1
|
Dirandeh E, Ansari-Pirsaraei Z, Teimouri A, Dadashpour N, Thatcher WW. Effects of increasing the dietary contents of metabolizable energy and protein during the peripartum period on mammary gland development in Sistani goats. Sci Rep 2025; 15:14722. [PMID: 40289192 PMCID: PMC12034761 DOI: 10.1038/s41598-025-96795-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
The experiment was designed to consider effects of different amounts of metabolizable energy and protein on mammary gland development, mRNA abundance of genes related to vascular function and angiogenesis of Sistani goats. A total of 32 pregnant Sistani goats were selected on day 100 of gestation and allocated randomly to four treatments: (1) basal diet that meets metabolizable energy (ME) and protein (MP) requirements of goats during late gestation, as recommended by the National Research Council = NRC (2007) (C). (2) A diet with 10% higher levels of ME than recommendations (E). (3) A diet with 10% higher levels of MP than recommendations (P), and (4) a co-addition diet with 10% higher levels of both ME and MP than recommendations (EP). The four diets were fed from 100 days prepartum to 30 days after parturition. Mammary biopsies were obtained 24 h after parturition. Feed intake (g/day), colostrum (kg/day) and milk (kg/month) productions increased when extra E and P were provided together prepartum and early postpartum (P < 0.05). Relative mRNA abundances of vascular endothelial growth factor (VEGF, 4.68-fold), tyrosine kinase tie2 receptor (RTK, 4.61-fold), vascular endothelial growth factor receptor 1 (VEGFR1, 5.15-fold), angiopoietin I (ANGPT1, 4.86-fold), cyclooxygenase I (COX1, 4.5-fold), prostacyclin synthase (PTGIS, 5.66-fold), thromboxane A2 synthase (TBXAS, 98-fold) increased significantly in the mammary gland with the co-addition of E and P. Histological results documented that alveolar area (epithelium + luminal space, µm2), average number of epithelial cells per alveolus, and relative area occupied by overall epithelial cells (per slide) were increased significantly, when co-addition of E and P were fed in late gestation. In summary, increasing the dietary supply of ME and MP peripartum resulted in greater milk production, stimulated mammary gland alveolar development and affected the mRNA abundance of genes associated with both angiogenesis and milk secretion in Sistani goats.
Collapse
Affiliation(s)
- Essa Dirandeh
- Department of Animal Science, Sari Agricultural Sciences and Natural Resources University, P.O. BOX: 578, Sari, Mazandaran, Iran.
| | - Zarbakht Ansari-Pirsaraei
- Department of Animal Science, Sari Agricultural Sciences and Natural Resources University, P.O. BOX: 578, Sari, Mazandaran, Iran
| | - Asad Teimouri
- Department of Animal Science, Sari Agricultural Sciences and Natural Resources University, P.O. BOX: 578, Sari, Mazandaran, Iran
| | | | - William W Thatcher
- Department of Animal Science, University of Florida, P.O. BOX: 578, Florida, USA
| |
Collapse
|
2
|
Sheehan BJ, Edwards B, Medrano IS, El-Saidi MA, Zaidan WR, El-Ezzi AA, Kuddus RH. Association between two single nucleotide polymorphisms of the Prostaglandin-Endoperoxide Synthase 1 and 2 genes and cell proliferative prostatic diseases in Lebanon. Oncotarget 2025; 16:262-272. [PMID: 40184332 PMCID: PMC11970937 DOI: 10.18632/oncotarget.28710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/13/2025] [Indexed: 04/06/2025] Open
Abstract
The polymorphic genes PTGS1 and PTGS2 encode cyclooxygenases COX-1 and COX-2, respectively. Overexpression of these cyclooxygenases is linked to inflammation and neoplasms. This study investigated the potential association between the single nucleotide polymorphism (SNP) -842A>G (rs10306114) of the PTGS1 gene and SNP-765G>C (rs20417) of the PTGS2 gene with prostate cancer (PCa) and benign prostate hyperplasia (BPH). Blood leucocyte DNA from 56 healthy individuals, 61 individuals with PCa, and 51 individuals with BPH were genotyped using the PCR-RFLP method. Associations were inferred by calculating odds ratios (OR) and relative risks (RR) of genotype distributions and allele frequencies. The genotypes for both SNPs were in Hardy-Weinberg equilibrium for all groups. No significant association was observed between the A or G alleles or the AA, AG, or GG genotypes of the SNP-842A>G of the PTGS1 gene and prostatic diseases. However, the C allele of SNP-765G>C of the PTGS2 gene was significantly associated with an increased risk of BPH (OR = 2.30, p-value = 0.01). Differences in the ratios of GG/GC and GG/(GC+CC) genotypes also suggested a potential association between the C allele and PCa (p-value <0.1), and the combined affected (PCa+BPH) group (p-value <0.04). The small sample size and sampling from one ethnic group are limitations of this study.
Collapse
Affiliation(s)
- Brock J. Sheehan
- Department of Biology, Utah Valley University, Orem, UT 84058, USA
| | - Bryson Edwards
- Department of Biology, Utah Valley University, Orem, UT 84058, USA
| | | | - Mohammed A. El-Saidi
- Department of Strategic Management and Operations, Utah Valley University, Orem, UT 84058, USA
| | - Wissam R. Zaidan
- Radioimmunoassay Laboratory, Lebanese Atomic Energy Commission, Beirut, Lebanon
| | - Asmahan A. El-Ezzi
- Radioimmunoassay Laboratory, Lebanese Atomic Energy Commission, Beirut, Lebanon
- Department of Chemistry and Biochemistry, Lebanese University, Hadath, Lebanon
| | - Ruhul H. Kuddus
- Department of Biology, Utah Valley University, Orem, UT 84058, USA
| |
Collapse
|
3
|
Micha JP, Rettenmaier MA, Bohart RD, Goldstein BH. Does Aspirin Use Reduce the Risk for Ovarian Cancer? Cancer Invest 2024; 42:643-646. [PMID: 38965997 DOI: 10.1080/07357907.2024.2375573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Ovarian cancer is an aggressive malignancy and the leading cause of death among gynecologic cancers. Researchers have evaluated prophylactic medications that potentially avert the manifestation of ovarian cancer, but currently, there are no reliable screening measures for this disease. Nevertheless, the largest study involving aspirin use and ovarian cancer reported a substantive risk reduction from enduring aspirin use. Since there are countervailing data to impugn the potential benefits of aspirin use in staving off ovarian cancer, further research should scrutinize the use of this medication as a prophylactic intervention, especially in women who are at higher risk for developing the disease.
Collapse
Affiliation(s)
- John P Micha
- Gynecologic Oncology, Women's Cancer Research Foundation, Laguna Beach, California, USA
| | - Mark A Rettenmaier
- Gynecologic Oncology, Women's Cancer Research Foundation, Laguna Beach, California, USA
| | | | - Bram H Goldstein
- Gynecologic Oncology, Women's Cancer Research Foundation, Laguna Beach, California, USA
| |
Collapse
|
4
|
Razavirad A, Rismanchi S, Mortazavi P, Muhammadnejad A. Canine Mammary Tumors as a Potential Model for Human Breast Cancer in Comparative Oncology. Vet Med Int 2024; 2024:9319651. [PMID: 38766503 PMCID: PMC11101259 DOI: 10.1155/2024/9319651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/17/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024] Open
Abstract
Clinical and molecular similarities between canine mammary tumors (CMTs) and human breast cancer (HBC) propel scientists to further study their application in comparative oncology as a model for human breast cancer. In total, 64 canine mammary tumors were selected to study the most common markers, which are applicable for human breast cancer treatment, including estrogen and progesterone receptors (ER and PR), human epidermal growth factor (HER2/neu), Ki67, and cyclooxygenase 2 (Cox2). Immunohistochemistry (IHC) was used to assess the protein expression. The Veterinary Nottingham Prognostic Index (Vet-NPI) was also computed. Moreover, univariate and multivariable Cox proportional hazard analyses were applied to estimate hazard ratios (HRs). The results demonstrated that Ki67 was strongly expressed in the triple-negative tumors, and Ki67 protein expression continuously increased over the increase of Cox2 protein expression (p < 0.001). Further analysis revealed a significant difference among canine mammary subtypes and Vet-NPI, in which triple-negative tumors displayed the highest mean score compared to other subtypes (p < 0.001). In addition, the multivariable analysis revealed that the regional mastectomy procedure (adjusted HR = 2.78 (1.14-6.8)), the triple-negative tumors (adjusted HR = 48.08 (7.74-298.8)), strong Ki67 protein expression group (adjusted HR = 7.88 (2.02-30.68)), and strong Cox2 protein expression group (adjusted HR = 29.35 (5.18-166.4)) demonstrated significantly lower disease-free survival rates compared to other corresponding groups. Overall, canine mammary tumors showed strong similarities to human breast cancer in terms of clinical and molecular aspects; therefore, they could be suggested as a model for human breast cancer in comparative oncology.
Collapse
Affiliation(s)
- Amirhossein Razavirad
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanaz Rismanchi
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Pejman Mortazavi
- Department of Pathobiology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ahad Muhammadnejad
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Moisand A, Madéry M, Boyer T, Domblides C, Blaye C, Larmonier N. Hormone Receptor Signaling and Breast Cancer Resistance to Anti-Tumor Immunity. Int J Mol Sci 2023; 24:15048. [PMID: 37894728 PMCID: PMC10606577 DOI: 10.3390/ijms242015048] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Breast cancers regroup many heterogeneous diseases unevenly responding to currently available therapies. Approximately 70-80% of breast cancers express hormone (estrogen or progesterone) receptors. Patients with these hormone-dependent breast malignancies benefit from therapies targeting endocrine pathways. Nevertheless, metastatic disease remains a major challenge despite available treatments, and relapses frequently ensue. By improving patient survival and quality of life, cancer immunotherapies have sparked considerable enthusiasm and hope in the last decade but have led to only limited success in breast cancers. In addition, only patients with hormone-independent breast cancers seem to benefit from these immune-based approaches. The present review examines and discusses the current literature related to the role of hormone receptor signaling (specifically, an estrogen receptor) and the impact of its modulation on the sensitivity of breast cancer cells to the effector mechanisms of anti-tumor immune responses and on the capability of breast cancers to escape from protective anti-cancer immunity. Future research prospects related to the possibility of promoting the efficacy of immune-based interventions using hormone therapy agents are considered.
Collapse
Affiliation(s)
- Alexandra Moisand
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 33076 Bordeaux, France; (A.M.); (M.M.); (T.B.); (C.D.)
- Cancer Biology Graduate Program, UB Grad 2.0, University of Bordeaux, 33076 Bordeaux, France
| | - Mathilde Madéry
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 33076 Bordeaux, France; (A.M.); (M.M.); (T.B.); (C.D.)
- Cancer Biology Graduate Program, UB Grad 2.0, University of Bordeaux, 33076 Bordeaux, France
| | - Thomas Boyer
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 33076 Bordeaux, France; (A.M.); (M.M.); (T.B.); (C.D.)
- Cancer Biology Graduate Program, UB Grad 2.0, University of Bordeaux, 33076 Bordeaux, France
| | - Charlotte Domblides
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 33076 Bordeaux, France; (A.M.); (M.M.); (T.B.); (C.D.)
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
| | - Céline Blaye
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 33076 Bordeaux, France; (A.M.); (M.M.); (T.B.); (C.D.)
| | - Nicolas Larmonier
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 33076 Bordeaux, France; (A.M.); (M.M.); (T.B.); (C.D.)
- Cancer Biology Graduate Program, UB Grad 2.0, University of Bordeaux, 33076 Bordeaux, France
| |
Collapse
|
6
|
Lee H, Hwang M, Jang S, Um SW. Immune Regulatory Function of Cancer- Associated Fibroblasts in Non-small Cell Lung Cancer. Tuberc Respir Dis (Seoul) 2023; 86:304-318. [PMID: 37345462 PMCID: PMC10555526 DOI: 10.4046/trd.2022.0129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 02/25/2023] [Accepted: 06/18/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are key components of the tumor microenvironment and significantly contribute to immune evasion. We investigated the effects of CAFs on the immune function of CD4+ and CD8+ T cells in non-small cell lung cancer (NSCLC). METHODS We isolated CAFs and normal fibroblasts (NFs) from tumors and normal lung tissues of NSCLC patients, respectively. CAFs were co-cultured with activated T cells to evaluate their immune regulatory function. We investigated the effect of CAF conditioned medium (CAF-CM) on the cytotoxicity of T cells. CAFs were also co-cultured with activated peripheral blood mononuclear cells and further incubated with cyclooxygenase- 2 (COX2) inhibitors to investigate the potential role of COX2 in immune evasion. RESULTS CAFs and NFs were isolated from the lung tissues (n=8) and lymph nodes (n=3) of NSCLC patients. Immune suppressive markers, such as COX2 and programmed death-ligand 1 (PD-L1), were increased in CAFs after co-culture with activated T cells. Interestingly, CAFs promoted the expression of programmed death-1 in CD4+ and CD8+ T cells, and strongly inhibited T cell proliferation in allogenic and autologous pairs of CAFs and T cells. CAF-CM decreased the cytotoxicity of T cells. COX2 inhibitors partially restored the proliferation of CD4+ and CD8+ T cells, and downregulated the expression of COX2, prostaglandin E synthase, prostaglandin E2, and PD-L1 in CAFs. CONCLUSION CAFs promote immune evasion by suppressing the function of CD4+ and CD8+ T cells via their effects on COX2 and PD-L1 in NSCLC. The immunosuppressive function of CAFs could be alleviated by COX2 inhibitors.
Collapse
Affiliation(s)
- Hyewon Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Mina Hwang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seonae Jang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang-Won Um
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
8
|
Kwon H, An J, Kim JS, Kang IC. Inhibitory effects of NSAID-conjugated SN-38 on the viability of A549 Non-small cell lung cancer cells. Biochem Biophys Rep 2023; 35:101517. [PMID: 37521373 PMCID: PMC10374863 DOI: 10.1016/j.bbrep.2023.101517] [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: 05/23/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023] Open
Abstract
The goal of this paper was to look into the anti-tumor mechanism of Non-Steroidal Anti-Inflammatory Drug (NSAID)-conjugated SN-38 Prodrug in A549 lung cancer cells. We found that Indomethacine-SN-38 (IndoSN-38) and Naproxen-SN-38(NaproSN-38) as a theranostic prodrug targeting cyclooxygenase-2(COX-2) in cancer cells inhibited A549 cell viability in a dose-dependent fashion. IndoSN-38 and NaproSN-38 inhibited A549 cell viability in a dose-dependent fashion. The suppression of A549 cell viability was due to induction of the cell apoptosis by enhancing the activities of Caspase 3 and Caspase 8. The cell cycle arrest of sub-G1 was found in the cells treated with IndoSN-38 or NaproSN-38. Collectively, these data suggested that the anti-proliferative activities of the NSAID-conjugated SN-38 prodrugs were due to promotion of cell death and arresting the cell cycle which was similar with those of SN-38.
Collapse
Affiliation(s)
- Hae–Won Kwon
- Department of Bioconvergence System, Graduate School, and BioChip Research Center, Hoseo University, Asan, 336-795, South Korea
| | - Jusung An
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - In-Cheol Kang
- Department of Bioconvergence System, Graduate School, and BioChip Research Center, Hoseo University, Asan, 336-795, South Korea
- InnoPharmaScreen Inc, Incheon, 21988, South Korea
| |
Collapse
|
9
|
Abbasi N, Najafi R. Cupping Therapy as an Anti-inflammation Therapy and Immunomodulator in Cancer Patients. J Gastrointest Cancer 2023; 54:3-5. [PMID: 34596827 DOI: 10.1007/s12029-021-00701-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Nooshin Abbasi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Avenue, Hamadan, Iran
- Department of Neurosciences-DNS, University of Padua, Padua, Italy
| | - Rezvan Najafi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Avenue, Hamadan, Iran.
| |
Collapse
|
10
|
Advances in Molecular Regulation of Prostate Cancer Cells by Top Natural Products of Malaysia. Curr Issues Mol Biol 2023; 45:1536-1567. [PMID: 36826044 PMCID: PMC9954984 DOI: 10.3390/cimb45020099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Prostate cancer (PCa) remains both a global health burden and a scientific challenge. We present a review of the molecular targets driving current drug discovery to fight this disease. Moreover, the preventable nature of most PCa cases represents an opportunity for phytochemicals as chemopreventive when adequately integrated into nutritional interventions. With a renovated interest in natural remedies as a commodity and their essential role in cancer drug discovery, Malaysia is looking towards capitalizing on its mega biodiversity, which includes the oldest rainforest in the world and an estimated 1200 medicinal plants. We here explore whether the list of top Malay plants prioritized by the Malaysian government may fulfill the potential of becoming newer, sustainable sources of prostate cancer chemotherapy. These include Andrographis paniculate, Centella asiatica, Clinacanthus nutans, Eurycoma longifolia, Ficus deltoidea, Hibiscus sabdariffa, Marantodes pumilum (syn. Labisia pumila), Morinda citrifolia, Orthosiphon aristatus, and Phyllanthus niruri. Our review highlights the importance of resistance factors such as Smac/DIABLO in cancer progression, the role of the CXCL12/CXCR4 axis in cancer metastasis, and the regulation of PCa cells by some promising terpenes (andrographolide, Asiatic acid, rosmarinic acid), flavonoids (isovitexin, gossypin, sinensetin), and alkylresorcinols (labisiaquinones) among others.
Collapse
|
11
|
Lee JK, Choi WS, Song JY, Kwon OS, Lee YJ, Lee JS, Lee S, Choi SR, Lee CH, Lee JY. Anti-inflammatory effects of Athyrium yokoscense extract via inhibition of the Erk1/2 and NF-κB pathways in bisphenol A-stimulated A549 cells. Toxicol Res 2023; 39:135-146. [PMID: 36726827 PMCID: PMC9839918 DOI: 10.1007/s43188-022-00154-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/09/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
Bisphenol A is an environmental endocrine disruptor that has similar functions to estrogen in humans. However, few studies have investigated pulmonary inflammation induced by BPA, and the effect of Athyrium yokoscense extract on this inflammatory response is unknown. In this study, we investigated this effect in A549 human alveolar epithelial cells. BPA at concentrations higher than 100 µM were cytotoxic to A549 cells at 24 and 48 h after treatment; however, AYE (100 µg/mL) had a protective effect against BPA-induced cytotoxicity. AYE also inhibited the generation of intracellular reactive oxygen species, expressions of cyclooxygenase-2 and extracellular signal-regulated kinase1/2 proteins, activities of phospholipase A2, COX-2, nuclear factor kappa-light-chain-enhancer of activated B cells, and proinflammatory mediators including prostaglandin E2, tumor necrosis factor-α, and interleukin-6 induced by BPA in A549 cells. This study demonstrated that BPA, which induces chronic lung disease, causes oxidative stress and inflammatory response in lung epithelial cell line, and found that AYE reduces BPA-induced oxidative stress and inflammatory response by down-regulating the Erk1/2 and NF-κB pathways.
Collapse
Affiliation(s)
- Jung-Kyu Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Won Seok Choi
- College of Pharmacy, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Jin Yong Song
- College of Pharmacy, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Oh Seong Kwon
- College of Pharmacy, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Yeon Jin Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974 Republic of Korea
| | - Jong Seok Lee
- National Institute of Biological Resources, Incheon, 22689 Republic of Korea
| | - Sarah Lee
- National Institute of Biological Resources, Incheon, 22689 Republic of Korea
| | - Se Rin Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029 Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029 Republic of Korea
| | - Ji-Yun Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974 Republic of Korea
| |
Collapse
|
12
|
DNA Polymerase Theta Plays a Critical Role in Pancreatic Cancer Development and Metastasis. Cancers (Basel) 2022; 14:cancers14174077. [PMID: 36077614 PMCID: PMC9454495 DOI: 10.3390/cancers14174077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), due to its genomic heterogeneity and lack of effective treatment, despite decades of intensive research, will become the second leading cause of cancer-related deaths by 2030. Step-wise acquisition of mutations, due to genomic instability, is considered to drive the development of PDAC; the KRAS mutation occurs in 95 to 100% of human PDAC, and is already detectable in early premalignant lesions designated as pancreatic intraepithelial neoplasia (PanIN). This mutation is possibly the key event leading to genomic instability and PDAC development. Our study aimed to investigate the role of the error-prone DNA double-strand breaks (DSBs) repair pathway, alt-EJ, in the presence of the KRAS G12D mutation in pancreatic cancer development. Our findings show that oncogenic KRAS contributes to increasing the expression of Polθ, Lig3, and Mre11, key components of alt-EJ in both mouse and human PDAC models. We further confirm increased catalytic activity of alt-EJ in a mouse and human model of PDAC bearing the KRAS G12D mutation. Subsequently, we focused on estimating the impact of alt-EJ inactivation by polymerase theta (Polθ) deletion on pancreatic cancer development, and survival in genetically engineered mouse models (GEMMs) and cancer patients. Here, we show that even though Polθ deficiency does not fully prevent the development of pancreatic cancer, it significantly delays the onset of PanIN formation, prolongs the overall survival of experimental mice, and correlates with the overall survival of pancreatic cancer patients in the TCGA database. Our study clearly demonstrates the role of alt-EJ in the development of PDAC, and alt-EJ may be an attractive therapeutic target for pancreatic cancer patients.
Collapse
|
13
|
Kaur J, Bhardwaj A, Wuest F. Fluorine-18 Labelled Radioligands for PET Imaging of Cyclooxygenase-2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123722. [PMID: 35744851 PMCID: PMC9227202 DOI: 10.3390/molecules27123722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022]
Abstract
Molecular imaging probes enable the early and accurate detection of disease-specific biomarkers and facilitate personalized treatment of many chronic diseases, including cancer. Among current clinically used functional imaging modalities, positron emission tomography (PET) plays a significant role in cancer detection and in monitoring the response to therapeutic interventions. Several preclinical and clinical studies have demonstrated the crucial involvement of cyclooxygenase-2 (COX-2) isozyme in cancer development and progression, making COX-2 a promising cancer biomarker. A variety of COX-2-targeting PET radioligands has been developed based on anti-inflammatory drugs and selective COX-2 inhibitors. However, many of those suffer from non-specific binding and insufficient metabolic stability. This article highlights examples of COX-2-targeting PET radioligands labelled with the short-lived positron emitter 18F, including radiosynthesis and PET imaging studies published in the last decade (2012–2021).
Collapse
Affiliation(s)
- Jatinder Kaur
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
- Correspondence: (J.K.); (F.W.)
| | - Atul Bhardwaj
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Correspondence: (J.K.); (F.W.)
| |
Collapse
|
14
|
Fluorochrome Selection for Imaging Intraoperative Ovarian Cancer Probes. Pharmaceuticals (Basel) 2022; 15:ph15060668. [PMID: 35745587 PMCID: PMC9230671 DOI: 10.3390/ph15060668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
The identification and removal of all gross and microscopic tumor to render the patient disease free represents a huge challenge in ovarian cancer treatment. The presence of residual disease is an independent negative prognostic factor. Herein, we describe the synthesis and the “in vitro” evaluation of compounds as cyclooxygenase (COX)-1 inhibitors, the COX-1 isoform being an ovarian cancer biomarker, each bearing fluorochromes with different fluorescence features. Two of these compounds N-[4-(9-dimethylimino-9H-benzo[a]phenoxazin-5-ylamino) butyl]-2-(3,4-bis(4-methoxyphenyl)isoxazol-5-yl)acetamide chloride (RR11) and 3-(6-(4-(2-(3,4-bis(4-methoxyphenyl)isoxazole-5-yl)acetamido)butyl)amino-6-oxohexyl)-2-[7-(1,3-dihydro-1,1-dimethyl-3-ethyl 2H-benz[e]indolin-2-yl-idene)-1,3,5-heptatrienyl]-1,1-dimethyl-3-(6-carboxilato-hexyl)-1H-benz[e]indolium chloride, 23 (MSA14) were found to be potent and selective inhibitors of cyclooxygenase (COX)-1 “in vitro”, and thus were further investigated “in vivo”. The IC50 values were 0.032 and 0.087 µM for RR11 and 23 (MSA 14), respectively, whereas the COX-2 IC50 for RR11 is 2.4 µM while 23 (MSA14) did not inhibit COX-2 even at a 50 µM concentration. Together, this represented selectivity index = 75 and 874, respectively. Structure-based virtual screening (SBVS) performed with the Fingerprints for Ligands and Proteins (FLAP) software allowed both to differentiate highly active compounds from less active and inactive structures and to define their interactions inside the substrate-binding cavity of hCOX1. Fluorescent probes RR11 and 23 (MSA14), were used for preliminary near-infrared (NIR) fluorescent imaging (FLI) in human ovarian cancer (OVCAR-3 and SKOV-3) xenograft models. Surprisingly, a tumor-specific signal was observed for both tested fluorescent probes, even though this signal is not linked to the presence of COX-1.
Collapse
|
15
|
PTGES3 is a Putative Prognostic Marker in Breast Cancer. J Surg Res 2022; 271:154-162. [DOI: 10.1016/j.jss.2021.08.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/30/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022]
|
16
|
Gallyas F, Ramadan FHJ, Andreidesz K, Hocsak E, Szabo A, Tapodi A, Kiss GN, Fekete K, Bognar R, Szanto A, Bognar Z. Involvement of Mitochondrial Mechanisms and Cyclooxygenase-2 Activation in the Effect of Desethylamiodarone on 4T1 Triple-Negative Breast Cancer Line. Int J Mol Sci 2022; 23:ijms23031544. [PMID: 35163464 PMCID: PMC8836269 DOI: 10.3390/ijms23031544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 12/10/2022] Open
Abstract
Novel compounds significantly interfering with the mitochondrial energy production may have therapeutic value in triple-negative breast cancer (TNBC). This criterion is clearly fulfilled by desethylamiodarone (DEA), which is a major metabolite of amiodarone, a widely used antiarrhythmic drug, since the DEA previously demonstrated anti-neoplastic, anti-metastasizing, and direct mitochondrial effects in B16F10 melanoma cells. Additionally, the more than fifty years of clinical experience with amiodarone should answer most of the safety concerns about DEA. Accordingly, in the present study, we investigated DEA’s potential in TNBC by using a TN and a hormone receptor positive (HR+) BC cell line. DEA reduced the viability, colony formation, and invasive growth of the 4T1 cell line and led to a higher extent of the MCF-7 cell line. It lowered mitochondrial transmembrane potential and induced mitochondrial fragmentation. On the other hand, DEA failed to significantly affect various parameters of the cellular energy metabolism as determined by a Seahorse live cell respirometer. Cyclooxygenase 2 (COX-2), which was upregulated by DEA in the TNBC cell line only, accounted for most of 4T1’s DEA resistance, which was counteracted by the selective COX-2 inhibitor celecoxib. All these data indicate that DEA may have potentiality in the therapy of TNBC.
Collapse
Affiliation(s)
- Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
- LERN-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
| | - Fadi H. J. Ramadan
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Kitti Andreidesz
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Eniko Hocsak
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Aliz Szabo
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Antal Tapodi
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Gyongyi N. Kiss
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Rita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Arpad Szanto
- Urology Clinic, UP Medical Center, University of Pecs Medical School, 7624 Pecs, Hungary;
| | - Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
- Correspondence: ; Tel.: +36-72-536-276
| |
Collapse
|
17
|
Stransky N, Ruth P, Schwab M, Löffler MW. Can Any Drug Be Repurposed for Cancer Treatment? A Systematic Assessment of the Scientific Literature. Cancers (Basel) 2021; 13:6236. [PMID: 34944859 PMCID: PMC8699650 DOI: 10.3390/cancers13246236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022] Open
Abstract
Drug repurposing is a complementary pathway for introducing new drugs against cancer. Broad systematic assessments of ongoing repurposing efforts in oncology are lacking, but may be helpful to critically appraise current and future efforts. Hence, we conducted a systematic PubMed search encompassing 100 frequently prescribed and 100 randomly selected drugs, and assessed the published preclinical anti-cancer effects. Furthermore, we evaluated all the identified original articles for methodological quality. We found reports indicating anti-cancer effects for 138/200 drugs, especially among frequently prescribed drugs (81/100). Most were reports suggesting single-agent activity of the drugs (61%). Basic information, such as the cell line used or control treatments utilized, were reported consistently, while more detailed information (e.g., excluded data) was mostly missing. The majority (56%) of in vivo studies reported randomizing animals, while only few articles stated that the experiments were conducted in a blinded fashion. In conclusion, we found promising reports of anti-cancer effects for the majority of the assessed drugs, but speculate that many of them are false-positive findings. Reward systems should be adjusted to encourage the widespread usage of high reporting quality and bias-reducing methodologies, aiming to decrease the rate of false-positive results, and thereby increasing the trust in the findings.
Collapse
Affiliation(s)
- Nicolai Stransky
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany;
- Department of Radiation Oncology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany;
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, Germany;
- Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Faculty of Medicine, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Markus W. Löffler
- Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Faculty of Medicine, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, 72076 Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| |
Collapse
|
18
|
Tsai MM, Lin HC, Yu MC, Lin WJ, Chu MY, Tsai CC, Cheng CY. Anticancer Effects of Helminthostachys zeylanica Ethyl acetate Extracts on Human Gastric Cancer Cells through Downregulation of the TNF-α-activated COX-2-cPLA2-PGE 2 Pathway. J Cancer 2021; 12:7052-7068. [PMID: 34729107 PMCID: PMC8558661 DOI: 10.7150/jca.64638] [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: 07/06/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Gastric cancer (GC) is the second most prevalent cancer worldwide and the eighth most common cause of tumor-related death in Taiwan. Helminthostachys zeylanica, a flavonoid compound, has anti-inflammatory, immunomodulatory, and anticancer effects. We examined whether an extract of H. zeylanica (E1 and E2) has potential as a treatment for GC. Methods: We investigated the effects (pro-apoptosis, pro-autophagy, and antiproliferation ability) of H. zeylanica-E2 on cell viability in healthy gastric epithelial (GES-1) and GC cells (AGS and BGC823). H. zeylanica-E2 was toxic to GC cells but had little or no toxicity to normal cells. Results: In this study, H. zeylanica-E2 induced apoptosis through caspase 3/7, Bcl-2, Bax, cyclooxygenase-2 (COX-2), and cleaved poly (ADP-ribose) polymerase pathways in GC cells. In addition, it increased autophagy by stimulating autophagy-related protein (ATG)5, ATG7, LC3-I/LC3-II, and inhibiting COX-2 activity in GC cells. We also found that H. zeylanica-E2 exhibited antiproliferation ability through cell cycle arrest in G0/G1 and G2/M and suppressed the migration of GC cells. The anticancer effects of H. zeylanica-E2 in GC cells might be mediated partly through inhibition of tumor necrosis factor-α (TNF-α)-activated proinflammatory cytosolic phospholipase A2 (cPLA2)-COX-2-prostaglandin E2 (PGE2) pathway. Conclusions: Our results suggest that H. zeylanica-E2 has potential as a novel adjunctive agent for the treatment of GC.
Collapse
Affiliation(s)
- Ming-Ming Tsai
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan and Department of General Surgery, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Horng-Chyuan Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Chin Yu
- Department of Surgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wan-Jung Lin
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Mei-Yi Chu
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Ching-Ching Tsai
- Department of Nursing, College of Nursing, Chang Gung University of Science and Technology, and Department of Cardiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ching-Yi Cheng
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Pulmonary Infection and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
19
|
Interactions between exposure to polycyclic aromatic hydrocarbons and xenobiotic metabolism genes, and risk of breast cancer. Breast Cancer 2021; 29:38-49. [PMID: 34351578 DOI: 10.1007/s12282-021-01279-0] [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: 08/13/2020] [Accepted: 07/25/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Polycyclic aromatic hydrocarbons (PAHs) are a group of environmental pollutants associated with multiple cancers, including female breast cancer. Several xenobiotic metabolism genes (XMGs), including the CYP450 family, play an important role in activating and detoxifying PAHs, and variations in the activity of the enzymes they encode can impact this process. This study aims to examine the association between XMGs and breast cancer, and to assess whether these variants modify the effects of PAH exposure on breast cancer risk. METHODS In a case-control study in Vancouver, British Columbia, and Kingston, Ontario, 1037 breast cancer cases and 1046 controls had DNA extracted from blood or saliva and genotyped for 138 single nucleotide polymorphisms (SNPs) and tagSNPs in 27 candidate XMGs. Occupational PAH exposure was assessed using a measurement-based job-exposure matrix. RESULTS An association between genetic variants and breast cancer was observed among six XMGs, including increased risk among the minor allele carriers of AKR1C3 variant rs12387 (OR 2.71, 95% CI 1.42-5.19) and AKR1C4 variant rs381267 (OR 2.50, 95% CI 1.23-5.07). Heterogeneous effects of occupational PAH exposure were observed among carriers of AKR1C3/4 variants, as well as the PTGS2 variant rs5275. CONCLUSION Our findings support an association between SNPs of XMGs and female breast cancer, including novel genetic variants that modify the toxicity of PAH exposure. These results highlight the interplay between genetic and environmental factors, which can be helpful in understanding the modifiable risks of breast cancer and its complex etiology.
Collapse
|
20
|
Ertürk Ö, Değirmenci A, Yurdakul Ertürk E, Atlı Şekeroğlu Z, Çol Ayvaz M, Kontaş Yedier S. Antimicrobial, antioxidant and cytotoxic activities of some analgesic or anti‐inflammatory drugs. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00755-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
21
|
Wang Y, Cui L, Georgiev P, Singh L, Zheng Y, Yu Y, Grein J, Zhang C, Muise ES, Sloman DL, Ferguson H, Yu H, Pierre CS, Dakle PJ, Pucci V, Baker J, Loboda A, Linn D, Brynczka C, Wilson D, Haines BB, Long B, Wnek R, Sadekova S, Rosenzweig M, Haidle A, Han Y, Ranganath SH. Combination of EP 4 antagonist MF-766 and anti-PD-1 promotes anti-tumor efficacy by modulating both lymphocytes and myeloid cells. Oncoimmunology 2021; 10:1896643. [PMID: 33796403 PMCID: PMC7993229 DOI: 10.1080/2162402x.2021.1896643] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Prostaglandin E2 (PGE2), an arachidonic acid pathway metabolite produced by cyclooxygenase (COX)-1/2, has been shown to impair anti-tumor immunity through engagement with one or more E-type prostanoid receptors (EP1-4). Specific targeting of EP receptors, as opposed to COX-1/2 inhibition, has been proposed to achieve preferential antagonism of PGE2–mediated immune suppression. Here we describe the anti-tumor activity of MF-766, a potent and highly selective small-molecule inhibitor of the EP4 receptor. EP4 inhibition by MF-766 synergistically improved the efficacy of anti-programmed cell death protein 1 (PD-1) therapy in CT26 and EMT6 syngeneic tumor mouse models. Multiparameter flow cytometry analysis revealed that treatment with MF-766 promoted the infiltration of CD8+ T cells, natural killer (NK) cells and conventional dendritic cells (cDCs), induced M1-like macrophage reprogramming, and reduced granulocytic myeloid-derived suppressor cells (MDSC) in the tumor microenvironment (TME). In vitro experiments demonstrated that MF-766 restored PGE2-mediated inhibition of lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α production in THP-1 cells and human blood, and PGE2-mediated inhibition of interleukin (IL)-2-induced interferon (IFN)-γ production in human NK cells. MF-766 reversed the inhibition of IFN-γ in CD8+ T-cells by PGE2 and impaired suppression of CD8+ T-cells induced by myeloid-derived suppressor cells (MDSC)/PGE2. In translational studies using primary human tumors, MF-766 enhanced anti-CD3-stimulated IFN-γ, IL-2, and TNF-α production in primary histoculture and synergized with pembrolizumab in a PGE2 high TME. Our studies demonstrate that the combination of EP4 blockade with anti-PD-1 therapy enhances antitumor activity by differentially modulating myeloid cell, NK cell, cDC and T-cell infiltration profiles.
Collapse
Affiliation(s)
- Yun Wang
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Long Cui
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Peter Georgiev
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Latika Singh
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Yanyan Zheng
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Ying Yu
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Jeff Grein
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Chunsheng Zhang
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Eric S Muise
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, USA
| | - David L Sloman
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Heidi Ferguson
- Department of Pharmaceutical Science, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Hongshi Yu
- Department of Pharmaceutical Science, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Cristina St Pierre
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Pranal J Dakle
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Vincenzo Pucci
- Department of Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts, USA
| | - James Baker
- Department of Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Andrey Loboda
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Doug Linn
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Christopher Brynczka
- Dept. Safety and Exploratory Pharmacology, Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Doug Wilson
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Brian B Haines
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Brian Long
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Richard Wnek
- Department of Translational Biomarkers, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Svetlana Sadekova
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Michael Rosenzweig
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Andrew Haidle
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Yongxin Han
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, USA
| | - Sheila H Ranganath
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA
| |
Collapse
|
22
|
Picado C, Roca-Ferrer J. Role of the Cyclooxygenase Pathway in the Association of Obstructive Sleep Apnea and Cancer. J Clin Med 2020; 9:E3237. [PMID: 33050416 PMCID: PMC7601393 DOI: 10.3390/jcm9103237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022] Open
Abstract
The objective of this review is to examine the findings that link obstructive sleep apnea (OSA) with cancer and the role played by the cyclooxygenase (COX) pathway in this association. Epidemiological studies in humans suggest a link between OSA and increased cancer incidence and mortality. Studies carried out in animal models have shown that intermittent hypoxia (IH) induces changes in several signaling pathways involved in the regulation of host immunological surveillance that results in tumor establishment and invasion. IH induces the expression of cyclooxygenase 2 (COX-2) that results in an increased synthesis of prostaglandin E2 (PGE2). PGE2 modulates the function of multiple cells involved in immune responses including T lymphocytes, NK cells, dendritic cells, macrophages, and myeloid-derived suppressor cells. In a mouse model blockage of COX-2/PGE2 abrogated the pro-oncogenic effects of IH. Despite the fact that aspirin inhibits PGE2 production and prevents the development of cancer, none of the epidemiological studies that investigated the association of OSA and cancer included aspirin use in the analysis. Studies are needed to investigate the regulation of the COX-2/PGE2 pathway and PGE2 production in patients with OSA, to better define the role of this axis in the physiopathology of OSA and the potential role of aspirin in preventing the development of cancer.
Collapse
Affiliation(s)
- César Picado
- Hospital Clinic, Department of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto Carlos III, 28029 Madrid, Spain
| | - Jordi Roca-Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
23
|
Bitis arietans Snake Venom Induces an Inflammatory Response Which Is Partially Dependent on Lipid Mediators. Toxins (Basel) 2020; 12:toxins12090594. [PMID: 32937985 PMCID: PMC7551280 DOI: 10.3390/toxins12090594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 01/04/2023] Open
Abstract
Bitis arietans is a snake of medical importance, as it is responsible for more accidents in humans and domestic animals than all other African snakes put together. The accidents are characterized by local and systemic alterations, such as inflammation, cardiovascular and hemostatic disturbances, which can lead victims to death or permanent disability. However, little is known about the envenomation mechanism, especially regarding the inflammatory response, which is related to severe clinical conditions triggered by the venom. Therefore, the aim of the present study was to evaluate the inflammatory response related to the B. arietans envenomation using a peritonitis mice model. By pharmacological interventions and use of mice genetically deficient of the 5-lipoxygenase enzyme (5-LO-/-) or platelet-activating factor (PAF) receptor (PAFR-/- the participation of eicosanoids and PAF in this response was also investigated. The obtained results demonstrated that the venom induces an in vivo inflammatory response, characterized by an early increased vascular permeability, followed by an accumulation of polymorphonuclear (PMN) cells in the peritoneal cavity, accompanied by the production of the eicosanoids LTB4, LTC4, TXB2 and PGE2, as well as the local and systemic production of IL-6 and MCP-1. These inflammatory events were attenuated by the pre-treatment with anti-inflammatory drugs that interfere in lipid mediators' functions. However, 5-LO-/- mice did not show a reduction of inflammatory response induced by the venom, while PAFR-/- mice showed a reduction in both the PMN leukocytes number and the local and systemic production of IL-6 and MCP-1. This study demonstrated that the Bitis arietans venom contains toxins that trigger an inflammatory process, which is partially dependent on lipid mediators, and may contribute to the envenomation pathology.
Collapse
|
24
|
Majerník M, Jendželovský R, Fedoročko P. Potentiality, Limitations, and Consequences of Different Experimental Models to Improve Photodynamic Therapy for Cancer Treatment in Relation to Antiangiogenic Mechanism. Cancers (Basel) 2020; 12:cancers12082118. [PMID: 32751731 PMCID: PMC7463805 DOI: 10.3390/cancers12082118] [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: 07/09/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
The relevance of experimentally gained information represents a long-term debating issue in the field of molecular biology research. The loss of original conditions in the in vitro environment affects various biological mechanisms and cellular interactions. Consequently, some biochemical mechanisms are lost or critically altered. Analyses in these modified conditions could, therefore, distort the relevancy of experimentally gained information. In some cases, the similarities with original conditions are so small that utilization of simpler in vitro models seems impossible, or could occur in a very limited way. To conclude, the study of more complex phenomena places higher demands on the complexity of the experimental model. The latest information highlights the fact that the tumor angiogenesis mechanism has very complex features. This complexity can be associated with a wide range of angiogenic factors expressed by a variety of malignant and non-malignant cells. Our article summarizes the results from various experimental models that were utilized to analyze a photodynamic therapy effect on tumor angiogenic mechanisms. Additionally, based on the latest information, we present the most important attributes and limitations of utilized experimental models. We also evaluate the essential problems associated with angiogenic mechanism induction after photodynamic therapy application.
Collapse
|
25
|
Chen YC, Chan CH, Lim YB, Yang SF, Yeh LT, Wang YH, Chou MC, Yeh CB. Risk of Breast Cancer in Women with Mastitis: A Retrospective Population-Based Cohort Study. ACTA ACUST UNITED AC 2020; 56:medicina56080372. [PMID: 32722165 PMCID: PMC7466309 DOI: 10.3390/medicina56080372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/16/2022]
Abstract
Background and objectives: Breast cancer is a common cancer in women and has been the fourth leading cause of death in Taiwanese women. Risk factors for breast cancer include family history of breast cancer, genetic factors, and not breastfeeding. Several studies have reported an association between repeated inflammation at a young age, especially among lactating women, and cancer; however, the number of studies about the association of mastitis and breast cancer in nonlactating women is still limited. Therefore, the aim of this study was to determine the relationship between mastitis in women aged ≥40 years and breast cancer. Materials and Methods: This was a retrospective cohort study design. The data source was the Longitudinal Health Insurance Database 2010 (LHID 2010), comprising data collected by Taiwan’s National Health Insurance program. Cases of newly diagnosed mastitis in women aged ≥40 years (ICD-9-CM code = 611.0) were selected from the years 2010 to 2012. Women not diagnosed with mastitis were selected as the control group, and their data for the years 2009 to 2013 were obtained through the database. In addition, the non-mastitis group was matched 1:10 by age. Results: A total of 8634 participants were selected from the LHID 2010, which included 734 cases with mastitis and 7900 cases without mastitis. After adjustment for age, hypertension, hyperlipidemia, diabetes, hypothyroidism, and autoimmune diseases, the Cox proportional hazard model showed that patients with mastitis had a higher risk of breast cancer (aHR = 3.71, 95% CI = 1.9–7.02) compared with the non-mastitis group. The Kaplan–Meier curve also showed that women with mastitis had a higher risk of developing breast cancer. Conclusions: This study confirmed that women with mastitis have a higher risk of developing breast cancer. Therefore, women aged ≥40 years could reduce breast cancer risk by taking precautions to prevent mammary gland infection and mastitis.
Collapse
Affiliation(s)
- Ying-Cheng Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-C.C.); (Y.-B.L.); (S.-F.Y.); (L.-T.Y.)
- Department of Surgery, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Chi-Ho Chan
- Department of Microbiology and Immunology, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
| | - Yu-Bing Lim
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-C.C.); (Y.-B.L.); (S.-F.Y.); (L.-T.Y.)
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-C.C.); (Y.-B.L.); (S.-F.Y.); (L.-T.Y.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
| | - Liang-Tsai Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-C.C.); (Y.-B.L.); (S.-F.Y.); (L.-T.Y.)
- Department of Anesthesiology, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-C.C.); (Y.-B.L.); (S.-F.Y.); (L.-T.Y.)
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (M.-C.C.); (C.-B.Y.)
| | - Chao-Bin Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-C.C.); (Y.-B.L.); (S.-F.Y.); (L.-T.Y.)
- Department of Emergency Medicine, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (M.-C.C.); (C.-B.Y.)
| |
Collapse
|
26
|
Xie C, Ge M, Jin J, Xu H, Mao L, Geng S, Wu J, Zhu J, Li X, Zhong C. Mechanism investigation on Bisphenol S-induced oxidative stress and inflammation in murine RAW264.7 cells: The role of NLRP3 inflammasome, TLR4, Nrf2 and MAPK. JOURNAL OF HAZARDOUS MATERIALS 2020; 394:122549. [PMID: 32283380 DOI: 10.1016/j.jhazmat.2020.122549] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/20/2020] [Accepted: 03/15/2020] [Indexed: 06/11/2023]
Abstract
Bisphenol S is considered as a safer alternative to bisphenol A. In the present study, we used murine macrophages to investigate the effects of BPS exposure on oxidative stress and inflammatory response as well as the underlying mechanism. Cells were exposed to BPS at various concentrations for short period of times. Results showed that 10-8 M BPS triggered oxidative stress by increasing ROS/RNS production, increased the levels of oxidant enzyme NOX1/2, and decreased the levels of antioxidant enzymes SOD1/2, CAT and GSH-Px. 10-8 M BPS exposure significantly induced the production of proinflammatory mediators. Activation of the NLRP3 inflammasome, TLR4, and MAPK pathways was involved in this process. Furthermore, we illustrated that NAC pretreatment diminished these effects triggered by BPS exposure. Collectively, our data suggested that BPS at a dose relevant to human serum concentration induced oxidative stress and inflammatory response in macrophages. These novel findings shed light on the concerns regarding the potential adverse effects of BPS exposure that requires further careful attention.
Collapse
Affiliation(s)
- Chunfeng Xie
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China
| | - Miaomiao Ge
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China
| | - Jianliang Jin
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Haie Xu
- Department of Clinical Nutrition, The Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210011, China
| | - Li Mao
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, 223300, China
| | - Shanshan Geng
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China
| | - Jieshu Wu
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China
| | - Jianyun Zhu
- Suzhou Digestive Diseases and Nutrition Research Center, North District of Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, No. 242 Guangji Road, Suzhou, Jiangsu, 215008, China.
| | - Xiaoting Li
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China.
| | - Caiyun Zhong
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China; Center for Global Health, Nanjing Medical University, 818 East Tianyuan Rd, Jiangning, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
27
|
Tyagi A, Kamal MA, Poddar NK. Integrated Pathways of COX-2 and mTOR: Roles in Cell Sensing and Alzheimer's Disease. Front Neurosci 2020; 14:693. [PMID: 32742252 PMCID: PMC7364283 DOI: 10.3389/fnins.2020.00693] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Cyclooxygenases (COX) are enzymes catalyzing arachidonic acid into prostanoids. COX exists in three isoforms: COX-1, 2, and 3. COX-1 and COX-2 have been widely studied in order to explore and understand their involvement in Alzheimer’s disease (AD), a progressive neuroinflammatory dementia. COX-2 was traditionally viewed to be expressed only under pathological conditions and to have detrimental effects in AD pathophysiology and neurodegeneration. However, an increasing number of reports point to much more complex roles of COX-2 in AD. Mammalian/mechanistic target of rapamycin (mTOR) has been considered as a hub which integrates multiple signaling cascades, some of which are also involved in AD progression. COX-2 and mTOR are both involved in environmental sensing, growth, and metabolic processes of the cell. They are also known to act in cooperation in many different cancers and thus, their role together in normal cellular functions as well as AD has been explored in this review. Some of the therapeutic approaches targeting COX-2 and mTOR in AD and cancer are also discussed.
Collapse
Affiliation(s)
- Arti Tyagi
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Mohammad A Kamal
- King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Enzymoics, Hebersham, NSW, Australia
| | | |
Collapse
|
28
|
Microarray analysis of the expression profile of immune-related gene in rapid recurrence early-stage lung adenocarcinoma. J Cancer Res Clin Oncol 2020; 146:2299-2310. [PMID: 32556504 PMCID: PMC7382661 DOI: 10.1007/s00432-020-03287-7] [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: 12/29/2019] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Background Although much progress has been made in the diagnosis of early-stage lung adenocarcinoma (ES-LUAD), the prognosis for ES-LUAD patients with rapid recurrence is still poor. Importantly, there is currently no effective and precise method to screen patients who may develop rapid recurrence. Therefore, it is necessary to identify potential differentially expressed genes (DEGs) in ES-LUAD patients with rapid recurrence and non-rapid recurrence. Methods Affymetrix GeneChip Human Transcriptome Array was used to identify DEGs between ES-LUAD patients with rapid recurrence and non-rapid recurrence. Rapid recurrence was defined as recurrence-free survival (RFS) ≦ 1 year and non-rapid recurrence was defined as RFS ≧ 3 years. The biological functions of the DEGs were analyzed by GO and KEGG pathway enrichment analyses. The protein–protein interaction (PPI) network of identified DEGs was conducted by STRING and Cytoscape software. The expression level of crucial hub genes and tumor-infiltrating lymphocytes (TILs) was verified by immunohistochemistry (IHC). Results A total of 416 DEGs were identified between ES-LUAD patients with and without rapid recurrence. The results of GO analysis revealed that 2 of the top 10 categories in the domain of cellular component, 2 of the top 10 in the domain of molecular function, and 9 of the top 10 in the domain of biological process were functionally related to immunity. The results of KEGG analysis showed that 6 of the top 8 pathways were functionally involved in immune regulation and inflammatory response. The PPI network analysis identified ten crucial nodal protein, including EGFR, MMP9, IL-1β, PTGS2, MMP1, and 5 histone proteins, which constituted 25 key interactions. IL-1β and PTGS2 expression were closely related to immunity and IHC analysis further revealed that low expression of IL-1β and PTGS2 is associated with rapid recurrence. Kaplan–Meier analysis further revealed that LUAD patients with lower IL-1β or PTGS2 expression had a worse RFS. When the TIL density of CD3+, CD4+, CD8+ and CD20+ subsets was less than 20%, ES-LUAD patients have a higher probability of rapid recurrence. Conclusion There were significant differences in the expression of immune-related genes between patients with rapid recurrence and patient with non-rapid recurrence. Immune-related genes such as IL-1β and PTGS2 and TIL density (20%) play important roles in rapid recurrence of ES-LUAD. This study provided a theoretical basis for distinguishing the two types of patients from an immunological perspective. Electronic supplementary material The online version of this article (10.1007/s00432-020-03287-7) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Shao W, Kuhn C, Mayr D, Ditsch N, Kailuwait M, Wolf V, Harbeck N, Mahner S, Jeschke U, Cavaillès V, Sixou S. Cytoplasmic PPARγ is a marker of poor prognosis in patients with Cox-1 negative primary breast cancers. J Transl Med 2020; 18:94. [PMID: 32085795 PMCID: PMC7035771 DOI: 10.1186/s12967-020-02271-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/14/2020] [Indexed: 01/16/2023] Open
Abstract
Background The aim of this study was to investigate the expression of the nuclear receptor PPARγ, together with that of the cyclooxygenases Cox-1 and Cox-2, in breast cancer (BC) tissues and to correlate the data with several clinicobiological parameters including patient survival. Methods In a well characterized cohort of 308 primary BC, PPARγ, Cox-1 and Cox-2 cytoplasmic and nuclear expression were evaluated by immunohistochemistry. Correlations with clinicopathological and aggressiveness features were analyzed, as well as survival using Kaplan–Meier analysis. Results PPARγ was expressed in almost 58% of the samples with a predominant cytoplasmic location. Cox-1 and Cox-2 were exclusively cytoplasmic. Cytoplasmic PPARγ was inversely correlated with nuclear PPARγ and ER expression, but positively with Cox-1, Cox-2, and other high-risk markers of BC, e.g. HER2, CD133, and N-cadherin. Overall survival analysis demonstrated that cytoplasmic PPARγ had a strong correlation with poor survival in the whole cohort, and even stronger in the subgroup of patients with no Cox-1 expression where cytoplasmic PPARγ expression appeared as an independent marker of poor prognosis. In support of this cross-talk between PPARγ and Cox-1, we found that Cox-1 became a marker of good prognosis only when cytoplasmic PPARγ was expressed at high levels. Conclusion Altogether, these data suggest that the relative expression of cytoplasmic PPARγ and Cox-1 may play an important role in oncogenesis and could be defined as a potential prognosis marker to identify specific high risk BC subgroups.
Collapse
Affiliation(s)
- Wanting Shao
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Christina Kuhn
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Doris Mayr
- Department of Pathology, LMU Munich, Munich, Germany
| | - Nina Ditsch
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Magdalena Kailuwait
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Verena Wolf
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Nadia Harbeck
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Sven Mahner
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Udo Jeschke
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany.
| | - Vincent Cavaillès
- IRCM-Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université Montpellier, Parc Euromédecine, 208 rue des Apothicaires, 34298, Montpellier Cedex 5, France
| | - Sophie Sixou
- Breast Center, Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany.,Faculté des Sciences Pharmaceutiques, Université Paul Sabatier Toulouse III, 31062, Toulouse Cedex 09, France.,Cholesterol Metabolism and Therapeutic Innovations, Cancer Research Center of Toulouse (CRCT), UMR 1037, CNRS, Inserm, UPS, Université de Toulouse, 31037, Toulouse, France
| |
Collapse
|
30
|
Prostaglandin F 2α-PTGFR signaling promotes proliferation of endometrial epithelial cells of cattle through cell cycle regulation. Anim Reprod Sci 2020; 213:106276. [PMID: 31987327 DOI: 10.1016/j.anireprosci.2020.106276] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 11/14/2019] [Accepted: 01/07/2020] [Indexed: 01/28/2023]
Abstract
There is production of prostaglandin F2α (PGF2α) and there is PGF2α receptor (PTGFR) mRNA transcript in endometrial epithelial cells of cattle. The aims of the present study were to (1) determine whether PGF2α-PTGFR signaling modulates the proliferation of endometrial epithelial cells and (2) increase knowledge of PGF2α-PTGFR signaling on the physiological and pharmacological processes in the endometrium of cattle. Amount of cellular proliferation was determined using real-time cell analysis and cell proliferation reagent WST-1 procedures. Abundance of cyclins, cyclin-dependent kinases (CDKs), cyclin-kinase inhibitors, proliferating cell nuclear antigen (PCNA), cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), PTGFR, epidermal growth factor (EGF) mRNA and protein abundances were evaluated using real-time RT-PCR and western blot analyses. The PGF2α-PTGFR signaling promoted the proliferation of endometrial epithelial cells by inducing changes in abundance of mRNA transcript and protein that resulted in an increase in the abundance for the cyclins (A, B1, D1, D3), CDKs (1, 2, 4, 6), and PCNA; decrease in abundance for p21; and increase in abundance for EGF, COX-1, COX-2, and PTGFR. There was a direct molecular association between PGF2α-PTGFR signaling and cell cycle regulation in endometrial epithelial cells of cattle. In addition, findings improve the understanding of PGF2α-PTGFR signaling in the physiological and pharmacological processes of the endometrium of cattle.
Collapse
|
31
|
Santoro AL, Drummond RD, Silva IT, Ferreira SS, Juliano L, Vendramini PH, Lemos MBDC, Eberlin MN, Andrade VP. In Situ DESI-MSI Lipidomic Profiles of Breast Cancer Molecular Subtypes and Precursor Lesions. Cancer Res 2020; 80:1246-1257. [PMID: 31911556 DOI: 10.1158/0008-5472.can-18-3574] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 11/08/2019] [Accepted: 12/23/2019] [Indexed: 11/16/2022]
Abstract
Clinically meaningful molecular subtypes for classification of breast cancers have been established, however, initiation and progression of these subtypes remain poorly understood. The recent development of desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) facilitates the convergence of analytical chemistry and traditional pathology, allowing chemical profiling with minimal tissue pretreatment in frozen samples. Here, we characterized the chemical composition of molecular subtypes of breast cancer with DESI-MSI. Regions of interest were identified, including invasive breast cancer (IBC), ductal carcinoma in situ (DCIS), and adjacent benign tissue (ABT), and metabolomic profiles at 200 μm elaborated using Biomap software and the Lasso method. Top ions identified in IBC regions included polyunsaturated fatty acids, deprotonated glycerophospholipids, and sphingolipids. Highly saturated lipids, as well as antioxidant molecules [taurine (m/z 124.0068), uric acid (m/z 167.0210), ascorbic acid (m/z 175.0241), and glutathione (m/z 306.0765)], were able to distinguish IBC from ABT. Moreover, luminal B and triple-negative subtypes showed more complex lipid profiles compared with luminal A and HER2 subtypes. DCIS and IBC were distinguished on the basis of cell signaling and apoptosis-related ions [fatty acids (341.2100 and 382.3736 m/z) and glycerophospholipids (PE (P-16:0/22:6, m/z 746.5099, and PS (38:3), m/z 812.5440)]. In summary, DESI-MSI identified distinct lipid composition between DCIS and IBC and across molecular subtypes of breast cancer, with potential implications for breast cancer pathogenesis. SIGNIFICANCE: These findings present the first in situ metabolomic findings of the four molecular subtypes of breast cancer, DCIS, and normal tissue, and add to the understanding of their pathogenesis.
Collapse
Affiliation(s)
| | - Rodrigo D Drummond
- Laboratory of Computational Biology and Bioinformatics, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Israel Tojal Silva
- Laboratory of Computational Biology and Bioinformatics, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Severino S Ferreira
- Department of Surgical Pathology, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Luiz Juliano
- Department of Surgical Pathology, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Pedro H Vendramini
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Marcos N Eberlin
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, Brazil.,Mackenzie Presbyterian University, School of Engineering, São Paulo, Brazil
| | - Victor Piana Andrade
- Department of Surgical Pathology, A.C. Camargo Cancer Center, São Paulo, Brazil.
| |
Collapse
|
32
|
Mirshahidi S, de Necochea-Campion R, Moretta A, Williams NL, Reeves ME, Otoukesh S, Mirshahidi HR, Khosrowpour S, Duerksen-Hughes P, Zuckerman LM. Inhibitory Effects of Indomethacin in Human MNNG/HOS Osteosarcoma Cell Line In Vitro. Cancer Invest 2019; 38:23-36. [DOI: 10.1080/07357907.2019.1698592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Saied Mirshahidi
- Biospecimen Laboratory, Loma Linda University Cancer Center, Loma Linda University School of Medicine, Loma Linda, CA, USA
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Rosalia de Necochea-Campion
- Biospecimen Laboratory, Loma Linda University Cancer Center, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Annie Moretta
- Biospecimen Laboratory, Loma Linda University Cancer Center, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Nadine L. Williams
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Mark E. Reeves
- Biospecimen Laboratory, Loma Linda University Cancer Center, Loma Linda University School of Medicine, Loma Linda, CA, USA
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- Division of Surgical Oncology, Department of Surgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Salman Otoukesh
- Division of Hematology and Oncology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Hamid R. Mirshahidi
- Division of Hematology and Oncology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Shahrzad Khosrowpour
- Leatherby Libraries/Collection Management Division, Chapman University, Orange, CA, USA
| | | | - Lee M. Zuckerman
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
33
|
ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6175804. [PMID: 31467634 PMCID: PMC6701375 DOI: 10.1155/2019/6175804] [Citation(s) in RCA: 509] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) are by-products of normal cell activity. They are produced in many cellular compartments and play a major role in signaling pathways. Overproduction of ROS is associated with the development of various human diseases (including cancer, cardiovascular, neurodegenerative, and metabolic disorders), inflammation, and aging. Tumors continuously generate ROS at increased levels that have a dual role in their development. Oxidative stress can promote tumor initiation, progression, and resistance to therapy through DNA damage, leading to the accumulation of mutations and genome instability, as well as reprogramming cell metabolism and signaling. On the contrary, elevated ROS levels can induce tumor cell death. This review covers the current data on the mechanisms of ROS generation and existing antioxidant systems balancing the redox state in mammalian cells that can also be related to tumors.
Collapse
|
34
|
|
35
|
Hurst EA, Pang LY, Argyle DJ. The selective cyclooxygenase-2 inhibitor mavacoxib (Trocoxil) exerts anti-tumour effects in vitro independent of cyclooxygenase-2 expression levels. Vet Comp Oncol 2019; 17:194-207. [PMID: 30767381 DOI: 10.1111/vco.12470] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/20/2018] [Accepted: 02/11/2019] [Indexed: 12/14/2022]
Abstract
The inducible inflammatory enzyme cyclooxygenase-2 (COX-2) and its product prostaglandin E2 (PGE2 ) are prominent tumour promoters, and expression of COX-2 is elevated in a number of tumours of both humans and canines. Targeting COX-2 in cancer is an attractive option because of readily available non-steroidal anti-inflammatory drugs (NSAIDs), and there is a clear epidemiological link between NSAID use and cancer risk. In this study, we aim to establish the anti-tumourigenic effects of the selective, long-acting COX-2 inhibitor mavacoxib. We show here that mavacoxib is cytotoxic to a panel of human and canine osteosarcoma, mammary and bladder carcinoma cancer cell lines; that it can induce apoptosis and inhibit the migration of these cells. Interestingly, we establish that mavacoxib can exert these effects independently of elevated COX-2 expression. This study highlights the potential novel use of mavacoxib as a cancer therapeutic, suggesting that mavacoxib may be an effective anti-cancer agent independent of tumour COX-2 expression.
Collapse
Affiliation(s)
- Emma A Hurst
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Lisa Y Pang
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - David J Argyle
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
36
|
Gupta R, Cristea M, Frankel P, Ruel C, Chen C, Wang Y, Morgan R, Leong L, Chow W, Koczywas M, Koehler S, Lim D, Luu T, Martel C, McNamara M, Somlo G, Twardowski P, Yen Y, Idorenyi A, Raechelle T, Carroll M, Chung V. Randomized trial of oral cyclophosphamide versus oral cyclophosphamide with celecoxib for recurrent epithelial ovarian, fallopian tube, and primary peritoneal cancer. Cancer Treat Res Commun 2019; 21:100155. [PMID: 31279962 PMCID: PMC9018111 DOI: 10.1016/j.ctarc.2019.100155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 04/26/2023]
Abstract
BACKGROUND Oral metronomic chemotherapy, which has low toxicity, has demonstrated promising anti-tumor and anti-angiogenic properties that may lead to prolonged progression-free survival and improved response rates in patients with recurrent epithelial ovarian cancer (EOC). These effects may be enhanced by the co-administration of anti-angiogenic agents. METHODS We conducted a randomized phase II clinical trial to evaluate the therapeutic benefit of oral metronomic cyclophosphamide (CTX) alone and with the anti-angiogenic drug celecoxib in patients with gynecological malignancies. 52 patients were randomly assigned to two treatments arms: 50 mg oral CTX daily alone (Arm A) or with 400 mg celecoxib twice daily (Arm B). The primary endpoint was response rate. Secondary endpoints included toxicity, time to treatment failure, and overall survival. RESULTS In Arm A (n = 26), 3 patients (12%) had stable disease >6 months and 1 (4%) had a partial response. In Arm B, 5 (19%) had stable disease >6 months and 1 patient (4%) had a partial response. There were no significant between-group differences in overall survival (9.69 months [95% CI 3.84-13.18] vs. 12.55 months [6.67-17.61]) or in median time to treatment failure (1.84 months [1.68-2.76] vs. 1.92 months [1.64-5.22]). The most common adverse events were nausea, vomiting, and abdominal pain. CONCLUSIONS Oral metronomic CTX has activity with no major toxicities in heavily pretreated recurrent gynecological cancers and may be considered in patients with indolent disease. We did not observe any additional benefit of celecoxib treatment, though this may be due to small sample sizes.
Collapse
Affiliation(s)
- Rohan Gupta
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Mihaela Cristea
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Paul Frankel
- Information Sciences, City of Hope National Cancer Center, Duarte, CA, United States
| | - Christopher Ruel
- Information Sciences, City of Hope National Cancer Center, Duarte, CA, United States
| | - Chen Chen
- Clinical Informatics, City of Hope National Cancer Center, Duarte, CA, United States
| | - Yingyu Wang
- Clinical Informatics, City of Hope National Cancer Center, Duarte, CA, United States
| | - Robert Morgan
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Lucille Leong
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Warren Chow
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Marianna Koczywas
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Steve Koehler
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Dean Lim
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Thehang Luu
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Cynthia Martel
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Mark McNamara
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - George Somlo
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Przemyslaw Twardowski
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Yun Yen
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Amanam Idorenyi
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Tinsley Raechelle
- Clinical Trial Office, City of Hope National Cancer Center, Duarte, CA, United States
| | - Mary Carroll
- Clinical Trial Office, City of Hope National Cancer Center, Duarte, CA, United States
| | - Vincent Chung
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States.
| |
Collapse
|
37
|
Cyclooxygenase-1 (COX-1) and COX-1 Inhibitors in Cancer: A Review of Oncology and Medicinal Chemistry Literature. Pharmaceuticals (Basel) 2018; 11:ph11040101. [PMID: 30314310 PMCID: PMC6316056 DOI: 10.3390/ph11040101] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 12/12/2022] Open
Abstract
Prostaglandins and thromboxane are lipid signaling molecules deriving from arachidonic acid by the action of the cyclooxygenase isoenzymes COX-1 and COX-2. The role of cyclooxygenases (particularly COX-2) and prostaglandins (particularly PGE₂) in cancer-related inflammation has been extensively investigated. In contrast, COX-1 has received less attention, although its expression increases in several human cancers and a pathogenetic role emerges from experimental models. COX-1 and COX-2 isoforms seem to operate in a coordinate manner in cancer pathophysiology, especially in the tumorigenesis process. However, in some cases, exemplified by the serous ovarian carcinoma, COX-1 plays a pivotal role, suggesting that other histopathological and molecular subtypes of cancer disease could share this feature. Importantly, the analysis of functional implications of COX-1-signaling, as well as of pharmacological action of COX-1-selective inhibitors, should not be restricted to the COX pathway and to the effects of prostaglandins already known for their ability of affecting the tumor phenotype. A knowledge-based choice of the most appropriate tumor cell models, and a major effort in investigating the COX-1 issue in the more general context of arachidonic acid metabolic network by using the systems biology approaches, should be strongly encouraged.
Collapse
|
38
|
Wang Y, Mandal AK, Son YO, Pratheeshkumar P, Wise JTF, Wang L, Zhang Z, Shi X, Chen Z. Roles of ROS, Nrf2, and autophagy in cadmium-carcinogenesis and its prevention by sulforaphane. Toxicol Appl Pharmacol 2018; 353:23-30. [PMID: 29885333 PMCID: PMC6281793 DOI: 10.1016/j.taap.2018.06.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
Abstract
Environmental and occupational exposures to cadmium increase the risk of various cancers, including lung cancer. The carcinogenic mechanism of cadmium, including its prevention remains to be investigated. Using fluorescence and electron spin resonance spin trapping, the present study shows that in immortalized lung cells (BEAS-2BR cells), exposure cadmium generated reactive oxygen species (ROS). Through ROS generation, cadmium increased the protein level of TNF-α, which activated NF-κB and its target protein COX-2, creating an inflammatory microenvironment. As measured by anchorage-independent colony formation assay, cadmium induced malignant cell transformation. Inhibition of ROS by antioxidants inhibited transformation, showing that ROS were important in the mechanism of this process. The inflammatory microenvironment created by cadmium may also contribute to the mechanism of the transformation. Using tandem fluorescence protein mCherry-GFP-LC3 construct, the present study shows that cadmium-transformed cells had a property of autophagy deficiency, resulting in accumulation of autophagosomes and increased p62. This protein upregulated Nrf2, which also upregulated p62 through positive feed-back mechanism. Constitutive Nrf2 activation increased its downstream anti-apoptotic proteins, Bcl-2 and Bcl-xl, resulting in apoptosis resistance. In untransformed BEAS-2BR cells, sulforaphane, a natural compound, increased autophagy, activated Nrf2, and decreased ROS. In cadmium-transformed BEAS-2BR cells, sulforaphane restored autophagy, decreased Nrf2, and decreased apoptosis resistance. In untransformed cells, this sulforaphane induced inducible Nrf2 to decrease ROS and possibly malignant cell transformation. In cadmium-transformed cells, it decreased constitutive Nrf2 and reduced apoptosis resistance. The dual roles of sulforaphane make this natural compound a valuable agent for prevention against cadmium-induced carcinogenesis.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, People's Republic of China; Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Ardhendu Kumar Mandal
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - James T F Wise
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Division of Nutritional Sciences, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536-0305,USA
| | - Lei Wang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| | - Zhimin Chen
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, People's Republic of China.
| |
Collapse
|
39
|
Cyclooxygenase-2 Expression Is a Predictive Marker for Late Recurrence in Colorectal Cancer. Gastroenterol Res Pract 2018; 2018:7968149. [PMID: 30034467 PMCID: PMC6035850 DOI: 10.1155/2018/7968149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/26/2018] [Accepted: 06/06/2018] [Indexed: 11/17/2022] Open
Abstract
Introduction Cyclooxygenase-2 (COX-2) expression is elevated in colorectal cancer (CRC). However, data about the relation between COX-2 expression and the impact on the biologic behavior of recurrent disease are inconclusive as yet. The aim of this study is to investigate the relationship between the status of COX-2 expression in the primary CRC and the characteristics of recurrence after curative resection of stage I to III CRC. Materials and Methods Ninety-eight patients with recurrence in 376 CRC patients, who underwent curative surgery between January 1991 and August 2001, were retrospectively assessed. Immunohistochemical staining, performed for the presence of COX-2 on tissue microarrays, was analyzed. Results Forty-six patients showed elevated COX-2 expression, and 52 patients did not. The mean time to recurrence was significantly longer in the positive group than in the negative group (34.1 months ± 30.0 versus 21.9 months ± 17.4; P = 0.019). Positive COX-2 expression was correlated with late recurrence (>3 years after surgery) [43.5% versus 13.5%; P = 0.001]. In multivariate analysis, COX-2 expression was an independent factor associated with late recurrence (OR 4.656; 95% CI, 1.696 to 12.779; P = 0.003). Recurrence pattern and postrecurrence survival were not different between the two groups. Conclusions Elevated COX-2 expression in itself is not a prognostic factor, but COX-2 expression in tumor tissue may be an independent predictive marker of late recurrence for patients with stage I to III CRC.
Collapse
|
40
|
Yoo HJ, Kim M, Kim M, Kang M, Jung KJ, Hwang SM, Jee SH, Lee JH. Analysis of metabolites and metabolic pathways in breast cancer in a Korean prospective cohort: the Korean Cancer Prevention Study-II. Metabolomics 2018; 14:85. [PMID: 30830383 DOI: 10.1007/s11306-018-1382-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 06/05/2018] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Since blood is in contact with all tissues in the body and is considered to dynamically reflect the body's pathophysiological status, serum metabolomics changes are important and have diagnostic value in early cancer detection. OBJECTIVES In this prospective study, we investigated the application of metabolomics to differentiate subjects with incident breast cancer (BC) from subjects who remained free of cancer during a mean follow-up period of 7 years with the aim of identifying valuable biomarkers for BC. METHODS Baseline serum samples from 84 female subjects with incident BC (BC group) and 88 cancer-free female subjects (control group) were used. Metabolic alterations associated with BC were investigated via metabolomics analysis of the baseline serum samples using ultra-performance liquid chromatography-linear-trap quadrupole-Orbitrap mass spectrometry. RESULTS A total of 57 metabolites were identified through the metabolic analysis. Among them, 20 metabolite levels were significantly higher and 22 metabolite levels were significantly lower in the BC group than in the control group at baseline. Ten metabolic pathways, including amino acid metabolism, arachidonic acid (AA) metabolism, fatty acid metabolism, linoleic acid metabolism, and retinol metabolism, showed significant differences between the BC group and the control group. Logistic regression revealed that the incidence of BC was affected by leucine, AA, prostaglandin (PG)J2, PGE2, and γ-linolenic acid (GLA). CONCLUSIONS This prospective study showed the clinical relevance of dysregulation of various metabolisms on the incidence of BC. Additionally, leucine, AA, PGJ2, PGE2, and GLA were identified as independent variables affecting the incidence of BC.
Collapse
Affiliation(s)
- Hye Jin Yoo
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, South Korea
| | - Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Minkyung Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Minsik Kang
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, South Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Keum Ji Jung
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Se-Mi Hwang
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Sun Ha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Jong Ho Lee
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, South Korea.
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea.
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
41
|
Inceler N, Ozkan Y, Turan NN, Kahraman DC, Cetin-Atalay R, Baytas SN. Design, synthesis and biological evaluation of novel 1,3-diarylpyrazoles as cyclooxygenase inhibitors, antiplatelet and anticancer agents. MEDCHEMCOMM 2018; 9:795-811. [PMID: 30108969 DOI: 10.1039/c8md00022k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/16/2018] [Indexed: 11/21/2022]
Abstract
With the aim of achieving new compounds possessing both anti-inflammatory and antiplatelet activities, we synthesized (E)-3-[3-(pyridin-3/4-yl)-1-(phenyl/sulfonylmethylphenyl)-1H-pyrazol-4-yl]acrylamides, and evaluated their COX-1 and COX-2 inhibitory and antiplatelet activities. Since COX-2 inhibitory and antiplatelet compounds have anticancer potential, we also screened their antiproliferative effects against three human cancer cell lines. Compounds 5n, 5p, 5s, 10d, 10g and 10i were determined as dual COX-2 inhibitor/antiplatelet compounds. Compound 10h appeared to be a compound that exhibited antiplatelet activity without inhibiting the COX enzyme. Compounds 5h, 10a and 10i were the most effective derivatives which displayed antiproliferative activity against Huh7, MCF7 and HCT116 cells. Particularly, compound 10i, as the compound exhibiting the highest cytotoxic, antiplatelet and COX-2 inhibitory activity, was remarkable.
Collapse
Affiliation(s)
- Nazan Inceler
- Division of Pharmaceutical Sciences , Department of Pharmaceutical Chemistry , Faculty of Pharmacy , Gazi University , 06330 , Ankara , Turkey .
| | - Yesim Ozkan
- Department of Biochemistry , Faculty of Pharmacy , Gazi University , 06330 , Ankara , Turkey
| | - Nilufer Nermin Turan
- Department of Pharmacology , Faculty of Pharmacy , Gazi University , 06330 , Ankara , Turkey
| | - Deniz Cansen Kahraman
- Cancer Systems Biology Laboratory , Graduate School of Informatics , METU , 06800 , Ankara , Turkey
| | - Rengul Cetin-Atalay
- Cancer Systems Biology Laboratory , Graduate School of Informatics , METU , 06800 , Ankara , Turkey
| | - Sultan Nacak Baytas
- Division of Pharmaceutical Sciences , Department of Pharmaceutical Chemistry , Faculty of Pharmacy , Gazi University , 06330 , Ankara , Turkey .
| |
Collapse
|
42
|
Joy M, Elrashedy AA, Mathew B, Pillay AS, Mathews A, Dev S, Soliman ME, Sudarsanakumar C. Discovery of new class of methoxy carrying isoxazole derivatives as COX-II inhibitors: Investigation of a detailed molecular dynamics study. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2017.11.109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
43
|
Abstract
Chemotherapy and hormonal therapy have significantly decreased breast cancer mortality, although with considerable side effects and financial costs. In the USA, over three million women are living after a breast cancer diagnosis and are eager for new treatments that are low in toxicity and cost. Multiple observational studies have reported improved breast cancer survival with regular aspirin use. Furthermore, pooled data from five large randomized trials of aspirin for cardiovascular disease showed that subjects on aspirin had decreased risk of cancer mortality and decreased risk of metastatic cancer. Although the potential mechanism for aspirin preventing breast cancer is not known, possible pathways may involve platelets, inflammation, cyclooxygenase (COX) 2, hormones, or PI3 kinase. This review article summarizes the current epidemiologic and clinical trial evidence as well as possible underlying mechanisms that justify current phase III randomized trials of aspirin to improve breast cancer survival.
Collapse
|
44
|
Tury S, Becette V, Assayag F, Vacher S, Benoist C, Kamal M, Marangoni E, Bièche I, Lerebours F, Callens C. Combination of COX-2 expression and PIK3CA mutation as prognostic and predictive markers for celecoxib treatment in breast cancer. Oncotarget 2018; 7:85124-85141. [PMID: 27835884 PMCID: PMC5356723 DOI: 10.18632/oncotarget.13200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 10/26/2016] [Indexed: 12/12/2022] Open
Abstract
COX-2 expression level and prognostic value are still a matter of debate in breast cancer (BC). We addressed these points in the context of PIK3CA mutational status. Based on an interesting study of aspirin efficacy in colorectal cancer, we hypothesized that celecoxib antitumoral activity may be restricted to PIK3CA mutated BC. COX-2 mRNA expression was analyzed in 446 BC samples and in 61 BC patient-derived xenografts (PDX) using quantitative RT-PCR. The prognostic impact of COX-2 expression level was assessed independently and according to PIK3CA mutational status in our cohort and in a validation set of 817 BC. The antitumoral activity of celecoxib was tested in two triple-negative (TN) PDX with a PIK3CA wild-type (wt) or mutated genotype. COX-2 mRNA was overexpressed in 2% of BC and significantly associated with TN subtype. Metastasis-free survival (MFS) was significantly better in patients with high COX-2 expression level, the prognosis of whom was similar to patients with PIK3CA mutations. TCGA validation cohort confirmed that patients with low COX-2 expression PIK3CA wt tumors had the worse disease-free survival (DFS) compared to all other subgroups. Celecoxib had a significant antitumoral effect in PIK3CA mutated PDX only. Celecoxib antitumoral activity involved S6 ribosomal protein and AKT phosphorylation. Low expression of COX-2 has a significant negative impact on the MFS/DFS of BC patients. Antitumoral effect of celecoxib is restricted to PIK3CA mutated PDX. These results suggest that PIK3CA mutation may be a new predictive biomarker for celecoxib efficacy.
Collapse
Affiliation(s)
- Sandrine Tury
- Pharmacogenomic Unit, Genetics Laboratory, Institut Curie, Paris, France
| | - Véronique Becette
- Department of Pathology, Institut Curie, Hôpital René Huguenin, Saint-Cloud, France
| | - Franck Assayag
- Laboratory of Preclinical Investigations, Translational Research Department, Institut Curie, Paris, France
| | - Sophie Vacher
- Pharmacogenomic Unit, Genetics Laboratory, Institut Curie, Paris, France
| | - Camille Benoist
- Pharmacogenomic Unit, Genetics Laboratory, Institut Curie, Paris, France
| | - Maud Kamal
- Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigations, Translational Research Department, Institut Curie, Paris, France
| | - Ivan Bièche
- Pharmacogenomic Unit, Genetics Laboratory, Institut Curie, Paris, France
| | - Florence Lerebours
- Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France
| | - Céline Callens
- Pharmacogenomic Unit, Genetics Laboratory, Institut Curie, Paris, France
| |
Collapse
|
45
|
Kochel TJ, Reader JC, Ma X, Kundu N, Fulton AM. Multiple drug resistance-associated protein (MRP4) exports prostaglandin E2 (PGE2) and contributes to metastasis in basal/triple negative breast cancer. Oncotarget 2018; 8:6540-6554. [PMID: 28029661 PMCID: PMC5351651 DOI: 10.18632/oncotarget.14145] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 11/22/2016] [Indexed: 02/02/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) and its primary enzymatic product, prostaglandin E2 (PGE2), are associated with a poor prognosis in breast cancer. In order to elucidate the factors contributing to intratumoral PGE2 levels, we evaluated the expression of COX-2/PGE2 pathway members MRP4, the prostaglandin transporter PGT, 15-PGDH (PGE2 metabolism), the prostaglandin E receptor EP4, COX-1, and COX-2 in normal, luminal, and basal breast cancer cell lines. The pattern of protein expression varied by cell line reflecting breast cancer heterogeneity. Overall, basal cell lines expressed higher COX-2, higher MRP4, lower PGT, and lower 15-PGDH than luminal cell lines resulting in higher PGE2 in the extracellular environment. Genetic or pharmacologic suppression of MRP4 expression or activity in basal cell lines led to less extracellular PGE2. The key finding is that xenografts derived from a basal breast cancer cell line with stably suppressed MRP4 expression showed a marked decrease in spontaneous metastasis compared to cells with unaltered MRP4 expression. Growth properties of primary tumors were not altered by MRP4 manipulation. In addition to the well-established role of high COX-2 in promoting metastasis, these data identify an additional mechanism to achieve high PGE2 in the tumor microenvironment; high MRP4, low PGT, and low 15-PGDH. MRP4 should be examined further as a potential therapeutic target in basal breast cancer.
Collapse
Affiliation(s)
- Tyler J Kochel
- University of Maryland School of Medicine, Department of Pathology, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jocelyn C Reader
- University of Maryland School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Xinrong Ma
- University of Maryland School of Medicine, Department of Pathology, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Namita Kundu
- University of Maryland School of Medicine, Department of Pathology, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Amy M Fulton
- University of Maryland School of Medicine, Department of Pathology, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| |
Collapse
|
46
|
Huang XZ, Chen Y, Wu J, Zhang X, Wu CC, Zhang CY, Sun SS, Chen WJ. Aspirin and non-steroidal anti-inflammatory drugs use reduce gastric cancer risk: A dose-response meta-analysis. Oncotarget 2018; 8:4781-4795. [PMID: 27902474 PMCID: PMC5354871 DOI: 10.18632/oncotarget.13591] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022] Open
Abstract
Background The association between non-steroidal anti-inflammatory drugs (NSAIDs) and gastric cancer (GC) risk is controversial. The aim of this study is to evaluate the chemopreventive effect of NSAIDs for GC. Methods A literature search was performed for relevant studies using the PubMed and Embase database (up to March 2016). Risk ratios (RRs) and 95% confidence intervals (CIs) were used as the effect measures. The dose–response analysis and subgroup analysis were also performed. Results Twenty-four studies were included. Our results indicated that NSAIDs could reduce GC risk (any NSAIDs: RR=0.78, 96%CI=0.72-0.85; aspirin: RR=0.70, 95%CI=0.62-0.80; non-aspirin NSAIDs: RR=0.86, 95%CI=0.80-0.94), especially for non-cardia GC risk. Moreover, the dose-response analysis indicated the risk of GC decreased by 11% and 5% for 2 years increment of any NSAIDs and aspirin use, respectively. There were nonlinear relationships between the frequency of any NSAIDs use and aspirin use and GC risk (P for non-linearity<0.01), with a threshold effect of 5 times/week. A monotonically decreasing trend was observed only for the frequency of less than 5 times/week. Conclusions Our results indicate that NSAIDs is inversely associated with GC risk, especially for non-cardia GC risk. NSAIDs use may become a feasible approach to prevent GC.
Collapse
Affiliation(s)
- Xuan-Zhang Huang
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| | - You Chen
- The Wenzhou Dental Hospital, Wenzhou City 325027, P.R. China
| | - Jian Wu
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| | - Xi Zhang
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| | - Cong-Cong Wu
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| | - Chao-Ying Zhang
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| | - Shuang-Shuang Sun
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| | - Wen-Jun Chen
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325027, P.R. China
| |
Collapse
|
47
|
Grabosch SM, Shariff OM, Helm CW, Cochrane Gynaecological, Neuro‐oncology and Orphan Cancer Group. Non-steroidal anti-inflammatory agents to induce regression and prevent the progression of cervical intraepithelial neoplasia. Cochrane Database Syst Rev 2018; 2:CD004121. [PMID: 29431861 PMCID: PMC6483561 DOI: 10.1002/14651858.cd004121.pub4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND This is an updated version of the original Cochrane review published in 2014, Issue 4. Cervical intraepithelial neoplasia (CIN) precedes the development of invasive carcinoma of the cervix. Current treatment of CIN is quite effective, but there is morbidity for the patient related to pain, bleeding, infection, cervical stenosis and premature birth in a subsequent pregnancy. Effective treatment with medications, rather than surgery, would be beneficial. OBJECTIVES To evaluate the effectiveness and safety of non-steroidal anti-inflammatory agents (NSAIDs), including cyclooxygenase-2 (COX-2) inhibitors, to induce regression and prevent the progression of CIN. SEARCH METHODS Previously, we searched the Cochrane Gynaecological Cancer Group Trials Register, the Cochrane Central Register of Controlled Trials (CENTRAL) (2013, Issue 11), MEDLINE (November, 2013) and Embase (November week 48, 2013). An updated search was performed in August 2017 for CENTRAL (2017, Issue 8), MEDLINE (July, week 3, 2017) and Embase (July week 31, 2017). Trial registries and journals were also searched as part of the update. SELECTION CRITERIA Randomised controlled trials (RCTs) or controlled trials of NSAIDs in the treatment of CIN. DATA COLLECTION AND ANALYSIS Three review authors independently abstracted data and assessed risks of bias in accordance with Cochrane methodology. Outcome data were pooled using fixed-effect meta-analyses. MAIN RESULTS In three RCTs, 171 women over the age of 18 years were randomised to receive celecoxib 400 mg daily for 14 to 18 weeks versus placebo (one study, 130 participants), celecoxib 200 mg twice daily by mouth for six months versus placebo (one study, 25 participants), or rofecoxib 25 mg once daily by mouth for three months versus placebo (one study, 16 participants). The study with rofecoxib was discontinued when the medicine was withdrawn from the market in 2004. The trials ran from June 2005 to April 2012, June 2002 to October 2003, and May to October 2004, respectively. We have chosen to include the data from the rofecoxib study as outcomes may be similar when other such NSAIDs are utilised.Partial or complete regression of CIN 2 or CIN 3 occurred in 31 out of 70 (44%) in the treatment arms and 19 of 62 (31%) in the placebo arms (risk ratio (RR) 1.45, 95% confidence interval (CI) 0.93 to 2.27; P value 0.10), three studies, 132 participants; moderate-certainty evidence). Complete regression of CIN 2 or CIN 3 occurred in 15 of 62 (24%) of those receiving celecoxib versus 10 of 54 (19%) of those receiving placebo (RR 1.31, 95% CI 0.65 to 2.67; P value 0.45, two studies, 116 participants; moderate-certainty evidence). Partial regression of CIN 2 or CIN 3 occurred in 14 of 62 (23%) of those receiving celecoxib versus 8 of 54 (15%) of those receiving placebo (RR 1.56, 95% CI 0.72 to 3.4; P value 0.26), two studies, 116 participants; moderate-certainty evidence).Progression to a higher grade of CIN, but not to invasive cancer, occurred in one of 12 (8%) of those receiving celecoxib and two of 13 (15%) receiving placebo (RR 0.54, 95% CI 0.05 to 5.24; P value 0.60, one study, 25 participants; very low-certainty evidence). Two studies reported no cases of progression to invasive cancer within the timeframe of the study. No toxicity was reported in the two original articles. The trial added in this update had one Grade 3 gastrointestinal adverse effect in the treatment arm, but otherwise had similar Grade 1 to 2 side effects between treatment and placebo groups. Although the studies were well-conducted and randomised, some risk of bias was detected in all studies. Furthermore, the duration of the studies was short, which may mask identifying progression to cancer.The addition of the trial in this update quadrupled the number of patients in the original review and was a well-designed multicentre trial thus, increasing the overall certainty of evidence from very low to moderate for this review. AUTHORS' CONCLUSIONS There are currently no convincing data to support a benefit for NSAIDs in the treatment of CIN. With the addition of this new, larger randomised trial we would rate this as overall moderate-certainty evidence by the GRADE criteria.
Collapse
Affiliation(s)
- Shannon M Grabosch
- Magee‐Womens Hospital of UPMCDepartment of Obstetrics, Gynecology, and Reproductive Sciences300 Halket StPittsburghPennsylvaniaUSA15213
| | - Osman M Shariff
- University of Louisville School of Medicine3646 Warner AveLouisvilleKentuckyUSA40207
| | - C. William Helm
- Princess Alexandra Wing, Royal Cornwall HospitalGynaecological OncologyTuroCornwallUKTR1 3LJ
| | | |
Collapse
|
48
|
Demirbolat GM, Altintas L, Yilmaz S, Degim IT. Development of Orally Applicable, Combinatorial Drug-Loaded Nanoparticles for the Treatment of Fibrosarcoma. J Pharm Sci 2018; 107:1398-1407. [PMID: 29339136 DOI: 10.1016/j.xphs.2018.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/16/2017] [Accepted: 01/03/2018] [Indexed: 12/15/2022]
Abstract
Nanoparticulate systems have been receiving a significant attention especially for the treatment of cancer but one of the main hurdles is to produce these developed and high-tech nanosystems in large quantities. Anticancer drug formulations are generally designed for parenteral administrations but oral administration is still the most convenient route. In this study, orally applicable nano-sized chitosan nanoparticles (NPs) were successfully prepared using Nano Spray Dryer. It is possible to produce these NPs in large quantities by simply increasing the processing time using the machine without changing any parameter. A chemotherapeutic agent (imatinib mesylate; IMA) and nonsteroidal anti-inflammatory drug (dexketoprofen trometamol) were loaded together in these NPs. NPs were also functionalized with polyethylene glycol and folic acid to obtain long circulating NPs and tumor targeting. The antitumoral activities of formulations showed that these developed NPs can enhance the effectiveness. Animal experiments were performed on fibrosarcoma-bearing mice model, and the treatment with 0.8 mg/μL/kg IMA-loaded chitosan NPs was found to be successful to slow down the growth of tumors. The tumor tissues were removed from the animals and enzymatic activities were evaluated. The inhibitory effect of tyrosine kinase was found to be enhanced from 36.4% to 68.4% when IMA was used in combination with dexketoprofen trometamol. Furthermore, all dried NPs were found to be stable for more than a year at 25°C. Presented results show that these developed combinatorial drug-loaded NPs can be used for the treatment of fibrosarcoma, and these data can provide an insight, new strategies for productions or alternatives in cancer treatment.
Collapse
Affiliation(s)
- Gulen Melike Demirbolat
- Department of Pharmaceutical Technology, Gazi University Faculty of Pharmacy, 06330, Ankara, Turkey
| | - Levent Altintas
- Department of Pharmacology and Toxicology, Ankara University Faculty of Veterinary Medicine, 06110, Ankara, Turkey
| | - Sukran Yilmaz
- Cells and Virus Bank Division, Food and Mouth Diseases Institute, 06520, Ankara, Turkey
| | - Ismail Tuncer Degim
- Department of Pharmaceutical Technology, Biruni University Faculty of Pharmacy, 34010, Topkapi, Istanbul, Turkey.
| |
Collapse
|
49
|
Garami A, Steiner AA, Romanovsky AA. Fever and hypothermia in systemic inflammation. HANDBOOK OF CLINICAL NEUROLOGY 2018; 157:565-597. [PMID: 30459026 DOI: 10.1016/b978-0-444-64074-1.00034-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Systemic inflammation-associated syndromes (e.g., sepsis and septic shock) often have high mortality and remain a challenge in emergency medicine. Systemic inflammation is usually accompanied by changes in body temperature: fever or hypothermia. In animal studies, systemic inflammation is often modeled by administering bacterial lipopolysaccharide, which triggers autonomic and behavioral thermoeffector responses and causes either fever or hypothermia, depending on the dose and ambient temperature. Fever and hypothermia are regulated changes of body temperature, which correspond to mild and severe forms of systemic inflammation, respectively. Mediators of fever and hypothermia are called endogenous pyrogens and cryogens; they are produced when the innate immune system recognizes an infectious pathogen. Upon an inflammatory challenge, hepatic and pulmonary macrophages (and later brain endothelial cells) start to release lipid mediators, of which prostaglandin (PG) E2 plays the key role, and cytokines. Blood PGE2 enters the brain and triggers fever. At later stages of fever, PGE2 synthesized within the blood-brain barrier maintains fever. In both cases, PGE2 is synthesized by cyclooxygenase-2 and microsomal PGE2synthase-1. Mediators of hypothermia are not well established. Both fever and hypothermia are beneficial host defense responses. Based on evidence from studies in laboratory animals and clinical trials in humans, fever is beneficial for fighting mild infection. Based mainly on animal studies, hypothermia is beneficial in severe systemic inflammation and infection.
Collapse
Affiliation(s)
- Andras Garami
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary.
| | - Alexandre A Steiner
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrej A Romanovsky
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| |
Collapse
|
50
|
Uçan B, Özbek M, Şahin M, Kızılgül M, Çakal E. Cyclooxygenase-2 (COX-2) gene polymorphism in patients with differentiated thyroid carcinomas in the Turkish population. Turk J Med Sci 2017; 47:1848-1853. [PMID: 29306248 DOI: 10.3906/sag-1708-49] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Background/aim: The most common thyroid malignancies are papillary and follicular thyroid carcinomas. They account for approximately 85%-90% of all thyroid cancers. Recent studies have reported the relevance of cyclooxygenase-2 (COX-2) gene polymorphism in human carcinogenesis. The aim of this study was to investigate the relationship between thyroid carcinoma and COX-2 gene polymorphism in the Turkish population.Materials and methods: We included a total of 96 differentiated thyroid cancer patients (mean age: 46.9 ± 10.3 years; 14 males, 82 females) and 83 healthy control subjects (mean age: 46.0 ± 10.6 years; 39 males, 44 females). The frequency of -765G>C, -8473T>C, and 1195A>G gene polymorphisms in the COX-2 promoter region was investigated in thyroid cancer patients and the control group using the high-resolution melting method. Results: COX-2-765G>C and COX-2-1195A>G gene polymorphisms were similar between thyroid cancer patients and the control group. There was a statistically significant difference between COX-2-8473T>C gene polymorphism in the thyroid cancer group and the control group (P = 0.012).Conclusion: The single nucleotide gene polymorphism COX-2-8473T>C might contribute to genetic susceptibility to differentiated thyroid cancer in the Turkish population.
Collapse
|