1
|
Xie H, Lin F, Shi F, Johnstone E, Wang Y, An Y, Su J, Liu J, Dong Q, Liu J. Synthesis, biological evaluation and mechanism study based on network pharmacology of amino acids esters of 20(S)-protopanaxadiol as novel anticancer agents. Fitoterapia 2025; 180:106274. [PMID: 39537112 DOI: 10.1016/j.fitote.2024.106274] [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: 01/16/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
As one of the metabolites of ginseng, 20(S)-protopanaxadiol (PPD) is a compound with dammarane-type tetracyclic triterpene, which performs a wide range of anticancer activities. In this study, PPD was used as a lead. A series of compounds were synthesized respectively with 11 amino acids through esterification and were evaluated for their cytotoxicity against several cancer cell lines. One of the synthetic products (PL) exhibited potent inhibitory effect on Huh-7 cells relative to that of PPD in vitro. Subsequently, the Annexin V-FITC /PI staining assay was used to verify that PL induced apoptosis of Huh-7 cells in a dose-dependent manner. A UPLC-Q/TOF-MS analysis method was established and validated for assessing pharmacokinetic properties after the administration of PPD and PL in rats. The results showed that compared with PPD, T1/2of PL in rats was prolonged, and the peak time was delayed, resulting in broader tissue distribution of the compound in the body. In addition, the targets of PL against several cancers were predicted and analyzed via network pharmacology. Molecular docking simulations demonstrated that PL interacted with the active sites of the above targets. In conclusion, this study provided a theoretical basis for the development and clinical application of anti-tumor activity of PPD.
Collapse
Affiliation(s)
- Hongliu Xie
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Fang Lin
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Fei Shi
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | | | - Yaqi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Yang An
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Jun Su
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China; Basic Medicine Department, Fenyang College of Shanxi Medical University, Fenyang 032200, PR China
| | - Jiayin Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Qinghai Dong
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Jihua Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
2
|
Caliò A, Marletta S, Stefanizzi L, Marcolini L, Rotellini M, Serio G, Bariani E, Vicentini C, Pedron S, Martelli FM, Antonini P, Brunelli M, Martignoni G. Comparison of Primary and Metastatic Fumarate Hydratase-Deficient Renal Cell Carcinomas Documents Morphologic Divergence and Potential Diagnostic Pitfall With Peritoneal Mesothelioma. Mod Pathol 2024; 37:100561. [PMID: 38996839 DOI: 10.1016/j.modpat.2024.100561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Fumarate hydratase (FH)-deficient renal cell carcinomas are rare neoplasms characterized by wide morphologic heterogeneity and pathogenetic mutations in the FH gene. They often show aggressive behavior with rapid diffusion to distant organs, so novel therapeutic scenarios have been explored, including EGFR inhibitors and PD-L1 expression for targeted immunotherapy. Herein, we investigated a series of 11 primary FH-deficient renal cell carcinomas and 7 distant metastases to evaluate tumor heterogeneity even in metastatic sites and estimate the specific spread rates to various organs. Furthermore, the tumors were tested for immunohistochemical PD-L1 expression and EGFR mutations. Most metastatic cases involved the abdominal lymph nodes (4/7; 57%), followed by the peritoneum (3/7; 42%), the liver (2/7; 29%), and the lungs (1/7; 14%). Six metastatic localizations were histologically documented, revealing a morphologic heterogeneous architecture often differing from that of the corresponding primary renal tumor. Peritoneal involvement morphologically resembled a benign reactive mesothelial process or primary peritoneal mesothelioma, thus advocating to perform an accurate immunohistochemical panel, including PAX8 and FH, to reach a proper diagnosis. A pure low-grade succinate dehydrogenase-looking primary FH-deficient renal cell carcinoma was also recorded. As for therapy, significant PD-L1 labeling was found in 60% of primary renal tumors, whereas none of them carried pathogenetic EGFR mutations. Our data show that FH-deficient renal cell carcinoma may be morphologically heterogeneous in metastases as well, which involve the lymph nodes, the liver, and the peritoneum more frequently than other renal tumors. Due to the high frequency of this latter (42%), pathologists should always be concerned about ruling out mesothelial-derived mimickers, and the occurrence of rarer, primary, low-grade-looking types. Finally, contrary to EGFR mutations, PD-L1 expression could be a possible predictive biomarker for the therapy of these tumors.
Collapse
Affiliation(s)
- Anna Caliò
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Stefano Marletta
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy; Division of Pathology, Humanitas Istituto Clinico Catanese, Catania, Italy
| | - Lavinia Stefanizzi
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Lisa Marcolini
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Matteo Rotellini
- Anatomia Patologica Massa Carrara Azienda Toscana Nord Ovest, Italy
| | - Gabriella Serio
- Pathology Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Italy
| | - Elena Bariani
- Unit of Anatomic Pathology, Department of Oncology, Bellaria Hospital, Bologna, Italy
| | - Caterina Vicentini
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Serena Pedron
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Filippo M Martelli
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Pietro Antonini
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Matteo Brunelli
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Guido Martignoni
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy; Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy.
| |
Collapse
|
3
|
Li Q, Liu H, Wang H, Xiong W, Dai L, Zhang X, Wang P, Ye H, Shi J, Fang Z, Wang K. Anti-BIRC5 autoantibody serves as a valuable biomarker for diagnosing AFP-negative hepatocellular carcinoma. PeerJ 2024; 12:e17494. [PMID: 38832035 PMCID: PMC11146321 DOI: 10.7717/peerj.17494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/09/2024] [Indexed: 06/05/2024] Open
Abstract
Background Autoantibodies targeting tumor-associated antigens (TAAbs) have emerged as promising biomarkers for early cancer detection. This research aimed to assess the diagnostic capacity of anti-BIRC5 autoantibody in detecting AFP-negative hepatocellular carcinoma (ANHCC). Methods This research was carried out in three stages (discovery phase, validation phase, and evaluation phase) and included a total of 744 participants. Firstly, the anti-BIRC5 autoantibody was discovered using protein microarray, exhibiting a higher positive rate in ANHCC samples (ANHCCs) compared to normal control samples (NCs). Secondly, the anti-BIRC5 autoantibody was validated through enzyme-linked immunosorbent assay (ELISA) in 85 ANHCCs and 85 NCs from two clinical centers (Zhengzhou and Nanchang). Lastly, the diagnostic usefulness of the anti-BIRC5 autoantibody for hepatocellular carcinoma (HCC) was evaluated by ELISA in a cohort consisting of an additional 149 AFP-positive hepatocellular carcinoma samples (APHCCs), 95 ANHCCs and 244 NCs. The association of elevated autoantibody to high expression of BIRC5 in HCC was further explored by the database from prognosis, immune infiltration, DNA methylation, and gene mutation level. Results In the validation phase, the area under the ROC curve (AUC) of anti-BIRC5 autoantibody to distinguish ANHCCs from NCs in Zhengzhou and Nanchang centers was 0.733 and 0.745, respectively. In the evaluation phase, the AUCs of anti-BIRC5 autoantibody for identifying ANHCCs and HCCs from NCs were 0.738 and 0.726, respectively. Furthermore, when combined with AFP, the AUC for identifying HCCs from NCs increased to 0.914 with a sensitivity of 77.5% and specificity of 91.8%. High expression of BIRC5 gene is not only correlated with poor prognosis of HCCs, but also significantly associated with infiltration of immune cells, DNA methylation, and gene mutation. Conclusion The findings suggest that the anti-BIRC5 autoantibody could serve as a potential biomarker for ANHCC, in addition to its supplementary role alongside AFP in the diagnosis of HCC. Next, we can carry out specific verification and explore the function of anti-BIRC5 autoantibody in the occurrence and development of HCC.
Collapse
Affiliation(s)
- Qing Li
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou, China
| | - Haiyan Liu
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Han Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Wenzhuo Xiong
- School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou, China
| | - Xiuzhi Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou, China
- Department of Pathology, Henan Medical College, Zhengzhou, Henan, China
| | - Peng Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hua Ye
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jianxiang Shi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou, China
| | - Zhihao Fang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Keyan Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou, China
| |
Collapse
|
4
|
Pachimatla AG, Fenstermaker R, Ciesielski M, Yendamuri S. Survivin in lung cancer: a potential target for therapy and prevention-a narrative review. Transl Lung Cancer Res 2024; 13:362-374. [PMID: 38496694 PMCID: PMC10938099 DOI: 10.21037/tlcr-23-621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024]
Abstract
Background and Objective A versatile biomarker, survivin, is highly expressed in proliferating cells of multiple cancers in humans and animals. It is an apoptosis-regulating protein, engaging in a cascade of reactions that involve several other genes and protein interactions. Currently, researchers are investigating its therapeutic potential due to the evidence linking its overexpression to advanced-stage lung cancer. This review is centered around examining survivin-related molecular mechanisms and its therapeutic role specifically in lung cancer. Our objective is to discuss the role of survivin in prognosis and treatment response, shedding light on immune-targeted therapies, as well as outlining future directions for survivin-based vaccines in lung cancer. Methods The PubMed database and the United States National Library of Medicine search engine at the National Institutes of Health were searched on 24 August 2023 to identify published research studies. Searching "((((((airway [Title/Abstract]) OR (lung [Title/Abstract])) OR (pulm[Title/Abstract])) OR (bronch[Title/Abstract])) OR (nslc[Title/Abstract])) AND (((cancer[Title/Abstract]) OR (carcino[Title/Abstract])) OR (oncol[Title/Abstract]))) AND (survivin[Title/Abstract])" gave 728 results. After screening the title and abstracts and excluding the review articles 168 titles were shortlisted and full text studied. The discussions are added to relevant sections. Key Content and Findings Survivin is a cell cycle-dependent, inhibitor of apoptosis protein that contributes to carcinogenesis, tumor vascularization, metastasis, and treatment resistance. Several treatments that impact survivin either directly or indirectly have been reported as effective in treating lung cancer. Immunity-based therapy, a novel approach known for its targeted nature and minimal side effects, is currently under investigation for lung cancer treatment. Emerging survivin-centered vaccines exhibit promising attributes in terms of safety, effectiveness, and ability to stimulate an immune response. These factors point towards a significant potential for advancing the future of lung cancer prevention and enhancing overall survival rates. Conclusions Nuclear survivin is a potential biomarker for advanced non-small cell lung cancer. It plays a role in determining drug responsiveness and is found to be significantly elevated in cases of resistance to chemotherapy. Multiple compounds and immunization strategies have been identified to impact lung cancer cells; however, they are currently in the early stages of phase I or phase II clinical trials. The substantial promise of survivin-based immunogenicity-focused treatments warrants in-depth investigation and exploration.
Collapse
Affiliation(s)
- Akhil Goud Pachimatla
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Robert Fenstermaker
- Department of Neurosurgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael Ciesielski
- Department of Neurosurgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| |
Collapse
|
5
|
Tzenaki N, Xenou L, Goulielmaki E, Tsapara A, Voudouri I, Antoniou A, Valianatos G, Tzardi M, De Bree E, Berdiaki A, Makrigiannakis A, Papakonstanti EA. A combined opposite targeting of p110δ PI3K and RhoA abrogates skin cancer. Commun Biol 2024; 7:26. [PMID: 38182748 PMCID: PMC10770346 DOI: 10.1038/s42003-023-05639-8] [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: 03/07/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024] Open
Abstract
Malignant melanoma is the most aggressive and deadly skin cancer with an increasing incidence worldwide whereas SCC is the second most common non-melanoma human skin cancer with limited treatment options. Here we show that the development and metastasis of melanoma and SCC cancers can be blocked by a combined opposite targeting of RhoA and p110δ PI3K. We found that a targeted induction of RhoA activity into tumours by deletion of p190RhoGAP-a potent inhibitor of RhoA GTPase-in tumour cells together with adoptive macrophages transfer from δD910A/D910A mice in mice bearing tumours with active RhoA abrogated growth progression of melanoma and SCC tumours. Τhe efficacy of this combined treatment is the same in tumours lacking activating mutations in BRAF and in tumours harbouring the most frequent BRAF(V600E) mutation. Furthermore, the efficiency of this combined treatment is associated with decreased ATX expression in tumour cells and tumour stroma bypassing a positive feedback expression of ATX induced by direct ATX pharmacological inactivation. Together, our findings highlight the importance of targeting cancer cells and macrophages for skin cancer therapy, emerge a reverse link between ATX and RhoA and illustrate the benefit of p110δ PI3K inhibition as a combinatorial regimen for the treatment of skin cancers.
Collapse
Affiliation(s)
- Niki Tzenaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Lydia Xenou
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Evangelia Goulielmaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Anna Tsapara
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Irene Voudouri
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Angelika Antoniou
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - George Valianatos
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Maria Tzardi
- Department of Pathology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Eelco De Bree
- Department of Surgical Oncology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Aikaterini Berdiaki
- Department of Obstetrics and Gynaecology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Antonios Makrigiannakis
- Department of Obstetrics and Gynaecology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | | |
Collapse
|
6
|
Samar MR, Khan W, Rashid YA, Mohammad ATV. Unmasking the enigma: A case of Fumarate Hydratase-deficient renal cell carcinoma. Int J Surg Case Rep 2023; 113:109054. [PMID: 37992668 DOI: 10.1016/j.ijscr.2023.109054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023] Open
Abstract
INTRODUCTION Fumarate Hydratase-deficient-renal cell carcinoma (FH-dRCC) is an uncommon and extremely aggressive variant of renal cell carcinoma (RCC) resulting from inactivating mutations in the tumor suppressor gene, fumarate hydratase (FH). CASE PRESENTATION We report a case of a young male who presented with the complaint of painless hematuria. Upon workup, he was found to have a renal mass with bony metastases. The histopathology was consistent with renal cell carcinoma with features of FH-deficient variant. Germline testing showed a pathogenic mutation in the FH gene. He was started on a treatment combination of Pembrolizumab and Axitinib along with Zoledronate for bone metastasis. His response to the combination therapy was short with early progression of disease. He was switched to a second-line treatment Bevacizumab and Erlotinib, which achieved significant disease response. DISCUSSION Systemic therapy is the mainstay of treatment for metastatic disease. Although the novel agents approved for other subsets of RCC have been used, the responses are grim. There is no consensus on the sequence of further lines of treatment for FH-dRCC and is based on the physician's choice, availability of the drugs, cost, toxicity, and tolerance of the patient. CONCLUSION This case report emphasizes the occurrence, presentation, management and prognosis of FH-dRCC, which is an aggressive entity, presenting at a young age with early distant metastases, not diagnosed appropriately due to its poorly characterized cytologic features. Being an infrequent neoplasm, it is an area that warrants oncological exploration to improve outcomes in these individuals. The combination of Erlotinib and Bevacizumab provides promising outcomes in terms of progression-free survival.
Collapse
Affiliation(s)
- Mirza Rameez Samar
- Department of Medical Oncology, The Aga Khan University Hospital, Pakistan.
| | - Wajiha Khan
- Department of Medicine And Surgery, Dow University of Health Sciences, Karachi, Pakistan
| | | | | |
Collapse
|
7
|
Qu A, Zhang S, Zou H, Li S, Chen D, Zhang Y, Li S, Zhang H, Yang J, Yang Y, Huang Y, Li X, Zhang Y. Outcome benefits of bevacizumab biosimilar (SIBP04) combined with carboplatin and paclitaxel in advanced non-squamous non-small-cell lung cancer patients with EGFR mutation: subgroup analysis of a prospective, randomized phase III clinical trial. J Cancer Res Clin Oncol 2023; 149:12713-12721. [PMID: 37452849 DOI: 10.1007/s00432-023-05103-4] [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: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE SIBP04 is a bevacizumab biosimilar, and bevacizumab combined with carboplatin and paclitaxel in advanced non-squamous non-small-cell lung cancer (nsqNSCLC) has been recommended as the first-line treatment choice. However, the efforts of bevacizumab combined with carboplatin and paclitaxel for nsqNSCLC patients with EGFR mutation remained unclear. Here we report an EGFR mutation subgroup analysis of a prospective, randomized phase III clinical trial (NCT05318443). METHODS In this randomized, double-blind, multi-center, parallel controlled, phase III clinical trial, locally advanced, metastatic NSCLC patients were enrolled, and EGFR expression was examined and considered as a stratification factor. All patients received 4 to 6 cycles of paclitaxel and carboplatin plus SIBP04 or bevacizumab 15 mg/kg intravenously followed by SIBP04 15 mg/kg maintenance until intolerable toxicity, disease progression or death. Patients with EGFR mutation and wild-type were assessed for progression-free survival (PFS) and overall survival (OS). RESULTS EGFR expression was examined in 398 NSCLC patients (142 with EGFR mutation, 256 with EGFR wild type). PFS in EGFR mutation patients was significantly longer than EGFR wild-type patients (10.91 vs. 7.82 months; HR = 0.692, 95% CI 0.519-0.921, P = 0.011). The median OS in patients with EGFR mutation was not reached while that of EGFR wild-type group was 17.54 months (HR = 0.398, 95% CI 0.275-0.575, P < 0.001). However, there were no significant differences in objective response rate (61.97% vs. 55.86%, P = 0.237) or disease control rate (90.14% vs. 89.84%, P = 0.925). CONCLUSION Bevacizumab combined with chemotherapy significantly prolonged the PFS and OS of advanced nsqNSCLC patients with EGFR mutation.
Collapse
Affiliation(s)
- Aidong Qu
- Shanghai Institute of Biological Products Company Limited, 1262 Yanan West Changning District Rd, Shanghai, 200052, China
| | - Shiying Zhang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Hongxia Zou
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Sixiu Li
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Dandan Chen
- Shanghai Institute of Biological Products Company Limited, 1262 Yanan West Changning District Rd, Shanghai, 200052, China
| | - Yaowen Zhang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Songsong Li
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Huijun Zhang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Ji Yang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China
| | - Yunkai Yang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China.
| | - Yubao Huang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China.
| | - Xiuling Li
- Shanghai Institute of Biological Products Company Limited, 1262 Yanan West Changning District Rd, Shanghai, 200052, China.
| | - Yuntao Zhang
- China National Biotec Group Company Limited, B2 Shuangqiao Rd, Chaoyang District, Beijing, 10020, China.
| |
Collapse
|
8
|
Mittal S, Mallia MB. Molecular imaging of tumor hypoxia: Evolution of nitroimidazole radiopharmaceuticals and insights for future development. Bioorg Chem 2023; 139:106687. [PMID: 37406518 DOI: 10.1016/j.bioorg.2023.106687] [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: 04/28/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Though growing evidence has been collected in support of the concept of dose escalation based on the molecular level images indicating hypoxic tumor sub-volumes that could be radio-resistant, validation of the concept is still a work in progress. Molecular imaging of tumor hypoxia using radiopharmaceuticals is expected to provide the required input to plan dose escalation through Image Guided Radiation Therapy (IGRT) to kill/control the radio-resistant hypoxic tumor cells. The success of the IGRT, therefore, is heavily dependent on the quality of images obtained using the radiopharmaceutical and the extent to which the image represents the true hypoxic status of the tumor in spite of the heterogeneous nature of tumor hypoxia. Available literature on radiopharmaceuticals for imaging hypoxia is highly skewed in favor of nitroimidazole as the pharmacophore given their ability to undergo oxygen dependent reduction in hypoxic cells. In this context, present review on nitroimidazole radiopharmaceuticals would be immensely helpful to the researchers to obtain a birds-eye view on what has been achieved so far and what can be tried differently to obtain a better hypoxia imaging agent. The review also covers various methods of radiolabeling that could be utilized for developing radiotracers for hypoxia targeting applications.
Collapse
Affiliation(s)
- Sweety Mittal
- Radiopharmaceuticals Division, Bhabha Atomic Research Center, Mumbai 400085, India.
| | - Madhava B Mallia
- Radiopharmaceuticals Division, Bhabha Atomic Research Center, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
9
|
Chen LC, Mokgautsi N, Kuo YC, Wu ATH, Huang HS. In Silico Evaluation of HN-N07 Small Molecule as an Inhibitor of Angiogenesis and Lymphangiogenesis Oncogenic Signatures in Non-Small Cell Lung Cancer. Biomedicines 2023; 11:2011. [PMID: 37509650 PMCID: PMC10376976 DOI: 10.3390/biomedicines11072011] [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: 06/21/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Tumor angiogenesis and lymphangiogenesis pathways have been identified as important therapeutic targets in non-small cell lung cancer (NSCLC). Bevacizumab, which is a monoclonal antibody, was the initial inhibitor of angiogenesis and lymphangiogenesis that received approval for use in the treatment of advanced non-small cell lung cancer (NSCLC) in combination with chemotherapy. Despite its usage, patients may still develop resistance to the treatment, which can be attributed to various histological subtypes and the initiation of treatment at advanced stages of cancer. Due to their better specificity, selectivity, and safety compared to chemotherapy, small molecules have been approved for treating advanced NSCLC. Based on the development of multiple small-molecule antiangiogenic drugs either in house and abroad or in other laboratories to treat NSCLC, we used a quinoline-derived small molecule-HN-N07-as a potential target drug for NSCLC. Accordingly, we used computational simulation tools and evaluated the drug-likeness properties of HN-N07. Moreover, we identified target genes, resulting in the discovery of the target BIRC5/HIF1A/FLT4 pro-angiogenic genes. Furthermore, we used in silico molecular docking analysis to determine whether HN-N07 could potentially inhibit BIRC5/HIF1A/FLT4. Interestingly, the results of docking HN-N07 with the BIRC5, FLT4, and HIF1A oncogenes revealed unique binding affinities, which were significantly higher than those of standard inhibitors. In summary, these results indicate that HN-N07 shows promise as a potential inhibitor of oncogenic signaling pathways in NSCLC. Ongoing studies that involve in vitro experiments and in vivo investigations using tumor-bearing mice are in progress, aiming to evaluate the therapeutic effectiveness of the HN-N07 small molecule.
Collapse
Affiliation(s)
- Lung-Ching Chen
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan
| | - Ntlotlang Mokgautsi
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Alexander T H Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
10
|
Gupta R, Kadhim MM, Turki Jalil A, Qasim Alasheqi M, Alsaikhan F, Khalimovna Mukhamedova N, Alexis Ramírez-Coronel A, Hassan Jawhar Z, Ramaiah P, Najafi M. The interactions of docetaxel with tumor microenvironment. Int Immunopharmacol 2023; 119:110214. [PMID: 37126985 DOI: 10.1016/j.intimp.2023.110214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
There are several interactions within the tumor microenvironment (TME) that affect the response of cancer cells to therapy. There are also a large number of cells and secretions in TME that increase resistance to therapy. Following the release of immunosuppressive, pro-angiogenic, and metastatic molecules by certain cells such as tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and cancer cells, immune evasion, angiogenesis, and metastasis may be induced. However, natural killer (NK) cells and cytotoxic CD8 + T lymphocytes (CTLs) can responsively release anticancer molecules. In addition, anticancer drugs can modulate these cells and their interactions in favor of either cancer resistance or therapy. Docetaxel belongs to taxanes, a class of anti-tumor drugs, which acts through the polymerization of tubulin and the induction of cell cycle arrest. Also, it has been revealed that taxanes including docetaxel affect cancer cells and the other cells within TME through some other mechanisms such as modulation of immune system responses, angiogenesis, and metastasis. In this paper, we explain the basic mechanisms of docetaxel interactions with malignant cells. Besides, we review the diverse effects of docetaxel on TME and cancer cells in consequence. Lastly, the modulatory effects of docetaxel alone or in conjunction with other anticancer agents on anti-tumor immunity, cancer cell resistance, angiogenesis, and metastasis will be discussed.
Collapse
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, 281406 U. P., India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla 51001, Iraq.
| | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia.
| | | | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca 010107, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Medillin 050001, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Azogues 030102, Ecuador
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil 44001, Iraq; Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil 44001, Iraq
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran.
| |
Collapse
|
11
|
Role of hypoxia-inducible factor-1α and survivin in enhancing radiosensitivity of breast cancer. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2023. [DOI: 10.1016/j.jrras.2023.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
12
|
Cao Q, Mushajiang M, Tang CQ, Ai XQ. Role of hypoxia-inducible factor-1α and survivin in breast cancer recurrence and prognosis. Heliyon 2023; 9:e14132. [PMID: 36950571 PMCID: PMC10025039 DOI: 10.1016/j.heliyon.2023.e14132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Objective To analyze the expression of hypoxia-inducible factor-1α (HIF-1α) and survivin in breast cancer, and different molecular subtypes of breast cancer and to assess their relationship with recurrence and prognosis. Methods The expression levels of HIF-1α and survivin genes in breast cancer were investigated using bioinformatics. Their protein expression levels were then verified through immunohistochemistry (IHC), and their relationship with recurrence and prognosis was assessed. Results Expression levels of HIF-1α and survivin genes and proteins were increased in breast cancer tissues compared with normal tissues. Both were associated with clinical features of breast cancer and differentially expressed in different molecular subtypes of breast cancer, and both are related to the signal pathway of breast cancer growth and invasion. HIF-1α and survivin gene and protein expression levels were correlated, and both were associated with breast cancer recurrence (R = 0.380, P < 0.05; R = 0.673, P < 0.05, respectively). According to The Cancer Genome Atlas (TCGA) database, HIF1A and BIRC5 gene were not associated with breast cancer prognosis (P ≥ 0.05); however, HIF-1α and survivin protein were associated with recurrence patient's overall survival (OS) (P < 0.05). Conclusion HIF-1α and survivin are highly expressed in breast cancer and can be used as potential biomarkers to predict recurrence and assess prognosis.
Collapse
Affiliation(s)
- Qian Cao
- Department of Breast Radiotherapy, The Third Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011, Xinjiang, China
| | - Munire Mushajiang
- Department of Breast Radiotherapy, The Third Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011, Xinjiang, China
| | - Cheng-qiong Tang
- Department of Radiological Physics and Technology, The Third Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011, Xinjiang, China
| | - Xiu-qing Ai
- Department of Breast Radiotherapy, The Third Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011, Xinjiang, China
- Corresponding author. Department of Breast Radiotherapy, The Third Affiliated Teaching Hospital of Xinjiang Medical University(Affiliated Cancer Hospital), No. 789, Suzhou East Street, Xinshi District, Urumqi, 830011 Xinjiang, China
| |
Collapse
|
13
|
Jafarzadeh A, Bazargan N, Chatrabnous N, Jafarzadeh S, Nemati M. Contribution of survivin to the immune system, allergies and autoimmune diseases. Hum Immunol 2023; 84:301-310. [PMID: 36754653 DOI: 10.1016/j.humimm.2023.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/30/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
In addition to malignancies, survivin (a member of the apoptosis inhibitor family) has been implicated in the pathogenesis of inflammatory disorders, including autoimmune and allergic diseases. Survivin is constantly expressed in the proliferating hematopoietic progenitor cells, and it is re-expressed in the mature cells of the innate and adaptive immunity, upon activation. Survivin enhances the expression of co-stimulatory molecules and MHC class II molecules in dendritic cells, and promotes the lifespan of macrophages, neutrophils, and eosinophils, while suppressing natural killer (NK) cell activity. Survivin has been implicated in T cell maturation, T cell expansion, effector CD4+ T cell differentiation, maintenance of memory CD4+ T and CD8+ T cells, as well as antibody production. Upregulated expression of survivin was indicated in the T cells as well as various samples collected from allergic patients. Survivin can contribute to the pathogenesis of allergic diseases via the promotion of the Th2 polarization, promoting IL-4 expression, compromising activation-induced cell death (AICD) in Th2 cells, and preventing apoptosis of eosinophils, as well as, amplification of eosinophilia. Moreover, survivin can interfere with clonal deletion of autoreactive T and B cells, as well as suppress Treg cell development and activity supporting the development of autoimmune diseases. This review discusses the role of survivin in immunity, allergy and autoimmunity as well as provides evidence that survivin may be considered as a novel therapeutic target for the treatment of allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Nasrin Bazargan
- Department of Internal Medicine, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Nazanin Chatrabnous
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Patel SA, Nilsson MB, Le X, Cascone T, Jain RK, Heymach JV. Molecular Mechanisms and Future Implications of VEGF/VEGFR in Cancer Therapy. Clin Cancer Res 2023; 29:30-39. [PMID: 35969170 DOI: 10.1158/1078-0432.ccr-22-1366] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/28/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023]
Abstract
Angiogenesis, the sprouting of new blood vessels from existing vessels, is one of six known mechanisms employed by solid tumors to recruit blood vessels necessary for their initiation, growth, and metastatic spread. The vascular network within the tumor facilitates the transport of nutrients, oxygen, and immune cells and is regulated by pro- and anti-angiogenic factors. Nearly four decades ago, VEGF was identified as a critical factor promoting vascular permeability and angiogenesis, followed by identification of VEGF family ligands and their receptors (VEGFR). Since then, over a dozen drugs targeting the VEGF/VEGFR pathway have been approved for approximately 20 solid tumor types, usually in combination with other therapies. Initially designed to starve tumors, these agents transiently "normalize" tumor vessels in preclinical and clinical studies, and in the clinic, increased tumor blood perfusion or oxygenation in response to these agents is associated with improved outcomes. Nevertheless, the survival benefit has been modest in most tumor types, and there are currently no biomarkers in routine clinical use for identifying which patients are most likely to benefit from treatment. However, the ability of these agents to reprogram the immunosuppressive tumor microenvironment into an immunostimulatory milieu has rekindled interest and has led to the FDA approval of seven different combinations of VEGF/VEGFR pathway inhibitors with immune checkpoint blockers for many solid tumors in the past 3 years. In this review, we discuss our understanding of the mechanisms of response and resistance to blocking VEGF/VEGFR, and potential strategies to develop more effective therapeutic approaches.
Collapse
Affiliation(s)
- Sonia A Patel
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monique B Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Orofiamma LA, Vural D, Antonescu CN. Control of cell metabolism by the epidermal growth factor receptor. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119359. [PMID: 36089077 DOI: 10.1016/j.bbamcr.2022.119359] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/24/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
The epidermal growth factor receptor (EGFR) triggers the activation of many intracellular signals that control cell proliferation, growth, survival, migration, and differentiation. Given its wide expression, EGFR has many functions in development and tissue homeostasis. Some of the cellular outcomes of EGFR signaling involve alterations of specific aspects of cellular metabolism, and alterations of cell metabolism are emerging as driving influences in many physiological and pathophysiological contexts. Here we review the mechanisms by which EGFR regulates cell metabolism, including by modulation of gene expression and protein function leading to control of glucose uptake, glycolysis, biosynthetic pathways branching from glucose metabolism, amino acid metabolism, lipogenesis, and mitochondrial function. We further examine how this regulation of cell metabolism by EGFR may contribute to cell proliferation and differentiation and how EGFR-driven control of metabolism can impact certain diseases and therapy outcomes.
Collapse
Affiliation(s)
- Laura A Orofiamma
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Dafne Vural
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada.
| |
Collapse
|
16
|
Liu J, Wang H, Zhang M, Li Y, Wang R, Chen H, Wang B, Gao X, Song S, Wang Y, Ren Y, Li J, Liu P. Metformin and simvastatin synergistically suppress endothelin 1-induced hypoxia and angiogenesis in multiple cancer types. Cancer Sci 2022; 114:640-653. [PMID: 36156330 PMCID: PMC9899631 DOI: 10.1111/cas.15602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/08/2022] [Accepted: 09/20/2022] [Indexed: 01/07/2023] Open
Abstract
Multiple cancers have been reported to be associated with angiogenesis and are sensitive to anti-angiogenic therapies. Vascular normalization, by restoring proper tumor perfusion and oxygenation, could limit tumor cell invasiveness and improve the effectiveness of anticancer treatments. However, the underlying anticancer mechanisms of antiangiogenic drugs are still unknown. Metformin (MET) and simvastatin (SVA), two metabolic-related drugs, have been shown to play important roles in modulating the hypoxic tumor microenvironment and angiogenesis. Whether the combination of MET and SVA could exert a more effective antitumor effect than individual treatments has not been examined. The antitumor effect of the synergism of SVA and MET was detected in mouse models, breast cancer patient-derived organoids, and multiple tumor cell lines compared with untreated, SVA, or MET alone. RNA sequencing revealed that the combination of MET and SVA (but not MET or SVA alone) inhibited the expression of endothelin 1 (ET-1), an important regulator of angiogenesis and the hypoxia-related pathway. We demonstrate that the MET and SVA combination showed synergistic effects on inhibiting tumor cell proliferation, promoting apoptosis, alleviating hypoxia, decreasing angiogenesis, and increasing vessel normalization compared with the use of a single agent alone. The MET and SVA combination suppressed ET-1-induced hypoxia-inducible factor 1α expression by increasing prolyl hydroxylase 2 (PHD2) expression. Furthermore, the MET and SVA combination showed a more potent anticancer effect compared with bosentan. Together, our findings suggest the potential application of the MET and SVA combination in antitumor therapy.
Collapse
Affiliation(s)
- Jie Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Huxia Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Mammary DepartmentShaanxi Provincial Cancer HospitalXi'anShaanxi ProvinceChina
| | - Miao Zhang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yazhao Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Ruiqi Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - He Chen
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Bo Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Xiaoqian Gao
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Shaoran Song
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yaochun Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yu Ren
- Department of Surgical OncologyThe First Affiliated Hospital of Xi' an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Juan Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Peijun Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| |
Collapse
|
17
|
Dekker Y, Le Dévédec SE, Danen EHJ, Liu Q. Crosstalk between Hypoxia and Extracellular Matrix in the Tumor Microenvironment in Breast Cancer. Genes (Basel) 2022; 13:genes13091585. [PMID: 36140753 PMCID: PMC9498429 DOI: 10.3390/genes13091585] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/28/2022] [Accepted: 08/31/2022] [Indexed: 11/24/2022] Open
Abstract
Even though breast cancer is the most diagnosed cancer among women, treatments are not always successful in preventing its progression. Recent studies suggest that hypoxia and the extracellular matrix (ECM) are important in altering cell metabolism and tumor metastasis. Therefore, the aim of this review is to study the crosstalk between hypoxia and the ECM and to assess their impact on breast cancer progression. The findings indicate that hypoxic signaling engages multiple mechanisms that directly contribute to ECM remodeling, ultimately increasing breast cancer aggressiveness. Second, hypoxia and the ECM cooperate to alter different aspects of cell metabolism. They mutually enhance aerobic glycolysis through upregulation of glucose transport, glycolytic enzymes, and by regulating intracellular pH. Both alter lipid and amino acid metabolism by stimulating lipid and amino acid uptake and synthesis, thereby providing the tumor with additional energy for growth and metastasis. Third, YAP/TAZ signaling is not merely regulated by the tumor microenvironment and cell metabolism, but it also regulates it primarily through its target c-Myc. Taken together, this review provides a better understanding of the crosstalk between hypoxia and the ECM in breast cancer. Additionally, it points to a role for the YAP/TAZ mechanotransduction pathway as an important link between hypoxia and the ECM in the tumor microenvironment, driving breast cancer progression.
Collapse
Affiliation(s)
- Yasmin Dekker
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sylvia E. Le Dévédec
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Erik H. J. Danen
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Correspondence: (E.H.J.D.); (Q.L.)
| | - Qiuyu Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100102, China
- Correspondence: (E.H.J.D.); (Q.L.)
| |
Collapse
|
18
|
Lindner AK, Tulchiner G, Seeber A, Siska PJ, Thurnher M, Pichler R. Targeting strategies in the treatment of fumarate hydratase deficient renal cell carcinoma. Front Oncol 2022; 12:906014. [PMID: 35912170 PMCID: PMC9337267 DOI: 10.3389/fonc.2022.906014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/29/2022] [Indexed: 11/26/2022] Open
Abstract
Fumarate hydratase (FH) - deficient renal cell carcinoma (FHdRCC) is a rare aggressive subtype of RCC caused by a germline or sporadic loss-of-function mutation in the FH gene. Here, we summarize how FH deficiency results in the accumulation of fumarate, which in turn leads to activation of hypoxia-inducible factor (HIF) through inhibition of prolyl hydroxylases. HIF promotes tumorigenesis by orchestrating a metabolic switch to glycolysis even under normoxia, a phenomenon well-known as the Warburg effect. HIF activates the transcription of many genes, including vascular endothelial growth factor (VEGF). Crosstalk between HIF and epidermal growth factor receptor (EGFR) has also been described as a tumor-promoting mechanism. In this review we discuss therapeutic options for FHdRCC with a focus on anti-angiogenesis and EGFR-blockade. We also address potential targets that arise within the metabolic escape routes taken by FH-deficient cells for cell growth and survival.
Collapse
Affiliation(s)
- Andrea Katharina Lindner
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Gennadi Tulchiner
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Seeber
- Department of Haematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter J. Siska
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
- *Correspondence: Renate Pichler,
| |
Collapse
|
19
|
Wiggs A, Molina S, Sumner SJ, Rushing BR. A Review of Metabolic Targets of Anticancer Nutrients and Nutraceuticals in Pre-Clinical Models of Triple-Negative Breast Cancer. Nutrients 2022; 14:1990. [PMID: 35631131 PMCID: PMC9146055 DOI: 10.3390/nu14101990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/19/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is notoriously aggressive and has poorer outcomes as compared with other breast cancer subtypes. Due to a lack of targeted therapies, TNBC is often treated with chemotherapeutics as opposed to hormone therapy or other targeted therapies available to individuals with estrogen receptor positive (ER+) breast cancers. Because of the lack of treatment options for TNBC, other therapeutic avenues are being explored. Metabolic reprogramming, a hallmark of cancer, provides potential opportunities to target cancer cells more specifically, increasing efficacy and reducing side effects. Nutrients serve a significant role in metabolic processes involved in DNA transcription, protein folding, and function as co-factors in enzyme activity, and may provide novel strategies to target cancer cell metabolism in TNBC. This article reviews studies that have investigated how nutrients/nutraceuticals target metabolic processes in TNBC cells alone or in combination with existing drugs to exert anticancer effects. These agents have been shown to cause perturbations in many metabolic processes related to glucose metabolism, fatty acid metabolism, as well as autophagy and oxidative stress-related metabolism. With this information, we present the potential of nutrients as metabolism-directed anticancer agents and the potential for using these agents alone or in cocktails as a new direction for TNBC therapy.
Collapse
Affiliation(s)
- Alleigh Wiggs
- Department of Nutrition, University of North Carolina-Chapel Hill, Durham, NC 27599, USA
| | - Sabrina Molina
- Nutrition Research Institute, University of North Carolina-Chapel Hill, Kannapolis, NC 280821, USA
| | - Susan J. Sumner
- Department of Nutrition, University of North Carolina-Chapel Hill, Durham, NC 27599, USA
- Nutrition Research Institute, University of North Carolina-Chapel Hill, Kannapolis, NC 280821, USA
| | - Blake R. Rushing
- Department of Nutrition, University of North Carolina-Chapel Hill, Durham, NC 27599, USA
- Nutrition Research Institute, University of North Carolina-Chapel Hill, Kannapolis, NC 280821, USA
| |
Collapse
|
20
|
Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Marciniak B, Drozda R, Kontek R. Targeting carbonic anhydrase IX and XII isoforms with small molecule inhibitors and monoclonal antibodies. J Enzyme Inhib Med Chem 2022; 37:1278-1298. [PMID: 35506234 PMCID: PMC9090362 DOI: 10.1080/14756366.2022.2052868] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbonic anhydrases IX and CAXII (CAIX/CAXII) are transmembrane zinc metalloproteins that catalyze a very basic but crucial physiological reaction: the conversion of carbon dioxide into bicarbonate with a release of the proton. CA, especially CAIX and CAXII isoforms gained the attention of many researchers interested in anticancer drug design due to pivotal functions of enzymes in the cancer cell metastasis and response to hypoxia, and their expression restricted to malignant cells. This offers an opportunity to develop new targeted therapies with fewer side effects. Continuous efforts led to the discovery of a series of diverse compounds with the most abundant sulphonamide derivatives. Here we review current knowledge considering small molecule and antibody-based targeting of CAIX/CAXII in cancer.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland.,Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Somdutt Mujwar
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| |
Collapse
|
21
|
Lee HH, Cho H. Apigenin Increases Natural Killer Cytotoxicity to Human Hepatocellular Carcinoma Expressing HIF-1α through High Interaction of CD95/CD95L. J Microbiol Biotechnol 2022; 32:397-404. [PMID: 35283421 PMCID: PMC9628789 DOI: 10.4014/jmb.2201.01010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cell activity is more attenuated in hepatocellular carcinoma (HCC) patients than normal. Hypoxic-inducible factor (HIF)-1α is highly expressed in tumors to maintain their metabolism in a hypoxic environment. The expression of HIF-1α in cancers can lead to cell growth, proliferation, invasion/metastasis and immune escape. Although apigenin, a flavonoid, is known to have various biological activities, it has not been demonstrated in NK cell immune activity in HCC cells. In this study, NK-92 cells were directly cocultured with HCC SK-Hep1 cells for 24 h to evaluate NK cell activity in HCC cells or HCC cells expressing HIF-1α by apigenin. NK cell cytotoxicity to HCC cells expressing HIF-1α was significantly increased, and NK cell-activating receptors, NKG2D, NKp30 and NKp44 were highly expressed. The activating effect of apigenin on NK cells substantially induced apoptosis in HCC cells expressing HIF-1α through high expression of CD95L on the surface of NK-92 cells. Moreover, apigenin excellently inhibited the level of TGF-β1 in a coculture of NK cells and HCC cells. In conclusion, apigenin seems to be a good compound that increases NK cell cytotoxicity to HCC cells by controlling HIF-1α expression.
Collapse
Affiliation(s)
- Hwan Hee Lee
- Department of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea,Duksung Innovative Drug Center, Duksung Women’s University, Seoul 01369, Republic of Korea,Corresponding authors H.H. Lee Phone: +82-2-901-8734 Fax: +82-2-901-8386 E-mail:
| | - Hyosun Cho
- Department of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea,Duksung Innovative Drug Center, Duksung Women’s University, Seoul 01369, Republic of Korea,
H. Cho Phone: +82-2-901-8678 Fax: +82-2-901-8386 E-mail:
| |
Collapse
|
22
|
Golchert J, Staar D, Bennewitz J, Hartmann M, Hoffmann N, Ameling S, Völker U, Peters J, Wanka H. Overexpression of Renin-B Induces Warburg-like Effects That Are Associated with Increased AKT/mTOR Signaling. Cells 2022; 11:cells11091459. [PMID: 35563765 PMCID: PMC9103744 DOI: 10.3390/cells11091459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/08/2022] [Accepted: 04/19/2022] [Indexed: 11/29/2022] Open
Abstract
The classical secretory renin-a is known to be involved in angiotensin generation, thereby regulating not only blood pressure, but also promoting oxidative stress as well as apoptotic and necrotic cell death. In contrast, another cytosolic renin isoform named renin-b has been described, exerting protective effects under ischemia-related conditions in H9c2 cardiomyoblasts. Using microarray-based transcriptome analyses, we aimed to identify the signaling pathways involved in mediating cardioprotection in H9c2 cells overexpressing renin-b. By transcriptome profiling, we identified increased gene expression of several genes encoding glycolytic enzymes and glucose transporters, while the transcript levels of TCA-cycle enzymes were decreased. Complementing data from metabolic analyses revealed enhanced glucose consumption and lactate accumulation due to renin-b overexpression. Renin-b overexpression further stimulated AKT/mTOR signaling, where numerous genes involved in this pathway showed altered transcript levels. For AKT, we also detected enhanced phosphorylation levels by means of Western blotting, suggesting an activation of this kinase. Moreover, analysis of the ROS levels identified an increase in ROS accumulation in renin-b-overexpressing cells. Altogether, our data demonstrate that renin-b overexpression induces the metabolic remodeling of H9c2 cells similar to that seen under oxygen deprivation. This metabolic phenotype exerting so-called aerobic glycolysis is also known as the Warburg effect.
Collapse
Affiliation(s)
- Janine Golchert
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
| | - Doreen Staar
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
| | - Jonathan Bennewitz
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
| | - Miriam Hartmann
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
| | - Nadin Hoffmann
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
| | - Sabine Ameling
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany; (S.A.); (U.V.)
- Partner Site Greifswald, DZHK (German Center for Cardiovascular Research), 17475 Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany; (S.A.); (U.V.)
- Partner Site Greifswald, DZHK (German Center for Cardiovascular Research), 17475 Greifswald, Germany
| | - Jörg Peters
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
- Correspondence:
| | - Heike Wanka
- Institute of Physiology, University Medicine Greifswald, 17475 Greifswald, Germany; (J.G.); (D.S.); (J.B.); (M.H.); (N.H.); (H.W.)
| |
Collapse
|
23
|
Kit OI, Pushkin AA, Alliluyev IA, Timoshkina NN, Gvaldin DY, Rostorguev EE, Kuznetsova NS. Differential expression of microRNAs targeting genes associated with the development of high-grade gliomas. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00245-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Highly malignant high-grade gliomas are tumors of the central nervous system (CNS). They are solid tumors arising from transformed cells of the brain and/or the spinal cord. In recent years, the expression of genes and regulating miRNAs in glial brain tumors has been actively studied. The present study is devoted to assessing the expression levels of miR-215-5p, miR-22-3p, miR-122-5p, miR-107, miR-324-5p, miR-34a-5p, miR-155. -5p, miR-21-5p, miR-497-5p, miR-330-3p, miR-146a-5p, miR-92a-1-5p, miR-326 and target genes EGFR, SMAD4, SMAD7, SMO, NOTCH1, NOTCH2, HIF1A, EGLIN1/3, KDM1B, KDM1A, MSI1, MSI2, TET1 in high-grade glioma tissues.
Results
As a result of the analysis of the levels of relative expression of the studied genes, there are significant changes (p < 0.05) in tumor tissue for genes: EGFR, SMAD4, SMAD7, SMO, HIF1A, EGLN1/3. We obtained data on a significant change (p < 0.05) in the levels of relative expression for microRNA: hsa-miR-215-5p, hsa-miR-22-3p, hsa-miR-122-5p, hsa-miR-107, hsa-miR-324-5p, hsa-miR-155-5p, hsa-miR-21-5p, hsa-miR-330-3p, hsa-miR-326. Data on the association of overall survival in patients with high-grade glioma and the level of relative expression of the EGFR and HIF1A genes were obtained. The obtained data demonstrate the association of overall survival of patients with high-grade glioma and the level of relative expression of EGFR, HIF1A and hsa-miR-22-3p, hsa-miR-107 and hsa-miR-330-3p.
Conclusions
The obtained data on the expression of genes and microRNAs expand the understanding of the biology of the development of high-grade glial tumors. These data demonstrate new potential therapeutic and prognostic goals in high-grade gliomas.
Collapse
|
24
|
Li Y, Jiang Y, Zheng Z, Du N, Guan S, Guo W, Tang X, Cui J, Zhang L, Liu K, Yu Q, Gan Z. Co-Delivery of Precisely Prescribed Multi-Prodrug Combination by an Engineered Nanocarrier enables Efficient Individualized Cancer Chemotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110490. [PMID: 35044690 DOI: 10.1002/adma.202110490] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Indexed: 06/14/2023]
Abstract
The limited anticancer drug library and the frequent occurrence of drug resistance have driven monotherapy-based cancer therapy into a difficult situation. Considering the formidable process of new drug discovery, combination therapy using currently available drugs is a potential alternative. Nevertheless, the barrier between in vitro combination screening and precise in vivo delivery remains insurmountable in the current free-drug- or nanoparticle (NP)-based combination therapy, which substantially hinders the application of combination therapy. Herein, a novel, precise drug delivery strategy to realize efficient and individualized combination therapy is proposed. Nanomedicine (NM) is engineered using a microfluidics-based mixer by combining rationally designed polymeric prodrugs of three commercial chemotherapeutics and a cascade-responsive block copolymer; the NM possesses ratiometric drug loading and synchronized drug release. In addition to quantitative drug loading and precisely controlled drug combination, consistent nanoproperties of these NPs make their in vivo fate predictable. Consequently, tumor growth and metastasis can be effectively inhibited by precisely prescribed NPs derived from in vitro combination screening. This proof-of-concept study clearly reveals the feasibility of overcoming the current drug-library limitations through precise delivery of any predetermined drug combination, facilitating translational research of individualized combination therapy.
Collapse
Affiliation(s)
- Yuqiang Li
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yitong Jiang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ziyan Zheng
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Nan Du
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Shuli Guan
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wenxuan Guo
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaohu Tang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jiajunzi Cui
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lanqiong Zhang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kunpeng Liu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
25
|
Lappano R, Todd LA, Stanic M, Cai Q, Maggiolini M, Marincola F, Pietrobon V. Multifaceted Interplay between Hormones, Growth Factors and Hypoxia in the Tumor Microenvironment. Cancers (Basel) 2022; 14:539. [PMID: 35158804 PMCID: PMC8833523 DOI: 10.3390/cancers14030539] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hormones and growth factors (GFs) are signaling molecules implicated in the regulation of a variety of cellular processes. They play important roles in both healthy and tumor cells, where they function by binding to specific receptors on target cells and activating downstream signaling cascades. The stages of tumor progression are influenced by hormones and GF signaling. Hypoxia, a hallmark of cancer progression, contributes to tumor plasticity and heterogeneity. Most solid tumors contain a hypoxic core due to rapid cellular proliferation that outgrows the blood supply. In these circumstances, hypoxia-inducible factors (HIFs) play a central role in the adaptation of tumor cells to their new environment, dramatically reshaping their transcriptional profile. HIF signaling is modulated by a variety of factors including hormones and GFs, which activate signaling pathways that enhance tumor growth and metastatic potential and impair responses to therapy. In this review, we summarize the role of hormones and GFs during cancer onset and progression with a particular focus on hypoxia and the interplay with HIF proteins. We also discuss how hypoxia influences the efficacy of cancer immunotherapy, considering that a hypoxic environment may act as a determinant of the immune-excluded phenotype and a major hindrance to the success of adoptive cell therapies.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Lauren A. Todd
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Mia Stanic
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Qi Cai
- Kite Pharma Inc., Santa Monica, CA 90404, USA; (Q.C.); (F.M.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | | | | |
Collapse
|
26
|
Olszewska A, Borkowska A, Granica M, Karolczak J, Zglinicki B, Kieda C, Was H. Escape From Cisplatin-Induced Senescence of Hypoxic Lung Cancer Cells Can Be Overcome by Hydroxychloroquine. Front Oncol 2022; 11:738385. [PMID: 35127467 PMCID: PMC8813758 DOI: 10.3389/fonc.2021.738385] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/23/2021] [Indexed: 12/23/2022] Open
Abstract
Chemotherapy is the commonly used treatment for advanced lung cancer. However, it produces side effects such as the development of chemoresistance. A possible responsible mechanism may be therapy-induced senescence (TIS). TIS cells display increased senescence-associated β-galactosidase (SA-β-gal) activity and irreversible growth arrest. However, recent data suggest that TIS cells can reactivate their proliferative potential and lead to cancer recurrence. Our previous study indicated that reactivation of proliferation by TIS cells might be related with autophagy modulation. However, exact relationship between both processes required further studies. Therefore, the aim of our study was to investigate the role of autophagy in the senescence-related chemoresistance of lung cancer cells. For this purpose, human and murine lung cancer cells were treated with two commonly used chemotherapeutics: cisplatin (CIS), which forms DNA adducts or docetaxel (DOC), a microtubule poison. Hypoxia, often overlooked in experimental settings, has been implicated as a mechanism responsible for a significant change in the response to treatment. Thus, cells were cultured under normoxic (~19% O2) or hypoxic (1% O2) conditions. Herein, we show that hypoxia increases resistance to CIS. Lung cancer cells cultured under hypoxic conditions escaped from CIS-induced senescence, displayed reduced SA-β-gal activity and a decreased percentage of cells in the G2/M phase of the cell cycle. In turn, hypoxia increased the proliferation of lung cancer cells and the proportion of cells proceeding to the G0/G1 phase. Further molecular analyses demonstrated that hypoxia inhibited the prosenescent p53/p21 signaling pathway and induced epithelial to mesenchymal transition in CIS-treated cancer cells. In cells treated with DOC, such effects were not observed. Of importance, pharmacological autophagy inhibitor, hydroxychloroquine (HCQ) was capable of overcoming short-term CIS-induced resistance of lung cancer cells in hypoxic conditions. Altogether, our data demonstrated that hypoxia favors cancer cell escape from CIS-induced senescence, what could be overcome by inhibition of autophagy with HCQ. Therefore, we propose that HCQ might be used to interfere with the ability of senescent cancer cells to repopulate following exposure to DNA-damaging agents. This effect, however, needs to be tested in a long-term perspective for preclinical and clinical applications.
Collapse
Affiliation(s)
- Aleksandra Olszewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Agata Borkowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Monika Granica
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Justyna Karolczak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Bartosz Zglinicki
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Halina Was
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- *Correspondence: Halina Was,
| |
Collapse
|
27
|
Medeiros PJ, Pascetta SA, Kirsh SM, Al-Khazraji BK, Uniacke J. Expression of hypoxia inducible factor-dependent Neuropeptide Y Receptors Y1 and Y5 sensitizes hypoxic cells to NPY stimulation. J Biol Chem 2022; 298:101645. [PMID: 35093384 PMCID: PMC8861119 DOI: 10.1016/j.jbc.2022.101645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 11/17/2022] Open
Abstract
Neuropeptide Y (NPY) is an abundant neurohormone in the central and peripheral nervous system involved in feeding behavior, energy balance, nociception, and anxiety. Several NPY receptor (NPYR) subtypes display elevated expression in many cancers including in breast tumors where it is exploited for imaging and diagnosis. Here, we address how hypoxia, a common feature of the tumor microenvironment, influences the expression of the NPYRs. We show that NPY1R and NPY5R mRNA abundance is induced by hypoxia in a hypoxia inducible factor (HIF)-dependent manner in breast cancer cell lines MCF7 and MDA-MB-231. We demonstrate that HIFs bind to several genomic regions upstream of the NPY1R and NPY5R transcription start sites. In addition, the MAPK/ERK pathway is activated more rapidly upon NPY5R stimulation in hypoxic cells compared with normoxic cells. This pathway requires insulin-like growth factor 1 receptor (IGF1R) activity in normoxia, but not in hypoxic cells, which display resistance to the radiosensitizer and IGF1R inhibitor AG1024. Furthermore, hypoxic cells proliferate and migrate more when stimulated with NPY relative to normoxic cells and exhibit a more robust response to a Y5-specific agonist. Our data suggest that hypoxia-induced NPYRs render hypoxic cells more sensitive to NPY stimulation. Considering that breast tissue receives a constant supply of NPY, hypoxic breast tumors are the perfect storm for hyperactive NPYR. This study not only highlights a new relationship between the HIFs and NPYR expression and activity but may inform the use of chemotherapeutics targeting NPYRs and hypoxic cells.
Collapse
Affiliation(s)
- Philip J Medeiros
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Sydney A Pascetta
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Sarah M Kirsh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | | | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
28
|
Abstract
Cellular activities are finely regulated by numerous signaling pathways to support specific functions of complex life processes. Viruses are obligate intracellular parasites. Each step of viral replication is ultimately governed by the interaction of a virus with its host cells. Because of the demands of viral replication, the nutritional needs of virus-infected cells differ from those of uninfected cells. To improve their chances of survival and replication, viruses have evolved to commandeer cellular processes, including cell metabolism, augmenting these processes to support their needs. This article summarizes recent findings regarding virus-induced alterations to major cellular metabolic pathways focusing on how viruses modulate various signaling cascades to induce these changes. We begin with a general introduction describing the role played by signaling pathways in cellular metabolism. We then discuss how different viruses target these signaling pathways to reprogram host metabolism to favor the viral needs. We highlight the gaps in understanding metabolism-related virus-host interactions and discuss how studying these changes will enhance our understanding of fundamental processes involved in metabolic regulation. Finally, we discuss the potential to harness these processes to combat viral diseases, as well as other diseases, including metabolic disorders and cancers.
Collapse
|
29
|
Zhao S, Zhou L, Dicker DT, Lev A, Zhang S, Ross E, El-Deiry WS. Anti-cancer efficacy including Rb-deficient tumors and VHL-independent HIF1α proteasomal destabilization by dual targeting of CDK1 or CDK4/6 and HSP90. Sci Rep 2021; 11:20871. [PMID: 34686682 PMCID: PMC8536770 DOI: 10.1038/s41598-021-00150-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
A prevalent characteristic of solid tumors is intra-tumoral hypoxia. Hypoxia-inducible factor 1α (HIF1α) predominantly mediates the adaptive response to O2 oscillation and is linked to multiple malignant hallmarks. Here we describe a strategy to robustly target HIF1α by dual inhibition of CDK(s) and heat shock protein 90 (HSP90). We show that CDK1 may contribute to HSP90-mediated HIF1α stabilization. CDK1 knockdown enhances the decrease of HIF1α by HSP90 inhibition. Dual inhibition of CDK1 and HSP90 significantly increases apoptosis and synergistically inhibits cancer cell viability. Similarly, targeting CDK4/6 using FDA-approved inhibitors in combination with HSP90 inhibition shows a class effect on HIF1α inhibition and cancer cell viability suppression not only in colorectal but also in various other cancer types, including Rb-deficient cancer cells. Dual inhibition of CDK4/6 and HSP90 suppresses tumor growth in vivo. In summary, combined targeting of CDK(s) (CDK1 or CDK4/6) and HSP90 remarkably inhibits the expression level of HIF1α and shows promising anti-cancer efficacy with therapeutic potential.
Collapse
Affiliation(s)
- Shuai Zhao
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Pathobiology Graduate Program, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - David T Dicker
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA
| | - Avital Lev
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Eric Ross
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA. .,Pathobiology Graduate Program, Brown University, Providence, RI, USA. .,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA. .,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA. .,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA. .,Fox Chase Cancer Center, Philadelphia, PA, USA. .,Hematology/Oncology Division, Lifespan Cancer Institute, Providence, RI, USA.
| |
Collapse
|
30
|
Hala D, Faulkner P, He K, Kamalanathan M, Brink M, Simons K, Apaydin M, Hernout B, Petersen LH, Ivanov I, Qian X. An integrated in vivo and in silico analysis of the metabolism disrupting effects of CPI-613 on embryo-larval zebrafish (Danio rerio). Comp Biochem Physiol C Toxicol Pharmacol 2021; 248:109084. [PMID: 34051378 DOI: 10.1016/j.cbpc.2021.109084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/12/2023]
Abstract
CPI-613 is a mitochondrial metabolism disrupter that inhibits tricarboxylic acid (TCA) cycle activity. The consequences of TCA cycle disruption on various metabolic pathways and overall organismal physiology are not fully known. The present study integrates in vivo experimental data with an in silico stoichiometric metabolism model of zebrafish to study the metabolic pathways perturbed under CPI-613 exposure. Embryo-larval life stages of zebrafish (Danio rerio) were exposed to 1 μM CPI-613 for 20 days. Whole-organism respirometry measurements showed an initial suppression of O2 consumption at Day 5 of exposure, followed by recovery comparable to the solvent control (0.01% DMSO) by Day 20. Comparison of whole-transcriptome RNA-sequencing at Day 5 vs. 20 of exposure showed functional categories related to O2 binding and transport, antioxidant activity, FAD binding, and hemoglobin complexes, to be commonly represented. Metabolic enzyme gene expression changes and O2 consumption rate was used to parametrize two in silico stoichiometric metabolic models representative of Day 5 or 20 of exposure. Computational simulations predicted impaired ATP synthesis, α-ketoglutarate dehydrogenase (KGDH) activity, and fatty acid β-oxidation at Day 5 vs. 20 of exposure. These results show that the targeted disruption of KGDH may also impact oxidative phosphorylation (ATP synthesis) and fatty acid metabolism (β-oxidation), in turn influencing cellular bioenergetics and the observed reduction in whole-organism O2 consumption rate. The results of this study provide an integrated in vivo and in silico framework to study the impacts of metabolic disruption on organismal physiology.
Collapse
Affiliation(s)
- David Hala
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA; Department of Ecology and Conservation Biology, Texas A&M University, College Station, TX, USA.
| | - Patricia Faulkner
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA
| | - Kai He
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Manoj Kamalanathan
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA
| | - Mikeelee Brink
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA
| | - Kristina Simons
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA
| | - Meltem Apaydin
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Beatrice Hernout
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA; Institute for a Sustainable Environment, Department of Biology, Clarkson University, Potsdam, NY, USA
| | - Lene H Petersen
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA
| | - Ivan Ivanov
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX, USA
| | - Xiaoning Qian
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, TX, USA
| |
Collapse
|
31
|
Cha JE, Bae WY, Choi JS, Lee SH, Jeong JW. Angiogenic activities are increased via upregulation of HIF-1α expression in gefitinib-resistant non-small cell lung carcinoma cells. Oncol Lett 2021; 22:671. [PMID: 34345296 PMCID: PMC8323004 DOI: 10.3892/ol.2021.12932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) have been used to treat patients with non-small cell lung cancer (NSCLC) and activating EGFR mutations; however, the emergence of secondary mutations in EGFR or the acquisition of resistance to EGFR-TKIs can develop and is involved in clinical failure. Since angiogenesis is associated with tumor progression and the blockade of antitumor drugs, inhibition of angiogenesis could be a rational strategy for developing anticancer drugs combined with EGFR-TKIs to treat patients with NSCLC. The signaling pathway mediated by hypoxia-inducible factor-1 (HIF-1) is essential for tumor angiogenesis. The present study aimed to identify the dependence of gefitinib resistance on HIF-1α activity using angiogenesis assays, western blot analysis, colony formation assay, xenograft tumor mouse model and immunohistochemical analysis of tumor tissues. In the NSCLC cell lines, HIF-1α protein expression levels and hypoxia-induced angiogenic activities were found to be increased. In a xenograft mouse tumor model, tumor tissues derived from gefitinib-resistant PC9 cells showed increased protein expression of HIF-1α and angiogenesis within the tumors. Furthermore, inhibition of HIF-1α suppressed resistance to gefitinib, whereas overexpression of HIF-1α increased resistance to gefitinib. The results from the present study provides evidence that HIF-1α was associated with the acquisition of resistance to gefitinib and suggested that inhibiting HIF-1α alleviated gefitinib resistance in NSCLC cell lines.
Collapse
Affiliation(s)
- Jeong Eun Cha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woom-Yee Bae
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae-Sun Choi
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.,Medical Science Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
32
|
Wu D, Dasgupta A, Read AD, Bentley RET, Motamed M, Chen KH, Al-Qazazi R, Mewburn JD, Dunham-Snary KJ, Alizadeh E, Tian L, Archer SL. Oxygen sensing, mitochondrial biology and experimental therapeutics for pulmonary hypertension and cancer. Free Radic Biol Med 2021; 170:150-178. [PMID: 33450375 PMCID: PMC8217091 DOI: 10.1016/j.freeradbiomed.2020.12.452] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
The homeostatic oxygen sensing system (HOSS) optimizes systemic oxygen delivery. Specialized tissues utilize a conserved mitochondrial sensor, often involving NDUFS2 in complex I of the mitochondrial electron transport chain, as a site of pO2-responsive production of reactive oxygen species (ROS). These ROS are converted to a diffusible signaling molecule, hydrogen peroxide (H2O2), by superoxide dismutase (SOD2). H2O2 exits the mitochondria and regulates ion channels and enzymes, altering plasma membrane potential, intracellular Ca2+ and Ca2+-sensitization and controlling acute, adaptive, responses to hypoxia that involve changes in ventilation, vascular tone and neurotransmitter release. Subversion of this O2-sensing pathway creates a pseudohypoxic state that promotes disease progression in pulmonary arterial hypertension (PAH) and cancer. Pseudohypoxia is a state in which biochemical changes, normally associated with hypoxia, occur despite normal pO2. Epigenetic silencing of SOD2 by DNA methylation alters H2O2 production, activating hypoxia-inducible factor 1α, thereby disrupting mitochondrial metabolism and dynamics, accelerating cell proliferation and inhibiting apoptosis. Other epigenetic mechanisms, including dysregulation of microRNAs (miR), increase pyruvate dehydrogenase kinase and pyruvate kinase muscle isoform 2 expression in both diseases, favoring uncoupled aerobic glycolysis. This Warburg metabolic shift also accelerates cell proliferation and impairs apoptosis. Disordered mitochondrial dynamics, usually increased mitotic fission and impaired fusion, promotes disease progression in PAH and cancer. Epigenetic upregulation of dynamin-related protein 1 (Drp1) and its binding partners, MiD49 and MiD51, contributes to the pathogenesis of PAH and cancer. Finally, dysregulation of intramitochondrial Ca2+, resulting from impaired mitochondrial calcium uniporter complex (MCUC) function, links abnormal mitochondrial metabolism and dynamics. MiR-mediated decreases in MCUC function reduce intramitochondrial Ca2+, promoting Warburg metabolism, whilst increasing cytosolic Ca2+, promoting fission. Epigenetically disordered mitochondrial O2-sensing, metabolism, dynamics, and Ca2+ homeostasis offer new therapeutic targets for PAH and cancer. Promoting glucose oxidation, restoring the fission/fusion balance, and restoring mitochondrial calcium regulation are promising experimental therapeutic strategies.
Collapse
Affiliation(s)
- Danchen Wu
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Asish Dasgupta
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Austin D Read
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Rachel E T Bentley
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Mehras Motamed
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Ruaa Al-Qazazi
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Jeffrey D Mewburn
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada
| | - Kimberly J Dunham-Snary
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Elahe Alizadeh
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Queen's University, 116 Barrie Street, Kingston, ON, K7L 3J9, Canada
| | - Lian Tian
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Stephen L Archer
- Department of Medicine, Queen's University, 94 Stuart St., Kingston, Ontario, K7L 3N6, Canada.
| |
Collapse
|
33
|
Abstract
Survivin is one of the rare proteins that is differentially expressed in normal and cancer cells and is directly or indirectly involved in numerous pathways required for tumor maintenance. It is expressed in almost all cancers and its expression has been detected at early stages of cancer. These traits make survivin an exceptionally attractive target for cancer therapeutics. Even with these promising features to be an oncotherapeutic target, there has been limited success in the clinical trials targeting survivin. Only recently it has emerged that survivin was not being specifically targeted which could have resulted in the negative clinical outcome. Also, focus of research has now shifted from survivin expression in the overall heterogeneous tumor cell populations to survivin expression in cancer stem cells as these cells have proved to be the major drivers of tumors. Therefore, in this review we have analyzed the expression of survivin in normal and cancer cells with a particular focus on its expression in cancer stem cell compartment. We have discussed the major signaling pathways involved in regulation of survivin. We have explored the current development status of various types of interventions for inhibition of survivin. Furthermore, we have discussed the challenges involving the development of potent and specific survivin inhibitors for cancer therapeutics. Finally we have given insights for some of the promising future anticancer treatments.
Collapse
|
34
|
Isomura H, Taguchi A, Kajino T, Asai N, Nakatochi M, Kato S, Suzuki K, Yanagisawa K, Suzuki M, Fujishita T, Yamaguchi T, Takahashi M, Takahashi T. Conditional Ror1 knockout reveals crucial involvement in lung adenocarcinoma development and identifies novel HIF-1α regulator. Cancer Sci 2021; 112:1614-1623. [PMID: 33506575 PMCID: PMC8019194 DOI: 10.1111/cas.14825] [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: 11/29/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 12/13/2022] Open
Abstract
We previously reported that ROR1 is a crucial downstream gene for the TTF‐1/NKX2‐1 lineage‐survival oncogene in lung adenocarcinoma, while others have found altered expression of ROR1 in multiple cancer types. Accumulated evidence therefore indicates ROR1 as an attractive molecular target, though it has yet to be determined whether targeting Ror1 can inhibit tumor development and growth in vivo. To this end, genetically engineered mice carrying homozygously floxed Ror1 alleles and an SP‐C promoter–driven human mutant EGFR transgene were generated. Ror1 ablation resulted in marked retardation of tumor development and progression in association with reduced malignant characteristics and significantly better survival. Interestingly, gene set enrichment analysis identified a hypoxia‐induced gene set (HALLMARK_HYPOXIA) as most significantly downregulated by Ror1 ablation in vivo, which led to findings showing that ROR1 knockdown diminished HIF‐1α expression under normoxia and clearly hampered HIF‐1α induction in response to hypoxia in human lung adenocarcinoma cell lines. The present results directly demonstrate the importance of Ror1 for in vivo development and progression of lung adenocarcinoma, and also identify Ror1 as a novel regulator of Hif‐1α. Thus, a future study aimed at the development of a novel therapeutic targeting ROR1 for treatment of solid tumors such as seen in lung cancer, which are frequently accompanied with a hypoxic tumor microenvironment, is warranted.
Collapse
Affiliation(s)
- Hisanori Isomura
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Molecular Diagnostics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Ayumu Taguchi
- Division of Molecular Diagnostics, Aichi Cancer Center Research Institute, Nagoya, Japan.,Division of Advanced Cancer Diagnostics, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taisuke Kajino
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Molecular Diagnostics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Naoya Asai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Pathology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiichi Kato
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Keiko Suzuki
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyoshi Yanagisawa
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Motoshi Suzuki
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Molecular Oncology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Teruaki Fujishita
- Division of Pathophysiology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Tomoya Yamaguchi
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Cancer Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| | - Masahide Takahashi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Takahashi
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
35
|
Abbaszade Z, Bagca BG, Avci CB. Molecular biological investigation of temozolomide and KC7F2 combination in U87MG glioma cell line. Gene 2021; 776:145445. [PMID: 33484758 DOI: 10.1016/j.gene.2021.145445] [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: 05/15/2020] [Revised: 12/25/2020] [Accepted: 01/13/2021] [Indexed: 11/25/2022]
Abstract
Glioblastom Multiforme (GBM) is the most invasive and malignant member of the IV grade of the subclass Astrocytoma according to the last assessment of the 2016 WHO report. Due to the resistance to treatment and weak response, as well as the topographical structure of the blood brain barrier, the treatment is also difficult due to the severe clinical manifestation, and new treatment methods and new therapeutic agents are needed. Temozolomide (TMZ) is widely used in the treatment of glioblastoma and is considered as the primary treatment modality. TMZ, a member of the class of cognitive agents, is currently considered the most effective drug because it can easily pass through the blood brain barrier. Glucose metabolism is a complex energy producing machine that, a glucose molecule produces 38 molecules of ATP after full glycolytic catabolism. According to Otto Warburg's numerous studies cancer cells perform the first glycolytic step without entering the mitochondrial step. These cells produce lactic acid and make the micro-media more acidic even in aerobic conditions. This phenomenon is attributed to the Warburg hypothesis and either as aerobic glycolysis. Although glycolysis enzymes are the primary actors of this phenotypic expression, some genetic and epigenetic factors are no exception. We experimentally used KC7F2 active ingredient to target cancer metabolism. In our study, we evaluated cancer metabolism in combination with the effect of TMZ chemotherapeutic agent, examining the effect of two different agents separately and in combination to observe the effects of cancer cell proliferation, survival, apoptosis and expression of metabolism genes on expression. We observed that the combined effect of reduced the effective dose of the TMZ alkylating agent and that the effect was increased and the effect of the combined teraphy is assessed from a metabolic point of view and that it suppresses aerobic glycolysis.
Collapse
Affiliation(s)
- Zaka Abbaszade
- Kazımdirik, Ege Ünv. Hst. No:9, 35100 Bornova/Izmir, Turkey.
| | | | | |
Collapse
|
36
|
Ma Y, Wu T, Zhou H, He G, Li Y, Wang B, Guo Q, Chen B, Li W. Canstatin represses glioma growth by inhibiting formation of VM-like structures. Transl Neurosci 2021; 12:309-319. [PMID: 34434564 PMCID: PMC8357010 DOI: 10.1515/tnsci-2020-0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vasculogenic mimicry (VM) is different from classical tumor angiogenesis and does not depend on endothelial cells. VM is closely related to the prognosis of various cancers. Canstatin was first identified as an endogenous angiogenesis inhibitor. In the present study, the inhibitory effect of canstatin on VM formation was evaluated. Human glioblastoma cell lines U87 and U251 were letivirally transduced to overexpress canstatin gene or GFP as control. In vitro assays showed that canstatin overexpression reduced the tube formation of U87 and U251 cells in Matrigel. A xenograft glioma model was created by subcutaneous injection of lentivirally modified U87 cells into nude mice. The results of in vivo experiments showed that canstatin gene introduction inhibited the growth of glioma xenografts. In tumor xenografts overexpressing canstatin, U87-mediated formation of VM-like structures and VM-related VEGF (vascular endothelial growth factor) expression were remarkably reduced. Canstatin overexpression also decreased the phosphorylation of Akt and reduced the expression of Survivin in vitro. In addition, HIF-1α production and MMP-2 secretion were decreased by canstatin overexpression. Therefore, these results suggested a protective role of canstatin during VM-like structure formation of glioma probably via inhibiting signaling pathways inducing vasculogenic mimicry.
Collapse
Affiliation(s)
- Yuqiang Ma
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Tao Wu
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Houjie Zhou
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Guilu He
- GuangDong 999 Brain Hospital, Guangzhou 510510, People's Republic of China
| | - Yifei Li
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Bocheng Wang
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Qiang Guo
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Baodong Chen
- Neurosurgery Department, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Weiping Li
- Neurosurgery Department, Shenzhen Second People's Hospital, Sungang Road, Futian District, Guangzhou 510510, People's Republic of China
| |
Collapse
|
37
|
JCPyV T-Antigen Activation of the Anti-Apoptotic Survivin Promoter-Its Role in the Development of Progressive Multifocal Leukoencephalopathy. Viruses 2020; 12:v12111253. [PMID: 33153187 PMCID: PMC7693140 DOI: 10.3390/v12111253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
Progressive Multifocal Leukoencephalopathy (PML) is a fatal demyelinating disease of the CNS, resulting from the lytic infection of oligodendrocytes by the human neurotropic polyomavirus JC (JCPyV), typically associated with severe immunocompromised states and, in recent years, with the use of immunotherapies. Apoptosis is a homeostatic mechanism to dispose of senescent or damaged cells, including virally infected cells, triggered in the vast majority of viral infections of the brain. Previously, we showed upregulation of the normally dormant anti-apoptotic protein Survivin in cases of PML, which—in vitro—resulted in protection from apoptosis in JCPyV-infected primary cultures of astrocytes and oligodendrocytes. In the present study, we first demonstrate the absence of apoptotic DNA fragmentation and the lack of caspase activity in 16 cases of PML. We also identified the viral protein large T-Antigen as being responsible for the activation of the Survivin promoter. Chromatin Immunoprecipitation assay shows a direct binding between T-Antigen and the Survivin promoter DNA. Finally, we have identified the specific region of T-Antigen, spanning from amino acids 266 and 688, which binds to Survivin and translocates it to the nucleus, providing evidence of a mechanism that results in the efficient replication of JCPyV and a potential target for novel therapies.
Collapse
|
38
|
Liu SH, Yu J, Creeden JF, Sutton JM, Markowiak S, Sanchez R, Nemunaitis J, Kalinoski A, Zhang JT, Damoiseaux R, Erhardt P, Brunicardi FC. Repurposing metformin, simvastatin and digoxin as a combination for targeted therapy for pancreatic ductal adenocarcinoma. Cancer Lett 2020; 491:97-107. [PMID: 32829010 DOI: 10.1016/j.canlet.2020.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/23/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Patients with pancreatic adenocarcinoma (PDAC) have a 5-year survival rate of 8%, the lowest of any cancer in the United States. Traditional chemotherapeutic regimens, such as gemcitabine- and fluorouracil-based regimens, often only prolong survival by months. Effective precision targeted therapy is therefore urgently needed to substantially improve survival. In an effort to expedite approval and delivery of targeted therapy to patients, we utilized a platform to develop a novel combination of FDA approved drugs that would target pancreaticoduodenal homeobox1 (PDX1) and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) utilizing super-promoters of the target genes to interrogate an FDA approved drug library. We identified and selected metformin, simvastatin and digoxin (C3) as a novel combination of FDA approved drugs, which were shown to effectively target PDX1 and BIRC5 in human PDAC tumors in mice with no toxicity.
Collapse
Affiliation(s)
- Shi-He Liu
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - Juehua Yu
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Justin F Creeden
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jeffrey M Sutton
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Stephen Markowiak
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Robbi Sanchez
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Andrea Kalinoski
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jian-Ting Zhang
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Paul Erhardt
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, 43614, USA
| | - F Charles Brunicardi
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| |
Collapse
|
39
|
Role of Adipose Tissue-Derived Autotaxin, Lysophosphatidate Signaling, and Inflammation in the Progression and Treatment of Breast Cancer. Int J Mol Sci 2020; 21:ijms21165938. [PMID: 32824846 PMCID: PMC7460696 DOI: 10.3390/ijms21165938] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022] Open
Abstract
Autotaxin (ATX) is a secreted enzyme that produces lysophosphatidate (LPA), which signals through six G-protein coupled receptors, promoting tumor growth, metastasis, and survival from chemotherapy and radiotherapy. Many cancer cells produce ATX, but breast cancer cells express little ATX. In breast tumors, ATX is produced by tumor-associated stroma. Breast tumors are also surrounded by adipose tissue, which is a major bodily source of ATX. In mice, a high-fat diet increases adipocyte ATX production. ATX production in obesity is also increased because of low-level inflammation in the expanded adipose tissue. This increased ATX secretion and consequent LPA signaling is associated with decreased adiponectin production, which results in adverse metabolic profiles and glucose homeostasis. Increased ATX production by inflamed adipose tissue may explain the obesity-breast cancer association. Breast tumors produce inflammatory mediators that stimulate ATX transcription in tumor-adjacent adipose tissue. This drives a feedforward inflammatory cycle since increased LPA signaling increases production of more inflammatory mediators and cyclooxygenase-2. Inhibiting ATX activity, which has implications in breast cancer adjuvant treatments, attenuates this cycle. Targeting ATX activity and LPA signaling may potentially increase chemotherapy and radiotherapy efficacy, and decrease radiation-induced fibrosis morbidity independently of breast cancer type because most ATX is not derived from breast cancer cells.
Collapse
|
40
|
Docosahexaenoic acid (DHA) inhibits pro-angiogenic effects of breast cancer cells via down-regulating cellular and exosomal expression of angiogenic genes and microRNAs. Life Sci 2020; 258:118094. [PMID: 32673663 DOI: 10.1016/j.lfs.2020.118094] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022]
Abstract
AIMS Docosahexaenoic acid (DHA) as an omega 3 free fatty acid has been reported to exert anti-angiogenesis effects. However, our current understanding regarding the precise mechanisms of such effects is still limited. Exosomes secreted by cancer cells may act as angiogenesis promoters. The aim of the study was to determine altered expression levels of HIF-1α, TGF-β, VEGFR, Snail1, Snail2 and SOX2 and their regulating microRNAs in MDA-MB-231 and BT-474 cell lines after treatment with DHA in both normoxic and hypoxic conditions. MAIN METHODS Human breast cancer cell lines including MDA-MB-231 and BT-474 were treated for 24 h with 100 uM DHA under normoxic and hypoxic conditions. Exosomes were isolated from untreated and treated cells and characterized by transmission electron microscopy (TEM) and western blotting. RNAs from cells and isolated exosomes were extracted and cDNAs were synthesized. Expression levels of miRNAs and their pro-angiogenic target genes were analyzed using quantitative real-time PCR (qRT-PCR). KEY FINDINGS We showed significant decrease in the expression of pro-angiogenic genes including HIF1-α, TGF-β, SOX2, Snail1, Snail2 and VEGFR in cells and also their secreted exosomes after treatment with DHA in normoxic and hypoxic conditions. Also the expression levels of tumor suppressor miRs including miR-101, miR-199, miR-342 were increased and the expression levels of oncomiRs including mir-382 and miR-21 were decreased after treatment with DHA in cells and exosomes. SIGNIFICANCE DHA can alter the expression of pro-angiogenic genes and microRNA contents in breast cancer cells and their derived-exosomes in favor of the inhibition of angiogenesis. Our data demonstrated new insight into DHA's anti-cancer action to target not only breast cancer cells but also their derived exosomes to suppress tumor angiogenesis.
Collapse
|
41
|
Maggs L, Ferrone S. Improving the Clinical Significance of Preclinical Immunotherapy Studies through Incorporating Tumor Microenvironment-like Conditions. Clin Cancer Res 2020; 26:4448-4453. [PMID: 32571789 DOI: 10.1158/1078-0432.ccr-20-0358] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/03/2020] [Accepted: 06/05/2020] [Indexed: 01/22/2023]
Abstract
Frequently, the results generated when testing novel antitumor immunotherapies in vitro do not correlate with data collected in in vivo models and/or in clinical settings. It is our hypothesis that this discrepancy is caused by the use of in vitro conditions, such as normoxia, a two-dimensional surface, optimal growth media, and lack of cell complexity and heterogeneity. These conditions do not accurately reflect the tumor microenvironment (TME) that the tested immunotherapeutic strategies experience in vivo While there are many variables which can have an impact upon the antitumor efficacy of an immunotherapy, the immunosuppressive TME is one in which several of the conditions commonly found in vivo can be mimicked in vitro These conditions, which include hypoxia, low pH, low glucose, presence of adenosine, cell complexity and heterogeneity, as well as the three-dimensional structure of TME, can all affect immune cell-tumor cell interactions. Here, we discuss the impact that these conditions, either individually or in combination, can have on these interactions. Furthermore, we propose that performing in vitro assays under TME-like conditions improves the clinical relevance of the yielded results. This, in turn, contributes to accelerate the speed, reduce the cost, and increase efficiency of screening novel immunotherapies and eventually the development of prospective clinical trials.
Collapse
Affiliation(s)
- Luke Maggs
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
42
|
Cui J, Wang J, Lin C, Liu J, Zuo W. Osteopontin Mediates Cetuximab Resistance via the MAPK Pathway in NSCLC Cells. Onco Targets Ther 2020; 12:10177-10185. [PMID: 32063712 PMCID: PMC6884967 DOI: 10.2147/ott.s228437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/05/2019] [Indexed: 01/21/2023] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. The high expression of osteopontin (OPN) is an important factor that aggravates drug resistance and causes a poor prognosis in this disease. Therefore, understanding the molecular mechanism of OPN is critical for the treatment and prognosis of NSCLC. Methods We used bioinformatics analysis to verify the expression of OPN in normal lung tissues and lung cancer tissues. Then we overexpressed and knocked down OPN in cell lines to detect cell proliferation, migration, invasion, and effects on signaling pathways. Finally, malignant progression and drug resistance induced by OPN were investigated by the wound healing assay, transwell assay, clone formation assay, and Western blot analysis. Results We verified that OPN was upregulated in NSCLC tissues, and its overexpression induced NSCLC cell proliferation, migration, and invasion via the mitogen-activated protein kinase (MAPK) pathway. Furthermore, overexpression of OPN reduced the sensitivity of NSCLC cells to cetuximab by upregulating MAPK pathway-related proteins. These results suggested that OPN promoted malignant progression and mediated drug resistance via the MAPK signaling pathway in NSCLC cells. Conclusion This study reveals the important role of OPN in NSCLC cells, making it a potential target for improving chemotherapy efficiency in patients with NSCLC.
Collapse
Affiliation(s)
- Jian Cui
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Jun Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Chao Lin
- Department of General Practice, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Jixiang Liu
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Wei Zuo
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| |
Collapse
|
43
|
Yu Y, Guan H, Jiang L, Li X, Xing L, Sun X. Nimotuzumab, an EGFR‑targeted antibody, promotes radiosensitivity of recurrent esophageal squamous cell carcinoma. Int J Oncol 2020; 56:945-956. [PMID: 32319582 DOI: 10.3892/ijo.2020.4981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/12/2019] [Indexed: 11/06/2022] Open
Abstract
Local tumor recurrence is one of the main causes for the failure of esophageal cancer treatment following radiotherapy. Previous studies have demonstrated that epidermal growth factor receptor (EGFR)‑targeted therapy combined with radiotherapy is expected to become an effective means to control tumor recurrence. The aim of the present study was to investigate the effect and mechanism of nimotuzumab (an EGFR‑targeted antibody) in the treatment of recurrent esophageal carcinoma. The radiation responses of two esophageal squamous carcinoma cell lines, EC109 and TE‑1, with or without nimotuzumab, were first evaluated by CCK‑8 assay. Colony formation and apoptosis were used to measure anti‑proliferation effects. It was demonstrated that nimotuzumab arrested the cell cycle at the G2 phase in vitro. Western blotting and immunofluorescence analysis were used to determine signaling pathway changes. It was observed that nimotuzumab inhibited phosphorylation of EGFR in EC109 cells. Furthermore, recurrent tumor models were established and it was identified that the degree of tumor hypoxia was positively associated with EGFR overexpression. In EC109 cell xenografts, nimotuzumab combined with radiation led to a significant delay in recurrent tumor growth compared with that of radiation alone (P<0.001 for 0 Gy pre‑irradiation, P=0.005 for 20 Gy pre‑irradiation, P=0.005 for 10 Gy pre‑irradiation). These results suggest that nimotuzumab combined with radiation may be an effective means to control recurrent esophageal squamous cell carcinoma with EGFR overexpression.
Collapse
Affiliation(s)
- Yang Yu
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250031, P.R. China
| | - Hui Guan
- Department of Radiation Oncology, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| | - Liyang Jiang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xiaolin Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
44
|
Whiting D, Sriprasad S. Molecular biology and targeted therapy in metastatic renal cell carcinoma. JOURNAL OF CLINICAL UROLOGY 2020. [DOI: 10.1177/2051415819849322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The treatment of metastatic renal cell carcinoma is challenging as it has proven to be relatively resistant to conventional oncological treatments. An improved understanding of the molecular biology of renal cell carcinoma has led to the development of a number of targeted therapies in metastatic renal cell carcinoma. This includes vascular endothelial growth factor inhibitors, tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors and most recently immune checkpoint inhibitors. This article will review the mechanisms of development and progression of renal cell carcinoma as well as the mechanisms of current approved treatments in metastatic disease.Level of evidence: Not applicable for this multicentre audit.
Collapse
Affiliation(s)
- D Whiting
- Department of Urology, Darent Valley Hospital, UK
| | - S Sriprasad
- Department of Urology, Darent Valley Hospital, UK
| |
Collapse
|
45
|
Kobliakov VA. The Mechanisms of Regulation of Aerobic Glycolysis (Warburg Effect) by Oncoproteins in Carcinogenesis. BIOCHEMISTRY (MOSCOW) 2019; 84:1117-1128. [DOI: 10.1134/s0006297919100018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Causes and Consequences of A Glutamine Induced Normoxic HIF1 Activity for the Tumor Metabolism. Int J Mol Sci 2019; 20:ijms20194742. [PMID: 31554283 PMCID: PMC6802203 DOI: 10.3390/ijms20194742] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/01/2019] [Accepted: 09/15/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factor hypoxia-inducible factor 1 (HIF1) is the crucial regulator of genes that are involved in metabolism under hypoxic conditions, but information regarding the transcriptional activity of HIF1 in normoxic metabolism is limited. Different tumor cells were treated under normoxic and hypoxic conditions with various drugs that affect cellular metabolism. HIF1α was silenced by siRNA in normoxic/hypoxic tumor cells, before RNA sequencing and bioinformatics analyses were performed while using the breast cancer cell line MDA-MB-231 as a model. Differentially expressed genes were further analyzed and validated by qPCR, while the activity of the metabolites was determined by enzyme assays. Under normoxic conditions, HIF1 activity was significantly increased by (i) glutamine metabolism, which was associated with the release of ammonium, and it was decreased by (ii) acetylation via acetyl CoA synthetase (ACSS2) or ATP citrate lyase (ACLY), respectively, and (iii) the presence of L-ascorbic acid, citrate, or acetyl-CoA. Interestingly, acetylsalicylic acid, ibuprofen, L-ascorbic acid, and citrate each significantly destabilized HIF1α only under normoxia. The results from the deep sequence analyses indicated that, in HIF1-siRNA silenced MDA-MB-231 cells, 231 genes under normoxia and 1384 genes under hypoxia were transcriptionally significant deregulated in a HIF1-dependent manner. Focusing on glycolysis genes, it was confirmed that HIF1 significantly regulated six normoxic and 16 hypoxic glycolysis-associated gene transcripts. However, the results from the targeted metabolome analyses revealed that HIF1 activity affected neither the consumption of glucose nor the release of ammonium or lactate; however, it significantly inhibited the release of the amino acid alanine. This study comprehensively investigated, for the first time, how normoxic HIF1 is stabilized, and it analyzed the possible function of normoxic HIF1 in the transcriptome and metabolic processes of tumor cells in a breast cancer cell model. Furthermore, these data imply that HIF1 compensates for the metabolic outcomes of glutaminolysis and, subsequently, the Warburg effect might be a direct consequence of the altered amino acid metabolism in tumor cells.
Collapse
|
47
|
Brooks JM, Menezes AN, Ibrahim M, Archer L, Lal N, Bagnall CJ, von Zeidler SV, Valentine HR, Spruce RJ, Batis N, Bryant JL, Hartley M, Kaul B, Ryan GB, Bao R, Khattri A, Lee SP, Ogbureke KUE, Middleton G, Tennant DA, Beggs AD, Deeks J, West CML, Cazier JB, Willcox BE, Seiwert TY, Mehanna H. Development and Validation of a Combined Hypoxia and Immune Prognostic Classifier for Head and Neck Cancer. Clin Cancer Res 2019; 25:5315-5328. [PMID: 31182433 DOI: 10.1158/1078-0432.ccr-18-3314] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/22/2019] [Accepted: 06/06/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Intratumoral hypoxia and immunity have been correlated with patient outcome in various tumor settings. However, these factors are not currently considered for treatment selection in head and neck cancer (HNC) due to lack of validated biomarkers. Here we sought to develop a hypoxia-immune classifier with potential application in patient prognostication and prediction of response to targeted therapy. EXPERIMENTAL DESIGN A 54-gene hypoxia-immune signature was constructed on the basis of literature review. Gene expression was analyzed in silico using the The Cancer Genome Atlas (TCGA) HNC dataset (n = 275) and validated using two independent cohorts (n = 130 and 123). IHC was used to investigate the utility of a simplified protein signature. The spatial distribution of hypoxia and immune markers was examined using multiplex immunofluorescence staining. RESULTS Unsupervised hierarchical clustering of TCGA dataset (development cohort) identified three patient subgroups with distinct hypoxia-immune phenotypes and survival profiles: hypoxialow/immunehigh, hypoxiahigh/immunelow, and mixed, with 5-year overall survival (OS) rates of 71%, 51%, and 49%, respectively (P = 0.0015). The prognostic relevance of the hypoxia-immune gene signature was replicated in two independent validation cohorts. Only PD-L1 and intratumoral CD3 protein expression were associated with improved OS on multivariate analysis. Hypoxialow/immunehigh and hypoxiahigh/immunelow tumors were overrepresented in "inflamed" and "immune-desert" microenvironmental profiles, respectively. Multiplex staining demonstrated an inverse correlation between CA-IX expression and prevalence of intratumoral CD3+ T cells (r = -0.5464; P = 0.0377), further corroborating the transcription-based classification. CONCLUSIONS We developed and validated a hypoxia-immune prognostic transcriptional classifier, which may have clinical application to guide the use of hypoxia modification and targeted immunotherapies for the treatment of HNC.
Collapse
Affiliation(s)
- Jill M Brooks
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Albert N Menezes
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Maha Ibrahim
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Lucinda Archer
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Neeraj Lal
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Christopher J Bagnall
- Human Biomaterials Resource Centre, University of Birmingham, Birmingham, United Kingdom
| | - Sandra V von Zeidler
- Department of Pathology, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Helen R Valentine
- Division of Cancer Sciences, University of Manchester, Christie Hospital, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rachel J Spruce
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nikolaos Batis
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jennifer L Bryant
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Margaret Hartley
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Baksho Kaul
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Gordon B Ryan
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Riyue Bao
- The University of Chicago Medicine, Chicago, Illinois
| | - Arun Khattri
- The University of Chicago Medicine, Chicago, Illinois
| | - Steven P Lee
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Kalu U E Ogbureke
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Andrew D Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jonathan Deeks
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Catharine M L West
- Division of Cancer Sciences, University of Manchester, Christie Hospital, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jean-Baptiste Cazier
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Benjamin E Willcox
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | | | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom.
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
48
|
Dong L, Du M, Lv Q. Picropodophyllin inhibits type I endometrial cancer cell proliferation via disruption of the PI3K/Akt pathway. Acta Biochim Biophys Sin (Shanghai) 2019; 51:753-760. [PMID: 31168597 DOI: 10.1093/abbs/gmz055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
The type-I insulin-like growth factor receptor (IGF-IR) is overexpressed in endometrial cancer. High IGF-IR expression was considered as an important prognostic factor for tumor progression. The purpose of this study was to investigate the role and molecular mechanism of IGF-IR inhibitor picropodophyllin (PPP) in the growth and development of endometrial cancer. High expression of IGF-IR was observed in endometrial cancer tissues, as well as in ECC-1 and KLE cell lines. PPP suppressed the number of clones of ECC-1 and KLE cell lines; however, it had no significant effect on HEC-1-A cell line, which expressed lower IGF-IR than ECC-1 and KLE cell lines. Furthermore, PPP reduced cell proliferation capacity, inhibited the IGF-IR mRNA expression, and suppressed protein phosphorylation of IGF-IR and Akt in the three cell lines. In addition, PPP inhibited the protein expression of survivin in KLE cell line after 1 h of exposure, though this effect did not last for prolonged time. In conclusion, IGF-IR was mostly overexpressed in type I endometrial cancer. High IGF-IR expression was an important prognostic factor of tumor progression. PPP mediated the down-regulation of IGF-IR phosphorylation and inhibited cell proliferation via the PI3K/Akt signal pathway. PPP may have the potential to become a clinical treatment target in endometrial carcinoma.
Collapse
Affiliation(s)
- Lin Dong
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meirong Du
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Qianzhou Lv
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Galloway NR, Ball KF, Stiff T, Wall NR. Yin Yang 1 (YY1): Regulation of Survivin and Its Role In Invasion and Metastasis. Crit Rev Oncog 2019; 22:23-36. [PMID: 29604934 DOI: 10.1615/critrevoncog.2017020836] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite significant clinical and basic science advancements, cancer remains a devastating disease that affects people of all ages, races, and backgrounds. The pathogenesis of cancer has recently been described to result from eight biological capabilities or hallmarks and two enabling characteristics. These eight hallmarks are: deregulation of cellular energetics, avoiding immune destruction, enabling replicative immortality, inducing angiogenesis, sustaining proliferative signaling, evading growth suppressors, resisting cell death, and activating invasion and metastasis. The enabling characteristics are: genome instability and mutation and tumor-promoting inflammation. Survivin, the fourth most common transcript found in cancer cells, is a protein that is thought to be involved in the enhanced proliferation, survival, and metastasis and possibly other key hallmarks of cancer cells. Understanding how this gene is turned on and off is vitally important for attempt improving cancer management and therapy. Our work has identified a novel transcriptional regulator of survivin called Yin Yang 1 (YY1), which has been observed to activate some gene promoters and repress others and is gaining increasing interest as a target of cancer therapy. Our work shows for the first time that YY1 represses survivin transcription by physically interacting with the survivin promoter. Furthermore, YY1 appears to contribute to basal survivin transcriptional activity, indicating that disruption of its binding may in part contribute to survivin overexpression after cellular stress events including chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Nicholas R Galloway
- Department of Basic Science and Division of Biochemistry, Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California 92350
| | - Kathryn F Ball
- Department of Basic Science and Division of Biochemistry, Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California 92350
| | - TessaRae Stiff
- Department of Basic Science and Division of Biochemistry, Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California 92350
| | - Nathan R Wall
- Department of Basic Science and Division of Biochemistry, Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California 92350
| |
Collapse
|
50
|
Al Tameemi W, Dale TP, Al-Jumaily RMK, Forsyth NR. Hypoxia-Modified Cancer Cell Metabolism. Front Cell Dev Biol 2019; 7:4. [PMID: 30761299 PMCID: PMC6362613 DOI: 10.3389/fcell.2019.00004] [Citation(s) in RCA: 334] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/10/2019] [Indexed: 12/20/2022] Open
Abstract
While oxygen is critical to the continued existence of complex organisms, extreme levels of oxygen within a system, known as hypoxia (low levels of oxygen) and hyperoxia (excessive levels of oxygen), potentially promote stress within a defined biological environment. The consequences of tissue hypoxia, a result of a defective oxygen supply, vary in response to the gravity, extent and environment of the malfunction. Persistent pathological hypoxia is incompatible with normal biological functions, and as a result, multicellular organisms have been compelled to develop both organism-wide and cellular-level hypoxia solutions. Both direct, including oxidative phosphorylation down-regulation and inhibition of fatty-acid desaturation, and indirect processes, including altered hypoxia-sensitive transcription factor expression, facilitate the metabolic modifications that occur in response to hypoxia. Due to the dysfunctional vasculature associated with large areas of some cancers, sections of these tumors continue to develop in hypoxic environments. Crucial to drug development, a robust understanding of the significance of these metabolism changes will facilitate our understanding of cancer cell survival. This review defines our current knowledge base of several of the hypoxia-instigated modifications in cancer cell metabolism and exemplifies the correlation between metabolic change and its support of the hypoxic-adapted malignancy.
Collapse
Affiliation(s)
- Wafaa Al Tameemi
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Tina P. Dale
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Rakad M. Kh Al-Jumaily
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
- Department of Biology, College of Science, University of Baghdad, Baghdad, Iraq
| | - Nicholas R. Forsyth
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| |
Collapse
|