1
|
Kaur C, Sahu SK, Bansal K, DeLiberto LK, Zhang J, Tewari D, Bishayee A. Targeting Peroxisome Proliferator-Activated Receptor-β/δ, Reactive Oxygen Species and Redox Signaling with Phytocompounds for Cancer Therapy. Antioxid Redox Signal 2024; 41:342-395. [PMID: 38299535 DOI: 10.1089/ars.2023.0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Significance: Peroxisome proliferator-activated receptors (PPARs) have a moderately preserved amino-terminal domain, an extremely preserved DNA-binding domain, an integral hinge region, and a distinct ligand-binding domain that are frequently encountered with the other nuclear receptors. PPAR-β/δ is among the three nuclear receptor superfamily members in the PPAR group. Recent Advances: Emerging studies provide an insight on natural compounds that have gained increasing attention as potential anticancer agents due to their ability to target multiple pathways involved in cancer development and progression. Critical Issues: Modulation of PPAR-β/δ activity has been suggested as a potential therapeutic strategy for cancer management. This review focuses on the ability of bioactive phytocompounds to impact reactive oxygen species (ROS) and redox signaling by targeting PPAR-β/δ for cancer therapy. The rise of ROS in cancer cells may play an important part in the initiation and progression of cancer. However, excessive levels of ROS stress can also be toxic to the cells and cancer cells with increased oxidative stress are likely to be more vulnerable to damage by further ROS insults induced by exogenous agents, such as phytocompounds and therapeutic agents. Therefore, redox modulation is a way to selectively kill cancer cells without causing significant toxicity to normal cells. However, use of antioxidants together with cancer drugs may risk the effect of treatment as both act through opposite mechanisms. Future Directions: It is advisable to employ more thorough and detailed methodologies to undertake mechanistic explorations of numerous phytocompounds. Moreover, conducting additional clinical studies is recommended to establish optimal dosages, efficacy, and the impact of different phytochemicals on PPAR-β/δ.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sanjeev Kumar Sahu
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Keshav Bansal
- Department of Pharmaceutics, Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Lindsay K DeLiberto
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| |
Collapse
|
2
|
Popović L, Wintgens JP, Wu Y, Brankatschk B, Menninger S, Degenhart C, Jensen N, Wichert SP, Klebl B, Rossner MJ, Wehr MC. Profiling of ERBB receptors and downstream pathways reveals selectivity and hidden properties of ERBB4 antagonists. iScience 2024; 27:108839. [PMID: 38303712 PMCID: PMC10831936 DOI: 10.1016/j.isci.2024.108839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/20/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
ERBB receptor tyrosine kinases are involved in development and diseases like cancer, cardiovascular, neurodevelopmental, and mental disorders. Although existing drugs target ERBB receptors, the next generation of drugs requires enhanced selectivity and understanding of physiological pathway responses to improve efficiency and reduce side effects. To address this, we developed a multilevel barcoded reporter profiling assay, termed 'ERBBprofiler', in living cells to monitor the activity of all ERBB targets and key physiological pathways simultaneously. This assay helps differentiate on-target therapeutic effects from off-target and off-pathway side effects of ERBB antagonists. To challenge the assay, eight established ERBB antagonists were profiled. Known effects were confirmed, and previously uncharacterized properties were discovered, such as pyrotinib's preference for ERBB4 over EGFR. Additionally, two lead compounds selectively targeting ERBB4 were profiled, showing promise for clinical trials. Taken together, this multiparametric profiling approach can guide early-stage drug development and lead to improved future therapeutic interventions.
Collapse
Affiliation(s)
- Lukša Popović
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Jan P. Wintgens
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Yuxin Wu
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Ben Brankatschk
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Sascha Menninger
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Carsten Degenhart
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Niels Jensen
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Sven P. Wichert
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Bert Klebl
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Moritz J. Rossner
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Michael C. Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| |
Collapse
|
3
|
Rauf A, Khalil AA, Awadallah S, Khan SA, Abu‐Izneid T, Kamran M, Hemeg HA, Mubarak MS, Khalid A, Wilairatana P. Reactive oxygen species in biological systems: Pathways, associated diseases, and potential inhibitors-A review. Food Sci Nutr 2024; 12:675-693. [PMID: 38370049 PMCID: PMC10867483 DOI: 10.1002/fsn3.3784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 02/20/2024] Open
Abstract
Reactive oxygen species (ROS) are produced under normal physiological conditions and may have beneficial and harmful effects on biological systems. ROS are involved in many physiological processes such as differentiation, proliferation, necrosis, autophagy, and apoptosis by acting as signaling molecules or regulators of transcription factors. In this case, maintaining proper cellular ROS levels is known as redox homeostasis. Oxidative stress occurs because of the imbalance between the production of ROS and antioxidant defenses. Sources of ROS include the mitochondria, auto-oxidation of glucose, and enzymatic pathways such as nicotinamide adenine dinucleotide phosphate reduced (NAD[P]H) oxidase. The possible ROS pathways are NF-κB, MAPKs, PI3K-Akt, and the Keap1-Nrf2-ARE signaling pathway. This review covers the literature pertaining to the possible ROS pathways and strategies to inhibit them. Additionally, this review summarizes the literature related to finding ROS inhibitors.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of ChemistryUniversity of SwabiAnbarPakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Samir Awadallah
- Department of Medical Lab Sciences, Faculty of Allied Medical SciencesZarqa UniversityZarqaJordan
| | - Shahid Ali Khan
- Department of Chemistry, School of Natural SciencesNational University of Science and Technology (NUST)IslamabadPakistan
| | - Tareq Abu‐Izneid
- Pharmaceutical Sciences, College of PharmacyAl Ain UniversityAl Ain, Abu DhabiUAE
| | - Muhammad Kamran
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical SciencesTaibah UniversityAl‐Medinah Al‐MonawaraSaudi Arabia
| | | | - Ahood Khalid
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| |
Collapse
|
4
|
Abdelazim SA, Shaker OG, Ali O, El-Tawil M, Senousy MA. Differential expression of serum miR-486 and miR-25 in ulcerative colitis and Crohn's disease: Correlations with disease activity, extent, and location. Pathol Res Pract 2023; 252:154910. [PMID: 37939427 DOI: 10.1016/j.prp.2023.154910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
Novel reliable biomarkers of inflammatory bowel disease (IBD) are clinically imperative due to potential limitations of endoscopic techniques. MicroRNAs (miRNAs) have emerged as non-invasive biomarkers of IBD; however, the full disease-specific miRNAs signature for IBD subtypes remains elusive. We evaluated the diagnostic role of circulating miR-486 and miR-25 in IBD patients and their potential ability to discriminate IBD subtypes; ulcerative colitis (UC) and Crohn's disease (CD). Sixty UC patients, 60 CD patients, and 60 healthy controls were recruited. Serum miRNA expression was determined using RT-qPCR. Bioinformatics was employed for target gene and protein-protein interaction (PPI) network analyses. Serum miR-486 was upregulated in CD patients, but didn't change in UC patients compared to controls. Conversely, serum miR-25 was decreased in both CD and UC patients compared to controls. Only miR-486 was differentially expressed between UC and CD patients. Receiver-operating characteristic analysis revealed that serum miR-486 was superior in CD diagnosis (AUC=0.945) and significantly distinguished CD and UC patients, whereas miR-25 showed discriminative potential for both UC and CD from controls. In the multivariate logistic analysis only miR-486 was associated with the risk of CD diagnosis. Serum miR-486 was correlated with CD activity index and location of disease in CD patients, whereas miR-25 was correlated with the type/extent of UC. PPI network analysis revealed common target genes and signaling pathways for both miRNAs. Conclusively, serum miR-486 and miR-25 might serve as new biomarkers of IBD, with serum miR-486 could be employed in risk stratification of IBD subtypes and has the ground for clinical utility in CD diagnosis, whereas miR-25 has potential for UC and CD diagnosis.
Collapse
Affiliation(s)
- Samy A Abdelazim
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Omaima Ali
- Department of Biochemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia 41636, Egypt; General division for Biological Control and Research, Egyptian Drug Authority, Cairo 12618 Egypt
| | - Mai El-Tawil
- Neurology department, Kasr Al-Ainy Hospital, Cairo, Egypt
| | - Mahmoud A Senousy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 11786, Egypt
| |
Collapse
|
5
|
Shang P, Lou C, Yin L, Tao C, Dong Y, Yang G, Yuan Z. Advanced renal pelvis carcinoma patient with an ERBB2 insertion mutation: a case report. J Int Med Res 2023; 51:3000605231204502. [PMID: 37917812 PMCID: PMC10623987 DOI: 10.1177/03000605231204502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/13/2023] [Indexed: 11/04/2023] Open
Abstract
The prognosis of renal pelvis cancer is poor and therapeutic options are limited, especially for patients with advanced disease. In this report, we present a case of advanced renal pelvis carcinoma in a male patient in his 60s, characterized by an activating mutation in ERBB2. Clinical evaluation resulted in a pathological diagnosis of renal pelvis carcinoma with liver metastasis. Immunohistochemistry staining results suggested that CK, P63, and PAX8 were positively expressed, while Sy, CK7, CK20, S100, PAX8, and HEP1 were negatively expressed. Furthermore, next-generation sequencing results showed an activating mutation in the ERBB2 gene. The patient initially received a trastuzumab-based combination therapy, which led to a significant reduction in ERBB2 mutation frequency and a stable condition after three treatment cycles. However, following continuous treatment for 4 months, the patient developed drug resistance that resulted in disease relapse. Subsequently, the patient received apatinib treatment, but the therapeutic response was not satisfactory. The patient's condition underwent rapid deterioration and he ultimately succumbed to the disease. This case underscores the potential benefit of trastuzumab for treating ERBB2-mutated advanced renal pelvis cancer, but further highlights that overcoming drug resistance remains a crucial challenge for long-term efficacy.
Collapse
Affiliation(s)
- Peipei Shang
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| | - Cheng Lou
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| | - Lei Yin
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| | - Chenjie Tao
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| | - Yulong Dong
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| | - Guang Yang
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| | - Zhengang Yuan
- Department of Oncology, Shanghai Eastern Hepatobiliary Surgery Hospital, Anting Town, Jiading District, Shanghai, China
| |
Collapse
|
6
|
Gholami M, Elyasigorji Z, Amoli AD, Farzaneh P. Effects of Alkanna bracteosa extract on the expression level of HSP90α and HER2 genes in human gastric cancer cell line. ADVANCES IN TRADITIONAL MEDICINE 2022. [PMCID: PMC9473469 DOI: 10.1007/s13596-022-00657-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gastric cancer was classified as the third most deadly cancer among all other cancer types. The HSP90 and HER2 genes play essential roles in the stability and function of high-expression proteins that cause malignancy. The aim of this research was to investigate the influence of the alcoholic Alkanna bracteosa extract on the expression of HSP90α and HER2 genes in AGS cell line. Therefore, the methanolic extraction was isolated from aerial parts of the plant and AGS and HuGu cell lines were analyzed using 102.4–0.05 mg ml−1 dose concentrations in serial dilution; to measure the cell toxicity by MTT assay. Furthermore, real-time PCR analysis measured the expression level of HSP90α and HER2 genes using the IC50 dose concentrations. Quantification of apoptosis was analyzed by Annexin/PI kit in flow cytometry and DNA fragmentation tests. The results of MTT assay represented the IC50 dose concentration of 0.8 and 3.2 mg ml−1 for AGS and HuGu respectively. The rate of HER2 gene expression was significantly decreased in AGS cells treated with 0.8 mg ml−1 dose concentration compared to control. The exposure of AGS treated cells with 0.8 mg ml−1 dose concentration after 24 h represented 24.3% apoptosis and 13.3% necrosis. The agarose gel represented the DNA fragmentation pattern of apoptosis. This study demonstrated the significant differences between the cell viability rate, gene expression level, and apoptosis of the Alkanna bracteosa extract on AGS cells. These results demonstrated the first report of which the Alkanna braceteosa would be an effective candidate for possible treatment of Gastric cancer.
Collapse
Affiliation(s)
- Mina Gholami
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
- Department of Molecular and Cellular Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Zahra Elyasigorji
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | | | - Parvaneh Farzaneh
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| |
Collapse
|
7
|
Sidhanth C, Bindhya S, Krishnapriya S, Manasa P, Shabna A, Alifia J, Patole C, Kumar V, Garg M, Ganesan TS. Phosphoproteome of signaling by ErbB2 in ovarian cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140768. [PMID: 35158093 DOI: 10.1016/j.bbapap.2022.140768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/07/2022] [Accepted: 02/04/2022] [Indexed: 06/14/2023]
Abstract
The gene for receptor tyrosine kinase ErbB2 is amplified in breast and ovarian tumours. The linear pathway by which signals are transduced through ErbB2 are well known. However, second generation questions that address spatial aspects of signaling remain. To address this, we have undertaken a mass spectrometry approach to identify phosphoproteins specific for ErbB2 using the inhibitors Lapatinib and CP724714 in ovarian cancer cells. The ErbB2 specific proteins identified in SKOV-3 cells were Myristoylated alanine-rich C-kinase substrate, Protein capicua homolog, Protein peptidyl isomerase G, Protein PRRC2C, Chromobox homolog1 and PRP4 homolog. We have evaluated three phosphoproteins PKM2, Aldose reductase and MARCKS in SKOV-3 cells. We observed that PKM2 was phosphorylated by EGF but was not inhibited by Lapatinib and CP724714. The activity of aldose reductase in reducing NADPH as a substrate was significantly higher in EGF stimulated cells which was inhibited by Lapatinib and CP724714 but not by Geftinib (EGFR inhibitor). MARCKS was phosphorylated on stimulation of SKOV-3 cells with EGF that was inhibited by Lapatinib and CP724714 which was dependent on the kinase activity of ErbB2. These results have identified phosphoproteins that are specific to ErbB2 which have not been previously reported and sets the basis for future experiments.
Collapse
Affiliation(s)
- C Sidhanth
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - S Bindhya
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - S Krishnapriya
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - P Manasa
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - A Shabna
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - J Alifia
- Mass Spectrometry Facility Proteomics, National Centre for Biological Sciences (NCBS), Bangalore, India
| | - C Patole
- Mass Spectrometry Facility Proteomics, National Centre for Biological Sciences (NCBS), Bangalore, India
| | - V Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
| | - M Garg
- Amity Institute of Molecular Medicine & Stem cell Research, Amity University, Delhi, India
| | - T S Ganesan
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India.
| |
Collapse
|
8
|
LASP-1 interacts with ErbB2 in ovarian cancer cells. Biochem J 2021; 479:23-38. [PMID: 34881777 DOI: 10.1042/bcj20210173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022]
Abstract
LASP-1 was identified as a protein following mass spectrometric analysis of phosphoproteins consequent to signaling by ErbB2 in SKOV-3 cells. It has been previously identified as an oncogene and is located on chromosomal arm 17q 0.76Mb centromeric to ErbB2. It is expressed in serous ovarian cancer cell lines as a 40kDa protein. In SKOV-3 cells, it was phosphorylated and was inhibited by Lapatinib and CP7274714. LASP-1 co-immunoprecipitated with ErbB2 in SKOV-3 cells, suggesting a direct interaction. This interaction and phosphorylation were independent of the kinase activity of ErbB2. Moreover, the binding of LASP-1 to ErbB2 was independent of the tyrosine phosphorylation of LASP-1. LASP-1 was neither expressed on the surface epithelium of the normal ovary nor in the fallopian tube. It was expressed in 28% of ovarian tumours (n=101) that did not significantly correlate with other clinical factors. In tumours from patients with invasive ductal carcinoma of the breast who had ErbB2 amplification (3+), LASP-1 was expressed in 3/20 (p <0.001). Analysis of the expression of an independent dataset of ovarian and breast tumors from TCGA showed the significant co-occurrence of ErbB2 and LASP-1 (p<0.01). These results suggest that LASP-1 and ErbB2 interaction could be important in the pathogenesis of ovarian cancer.
Collapse
|
9
|
Huang W, Zhang Q, Wu G, Chen PP, Li J, McCabe Gillen K, Spincemaille P, Chiang GC, Gupta A, Wang Y, Chen F. DCE-MRI quantitative transport mapping for noninvasively detecting hypoxia inducible factor-1α, epidermal growth factor receptor overexpression, and Ki-67 in nasopharyngeal carcinoma patients. Radiother Oncol 2021; 164:146-154. [PMID: 34592360 DOI: 10.1016/j.radonc.2021.09.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/13/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has the potential to noninvasively detect expression of hypoxia inducible factor-1-alpha (HIF-1α), epidermal growth factor receptor (EGFR), and Ki-67 in nasopharyngeal carcinoma (NPC) by quantitatively measuring tumor blood flow, vascularity, and permeability. PURPOSE We aim to explore the utility of DCE-MRI in detecting HIF-1α, EGFR, and Ki-67 expression levels using traditional Kety's/Tofts' modeling and quantitative transport mapping (QTM). MATERIALS AND METHODS Eighty-nine NPC patients underwent DCE-MRI before treatment were enrolled. DCE-MRI was processed to generate the following kinetic parameters: |u| and D from the QTM model, tumor blood flow (TBF) from Kety's model, and Ktrans, Ve, and Kep from Tofts' model. Pretreatment levels of HIF-1α, EGFR, and Ki-67 were assessed by immunohistochemistry and classified into low and high expression groups. RESULTS |u| (p < 0.001) and TBF (p = 0.015) values were significantly higher in the HIF-1α high-expression group compared to low-expression group. Only Ktrans (p = 0.016) was significantly higher in the EGFR high-expression group. Only |u| (p < 0.001) values were significantly higher in the Ki-67 high-expression group compared to low-expression group. Multiple linear regression analyses showed that |u| independently correlated with HIF-1α and Ki-67 expression, and Ktrans independently correlated with EGFR. The areas under the ROC curves of |u| for HIF-1α and Ki-67, and Ktrans for EGFR were 0.83, 0.74, and 0.70, respectively. CONCLUSION |u| and Ktrans derived from DCE-MRI may be considered as noninvasive imaging markers for detecting hypoxia and proliferation in NPC patients.
Collapse
Affiliation(s)
- Weiyuan Huang
- Department of Radiology, Weill Medical College of Cornell University, New York, USA; Department of Radiology, Hainan General Hospital (Affiliated Hainan Hospital of Hainan Medical University), China.
| | - Qihao Zhang
- Department of Radiology, Weill Medical College of Cornell University, New York, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, USA
| | - Gang Wu
- Department of Radiotherapy, Hainan General Hospital (Affiliated Hainan Hospital of Hainan Medical University), China
| | - Pian Pian Chen
- Department of Pathology, Hainan General Hospital (Affiliated Hainan Hospital of Hainan Medical University), China
| | - Jiao Li
- Department of Pathology, Hainan General Hospital (Affiliated Hainan Hospital of Hainan Medical University), China
| | - Kelly McCabe Gillen
- Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Pascal Spincemaille
- Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Gloria C Chiang
- Departments of Radiology, Weill Medical College of Cornell University/New York-Presbyterian Hospital, New York, USA
| | - Ajay Gupta
- Departments of Radiology, Weill Medical College of Cornell University/New York-Presbyterian Hospital, New York, USA
| | - Yi Wang
- Department of Radiology, Weill Medical College of Cornell University, New York, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, USA.
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Affiliated Hainan Hospital of Hainan Medical University), China.
| |
Collapse
|
10
|
Danesi R, Fogli S, Indraccolo S, Del Re M, Dei Tos AP, Leoncini L, Antonuzzo L, Bonanno L, Guarneri V, Pierini A, Amunni G, Conte P. Druggable targets meet oncogenic drivers: opportunities and limitations of target-based classification of tumors and the role of Molecular Tumor Boards. ESMO Open 2021; 6:100040. [PMID: 33540286 PMCID: PMC7859305 DOI: 10.1016/j.esmoop.2020.100040] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The therapeutic landscape of cancer is changing rapidly due to the growing number of approved drugs capable of targeting specific genetic alterations. This aspect, together with the development of noninvasive methods for the assessment of somatic mutations in the peripheral blood of patients, generated a growing interest toward a new tumor-agnostic classification system based on ‘predictive’ biomarkers. The current review article discusses this emerging alternative approach to the classification of cancer and its implications for the selection of treatments. It is suggested that different types of cancers sharing the same molecular profiles could benefit from the same targeted drugs. Although recent clinical trials have demonstrated that this approach cannot be generalized, there are also specific examples that demonstrate the clinical utility of this alternative vision. In this rapidly evolving scenario, a multidisciplinary approach managed by institutional Molecular Tumor Boards is fundamental to interpret the biological and clinical relevance of genetic alterations and the complexity of their relationship with treatment response. The identification of oncogenic drivers offers the opportunity to develop target-specific drugs. The inhibition of crucial pathways realizes the principle of druggable target to exploit cancer vulnerability. The approval of new anticancer agents based on target-based concept represents a paradigm shift in cancer therapy. However, only few drugs have been approved so far on an agnostic basis and the concept of biomarker cannot be generalized. Tumor Molecular Boards will have an increasing role in the identification of new therapeutic options in selected patients.
Collapse
Affiliation(s)
- R Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Fogli
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Indraccolo
- Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - M Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A P Dei Tos
- Department of Medicine, School of Medicine, University of Padua, Padua, Italy
| | - L Leoncini
- Department of Medical Biotechnology, Anatomic Pathology Division, University of Siena, Siena, Italy
| | - L Antonuzzo
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - L Bonanno
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - V Guarneri
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - A Pierini
- Integrated Access, Roche, Monza, Italy
| | - G Amunni
- Institute for the Study, Prevention and Oncology Network (ISPRO), Florence, Italy.
| | - P Conte
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| |
Collapse
|
11
|
Kirtonia A, Sethi G, Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci 2020; 77:4459-4483. [PMID: 32358622 PMCID: PMC11105050 DOI: 10.1007/s00018-020-03536-5] [Citation(s) in RCA: 258] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/29/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
Abstract
Redox homeostasis is an essential requirement of the biological systems for performing various normal cellular functions including cellular growth, differentiation, senescence, survival and aging in humans. The changes in the basal levels of reactive oxygen species (ROS) are detrimental to cells and often lead to several disease conditions including cardiovascular, neurological, diabetes and cancer. During the last two decades, substantial research has been done which clearly suggests that ROS are essential for the initiation, progression, angiogenesis as well as metastasis of cancer in several ways. During the last two decades, the potential of dysregulated ROS to enhance tumor formation through the activation of various oncogenic signaling pathways, DNA mutations, immune escape, tumor microenvironment, metastasis, angiogenesis and extension of telomere has been discovered. At present, surgery followed by chemotherapy and/or radiotherapy is the major therapeutic modality for treating patients with either early or advanced stages of cancer. However, the majority of patients relapse or did not respond to initial treatment. One of the reasons for recurrence/relapse is the altered levels of ROS in tumor cells as well as in cancer-initiating stem cells. One of the critical issues is targeting the intracellular/extracellular ROS for significant antitumor response and relapse-free survival. Indeed, a large number of FDA-approved anticancer drugs are efficient to eliminate cancer cells and drug resistance by increasing ROS production. Thus, the modulation of oxidative stress response might represent a potential approach to eradicate cancer in combination with FDA-approved chemotherapies, radiotherapies as well as immunotherapies.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Campus, Sector-125, Noida, Uttar Pradesh, 201313, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Campus, Sector-125, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
12
|
Del Re M, Cucchiara F, Petrini I, Fogli S, Passaro A, Crucitta S, Attili I, De Marinis F, Chella A, Danesi R. erbB in NSCLC as a molecular target: current evidences and future directions. ESMO Open 2020; 5:e000724. [PMID: 32820012 PMCID: PMC7443272 DOI: 10.1136/esmoopen-2020-000724] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
A number of treatments have been developed for HER1, 2 and 3-driven non-small cell lung cancer (NSCLC), of which the most successful have been the epidermal growth factor receptor-tyrosine kinase inhibitors in HER1-mutant tumours resulting in highly improved progression-free survival. Human epidermal growth factor (HER)2 and 3-driven tumours represent the minority of NSCLC, and effective therapies in these patients still represent an unmet medical need. The encouraging results seen with anti-HER2 and anti-HER3 monoclonal antibodies need to be validated in larger studies, even if the greatest obstacle is represented by the exiguous number of patients bearing deregulated HER2/3 system and abnormalities of signal transduction pathway. Considering NSCLC tumour heterogeneity, which affects response and resistance to treatment, combined multiparametric approaches, such as liquid biopsy together with radiomics, may provide a better understanding of the tumour dynamics and clonal selection during the treatments.
Collapse
Affiliation(s)
- Marzia Del Re
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Federico Cucchiara
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Stefano Fogli
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | - Stefania Crucitta
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Ilaria Attili
- Division of Thoracic Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | - Antonio Chella
- Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
13
|
Wang MN, Kuang Y, Gong LY, Hua Y, Pei Q, Guo CX, Cao Y, Huang J, Yang GP. First-in-human, phase I single-ascending-dose study of the safety, pharmacokinetics, and relative bioavailability of selatinib, a dual EGFR-ErbB2 inhibitor in healthy subjects. Invest New Drugs 2020; 38:1826-1835. [PMID: 32535812 PMCID: PMC7575490 DOI: 10.1007/s10637-020-00959-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023]
Abstract
We assessed the pharmacokinetics and safety of a single oral administration of selatinib to healthy Chinese subjects and evaluated the potential bioavailability advantage of selatinib relative to lapatinib. Healthy subjects aged 18-40 years were enrolled in this two-part study: Part 1, a single ascending dose (50-500 mg), randomized, double-blind, placebo-control study with 64 subjects; and Part 2, an open-label, positive control, randomized, three-treatment, three-period, three-sequence crossover design study, with 6 subjects administered a single 500-mg dose of selatinib tablets (A), selatinib suspension (B), or lapatinib tablets C) per cycle. In part 1, selatinib was well-tolerated up to the planned maximum dose of 500 mg; thus the maximum tolerated dose was not attained. Twenty-two adverse events were observed in 19 (36.5%) of the 52 subjects administered the test drug. The most common drug-related adverse event was diarrhea. The mean selatinib peak plasma concentration was 69.4-494 ng/mL, which was achieved in a median peak time of 3.5-4.5 h, with a mean elimination half-life between 13.8 and 15.8 h. In Part 2, A and B showed similar bioavailability. Plasma exposure to the active drug (selatinib plus the metabolite, lapatinib) after A intake was more than two-fold higher than that of the same dose of C. In the dose range of 50-500 mg, selatinib was safe and well-tolerated by healthy Chinese subjects, and it conformed with linear pharmacokinetics. Active exposure to selatinib was much greater than that to lapatinib, supporting its development as an adjuvant for anticancer treatment.
Collapse
Affiliation(s)
- Meng-Na Wang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yun Kuang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Li-Ying Gong
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China.,Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Ye Hua
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Qi Pei
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Cheng-Xian Guo
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yu Cao
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Jie Huang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China.
| | - Guo-Ping Yang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
14
|
Plant lectins and their usage in preparing targeted nanovaccines for cancer immunotherapy. Semin Cancer Biol 2020; 80:87-106. [PMID: 32068087 DOI: 10.1016/j.semcancer.2020.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/30/2020] [Accepted: 02/06/2020] [Indexed: 01/06/2023]
Abstract
Plant lectins, a natural source of glycans with a therapeutic potential may lead to the discovery of new targeted therapies. Glycans extracted from plant lectins are known to act as ligands for C-type lectin receptors (CLRs) that are primarily present on immune cells. Plant-derived glycosylated lectins offer diversity in their N-linked oligosaccharide structures that can serve as a unique source of homogenous and heterogenous glycans. Among the plant lectins-derived glycan motifs, Man9GlcNAc2Asn exhibits high-affinity interactions with CLRs that may resemble glycan motifs of pathogens. Thus, such glycan domains when presented along with antigens complexed with a nanocarrier of choice may bewilder the immune cells and direct antigen cross-presentation - a cytotoxic T lymphocyte immune response mediated by CD8+ T cells. Glycan structure analysis has attracted considerable interest as glycans are looked upon as better therapeutic alternatives than monoclonal antibodies due to their cost-effectiveness, reduced toxicity and side effects, and high specificity. Furthermore, this approach will be useful to understand whether the multivalent glycan presentation on the surface of nanocarriers can overcome the low-affinity lectin-ligand interaction and thereby modulation of CLR-dependent immune response. Besides this, understanding how the heterogeneity of glycan structure impacts the antigen cross-presentation is pivotal to develop alternative targeted therapies. In the present review, we discuss the findings on structural analysis of glycans from natural lectins performed using GlycanBuilder2 - a software tool based on a thorough literature review of natural lectins. Additionally, we discuss how multiple parameters like the orientation of glycan ligands, ligand density, simultaneous targeting of multiple CLRs and design of antigen delivery nanocarriers may influence the CLR targeting efficacy. Integrating this information will eventually set the ground for new generation immunotherapeutic vaccine design for the treatment of various human malignancies.
Collapse
|
15
|
Floris A, Luo J, Frank J, Zhou J, Orrù S, Biancolella M, Pucci S, Orlandi A, Campagna P, Balzano A, Ramani K, Tomasi ML. Star-related lipid transfer protein 10 (STARD10): a novel key player in alcohol-induced breast cancer progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:4. [PMID: 30611309 PMCID: PMC6321732 DOI: 10.1186/s13046-018-1013-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/17/2018] [Indexed: 01/30/2023]
Abstract
Background Ethanol abuse promotes breast cancer development, metastasis and recurrence stimulating mammary tumorigenesis by mechanisms that remain unclear. Normally, 35% of breast cancer is Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2)-positive that predisposes to poor prognosis and relapse, while ethanol drinking leads to invasion of their ERBB2 positive cells triggering the phosphorylation status of mitogen-activated protein kinase. StAR-related lipid transfer protein 10 (STARD10) is a lipid transporter of phosphatidylcholine (PC) and phosphatidylethanolamine (PE); changes on membrane composition of PC and PE occur before the morphological tumorigenic events. Interestingly, STARD10 has been described to be highly expressed in 35–40% of ERBB2-positive breast cancers. In this study, we demonstrate that ethanol administration promotes STARD10 and ERBB2 expression that is significantly associated with increased cell malignancy and aggressiveness. Material and methods We investigated the effect of ethanol on STARD10-ERBB2 cross-talk in breast cancer cells, MMTV-neu transgenic mice and in clinical ERBB2-positive breast cancer specimens with Western Blotting and Real-time PCR. We also examined the effects of their knockdown and overexpression on transient transfected breast cancer cells using promoter activity, MTT, cell migration, calcium and membrane fluidity assays in vitro. Results Ethanol administration induces STARD10 and ERBB2 expression in vitro and in vivo. ERBB2 overexpression causes an increase in STARD10 expression, while overexpression of ERBB2’s downstream targets, p65, c-MYC, c-FOS or c-JUN induces STARD10 promoter activity, correlative of enhanced ERBB2 function. Ethanol and STARD10-mediated cellular membrane fluidity and intracellular calcium concentration impact ERBB2 signaling pathway as evaluated by enhanced p65 nuclear translocation and binding to both ERBB2 and STARD10 promoters. Conclusion Our finding proved that STARD10 and ERBB2 positively regulate each other’s expression and function. Taken together, our data demonstrate that ethanol can modulate ERBB2’s function in breast cancer via a novel interplay with STARD10. Electronic supplementary material The online version of this article (10.1186/s13046-018-1013-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea Floris
- Department of Medicine, Cedars-Sinai Medical Center, DAVIS Research Building 3096A, 8700 Beverly Blv, Los Angeles, CA, 90048, USA.,Medical Genetics, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jacqueline Frank
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jennifer Zhou
- Department of Medicine, Cedars-Sinai Medical Center, DAVIS Research Building 3096A, 8700 Beverly Blv, Los Angeles, CA, 90048, USA
| | - Sandro Orrù
- Medical Genetics, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Sabina Pucci
- Department of Biology, University of Tor Vergata, Rome, Italy.,Department of Biomedicine, University of Tor Vergata, Rome, Italy
| | - Augusto Orlandi
- Department of Biology, University of Tor Vergata, Rome, Italy.,Department of Biomedicine, University of Tor Vergata, Rome, Italy
| | - Paolo Campagna
- Casa di Cura Polispecialistica Sant'Elena, Quartu, Italy
| | | | - Komal Ramani
- Department of Medicine, Cedars-Sinai Medical Center, DAVIS Research Building 3096A, 8700 Beverly Blv, Los Angeles, CA, 90048, USA
| | - Maria Lauda Tomasi
- Department of Medicine, Cedars-Sinai Medical Center, DAVIS Research Building 3096A, 8700 Beverly Blv, Los Angeles, CA, 90048, USA.
| |
Collapse
|