1
|
Pandit-Taskar N, O'Donoghue JA, Chetty D, Max S, Wanik D, Ilovich O, Russell M, Nyima T, Divgi CR, Yu M, Morris MJ. A Phase 0 Study to Assess the Biodistribution and Pharmacokinetics of a Radiolabeled Antibody Targeting Human Kallikrein 2 in Participants with Metastatic Castration-Resistant Prostate Cancer. J Nucl Med 2024; 65:1051-1056. [PMID: 38782459 PMCID: PMC11218722 DOI: 10.2967/jnumed.124.267416] [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: 01/25/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Despite the inclusion of multiple agents within the prostate cancer treatment landscape, new treatment options are needed to address the unmet need for patients with metastatic castration-resistant prostate cancer (mCRPC). Although prostate-specific membrane antigen is the only cell-surface target to yield clinical benefit in men with advanced prostate cancer, additional targets may further advance targeted immune, cytotoxic, radiopharmaceutical, and other tumor-directed therapies for these patients. Human kallikrein 2 (hK2) is a novel prostate-specific target with little to no expression in nonprostate tissues. This first-in-human phase 0 trial uses an 111In-radiolabeled anti-hK2 monoclonal antibody, [111In]-DOTA-h11B6, to credential hK2 as a potential target for prostate cancer treatment. Methods: Participants with progressive mCRPC received a single infusion of 2 mg of [111In]-DOTA-h11B6 (185 MBq of 111In), with or without 8 mg of unlabeled h11B6 to assess antibody mass effects. Sequential imaging and serial blood samples were collected to determine [111In]-DOTA-h11B6 biodistribution, dosimetry, serum radioactivity, and pharmacokinetics. Safety was assessed within a 2-wk follow-up period from the time of [111In]-DOTA-h11B6 administration. Results: Twenty-two participants received [111In]-DOTA-h11B6 and are included in this analysis. Within 6-8 d of administration, [111In]-DOTA-h11B6 visibly accumulated in known mCRPC lesions, with limited uptake in other organs. Two treatment-emergent adverse events unrelated to treatment occurred, including tumor-related bleeding in 1 patient, which led to early study discontinuation. Serum clearance, biodistribution, and tumor targeting were independent of total antibody mass (2 or 10 mg). Conclusion: This first-in-human study demonstrates that tumor-associated hK2 can be identified and targeted using h11B6 as a platform as the h11B6 antibody selectively accumulated in mCRPC metastases with mass-independent clearance kinetics. These data support the feasibility of hK2 as a target for imaging and hK2-directed agents as potential therapies in patients with mCRPC.
Collapse
Affiliation(s)
- Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical Center, New York, New York
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dushen Chetty
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Steven Max
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | | | | | - Michael Russell
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Tenzin Nyima
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Margaret Yu
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Michael J Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
- Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
2
|
Valente-Santos J, Vitorino R, Sousa-Mendes C, Oliveira P, Colaço B, Faustino-Rocha AI, Neuparth MJ, Leite-Moreira A, Duarte JA, Ferreira R, Amado F. Long-Term Exposure to Supraphysiological Levels of Testosterone Impacts Rat Submandibular Gland Proteome. Int J Mol Sci 2023; 25:550. [PMID: 38203721 PMCID: PMC10778877 DOI: 10.3390/ijms25010550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
The salivary glands play a central role in the secretion of saliva, whose composition and volume affect oral and overall health. A lesser-explored dimension encompasses the possible changes in salivary gland proteomes in response to fluctuations in sex hormone levels. This study aimed to examine the effects of chronic exposure to testosterone on salivary gland remodeling, particularly focusing on proteomic adaptations. Therefore, male Wistar rats were implanted with subcutaneous testosterone-releasing devices at 14 weeks of age. Their submandibular glands were histologically and molecularly analyzed 47 weeks later. The results underscored a significant increase in gland mass after testosterone exposure, further supported by histologic evidence of granular duct enlargement. Despite increased circulating sex hormones, there was no detectable shift in the tissue levels of estrogen alpha and androgen receptors. GeLC-MS/MS and subsequent bioinformatics identified 308 proteins in the submandibular glands, 12 of which were modulated by testosterone. Of note was the pronounced upregulation of Klk3 and the downregulation of Klk6 and Klk7 after testosterone exposure. Protein-protein interaction analysis with the androgen receptor suggests that Klk3 is a potential target of androgenic signaling, paralleling previous findings in the prostate. This exploratory analysis sheds light on the response of salivary glands to testosterone exposure, providing proteome-level insights into the associated weight and histological changes.
Collapse
Affiliation(s)
- João Valente-Santos
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.-S.); (R.F.)
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Cláudia Sousa-Mendes
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.-M.); (A.L.-M.)
| | - Paula Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; (P.O.); (A.I.F.-R.)
| | - Bruno Colaço
- Veterinary and Animal Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Department of Animal Science, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Ana I. Faustino-Rocha
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; (P.O.); (A.I.F.-R.)
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology, University of Évora, 7006-554 Évora, Portugal
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal; (M.J.N.); (J.A.D.)
- TOXRUN—Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
| | - Adelino Leite-Moreira
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.-M.); (A.L.-M.)
| | - José Alberto Duarte
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal; (M.J.N.); (J.A.D.)
- TOXRUN—Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.-S.); (R.F.)
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.-S.); (R.F.)
| |
Collapse
|
3
|
Inderjeeth A, Iravani A, Subramaniam S, Conduit C, Sandhu S. Novel radionuclide therapy combinations in prostate cancer. Ther Adv Med Oncol 2023; 15:17588359231187202. [PMID: 37547444 PMCID: PMC10399256 DOI: 10.1177/17588359231187202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/21/2023] [Indexed: 08/08/2023] Open
Abstract
Prostate cancer remains the commonest cancer diagnosed in males and a leading cause of cancer-related death. Men with metastatic castration-resistant prostate cancer (mCRPC) who have progressed on chemotherapy and androgen receptor pathway inhibitors (ARPI) have limited treatment options, significant morbidity, and poor outcomes. Prostate-specific membrane antigen (PSMA)-directed radionuclide therapy (RNT) is emerging as an efficacious and well-tolerated therapy; however, disease progression is universal. Several ongoing RNT trials focus on combination strategies to improve efficacy and durability of treatment response, including combinations with ARPIs, chemotherapy, immunotherapy, and targeted therapies. Further, efforts are underway to expand the role of PSMA-directed RNT to earlier stages of disease including hormone-sensitive and localized prostate cancer. In this review, we discuss the rationale and ongoing RNT combination therapeutic trials in prostate cancer and summarize the efficacy and toxicity associated with RNT.
Collapse
Affiliation(s)
- Andrisha–Jade Inderjeeth
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Amir Iravani
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Shalini Subramaniam
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Bankstown-Lidcombe Hospital, Bankstown, NSW, Australia
| | - Ciara Conduit
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
4
|
Gut M, Dreier B, Furler S, Sobek J, Plückthun A, Holland JP. Designed ankyrin repeat proteins for detecting prostate-specific antigen expression in vivo. RSC Chem Biol 2023; 4:494-505. [PMID: 37415866 PMCID: PMC10320840 DOI: 10.1039/d3cb00010a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/16/2023] [Indexed: 07/08/2023] Open
Abstract
Late-stage prostate cancer often acquires resistance to conventional chemotherapies and transforms into a hormone-refractory, drug-resistant, and non-curative disease. Developing non-invasive tools to detect the biochemical changes that correlate with drug efficacy and reveal the onset of drug resistance would have important ramifications in managing the treatment regimen for individual patients. Here, we report the selection of new Designed Ankyrin Repeat Proteins (DARPins) that show high affinity toward prostate-specific antigen (PSA), a biomarker used in clinical monitoring of prostate cancer. Ribosome display and in vitro screening tools were used to select PSA-binding DARPins based on their binding affinity, selectivity, and chemical constitution. Surface plasmon resonance measurements demonstrated that the four lead candidates bind to PSA with nanomolar affinity. DARPins were site-specifically functionalised at a unique C-terminal cysteine with a hexadentate aza-nonamacrocyclic chelate (NODAGA) for subsequent radiolabelling with the positron-emitting radionuclide 68Ga. [68Ga]GaNODAGA-DARPins showed high stability toward transchelation and were stable in human serum for >2 h. Radioactive binding assays using streptavidin-loaded magnetic beads confirmed that the functionalisation and radiolabelling did not compromise the specificity of [68Ga]GaNODAGA-DARPins toward PSA. Biodistribution experiments in athymic nude mice bearing subcutaneous prostate cancer xenografts derived from the LNCaP cell line revealed that three of the four [68Ga]GaNODAGA-DARPins displayed specific tumour-binding in vivo. For DARPin-6, tumour-uptake in the normal group reached 4.16 ± 0.58% ID g-1 (n = 3; 2 h post-administration) and was reduced by ∼50% by competitive binding with a low molar activity formulation (blocking group: 2.47 ± 0.42% ID g-1; n = 3; P value = 0.018). Collectively, the experimental results support the future development of new PSA-specific imaging agents for potential use in monitoring the efficacy of androgen receptor (AR)-targeted therapies.
Collapse
Affiliation(s)
- Melanie Gut
- University of Zurich, Department of Chemistry Winterthurerstrasse 190 CH-8057, Zurich Switzerland https://www.hollandlab.org https://twitter.com/HollandLab_ +41-44-63-53990
| | - Birgit Dreier
- University of Zurich, Department of Biochemistry Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Sven Furler
- University of Zurich, Department of Biochemistry Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Jens Sobek
- Functional Genomics Center Zurich, Eidgenössische Technische Hochschule (ETH) Zurich and University of Zurich Zurich Switzerland
| | - Andreas Plückthun
- University of Zurich, Department of Biochemistry Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Jason P Holland
- University of Zurich, Department of Chemistry Winterthurerstrasse 190 CH-8057, Zurich Switzerland https://www.hollandlab.org https://twitter.com/HollandLab_ +41-44-63-53990
| |
Collapse
|
5
|
Rasmussen M, Fredsøe J, Tin AL, Vickers AJ, Ulhøi B, Borre M, Eastham J, Ehdaie B, Guillonneau B, Laudone V, Scardino PT, Touijer K, Sørensen KD, Lilja H. Independent validation of a pre-specified four-kallikrein marker model for prediction of adverse pathology and biochemical recurrence. Br J Cancer 2022; 126:1004-1009. [PMID: 34903844 PMCID: PMC8980060 DOI: 10.1038/s41416-021-01661-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Accurate markers for prostate cancer (PC) risk stratification could aid decision-making for initial management strategies. The 4Kscore has an undefined role in predicting outcomes after radical prostatectomy (RP). METHODS We included 1476 patients with 4Kscore measured prior to RP at two institutions. The 4Kscore was assessed for prediction of adverse pathology at RP and biochemical recurrence (BCR) relative to a clinical model. We pre-specified that all analyses would be assessed in biopsy Grade Group 1 (GG1) or 2 (GG2) PC patients, separately. RESULTS The 4Kscore increased discrimination for adverse pathology in all patients (delta area under the receiver operative curve (AUC) 0.009, 95% confidence interval (CI) 0.002, 0.016; clinical model AUC 0.767), driven by GG1 (delta AUC 0.040, 95% CI 0.006, 0.073) rather than GG2 patients (delta AUC 0.005, 95% CI -0.012, 0.021). Adding 4Kscore improved prediction of BCR in all patients (delta C-index 0.014, 95% CI 0.007, 0.021; preop-BCR nomogram C-index 0.738), again with larger changes in GG1 than in GG2. CONCLUSIONS This study validates prior investigations on the use of 4Kscore in men with biopsy-confirmed PC. Men with GG1 PC and a high 4Kscore may benefit from additional testing to guide treatment selection. Further research is warranted regarding the value of the 4Kscore in men with biopsy GG2 PC.
Collapse
Affiliation(s)
- Martin Rasmussen
- grid.154185.c0000 0004 0512 597XDepartment of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark ,grid.7048.b0000 0001 1956 2722Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Fredsøe
- grid.154185.c0000 0004 0512 597XDepartment of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark ,grid.7048.b0000 0001 1956 2722Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Amy L. Tin
- grid.51462.340000 0001 2171 9952Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Andrew J. Vickers
- grid.51462.340000 0001 2171 9952Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Benedicte Ulhøi
- grid.154185.c0000 0004 0512 597XDepartment of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Borre
- grid.7048.b0000 0001 1956 2722Department of Clinical Medicine, Aarhus University, Aarhus, Denmark ,grid.154185.c0000 0004 0512 597XDepartment of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - James Eastham
- grid.51462.340000 0001 2171 9952Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Behfar Ehdaie
- grid.51462.340000 0001 2171 9952Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Bertrand Guillonneau
- grid.51462.340000 0001 2171 9952Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY USA ,grid.6363.00000 0001 2218 4662Uro-Oncology Department, Charité University Hospital, Berlin, Germany
| | - Vincent Laudone
- grid.51462.340000 0001 2171 9952Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Peter T. Scardino
- grid.51462.340000 0001 2171 9952Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Karim Touijer
- grid.51462.340000 0001 2171 9952Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Karina D. Sørensen
- grid.154185.c0000 0004 0512 597XDepartment of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark ,grid.7048.b0000 0001 1956 2722Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hans Lilja
- grid.51462.340000 0001 2171 9952Departments of Laboratory Medicine, Surgery, and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA ,grid.4514.40000 0001 0930 2361Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
6
|
Song H, Weinstein HNW, Allegakoen P, Wadsworth MH, Xie J, Yang H, Castro EA, Lu KL, Stohr BA, Feng FY, Carroll PR, Wang B, Cooperberg MR, Shalek AK, Huang FW. Single-cell analysis of human primary prostate cancer reveals the heterogeneity of tumor-associated epithelial cell states. Nat Commun 2022; 13:141. [PMID: 35013146 PMCID: PMC8748675 DOI: 10.1038/s41467-021-27322-4] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 10/29/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer is the second most common malignancy in men worldwide and consists of a mixture of tumor and non-tumor cell types. To characterize the prostate cancer tumor microenvironment, we perform single-cell RNA-sequencing on prostate biopsies, prostatectomy specimens, and patient-derived organoids from localized prostate cancer patients. We uncover heterogeneous cellular states in prostate epithelial cells marked by high androgen signaling states that are enriched in prostate cancer and identify a population of tumor-associated club cells that may be associated with prostate carcinogenesis. ERG-negative tumor cells, compared to ERG-positive cells, demonstrate shared heterogeneity with surrounding luminal epithelial cells and appear to give rise to common tumor microenvironment responses. Finally, we show that prostate epithelial organoids harbor tumor-associated epithelial cell states and are enriched with distinct cell types and states from their parent tissues. Our results provide diagnostically relevant insights and advance our understanding of the cellular states associated with prostate carcinogenesis.
Collapse
Affiliation(s)
- Hanbing Song
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Hannah N. W. Weinstein
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Paul Allegakoen
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Marc H. Wadsworth
- grid.116068.80000 0001 2341 2786The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.66859.340000 0004 0546 1623Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142 USA
| | - Jamie Xie
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Heiko Yang
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Ethan A. Castro
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Kevin L. Lu
- grid.266102.10000 0001 2297 6811Department of Pathology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Bradley A. Stohr
- grid.266102.10000 0001 2297 6811Department of Pathology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Felix Y. Feng
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Departments of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Peter R. Carroll
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Bruce Wang
- grid.266102.10000 0001 2297 6811Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA 94143 USA
| | - Matthew R. Cooperberg
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.410372.30000 0004 0419 2775Division of Hematology and Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121 USA
| | - Alex K. Shalek
- grid.116068.80000 0001 2341 2786The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.66859.340000 0004 0546 1623Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142 USA
| | - Franklin W. Huang
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.410372.30000 0004 0419 2775Division of Hematology and Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121 USA
| |
Collapse
|
7
|
Nair RT, Dawkins AA, Ganesh HS. Editorial for "Diagnostic Performance of Prostate MRI Radiomics, Four Kallikrein Panel and Radiologist in the Detection of Prostate Cancer: A Retrospective External Validation Multi-center Study of Men With a Clinical Suspicion of Prostate Cancer". J Magn Reson Imaging 2021; 55:478-479. [PMID: 34448318 DOI: 10.1002/jmri.27898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/11/2022] Open
Affiliation(s)
- Rashmi T Nair
- Division of Abdominal Radiology, Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Adrian A Dawkins
- Division of Abdominal Radiology, Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Halemane S Ganesh
- Division of Abdominal Radiology, Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
8
|
Camacho L, Zabala-Letona A, Cortazar AR, Astobiza I, Dominguez-Herrera A, Ercilla A, Crespo J, Viera C, Fernández-Ruiz S, Martinez-Gonzalez A, Torrano V, Martín-Martín N, Gomez-Muñoz A, Carracedo A. Identification of Androgen Receptor Metabolic Correlome Reveals the Repression of Ceramide Kinase by Androgens. Cancers (Basel) 2021; 13:cancers13174307. [PMID: 34503116 PMCID: PMC8431577 DOI: 10.3390/cancers13174307] [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: 06/01/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent cancers in men. Androgen receptor signaling plays a major role in this disease, and androgen deprivation therapy is a common therapeutic strategy in recurrent disease. Sphingolipid metabolism plays a central role in cell death, survival, and therapy resistance in cancer. Ceramide kinase (CERK) catalyzes the phosphorylation of ceramide to ceramide 1-phosphate, which regulates various cellular functions including cell growth and migration. Here we show that activated androgen receptor (AR) is a repressor of CERK expression. We undertook a bioinformatics strategy using PCa transcriptomics datasets to ascertain the metabolic alterations associated with AR activity. CERK was among the most prominent negatively correlated genes in our analysis. Interestingly, we demonstrated through various experimental approaches that activated AR reduces the mRNA expression of CERK: (i) expression of CERK is predominant in cell lines with low or negative AR activity; (ii) AR agonist and antagonist repress and induce CERK mRNA expression, respectively; (iii) orchiectomy in wildtype mice or mice with PCa (harboring prostate-specific Pten deletion) results in elevated Cerk mRNA levels in prostate tissue. Mechanistically, we found that AR represses CERK through interaction with its regulatory elements and that the transcriptional repressor EZH2 contributes to this process. In summary, we identify a repressive mode of AR that influences the expression of CERK in PCa.
Collapse
Affiliation(s)
- Laura Camacho
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Biochemistry and Molecular Biology Department, University of the Basque Country, 48040 Bilbao, Spain; (A.D.-H.); (A.G.-M.)
| | - Amaia Zabala-Letona
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ana R. Cortazar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ianire Astobiza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
| | - Asier Dominguez-Herrera
- Biochemistry and Molecular Biology Department, University of the Basque Country, 48040 Bilbao, Spain; (A.D.-H.); (A.G.-M.)
| | - Amaia Ercilla
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Jana Crespo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
| | - Cristina Viera
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
| | - Sonia Fernández-Ruiz
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ainara Martinez-Gonzalez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
| | - Veronica Torrano
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Biochemistry and Molecular Biology Department, University of the Basque Country, 48040 Bilbao, Spain; (A.D.-H.); (A.G.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Antonio Gomez-Muñoz
- Biochemistry and Molecular Biology Department, University of the Basque Country, 48040 Bilbao, Spain; (A.D.-H.); (A.G.-M.)
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (L.C.); (A.Z.-L.); (A.R.C.); (I.A.); (A.E.); (J.C.); (C.V.); (S.F.-R.); (A.M.-G.); (V.T.); (N.M.-M.)
- Biochemistry and Molecular Biology Department, University of the Basque Country, 48040 Bilbao, Spain; (A.D.-H.); (A.G.-M.)
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- Correspondence:
| |
Collapse
|
9
|
Moradi A, Srinivasan S, Clements J, Batra J. Beyond the biomarker role: prostate-specific antigen (PSA) in the prostate cancer microenvironment. Cancer Metastasis Rev 2020; 38:333-346. [PMID: 31659564 DOI: 10.1007/s10555-019-09815-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prostate-specific antigen (PSA) blood test is the accepted biomarker of tumor recurrence. PSA levels in serum correlate with disease progression, though its diagnostic accuracy is questionable. As a result, significant progress has been made in developing modified PSA tests such as PSA velocity, PSA density, 4Kscore, PSA glycoprofiling, Prostate Health Index, and the STHLM3 test. PSA, a serine protease, is secreted from the epithelial cells of the prostate. PSA has been suggested as a molecular target for prostate cancer therapy due to the fact that it is not only active in prostate tissue but also has a pivotal role on prostate cancer signaling pathways including proliferation, invasion, metastasis, angiogenesis, apoptosis, immune response, and tumor microenvironment regulation. Here, we summarize the current standing of PSA in prostate cancer progression as well as its utility in prostate cancer therapeutic approaches with an emphasis on the role of PSA in the tumor microenvironment.
Collapse
Affiliation(s)
- Afshin Moradi
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia. .,Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
10
|
Wu L, Xiang S, Hu X, Mo M, Zhao C, Cai Y, Tong S, Jiang H, Chen L, Wang Z, Xiong W, Ou Z. Prostate-specific antigen modulates the osteogenic differentiation of MSCs via the cadherin 11-Akt axis. Clin Transl Med 2020; 10:363-373. [PMID: 32508049 PMCID: PMC7240859 DOI: 10.1002/ctm2.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A high prevalence of osteoblastic bone metastases is characteristic of prostate cancer. Prostate-specific antigen (PSA) is a serine protease uniquely produced by prostate cancer cells and is an important serological marker for prostate cancer. However, whether PSA modulates the osteogenic process remains largely unknown. In this study, we explored the effect of PSA on modulating the osteoblastic differentiation of mesenchymal stem cells (MSCs). In this study, we used flow cytometry, CCK-8 assay, Alizarin red S (ARS) staining and quantification, alkaline phosphatase (ALP) activity and staining, Western blotting, and quantitative real-time PCR (qRT-PCR) to explore the effect of PSA on osteogenic differentiation of MSCs. RESULTS We first demonstrated that although PSA did not affect the proliferation, morphology, or phenotype of MSCs, it significantly promoted the osteogenic differentiation of MSCs in a concentration-dependent manner. Furthermore, we demonstrated that PSA promoted the osteogenic differentiation of MSCs by elevating the expression of Cadherin 11 in MSCs and, thus, activating the Akt signaling pathway. CONCLUSIONS In conclusion, we demonstrated that PSA could promote the osteogenesis of MSCs through Akt signaling pathway activation by elevating the expression of cadherin-11 in MSCs. These findings imply a possible role of PSA in osteoblastic bone metastases in prostate cancer.
Collapse
Affiliation(s)
- Longxiang Wu
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Shiqi Xiang
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaP.R. China
| | - Xiheng Hu
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Miao Mo
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Cheng Zhao
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Yi Cai
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Shiyu Tong
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Huichuan Jiang
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Linxiao Chen
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Zhi Wang
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Wei Xiong
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Zhenyu Ou
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| |
Collapse
|
11
|
Matin F, Jeet V, Srinivasan S, Cristino AS, Panchadsaram J, Clements JA, Batra J. MicroRNA-3162-5p-Mediated Crosstalk between Kallikrein Family Members Including Prostate-Specific Antigen in Prostate Cancer. Clin Chem 2019; 65:771-780. [PMID: 31018918 DOI: 10.1373/clinchem.2018.295824] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND MicroRNAs mediate biological processes through preferential binding to the 3' untranslated region (3' UTR) of target genes. Studies have shown their association with prostate cancer (PCa) risk through single-nucleotide polymorphisms (SNPs), known as miRSNPs. In a European cohort, 22 PCa risk-associated miRSNPs have been identified. The most significant miRSNP in the 3' UTR of Kallikrein-related peptidase 3 (KLK3) created a binding site for miR-3162-5p. Here we investigated the miR-3162-5p-KLK interaction and the clinical implication of miR-3162-5p in PCa. METHODS We tested the role of miR-3162-5p in PCa etiology using IncuCyte live-cell imaging and anchorage-independent growth assays. The effect of miR-3162-5p on KLK and androgen receptor (AR) expression was measured by RT-quantitative (q)PCR and target pulldown assays. KLK3 proteolytic activity was determined by DELFIA® immunoassay. Mass spectrometry identified pathways affected by miR-3162-5p. miR-3162-5p expression was measured in clinical samples using RT-qPCR. RESULTS miR-3162-5p affected proliferation, migration, and colony formation of LNCaP cells by regulating the expression of KLK2-4 and AR by direct targeting. KLK3 protein expression was regulated by miR-3162-5p consistent with lower KLK3 proteolytic activity observed in LNCaP-conditioned media. KLK/AR pulldown and mass spectrometry analysis showed a potential role of miR-3162-5p in metabolic pathways via KLK/AR and additional targets. Increased miR-3162-5p expression was observed in prostate tumor tissues with higher Gleason grade. CONCLUSIONS Our study provides an insight into possible involvement of miR-3162-5p in PCa etiology by targeting KLKs and AR. It highlights clinical utility of miR-3162-5p and its interactive axis as a new class of biomarkers and therapeutic targets for PCa.
Collapse
Affiliation(s)
- Farhana Matin
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | - Varinder Jeet
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | - Alexandre S Cristino
- University of Queensland Diamantina Institute (UQDI), Faculty of Medicine, Translational Research Institute, University of Queensland, Brisbane, Australia
| | - Janaththani Panchadsaram
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | | | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia;
| | | |
Collapse
|
12
|
Udager AM, Tomlins SA. Molecular Biomarkers in the Clinical Management of Prostate Cancer. Cold Spring Harb Perspect Med 2018; 8:a030601. [PMID: 29311125 PMCID: PMC6211380 DOI: 10.1101/cshperspect.a030601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer, one of the most common noncutaneous malignancies in men, is a heterogeneous disease with variable clinical outcome. Although the majority of patients harbor indolent tumors that are essentially cured by local therapy, subsets of patients present with aggressive disease or recur/progress after primary treatment. With this in mind, modern clinical approaches to prostate cancer emphasize the need to reduce overdiagnosis and overtreatment via personalized medicine. Advances in our understanding of prostate cancer pathogenesis, coupled with recent technologic innovations, have facilitated the development and validation of numerous molecular biomarkers, representing a range of macromolecules assayed from a variety of patient sample types, to help guide the clinical management of prostate cancer, including early detection, diagnosis, prognostication, and targeted therapeutic selection. Herein, we review the current state of the art regarding prostate cancer molecular biomarkers, emphasizing those with demonstrated utility in clinical practice.
Collapse
Affiliation(s)
- Aaron M Udager
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-5054
| | - Scott A Tomlins
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-5054
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan 48109-5948
- Comprehensive Cancer Center, Michigan Medicine, Ann Arbor, Michigan 48109-0944
- Michigan Center for Translational Pathology, Ann Arbor, Michigan 48109-5940
| |
Collapse
|
13
|
Thorek DLJ, Ku AT, Mitsiades N, Veach D, Watson PA, Metha D, Strand SE, Sharma SK, Lewis JS, Abou DS, Lilja HG, Larson SM, McDevitt MR, Ulmert D. Harnessing Androgen Receptor Pathway Activation for Targeted Alpha Particle Radioimmunotherapy of Breast Cancer. Clin Cancer Res 2018; 25:881-891. [PMID: 30254080 DOI: 10.1158/1078-0432.ccr-18-1521] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/30/2018] [Accepted: 09/17/2018] [Indexed: 01/26/2023]
Abstract
PURPOSE The impact of androgen receptor (AR) activity in breast cancer biology is unclear. We characterized and tested a novel therapy to an AR-governed target in breast cancer.Experimental Design: We evaluated the expression of prototypical AR gene products human kallikrein 2 (hK2) and PSA in breast cancer models. We screened 13 well-characterized breast cancer cell lines for hK2 and PSA production upon in vitro hormone stimulation by testosterone [dihydrotestosterone (DHT)]. AR-positive lines were further evaluated by exposure to estrogen (17β-Estradiol) and the synthetic progestin D-Norgestrel. We then evaluated an anti-hK2-targeted radiotherapy platform (hu11B6), labeled with alpha (α)-particle emitting Actinium-225, to specifically treat AR-expressing breast cancer xenografts under hormone stimulation. RESULTS D-Norgestrel and DHT activated the AR pathway, while 17β-Estradiol did not. Competitive binding for AR protein showed similar affinity between DHT and D-Norgestrel, indicating direct AR-ligand interaction. In vivo production of hK2 was sufficient to achieve site-specific delivery of therapeutic radionuclide to tumor tissue at >20-fold over background muscle uptake; effecting long-term local tumor control. CONCLUSIONS [225Ac]hu11B6 targeted radiotherapy was potentiated by DHT and by D-Norgestrel in murine xenograft models of breast cancer. AR activity in breast cancer correlates with kallikrein-related peptidase-2 and can be activated by D-Norgestrel, a common contraceptive, and AR induction can be harnessed for hK2-targeted breast cancer α-emitter radiotherapy.
Collapse
Affiliation(s)
- Daniel L J Thorek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Anson T Ku
- Division of Clinical Chemistry, Department of Translational Medicine, Lund University, Lund, Sweden.,Clinical Research Center, Lund University, Malmö, Sweden
| | - Nicholas Mitsiades
- Departments of Medicine and Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Darren Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Philip A Watson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dipti Metha
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sven-Erik Strand
- Division of Oncology and Pathology, and Medical Radiation Physics, Department of Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Sai Kiran Sharma
- Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York.,Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Diane S Abou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Hans G Lilja
- Departments of Laboratory Medicine, Surgery and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Nuffield Department of Surgical Sciences, Oxford University, John Radcliffe Hospital, Headington, Oxford, United Kingdom.,Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York.,Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York.,Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael R McDevitt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - David Ulmert
- Division of Oncology and Pathology, and Medical Radiation Physics, Department of Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden. .,Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, California.,Jonsson Comprehensive Cancer Center, David Geffen UCLA School of Medicine, Los Angeles, California.,Ahmanson Translational Imaging Division, David Geffen UCLA School of Medicine, Los Angeles, California
| |
Collapse
|
14
|
Filippou PS, Ren AH, Soosaipillai A, Papaioannou MD, Korbakis D, Safar R, Diamandis EP, Conner JR. Expression profile of human tissue kallikrein 15 provides preliminary insights into its roles in the prostate and testis. Clin Biochem 2018; 59:78-85. [DOI: 10.1016/j.clinbiochem.2018.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/07/2018] [Accepted: 06/25/2018] [Indexed: 02/02/2023]
|
15
|
Electrochemical immunoassay for detection of prostate specific antigen based on peptide nanotube-gold nanoparticle-polyaniline immobilized pencil graphite electrode. J Colloid Interface Sci 2018; 510:318-326. [DOI: 10.1016/j.jcis.2017.09.079] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 01/24/2023]
|
16
|
Thorek DLJ, Watson PA, Lee SG, Ku AT, Bournazos S, Braun K, Kim K, Sjöström K, Doran MG, Lamminmäki U, Santos E, Veach D, Turkekul M, Casey E, Lewis JS, Abou DS, van Voss MRH, Scardino PT, Strand SE, Alpaugh ML, Scher HI, Lilja H, Larson SM, Ulmert D. Internalization of secreted antigen-targeted antibodies by the neonatal Fc receptor for precision imaging of the androgen receptor axis. Sci Transl Med 2017; 8:367ra167. [PMID: 27903863 DOI: 10.1126/scitranslmed.aaf2335] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 07/14/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022]
Abstract
Targeting the androgen receptor (AR) pathway prolongs survival in patients with prostate cancer, but resistance rapidly develops. Understanding this resistance is confounded by a lack of noninvasive means to assess AR activity in vivo. We report intracellular accumulation of a secreted antigen-targeted antibody (SATA) that can be used to characterize disease, guide therapy, and monitor response. AR-regulated human kallikrein-related peptidase 2 (free hK2) is a prostate tissue-specific antigen produced in prostate cancer and androgen-stimulated breast cancer cells. Fluorescent and radio conjugates of 11B6, an antibody targeting free hK2, are internalized and noninvasively report AR pathway activity in metastatic and genetically engineered models of cancer development and treatment. Uptake is mediated by a mechanism involving the neonatal Fc receptor. Humanized 11B6, which has undergone toxicological tests in nonhuman primates, has the potential to improve patient management in these cancers. Furthermore, cell-specific SATA uptake may have a broader use for molecularly guided diagnosis and therapy in other cancers.
Collapse
Affiliation(s)
- Daniel L J Thorek
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philip A Watson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sang-Gyu Lee
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anson T Ku
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stylianos Bournazos
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA
| | - Katharina Braun
- Department of Urology, University Hospital of the Ruhr-University of Bochum, Marien Hospital Herne, Herne, Germany
| | - Kwanghee Kim
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Michael G Doran
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Urpo Lamminmäki
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Elmer Santos
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Darren Veach
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mesruh Turkekul
- Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Emily Casey
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason S Lewis
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Diane S Abou
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Marise R H van Voss
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Peter T Scardino
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Sven-Erik Strand
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Mary L Alpaugh
- Departments of Biology and Biomedical and Translational Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hans Lilja
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, U.K.,Department of Laboratory Medicine, Lund University, Malmö, Sweden
| | - Steven M Larson
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - David Ulmert
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Division of Urological Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
17
|
Muñoz D, Serrano MK, Hernandez ME, Haller R, Swanson T, Slaton JW, Sinha AA, Wilson MJ. Matrix metalloproteinase and heparin-stimulated serine proteinase activities in post-prostate massage urine of men with prostate cancer. Exp Mol Pathol 2017; 103:300-305. [DOI: 10.1016/j.yexmp.2017.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/15/2017] [Indexed: 10/18/2022]
|
18
|
Renné T, Scorilas A, Schmitt M. The kallikreins: old proteases with new clinical potentials. Thromb Haemost 2017; 110:396-8. [DOI: 10.1160/th13-07-0583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/19/2013] [Indexed: 11/05/2022]
|
19
|
Koistinen H, Wallén E, Ylikangas H, Meinander K, Lahtela-Kakkonen M, Närvänen A, Stenman UH. Development of molecules stimulating the activity of KLK3 - an update. Biol Chem 2017; 397:1229-1235. [PMID: 27383882 DOI: 10.1515/hsz-2016-0189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/30/2016] [Indexed: 01/03/2023]
Abstract
Kallikrein-related peptidase-3 (KLK3, known also as prostate-specific antigen, PSA) is highly expressed in the prostate. KLK3 possess antiangiogenic activity, which we have found to be related to its proteolytic activity. Thus, it may be possible to slow down the growth of prostatic tumors by enhancing this activity. We have developed peptides that enhance the proteolytic activity of KLK3. As these peptides are degraded in circulation and rapidly excreted, we have started to modify them and have succeeded in creating bioactive and more stable pseudopeptides. We have also identified small molecules stimulating the activity of KLK3, especially in synergy with peptides.
Collapse
|
20
|
Falco M, Palma G, Rea D, De Biase D, Scala S, D'Aiuto M, Facchini G, Perdonà S, Barbieri A, Arra C. Tumour biomarkers: homeostasis as a novel prognostic indicator. Open Biol 2016; 6:160254. [PMID: 27927793 PMCID: PMC5204124 DOI: 10.1098/rsob.160254] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
The term 'personalized medicine' refers to a medical procedure that consists in the grouping of patients based on their predicted individual response to therapy or risk of disease. In oncologic patients, a 'tailored' therapeutic approach may potentially improve their survival and well-being by not only reducing the tumour, but also enhancing therapeutic response and minimizing the adverse effects. Diagnostic tests are often used to select appropriate and optimal therapies that rely both on patient genome and other molecular/cellular analysis. Several studies have shown that lifestyle and environmental factors can influence the epigenome and that epigenetic events may be involved in carcinogenesis. Thus, in addition to traditional biomarkers, epigenetic factors are raising considerable interest, because they could potentially be used as an excellent tool for cancer diagnosis and prognosis. In this review, we summarize the role of conventional cancer genetic biomarkers and their association with epigenomics. Furthermore, we will focus on the so-called 'homeostatic biomarkers' that result from the physiological response to cancer, emphasizing the concept that an altered 'new' homeostasis influence not only tumour environment, but also the whole organism.
Collapse
Affiliation(s)
- Michela Falco
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Giuseppe Palma
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Domenica Rea
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples 'Federico II', Via Delpino 1, 80137 Naples, Italy
| | - Stefania Scala
- Molecular lmmunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, IRCCS Naples 'Fondazione G. Pascale', Naples, italy, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Massimiliano D'Aiuto
- Division of Breast Surgery, Department of Breast Disease, National Cancer Institute, IRCCS, 'Fondazione Pascale', Naples, Italy
| | - Gaetano Facchini
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, , Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Antonio Barbieri
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Claudio Arra
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| |
Collapse
|
21
|
Bachour DM, Chahin E, Al-Fahoum S. Human Kallikrein-2, Prostate Specific Antigen and Free- Prostate Specific Antigen in Combination to Discriminate Prostate Cancer from Benign Diseases in Syrian Patients. Asian Pac J Cancer Prev 2016; 16:7085-8. [PMID: 26514494 DOI: 10.7314/apjcp.2015.16.16.7085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The high incidence of prostate cancer as the most common malignancy in males in many countries raises the question of developing reliable detection tests. The prostate specific antigen (PSA) test is the most widely used for screening for prostate cancer; however, its low specificity elevates the number of unnecessarily biopsies. Serum human kallikrein-2 (hK2) is considered as a promising marker, and especially its ratio to fPSA, for predicting the presence of malignancy to select the best choice referring to biopsy or surveillance. In this study, we investigated the role of hK2 and its combinations with other markers to discriminate prostate cancer from benign diseases in Syrian patients. MATERIALS AND METHODS In this prospective oriented cross-sectional cohort study, serum samples were collected from patients referred to many Hospitals in Damascus, Syria, between May 2011 and March 2012, and diagnosed with biopsy proven benign prostate hyperplasia (BPH) or prostate cancer (PCa). Serum was analyzed for hK2, PSA and fPSA, and the ratios of fPSA/PSA and hK2/fPSA were calculated. RESULTS We found that mean hK2/fPSA ratios were significantly higher (P=0.01) in prostate cancer patients than in the BPH or control groups. Also the ratio hk2/fPSA gave the largest area under the curve (AUC:0.96) which was significantly larger than for fPSA/PSA (AUC:0.41) indicative of higher specificity. CONCLUSIONS Our results demonstrate that the ratio of hK2/fPSA might be superior to the use of fPSA/PSA alone. The hK2 could be shown to enhance the early detection of prostate cancer; especially the ratio hK2/fPSA improves specificity and hence may reduce the number of negative biopsies.
Collapse
Affiliation(s)
- Dala-Maria Bachour
- Master Student, Faculty of Pharmacy, Damascus University, Syria E-mail :
| | | | | |
Collapse
|
22
|
Ylikangas H, Mattsson JM, Stenman UH, Koistinen H, Poso A, Lahtela-Kakkonen M. Virtual Screening of Small Drug-Like Compounds Stimulating the Enzymatic Activity of Kallikrein-Related Peptidase 3 (KLK3). ChemMedChem 2016; 11:2043-9. [DOI: 10.1002/cmdc.201600181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/29/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Henna Ylikangas
- School of Pharmacy; University of Eastern Finland; P.O. Box 1627 70211 Kuopio Finland
| | - Johanna M. Mattsson
- Department of Clinical Chemistry; Biomedicum Helsinki; University ofHelsinki and; Helsinki University Central Hospital; 00014 Helsinki Finland
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry; Biomedicum Helsinki; University ofHelsinki and; Helsinki University Central Hospital; 00014 Helsinki Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry; Biomedicum Helsinki; University ofHelsinki and; Helsinki University Central Hospital; 00014 Helsinki Finland
| | - Antti Poso
- School of Pharmacy; University of Eastern Finland; P.O. Box 1627 70211 Kuopio Finland
- University Hospital Tübingen; Department of Internal Medicine 1; Division of Molecular Oncology of Solid Tumors; Otfried-Müller-Strasse 10 72076 Tübingen Germany
| | | |
Collapse
|
23
|
Verze P, Cai T, Lorenzetti S. The role of the prostate in male fertility, health and disease. Nat Rev Urol 2016; 13:379-86. [PMID: 27245504 DOI: 10.1038/nrurol.2016.89] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
24
|
Involvement of Kallikrein-Related Peptidases in Normal and Pathologic Processes. DISEASE MARKERS 2015; 2015:946572. [PMID: 26783378 PMCID: PMC4689925 DOI: 10.1155/2015/946572] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/16/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022]
Abstract
Human kallikrein-related peptidases (KLKs) are a subgroup of serine proteases that participate in proteolytic pathways and control protein levels in normal physiology as well as in several pathological conditions. Their complex network of stimulatory and inhibitory interactions may induce inflammatory and immune responses and contribute to the neoplastic phenotype through the regulation of several cellular processes, such as proliferation, survival, migration, and invasion. This family of proteases, which includes one of the most useful cancer biomarkers, kallikrein-related peptidase 3 or PSA, also has a protective effect against cancer promoting apoptosis or counteracting angiogenesis and cell proliferation. Therefore, they represent attractive therapeutic targets and may have important applications in clinical oncology. Despite being intensively studied, many gaps in our knowledge on several molecular aspects of KLK functions still exist. This review aims to summarize recent data on their involvement in different processes related to health and disease, in particular those directly or indirectly linked to the neoplastic process.
Collapse
|
25
|
Stephan C, Jung K, Ralla B. Current biomarkers for diagnosing of prostate cancer. Future Oncol 2015; 11:2743-55. [DOI: 10.2217/fon.15.203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is mostly detected by prostate-specific antigen (PSA) as one of the most widely used tumor markers. But PSA is limited with its low specificity. The prostate health index (phi) can improve specificity over percent free and total PSA and correlates with aggressive cancer. The urinary PCA3 also shows its utility to detect PCa but its correlation with aggressiveness and the low sensitivity at high values are limitations. While the detection of alterations of the androgen-regulated TMPRSS2 and ETS transcription factor genes in tissue of ˜50% of all PCa patients was one research milestone, the urinary assay should only be used in combination with PCA3. Both US FDA-approved markers phi and PCA3 perform equally.
Collapse
Affiliation(s)
- Carsten Stephan
- Department of Urology, Charité – Universitätsmedizin Berlin, CCM, Charitéplatz 1, D-10117 Berlin, Germany
- Berlin Institute for Urologic Research, Berlin, Germany
| | - Klaus Jung
- Department of Urology, Charité – Universitätsmedizin Berlin, CCM, Charitéplatz 1, D-10117 Berlin, Germany
- Berlin Institute for Urologic Research, Berlin, Germany
| | - Bernhard Ralla
- Department of Urology, Charité – Universitätsmedizin Berlin, CCM, Charitéplatz 1, D-10117 Berlin, Germany
| |
Collapse
|
26
|
Cotton RW, Fisher MB. Review: Properties of sperm and seminal fluid, informed by research on reproduction and contraception. Forensic Sci Int Genet 2015; 18:66-77. [DOI: 10.1016/j.fsigen.2015.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/12/2022]
|
27
|
Portillo-Lara R, Alvarez MM. Enrichment of the Cancer Stem Phenotype in Sphere Cultures of Prostate Cancer Cell Lines Occurs through Activation of Developmental Pathways Mediated by the Transcriptional Regulator ΔNp63α. PLoS One 2015; 10:e0130118. [PMID: 26110651 PMCID: PMC4481544 DOI: 10.1371/journal.pone.0130118] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/18/2015] [Indexed: 11/17/2022] Open
Abstract
Background Cancer stem cells (CSC) drive prostate cancer tumor survival and metastasis. Nevertheless, the development of specific therapies against CSCs is hindered by the scarcity of these cells in prostate tissues. Suspension culture systems have been reported to enrich CSCs in primary cultures and cell lines. However, the molecular mechanisms underlying this phenomenon have not been fully explored. Methodology/Principal Findings We describe a prostasphere assay for the enrichment of CD133+ CSCs in four commercial PCa cell lines: 22Rv1, DU145, LNCaP, and PC3. Overexpression of CD133, as determined by flow cytometric analysis, correlated with an increased clonogenic, chemoresistant, and invasive potential in vitro. This phenotype is concordant to that of CSCs in vivo. Gene expression profiling was then carried out using the Cancer Reference panel and the nCounter system from NanoString Technologies. This analysis revealed several upregulated transcripts that can be further explored as potential diagnostic markers or therapeutic targets. Furthermore, functional annotation analysis suggests that ΔNp63α modulates the activation of developmental pathways responsible for the increased stem identity of cells growing in suspension cultures. Conclusions/Significance We conclude that profiling the genetic mechanisms involved in CSC enrichment will help us to better understand the molecular pathways that underlie CSC pathophysiology. This platform can be readily adapted to enrich and assay actual patient samples, in order to design patient-specific therapies that are aimed particularly against CSCs.
Collapse
Affiliation(s)
- Roberto Portillo-Lara
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León, México
| | - Mario Moisés Alvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León, México; Harvard-MIT Health Sciences and Technology, Brigham and Women's Hospital, Cambridge, Massachusetts, United States of America
| |
Collapse
|
28
|
Stegeman S, Amankwah E, Klein K, O'Mara TA, Kim D, Lin HY, Permuth-Wey J, Sellers TA, Srinivasan S, Eeles R, Easton D, Kote-Jarai Z, Amin Al Olama A, Benlloch S, Muir K, Giles GG, Wiklund F, Gronberg H, Haiman CA, Schleutker J, Nordestgaard BG, Travis RC, Neal D, Pharoah P, Khaw KT, Stanford JL, Blot WJ, Thibodeau S, Maier C, Kibel AS, Cybulski C, Cannon-Albright L, Brenner H, Kaneva R, Teixeira MR, Spurdle AB, Clements JA, Park JY, Batra J. A Large-Scale Analysis of Genetic Variants within Putative miRNA Binding Sites in Prostate Cancer. Cancer Discov 2015; 5:368-79. [PMID: 25691096 PMCID: PMC4390388 DOI: 10.1158/2159-8290.cd-14-1057] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/02/2015] [Indexed: 02/02/2023]
Abstract
UNLABELLED Prostate cancer is the second most common malignancy among men worldwide. Genome-wide association studies have identified 100 risk variants for prostate cancer, which can explain approximately 33% of the familial risk of the disease. We hypothesized that a comprehensive analysis of genetic variations found within the 3' untranslated region of genes predicted to affect miRNA binding (miRSNP) can identify additional prostate cancer risk variants. We investigated the association between 2,169 miRSNPs and prostate cancer risk in a large-scale analysis of 22,301 cases and 22,320 controls of European ancestry from 23 participating studies. Twenty-two miRSNPs were associated (P<2.3×10(-5)) with risk of prostate cancer, 10 of which were within 7 genes previously not mapped by GWAS studies. Further, using miRNA mimics and reporter gene assays, we showed that miR-3162-5p has specific affinity for the KLK3 rs1058205 miRSNP T-allele, whereas miR-370 has greater affinity for the VAMP8 rs1010 miRSNP A-allele, validating their functional role. SIGNIFICANCE Findings from this large association study suggest that a focus on miRSNPs, including functional evaluation, can identify candidate risk loci below currently accepted statistical levels of genome-wide significance. Studies of miRNAs and their interactions with SNPs could provide further insights into the mechanisms of prostate cancer risk.
Collapse
Affiliation(s)
- Shane Stegeman
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Ernest Amankwah
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Kerenaftali Klein
- Statistics Unit, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Tracy A O'Mara
- Molecular Cancer Epidemiology Laboratory, Genetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Donghwa Kim
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Hui-Yi Lin
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | | | - Thomas A Sellers
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Srilakshmi Srinivasan
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Rosalind Eeles
- The Institute of Cancer Research, London, United Kingdom. Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, United Kingdom
| | - Doug Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | | | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | - Sara Benlloch
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | | | - Graham G Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria, Australia. Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Johanna Schleutker
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland. Institute of Biomedical Technology/BioMediTech, University of Tampere and FimLab Laboratories, Tampere, Finland
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - David Neal
- Surgical Oncology (Uro-Oncology: S4), University of Cambridge, Addenbrooke's Hospital, Cambridge; Cancer Research UK, Cambridge Research Institute, Cambridge, United Kingdom
| | - Paul Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | - Kay-Tee Khaw
- Cambridge Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | - William J Blot
- International Epidemiology Institute, Rockville, Maryland
| | | | - Christiane Maier
- Department of Urology, University Hospital Ulm, Ulm, Germany. Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Adam S Kibel
- Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts. Washington University, St. Louis, Missouri
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Lisa Cannon-Albright
- Division of Genetic Epidemiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Radka Kaneva
- Molecular Medicine Center and Department of Medical Chemistry and Biochemistry, Medical University-Sofia, Sofia, Bulgaria
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute, Porto; Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Amanda B Spurdle
- Molecular Cancer Epidemiology Laboratory, Genetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Jyotsna Batra
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
29
|
LeBeau AM, Denmeade SR. Protease-activated pore-forming peptides for the treatment and imaging of prostate cancer. Mol Cancer Ther 2014; 14:659-68. [PMID: 25537662 DOI: 10.1158/1535-7163.mct-14-0744] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023]
Abstract
A common hallmark of cancers with highly aggressive phenotypes is increased proteolysis in the tumor and the surrounding microenvironment. Prostate cancer has a number of proteases uniquely associated with it that may play various important roles in disease progression. In this report, we utilize the peritumoral proteolytic activity of prostate cancer to activate engineered peptide constructs for the treatment and noninvasive imaging of prostate cancer. Using a modular "propeptide" approach, a cationic diastereomeric pore-forming peptide domain was linked to an inactivating acidic peptide domain. The inactivating acidic peptide domain was engineered to be a cleavable substrate for the secreted serine protease prostate-specific antigen (PSA) or the transmembrane metalloprotease prostate-specific membrane antigen (PSMA). The propeptides were then evaluated in a direct comparison study. Both the PSA and PSMA activated propeptides were found to be cytotoxic to prostate cancer cells in vitro. In vivo, however, treatment of LNCaP and CWR22Rv1 xenografts with the PSMA propeptide resulted in a pronounced cytostatic effect when compared with xenografts treated with the PSA propeptide or the cationic diastereomeric peptide alone. The PSMA activated propeptide also proved to be an effective optical imaging probe in vivo when labeled with a near-infrared fluorophore. These data suggest that protease-activated pore-forming peptides could potentially be used for both imaging and treating prostate cancer.
Collapse
Affiliation(s)
- Aaron M LeBeau
- Department of Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota.
| | - Samuel R Denmeade
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
30
|
Vilhelmsson-Timmermand O, Santos E, Thorek DLJ, Evans-Axelsson S, Bjartell A, Lilja H, Larson SM, Strand SE, Tran TA, Ulmert D. Radiolabeled antibodies in prostate cancer: a case study showing the effect of host immunity on antibody bio-distribution. Nucl Med Biol 2014; 42:375-80. [PMID: 25577038 DOI: 10.1016/j.nucmedbio.2014.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 11/05/2014] [Accepted: 12/17/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVES Human tumors xenografted in immunodeficient mice are crucial models in nuclear medicine to evaluate the effectiveness of candidate diagnostic and therapeutic compounds. However, little attention has been focused on the biological profile of the host model and its potential effects on the bio-distribution and tumor targeting of the tracer compound under study. We specifically investigated the dissimilarity in bio-distribution of (111)In-DTPA-5A10, which targets free prostate specific antigen (fPSA), in two animal models. METHODS In vivo bio-distribution studies of (111)In-DTPA-5A10 were performed in immunodeficient BALB/c-nu or NMRI-nu mice with subcutaneous (s.c.) LNCaP tumors. Targeting-specificity of the tracer was assessed by quantifying the uptake in (a) mice with s.c. xenografts of PSA-negative DU145 cells as well as (b) BALB/c-nu or NMRI-nu mice co-injected with an excess of non-labeled 5A10. Finally, the effect of neonatal Fc-receptor (FcRn) inhibition on the bio-distribution of the conjugate was studied by saturating FcRn-binding capacity with nonspecific IgG1. RESULTS The inherent biological attributes of the mouse model substantially influenced the bio-distribution and pharmacokinetics of (111)In-DTPA-5A10. With LNCaP xenografts in BALB/c-nu mice (with intact B and NK cells but with deficient T cells) versus NMRI-nu mice (with intact B cells, increased NK cells and absent T cells), we observed a significantly higher hepatic accumulation (26 ± 3.9 versus 3.5 ± 0.4%IA/g respectively), and concomitantly lower tumor uptake (25 ± 11 versus 52 ± 10%IA/g respectively) in BALB/c-nu mice. Inhibiting FcRn by administration of nonspecific IgG1 just prior to (111)In-DTPA-5A10 did not change tumor accumulation significantly. CONCLUSIONS We demonstrated that the choice of immunodeficient mouse model importantly influence the bio-distribution of (111)In-DTPA-5A10. This study further highlighted important considerations in the evaluation of preclinical tracers, with respect to gaining information on their performance in the translational setting. Investigators utilizing xenograft models need to assess not only radiolabeling strategies, but also the host immunological status.
Collapse
Affiliation(s)
| | - Elmer Santos
- Department of Radiology (Nuclear Medicine Service), Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Daniel L J Thorek
- Department of Radiology and Radiological Sciences, Division of Nuclear Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Evans-Axelsson
- Department of Clinical Sciences, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Anders Bjartell
- Department of Clinical Sciences, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Hans Lilja
- Department of Surgery (Urology), Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Departments of Laboratory Medicine and Medicine (GU Oncology), Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom; Department of Laboratory Medicine, Lund University, Malmö, Sweden
| | - Steven M Larson
- Department of Radiology (Nuclear Medicine Service), Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Sven-Erik Strand
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Thuy A Tran
- Department of Medical Radiation Physics, Lund University, Lund, Sweden; Lund University Bioimaging Center, Lund University, Lund, Sweden
| | - David Ulmert
- Department of Surgery (Urology), Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Department of Clinical Sciences, Division of Urological Research, Lund University, Malmö, Sweden.
| |
Collapse
|
31
|
Mattsson JM, Ravela S, Hekim C, Jonsson M, Malm J, Närvänen A, Stenman UH, Koistinen H. Proteolytic activity of prostate-specific antigen (PSA) towards protein substrates and effect of peptides stimulating PSA activity. PLoS One 2014; 9:e107819. [PMID: 25237904 PMCID: PMC4169579 DOI: 10.1371/journal.pone.0107819] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/15/2014] [Indexed: 11/18/2022] Open
Abstract
Prostate-specific antigen (PSA or kallikrein-related peptidase-3, KLK3) exerts chymotrypsin-like proteolytic activity. The main biological function of PSA is the liquefaction of the clot formed after ejaculation by cleavage of semenogelins I and II in seminal fluid. PSA also cleaves several other substrates, which may explain its putative functions in prostate cancer and its antiangiogenic activity. We compared the proteolytic efficiency of PSA towards several protein and peptide substrates and studied the effect of peptides stimulating the activity of PSA with these substrates. An endothelial cell tube formation model was used to analyze the effect of PSA-degraded protein fragments on angiogenesis. We showed that PSA degrades semenogelins I and II much more efficiently than other previously identified protein substrates, e.g., fibronectin, galectin-3 and IGFBP-3. We identified nidogen-1 as a new substrate for PSA. Peptides B2 and C4 that stimulate the activity of PSA towards small peptide substrates also enhanced the proteolytic activity of PSA towards protein substrates. Nidogen-1, galectin-3 or their fragments produced by PSA did not have any effect on endothelial cell tube formation. Although PSA cleaves several other protein substrates, in addition to semenogelins, the physiological importance of this activity remains speculative. The PSA levels in prostate are very high, but several other highly active proteases, such as hK2 and trypsin, are also expressed in the prostate and may cleave protein substrates that are weakly cleaved by PSA.
Collapse
Affiliation(s)
- Johanna M. Mattsson
- Department of Clinical Chemistry, Biomedicum Helsinki, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Suvi Ravela
- Department of Clinical Chemistry, Biomedicum Helsinki, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Can Hekim
- Department of Clinical Chemistry, Biomedicum Helsinki, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Magnus Jonsson
- Department of Laboratory Medicine, Section for Clinical Chemistry, Lund University and Laboratory Medicine Skåne, Lund, Sweden
| | - Johan Malm
- Department of Laboratory Medicine, Section for Clinical Chemistry, Lund University and Laboratory Medicine Skåne, Lund, Sweden
| | - Ale Närvänen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry, Biomedicum Helsinki, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry, Biomedicum Helsinki, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- * E-mail:
| |
Collapse
|
32
|
Timmermand OV, Ulmert D, Evans-Axelsson S, Pettersson K, Bjartell A, Lilja H, Strand SE, Tran TA. Preclinical imaging of kallikrein-related peptidase 2 (hK2) in prostate cancer with a (111)In-radiolabelled monoclonal antibody, 11B6. EJNMMI Res 2014; 4:51. [PMID: 26116115 PMCID: PMC4452661 DOI: 10.1186/s13550-014-0051-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/07/2014] [Indexed: 12/01/2022] Open
Abstract
Background Prostate cancer is a leading cause of death in the male population of the western world. Human kallikrein-related peptidase 2 (hK2) is abundantly expressed in malignant prostatic tissue, and its gene, KLK2, is regulated by the androgen receptor. 11B6 is a murine IgG1 monoclonal antibody directed against free human hK2. In this study, we performed a preclinical evaluation of 111In-labelled 11B6 in mouse xenografts to investigate its potential in the clinical staging and assessment of metastatic prostate cancer. Methods 11B6 was radiolabelled with 111In through CHX-A″-DTPA chelation. In vivo biodistribution and uptake of 111In-DTPA-11B6 were measured until 168 h post-injection in NMRI nude mice bearing subcutaneous LNCaP xenografts. The binding specificity to hK2 was evaluated by both in vivo competitive binding assays with excess non-labelled 11B6 and hK2-negative DU145 xenografts. SPECT/CT imaging of subcutaneous and intra-tibial LNCaP xenografts was used to visualize the tumours. Results Tumour uptake of 111In-DTPA-11B6 in LNCaP xenografts was 19% ± 0.78%IA/g at 48 h, giving a tumour-to-blood ratio of 1.6, which increases to 2.4 at 1 week post-injection. Accumulation was low in other organs except for the salivary glands, which is probably the result of cross-reactivity with mouse kallikreins. Significantly lower tumour accumulation was observed in competitive assays and DU145 xenografts. SPECT/CT imaging could clearly visualize the subcutaneous and intra-tibial LNCaP xenografts. Conclusions Our study demonstrates the potential of 111In-DTPA-11B6 for the detection of metastatic prostate cancer and monitoring anti-androgen therapy, as it exhibits an increased uptake and accumulation in viable tumour when compared to normal tissue. A humanised version of the 11B6 monoclonal antibody is currently under evaluation. Electronic supplementary material The online version of this article (doi:10.1186/s13550-014-0051-5) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Fuhrman-Luck RA, Silva ML, Dong Y, Irving-Rodgers H, Stoll T, Hastie ML, Loessner D, Gorman JJ, Clements JA. Proteomic and other analyses to determine the functional consequences of deregulated kallikrein-related peptidase (KLK) expression in prostate and ovarian cancer. Proteomics Clin Appl 2014; 8:403-15. [PMID: 24535680 DOI: 10.1002/prca.201300098] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/23/2013] [Accepted: 11/30/2013] [Indexed: 02/06/2023]
Abstract
Rapidly developing proteomic tools are improving detection of deregulated kallikrein-related peptidase (KLK) expression, at the protein level, in prostate and ovarian cancer, as well as facilitating the determination of functional consequences downstream. MS-driven proteomics uniquely allows for the detection, identification, and quantification of thousands of proteins in a complex protein pool, and this has served to identify certain KLKs as biomarkers for these diseases. In this review, we describe applications of this technology in KLK biomarker discovery and elucidate MS-based techniques that have been used for unbiased, global screening of KLK substrates within complex protein pools. Although MS-based KLK degradomic studies are limited to date, they helped to discover an array of novel KLK substrates. Substrates identified by MS-based degradomics are reported with improved confidence over those determined by incubating a purified or recombinant substrate and protease of interest, in vitro. We propose that these novel proteomic approaches represent the way forward for KLK research, in order to correlate proteolysis of biological substrates with tissue-related consequences, toward clinical targeting of KLK expression and function for cancer diagnosis, prognosis, and therapies.
Collapse
Affiliation(s)
- Ruth Anna Fuhrman-Luck
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Prostate-Specific Antigen (PSA) Screening and New Biomarkers for Prostate Cancer (PCa). EJIFCC 2014; 25:55-78. [PMID: 27683457 PMCID: PMC4975191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PSA screening reduces PCa-mortality but the disadvantages overdiagnosis and overtreatment require multivariable risk-prediction tools to select appropriate treatment or active surveillance. This review explains the differences between the two largest screening trials and discusses the drawbacks of screening and its meta-analysisxs. The current American and European screening strategies are described. Nonetheless, PSA is one of the most widely used tumor markers and strongly correlates with the risk of harboring PCa. However, while PSA has limitations for PCa detection with its low specificity there are several potential biomarkers presented in this review with utility for PCa currently being studied. There is an urgent need for new biomarkers especially to detect clinically significant and aggressive PCa. From all PSA-based markers, the FDA-approved prostate health index (phi) shows improved specificity over percent free and total PSA. Another kallikrein panel, 4K, which includes KLK2 has recently shown promise in clinical research studies but has not yet undergone formal validation studies. In urine, prostate cancer gene 3 (PCA3) has also been validated and approved by the FDA for its utility to detect PCa. The potential correlation of PCA3 with cancer aggressiveness requires more clinical studies. The detection of the fusion of androgen-regulated genes with genes of the regulatory transcription factors in tissue of (~)50% of all PCa-patients is a milestone in PCa research. A combination of the urinary assays for TMPRSS2:ERG gene fusion and PCA3 shows an improved accuracy for PCa detection. Overall, the field of PCa biomarker discovery is very exciting and prospective.
Collapse
|
35
|
Stephan C, Ralla B, Jung K. Prostate-specific antigen and other serum and urine markers in prostate cancer. Biochim Biophys Acta Rev Cancer 2014; 1846:99-112. [PMID: 24727384 DOI: 10.1016/j.bbcan.2014.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 03/24/2014] [Accepted: 04/01/2014] [Indexed: 11/16/2022]
Abstract
Prostate-specific antigen (PSA) is one of the most widely used tumor markers, and strongly correlates with the risk of harboring from prostate cancer (PCa). This risk is visible already several years in advance but PSA has severe limitations for PCa detection with its low specificity and low negative predictive value. There is an urgent need for new biomarkers especially to detect clinically significant and aggressive PCa. From all PSA-based markers, the FDA-approved Prostate Health Index (phi) shows improved specificity over percent free and total PSA. Other serum kallikreins or sarcosine in serum or urine show more diverging data. In urine, the FDA-approved prostate cancer gene 3 (PCA3) has also proven its utility in the detection and management of early PCa. However, some aspects on its correlation with aggressiveness and the low sensitivity at very high values have to be re-examined. The detection of a fusion of the androgen regulated TMPRSS2 gene with the ERG oncogene (from the ETS family), which acts as transcription factor gene, in tissue of ~50% of all PCa patients was one milestone in PCa research. When combining the urinary assays for TMPRSS2:ERG and PCA3, an improved accuracy for PCa detection is visible. PCA3 and phi as the best available PCa biomarkers show an equal performance in direct comparisons.
Collapse
Affiliation(s)
- Carsten Stephan
- Department of Urology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute for Urologic Research, Berlin, Germany.
| | - Bernhard Ralla
- Department of Urology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus Jung
- Department of Urology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute for Urologic Research, Berlin, Germany
| |
Collapse
|
36
|
Duffy MJ. PSA in Screening for Prostate Cancer. Adv Clin Chem 2014. [DOI: 10.1016/b978-0-12-801401-1.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|