESMO Open 2022; 7:100360. [PMID: 34973512 PMCID: PMC8728435 DOI: 10.1016/j.esmoop.2021.100360] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 01/14/2023] Open
Abstract
Since completion of the Trastuzumab for Gastric Cancer study, trastuzumab with doublet chemotherapy (a fluoropyrimidine and a platinum) has been the gold-standard first-line therapy for patients with locally advanced unresectable or metastatic human epidermal growth factor receptor 2-positive (HER2+) gastroesophageal adenocarcinoma (GEA). The safety and efficacy of 23 studies of first-line trastuzumab plus doublet chemotherapy, without checkpoint inhibitors (n = 19) or with checkpoint inhibitors (n = 4), conducted in patients with locally advanced unresectable or metastatic HER2+ GEA, including phase II/III, prospective, and retrospective observational studies, were summarized. In studies without checkpoint inhibitors, the median duration of trastuzumab treatment ranged from 19.5 to 39.0 weeks and from 15.3 to 30.0 weeks for chemotherapy. In studies with checkpoint inhibitors, the median duration of pembrolizumab/trastuzumab/chemotherapy was 30 weeks, and 18 weeks for chemotherapy. In studies without checkpoint inhibitors, treatment-emergent adverse events (TEAEs) of grade ≥3 ranged from 32% to 84%. Serious adverse events (SAEs) ranged from 15% to 39%. Adverse events resulting in discontinuation ranged from 0% to 30%. Treatment-related deaths occurred in 0%-9% of patients. In studies with checkpoint inhibitors, TEAEs of grade ≥3 were 57%. SAEs ranged from 31% to 38%. Adverse events resulting in discontinuation ranged from 5% to 24%. Treatment-related deaths occurred in 0%-3% of patients. In studies without checkpoint inhibitors, objective response rate (ORR) ranged from 39% to 82%, median progression-free survival (PFS) from 5.7 to 11.6 months, and median overall survival (OS) from 11.2 to 27.6 months. In studies with checkpoint inhibitors, ORR ranged from 39% to 86%, median PFS from 8.0 to 13.0 months, and median OS from 19.3 to 27.3 months. This review provides a historical benchmark on safety and efficacy of available first-line chemotherapy-based standard of care for patients with locally advanced unresectable or metastatic HER2+ GEA.
Collapse
Affiliation(s)
- D V T Catenacci
- Department of Medicine, The University of Chicago Medical Center, Chicago, USA.
| | - H C Chung
- Department of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - L Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - M Moehler
- Johannes-Gutenberg University, Mainz, Germany
| | - H H Yoon
- Division of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, USA
| | | | - Y-K Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Gürler F, İlhan A, Güven DC, Turhan O, Kurt İnci B, Sütçüoğlu O, Yildiz F, Arik Z, Öksüzoğlu B, Yalçin Ş, Özdemir N, Yazici O, Özet A. Does docetaxel matter in metastatic gastric cancer? FOLFOX versus FLOT regimens as first-line treatment. Anticancer Drugs 2022; 33:e477-e485. [PMID: 34261917 DOI: 10.1097/cad.0000000000001143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We aimed to compare the efficacy and the safety of the FOLFOX and the FLOT regimens in metastatic gastric cancer (mGC) as first-line treatment. It was a retrospective multicenter observational study. The comparisons between groups were conducted in terms of progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and hematologic adverse events. Seventy-nine patients, diagnosed with mGC between March 2012 and December 2019, treated with FOLFOX (n = 43) or FLOT (n = 36) regimens as first-line treatment were included in the study. The mPFS was 10.9 months [95% confidence interval (CI), 5.8-16.1] in the FLOT arm and 7.1 months (95% CI, 5.1-9.1) in the FOLFOX arm (P < 0.001). The ORR was 63.9% in the FLOT arm and 30.2% in the FOLFOX arm (P = 0.003). The mOS was 13.3 months (95% CI, 11.3-15.4) in the FLOT arm and 10.9 months (95% CI, 8.2-13.5) in the FOLFOX arm (P = 0.103). The hematologic adverse events in all grades were 88.4% (n = 38) in the FOLFOX arm compared with 80.6% (n = 29) in the FLOT arm (P = 0.335). The FLOT regimen might be a preferred option in mGC with an improved PFS and ORR compared with the FOLFOX regimen.
Collapse
Affiliation(s)
| | - Ayşegül İlhan
- Department of Medical Oncology, HSU Dr Abdurrahman Yurtaslan Oncology Training & Research Hospital
| | - Deniz Can Güven
- Department of Medical Oncology, Hacettepe University Cancer Institute
| | - Okan Turhan
- Department of Internal Medicine, Gazi University, Ankara, Turkey
| | | | | | - Fatih Yildiz
- Department of Medical Oncology, HSU Dr Abdurrahman Yurtaslan Oncology Training & Research Hospital
| | - Zafer Arik
- Department of Medical Oncology, Hacettepe University Cancer Institute
| | - Berna Öksüzoğlu
- Department of Medical Oncology, HSU Dr Abdurrahman Yurtaslan Oncology Training & Research Hospital
| | - Şuayib Yalçin
- Department of Medical Oncology, Hacettepe University Cancer Institute
| | | | - Ozan Yazici
- Department of Medical Oncology, Gazi University
| | - Ahmet Özet
- Department of Medical Oncology, Gazi University
| |
Collapse
|
22
|
Yıldırım S, Yılmaz C. Trıplet or Doublet Chemotherapy Regimens in Metastatic Gastric Cancer. CLINICAL CANCER INVESTIGATION JOURNAL 2022. [DOI: 10.51847/rpfscains5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
Konopka K, Micek A, Ochenduszko S, Streb J, Potocki P, Kwinta Ł, Wysocki PJ. Combined Neutrophil-to-Lymphocyte and Platelet-Volume-to-Platelet Ratio (NLR and PVPR Score) Represents a Novel Prognostic Factor in Advanced Gastric Cancer Patients. J Clin Med 2021; 10:3902. [PMID: 34501353 PMCID: PMC8432226 DOI: 10.3390/jcm10173902] [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: 07/28/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Chemotherapy is a cornerstone of treatment in advanced gastric cancer (GC) with a proven impact on overall survival, however, reliable predictive markers are missing. The role of various inflammatory markers has been tested in gastric cancer patients, but there is still no general consensus on their true clinical applicability. High neutrophil-to-lymphocyte (NLR) and low (medium)-platelets-volume-to-platelet ratio (PVPR) are known markers of unspecific immune system activation, correlating significantly with outcomes in advanced GC patients. METHODS Metastatic GC patients (N:155) treated with chemotherapy +/- trastuzumab were enrolled in this retrospective study. Pre-treatment NLR and PVPR, as well as other inflammatory markers were measured in peripheral blood. Univariate Cox regression was conducted to find markers with a significant impact on overall survival (OS) and progression-free survival (PFS). Spearman correlation and Cohen's kappa was used to analyze multicollinearity. Multiple multivariable Cox regression models were built to study the combined impact of NLR and PVPR, as well as other known prognostic factors on OS. RESULTS Elevated NLR was significantly associated with increased risk of death (HR = 1.95; 95% CI: 1.17-3.24), and lower PVPR was significantly associated with improved outcomes (HR = 0.53; 95% CI: 0.32-0.90). A novel inflammatory marker, based on a combination of NLR and PVPR, allows for the classification of GC patients into three prognostic groups, characterized by median OS of 8.4 months (95% CI 5.8-11.1), 10.5 months (95% CI 8.8-12.1), and 15.9 months (95% CI 13.5-18.3). CONCLUSION The NLR and PVPR score (elevated NLR and decreased PVPR) is a marker of detrimental outcome of advanced GC patients treated with chemotherapy.
Collapse
Affiliation(s)
- Kamil Konopka
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland; (J.S.); (P.P.); (Ł.K.); (P.J.W.)
| | - Agnieszka Micek
- Department of Nursing Management and Epidemiology Nursing, Jagiellonian University Medical College, 31-007 Cracow, Poland;
| | | | - Joanna Streb
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland; (J.S.); (P.P.); (Ł.K.); (P.J.W.)
| | - Paweł Potocki
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland; (J.S.); (P.P.); (Ł.K.); (P.J.W.)
| | - Łukasz Kwinta
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland; (J.S.); (P.P.); (Ł.K.); (P.J.W.)
| | - Piotr J. Wysocki
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland; (J.S.); (P.P.); (Ł.K.); (P.J.W.)
| |
Collapse
|
24
|
Fahmy SA, Ponte F, Fawzy IM, Sicilia E, Azzazy HMES. Betaine host-guest complexation with a calixarene receptor: enhanced in vitro anticancer effect. RSC Adv 2021; 11:24673-24680. [PMID: 35481025 PMCID: PMC9036920 DOI: 10.1039/d1ra04614d] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
p-Sulfonatocalix[n]arenes have shown excellent potential for accommodating chemotherapeutic drugs through host-guest complexation and enhancing their anticancer activity. Betaine has been reported to exert an anticancer effect at high concentrations. In order to increase its concentration in cancer cells, we have complexed it with p-SC4, which releases its content in an acidic environment typical of cancer tissue. In this work, a host-guest complex of the chemically stable, natural, and safe active methyl donor (betaine) and p-sulfonatocalix[4]arenes (p-SC4) was designed and characterized using 1H NMR, UV, Job's plot analysis, DFT calculations, and molecular modeling for use in cancer therapeutics. The peak amplitude of the prepared host-guest complexes was linearly proportional to the concentration of betaine in the range of 1.0 × 10-5 M-1 to 2.5 × 10-4 M-1. The reaction stoichiometry between p-SC4 and betaine in the formed complex was 1 : 1. The stability constant for the complex is 8.9 × 104 M-1 which corresponds to a complexation free energy of -6.74 kcal mol-1. Complexation between betaine and p-SC4 was found to involve the insertion of the trimethylammonium group of betaine into the p-SC4 cavity, as supported by the experimental data. The complex displayed enhanced cytotoxic activities against breast adenocarcinoma cells (MCF-7) and cervical cancer cells (HeLa) compared to free betaine. In conclusion, the host-guest complexation of betaine with p-SC4 increases its concentration in cancer cells, which warrants further investigation for cancer therapy.
Collapse
Affiliation(s)
- Sherif Ashraf Fahmy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo AUC Avenue, PO. Box 74 New Cairo 11835 Egypt +20 2 2795 7565 +20 2 2615 2559
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria Arcavacata di Rende 87036 Italy
| | - Iten M Fawzy
- Pharmaceutical Chemistry Department, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt Cairo 12311 Egypt
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, University of Calabria Arcavacata di Rende 87036 Italy
| | - Hassan Mohamed El-Said Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo AUC Avenue, PO. Box 74 New Cairo 11835 Egypt +20 2 2795 7565 +20 2 2615 2559
| |
Collapse
|
25
|
Jimenez-Fonseca P, Carmona-Bayonas A, Martinez-Torron A, Alsina M, Custodio A, Serra O, Cacho Lavin D, Limón ML, Sauri T, López F, Visa L, Granja M, Martínez Lago N, Arrazubi V, Vidal Tocino R, Hernandez R, Aguado G, Cano JM, Martín Carnicero A, Mangas M, Pimentel P, Fernández Montes A, Macias Declara I, Longo F, Ramchandani A, Martín Richard M, Hurtado A, Azkarate A, Hernández Pérez C, Serrano R, Gallego J, on behalf of the AGAMENON-SEOM study group. External validity of clinical trials with diverse trastuzumab-based chemotherapy regimens in advanced gastroesophageal adenocarcinoma: data from the AGAMENON-SEOM registry. Ther Adv Med Oncol 2021; 13:17588359211019672. [PMID: 34211587 PMCID: PMC8216357 DOI: 10.1177/17588359211019672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Trastuzumab combined with cisplatin and fluoropyrimidines, either capecitabine or 5-fluorouracile (XP/FP), is the standard first-line treatment for advanced, HER2-positive, gastric cancer patients based on the ToGA trial. Despite the lack of phase III trials, many clinicians administer trastuzumab with alternative regimens. One meta-analysis suggests that substituting cisplatin for oxaliplatin might lead to greater efficacy and less toxicity. METHODS 594 patients with HER2-positive gastroesophageal adenocarcinoma were recruited from the AGAMENON-SEOM registry. The objective was to evaluate the external validity of clinical trials with chemotherapy and trastuzumab. RESULTS The regimens used in at least 5% of the patients were XP (27%), oxaliplatin and capecitabine (CAPOX) (26%), oxaliplatin and 5-fluorouracil (FOLFOX) (14%), FP (14%), triplet with anthracycline/docetaxel (7%), and carboplatin-FU (5%). Median exposure to trastuzumab was longer with FOLFOX (11.4 months, 95% CI, 9.1-21.0) versus ToGA regimens (7.5, 6.4-8.5), p < 0.001. Patients with HER2-IHC 3+ cancers had higher response rates than those with IHC 2+/FISH+, odds-ratio 1.97 (95% CI, 1.25-3.09). The results achieved with CAPOX-trastuzumab were comparable to those attained with ToGA regimens. FOLFOX-trastuzumab was superior to ToGA schemes in terms of overall survival (OS), with a greater magnitude of effect in IHC 2+/FISH+ tumors (HR 0.47, 0.24-0.92) compared with IHC 3+ (HR 0.69, 0.49-0.96), and in diffuse (HR 0.37, 0.20-0.69) versus intestinal-type tumors (HR 0.76, 0.54-1.06). CONCLUSION We have updated the external validity of clinical trials with trastuzumab in first-line treatment of gastric cancer. Our data confirm the comparable outcomes of ToGA regimens and CAPOX-trastuzumab in clinical practice and point toward a possible benefit of FOLFOX-trastuzumab, contingent on the subtypes typically less sensitive to trastuzumab, to be confirmed in clinical trials.
Collapse
Affiliation(s)
- Paula Jimenez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Alberto Carmona-Bayonas
- Medical Oncology Department, Hospital Universitario Morales Meseguer, Calle Marqués de los Vélez, s/n, Murcia, 30007, Spain
| | - Alba Martinez-Torron
- Pharmacy Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Maria Alsina
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Custodio
- Medical Oncology Department, Hospital Universitario La Paz, CIBERONC CB16/12/00398, Madrid, Spain
| | - Olbia Serra
- Medical Oncology Department, Catalan Institute of Oncology, L’Hospitalet, Spain
| | - Diego Cacho Lavin
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - María Luisa Limón
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Tamara Sauri
- Medical Oncology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Flora López
- Medical Oncology Department, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Laura Visa
- Medical Oncology Department, Hospital Universitario El Mar, Barcelona, Spain
| | - Mónica Granja
- Medical Oncology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Nieves Martínez Lago
- Medical Oncology Department, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Rosario Vidal Tocino
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Raquel Hernandez
- Medical Oncology Department, Hospital Universitario de Canarias, Tenerife, Spain
| | - Gema Aguado
- Medical Oncology Department, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Juana María Cano
- Medical Oncology Department, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | | | - Monserrat Mangas
- Medical Oncology Department, Hospital Galdakao-Usansolo, Usansolo, Spain
| | - Paola Pimentel
- Medical Oncology Department, Hospital General Universitario Santa Lucía, Cartagena, Spain
| | | | | | - Federico Longo
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Avinash Ramchandani
- Medical Oncology Department, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Marta Martín Richard
- Medical Oncology Department, Hospital Universitario Santa Creu i Sant Pau, Barcelona, Spain
| | - Alicia Hurtado
- Medical Oncology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - Aitor Azkarate
- Medical Oncology Department, Hospital Universitario Son Espases, Mallorca, Spain
| | - Carolina Hernández Pérez
- Medical Oncology Department, Hospital Universitario Nuestra Señora de the Candelaria, Tenerife, Spain
| | - Raquel Serrano
- Medical Oncology Department, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Javier Gallego
- Medical Oncology Department, Hospital General Universitario of Elche, Elche, Spain
| | | |
Collapse
|
26
|
van Schaik TA, Chen KS, Shah K. Therapy-Induced Tumor Cell Death: Friend or Foe of Immunotherapy? Front Oncol 2021; 11:678562. [PMID: 34141622 PMCID: PMC8204251 DOI: 10.3389/fonc.2021.678562] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
Combinatory treatments using surgery, radiotherapy and/or chemotherapy together with immunotherapy have shown encouraging results for specific subsets of tumors, but a significant proportion of tumors remains unsusceptible. Some of these inconsistencies are thought to be the consequence of an immunosuppressive tumor microenvironment (TME) caused by therapy-induced tumor cell death (TCD). An increased understanding of the molecular mechanisms governing TCD has provided valuable insights in specific signaling cascades activated by treatment and the subsequent effects on the TME. Depending on the treatment variables of conventional chemo-, radio- and immunotherapy and the genetic composition of the tumor cells, particular cell death pathways are activated. Consequently, TCD can either have tolerogenic or immunogenic effects on the local environment and thereby affect the post-treatment anti-tumor response of immune cells. Thus, identification of these events can provide new rationales to increase the efficacy of conventional therapies combined with immunotherapies. In this review, we sought to provide an overview of the molecular mechanisms initiated by conventional therapies and the impact of treatment-induced TCD on the TME. We also provide some perspectives on how we can circumvent tolerogenic effects by adequate treatment selection and manipulation of key signaling cascades.
Collapse
Affiliation(s)
- Thijs A van Schaik
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, MA, United States.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kok-Siong Chen
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, MA, United States.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, MA, United States.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
| |
Collapse
|
27
|
Wei G, Wu M, Zhu H, Han S, Chen J, Zhai C, Shi L. Off-Label Use of Antineoplastic Drugs to Treat Malignancies: Evidence From China Based on a Nationwide Medical Insurance Data Analysis. Front Pharmacol 2021; 12:616453. [PMID: 33897416 PMCID: PMC8060556 DOI: 10.3389/fphar.2021.616453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
Purpose: Cancer is a leading cause of morbidity and mortality worldwide. Off-label (OL) use of antineoplastic drugs to treat malignancies is prevalent. In this study, we quantified and characterized OL use of antineoplastic drugs to treat malignancies in China. Methods: This was a retrospective study using nationwide data collected from 2008 to 2010. Use of antineoplastic drugs was considered OL if they were used for indications not reflected in the package insert published by the National Medical Products Administration at the time of prescription. Descriptive analysis and Spearman rank correlation were used to evaluate the frequency and pattern of OL drug use. Results: In total, 51,382 patients with malignancies, 24 categories of antineoplastic drugs, and 77 types of malignancies treated with OL drugs were included in this study. Twenty commonly used antineoplastic drugs (ICD encoded as L01) were used OL in 10-61% of cases, and four commonly used endocrine therapy antineoplastic drugs (ICD encoded as L02) were used OL in 10-19% of cases. There was a significant negative correlation between the disease constituent ratio and the average OL use rate of antineoplastic drugs for various malignancies. In contrast, there was a significant positive correlation between the average OL use rate of antineoplastic drugs and the number of malignancies treated with OL drugs. Conclusion: This study provided information regarding OL use of antineoplastic drugs for treatment of malignancies, and showed that OL use was prevalent. In addition, uncommon malignancies were more likely to be treated with OL antineoplastic drugs. Furthermore, more commonly used antineoplastic drugs were more likely to be used OL.
Collapse
Affiliation(s)
- Guoxu Wei
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Min Wu
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - He Zhu
- International Research Center for Medicinal Administration, Peking University Health Science Center, Beijing, China
| | - Sheng Han
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
- International Research Center for Medicinal Administration, Peking University Health Science Center, Beijing, China
| | - Jing Chen
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
- International Research Center for Medicinal Administration, Peking University Health Science Center, Beijing, China
| | - Chenchen Zhai
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Luwen Shi
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
- International Research Center for Medicinal Administration, Peking University Health Science Center, Beijing, China
| |
Collapse
|
28
|
Grover M, Behl T, Sachdeva M, Bungao S, Aleya L, Setia D. Focus on Multi-targeted Role of Curcumin: a Boon in Therapeutic Paradigm. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:18893-18907. [PMID: 33595796 DOI: 10.1007/s11356-021-12809-w] [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: 05/22/2020] [Accepted: 02/01/2021] [Indexed: 06/12/2023]
Abstract
Curcumin is a polyphenolic compound that exhibited good anticancer potential against different types of cancers through its multi-targeted effect like the termination of cell proliferation, inflammation, angiogenesis, and metastasis, thereby acting as antiproliferative and cytotoxic in nature. The present review surveys the various drug combination tried with curcumin or its synthetic analogues and also the mechanism by which curcumin potentiates the effect of almost every drug. In addition, this article also focuses on aromatherapy which is gaining much popularity in cancer patients. After thoroughly studying several articles on combination therapy of curcumin through authenticated book chapters, websites, research, and review articles available at PubMed, ScienceDirect, etc., it has been observed that multi-targeted curcumin possess enormous anticancer potential and, with whatever drug it is given in combination, has always resulted in enhanced effect with reduced dose as well as side effects. It is also capable enough in overcoming the problem of chemoresistance. Besides this, aromatherapy also proved its potency in reducing cancer-related side effects. Combining all the factors together, we can conclude that combination therapy of drugs with curcumin should be explored extensively. In addition, aromatherapy can be used as an adjuvant or supplementary therapy to reduce the cancer complications in patients.
Collapse
Affiliation(s)
- Madhuri Grover
- B.S. Anangpuria Institute of Pharmacy, Alampur, Haryana, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | | | - Simona Bungao
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Dhruv Setia
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
29
|
Fahmy SA, Ponte F, Fawzy IM, Sicilia E, Bakowsky U, Azzazy HMES. Host-Guest Complexation of Oxaliplatin and Para-Sulfonatocalix[n]Arenes for Potential Use in Cancer Therapy. Molecules 2020; 25:E5926. [PMID: 33327642 PMCID: PMC7765097 DOI: 10.3390/molecules25245926] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 01/25/2023] Open
Abstract
P-sulfonatocalix[n]arenes have demonstrated a great potential for encapsulation of therapeutic drugs via host-guest complexation to improve solubility, stability, and bioavailability of encapsulated drugs. In this work, guest-host complexes of a third-generation anticancer drug (oxaliplatin) and p-4-sulfocalix[n]arenes (n = 4 and 6; p-SC4 and p-SC6, respectively) were prepared and investigated, using 1H NMR, UV, Job's plot analysis, and DFT calculations, for use as cancer therapeutics. The peak amplitude of the prepared host-guest complexes was linearly proportional to the concentration of oxaliplatin in the range of 1.0 × 10-5 M-1 to 2.1 × 10-4 M-1. The reaction stoichiometry between either p-SC4 or p-SC6 and oxaliplatin in the formed complexes was 1:1. The stability constants for the complexes were 5.07 × 104 M-1 and 6.3 × 104 M-1. These correspond to complexation free energy of -6.39 and -6.52 kcal/mol for p-SC4 and p-SC6, respectively. Complexation between oxaliplatin and p-SC4 or p-SC6 was found to involve hydrogen bonds. Both complexes exhibited enhanced biological and high cytotoxic activities against HT-29 colorectal cells and MCF-7 breast adenocarcinoma compared to free oxaliplatin, which warrants further investigation for cancer therapy.
Collapse
Affiliation(s)
- Sherif Ashraf Fahmy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt;
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria, 87036 Arcavacata di Rende, Italy; (F.P.); (E.S.)
| | - Iten M. Fawzy
- Pharmaceutical Chemistry Department, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo 12311, Egypt;
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, University of Calabria, 87036 Arcavacata di Rende, Italy; (F.P.); (E.S.)
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Hassan Mohamed El-Said Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt;
| |
Collapse
|
30
|
Fahmy S, Ponte F, Sicilia E, El-Said Azzazy HM. Experimental and Computational Investigations of Carboplatin Supramolecular Complexes. ACS OMEGA 2020; 5:31456-31466. [PMID: 33324858 PMCID: PMC7726934 DOI: 10.1021/acsomega.0c05168] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/13/2020] [Indexed: 05/28/2023]
Abstract
Supramolecular systems (macromolecules), such as calix[n]arenes (SCn), cyclodextrins (CDs), and cucurbiturils (CBs), are promising vehicles for anticancer drugs. In this work, guest-host complexes of carboplatin, a second-generation platinum-based anticancer drug, and p-4-sulfocalix[n]arenes (n = 4 and 6; PS4 and PS6, respectively) were prepared and studied using 1H NMR, UV, Job's plot analysis, HPLC, and density-functional theory calculations. The experimental and the computational studies suggest the formation of 1:1 complexes between carboplatin and each of PS4 and PS6. The stability constants of the formed complexes were estimated to be 5.3 × 104 M-1 and 9.8 × 104 M-1, which correspond to free energy of complexation of -6.40 and -6.81 kcal mol-1, in the case of PS4 and PS6, respectively. The interaction free energy depends on the different inclusion modes of carboplatin in the host cavities. UV-vis findings and atoms in molecules analysis showed that hydrogen bond interactions stabilize the host-guest complexes without the full inclusion in the host cavity. The in vitro anticancer study revealed that both complexes exhibited stronger anticancer activities against breast adenocarcinoma cells (MCF-7) and lung cancer cells (A-549) compared to free carboplatin, preluding to their potential use in cancer therapy.
Collapse
Affiliation(s)
- Sherif
Ashraf Fahmy
- Department
of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| | - Fortuna Ponte
- Department
of Chemistry and Chemical Technologies, University of Calabria, Arcavacata
di Rende 87036, Italy
| | - Emilia Sicilia
- Department
of Chemistry and Chemical Technologies, University of Calabria, Arcavacata
di Rende 87036, Italy
| | - Hassan Mohamed El-Said Azzazy
- Department
of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| |
Collapse
|
31
|
Safety and efficacy of S-1 plus oxaliplatin 130 mg/m 2 combination therapy in patients with previously untreated HER2-negative unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: a phase II trial (KSCC1501A). Int J Clin Oncol 2020; 26:345-354. [PMID: 33085058 DOI: 10.1007/s10147-020-01803-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND In a randomized pivotal global phase III study, S-1 and oxaliplatin 100 mg/m2 (SOX100) combination chemotherapy was as effective as S-1 and cisplatin for advanced gastric cancer (AGC) and showed a favorable safety profile. In this phase II study, we analyzed survival outcomes to assess the efficacy and safety of the SOX regimen with oxaliplatin 130 mg/m2 (SOX130) in AGC. METHODS Patients with HER2-negative AGC received 80 mg/m2/day S-1 orally on days 1-14 and 130 mg/m2 oxaliplatin intravenously on day 1 of each 21-day cycle until the criteria for treatment withdrawal were fulfilled. The primary endpoint was the response rate (RR), and the null hypothesis of RR in the current trial was 45%. The secondary endpoints were progression-free survival (PFS) and overall survival (OS). Adverse events (AEs) were recorded according to CTCAE version 4.0. RESULTS Seventy-one patients were enrolled from June 2015 to November 2016, but eight were excluded for ineligibility. Therefore, all final analyses were conducted with 63 patients. The confirmed RR was 46.0% (90% confidence interval [CI]: 36.1-56.3), and the disease control rate was 77.8% (90% CI: 68.1-85.1). The median PFS and OS were 4.9 (95% CI: 4.2-7.1) and 14.8 (95% CI: 11.1-18.9) months, respectively. Incidences of grade 3-4 AEs > 10% were anorexia (19.0%), peripheral neuropathy (12.7%), nausea (11.1%), and thrombocytopenia (11.1%). CONCLUSIONS This study represents the first evaluation of SOX130 in patients with HER2-negative AGC. SOX130 showed an acceptable safety profile, but the prespecified statistical efficacy targets were not achieved.
Collapse
|
32
|
Wang T, Wang J, Ren W, Liu ZL, Cheng YF, Zhang XM. Combination treatment with artemisinin and oxaliplatin inhibits tumorigenesis in esophageal cancer EC109 cell through Wnt/β-catenin signaling pathway. Thorac Cancer 2020; 11:2316-2324. [PMID: 32657048 PMCID: PMC7396387 DOI: 10.1111/1759-7714.13570] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 01/20/2023] Open
Abstract
Background Esophageal cancer (EC) is a prevalent malignant cancer worldwide. Interestingly, the antimalaria compound artemisinin (ART) is also reported to have anticancer potential, although its underlying mechanism in EC is unclear. In this study, we explored the anticancer role of ART in EC109 and further explored the combination of ART and oxaliplatin (OXA) for their synergetic anticancer functions. Methods Human EC cell line EC109 was used. After ART or oxaliplatin (OXA) treatment, cell proliferation, migration, and invasion were measured by MTT, transwell, and scratch wound assays, respectively. Flow cytometry was performed to examine the cell cycle and apoptosis. The mRNA and protein levels were determined using qRT‐PCR and western blotting. Results The migration and invasion abilities of EC109 were suppressed by ART. This was due to the inhibitory effect of ART on the Wnt/β‐catenin signaling pathway. The levels of β‐catenin, c‐myc, and survivin were also downregulated by ART. ART inhibits the proliferation of EC109 cells by arresting the cells in the G1‐phase of cell cycle. By using LiCl, an activator of the Wnt/β‐catenin pathway, we further verified that the inhibition of the Wnt/β‐catenin pathway was indeed due to ART. Remarkably, ART enhanced the anticancer effects of OXA in EC109 cells. OXA combined with ART was found to be more efficient in decreasing tumor growth compared to the individual drugs. Conclusions ART could suppress tumor progression by inhibiting Wnt/β‐catenin signaling pathway, and it may also enhance the antitumor effect of OXA in EC. Thus, ART could be a novel anticancer drug for EC treatment. Key points Significant findings of the study ART could be a novel anticancer drug for esophageal cancer (EC) treatment. What this study adds Combination treatment with artemisinin and oxaliplatin inhibits tumorigenesis in esophageal cancer EC109 cells through the Wnt/β‐catenin signaling pathway.
Collapse
Affiliation(s)
- Tao Wang
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Wang
- Department of Ultrasound, Shandong Province Coal Taishan Sanatorium, Taian, China
| | - Wei Ren
- Department of Radiotherapy, The People's Hospital of Lanling County, Linyi, China
| | - Zhu-Long Liu
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Feng Cheng
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Mei Zhang
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
33
|
von Döbeln GA, Wagenius G, Holtved E, Jacobsen AB, Nilsson M, Yu J, Baeksgaard L. Definitive chemoradiotherapy plus cetuximab for cancer in the oesophagus or gastro-oesophageal junction. Cancer Treat Res Commun 2020; 24:100187. [PMID: 32619832 DOI: 10.1016/j.ctarc.2020.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Chemoradiotherapy is standard treatment for localized oesophageal cancer unsuitable for surgery. We aimed to evaluate the efficacy of cetuximab in combination with chemoradiotherapy. METHODS This non-randomised multicentre phase II trial recruited patients aged 18-75 with WHO performance status 0-2 having squamous cell carcinoma or adenocarcinoma in the oesophagus or gastro-oesophageal junction, T2-4, N0-3, M0 not suitable for surgery. Chemotherapy was three 21-day cycles of fluorouracil 750 mg/m2 D1-5 and oxaliplatin D1 (cycle 1:130mg/m 2, cycle 2-3:85 mg/m 2). Radiotherapy was 50Gy in 2Gy/fraction, 5 days a week, concurrent with cycle 2 and 3 and weekly cetuximab. The primary objective was loco-regional control at one year. RESULTS 52 patients were included. 51 were eligible for toxicity and survival analysis and 46 for recurrence analysis. Full radiotherapy dose was delivered to 80%, 75% received all three cycles of chemotherapy and 75% received four or more doses of cetuximab. The most common related grade III-IV adverse events were gastro-intestinal(16), hypersensitivity(6) and infection(5). There were two drug-related deaths. Within six months from the end of treatment, six patients died from complications from fistulas. The loco-regional control rate at one year was 47.3%(95%CI 30.9%-62.1%). Overall survival at three years was 29.1%(95% CI 17.4-41.9%). CONCLUSIONS Oxaliplatin and fluorouracil given concurrent with radiotherapy and cetuximab had an acceptable safety profile and showed a clinical response in patients with locoregionally advanced oesophageal cancer unsuitable for surgery. However, the primary end-point was not met, and the addition of cetuximab to definitive chemoradiotherapy cannot be recommended.
Collapse
Affiliation(s)
| | - Gunnar Wagenius
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Holtved
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | - Magnus Nilsson
- Department of Clinical Sciences Intervention and Technology Karolinska Institutet Division of Upper Abdominal Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Jingru Yu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
34
|
Bossi P, Airoldi M, Aloe Spiriti MA, Antonuzzo A, Bonciarelli G, Campagna A, Cassano A, Murialdo R, Musio D, Silvano G. A multidisciplinary expert opinion on CINV and RINV, unmet needs and practical real-life approaches. Expert Opin Drug Saf 2020; 19:187-204. [PMID: 32005072 DOI: 10.1080/14740338.2020.1724955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Introduction: A range of combination chemotherapy regimens are currently used in clinical practice. However, international antiemetic guidelines often only categorize the emetogenic potential of single agents rather than the emetogenicity of combination chemotherapy regimens. To manage the nausea and vomiting induced by antineoplastic combinations, guidelines suggest antiemetics that are appropriate for the component drug with the highest emetogenic potential. Furthermore, antiemetic guidelines generally do not consider the influence of other factors, including individual patient characteristics, on the emetic effects of cancer treatments. Similarly, the emetogenic potential of radiotherapy is stratified only according to the site of radiation, while other factors contributing to emetic risk are overlooked.Areas covered: An Expert Panel was convened to examine unresolved issues and summarize the current clinical research on managing nausea and vomiting associated with combination chemotherapy and radiotherapy.Expert opinion: The panel identified the incidence of nausea and vomiting induced by multi-drug combination therapies currently used to treat cancer at different anatomic sites and by radiotherapy in the presence of other risk factors. Based on these data and the clinical experience of panel members, several suggestions are made for a practical approach to prevent or manage nausea and vomiting due to chemotherapy regimens and radiation therapy.
Collapse
Affiliation(s)
- Paolo Bossi
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - Mario Airoldi
- Oncology Departement, Città della Salute e della Scienza Hospital of Turin, Turin, Italy
| | - Maria Antonietta Aloe Spiriti
- Department of Clinical and Molecular Medicine, Azienda Universitaria Ospedaliera Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Andrea Antonuzzo
- Medical Oncology Unit 1 SSN, Pisa University Hospital, Pisa, Italy
| | | | - Alessia Campagna
- Department of Hematology, Azienda Universitaria Ospedaliera Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Alessandra Cassano
- Division of Medical Oncology, Catholic University of the Sacred Heart, Rome, Italy
| | - Roberto Murialdo
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Daniela Musio
- Department of Radiotherapy, Policlinico Umberto I "Sapienza" University of Rome, Rome, Italy
| | - Giovanni Silvano
- Radiation Oncology Unit, San Giuseppe Moscati Hospital, Taranto, Italy
| |
Collapse
|
35
|
Arienti C, Pignatta S, Tesei A. Epidermal Growth Factor Receptor Family and its Role in Gastric Cancer. Front Oncol 2019; 9:1308. [PMID: 31850207 PMCID: PMC6901979 DOI: 10.3389/fonc.2019.01308] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022] Open
Abstract
Despite the gradual decrease in incidence, gastric cancer is still the third leading cause of cancer death worldwide. Although chemotherapy enhances overall survival and quality of life in advanced disease, the median overall survival is < 12 months. In recent years, the human epidermal growth factor receptor (ErbB) family has been extensively investigated in gastric cancer. The ErbB family is composed of four closely-related members: ErbB-1 (HER1 or epidermal growth factor receptor, EGFR), ErbB-2 (HER2), ErbB-3 (HER3), and ErbB-4 (HER4), all of which play a critical role in regulating cell growth, proliferation and migration of tumors. It is well known that gastric cancer overexpresses HER in a heterogeneous pattern, especially EGFR, and HER2. HER3 is another important member of the ErbB family that preferentially activates the phosphatidylinositol 3-kinase (PI3K) pathway. Furthermore, its heterodimerization with HER2 seems fundamental for steering HER2-overexpressing breast cancer tumor growth. Less is known about the impact of HER4 on gastric cancer. Improved survival from the use of trastuzumab has paved the way for ErbB receptor family-targeted treatments in gastric cancer. However, unlike trastuzumab, ErbB receptor-targeted drugs have not consistently maintained the encouraging results obtained in preclinical and early clinical trials. This may be attributable to the intrinsic heterogeneity of gastric cancer and/or to the lack of standardized test quality for established biomarkers used to evaluate these biological targets. This review presents an overview of the most recent clinical studies on agents targeting the ErbB family in gastric cancer.
Collapse
Affiliation(s)
| | | | - Anna Tesei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
36
|
Marcq E, Audenaerde JRV, Waele JD, Jacobs J, Loenhout JV, Cavents G, Pauwels P, Meerbeeck JPV, Smits EL. Building a Bridge between Chemotherapy and Immunotherapy in Malignant Pleural Mesothelioma: Investigating the Effect of Chemotherapy on Immune Checkpoint Expression. Int J Mol Sci 2019; 20:E4182. [PMID: 31455014 PMCID: PMC6747385 DOI: 10.3390/ijms20174182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
In light of the promising results of immune checkpoint blockade (ICPB) in malignant pleural mesothelioma (MPM), we investigated the effect of different chemotherapeutic agents on the expression of immune checkpoints (ICPs) in order to rationally design a good treatment schedule for their combination with ICP blocking antibodies. Cisplatin, oxaliplatin and pemetrexed are interesting chemotherapeutic agents to combine with immunotherapy given their immunomodulatory capacities. We looked into cisplatin and pemetrexed because their combination is used as first-line treatment of MPM. Additionally, the effect of the immunogenic chemotherapeutic agent, oxaliplatin, was also studied. Three different MPM cell lines were used for representation of both epithelioid and sarcomatoid subtypes. The desired inhibitory concentrations of the chemotherapeutic agents were determined with the SRB-assay. Allogeneic co-cultures of MPM cells with healthy donor peripheral blood mononuclear cells (PBMC) were set up to assess the effect of these chemotherapeutic agents on the expression of ICPs (PD-1, LAG-3, TIM-3) and their ligands (PD-L1, PD-L2, galectin-9). Cisplatin might be a promising treatment to combine with ICP blocking antibodies since our MPM cell lines were most susceptible to this stand-alone treatment. We found that the expression of ICPs and their ligands on both MPM cells and PBMC was mostly downregulated or unaltered when treated with chemotherapeutic agents, though no clear trend could be determined.
Collapse
Affiliation(s)
- Elly Marcq
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium.
| | | | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Julie Jacobs
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Jinthe Van Loenhout
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Glenn Cavents
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
- Department of Pathology, Antwerp University Hospital, Antwerp 2650, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
- Department of Pulmonology & Thoracic Oncology, Antwerp University Hospital, Antwerp 2650, Belgium
| | - Evelien Lj Smits
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp 2650, Belgium
| |
Collapse
|
37
|
Koemans WJ, van der Kaaij RT, Boot H, Buffart T, Veenhof AAFA, Hartemink KJ, Grootscholten C, Snaebjornsson P, Retel VP, van Tinteren H, Vanhoutvin S, van der Noort V, Houwink A, Hahn C, Huitema ADR, Lahaye M, Los M, van den Barselaar P, Imhof O, Aalbers A, van Dam GM, van Etten B, Wijnhoven BPL, Luyer MDP, Boerma D, van Sandick JW. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy versus palliative systemic chemotherapy in stomach cancer patients with peritoneal dissemination, the study protocol of a multicentre randomised controlled trial (PERISCOPE II). BMC Cancer 2019; 19:420. [PMID: 31060544 PMCID: PMC6501330 DOI: 10.1186/s12885-019-5640-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/25/2019] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND At present, palliative systemic chemotherapy is the standard treatment in the Netherlands for gastric cancer patients with peritoneal dissemination. In contrast to lymphatic and haematogenous dissemination, peritoneal dissemination may be regarded as locoregional spread of disease. Administering cytotoxic drugs directly into the peritoneal cavity has an advantage over systemic chemotherapy since high concentrations can be delivered directly into the peritoneal cavity with limited systemic toxicity. The combination of a radical gastrectomy with cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) has shown promising results in patients with gastric cancer in Asia. However, the results obtained in Asian patients cannot be extrapolated to Western patients. The aim of this study is to compare the overall survival between patients with gastric cancer with limited peritoneal dissemination and/or tumour positive peritoneal cytology treated with palliative systemic chemotherapy, and those treated with gastrectomy, CRS and HIPEC after neoadjuvant systemic chemotherapy. METHODS In this multicentre randomised controlled two-armed phase III trial, 106 patients will be randomised (1:1) between palliative systemic chemotherapy only (standard treatment) and gastrectomy, CRS and HIPEC (experimental treatment) after 3-4 cycles of systemic chemotherapy.Patients with gastric cancer are eligible for inclusion if (1) the primary cT3-cT4 gastric tumour including regional lymph nodes is considered to be resectable, (2) limited peritoneal dissemination (Peritoneal Cancer Index < 7) and/or tumour positive peritoneal cytology are confirmed by laparoscopy or laparotomy, and (3) systemic chemotherapy was given (prior to inclusion) without disease progression. DISCUSSION The PERISCOPE II study will determine whether gastric cancer patients with limited peritoneal dissemination and/or tumour positive peritoneal cytology treated with systemic chemotherapy, gastrectomy, CRS and HIPEC have a survival benefit over patients treated with palliative systemic chemotherapy only. TRIAL REGISTRATION clinicaltrials.gov NCT03348150 ; registration date November 2017; first enrolment November 2017; expected end date December 2022; trial status: Ongoing.
Collapse
Affiliation(s)
- W. J. Koemans
- Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - R. T. van der Kaaij
- Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - H. Boot
- Department of Gastro-Intestinal Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - T. Buffart
- Department of Gastro-Intestinal Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - A. A. F. A. Veenhof
- Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - K. J. Hartemink
- Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - C. Grootscholten
- Department of Gastro-Intestinal Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - P. Snaebjornsson
- Department of Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - V. P. Retel
- Department of Psychosocial Research and Epidomiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - H. van Tinteren
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - S. Vanhoutvin
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - V. van der Noort
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - A. Houwink
- Department of Anaesthesiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - C. Hahn
- Department of Anaesthesiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - A. D. R. Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - M. Lahaye
- Department of Radiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - M. Los
- Department of Oncology, Sint Antonius Hospital, Koekoekslaan 1, Nieuwegein, 3435 CM The Netherlands
| | | | - O. Imhof
- Clinical perfusion, Heartbeat, Kerkstraat 3A, Eemnes, 3755 CK The Netherlands
| | - A. Aalbers
- Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| | - G. M. van Dam
- Department of Surgery, University Medical Center Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
| | - B. van Etten
- Department of Surgery, University Medical Center Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
| | - B. P. L. Wijnhoven
- Department of Surgery, Erasmus Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD The Netherlands
| | - M. D. P. Luyer
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, Eindhoven, 5623 EJ The Netherlands
| | - D. Boerma
- Department of Surgery, Sint Antonius Hospital, Koekoekslaan 1, Nieuwegein, 3435 CM The Netherlands
| | - J. W. van Sandick
- Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066CX The Netherlands
| |
Collapse
|
38
|
Han G, Shi J, Mi L, Li N, Shi H, Li C, Shan B, Yin F. Clinical efficacy and safety of paclitaxel liposomes as first-line chemotherapy in advanced gastric cancer. Future Oncol 2019; 15:1617-1627. [PMID: 31038363 DOI: 10.2217/fon-2018-0439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 02/11/2019] [Indexed: 01/19/2023] Open
Abstract
Aim: To compare the performance of first-line paclitaxel liposome + oxaliplatin and SOX (tegafur/gimeracil/oteracil + oxaliplatin) in advanced gastric cancer patients. Materials & methods: Stage IIb-IV gastric cancer patients underwent either first-line paclitaxel liposome + oxaliplatin (n = 52) or SOX (n = 69) between 2010-2013, and followed up until 2015 or death. Results: Both groups had similar objective response rate (p = 0.48) and disease control rate (p = 0.992) after two chemotherapy cycles, median progression-free survival (p = 0.495) and median overall survival (p = 0.208). Liposome group had significantly lower rate of grade I-II platelet decline and liver function damage (p = 0.04 and 0.019). Multivariate COX regression identified pre-treatment neutrophil-to-lymphocyte ratio as an independent prognostic factor. Conclusion: First-line paclitaxel liposome + oxaliplatin has comparable efficacy, but causes reduced adverse reactions in advanced gastric cancer as compared with SOX.
Collapse
Affiliation(s)
- Guangjie Han
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Jianfei Shi
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Lili Mi
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Ning Li
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Huacun Shi
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Cuizhen Li
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Baoen Shan
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| | - Fei Yin
- Department of Gastroenterology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Heibei, PR China
| |
Collapse
|
39
|
Zhang F, Zhang Y, Jia Z, Wu H, Gu K. Oxaliplatin-Based Regimen is Superior to Cisplatin-Based Regimen in Tumour Remission as First-line Chemotherapy for Advanced Gastric Cancer: A Meta-Analysis. J Cancer 2019; 10:1923-1929. [PMID: 31205551 PMCID: PMC6547983 DOI: 10.7150/jca.28896] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/19/2019] [Indexed: 01/20/2023] Open
Abstract
Background: This study was initially designed to examine whether oxaliplatin-based regimen was superior to cisplatin-based regimen in tumour remission as first-line chemotherapy for advanced gastric cancer (GC). Methods: Literature in EMBASE, PUBMED, Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, American Society of Clinical Oncology (ASCO) and European Society of Medical Oncology (ESMO) was searched. Only phase II or III randomized controlled trials (RCTs) comparing the effectiveness and safety between oxaliplatin-based and cisplatin-based regimens as first-line treatment for advanced GC were selected. Odds ratios (ORs) with 95% confidence intervals (CIs) were reported. The primary endpoints were complete remission rate (CRR), partial remission rate (PRR), objective response rate (ORR), and disease control rate (DCR). The second endpoint was the toxicity response. Results: 2,140 patients from six phase II or III RCTs were included. Compared to cisplatin-based therapy, subjects who received oxaliplatin-based treatment had significantly higher PRR (OR: 1.25, 95%CI: 1.05-1.48, P=0.01, I2=0%), ORR (OR: 1.21, 95%CI: 1.02-1.44, P=0.03, I2=0%) and DCR (OR: 1.76, 95%CI: 1.31-2.38, P=0.0002, I2=25%), but not CRR (OR: 0.70, 95%CI: 0.37-1.31, P=0.27, I2=0%). In addition, oxaliplatin-based therapy significantly decreased all grades of leukopenia, neutropenia, anemia, febrile neutropenia, nausea, stomatitis, creatinine elevation and thromboembolism, as well as grades 3-4 of leukopenia, neutropenia, anemia and febrile neutropenia than cisplatin-based regimen. However, oxaliplatin-based treatment strikingly increased the risk of thrombocytopenia, sensory neuropathy, diarrhea, fatigue and liver dysfunction. Conclusions: Oxaliplatin-based regimen is superior to cisplatin-based regimen in tumour remission as first-line chemotherapy for advanced GC, and is associated with less toxicity and better tolerability.
Collapse
Affiliation(s)
| | | | | | | | - Kangsheng Gu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| |
Collapse
|
40
|
Zangui M, Atkin SL, Majeed M, Sahebkar A. Current evidence and future perspectives for curcumin and its analogues as promising adjuncts to oxaliplatin: state-of-the-art. Pharmacol Res 2019; 141:343-356. [DOI: 10.1016/j.phrs.2019.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023]
|
41
|
Tanaka M, Okinaga T, Iwanaga K, Matsuo K, Toyono T, Sasaguri M, Ariyoshi W, Tominaga K, Enomoto Y, Matsumura Y, Nishihara T. Anticancer effect of novel platinum nanocomposite beads on oral squamous cell carcinoma cells. J Biomed Mater Res B Appl Biomater 2019; 107:2281-2287. [PMID: 30689290 DOI: 10.1002/jbm.b.34320] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 01/19/2023]
Abstract
Nanoparticles are used in industry and medicine, because of their physiochemical properties, such as size, charge, large surface area and surface reactivity. Recently, metal nanoparticles were reported to show cell toxicity on cancer cells. In this study, we focused novel platinum nanoparticles-conjugated latex beads (P2VPs), platinum nanocomposite (PtNCP) beads, and investigated the possibility to incorporate novel anti-cancer effect of these combined nanoparticles. Oral squamous cell carcinoma cell lines, HSC-3-M3 cells were injected subcutaneously into the back of nude mice to produce a xenograft model. PtNCP beads were injected locally and examined by measuring tumor volume and comparing pathological histology. PtNCP beads treatment suppressed tumor growth and identified increasing pathological necrotic areas, in vivo. PtNCP beads inhibited the cell viability of HSC-3-M3 cells in dose-dependent manner and induced the cytotoxicity with extracellular LDH value, in vitro. Furthermore, SEM images were morphologically observed in PtNCP beads-treated HSC-3-M3 cells. The aggregation of the PtNCP beads on the cell membrane, the destructions of the cell membrane and globular structures were observed in the SEM image. Our results indicated that a potential anti-cancer effect of the PtNCP beads, suggesting the possibility as a therapeutic tool for cancer cell-targeted therapy. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 2281-2287, 2019.
Collapse
Affiliation(s)
- Mai Tanaka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan.,Division of Oral and Maxillofacial Surgery, Department of Science of physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | | | - Kenjiro Iwanaga
- Division of Preventive Dentistry, Department of Oral Health and Development Sciences, Tohoku University, Sendai, Japan
| | - Kou Matsuo
- Division of Oral Pathology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Takashi Toyono
- Division of Oral Histology and Neurobiology, Kyushu Dental College, Kitakyushu, Japan
| | - Masaaki Sasaguri
- Division of Oral and Maxillofacial Surgery, Department of Science of physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Kazuhiro Tominaga
- Division of Oral and Maxillofacial Surgery, Department of Science of physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | - Yasushi Enomoto
- New Materials Development Center, Nippon Steel & Sumikin Chemical Co., Ltd, Chiba, Japan
| | - Yasufumi Matsumura
- New Materials Development Center, Nippon Steel & Sumikin Chemical Co., Ltd, Chiba, Japan
| | - Tatsuji Nishihara
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| |
Collapse
|
42
|
van den Ende T, Ter Veer E, Machiels M, Mali RMA, Abe Nijenhuis FA, de Waal L, Laarman M, Gisbertz SS, Hulshof MCCM, van Oijen MGH, van Laarhoven HWM. The Efficacy and Safety of (Neo)Adjuvant Therapy for Gastric Cancer: A Network Meta-analysis. Cancers (Basel) 2019; 11:E80. [PMID: 30641964 PMCID: PMC6356558 DOI: 10.3390/cancers11010080] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/05/2019] [Accepted: 01/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Alternatives in treatment-strategies exist for resectable gastric cancer. Our aims were: (1) to assess the benefit of perioperative, neoadjuvant and adjuvant treatment-strategies and (2) to determine the optimal adjuvant regimen for gastric cancer treated with curative intent. Methods: PubMed, EMBASE, CENTRAL, and ASCO/ESMO conferences were searched up to August 2017 for randomized-controlled-trials on the curative treatment of resectable gastric cancer. We performed two network-meta-analyses (NMA). NMA-1 compared perioperative, neoadjuvant and adjuvant strategies only if there was a direct comparison. NMA-2 compared different adjuvant chemo(radio)therapy regimens, after curative resection. Overall-survival (OS) and disease-free-survival (DFS) were analyzed using random-effects NMA on the hazard ratio (HR)-scale and calculated as combined HRs and 95% credible intervals (95% CrIs). Results: NMA-1 consisted of 9 direct comparisons between strategies for OS (14 studies, n = 4187 patients). NMA-2 consisted of 16 direct comparisons between adjuvant chemotherapy/chemoradiotherapy regimens for OS (37 studies, n = 10,761) and 14 for DFS (30 studies, n = 9714 patients). Compared to taxane-based-perioperative-chemotherapy, surgery-alone (HR = 0.58, 95% CrI = 0.38⁻0.91) and perioperative-chemotherapy regimens without a taxane (HR = 0.79, 95% CrI = 0.58⁻1.15) were inferior in OS. After curative-resection, the doublet oxaliplatin-fluoropyrimidine (for one-year) was the most efficacious adjuvant regimen in OS (HR = 0.47, 95% CrI = 0.28⁻0.80). Conclusions: For resectable gastric cancer, (1) taxane-based perioperative-chemotherapy was the most promising treatment strategy; and (2) adjuvant oxaliplatin-fluoropyrimidine was the most promising regimen after curative resection. More research is warranted to confirm or reproach these findings.
Collapse
Affiliation(s)
- Tom van den Ende
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Emil Ter Veer
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Mélanie Machiels
- Department of Radiotherapy, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Rosa M A Mali
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Frank A Abe Nijenhuis
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Laura de Waal
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Marety Laarman
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Suzanne S Gisbertz
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Maarten C C M Hulshof
- Department of Radiotherapy, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC) location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Calderon-Aparicio A, Cornejo A, Orue A, Rieber M. Anticancer response to disulfiram may be enhanced by co-treatment with MEK inhibitor or oxaliplatin: modulation by tetrathiomolybdate, KRAS/BRAF mutations and c-MYC/p53 status. Ecancermedicalscience 2019; 13:890. [PMID: 30792807 PMCID: PMC6369974 DOI: 10.3332/ecancer.2019.890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Indexed: 12/22/2022] Open
Abstract
Ammonium tetrathiomolybdate (TTM) and disulfiram (DSF) are copper (Cu) chelators in cancer clinical trials partly because Cu chelation: a) restricts the activity of Cu-binding MEK1/2 enzymes which drive tumourigenesis by KRAS or BRAF oncogenic mutations and b) enhances uptake of oxaliplatin (OxPt), clinically used in advanced KRAS-mutant colorectal carcinomas (CRC). Whereas TTM decreases intracellular Cu trafficking, DSF can reach other Cu-dependent intracellular proteins. Since the use of individual or combined Cu chelation may help or interfere with anti-cancer therapy, this study investigated whether TTM modifies the response to DSF supplemented with: 1) UO126, a known MEK1/2 inhibitor; 2) other Cu chelators like neocuproine (NC) or 1, 10-o-phenanthroline (OPT) in wt p53 melanoma cells differing in BRAF or KRAS mutations; 3) OxPt in mutant p53 CRC cells devoid of KRAS and BRAF mutations or harbouring either KRAS or BRAF mutations. TTM was not toxic against V600E-mut-BRAF A375 and G12D-mut-KRAS/high c-myc C8161 melanoma cells. Moreover, TTM protected both melanoma types from toxicity induced by DSF, NC and co-treatment with sub-lethal levels of DSF and the MEK inhibitor, UO126. Toxicity by co-treatment with DSF+OPT was poorly reversed by TTM in C8161 melanoma cells. In contrast to the greater toxicity of 0.1 μM DSF against mutant p53 CRC cells irrespective of their KRAS mutation, TTM did not protect G12V-mut-KRAS/high c-myc SW620 CRC from DSF+OxPt compared to KRAS-WT/BRAF-WT Caco-2 CRC. Our results show that DSF co-treatment with: a) MEK inhibitors may enhance tumour suppression; b) OxPt in CRC may counteract impaired response to cetuximab by KRAS/BRAF mutations and c) as a single treatment, TTM may be less effective than DSF and decreases the efficacy of the latter.
Collapse
Affiliation(s)
| | | | - Andrea Orue
- Instituto Venezolano de Investigaciones Cientificas, Tumor Cell Biology Laboratory, Caracas 1020-A, Venezuela.,These authors contributed equally to this work
| | - Manuel Rieber
- Instituto Venezolano de Investigaciones Cientificas, Tumor Cell Biology Laboratory, Caracas 1020-A, Venezuela.,These authors contributed equally to this work
| |
Collapse
|
44
|
Ghasemi E, Ghaffarifar F, Dalimi A, Sadraei J. In-vitro and In-vivo Antileishmanial Activity of a Compound Derived of Platinum, Oxaliplatin, against Leishmania Major. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2019; 18:2028-2041. [PMID: 32184867 PMCID: PMC7059061 DOI: 10.22037/ijpr.2019.15364.13046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This study aimed to evaluate the antileishmanial efficacy of oxaliplatin against Leishmania major (L. major) both in-vitro and in-vivo. The IC50, CC50, and SI of oxaliplatin against promastigotes, murine macrophages, Raw 264.7 cells, and intramacrophage amastigotes of L. major were investigated in-vitro. The effects of this drug on intracellular amastigotes were also assayed, and the percentage of infectivity and IIR were calculated. Flow cytometry was performed to assay apoptosis, using 50 and 100 µg/mL of oxaliplatin in the promastigotes and macrophages. In-vivo, the BALB/c mice were classified into three groups, receiving oxaliplatin, glucantime, and phosphate-buffered saline for one month, respectively. The lesion size, IFN-γ, and IL-4 levels, and parasite burden were also evaluated in the animals. After 72 h, the IC50 and CC50 of oxaliplatin against promastigotes and macrophages were respectively 0.5 and 66.78 µg/mL. The apoptosis of promastigotes and macrophages using 50 µg/mL of oxaliplatin was 7.25% and 2.14%, respectively, while apoptosis induced at 100 µg/mL was 15.48% and 2.80%, respectively. Similar to the glucantime group, the mice treated with oxaliplatin showed a lower parasite burden and smaller lesions, compared with the PBS group (p < 0.01). Furthermore, higher IFN-γ levels were reported in mice receiving oxaliplatin in comparison with those receiving PBS (p < 0.01). The current findings indicated the efficacy of oxaliplatin against promastigote and amastigote forms of Leishmania and L. major-induced leishmaniasis.
Collapse
Affiliation(s)
| | - Fatemeh Ghaffarifar
- Department of Parasitology and Entomology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | | |
Collapse
|
45
|
The interactions of novel mononuclear platinum-based complexes with DNA. BMC Cancer 2018; 18:1284. [PMID: 30577821 PMCID: PMC6303901 DOI: 10.1186/s12885-018-5194-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
Background Cisplatin has been widely used for the treatment of cancer and its antitumour activity is attributed to its capacity to form DNA adducts, predominantly at guanine residues, which impede cellular processes such as DNA replication and transcription. However, there are associated toxicity and drug resistance issues which plague its use. This has prompted the development and screening of a range of chemotherapeutic drug analogues towards improved efficacy. The biological properties of three novel platinum-based compounds consisting of varying cis-configured ligand groups, as well as a commercially supplied compound, were characterised in this study to determine their potential as anticancer agents. Methods The linear amplification reaction was employed, in conjunction with capillary electrophoresis, to quantify the sequence specificity of DNA adducts induced by these compounds using a DNA template containing telomeric repeat sequences. Additionally, the DNA interstrand cross-linking and unwinding efficiency of these compounds were assessed through the application of denaturing and native agarose gel electrophoresis techniques, respectively. Their cytotoxicity was determined in HeLa cells using a colorimetric cell viability assay. Results All three novel platinum-based compounds were found to induce DNA adduct formation at the tandem telomeric repeat sequences. The sequence specificity profile at these sites was characterised and these were distinct from that of cisplatin. Two of these compounds with the enantiomeric 1,2-diaminocyclopentane ligand (SS and RR-DACP) were found to induce a greater degree of DNA unwinding than cisplatin, but exhibited marginally lower DNA cross-linking efficiencies. Furthermore, the RR-isomer was more cytotoxic in HeLa cells than cisplatin. Conclusions The biological characteristics of these compounds were assessed relative to cisplatin, and a variation in the sequence specificity and a greater capacity to induce DNA unwinding was observed. These compounds warrant further investigations towards developing more efficient chemotherapeutic drugs.
Collapse
|
46
|
Park JK, Seo JS, Lee SK, Chan KK, Kuh HJ. Combinatorial Antitumor Activity of Oxaliplatin with Epigenetic Modifying Agents, 5-Aza-CdR and FK228, in Human Gastric Cancer Cells. Biomol Ther (Seoul) 2018; 26:591-598. [PMID: 30173503 PMCID: PMC6254647 DOI: 10.4062/biomolther.2018.061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/13/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Epigenetic silencing is considered to be a major mechanism for loss of activity in tumor suppressors. Reversal of epigenetic silencing by using inhibitors of DNA methyltransferase (DNMT) or histone deacetylases (HDACs) such as 5-Aza-CdR and FK228 has shown to enhance cytotoxic activities of several anticancer agents. This study aims to assess the combinatorial effects of gene-silencing reversal agents (5-Aza-CdR and FK228) and oxaliplatin in gastric cancer cells, i.e., Epstein-Barr virus (EBV)-negative SNU-638 and EBV-positive SNU-719 cells. The doublet combinatorial treatment of 5-Aza-CdR and FK228 exhibited synergistic effects in both cell lines, and this was further corroborated by Zta expression induction in SNU-719 cells. Three drug combinations as 5-Aza-CdR/FK228 followed by oxaliplatin, however, resulted in antagonistic effects in both cell lines. Simultaneous treatment with FK228 and oxaliplatin induced synergistic and additive effects in SNU-638 and SNU-719 cells, respectively. Three drug combinations as 5-Aza-CdR prior to FK228/oxaliplatin, however, again resulted in antagonistic effects in both cell lines. This work demonstrated that efficacy of doublet synergistic combination using DNMT or HDACs inhibitors can be compromised by adding the third drug in pre- or post-treatment approach in gastric cancer cells. This implies that the development of clinical trial protocols for triplet combinations using gene-silencing reversal agents should be carefully evaluated in light of their potential antagonistic effects.
Collapse
Affiliation(s)
- Jong Kook Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jung Seon Seo
- Department of Biomedicine & Health Science, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Suk Kyeong Lee
- Department of Biomedicine & Health Science, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kenneth K Chan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Science, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea.,Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea.,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
47
|
Real-Time Tumor Gene Expression Profiling to Direct Gastric Cancer Chemotherapy: Proof-of-Concept “3G” Trial. Clin Cancer Res 2018; 24:5272-5281. [DOI: 10.1158/1078-0432.ccr-18-0193] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/28/2018] [Accepted: 07/12/2018] [Indexed: 11/16/2022]
|
48
|
Liu D, Li X, Chen C, Li C, Zhou C, Zhang W, Zhao J, Fan J, Cheng K, Chen L. Target-specific delivery of oxaliplatin to HER2-positive gastric cancer cells in vivo using oxaliplatin-au-fe3o4-herceptin nanoparticles. Oncol Lett 2018; 15:8079-8087. [PMID: 29731915 DOI: 10.3892/ol.2018.8323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 12/14/2017] [Indexed: 01/14/2023] Open
Abstract
Gastric cancer is the fourth most common malignancy globally. In order to decrease the dosage and side effects of conventional chemotherapy, and achieve improved benefits from molecular targeted therapy, novel drug delivery systems were developed in the present study. Oxaliplatin-Au-Fe3O4-Herceptin® acts as a dual-functional nanoparticles (NPs) conjugate and possesses the capability of human epithelial growth factor receptor 2 (HER2) targeting and oxaliplatin delivery. The 8-20 nm Au-Fe3O4 were synthesized by decomposing iron pentacarbonyl on the surfaces of Au NPs in the presence of oleic acid and oleylamine. Following being coated with polyethylene glycol, the NPs possessed a ζ-potential of 13.8±1.6 mV and were demonstrated to exhibit no cytotoxicity when Fe concentration is <100 µg/ml via an MTS assay. Mass spectrometry analysis detected a peak at m/z 148,000, and Nuclear Magnetic Resonance indicated peaks at δ 3.51 (8.00H, s, 3-H), 2.97-3.02 (3.80H, t, 2-H) and 2.72-2.76 (3.72H, t, 1-H) following successful loading with Herceptin and oxaliplatin probes. A drug release assay via dialysis cassettes demonstrated that 25% of the oxaliplatin was released at pH 8.0, however >58% was released at pH 6.0 following 4 h incubation, indicating its pH-dependent release characteristic. The active targeting feature of oxaliplatin-Au-Fe3O4-Herceptin was verified in a subcutaneous xenograft mouse model containing SGC-7901 cells via detecting aggregated low intensity in T2-weighted magnetic resonance imaging, which was further confirmed by immunohistochemistry. Therefore, oxaliplatin-Au-Fe3O4-Herceptin is a promising multifunctional platform for simultaneous magnetic traceable and HER2 targeted chemotherapy for gastric cancer.
Collapse
Affiliation(s)
- Daren Liu
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaowen Li
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Changlei Chen
- Key Laboratory of Applied Chemistry of Zhejiang, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Chao Li
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Chuanbiao Zhou
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Weidong Zhang
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jiangang Zhao
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jie Fan
- Key Laboratory of Applied Chemistry of Zhejiang, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Kai Cheng
- Molecular Imaging Program, Canary Center for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University, Stanford, CA 94305, USA
| | - Li Chen
- Department of General Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
49
|
Management of advanced gastric cancer: An overview of major findings from meta-analysis. Oncotarget 2018; 7:78180-78205. [PMID: 27655725 PMCID: PMC5363654 DOI: 10.18632/oncotarget.12102] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/26/2016] [Indexed: 12/19/2022] Open
Abstract
This study aims to provide an overview of different treatment for advanced gastric cancer. In the present study, we systematically reviewed the major findings from relevant meta-analyses. A total of 54 relevant papers were searched via the PubMed, Web of Science, and Google scholar databases. They were classified according to the mainstay treatment modalities such as surgery, chemotherapy and others. Primary outcomes including overall survival, response rate, disease-free survival, recurrence-free survival, progression-free survival, time-to-progression, time-to failure, recurrence and safety were summarized. The recommendations and uncertainties regarding the treatment of advanced gastric cancer were also proposed. It was suggested that laparoscopic gastrectomy was a safe and technical alternative to open gastrectomy. Besides, neoadjuvant chemotherapy and adjuvant chemotherapy were thought to benefit the survival over surgery alone. And it was demonstrated in the study that targeted therapy like anti-angiogenic and anti-HER2 agents but anti-EGFR agent might have a significant survival benefit.
Collapse
|
50
|
Fahmy SA, Ponte F, Abd El-Rahman MK, Russo N, Sicilia E, Shoeib T. Investigation of the host-guest complexation between 4-sulfocalix[4]arene and nedaplatin for potential use in drug delivery. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 193:528-536. [PMID: 29306207 DOI: 10.1016/j.saa.2017.12.070] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/10/2017] [Accepted: 12/26/2017] [Indexed: 05/28/2023]
Affiliation(s)
- Sherif Ashraf Fahmy
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria, Arcavacata di Rende, 87036, Italy
| | - Mohamed K Abd El-Rahman
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr-El Aini Street, Cairo, Egypt 11562
| | - Nino Russo
- Department of Chemistry and Chemical Technologies, University of Calabria, Arcavacata di Rende, 87036, Italy; Division de Ciencias Basicas e Ingenieria, Departamento de Quimica, Universidad, Autonoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco No. 186, Col. Vicentina, CP 09340 Mexico, Distrito Federal, Mexico
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, University of Calabria, Arcavacata di Rende, 87036, Italy.
| | - Tamer Shoeib
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt.
| |
Collapse
|