1
|
Moghaddam S, Jalali A, O’Neill A, Murphy L, Gorman L, Reilly AM, Heffernan Á, Lynch T, Power R, O’Malley KJ, Taskèn KA, Berge V, Solhaug VA, Klocker H, Murphy TB, Watson RW. Integrating Serum Biomarkers into Prediction Models for Biochemical Recurrence Following Radical Prostatectomy. Cancers (Basel) 2021; 13:4162. [PMID: 34439316 PMCID: PMC8391749 DOI: 10.3390/cancers13164162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/14/2021] [Indexed: 12/13/2022] Open
Abstract
This study undertook to predict biochemical recurrence (BCR) in prostate cancer patients after radical prostatectomy using serum biomarkers and clinical features. Three radical prostatectomy cohorts were used to build and validate a model of clinical variables and serum biomarkers to predict BCR. The Cox proportional hazard model with stepwise selection technique was used to develop the model. Model evaluation was quantified by the AUC, calibration, and decision curve analysis. Cross-validation techniques were used to prevent overfitting in the Irish training cohort, and the Austrian and Norwegian independent cohorts were used as validation cohorts. The integration of serum biomarkers with the clinical variables (AUC = 0.695) improved significantly the predictive ability of BCR compared to the clinical variables (AUC = 0.604) or biomarkers alone (AUC = 0.573). This model was well calibrated and demonstrated a significant improvement in the predictive ability in the Austrian and Norwegian validation cohorts (AUC of 0.724 and 0.606), compared to the clinical model (AUC of 0.665 and 0.511). This study shows that the pre-operative biomarker PEDF can improve the accuracy of the clinical factors to predict BCR. This model can be employed prior to treatment and could improve clinical decision making, impacting on patients' outcomes and quality of life.
Collapse
Affiliation(s)
- Shirin Moghaddam
- School of Mathematical Sciences, University College Cork, T12XF62 Cork, Ireland
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Amirhossein Jalali
- School of Mathematical Sciences, University College Cork, T12XF62 Cork, Ireland
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Amanda O’Neill
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Lisa Murphy
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Laura Gorman
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Anne-Marie Reilly
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Áine Heffernan
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| | - Thomas Lynch
- Department of Urology, Trinity College, St James Hospital, D08 W9RT Dublin 8, Ireland;
| | - Richard Power
- Department of Urology, Royal College of Surgeons in Ireland, Beaumont Hospital, D09V2N0 Dublin 9, Ireland;
| | - Kieran J. O’Malley
- Department of Urology, University College Dublin, Mater Misericordiae University Hospital, D07YH5R Dublin 7, Ireland;
| | - Kristin A. Taskèn
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (K.A.T.); (V.B.)
- Department of Tumor Biology, Oslo University Hospital, 0379 Oslo, Norway
| | - Viktor Berge
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (K.A.T.); (V.B.)
- Department of Urology, Oslo University Hospital, 0379 Oslo, Norway;
| | - Vivi-Ann Solhaug
- Department of Urology, Oslo University Hospital, 0379 Oslo, Norway;
| | - Helmut Klocker
- Department of Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - T. Brendan Murphy
- UCD School of Mathematics and Statistics, University College Dublin, D04V1W8 Dublin 4, Ireland;
| | - R. William Watson
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, UCD, D04V1W8 Dublin 4, Ireland; (A.O.); (L.M.); (L.G.); (A.-M.R.); (Á.H.); (R.W.W.)
| |
Collapse
|
2
|
Brook N, Brook E, Dass CR, Chan A, Dharmarajan A. Pigment Epithelium-Derived Factor and Sex Hormone-Responsive Cancers. Cancers (Basel) 2020; 12:cancers12113483. [PMID: 33238558 PMCID: PMC7700359 DOI: 10.3390/cancers12113483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Oestrogens and androgens play important roles in normal and cancerous tissue and have been shown to negatively regulate pigment epithelium-derived factor (PEDF) expression in sex hormone-responsive tumours. PEDF suppresses tumour growth and its downregulation by oestrogen is implicated in tumorigenesis, metastasis, and progression. PEDF expression is reduced in cancerous tissue of the prostate, breast, ovary, and endometrium compared to their normal tissue counterparts, with a link between PEDF downregulation and sex hormone signalling observed in pre-clinical studies. PEDF reduces growth and metastasis of tumour cells by promoting apoptosis, inhibiting angiogenesis, increasing adhesion, and reducing migration. PEDF may also prevent treatment resistance in some cancers by downregulating oestrogen receptor signalling. By interacting with components of the tumour microenvironment, PEDF counteracts the proliferative and immunosuppressive effects of oestrogens, to ultimately reduce tumorigenesis and metastasis. In this review, we focus on sex hormone regulation of PEDF's anti-tumour action in sex hormone-responsive tumours.
Collapse
Affiliation(s)
- Naomi Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
| | - Emily Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
| | - Crispin R. Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
- Correspondence: (C.R.D.); (A.D.); Tel.: +61-8-9266-1489 (C.R.D.)
| | - Arlene Chan
- School of Medicine, Curtin University, Bentley, WA 6102, Australia;
- Breast Cancer Research Centre-Western Australia, Hollywood Private Hospital, Nedlands, WA 6009, Australia
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India
- Correspondence: (C.R.D.); (A.D.); Tel.: +61-8-9266-1489 (C.R.D.)
| |
Collapse
|
3
|
George DJ, Halabi S, Healy P, Barak I, Winters C, Anand M, Wilder R, Klein M, Martinez E, Nixon AB, Harrison MR, Szmulewitz R, Armstrong AJ. Phase 1b trial of docetaxel, prednisone, and pazopanib in men with metastatic castration-resistant prostate cancer. Prostate 2019; 79:1752-1761. [PMID: 31497882 DOI: 10.1002/pros.23899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/05/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Docetaxel prednisone is a standard of care for men with metastatic castration-resistant prostate cancer (mCRPC), and plasma vascular endothelial growth factor (VEGF) levels are a poor prognostic factor in this population; therefore, we evaluated the combination of docetaxel prednisone with pazopanib, an oral VEGF receptor inhibitor, for safety and preliminary efficacy. METHODS This is a two-site phase 1b Department of Defense Prostate Cancer Clinical Trials Consortium trial of docetaxel, prednisone, and pazopanib once daily and ongoing androgen deprivation therapy and prophylactic pegfilgrastim in men with mCRPC. The primary endpoint was safety and the determination of a maximum tolerated dose (MTD) through a dose-escalation and expansion design; secondary endpoints included progression-free and overall survival (OS), prostate specific antigen (PSA) declines, radiographic responses, and pharmacokinetic and plasma angiokine biomarker analyses. RESULTS Twenty-five men were treated over six dose levels. Pegfilgrastim was added to the regimen after myelosuppression limited dose escalation. With pegfilgrastim, our target MTD of docetaxel 75 mg/m2 q3 weeks; prednisone 10 mg daily; and pazopanib 800 mg daily was reached. Eleven additional patients were accrued at this dose level for a total of 36 patients. Dose-limiting toxicities included neutropenia, syncope, and hypertension. Three deaths attributed to study treatment occurred. The objective response rate was 31%; median PFS was 14.1 months (95% confidence interval [CI]: 7.1 and 22.2); and OS was 18.6 months (95% CI: 11.8 and 22.2). CONCLUSIONS The combination of docetaxel, prednisone, and pazopanib (with pegfilgrastim) was tolerable at full doses and demonstrated promising efficacy in a relatively poor risk patients with mCRPC. Further development of predictive biomarkers may enrich for patients who receive clinical benefit from this regimen.
Collapse
Affiliation(s)
- Daniel J George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, North Carolina
| | - Susan Halabi
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Department of Biostatistics, Duke University, Durham, North Carolina
| | - Patrick Healy
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Department of Biostatistics, Duke University, Durham, North Carolina
| | - Ian Barak
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Department of Biostatistics, Duke University, Durham, North Carolina
| | - Carolyn Winters
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Monika Anand
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Rhonda Wilder
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Melissa Klein
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Elia Martinez
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Andrew B Nixon
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, North Carolina
| | - Michael R Harrison
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, North Carolina
| | - Russell Szmulewitz
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
4
|
Kadimisetty K, Malla S, Bhalerao KS, Mosa IM, Bhakta S, Lee NH, Rusling JF. Automated 3D-Printed Microfluidic Array for Rapid Nanomaterial-Enhanced Detection of Multiple Proteins. Anal Chem 2018; 90:7569-7577. [PMID: 29779368 PMCID: PMC6104517 DOI: 10.1021/acs.analchem.8b01198] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We report here the fabrication and validation of a novel 3D-printed, automated immunoarray to detect multiple proteins with ultralow detection limits. This low cost, miniature immunoarray employs electrochemiluminescent (ECL) detection measured with a CCD camera and employs touch-screen control of a micropump to facilitate automated use. The miniaturized array features prefilled reservoirs to deliver sample and reagents to a paper-thin pyrolytic graphite microwell detection chip to complete sandwich immunoassays. The detection chip achieves high sensitivity by using single-wall carbon nanotube-antibody conjugates in the microwells and employing massively labeled antibody-decorated RuBPY-silica nanoparticles to generate ECL. The total cost of an array is $0.65, and an eight-protein assay can be done in duplicate for $0.14 per protein with limits of detection (LOD) as low as 78-110 fg mL-1 in diluted serum. The electronic control system costs $210 in components. Utility of the automated immunoarray was demonstrated by detecting an eight-protein prostate cancer biomarker panel in human serum samples in 25 min. The system is well suited to future clinical and point-of-care diagnostic testing and could be used in resource-limited environments.
Collapse
Affiliation(s)
- Karteek Kadimisetty
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Spundana Malla
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ketki S. Bhalerao
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Islam M. Mosa
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Chemistry, Tanta University, Tanta 31527, Egypt
| | - Snehasis Bhakta
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Norman H. Lee
- Department of Pharmacology & Physiology, George Washington University, Washington, D.C. 20037, United States
| | - James F. Rusling
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Surgery and Neag Cancer Center, UConn Health, Farmington, Connecticut 06032, United States
- School of Chemistry, National University of Ireland, Galway H91 TK33, Ireland
| |
Collapse
|
5
|
Breen KJ, O'Neill A, Murphy L, Fan Y, Boyce S, Fitzgerald N, Dorris E, Brady L, Finn SP, Hayes BD, Treacy A, Barrett C, Aziz MA, Kay EW, Fitzpatrick JM, Watson RWG. Investigating the role of the IGF axis as a predictor of biochemical recurrence in prostate cancer patients post-surgery. Prostate 2017; 77:1288-1300. [PMID: 28726241 DOI: 10.1002/pros.23389] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/22/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Between 20% and 35% of prostate cancer (PCa) patients who undergo treatment with curative intent (ie, surgery or radiation therapy) for localized disease will experience biochemical recurrence (BCR). Alterations in the insulin-like growth factor (IGF) axis and PTEN expression have been implicated in the development and progression of several human tumors including PCa. We examined the expression of the insulin receptor (INSR), IGF-1 receptor (IGF-1R), PTEN, and AKT in radical prostatectomy tissue of patients who developed BCR post-surgery. METHODS Tissue microarrays (TMA) of 130 patients post-radical prostatectomy (65 = BCR, 65 = non-BCR) were stained by immunohistochemistry for INSR, IGF-1R, PTEN, and AKT using optimized antibody protocols. INSR, IGF1-R, PTEN, and AKT expression between benign and cancerous tissue, and different Gleason grades was assessed. Kaplan-Meier survival curves were used to examine the relationship between proteins expression and BCR. RESULTS INSR (P < 0.001), IGF-1R (P < 0.001), and AKT (P < 0.05) expression was significantly increased and PTEN (P < 0.001) was significantly decreased in cancerous versus benign tissue. There was no significant difference in INSR, IGF-1R, or AKT expression in the cancerous tissue of non-BCR versus BCR patients (P = 0.149, P = 0.990, P = 0.399, respectively). There was a significant decrease in PTEN expression in the malignant tissue of BCR versus non-BCR patients (P = 0.011). Combinational analysis of the tissue proteins identified a combination of decreased PTEN and increased AKT or increased INSR was associated with worst outcome. We found that in each case, our hypothesized worst group was most likely to experience BCR and this was significant for combinations of PTEN+INSR and PTEN+AKT but not PTEN+IGF-1R (P = 0.023, P = 0.028, P = 0.078, respectively). CONCLUSIONS Low PTEN is associated with BCR and this association is strongly modified by high INSR and high AKT expression. Measurement of these proteins could help inform appropriate patient selection for postoperative adjuvant therapy and prevent BCR.
Collapse
Affiliation(s)
- Kieran J Breen
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Amanda O'Neill
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Lisa Murphy
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Yue Fan
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Susie Boyce
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- UCD School of Mathematical Sciences, Dublin, Ireland
| | - Noel Fitzgerald
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Emma Dorris
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Lauren Brady
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Brian D Hayes
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Ann Treacy
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ciara Barrett
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Mardiana Abdul Aziz
- Department of Histopathology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Elaine W Kay
- Department of Pathology, RCSI Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - John M Fitzpatrick
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - R William G Watson
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
6
|
Li QK, Chen L, Ao MH, Chiu JH, Zhang Z, Zhang H, Chan DW. Serum fucosylated prostate-specific antigen (PSA) improves the differentiation of aggressive from non-aggressive prostate cancers. Theranostics 2015; 5:267-76. [PMID: 25553114 PMCID: PMC4279190 DOI: 10.7150/thno.10349] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 11/01/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Clinically, it is still challenging to differentiate aggressive from non-aggressive prostate cancers (Pca) by non-invasive approaches. Our recent studies showed that overexpression of alpha (1-6) fucosyltransferase played an important role in Pca cells. In this study, we have investigated levels of glycoproteins and their fucosylated glycoforms in sera of Pca patients, as well as the potential utility of fucosylated glycoproteins in the identification of aggressive Pca. MATERIAL AND METHODS Serum samples from histomorphology-proven Pca cases were included. Prostate-specific antigen (PSA), tissue inhibitor of metallopeptidase 1 (TIMP1) and tissue plasminogen activator (tPA), and their fucosylated glycoforms were captured by Aleuria Aurantia Lectin (AAL), followed by the multiplex magnetic bead-based immunoassay. The level of fucosylated glycoproteins was correlated with patients' Gleason score of the tumor. RESULT Among three fucosylated glycoproteins, the fucosylated PSA was significantly increased and correlated with the tumor Gleason score (p<0.05). The ratio of fucosylated PSA showed a marked increase in aggressive tumors in comparison to non-aggressive tumors. ROC analysis also showed an improved predictive power of fucosylated PSA in the identification of aggressive Pca. CONCLUSIONS Our data demonstrated that fucosylated PSA has a better predictive power to differentiate aggressive tumors from non-aggressive tumors, than that of native PSA and two other glycoproteins. The fucosylated PSA has the potential to be used as a surrogate biomarker.
Collapse
Affiliation(s)
- Qing Kay Li
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Adamy A. Editorial comment. Urology 2013; 81:990-1; discussion 991. [PMID: 23490522 DOI: 10.1016/j.urology.2012.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|