1
|
Yanjun Y, Jing Z, Yifei S, Gangzhao G, Chenxin Y, Qiang W, Qiang Y, Shuwen H. Trace elements in pancreatic cancer. Cancer Med 2024; 13:e7454. [PMID: 39015024 PMCID: PMC11252496 DOI: 10.1002/cam4.7454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PCA) is an extremely aggressive malignant cancer with an increasing incidence and a low five-year survival rate. The main reason for this high mortality is that most patients are diagnosed with PCA at an advanced stage, missing early treatment options and opportunities. As important nutrients of the human body, trace elements play an important role in maintaining normal physiological functions. Moreover, trace elements are closely related to many diseases, including PCA. REVIEW This review systematically summarizes the latest research progress on selenium, copper, arsenic, and manganese in PCA, elucidates their application in PCA, and provides a new reference for the prevention, diagnosis and treatment of PCA. CONCLUSION Trace elements such as selenium, copper, arsenic and manganese are playing an important role in the risk, pathogenesis, diagnosis and treatment of PCA. Meanwhile, they have a certain inhibitory effect on PCA, the mechanism mainly includes: promoting ferroptosis, inducing apoptosis, inhibiting metastasis, and inhibiting excessive proliferation.
Collapse
Affiliation(s)
- Yao Yanjun
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Zhuang Jing
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Song Yifei
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Gu Gangzhao
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Yan Chenxin
- Shulan International Medical schoolZhejiang Shuren UniversityHangzhouChina
| | - Wei Qiang
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Yan Qiang
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
- Institut Catholique de Lille, Junia (ICL), Université Catholique de Lille, Laboratoire Interdisciplinaire des Transitions de Lille (LITL)LilleFrance
| |
Collapse
|
2
|
Haberkorn B, Löwen D, Meier L, Fromm MF, König J. Transcriptional Regulation of Liver-Type OATP1B3 (Lt-OATP1B3) and Cancer-Type OATP1B3 (Ct-OATP1B3) Studied in Hepatocyte-Derived and Colon Cancer-Derived Cell Lines. Pharmaceutics 2023; 15:pharmaceutics15030738. [PMID: 36986600 PMCID: PMC10051083 DOI: 10.3390/pharmaceutics15030738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Due to alternative splicing, the SLCO1B3 gene encodes two protein variants; the hepatic uptake transporter liver-type OATP1B3 (Lt-OATP1B3) and the cancer-type OATP1B3 (Ct-OATP1B3) expressed in several cancerous tissues. There is limited information about the cell type-specific transcriptional regulation of both variants and about transcription factors regulating this differential expression. Therefore, we cloned DNA fragments from the promoter regions of the Lt-SLCO1B3 and the Ct-SLCO1B3 gene and investigated their luciferase activity in hepatocellular and colorectal cancer cell lines. Both promoters showed differences in their luciferase activity depending on the used cell lines. We identified the first 100 bp upstream of the transcriptional start site as the core promoter region of the Ct-SLCO1B3 gene. In silico predicted binding sites for the transcription factors ZKSCAN3, SOX9 and HNF1α localized within these fragments were further analyzed. The mutagenesis of the ZKSCAN3 binding site reduced the luciferase activity of the Ct-SLCO1B3 reporter gene construct in the colorectal cancer cell lines DLD1 and T84 to 29.9% and 14.3%, respectively. In contrast, using the liver-derived Hep3B cells, 71.6% residual activity could be measured. This indicates that the transcription factors ZKSCAN3 and SOX9 are important for the cell type-specific transcriptional regulation of the Ct-SLCO1B3 gene.
Collapse
Affiliation(s)
| | | | | | | | - Jörg König
- Correspondence: ; Tel.: +49-9131-8522077
| |
Collapse
|
3
|
Jiang H, Wei H, Yang T, Qin Y, Wu Y, Chen W, Shi Y, Ronot M, Bashir MR, Song B. VICT2 Trait: Prognostic Alternative to Peritumoral Hepatobiliary Phase Hypointensity in HCC. Radiology 2023; 307:e221835. [PMID: 36786702 DOI: 10.1148/radiol.221835] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Background Peritumoral hepatobiliary phase (HBP) hypointensity is an established prognostic imaging feature in hepatocellular carcinoma (HCC), often associated with microvascular invasion (MVI). Similar prognostic features are needed for non-HBP MRI. Purpose To propose a non-hepatobiliary-specific MRI tool with similar prognostic value to peritumoral HBP hypointensity. Materials and Methods From December 2011 to November 2021, consecutive patients with HCC who underwent preoperative contrast-enhanced MRI were retrospectively enrolled and followed up until recurrence. All MRI scans were reviewed by two blinded radiologists with 7 and 10 years of experiences with liver MRI. A scoring system based on non-hepatobiliary-specific features that highly correlated with peritumoral HBP hypointensity was identified in a stratified sampling-derived training set of the gadoxetate disodium (EOB) group by means of multivariable logistic regression, and its values to predict MVI and recurrence-free survival (RFS) were assessed. Results There were 660 patients (551 men; median age, 53 years; IQR, 45-61 years) enrolled. Peritumoral portal venous phase hypoenhancement (odds ratio [OR] = 8.8), incomplete "capsule" (OR = 3.3), corona enhancement (OR, 2.6), and peritumoral mild-moderate T2 hyperintensity (OR, 2.2) (all P < .001) were associated with peritumoral HBP hypointensity and constituted the "VICT2 trait" (test set area under the receiver operating characteristic curve = 0.84; 95% CI: 0.78, 0.90). For the EOB group, both peritumoral HBP hypointensity (OR for MVI = 2.5, P = .02; hazard ratio for RFS = 2.5, P < .001) and the VICT2 trait (OR for MVI = 5.1, P < .001; hazard ratio for RFS = 2.3, P < .001) were associated with MVI and RFS, despite a higher specificity of the VICT2 trait for MVI (89% vs 80%, P = .01). These values of the VICT2 trait were confirmed in the extracellular contrast agent group (OR for MVI = 4.0; hazard ratio for RFS = 1.7; both P < .001). Conclusion Based on four non-hepatobiliary-specific MRI features, the VICT2 trait was comparable to peritumoral hepatobiliary phase hypointensity in predicting microvascular invasion and postoperative recurrence of hepatocellular carcinoma. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Harmath in this issue.
Collapse
Affiliation(s)
- Hanyu Jiang
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Hong Wei
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Ting Yang
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yun Qin
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yuanan Wu
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Weixia Chen
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yujun Shi
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Maxime Ronot
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Mustafa R Bashir
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Bin Song
- From the Department of Radiology (H.J., H.W., T.Y., Y.Q., W.C., B.S.) and Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC (Y.S.), West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; Big Data Research Center, University of Electronic Science and Technology of China, Chengdu, China (Y.W.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France (M.R.); Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC (M.R.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| |
Collapse
|
4
|
Koller S, Kendler J, Karacs J, Wolf A, Kreuzinger C, Von Der Decken I, Mungenast F, Mechtcheriakova D, Schreiner W, Gleiss A, Jäger W, Cacsire Castillo-Tong D, Thalhammer T. SLCO4A1 expression is associated with activated inflammatory pathways in high-grade serous ovarian cancer. Front Pharmacol 2022; 13:946348. [PMID: 36105223 PMCID: PMC9465617 DOI: 10.3389/fphar.2022.946348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with high-grade serous ovarian cancer (HGSOC) have a very poor overall survival. Current therapeutic approaches do not bring benefit to all patients. Although genetic alterations and molecular mechanisms are well characterized, the molecular pathological conditions are poorly investigated. Solute carrier organic anion transporter family member 4A1 (SLCO4A1) encodes OATP4A1, which is an uptake membrane transporter of metabolic products. Its expression may influence various signaling pathways associated with the molecular pathophysiological conditions of HGSOC and consequently tumor progression. RNA sequencing of 33 patient-derived HGSOC cell lines showed that SLCO4A1 expression was diverse by individual tumors, which was further confirmed by RT-qPCR, Western blotting and immunohistochemistry. Gene Set Enrichment Analysis revealed that higher SLCO4A1 level was associated with inflammation-associated pathways including NOD-like receptor, adipocytokine, TALL1, CD40, NF-κB, and TNF-receptor 2 signaling cascades, while low SLCO4A1 expression was associated with the mitochondrial electron transport chain pathway. The overall gene expression pattern in all cell lines was specific to each patient and remained largely unchanged during tumor progression. In addition, genes encoding ABCC3 along with SLCO4A1-antisense RNA 1, were associated with higher expression of the SLCO4A1, indicating their possible involvement in inflammation-associated pathways that are downstream to the prostaglandin E2/cAMP axis. Taken together, increased SLCO4A1/OATP4A1 expression is associated with the upregulation of specific inflammatory pathways, while the decreased level is associated with mitochondrial dysfunction. These molecular pathophysiological conditions are tumor specific and should be taken into consideration by the development of therapies against HGSOC.
Collapse
Affiliation(s)
- Stephanie Koller
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jonatan Kendler
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jasmine Karacs
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Andrea Wolf
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Caroline Kreuzinger
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Isabel Von Der Decken
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Felicitas Mungenast
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Schreiner
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Dan Cacsire Castillo-Tong
- Department of Obstetrics and Gynecology, Translational Gynecology Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- *Correspondence: Dan Cacsire Castillo-Tong, ; Theresia Thalhammer,
| | - Theresia Thalhammer
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Dan Cacsire Castillo-Tong, ; Theresia Thalhammer,
| |
Collapse
|
5
|
Zhou S, Shu Y. Transcriptional Regulation of Solute Carrier (SLC) Drug Transporters. Drug Metab Dispos 2022; 50:DMD-MR-2021-000704. [PMID: 35644529 PMCID: PMC9488976 DOI: 10.1124/dmd.121.000704] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 05/02/2022] [Accepted: 05/16/2022] [Indexed: 09/03/2023] Open
Abstract
Facilitated transport is necessitated for large size, charged, and/or hydrophilic drugs to move across the membrane. The drug transporters in the solute carrier (SLC) superfamily, mainly including organic anion-transporting polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), organic cation/carnitine transporters (OCTNs), peptide transporters (PEPTs), and multidrug and toxin extrusion proteins (MATEs), are critical facilitators of drug transport and distribution in human body. The expression of these SLC drug transporters is found in tissues throughout the body, with high abundance in the epithelial cells of major organs for drug disposition, such as intestine, liver, and kidney. These SLC drug transporters are clinically important in drug absorption, metabolism, distribution, and excretion. The mechanisms underlying their regulation have been revealing in recent years. Epigenetic and nuclear receptor-mediated transcriptional regulation of SLC drug transporters have particularly attracted much attention. This review focuses on the transcriptional regulation of major SLC drug transporter genes. Revealing the mechanisms underlying the transcription of those critical drug transporters will help us understand pharmacokinetics and pharmacodynamics, ultimately improving drug therapeutic effectiveness while minimizing drug toxicity. Significance Statement It has become increasingly recognized that solute carrier (SLC) drug transporters play a crucial, and sometimes determinative, role in drug disposition and response, which is reflected in decision-making during not only clinical drug therapy but also drug development. Understanding the mechanisms accounting for the transcription of these transporters is critical to interpret their abundance in various tissues under different conditions, which is necessary to clarify the pharmacological response, adverse effects, and drug-drug interactions for clinically used drugs.
Collapse
Affiliation(s)
- Shiwei Zhou
- Pharmaceutical Sciences, University of Maryland, United States
| | - Yan Shu
- Pharmaceutical Sciences, University of Maryland, United States
| |
Collapse
|
6
|
SLCO1B3 promotes colorectal cancer tumorigenesis and metastasis through STAT3. Aging (Albany NY) 2021; 13:22164-22175. [PMID: 34526411 PMCID: PMC8507254 DOI: 10.18632/aging.203502] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022]
Abstract
Solute carrier organic anion transporter family member 1B3 (SLCO1B3) is a gene that encodes an organic anion-transporting polypeptide (OATP) 1B3, a membrane-bound multi-specific transporter in hepatocytes. SLCO1B3 was first reported in hepatocytes. Later, it was found that its expression is higher in colorectal cancer (CRC) than in the adjacent normal tissue. However, the role of SLCO1B3 in CRC is not well elucidated. In this study, the correlation between SLCO1B3 and the overall survival (OS) of CRC patients was evaluated using data from the GEO database. This study evaluated the relationship between SLCO1B3 and the clinicopathological characteristics and prognosis of CRC patients. The effects of SLCO1B3 knockdown, on human CRC cell proliferation, migration, and invasion in vitro and CRC tumorigenesis and metastasis in vivo were also examined. In addition, next-generation sequencing was used to identify SLCO1B3 mediators. The results confirmed the association between SLCO1B3 and poor OS of CRC patients, and SLCO1B3 was identified as the top hub gene associated with the OS. The study showed that high SLCO1B3 expression was associated with poor tumor differentiation, advanced disease stage, tumor invasion, lymph node metastasis, and poor OS. Next-generation sequencing revealed that SLCO1B3 knockdown affected the expression of several genes involved in cancer invasion, metastasis, and DNA repair. Moreover, the western blot analysis showed that SLCO1B3 knockdown downregulated p-STAT3, MMP-2, and MMP-9. In summary, we demonstrated that SLCO1B3 acts as a novel carcinogen in the CRC that drives the CRC tumorigenesis and metastasis. SLCO1B3 inhibitors, alone or in combination with current drugs, may have therapeutic benefits in CRC.
Collapse
|
7
|
Organic Anion Transporting Polypeptide 1B1 Is a Potential Reporter for Dual MR and Optical Imaging. Int J Mol Sci 2021; 22:ijms22168797. [PMID: 34445497 PMCID: PMC8395777 DOI: 10.3390/ijms22168797] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 11/16/2022] Open
Abstract
Membrane proteins responsible for transporting magnetic resonance (MR) and fluorescent contrast agents are of particular importance because they are potential reporter proteins in noninvasive molecular imaging. Gadobenate dimeglumine (Gd-BOPTA), a liver-specific MR contrast agent, has been used globally for more than 10 years. However, the corresponding molecular transportation mechanism has not been validated. We previously reported that the organic anion transporting polypeptide (OATP) 1B3 has an uptake capability for both MR agents (Gd-EOB-DTPA) and indocyanine green (ICG), a clinically available near-infrared (NIR) fluorescent dye. This study further evaluated OATP1B1, another polypeptide of the OATP family, to determine its reporter capability. In the OATP1B1 transfected 293T transient expression model, both Gd-BOPTA and Gd-EOB-DTPA uptake were confirmed through 1.5 T MR imaging. In the constant OAPT1B1 and OATP1B3 expression model in the HT-1080 cell line, both HT-1080-OAPT1B1 and HT-1080-OATP1B3 were observed to ingest Gd-BOPTA and Gd-EOB-DTPA. Lastly, we validated the ICG uptake capability of both OATP1B1 and OATP1B3. OAPT1B3 exhibited a superior ICG uptake capability to that of OAPT1B1. We conclude that OATP1B1 is a potential reporter for dual MR and NIR fluorescent molecular imaging, especially in conjunction with Gd-BOPTA.
Collapse
|
8
|
Barbier RH, McCrea EM, Lee KY, Strope JD, Risdon EN, Price DK, Chau CH, Figg WD. Abiraterone induces SLCO1B3 expression in prostate cancer via microRNA-579-3p. Sci Rep 2021; 11:10765. [PMID: 34031488 PMCID: PMC8144422 DOI: 10.1038/s41598-021-90143-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/30/2021] [Indexed: 11/25/2022] Open
Abstract
Understanding mechanisms of resistance to abiraterone, one of the primary drugs approved for the treatment of castration resistant prostate cancer, remains a priority. The organic anion polypeptide 1B3 (OATP1B3, encoded by SLCO1B3) transporter has been shown to transport androgens into prostate cancer cells. In this study we observed and investigated the mechanism of induction of SLCO1B3 by abiraterone. Prostate cancer cells (22Rv1, LNCaP, and VCAP) were treated with anti-androgens and assessed for SLCO1B3 expression by qPCR analysis. Abiraterone treatment increased SLCO1B3 expression in 22Rv1 cells in vitro and in the 22Rv1 xenograft model in vivo. MicroRNA profiling of abiraterone-treated 22Rv1 cells was performed using a NanoString nCounter miRNA panel followed by miRNA target prediction. TargetScan and miRanda prediction tools identified hsa-miR-579-3p as binding to the 3'-untranslated region (3'UTR) of the SLCO1B3. Using dual luciferase reporter assays, we verified that hsa-miR-579-3p indeed binds to the SLCO1B3 3'UTR and significantly inhibited SLCO1B3 reporter activity. Treatment with abiraterone significantly downregulated hsa-miR-579-3p, indicating its potential role in upregulating SLCO1B3 expression. In this study, we demonstrated a novel miRNA-mediated mechanism of abiraterone-induced SLCO1B3 expression, a transporter that is also responsible for driving androgen deprivation therapy resistance. Understanding mechanisms of abiraterone resistance mediated via differential miRNA expression will assist in the identification of potential miRNA biomarkers of treatment resistance and the development of future therapeutics.
Collapse
Affiliation(s)
- Roberto H Barbier
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - Edel M McCrea
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - Kristi Y Lee
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - Jonathan D Strope
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - Emily N Risdon
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - Douglas K Price
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - Cindy H Chau
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA
| | - William D Figg
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 5A03, Bethesda, MD, 20892, USA.
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
9
|
Low expression of organic anion-transporting polypeptide 1B3 predicts a poor prognosis in hepatocellular carcinoma. World J Surg Oncol 2020; 18:127. [PMID: 32534581 PMCID: PMC7293789 DOI: 10.1186/s12957-020-01891-y] [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: 01/02/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022] Open
Abstract
Objective To detect the expression level of organic anion-transporting polypeptide 1B3 (OATP1B3) in hepatocellular carcinoma (HCC) and to determine the relationship between OATP1B3 expression, clinicopathological features, and prognosis. Methods Immunohistochemical (IHC) staining was performed to detect the expression of OATP1B3 in 131 HCC specimens and in 89 adjacent nontumorous tissues. Moreover, the expression levels of OATP1B3 in 30 pairs of tumor and matched adjacent nontumorous tissues were detected by quantitative real-time polymerase chain reaction, and 34 pairs of tumor and matched adjacent nontumorous tissues were detected by Western blotting. The χ2 test was applied to analyze the correlation between OATP1B3 expression and the clinical parameters of HCC patients. The prognostic value of OATP1B3 in HCC patients was estimated by Kaplan-Meier survival analysis and the Cox stepwise proportional hazards model. Results Compared with that in adjacent nontumorous tissues (25.8%, 23/89), OATP1B3 expression was significantly downregulated in tumor tissues (59.5%, 78/131) (P < 0.0001). Moreover, OATP1B3 expression was markedly correlated with tumor size, recurrence, tumor differentiation, and tumor node metastasis (TNM) stage (P < 0.05 for each). However, age, sex, tumor capsule status, HBsAg, cirrhosis, tumor number, vascular invasion, and serum alpha fetoprotein were not associated with OATP1B3 expression. The overall survival (OS) and disease-free survival (DFS) of HCC patients who had high expression of OATP1B3 were significantly longer than those of patients with low expression (33.0% vs 12.9%, P = 0.001; 18.8% vs 5.3%, P < 0.0001). Cox multivariate analysis showed that OATP1B3, invasion, and TNM stage (P < 0.05 for each) were independent prognostic factors of OS in HCC patients and that OATP1B3 and TNM stage (both P < 0.05) were independent prognostic factors of DFS in HCC patients. Conclusions The expression of OATP1B3 in HCC patients was significantly lower than that in adjacent nontumorous tissues. OATP1B3 expression may be a potential prognostic marker in HCC patients.
Collapse
|
10
|
Sun R, Ying Y, Tang Z, Liu T, Shi F, Li H, Guo T, Huang S, Lai R. The Emerging Role of the SLCO1B3 Protein in Cancer Resistance. Protein Pept Lett 2020; 27:17-29. [PMID: 31556849 PMCID: PMC6978646 DOI: 10.2174/0929866526666190926154248] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/08/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Currently, chemotherapy is one of the mainstays of oncologic therapies. But the efficacy of chemotherapy is often limited by drug resistance and severe side effects. Consequently, it is becoming increasingly important to investigate the underlying mechanism and overcome the problem of anticancer chemotherapy resistance. The solute carrier organic anion transporter family member 1B3 (SLCO1B3), a functional transporter normally expressed in the liver, transports a variety of endogenous and exogenous compounds, including hormones and their conjugates as well as some anticancer drugs. The extrahepatic expression of SLCO1B3 has been detected in different cancer cell lines and cancer tissues. Recently, accumulating data indicates that the abnormal expression and function of SLCO1B3 are involved in resistance to anticancer drugs, such as taxanes, camptothecin and its analogs, SN-38, and Androgen Deprivation Therapy (ADT) in breast, prostate, lung, hepatic, and colorectal cancer, respectively. Thus, more investigations have been implemented to identify the potential SLCO1B3-related mechanisms of cancer drug resistance. In this review, we focus on the emerging roles of SLCO1B3 protein in the development of cancer chemotherapy resistance and briefly discuss the mechanisms of resistance. Elucidating the function of SLCO1B3 in chemoresistance may bring out novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Ruipu Sun
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China.,Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Zhimin Tang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Ting Liu
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Fuli Shi
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Huixia Li
- Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Taichen Guo
- Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Shibo Huang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Pharmacy, Medical College, Nanchang University, Nanchang 330006, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences / Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| |
Collapse
|
11
|
Schulte RR, Ho RH. Organic Anion Transporting Polypeptides: Emerging Roles in Cancer Pharmacology. Mol Pharmacol 2019; 95:490-506. [PMID: 30782852 DOI: 10.1124/mol.118.114314] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/09/2019] [Indexed: 12/13/2022] Open
Abstract
The organic anion transporting polypeptides (OATPs) are a superfamily of drug transporters involved in the uptake and disposition of a wide array of structurally divergent endogenous and exogenous substrates, including steroid hormones, bile acids, and commonly used drugs, such as anti-infectives, antihypertensives, and cholesterol lowering agents. In the past decade, OATPs, primarily OATP1A2, OATP1B1, and OATP1B3, have emerged as potential mediators of chemotherapy disposition, including drugs such as methotrexate, doxorubicin, paclitaxel, docetaxel, irinotecan and its important metabolite 7-ethyl-10-hydroxycamptothecin, and certain tyrosine kinase inhibitors. Furthermore, OATP family members are polymorphic and numerous studies have shown OATP variants to have differential uptake, disposition, and/or pharmacokinetics of numerous drug substrates with important implications for interindividual differences in efficacy and toxicity. Additionally, certain OATPs have been found to be overexpressed in a variety of human solid tumors, including breast, liver, colon, pancreatic, and ovarian cancers, suggesting potential roles for OATPs in tumor development and progression and as novel targets for cancer therapy. This review focuses on the emerging roles for selected OATPs in cancer pharmacology, including preclinical and clinical studies suggesting roles in chemotherapy disposition, the pharmacogenetics of OATPs in cancer therapy, and OATP overexpression in various tumor tissues with implications for OATPs as therapeutic targets.
Collapse
Affiliation(s)
- Rachael R Schulte
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Richard H Ho
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
12
|
Svoboda M, Mungenast F, Gleiss A, Vergote I, Vanderstichele A, Sehouli J, Braicu E, Mahner S, Jäger W, Mechtcheriakova D, Cacsire-Tong D, Zeillinger R, Thalhammer T, Pils D. Clinical Significance of Organic Anion Transporting Polypeptide Gene Expression in High-Grade Serous Ovarian Cancer. Front Pharmacol 2018; 9:842. [PMID: 30131693 PMCID: PMC6090214 DOI: 10.3389/fphar.2018.00842] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/13/2018] [Indexed: 12/31/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is considered the most deadly and frequently occurring type of ovarian cancer and is associated with various molecular compositions and growth patterns. Evaluating the mRNA expression pattern of the organic anion transporters (OATPs) encoded by SLCO genes may allow for improved stratification of HGSOC patients for targeted invention. The expression of SLCO mRNA and genes coding for putative functionally related ABC-efflux pumps, enzymes, pregnane-X-receptor, ESR1 and ESR2 (coding for estrogen receptors ERα and ERß) and HER-2 were assessed using RT-qPCR. The expression levels were assessed in a cohort of 135 HGSOC patients to elucidate the independent impact of the expression pattern on the overall survival (OS). For identification of putative regulatory networks, Graphical Gaussian Models were constructed from the expression data with a tuning parameter K varying between meaningful borders (Pils et al., 2012; Auer et al., 2015, 2017; Kurman and Shih Ie, 2016; Karam et al., 2017; Labidi-Galy et al., 2017; Salomon-Perzynski et al., 2017; Sukhbaatar et al., 2017). The final value used (K = 4) was determined by maximizing the proportion of explained variation of the corresponding LASSO Cox regression model for OS. The following two networks of directly correlated genes were identified: (i) SLCO2B1 with ABCC3 implicated in estrogen homeostasis; and (ii) two ABC-efflux pumps in the immune regulation (ABCB2/ABCB3) with ABCC3 and HER-2. Combining LASSO Cox regression and univariate Cox regression analyses, SLCO5A1 coding for OATP5A1, an estrogen metabolite transporter located in the cytoplasm and plasma membranes of ovarian cancer cells, was identified as significant and independent prognostic factor for OS (HR = 0.68, CI 0.49-0.93; p = 0.031). Furthermore, results indicated the benefits of patients with high expression by adding 5.1% to the 12.8% of the proportion of explained variation (PEV) for clinicopathological parameters known for prognostic significance (FIGO stage, age and residual tumor after debulking). Additionally, overlap with previously described signatures that indicated a more favorable prognosis for ovarian cancer patients was shown for SLCO5A1, the network ABCB2/ABCB3/ABCC4/HER2 as well as ESR1. Furthermore, expression of SLCO2A1 and PGDH, which are important for PGE2 degradation, was associated with the non-miliary peritoneal tumor spreading. In conclusion, the present findings suggested that SLCOs and the related molecules identified as potential biomarkers in HGSOC may be useful for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Martin Svoboda
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Felicitas Mungenast
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Institute of Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Ignace Vergote
- Division of Gynaecological Oncology, Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospital Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Adriaan Vanderstichele
- Division of Gynaecological Oncology, Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospital Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jalid Sehouli
- Department of Gynecology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Elena Braicu
- Department of Gynecology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sven Mahner
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dan Cacsire-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynaecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Robert Zeillinger
- Molecular Oncology Group, Department of Obstetrics and Gynaecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Theresia Thalhammer
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Institute of Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria.,Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Alam K, Crowe A, Wang X, Zhang P, Ding K, Li L, Yue W. Regulation of Organic Anion Transporting Polypeptides (OATP) 1B1- and OATP1B3-Mediated Transport: An Updated Review in the Context of OATP-Mediated Drug-Drug Interactions. Int J Mol Sci 2018. [PMID: 29538325 PMCID: PMC5877716 DOI: 10.3390/ijms19030855] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Organic anion transporting polypeptides (OATP) 1B1 and OATP1B3 are important hepatic transporters that mediate the uptake of many clinically important drugs, including statins from the blood into the liver. Reduced transport function of OATP1B1 and OATP1B3 can lead to clinically relevant drug-drug interactions (DDIs). Considering the importance of OATP1B1 and OATP1B3 in hepatic drug disposition, substantial efforts have been given on evaluating OATP1B1/1B3-mediated DDIs in order to avoid unwanted adverse effects of drugs that are OATP substrates due to their altered pharmacokinetics. Growing evidences suggest that the transport function of OATP1B1 and OATP1B3 can be regulated at various levels such as genetic variation, transcriptional and post-translational regulation. The present review summarizes the up to date information on the regulation of OATP1B1 and OATP1B3 transport function at different levels with a focus on potential impact on OATP-mediated DDIs.
Collapse
Affiliation(s)
- Khondoker Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Xueying Wang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Pengyue Zhang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kai Ding
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA.
| | - Lang Li
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA.
| | - Wei Yue
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| |
Collapse
|
14
|
Park JE, Ryoo G, Lee W. Alternative Splicing: Expanding Diversity in Major ABC and SLC Drug Transporters. AAPS JOURNAL 2017; 19:1643-1655. [DOI: 10.1208/s12248-017-0150-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/10/2017] [Indexed: 01/18/2023]
|
15
|
Sissung TM, Ley AM, Strope JD, McCrea EM, Beedie S, Peer CJ, Shukla S, van Velkinburgh J, Reece K, Troutman S, Campbell T, Fernandez E, Huang P, Smith J, Thakkar N, Venzon DJ, Brenner S, Lee W, Merino M, Luo J, Jager W, Price DK, Chau CH, Figg WD. Differential Expression of OATP1B3 Mediates Unconjugated Testosterone Influx. Mol Cancer Res 2017; 15:1096-1105. [PMID: 28389619 PMCID: PMC5540879 DOI: 10.1158/1541-7786.mcr-16-0477] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/06/2017] [Accepted: 04/04/2017] [Indexed: 12/21/2022]
Abstract
Castration-resistant prostate cancer (CRPC) has greater intratumoral testosterone concentrations than similar tumors from eugonadal men; simple diffusion does not account for this observation. This study was undertaken to ascertain the androgen uptake kinetics, functional, and clinical relevance of de novo expression of the steroid hormone transporter OATP1B3 (SLCO1B3). Experiments testing the cellular uptake of androgens suggest that testosterone is an excellent substrate of OATP1B3 (Km = 23.2 μmol/L; Vmax = 321.6 pmol/mg/minute), and cells expressing a doxycycline-inducible SLCO1B3 construct had greater uptake of a clinically relevant concentration of 3H-testosterone (50 nmol/L; 1.6-fold, P = 0.0027). When compared with Slco1b2 (-/-) mice, Slco1b2 (-/-)/hSLCO1B3 knockins had greater hepatic uptake (15% greater AUC, P = 0.0040) and lower plasma exposure to 3H-testosterone (17% lower AUC, P = 0.0030). Of 82 transporters genes, SLCO1B3 is the second-most differentially expressed transporter in CRPC cell lines (116-fold vs. androgen-sensitive cells), with a differentially spliced cancer-type ct-SLCO1B3 making up the majority of SLCO1B3 expression. Overexpression of SLCO1B3 in androgen-responsive cells results in 1.5- to 2-fold greater testosterone uptake, whereas siRNA knockdown of SLCO1B3 in CRPC cells did not change intracellular testosterone concentration. Primary human prostate tumors express SLCO1B3 to a greater extent than ct-SLCO1B3 (26% of total SLCO1B3 expression vs. 0.08%), suggesting that androgen uptake in these tumor cells also is greater. Non-liver tumors do not differentially express SLCO1B3.Implications: This study suggests that de novo OATP1B3 expression in prostate cancer drives greater androgen uptake and is consistent with previous observations that greater OATP1B3 activity results in the development of androgen deprivation therapy resistance and shorter overall survival. Mol Cancer Res; 15(8); 1096-105. ©2017 AACR.
Collapse
Affiliation(s)
- Tristan M Sissung
- Clinical Pharmacology Program, Office of the Clinical Director, NCI, Bethesda, Maryland
| | - Ariel M Ley
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Jonathan D Strope
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Edel M McCrea
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Shaunna Beedie
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Cody J Peer
- Clinical Pharmacology Program, Office of the Clinical Director, NCI, Bethesda, Maryland
| | | | | | - Kelie Reece
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Sarah Troutman
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Tessa Campbell
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Elena Fernandez
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Phoebe Huang
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Jordan Smith
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Nilay Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - David J Venzon
- Biostatistics and Data Management Section, NCI, Bethesda, Maryland
| | - Stefan Brenner
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Wooin Lee
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Maria Merino
- Translational Surgical Pathology Section, NCI, Bethesda, Maryland
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Walter Jager
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Douglas K Price
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - Cindy H Chau
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| | - William D Figg
- Clinical Pharmacology Program, Office of the Clinical Director, NCI, Bethesda, Maryland.
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, NCI, Bethesda, Maryland
| |
Collapse
|
16
|
Zhang C, Zhao Y, Zhang H, Chen X, Zhao N, Tan D, Zhang H, Shi C. The Application of Heptamethine Cyanine Dye DZ-1 and Indocyanine Green for Imaging and Targeting in Xenograft Models of Hepatocellular Carcinoma. Int J Mol Sci 2017; 18:E1332. [PMID: 28635650 PMCID: PMC5486152 DOI: 10.3390/ijms18061332] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/07/2017] [Accepted: 06/18/2017] [Indexed: 12/31/2022] Open
Abstract
Near infrared fluorescence (NIRF) imaging has strong potential for widespread use in noninvasive tumor imaging. Indocyanine green (ICG) is the only Food and Drug Administration (FDA) -approved NIRF dye for clinical diagnosis; however, it is unstable and poorly targets tumors. DZ-1 is a novel heptamethine cyanine NIRF dye, suitable for imaging and tumor targeting. Here, we compared the fluorescence intensity and metabolism of DZ-1 and ICG. Additionally, we assayed their specificities and abilities to target tumor cells, using cultured hepatocellular carcinoma (HCC) cell lines, a nude mouse subcutaneous xenograft model of liver cancer, and a rabbit orthotopic transplantation model. We found that DZ-1 accumulates in tumor tissue and specifically recognizes HCC in subcutaneous and orthotopic models. The NIRF intensity of DZ-1 was one order of magnitude stronger than that of ICG, and DZ-1 showed excellent intraoperative tumor targeting in the rabbit model. Importantly, ICG accumulated at tumor sites, as well as in the liver and kidney. Furthermore, DZ-1 analog-gemcitabine conjugate (NIRG) exhibited similar tumor-specific targeting and imaging properties, including inhibition of tumor growth, in HCC patient-derived xenograft (PDX) mice. DZ-1 and NIRG demonstrated superior tumor-targeting specificity, compared to ICG. We show that DZ-1 is an effective molecular probe for specific imaging, targeting, and therapy in HCC.
Collapse
Affiliation(s)
- Caiqin Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - Yong Zhao
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - He Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - Xue Chen
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - Ningning Zhao
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - Dengxu Tan
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - Hai Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| | - Changhong Shi
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
17
|
Thakkar N, Slizgi JR, Brouwer KLR. Effect of Liver Disease on Hepatic Transporter Expression and Function. J Pharm Sci 2017; 106:2282-2294. [PMID: 28465155 DOI: 10.1016/j.xphs.2017.04.053] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/27/2022]
Abstract
Liver disease can alter the disposition of xenobiotics and endogenous substances. Regulatory agencies such as the Food and Drug Administration and the European Medicines Evaluation Agency recommend, if possible, studying the effect of liver disease on drugs under development to guide specific dose recommendations in these patients. Although extensive research has been conducted to characterize the effect of liver disease on drug-metabolizing enzymes, emerging data have implicated that the expression and function of hepatobiliary transport proteins also are altered in liver disease. This review summarizes recent developments in the field, which may have implications for understanding altered disposition, safety, and efficacy of new and existing drugs. A brief review of liver physiology and hepatic transporter localization/function is provided. Then, the expression and function of hepatic transporters in cholestasis, hepatitis C infection, hepatocellular carcinoma, human immunodeficiency virus infection, nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, and primary biliary cirrhosis are reviewed. In the absence of clinical data, nonclinical information in animal models is presented. This review aims to advance the understanding of altered expression and function of hepatic transporters in liver disease and the implications of such changes on drug disposition.
Collapse
Affiliation(s)
- Nilay Thakkar
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jason R Slizgi
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.
| |
Collapse
|
18
|
Murray M, Zhou F. Trafficking and other regulatory mechanisms for organic anion transporting polypeptides and organic anion transporters that modulate cellular drug and xenobiotic influx and that are dysregulated in disease. Br J Pharmacol 2017; 174:1908-1924. [PMID: 28299773 DOI: 10.1111/bph.13785] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/01/2017] [Accepted: 03/05/2017] [Indexed: 12/25/2022] Open
Abstract
Organic anion transporters (OATs) and organic anion-transporting polypeptides (OATPs), encoded by a number of solute carrier (SLC)22A and SLC organic anion (SLCO) genes, mediate the absorption and distribution of drugs and other xenobiotics. The regulation of OATs and OATPs is complex, comprising both transcriptional and post-translational mechanisms. Plasma membrane expression is required for cellular substrate influx by OATs/OATPs. Thus, interest in post-translational regulatory processes, including membrane targeting, endocytosis, recycling and degradation of transporter proteins, is increasing because these are critical for plasma membrane expression. After being synthesized, transporters undergo N-glycosylation in the endoplasmic reticulum and Golgi apparatus and are delivered to the plasma membrane by vesicular transport. Their expression at the cell surface is maintained by de novo synthesis and recycling, which occurs after clathrin- and/or caveolin-dependent endocytosis of existing protein. Several studies have shown that phosphorylation by signalling kinases is important for the internalization and recycling processes, although the transporter protein does not appear to be directly phosphorylated. After internalization, transporters that are targeted for degradation undergo ubiquitination, most likely on intracellular loop residues. Epigenetic mechanisms, including methylation of gene regulatory regions and transcription from alternate promoters, are also significant in the regulation of certain SLC22A/SLCO genes. The membrane expression of OATs/OATPs is dysregulated in disease, which affects drug efficacy and detoxification. Several transporters are expressed in the cytoplasmic subcompartment in disease states, which suggests that membrane targeting/internalization/recycling may be impaired. This article focuses on recent developments in OAT and OATP regulation, their dysregulation in disease and the significance for drug therapy.
Collapse
Affiliation(s)
- Michael Murray
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, NSW, 2006, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
19
|
Chun SE, Thakkar N, Oh Y, Park JE, Han S, Ryoo G, Hahn H, Maeng SH, Lim YR, Han BW, Lee W. The N-terminal region of organic anion transporting polypeptide 1B3 (OATP1B3) plays an essential role in regulating its plasma membrane trafficking. Biochem Pharmacol 2017; 131:98-105. [PMID: 28216016 DOI: 10.1016/j.bcp.2017.02.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/14/2017] [Indexed: 10/20/2022]
Abstract
Organic anion transporting polypeptide 1B3 (OATP1B3) is a major influx transporter mediating the hepatic uptake of various endogenous substrates as well as clinically important drugs such as statins and anticancer drugs. However, molecular mechanisms controlling the membrane trafficking of OATP1B3 have been largely unknown. Several reports recently indicated the presence of a distinct, cancer-type OATP1B3 variant lacking the N-terminal 28 amino acids compared to OATP1B3 expressed in non-malignant hepatocytes. Interestingly, the cancer-type OATP1B3 variant is located predominantly in the cytoplasm, implicating the involvement of the N-terminal region of OATP1B3 in its membrane trafficking. In the current study, we set out to experimentally validate the importance of the N-terminal region of OATP1B3 and to identify responsible sequence motif(s) in that region. A number of truncation or point mutants of OATP1B3 were transiently expressed in HEK293T, HCT-8 or MDCK II cells and their expression in cytoplasmic and surface membrane fractions were analyzed by immunoblotting. Our results indicated that the N-terminal sequence of OATP1B3, in particular, at the amino acid positions between 12 and 28, may be indispensable in its membrane trafficking. Moreover, our results using a fusion construct indicated that the first 50 amino acids of OATP1B3 are sufficient for its membrane localization. The importance of the N-terminal region in membranous localization was shared among the other OATP1B subfamily members, OATP1B1 and rat Oatp1b2. Our efforts to identify the responsible amino acid(s) or structure motif(s) in the N-terminal region did not pinpoint individual amino acids or motifs with putative secondary structures. Our current findings however demonstrate that the N-terminal region is important for the membrane localization of the OATP1B subfamily members and should facilitate future investigations of the mechanisms involved in the regulation and membrane trafficking of these important transporter proteins.
Collapse
Affiliation(s)
- Se-Eun Chun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Nilay Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Yunseok Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Songhee Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Gongmi Ryoo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Hyunggu Hahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Sang Hyun Maeng
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Young-Ran Lim
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Byung Woo Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Shi C, Wu JB, Pan D. Review on near-infrared heptamethine cyanine dyes as theranostic agents for tumor imaging, targeting, and photodynamic therapy. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:50901. [PMID: 27165449 DOI: 10.1117/1.jbo.21.5.050901] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/20/2016] [Indexed: 05/21/2023]
Abstract
A class of near-infrared fluorescence (NIRF) heptamethine cyanine dyes that are taken up and accumulated specifically in cancer cells without chemical conjugation have recently emerged as promising tools for tumor imaging and targeting. In addition to their fluorescence and nuclear imaging-based tumor-imaging properties, these dyes can be developed as drug carriers to safely deliver chemotherapy drugs to tumors. They can also be used as effective agents for photodynamic therapy with remarkable tumoricidal activity via photodependent cytotoxic activity. The preferential uptake of dyes into cancer but not normal cells is co-operatively mediated by the prevailing activation of a group of organic anion-transporting polypeptides on cancer cell membranes, as well as tumor hypoxia and increased mitochondrial membrane potential in cancer cells. Such mechanistic explorations have greatly advanced the current application and future development of NIRF dyes and their derivatives as anticancer theranostic agents. This review summarizes current knowledge and emerging advances in NIRF dyes, including molecular characterization, photophysical properties, multimodal development and uptake mechanisms, and their growing potential for preclinical and clinical use.
Collapse
Affiliation(s)
- Changhong Shi
- Fourth Military Medical University, Laboratory Animal Center, 169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Jason Boyang Wu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Uro-Oncology Research Program, Department of Medicine, Los Angeles, California 90048, United States
| | - Dongfeng Pan
- University of Virginia, Department of Radiology, Charlottesville, Virginia 22908, United States
| |
Collapse
|
21
|
Shi C, Wu JB, Chu GCY, Li Q, Wang R, Zhang C, Zhang Y, Kim HL, Wang J, Zhau HE, Pan D, Chung LWK. Heptamethine carbocyanine dye-mediated near-infrared imaging of canine and human cancers through the HIF-1α/OATPs signaling axis. Oncotarget 2015; 5:10114-26. [PMID: 25361418 PMCID: PMC4259409 DOI: 10.18632/oncotarget.2464] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/06/2014] [Indexed: 12/18/2022] Open
Abstract
Near-infrared (NIR) fluorescence imaging agents are promising tools for noninvasive cancer imaging. This study explored the specific uptake and retention of a NIR heptamethine carbocyanine MHI-148 dye by canine cancer cells and tissues and human prostate cancer (PCa) specimens and also the dye uptake mechanisms. The accumulation of MHI-148 was detected specifically in canine cancer cells and tissues and freshly harvested human PCa tissues xenografted in mice by NIR fluorescence microscopy and whole-body NIR optical imaging. Specific dye uptake in canine spontaneous tumors was further confirmed by PET imaging. Higher hypoxia-inducible factor-1α (HIF-1α) and organic anion-transporting polypeptide (OATP) protein and mRNA expression was demonstrated by multiplex quantum dots labeling and qPCR in tumors over that of normal tissues. Treating cancer cells with HIF-1α stabilizers activated HIF-1α downstream target genes, induced OATP superfamily gene expression and enhanced cellular uptake and retention of NIR dyes. Moreover, silencing HIF-1α by siRNA significantly decreased OATP mRNA expression and blocked NIR dye uptake in cancer cells. Together, these results demonstrated the preferential uptake of NIR dyes by canine and human cancer cells and tissues via the HIF-1α/OATPs signaling axis, which provides insights into future application of these dyes for cancer detection and treatment.
Collapse
Affiliation(s)
- Changhong Shi
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China. Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jason Boyang Wu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gina C-Y Chu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ruoxiang Wang
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Caiqin Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yi Zhang
- Department of Radiology, the University of Virginia, Charlottesville, VA 22908, USA
| | - Hyung L Kim
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dongfeng Pan
- Department of Radiology, the University of Virginia, Charlottesville, VA 22908, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
22
|
Wu JB, Shi C, Chu GCY, Xu Q, Zhang Y, Li Q, Yu JS, Zhau HE, Chung LWK. Near-infrared fluorescence heptamethine carbocyanine dyes mediate imaging and targeted drug delivery for human brain tumor. Biomaterials 2015. [PMID: 26197410 DOI: 10.1016/j.biomaterials.2015.07.028] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain tumors and brain metastases are among the deadliest malignancies of all human cancers, largely due to the cellular blood-brain and blood-tumor barriers that limit the delivery of imaging and therapeutic agents from the systemic circulation to tumors. Thus, improved strategies for brain tumor visualization and targeted treatment are critically needed. Here we identified and synthesized a group of near-infrared fluorescence (NIRF) heptamethine carbocyanine dyes and derivative NIRF dye-drug conjugates for effective imaging and therapeutic targeting of brain tumors of either primary or metastatic origin in mice, which is mechanistically mediated by tumor hypoxia and organic anion-transporting polypeptide genes. We also demonstrate that these dyes, when conjugated to chemotherapeutic agents such as gemcitabine, significantly restricted the growth of both intracranial glioma xenografts and prostate tumor brain metastases and prolonged survival in mice. These results show promise in the application of NIRF dyes as novel theranostic agents for the detection and treatment of brain tumors.
Collapse
Affiliation(s)
- Jason Boyang Wu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Changhong Shi
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Gina Chia-Yi Chu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Qijin Xu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yi Zhang
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - John S Yu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
23
|
Thakkar N, Lockhart AC, Lee W. Role of Organic Anion-Transporting Polypeptides (OATPs) in Cancer Therapy. AAPS JOURNAL 2015; 17:535-45. [PMID: 25735612 DOI: 10.1208/s12248-015-9740-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 02/11/2015] [Indexed: 12/31/2022]
Abstract
The superfamily of organic anion-transporting polypeptides (OATPs, gene symbol SLCO) includes important transporters handling a variety of endogenous and xenobiotic substrates. Currently, 11 human OATPs are known and their substrates include endogenous hormones and their conjugates, anticancer drugs, and imaging agents. The contribution of OATPs to the in vivo disposition of these substrates has been extensively investigated. An accumulating body of evidence also indicates that the expression of some OATPs may be up- or downregulated in several types of cancers, suggesting potential pathogenic roles during the development and progression of cancer. Given that the role of OATPs in handling cancer therapeutics has been already covered by several excellent reviews, this review will focus on the recent progresses on the topic, in particular the role of OATPs in the disposition of anticancer drugs, the impact of OATP genetic variations on the function of OATPs, and the OATPs differentially expressed in cancer and their potential roles in cancer development, progression, and treatment.
Collapse
Affiliation(s)
- Nilay Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA
| | | | | |
Collapse
|
24
|
Teft WA, Welch S, Lenehan J, Parfitt J, Choi YH, Winquist E, Kim RB. OATP1B1 and tumour OATP1B3 modulate exposure, toxicity, and survival after irinotecan-based chemotherapy. Br J Cancer 2015; 112:857-65. [PMID: 25611302 PMCID: PMC4453959 DOI: 10.1038/bjc.2015.5] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/01/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022] Open
Abstract
Background: Treatment of advanced and metastatic colorectal cancer with irinotecan is hampered by severe toxicities. The active metabolite of irinotecan, SN-38, is a known substrate of drug-metabolising enzymes, including UGT1A1, as well as OATP and ABC drug transporters. Methods: Blood samples (n=127) and tumour tissue (n=30) were obtained from advanced cancer patients treated with irinotecan-based regimens for pharmacogenetic and drug level analysis and transporter expression. Clinical variables, toxicity, and outcomes data were collected. Results: SLCO1B1 521C was significantly associated with increased SN-38 exposure (P<0.001), which was additive with UGT1A1*28. ABCC5 (rs562) carriers had significantly reduced SN-38 glucuronide and APC metabolite levels. Reduced risk of neutropenia and diarrhoea was associated with ABCC2–24C/T (odds ratio (OR)=0.22, 0.06–0.85) and CES1 (rs2244613; OR=0.29, 0.09–0.89), respectively. Progression-free survival (PFS) was significantly longer in SLCO1B1 388G/G patients and reduced in ABCC2–24T/T and UGT1A1*28 carriers. Notably, higher OATP1B3 tumour expression was associated with reduced PFS. Conclusions: Clarifying the association of host genetic variation in OATP and ABC transporters to SN-38 exposure, toxicity and PFS provides rationale for personalising irinotecan-based chemotherapy. Our findings suggest that OATP polymorphisms and expression in tumour tissue may serve as important new biomarkers.
Collapse
Affiliation(s)
- W A Teft
- Department of Medicine, Division of Clinical Pharmacology, London Health Sciences Centre-University Hospital, Western University, Room B9-132, 339 Windermere Road, London, Ontario, Canada N6A 5A5
| | - S Welch
- Department of Oncology, London Health Sciences Centre-Victoria Hospital, Western University, 800 Commissioners Road East, PO Box 5010, London, Ontario, Canada N6A 5W9
| | - J Lenehan
- Department of Oncology, London Health Sciences Centre-Victoria Hospital, Western University, 800 Commissioners Road East, PO Box 5010, London, Ontario, Canada N6A 5W9
| | - J Parfitt
- Department of Pathology, London Health Sciences Centre - University Hospital, Western University, 339 Windermere Road, London, Ontario, Canada N6A 5A5
| | - Y-H Choi
- Department of Epidemiology and Biostatistics, Kresge Building, Western University, London Ontario, Canada N6A 5C1
| | - E Winquist
- Department of Oncology, London Health Sciences Centre-Victoria Hospital, Western University, 800 Commissioners Road East, PO Box 5010, London, Ontario, Canada N6A 5W9
| | - R B Kim
- 1] Department of Medicine, Division of Clinical Pharmacology, London Health Sciences Centre-University Hospital, Western University, Room B9-132, 339 Windermere Road, London, Ontario, Canada N6A 5A5 [2] Department of Oncology, London Health Sciences Centre-Victoria Hospital, Western University, 800 Commissioners Road East, PO Box 5010, London, Ontario, Canada N6A 5W9 [3] Department of Physiology and Pharmacology, Medical Sciences Building, Western University, London, Ontario, Canada N6A 5C1
| |
Collapse
|
25
|
Abstract
Organic anion-transporting polypeptides or OATPs are central transporters in the disposition of drugs and other xenobiotics. In addition, they mediate transport of a wide variety of endogenous substrates. The critical role of OATPs in drug disposition has spurred research both in academia and in the pharmaceutical industry. Translational aspects with clinical questions are the focus in academia, while the pharmaceutical industry tries to define and understand the role these transporters play in pharmacotherapy. The present overview summarizes our knowledge on the interaction of food constituents with OATPs and on the OATP transport mechanisms. Further, it gives an update on the available information on the structure-function relationship of the OATPs and, finally, covers the transcriptional and posttranscriptional regulation of OATPs.
Collapse
Affiliation(s)
- Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zürich, Switzerland.
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
26
|
Nakanishi T, Tamai I. Putative roles of organic anion transporting polypeptides (OATPs) in cell survival and progression of human cancers. Biopharm Drug Dispos 2014; 35:463-84. [DOI: 10.1002/bdd.1915] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 08/01/2014] [Accepted: 08/12/2014] [Indexed: 01/19/2023]
Affiliation(s)
- Takeo Nakanishi
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kakuma-machi Kanazawa 920-1192 Japan
| | - Ikumi Tamai
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kakuma-machi Kanazawa 920-1192 Japan
| |
Collapse
|