1
|
Martinez P, Sabatier JM. Rethinking corticosteroids use in oncology. Front Pharmacol 2025; 16:1551111. [PMID: 40206059 PMCID: PMC11979161 DOI: 10.3389/fphar.2025.1551111] [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: 12/24/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Corticosteroids (CSs), widely used in oncology for their anti-inflammatory and immunosuppressive properties, help manage cancer-related symptoms and side effects. However, their long-term use may negatively affect patient survival and exacerbate tumor progression. Elevated glucose and glutamine metabolism, disruption of vitamin D levels, and alterations in the microbiome are some of the key factors contributing to these adverse outcomes. Approaches such as ketogenic diets, fasting, sartans, and vitamin D supplementation have shown promise in providing similar benefits to CSs while mitigating the risks associated with the mechanisms identified as contributing to tumor progression. This perspective underscores the necessity for a reevaluation of CSs use in cancer care and advocates for further research into safer, more effective therapeutic strategies.
Collapse
Affiliation(s)
| | - Jean-Marc Sabatier
- Institut de NeuroPhysiopathologie (INP), CNRS UMR 7051, Marseille, France
| |
Collapse
|
2
|
Huang X, Lin K, Chen W, Zhang D, Khan M, Ye X, Wang B, Chen C, Tian Y, Yuan Y, Lin J. Modulation of the local angiotensin II: Suppression of ferroptosis and radiosensitivity in nasopharyngeal carcinoma via the HIF-1α-HILPDA axis. Radiother Oncol 2025; 203:110686. [PMID: 39709027 DOI: 10.1016/j.radonc.2024.110686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 11/06/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE Radiotherapy presents a curative approach for nasopharyngeal carcinoma (NPC); however, the cellular radiosensitivity heterogeneity limits its efficacy. Thus, investigating the specific mechanisms of radioresistance in NPC is crucial for identifying and employing effective radiosensitizing agents to enhance treatment success. METHODS AND MATERIALS Radioresistant NPC cell lines HONE1-RR and SUNE1-RR were established. Quantitative reverse transcription-PCR (qRT-PCR), western blot, and enzyme-linked immuno sorbent assay (ELISA) were employed to detect the activation of the angiotensinogen (AGT) and local angiotensin II (Ang II). Transmission electron microscopy, ferrous ion detection, and lipid oxidation levels were utilized to detect radiation-induced ferroptosis in NPC. Bioinformatics analysis, along with qRT-PCR, western blotting, co-immunoprecipitation, and dual-luciferase assays were employed to explore downstream mechanisms. Colony formation assay, Cell Counting Kit-8 (CCK-8) assay, and a nude mouse xenograft model were utilized to assess NPC radiosensitivity. The expression of AGT, hypoxia-inducible factor-1 alpha (HIF-1α), hypoxia-inducible lipid droplet-associated protein (HILPDA), and glutathione peroxidase 4 (GPX4) in NPC tissues was detected through immunohistochemistry. RESULTS Activation of local Ang II was revealed to play a critical role in driving radioresistance in NPC cells modulating ferroptosis. This local Ang II established a positive feedback loop with HIF-1α through two parallel pathways; Ang II stabilizes HIF-1α by activating the MAPK pathway, and AGT directly binds HIF-1α to prevent its degradation. This AGT-HIF-1α loop regulated NPC cell ferroptosis via transcriptional regulation of HILPDA expression. Moreover, the co-administration of Ang II receptor antagonist (ARB) and ferroptosis inducers markedly increased NPC radiosensitivity.Additionally, the expression of AGT, HIF-1α, and HILPDA was closely correlated with the intensity of ferroptosis, radiosensitivity, and prognosis in NPC. CONCLUSIONS Our findings suggest that the AGT-HIF-1α-HILPDA pathway promotes radioresistance in NPC by enhancing lipid droplet accumulation, thereby suppressing ferroptosis. Targeting local Ang II alongside ferroptosis induction offers a promising strategy to improve radiosensitivity in NPC.
Collapse
Affiliation(s)
- Xiuting Huang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Kehai Lin
- Department of Oncology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000, China
| | - Weirui Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Donghui Zhang
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Xiaoxin Ye
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Baiyao Wang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Chengcong Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yunhong Tian
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Yawei Yuan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Jie Lin
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China.
| |
Collapse
|
3
|
Wu LY, Su BC, Yu HH, Cheng CC, Tsai CC, Hsu PL, Lee CW. Antihypertensive agent losartan promotes tongue squamous cell carcinoma cell proliferation via EGFR/ERK1/2/cyclin D1 signaling axis. J Oral Biosci 2024; 66:74-80. [PMID: 39245205 DOI: 10.1016/j.job.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE To study the effects of losartan, an angiotensin II receptor blocker, in the SCC4 and SCC25 human tongue squamous cell carcinoma cell lines. METHODS Cell proliferation was measured by MTS/PMS activity and trypan blue exclusion assays. The levels of the cell proliferation marker, cyclin D1, were analyzed by western blotting. Apoptosis was assessed by caspase-3 activation and Annexin V-FITC/propidium iodide double staining. Activation of epidermal growth factor receptor (EGFR) and ERK1/2 was validated by western blotting. RESULTS Moderate concentrations of losartan enhanced the proliferation of SCC4 and SCC25 cells. However, high losartan concentrations induced apoptosis in SCC4 cells. Losartan activated the EGFR/ERK1/2/cyclin D1 signaling axis, which in turn promoted cell proliferation. Afatinib (EGFR inhibitor) and U0126 (ERK1/2 inhibitor) abolished losartan-induced cell proliferation. In contrast, UC2288 (p21 inhibitor) enhanced it. CONCLUSIONS Losartan exhibited dual effects on tongue squamous cell carcinoma cells. Moderate losartan concentrations facilitated cell proliferation, whereas high concentrations induced cytotoxicity in tongue carcinoma cells.
Collapse
Affiliation(s)
- Luo-Yun Wu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Bor-Chyuan Su
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Hsien Yu
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Cheng Cheng
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chia-Chi Tsai
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Pei-Ling Hsu
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Chu-Wan Lee
- Department of Nursing, National Tainan Junior College of Nursing, 78, Section 2, Minzu Road, West Central District, Tainan, 70007, Taiwan.
| |
Collapse
|
4
|
Georgiou N, Chontzopoulou E, Routsi EA, Stavrakaki IG, Petsas E, Zoupanou N, Kakava MG, Tzeli D, Mavromoustakos T, Kiriakidi S. Exploring Hypertension: The Role of AT1 Receptors, Sartans, and Lipid Bilayers. ACS OMEGA 2024; 9:44876-44890. [PMID: 39554401 PMCID: PMC11561769 DOI: 10.1021/acsomega.4c06351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024]
Abstract
The rational design of AT1 receptor antagonists represents a pivotal approach in the development of therapeutic agents targeting cardiovascular pathophysiology. Sartans, a class of compounds engineered to inhibit the binding and activation of Angiotensin II on the AT1 receptor, have demonstrated significant clinical efficacy. This review explores the multifaceted role of sartans in mitigating hypertension and related complications. We highlight the integration of crystallography, computational simulations, and NMR spectroscopy to elucidate sartan-AT1 receptor interactions, providing a foundation for the next-generation antagonist design. The review also delves into the challenges posed by the high lipophilicity and suboptimal bioavailability of sartans, emphasizing advancements in nanotechnology and novel drug delivery systems. Additionally, we discuss the impact of lipid bilayers on the AT1 receptor conformation and drug binding, underscoring the importance of the lipidic environment in receptor-drug interactions. We suggest that optimizing drug design to account for these factors could enhance the therapeutic potential of AT1 receptor antagonists, paving the way for improved cardiovascular health outcomes.
Collapse
Affiliation(s)
- Nikitas Georgiou
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Eleni Chontzopoulou
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Efthymios Alexandros Routsi
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Irene Georgia Stavrakaki
- Industrial
Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Errikos Petsas
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Nikoletta Zoupanou
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Margarita Georgia Kakava
- Laboratory
of Organic Chemistry and Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Demeter Tzeli
- Laboratory
of Physical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis
Zografou, 15771 Athens, Greece
- Theoretical
and Physical Chemistry Institute, National
Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Thomas Mavromoustakos
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Sofia Kiriakidi
- Laboratory
of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
- Departamento
de Quimica Orgánica, Facultade de
Quimica, Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
5
|
Mansour AMA, Khattab MM, El-Khatib AS, Awaad AK, El-Refaie WM, El-Mezayen NS. Valsartan as a prophylactic treatment against breast cancer development and niche activation: What molecular sequels follow chronic AT-1R blockade? Life Sci 2024; 353:122939. [PMID: 39094905 DOI: 10.1016/j.lfs.2024.122939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/07/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
AIMS Transactivation of insulin-growth-factor-receptor (IGF-1R) by angiotensin-II-type-1-receptor (AT-1R) was only demonstrated in vascular-smooth-muscle cells and has never been tested in breast-cancer (BC). This investigation addressed the impact of chronic AT-1R blockade by valsartan (Val) on possible concurrent AT-1R/IGF-1R signaling inhibition, regressing BC-tumor-microenvironment (TME) cellular components activation, and hindering BC development. MAIN METHODS The effect of different Val doses (10, 20, 40 & 80 mg/kg/day for 490 days) was tested on dimethylbenz(a)anthracene (DMBA)-induced progesterone-promoted-BC in rats. The influence on intratumoral/circulating angiotensin-II (ANG-II) levels and AT-1R/Mas-R immunofluorescent-expression were assessed. The potential AT-1R/IGF-1R crosstalk within TME-BC-stem-cells (BCSCs) and cancer-associated-fibroblasts (CAFs) was evaluated by fluorescently marking these cells and locating the immunofluorescently-stained AT-1R/IGF-1R in them using confocal-laser-microscopy and further quantified by flow cytometry. In addition, the molecular alterations following blocking AT-1R were inspected including determining Src; crucial for IGF-1R transactivation by AT-1R, Notch-1; IGF-IR transcriptional-regulator, and PI3K/Akt &IL-6/STAT expression. Further, the suppression of CSCs' capabilities to maintain pluripotency, stemness features, epithelial-to-mesenchymal-transition (EMT), and angiogenesis was evaluated by assessing NANOG gene, aldehyde-dehydrogenase (ALDH), N-cadherin and vascular-endothelial-growth-factor (VEGF), respectively. Furthermore, the proliferative marker; Ki-67, was detected by immunostaining, and tumors were histologically graded using Elston-Ellis-modified-Scarff-Bloom-Richardson method. KEY FINDINGS Prophylactic Val significantly reduced tumor size, prolonged latency, reduced tumor histopathologic grade, decreased circulating/intratumoral-ANG-II levels, increased Mas-R, and decreased AT1R expression. AT-1R/IGF-1R were co-expressed with a high correlation coefficient on CAFs/BCSCs. Moreover, Val significantly attenuated IGF-1R transactivation and transcriptional regulation via Src and Notch-1 genes' downregulation and reduced Src/IGF-IR-associated PI3K/Akt and IL-6/STAT3 signaling. Further, Val significantly decreased intratumoral NANOG, ALDH, N-cadherin, VEGF, and Ki-67 levels. SIGNIFICANCE Chronic Val administration carries a potential for repurposing as adjuvant or conjunct therapy for patients at high risk for BC.
Collapse
Affiliation(s)
- Amira M A Mansour
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Egypt
| | - Wessam M El-Refaie
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Egypt
| | - Nesrine S El-Mezayen
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| |
Collapse
|
6
|
Fatehi R, Nouraei M, Panahiyan M, Rashedinia M, Firouzabadi N. Modulation of ACE2/Ang1-7/Mas and ACE/AngII/AT1 axes affects anticancer properties of sertraline in MCF-7 breast cancer cells. Biochem Biophys Rep 2024; 38:101738. [PMID: 38831897 PMCID: PMC11145238 DOI: 10.1016/j.bbrep.2024.101738] [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: 02/12/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
The renin-angiotensin system (RAS) is best known for playing a major role in maintaining the physiology of the cardiovascular system. Dysregulation of the RAS pathway has been proposed as a link to some malignancies and contributes to cancer metastasis. Breast cancer is considered as one of the leading causes of cancer death in women and its prevention remains yet a challenge. Elements of RAS are expressed in both normal breast tissue and cancerous cells, signifying the essential role of RAS in breast cancer pathology. Sertraline, a widely used antidepressant, has shown anti-proliferative properties on a variety of malignancies. This study aimed to investigate the effect of sertraline and its combination with agonists and antagonists of RAS (A779, Ang 1-7 and losartan) on viability of MCF-7 cells along with their effect on apoptosis and distribution of cell cycle. Our results indicated that sertraline, losartan and Ang 1-7 significantly decreased cell viability, induced apoptosis and cell cycle arrest. A779 blunted the effect of sertraline on cell viability, ROS generation and cell cycle arrest. Combination treatment of sertraline with losartan as well as Ang 1-7 caused a remarkable decline in cell viability. In conclusion, results of the present study support the anti-cancer properties of sertraline, losartan and Ang 1-7 via induction of apoptosis and cell cycle arrest.
Collapse
Affiliation(s)
- Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Nouraei
- Student Research Comittee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Panahiyan
- Student Research Comittee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Rashedinia
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Food and Supplements Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Panagi M, Mpekris F, Voutouri C, Hadjigeorgiou AG, Symeonidou C, Porfyriou E, Michael C, Stylianou A, Martin JD, Cabral H, Constantinidou A, Stylianopoulos T. Stabilizing Tumor-Resident Mast Cells Restores T-Cell Infiltration and Sensitizes Sarcomas to PD-L1 Inhibition. Clin Cancer Res 2024; 30:2582-2597. [PMID: 38578281 PMCID: PMC11145177 DOI: 10.1158/1078-0432.ccr-24-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/10/2024] [Accepted: 04/03/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE To explore the cellular cross-talk of tumor-resident mast cells (MC) in controlling the activity of cancer-associated fibroblasts (CAF) to overcome tumor microenvironment (TME) abnormalities, enhancing the efficacy of immune-checkpoint inhibitors in sarcoma. EXPERIMENTAL DESIGN We used a coculture system followed by further validation in mouse models of fibrosarcoma and osteosarcoma with or without administration of the MC stabilizer and antihistamine ketotifen. To evaluate the contribution of ketotifen in sensitizing tumors to therapy, we performed combination studies with doxorubicin chemotherapy and anti-PD-L1 (B7-H1, clone 10F.9G2) treatment. We investigated the ability of ketotifen to modulate the TME in human sarcomas in the context of a repurposed phase II clinical trial. RESULTS Inhibition of MC activation with ketotifen successfully suppressed CAF proliferation and stiffness of the extracellular matrix accompanied by an increase in vessel perfusion in fibrosarcoma and osteosarcoma as indicated by ultrasound shear wave elastography imaging. The improved tissue oxygenation increased the efficacy of chemoimmunotherapy, supported by enhanced T-cell infiltration and acquisition of tumor antigen-specific memory. Importantly, the effect of ketotifen in reducing tumor stiffness was further validated in sarcoma patients, highlighting its translational potential. CONCLUSIONS Our study suggests the targeting of MCs with clinically administered drugs, such as antihistamines, as a promising approach to overcome resistance to immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G. Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | | | | | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas Stylianou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- Basic and Translational Cancer Research Center, School of Sciences, European University of Cyprus, Nicosia, Cyprus
| | | | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Anastasia Constantinidou
- Bank of Cyprus Oncology Centre, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
8
|
Xia C, Zhao J, Huang Y, Miao H, Zhao F. Angiogenesis in nasopharyngeal carcinoma: insights, imaging, and therapeutic strategies. Front Oncol 2024; 14:1331064. [PMID: 38863627 PMCID: PMC11165036 DOI: 10.3389/fonc.2024.1331064] [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: 10/31/2023] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a highly prevalent head and neck malignancy in southern China frequently diagnosed at advanced stages owing to subtle early symptoms and associated metastasis. Angiogenesis emerges as a pivotal factor in NPC progression, with numerous angiogenesis-related factors showing aberrant expression and contributing to increased neovascularization within NPC tumors. These abnormal vessels not only nourish tumor growth but also facilitate metastasis, culminating in unfavorable patient outcomes. Multiple studies have demonstrated the applicability of various imaging techniques for assessing angiogenesis in NPC tumors, thus serving as a foundation for personalized treatment strategies and prognostic assessments. Anti-angiogenic therapies have exhibited significant potential for inhibiting NPC angiogenesis and exerting anti-tumor effects. To enhance efficacy, anti-angiogenic drugs are frequently combined with other treatment modalities to synergistically enhance anti-tumor effects while mitigating the side effects associated with single-agent therapies, consequently improving patient prognosis. Identifying the potential mechanisms and key targets underlying NPC angiogenesis and exploring more effective detection and treatment approaches holds promise for shaping the future of NPC diagnosis, treatment, and prognosis, thereby offering new avenues and perspectives for research and clinical practice.
Collapse
Affiliation(s)
- Chenxi Xia
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Jia Zhao
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Hongbin Miao
- Department of Otolaryngology-Head and Neck Surgery, Bishan hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Bishan, Chongqing, China
| | - Feipeng Zhao
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Department of Otolaryngology-Head and Neck Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
9
|
He Z, Feng D, Zhang C, Chen Z, Wang H, Hou J, Li S, Wei X. Recent strategies for evoking immunogenic Pyroptosis in antitumor immunotherapy. J Control Release 2024; 366:375-394. [PMID: 38142962 DOI: 10.1016/j.jconrel.2023.12.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/24/2023] [Accepted: 12/16/2023] [Indexed: 12/26/2023]
Abstract
Pyroptosis is a specific type of programmed cell death (PCD) characterized by distinct morphological changes, including cell swelling, membrane blebbing, DNA fragmentation, and eventual cell lysis. Pyroptosis is closely associated with human-related diseases, such as inflammation and malignancies. Since the initial observation of pyroptosis in Shigella flexneri-infected macrophages more than 20 years ago, various pyroptosis-inducing agents, including ions, small molecules, and biological nanomaterials, have been developed for tumor treatment. Given that pyroptosis can activate the body's robust immune response against tumor and promote the formation of the body's long-term immune memory in tumor treatment, its status as a type of immunogenic cell death is self-evident. Therefore, pyroptosis should be used as a powerful anti-tumor strategy. However, there still is a lack of a comprehensive summary of the most recent advances in pyroptosis-based cancer therapy. Therefore, it is vital to fill this gap and inspire future drug design to better induce tumor cells to undergo pyroptosis to achieve advanced anti-tumor effects. In this review, we summarize in detail the most recent advances in triggering tumor cell immunogenic pyroptosis for adequate tumor clearance based on various treatment modalities, and highlight material design and therapeutic advantages. Besides, we also provide an outlook on the prospects of this emerging field in the next development.
Collapse
Affiliation(s)
- Zhangxin He
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215000, China
| | - Dexiang Feng
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Chaoji Zhang
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhiqian Chen
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - He Wang
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China.
| | - Jianquan Hou
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215000, China.
| | - Xuedong Wei
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
10
|
Chiu TJ, Chen CH, Chen YJ, Wee Y, Wang CS, Luo SD. Prognosis of Midkine and AT1R expression in resectable head and neck squamous cell carcinoma. Cancer Cell Int 2023; 23:212. [PMID: 37743493 PMCID: PMC10518915 DOI: 10.1186/s12935-023-03060-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Research studies have demonstrated that Midkine (MDK) can influence the expression and activity of Renin-angiotensin system (RAS) components. Angiotensin II is involved in tumor growth and angiogenesis in different cancers. We previously observed Angiotensin II receptor blockers (ARBs) improve the survival rates of patients with oral cancers. These findings have prompted us to investigate whether MDK can influence the RAS pathway, mainly through its association with angiotensin II type 1 receptor (AT1R), which contributes to the observed poor prognosis in head and neck squamous cell carcinoma (HNSCC) patients. METHODS MDK and AT1R expressions were examined in 150 HNSCC patients post-operation by immunohistochemical staining between 1 January 2010 and 31 December 2016. We tested the over-expression and silencing of MDK to evaluate the AT1R expression and functional biological assays in HNSCC cell lines HSC-3 and SAS. RESULTS Positive expression of MDK is correlated with positive AT1R expression. MDK predicted poor NSCC patients' survival. Silencing MDK could suppress AT1R and pAKT expression and reduce the growth, migration, and invasion of HNSCC cells. ARB also inhibits MDK stimulating HNSCC cell proliferation. Overexpression of MDK could upregulate AT1R and pAKT. CONCLUSIONS MDK is an independent prognostic factor of HNSCC post-operation, and AT1R regulates HNSCC cell growth, invasion, and migration. Positive MDK and AT1R expressions are highly correlated. Mechanistically, the interaction between MDK and AT1R is crucial for MDK-mediated cell viability, and inhibiting AT1R can effectively counteract or abolish these effects. Furthermore, MDK exerts a regulatory role in the expression of AT1R, as well as in the growth and motility of HNSCC cells. These findings highlight the involvement of the interaction between MDK, AT1R, and the pAkt signaling pathways in HNSCC cell viability growth.
Collapse
Affiliation(s)
- Tai-Jan Chiu
- Department of Hematology‑Oncology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, 833, Taiwan
| | - Chang-Han Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Ju Chen
- Kaohsiung Cancer Prevention and Screening Center, Kaohsiung, 833, Taiwan
| | - Yinshen Wee
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Sheng-Dean Luo
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 833, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan.
- School of Traditional Chinese Medicine, Chang Gung University College of Medicine, Taoyuan, 33302, Taiwan.
| |
Collapse
|
11
|
Li S, Chen X, Chen J, Wu B, Liu J, Guo Y, Li M, Pu X. Multi-omics integration analysis of GPCRs in pan-cancer to uncover inter-omics relationships and potential driver genes. Comput Biol Med 2023; 161:106988. [PMID: 37201441 DOI: 10.1016/j.compbiomed.2023.106988] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/30/2023] [Accepted: 04/27/2023] [Indexed: 05/20/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest drug target family. Unfortunately, applications of GPCRs in cancer therapy are scarce due to very limited knowledge regarding their correlations with cancers. Multi-omics data enables systematic investigations of GPCRs, yet their effective integration remains a challenge due to the complexity of the data. Here, we adopt two types of integration strategies, multi-staged and meta-dimensional approaches, to fully characterize somatic mutations, somatic copy number alterations (SCNAs), DNA methylations, and mRNA expressions of GPCRs in 33 cancers. Results from the multi-staged integration reveal that GPCR mutations cannot well predict expression dysregulation. The correlations between expressions and SCNAs are primarily positive, while correlations of the methylations with expressions and SCNAs are bimodal with negative correlations predominating. Based on these correlations, 32 and 144 potential cancer-related GPCRs driven by aberrant SCNA and methylation are identified, respectively. In addition, the meta-dimensional integration analysis is carried out by using deep learning models, which predict more than one hundred GPCRs as potential oncogenes. When comparing results between the two integration strategies, 165 cancer-related GPCRs are common in both, suggesting that they should be prioritized in future studies. However, 172 GPCRs emerge in only one, indicating that the two integration strategies should be considered concurrently to complement the information missed by the other such that obtain a more comprehensive understanding. Finally, correlation analysis further reveals that GPCRs, in particular for the class A and adhesion receptors, are generally immune-related. In a whole, the work is for the first time to reveal the associations between different omics layers and highlight the necessity of combing the two strategies in identifying cancer-related GPCRs.
Collapse
Affiliation(s)
- Shiqi Li
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Xin Chen
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Jianfang Chen
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Binjian Wu
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Jing Liu
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Yanzhi Guo
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Xuemei Pu
- College of Chemistry, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
12
|
Zhang C, Chen W, Pan S, Zhang S, Xie H, Zhang Z, Lei W, Bao L, You Y. SEVs-mediated miR-6750 transfer inhibits pre-metastatic niche formation in nasopharyngeal carcinoma by targeting M6PR. Cell Death Dis 2023; 9:2. [PMID: 36609569 PMCID: PMC9823008 DOI: 10.1038/s41420-022-01262-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 01/07/2023]
Abstract
Reliable detection of circulating small extracellular vesicles (SEVs) and their miRNA cargo has been needed to develop potential specific non-invasive diagnostic and therapeutic marker for cancer metastasis. Here, we detected miR-6750, the precise molecular function of which was largely unknown, was significantly enriched in serum-SEVs from normal volunteers vs. patients with nasopharyngeal carcinoma (NPC). And we determined that miR-6750-SEVs attenuated NPC metastasis. Subsequently, miR-6750-SEVs was proven to inhibit angiogenesis and activate macrophage toward to M1 phenotype to inhibit pre-metastatic niche formation. After analyzing the expression level of miR-6750 in NPC cells, HUVECs and macrophage, we found that once miR-6750 level in NPC cells was close to or higher than normal nasopharyngeal epithelial cells (NP69), miR-6750-SEVs would be transferred from NPC cells to macrophage and then to HUVECs to modulate metastatic niche. Moreover, in vitro assays and BALB/c mouse tumor models revealed that miR-6750 directly targeted mannose 6-phosphate receptor (M6PR). Taken together, our findings revealed that miR-6750-M6PR axis can mediate NPC metastasis by remodeling tumor microenvironment (TME) via SEVs, which give novel sights to pathogenesis of NPC.
Collapse
Affiliation(s)
- Caiming Zhang
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenhui Chen
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Si Pan
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Siyu Zhang
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Haijing Xie
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Zixiang Zhang
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Wei Lei
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Lili Bao
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yiwen You
- grid.440642.00000 0004 0644 5481Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China ,grid.440642.00000 0004 0644 5481Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
13
|
Fu Y, Saxu R, Ridwan KA, Yao J, Chen X, Xu X, Zheng W, Yu P, Teng Y. Losartan Alleviates the Side Effects and Maintains the Anticancer Activity of Axitinib. Molecules 2022; 27:2764. [PMID: 35566115 PMCID: PMC9101101 DOI: 10.3390/molecules27092764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/17/2022] Open
Abstract
Axitinib is one of the most potent inhibitors of the vascular endothelial growth factor (VEGF) receptor and shows strong antitumor activity toward various malignant tumors. However, its severe side effects affect the quality of life and prognosis of patients. Losartan, which functions as a typical angiotensin receptor blocker, controls the average arterial pressure of patients with essential hypertension and protects against hypertension-related secondary diseases, including proteinuria and cardiovascular injury. To explore the effects of losartan on side effects caused by axitinib and its antitumor activity, several animal experiments were conducted. This study first analyzed and explored the effect of losartan on the amelioration of side effects in Wistar rats caused by axitinib. The results showed that the systolic blood pressure of Wistar rats was significantly increased by about 30 mmHg in 7 days of axitinib treatment, while the combination of losartan significantly reduced the blood pressure rise caused by axitinib. The Miles experimental model and mouse xenograft tumor model were further used to evaluate the effect of losartan on the antitumor effect of axitinib. The result clearly demonstrated that losartan has no significant influence on axitinib-related low vascular permeability and antitumor activity. In summary, our results showed that the combination of axitinib and losartan significantly reduced the side effects and maintained the antitumor effects of axitinib. This study provides information for overcoming VEGF receptor inhibitor-related side effects.
Collapse
Affiliation(s)
- Ying Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Rengui Saxu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Kadir Ahmad Ridwan
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Jiaping Yao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Xiaoxuan Chen
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Xueping Xu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Weida Zheng
- Medical College, Yanbian University, No. 977 Gongyuan Road, Yanji 133002, China;
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| |
Collapse
|
14
|
Munjal A, Khandia R, Paladhi S, Pandey M, Parihar A, Pathe C, Rajukumar K, Bin Emran T, Alqahtani T, Alqahtani AM, Alamri AH, Chidambara K, Dhama K. Evaluating the Effects of Hypotensive Drug Valsartan on Angiogenesis and Associated Breast Ductal Carcinoma Cell Metastasis. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.817.825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Fu L, Jin W, Zhang J, Zhu L, Lu J, Zhen Y, Zhang L, Ouyang L, Liu B, Yu H. Repurposing non-oncology small-molecule drugs to improve cancer therapy: Current situation and future directions. Acta Pharm Sin B 2022; 12:532-557. [PMID: 35256933 PMCID: PMC8897051 DOI: 10.1016/j.apsb.2021.09.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/05/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022] Open
Abstract
Drug repurposing or repositioning has been well-known to refer to the therapeutic applications of a drug for another indication other than it was originally approved for. Repurposing non-oncology small-molecule drugs has been increasingly becoming an attractive approach to improve cancer therapy, with potentially lower overall costs and shorter timelines. Several non-oncology drugs approved by FDA have been recently reported to treat different types of human cancers, with the aid of some new emerging technologies, such as omics sequencing and artificial intelligence to overcome the bottleneck of drug repurposing. Therefore, in this review, we focus on summarizing the therapeutic potential of non-oncology drugs, including cardiovascular drugs, microbiological drugs, small-molecule antibiotics, anti-viral drugs, anti-inflammatory drugs, anti-neurodegenerative drugs, antipsychotic drugs, antidepressants, and other drugs in human cancers. We also discuss their novel potential targets and relevant signaling pathways of these old non-oncology drugs in cancer therapies. Taken together, these inspiring findings will shed new light on repurposing more non-oncology small-molecule drugs with their intricate molecular mechanisms for future cancer drug discovery.
Collapse
|
16
|
Leveraging cellular mechano-responsiveness for cancer therapy. Trends Mol Med 2021; 28:155-169. [PMID: 34973934 DOI: 10.1016/j.molmed.2021.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022]
Abstract
Cells sense the biophysical properties of the tumor microenvironment (TME) and adopt these signals in their development, progression, and metastatic dissemination. Recent work highlights the mechano-responsiveness of cells in tumors and the underlying mechanisms. Furthermore, approaches to mechano-modulating diverse types of cell have emerged aiming to inhibit tumor growth and metastasis. These include targeting mechanosensitive machineries in cancer cells to induce apoptosis, intervening matrix stiffening incurred by cancer-associated fibroblasts (CAFs) in both primary and metastatic tumor sites, and modulating matrix mechanics to improve immune cell therapeutic efficacy. This review is envisaged to help scientists and clinicians in cancer research to advance understanding of the cellular mechano-responsiveness in TME, and to harness these concepts for cancer mechanotherapies.
Collapse
|
17
|
Wu CN, Wu SC, Chen WC, Yang YH, Chin JC, Chien CY, Fang FM, Li SH, Luo SD, Chiu TJ. Angiotensin II receptor blockers and oral squamous cell carcinoma survival: A propensity-score-matched cohort study. PLoS One 2021; 16:e0260772. [PMID: 34855858 PMCID: PMC8638984 DOI: 10.1371/journal.pone.0260772] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 11/16/2021] [Indexed: 01/05/2023] Open
Abstract
Objectives Angiotensin II receptor blockers (ARBs) improve the survival rates of patients with various cancers. However, it remains unclear whether ARBs confer a survival benefit on patients with oral squamous cell carcinoma (OSCC). Here, we assessed the associations between ARB use and survival in patients with OSCC of different stages. Materials and methods This was a 10-year retrospective cohort study of OSCC patients. We enrolled 7,558 patients diagnosed with oral cancer between January 2007 and December 2017 whose details had been entered into the Chang Gung Research Database. Seven hundred and fourteen patients were recruited from the Chang Gung Research Database after performing 1:1 propensity score-matching between ARB users and non-users. Cox’s regression models with adjusted covariates were employed to detect factors influencing the survival rates of patients with OSCC. Results Kaplan-Meier analysis revealed that the overall survival (OS) rate of 180-day ARB users increased (p = 0.038). Cox’s regression models indicated that ARB use, younger patients, early-stage OSCC, and patients without diabetes mellitus were independently prognostic of improved OS. Increased OS was more prominent in 180-day ARB users in stage III, Iva, and IVb categories. Conclusions ARB use for more than 180 days is associated with an increased survival rate and is a positive, independent prognostic factor in patients with OSCC. A further two-arm study should be conducted to confirm the clinical usefulness of ARBs in OSCC patients.
Collapse
Affiliation(s)
- Ching-Nung Wu
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wei-Chih Chen
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
- Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jo-Chi Chin
- Department of Anesthesiology, Park One International Hospital, Kaohsiung, Taiwan
| | - Chih-Yen Chien
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Fu-Min Fang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shau-Hsuan Li
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Sheng-Dean Luo
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (TJC); (SDL)
| | - Tai-Jan Chiu
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (TJC); (SDL)
| |
Collapse
|
18
|
Panza S, Malivindi R, Caruso A, Russo U, Giordano F, Győrffy B, Gelsomino L, De Amicis F, Barone I, Conforti FL, Giordano C, Bonofiglio D, Catalano S, Andò S. Novel Insights into the Antagonistic Effects of Losartan against Angiotensin II/AGTR1 Signaling in Glioblastoma Cells. Cancers (Basel) 2021; 13:cancers13184555. [PMID: 34572782 PMCID: PMC8469998 DOI: 10.3390/cancers13184555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Patients with high-grade glioma (HGG) such as glioblastoma (GBM) who undergo surgical resection with adjuvant therapy have a mean overall survival of 14.6 months and 100% of recurrence. Thus, these disappointing outcomes in terms of glioblastoma life expectancy require seeking novel pharmacological tools, including drug repurposing. In the present study, we identify a novel molecular mechanism through which Losartan antagonizes Angiotensin II (Ang II)/Angiotensin II type I receptor (AGTR1) signaling, overexpressed in GBM cells. For instance, we demonstrate how Losartan drastically inhibits the stimulatory effects of Ang II on aromatase activity and consequently reduces local estrogen production, sustaining cancer progression. Thus, it is reasonable to repurpose Losartan as an adjuvant pharmacological tool to be implemented prospectively in the novel therapeutic strategies adopted in GBM patients. Abstract New avenues for glioblastoma therapy are required due to the limited mortality benefit of the current treatments. The renin-angiotensin system (RAS) exhibits local actions and works as a paracrine system in different tissues and tumors, including glioma. The glioblastoma cell lines U-87 MG and T98G overexpresses Angiotensin II (Ang II)/Angiotensin II type I receptor (AGTR1) signaling, which enhances in vitro and in vivo local estrogen production through a direct up-regulation of the aromatase gene promoters p I.f and p I.4. In addition, Ang II/AGTR1 signaling transactivates estrogen receptor-α in a ligand-independent manner through mitogen-activated protein kinase (MAPK) activation. The higher aromatase mRNA expression in patients with glioblastoma was associated with the worst survival prognostic, according to The Cancer Genome Atlas (TCGA). An intrinsic immunosuppressive glioblastoma tumor milieu has been previously documented. We demonstrate how Ang II treatment in glioblastoma cells increases programmed death-ligand 1 (PD-L1) expression reversed by combined exposure to Losartan (LOS) in vitro and in vivo. Our findings highlight how LOS, in addition, antagonizes the previously documented neoangiogenetic, profibrotic, and immunosuppressive effects of Ang II and drastically inhibits its stimulatory effects on local estrogen production, sustaining glioblastoma cell growth. Thus, Losartan may represent an adjuvant pharmacological tool to be repurposed prospectively for glioblastoma treatment.
Collapse
Affiliation(s)
- Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Umberto Russo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary;
- Cancer Biomarker Research Group, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
- Correspondence: ; Tel.: +39-0984-496201; Fax: +39-0984-496203
| |
Collapse
|
19
|
Stokes WA, Molina E, McDermott JD, Morgan RL, Bickett T, Fakhoury KR, Amini A, Karam SD. Survival impact of angiotensin-converting enzyme inhibitors and angiotensin II receptor antagonists in head and neck cancer. Head Neck 2021; 43:3255-3275. [PMID: 34289190 DOI: 10.1002/hed.26809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Preclinical evidence suggests a link between the renin-angiotensin system and oncogenesis. We aimed to explore the impact of angiotensin-converting enzyme inhibitors (ACEi) and angiotensin II receptor blockers (ARB) in head and neck cancer (HNC). METHODS Over 5000 patients were identified from the Surveillance, Epidemiology, and End Results-Medicare linked dataset and categorized according to ACEi and ARB and diagnoses of chronic kidney disease (CKD) or hypertension (HTN). Overall survival (OS) and cancer-specific survival (CSS) were compared using Cox multivariable regression (MVA), expressed as hazard ratios (HR) with 95% confidence intervals (95%CI). RESULTS No significant MVA associations for OS or CSS were found for ACEi. Compared to patients with CKD/HTN taking ARB, those with CKD/HTN not taking ARB experienced worse OS (HR 1.28, 95%CI 1.09-1.51, p = 0.003) and CSS (HR 1.23, 95%CI 1.00-1.50, p = 0.050). CONCLUSIONS ARB usage is associated with improved OS and CSS among HNC patients with CKD or HTN.
Collapse
Affiliation(s)
- William A Stokes
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Elizabeth Molina
- Department of Health Systems, Management, and Policy, Colorado School of Public Health, Aurora, Colorado, USA
| | - Jessica D McDermott
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Rustain L Morgan
- Division of Nuclear Medicine, Department of Radiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Thomas Bickett
- Department of Radiation Oncology, University of Colorado Denver, Aurora, Colorado, USA
| | - Kareem R Fakhoury
- Department of Radiation Oncology, University of Colorado Denver, Aurora, Colorado, USA
| | - Arya Amini
- Department of Radiation Oncology, City of Hope Cancer Center, Duarte, California, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
20
|
Chen S, Bie M, Wang X, Fan M, Chen B, Shi Q, Jiang Y. PGRN exacerbates the progression of non-small cell lung cancer via PI3K/AKT/Bcl-2 antiapoptotic signaling. Genes Dis 2021; 9:1650-1661. [PMID: 36157487 PMCID: PMC9485207 DOI: 10.1016/j.gendis.2021.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
Progranulin (PGRN) is a growth factor that is involved in the progression of multiple tumors. However, the effects and molecular mechanisms by which PGRN induces lung cancer remain unclear. The expression level of PGRN was analyzed by conducting immunohistochemistry of the histological sections of lung tissues from non-small-cell lung carcinoma (NSCLC) patients. The proliferation, apoptosis, migration, and invasion of NSCLC cells were assessed by the MTT assay, Western blot, degree of wound healing, and Transwell assays. A nude mouse xenograft model was used to validate the role of PGRN in vivo. The expression level of PGRN was higher in male patients with lung adenocarcinoma than in those with lung squamous cell carcinoma; by contrast, no difference was observed in female patients. The overexpression of PGRN promoted the proliferation and anti-apoptosis of H520 (derived from lung squamous cell carcinoma) cells, whereas knockdown of PGRN inhibited the proliferation and anti-apoptosis of A549 (derived from lung adenocarcinoma) cells. Copanlisib (targeting PI3K) inhibited the increase in the expression of cell anti-apoptosis marker Bcl-2 induced by rhPGRN protein; the PI3K agonist 740 Y–P partially reversed the decrease in Bcl-2 expression induced by PGRN deficiency in both A549 and H520 cells. PGRN increased the expression of Ki-67, PCNA, and Bcl-2 in vivo. PGRN inhibited cell apoptosis depending on the PI3K/Akt/Bcl-2 signaling axis; PGRN positivity correlated with lung adenocarcinoma. PGRN is a potential biomarker for the treatment and diagnosis of NSCLC, especially in lung adenocarcinoma.
Collapse
Affiliation(s)
- Sicheng Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Mengjun Bie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Mengtian Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Bin Chen
- Ministry of Education Key Laboratory of Diagnostic Medicine, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Qiong Shi
- Ministry of Education Key Laboratory of Diagnostic Medicine, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Yingjiu Jiang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Corresponding author. Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, PR China. Fax: +86 023 63310999.
| |
Collapse
|