1
|
Cai S, Deng Y, Zou Z, Tian W, Tang Z, Li J, Tan Z, Wu Z, Han Z, Wen B, Feng Y, Liu R, Zhu X, Wu Y, Xiao H, He H, Ye J, Zhong W. Metformin inhibits the progression of castration-resistant prostate cancer by regulating PDE6D induced purine metabolic alternation and cGMP / PKG pathway activation. Cancer Lett 2025; 622:217694. [PMID: 40216151 DOI: 10.1016/j.canlet.2025.217694] [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: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
The castration-resistant prostate cancer (CRPC) remains an incurable disease. Metformin has demonstrated a potential therapeutic effect on CRPC. However, the poor clinical performance of metformin against cancer may be due to its clinical dose being much lower than the anticancer concentration used in pre-clinical experiments. The challenge is to determine a way to enhance sensitivity to metformin at an appropriate concentration on CRPC. In this study, a mouse model of low-dose metformin treatment for CRPC cells were established. Metabolomic-seq and transcriptomic-seq was used to investigate changes in CRPC xenografts. We discovered that low-dose metformin inhibits the progression of CRPC by regulating PDE6D, which induces alterations in purine metabolism and activates the cGMP/PKG pathway. Furthermore, we found that cells with high expression of PDE6D were more resistant to metformin. When combined with the PDE6D inhibitor TMX-4100, the inhibitory effect on tumors was enhanced, and TMX-4100 demonstrated favorable biosafety in animal models. In conclusion, we found that low-dose metformin inhibits the progression of CRPC by regulating PDE6D-induced alterations in purine metabolism and activating the cGMP/PKG pathway. Moreover, patients with high PDE6D expression may exhibit greater resistance to metformin. Combining metformin with TMX-4100 could further improve the inhibitory effects on tumors.
Collapse
Affiliation(s)
- Shanghua Cai
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Yulin Deng
- Department of Urology, The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710, Dongguan, Guangdong, China
| | - Zhihao Zou
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Weicheng Tian
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Zhenfeng Tang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Jinchuang Li
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Zeheng Tan
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Zhenjie Wu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Zhaodong Han
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Biyan Wen
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Yuanfa Feng
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Ren Liu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Xuejin Zhu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Yongding Wu
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Haiyin Xiao
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Huichan He
- Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China.
| | - Jianheng Ye
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, 999078, Macao Special Administrative Region of China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China.
| | - Weide Zhong
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, 999078, Macao Special Administrative Region of China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Leigh J, Lee SF, Fawaz A, Jia J, Theriau CF, Rodrigues J, Brown J, Ng TL. Assessment of the benefits of bone modifying agents in the management of advanced breast, prostate, and lung cancers. Curr Opin Support Palliat Care 2025; 19:117-129. [PMID: 39946089 DOI: 10.1097/spc.0000000000000749] [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] [Indexed: 05/09/2025]
Abstract
PURPOSE OF REVIEW Skeletal metastases occur in approximately 80% of advanced breast, 70% of advanced prostate, and 30% of lung cancers, and place patients at increased risk of skeletal related events (SRE). Bone modifying agents (BMAs) have been shown to prevent or delay SRE development. Our objective was to summarize the role of these agents in the management of these three cancers. RECENT FINDINGS Total 52 studies met our inclusion criteria. These highlighted the benefit of BMAs in reducing SREs in metastatic breast and castrate resistant prostate cancer (mCRPC), with less clear impact on reducing SRE in lung cancer, or on improving progression-free and overall survival due to significant heterogeneity in trial design and outcomes. Benefits in SRE reduction occurred with bisphosphonates and denosumab, however when compared, denosumab was superior. Denosumab however is not more cost effective, and multiple trials support potential de-escalation to either 12 weekly dosing or other reduced duration. SUMMARY There is a large body of evidence to support the role of BMAs in reducing SREs in metastatic breast and mCRPC. Impact on survival outcomes is heterogeneous, and future large database trials would be helpful in identifying which subgroups of patients truly have survival benefit from BMAs.
Collapse
Affiliation(s)
- Jennifer Leigh
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Shing Fung Lee
- Department of Radiation Oncology, National University Cancer Institute, National University Hospital, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ali Fawaz
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Jason Jia
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Christopher F Theriau
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Jessica Rodrigues
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Janet Brown
- Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Terry L Ng
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
Seaman K, Lin C, Song X, Sassi A, Du WW, Yang B, Sun Y, You L. Mechanical Loading of Osteocytes via Oscillatory Fluid Flow Regulates Early-Stage PC-3 Prostate Cancer Metastasis to Bone. Adv Biol (Weinh) 2025; 9:e2400824. [PMID: 39969425 PMCID: PMC12000999 DOI: 10.1002/adbi.202400824] [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: 12/19/2024] [Revised: 02/05/2025] [Indexed: 02/20/2025]
Abstract
Bone metastasis is a devastating complication for advanced-stage prostate cancer patients. Osteocytes, as the primary mechanosensors in bone, have been recently investigated for their role in prostate cancer bone metastasis. In vivo findings show potential benefits of exercise as a preventative intervention strategy for bone metastasis. In contrast, in vitro studies indicate direct prostate cancer-osteocyte interactions under mechanical loading promote prostate cancer growth and migration. These findings are not consistent with in vivo results and may be more reflective of late-stage metastatic colonization. Here, the role of flow-stimulated osteocytes during early-stage bone metastasis, particularly prostate cancer-endothelial interactions, is examined. Flow-stimulated osteocytes reduce PC-3 prostate cancer cell adhesion and trans-endothelial migration by 32.3% and 40% compared to static controls. Both MLO-Y4 and primary murine osteocytes under mechanical loading regulate the extravasation distance and frequency of PC-3 cells in a microfluidic tissue model. Application of vascular cellular adhesion molecule 1 (VCAM-1) neutralizing antibody abolishes the difference in cancer cell adhesion, extravasation frequency, and number of extravasated PC-3 cells between static and flow-stimulated groups. Taken together, the role of osteocytes in early-stage bone metastasis using PC-3 cells as a model is demonstrated here, bridging the gap between in vitro and in vivo findings.
Collapse
Affiliation(s)
- Kimberly Seaman
- Department of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioM5S 3G8Canada
| | - Chun‐Yu Lin
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5S 3G9Canada
| | - Xin Song
- Department of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioM5S 3G8Canada
| | - Amel Sassi
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5S 3G9Canada
| | - William W. Du
- Sunnybrook Research Institute and Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioM4N 3M5Canada
| | - Burton Yang
- Sunnybrook Research Institute and Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioM4N 3M5Canada
| | - Yu Sun
- Department of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioM5S 3G8Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5S 3G9Canada
| | - Lidan You
- Department of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioM5S 3G8Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5S 3G9Canada
- Department of Mechanical and Materials EngineeringQueen's UniversityKingstonOntarioK7L 3N6Canada
| |
Collapse
|
4
|
Askari E, Shakeri S, Roustaei H, Fotouhi M, Sadeghi R, Harsini S, Vali R. Superscan Pattern on Bone Scintigraphy: A Comprehensive Review. Diagnostics (Basel) 2024; 14:2229. [PMID: 39410633 PMCID: PMC11475626 DOI: 10.3390/diagnostics14192229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES The superscan pattern is a characteristic finding on bone scintigraphy, associated with a variety of metabolic bone diseases, malignancies, and other conditions. This pattern is characterized by a diffuse and intense uptake of radiotracer throughout the entire skeleton. Despite being a relatively rare finding, the superscan pattern can have significant clinical implications. METHODS This comprehensive review summarizes the available literature on the superscan pattern, focusing on its pathophysiology, clinical significance, and differential diagnoses. Relevant studies and case reports were analyzed to outline the diagnostic challenges associated with the interpretation of bone scintigraphy featuring the superscan pattern. RESULTS The literature highlights the clinical significance of the superscan pattern in various metabolic and oncologic conditions. Misinterpretation of this pattern can lead to diagnostic challenges, especially in distinguishing it from other pathologic conditions. Differential diagnosis remains crucial in the accurate interpretation and subsequent management of patients with this finding. CONCLUSIONS This review provides a comprehensive overview of the superscan pattern on bone scintigraphy, aiming to assist clinicians in recognizing and managing this rare yet clinically important finding.
Collapse
Affiliation(s)
- Emran Askari
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (E.A.); (S.S.); (H.R.); (R.S.)
| | - Sara Shakeri
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (E.A.); (S.S.); (H.R.); (R.S.)
| | - Hessamoddin Roustaei
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (E.A.); (S.S.); (H.R.); (R.S.)
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Maryam Fotouhi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran 1419733141, Iran;
| | - Ramin Sadeghi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (E.A.); (S.S.); (H.R.); (R.S.)
| | - Sara Harsini
- BC Cancer Research Institute, Vancouver, BC V5Z 1LL3, Canada;
| | - Reza Vali
- Department of Nuclear Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
5
|
Belue MJ, Harmon SA, Yang D, An JY, Gaur S, Law YM, Turkbey E, Xu Z, Tetreault J, Lay NS, Yilmaz EC, Phelps TE, Simon B, Lindenberg L, Mena E, Pinto PA, Bagci U, Wood BJ, Citrin DE, Dahut WL, Madan RA, Gulley JL, Xu D, Choyke PL, Turkbey B. Deep Learning-Based Detection and Classification of Bone Lesions on Staging Computed Tomography in Prostate Cancer: A Development Study. Acad Radiol 2024; 31:2424-2433. [PMID: 38262813 PMCID: PMC11214604 DOI: 10.1016/j.acra.2024.01.009] [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: 11/06/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
RATIONALE AND OBJECTIVES Efficiently detecting and characterizing metastatic bone lesions on staging CT is crucial for prostate cancer (PCa) care. However, it demands significant expert time and additional imaging such as PET/CT. We aimed to develop an ensemble of two automated deep learning AI models for 1) bone lesion detection and segmentation and 2) benign vs. metastatic lesion classification on staging CTs and to compare its performance with radiologists. MATERIALS AND METHODS This retrospective study developed two AI models using 297 staging CT scans (81 metastatic) with 4601 benign and 1911 metastatic lesions in PCa patients. Metastases were validated by follow-up scans, bone biopsy, or PET/CT. Segmentation AI (3DAISeg) was developed using the lesion contours delineated by a radiologist. 3DAISeg performance was evaluated with the Dice similarity coefficient, and classification AI (3DAIClass) performance on AI and radiologist contours was assessed with F1-score and accuracy. Training/validation/testing data partitions of 70:15:15 were used. A multi-reader study was performed with two junior and two senior radiologists within a subset of the testing dataset (n = 36). RESULTS In 45 unseen staging CT scans (12 metastatic PCa) with 669 benign and 364 metastatic lesions, 3DAISeg detected 73.1% of metastatic (266/364) and 72.4% of benign lesions (484/669). Each scan averaged 12 extra segmentations (range: 1-31). All metastatic scans had at least one detected metastatic lesion, achieving a 100% patient-level detection. The mean Dice score for 3DAISeg was 0.53 (median: 0.59, range: 0-0.87). The F1 for 3DAIClass was 94.8% (radiologist contours) and 92.4% (3DAISeg contours), with a median false positive of 0 (range: 0-3). Using radiologist contours, 3DAIClass had PPV and NPV rates comparable to junior and senior radiologists: PPV (semi-automated approach AI 40.0% vs. Juniors 32.0% vs. Seniors 50.0%) and NPV (AI 96.2% vs. Juniors 95.7% vs. Seniors 91.9%). When using 3DAISeg, 3DAIClass mimicked junior radiologists in PPV (pure-AI 20.0% vs. Juniors 32.0% vs. Seniors 50.0%) but surpassed seniors in NPV (pure-AI 93.8% vs. Juniors 95.7% vs. Seniors 91.9%). CONCLUSION Our lesion detection and classification AI model performs on par with junior and senior radiologists in discerning benign and metastatic lesions on staging CTs obtained for PCa.
Collapse
Affiliation(s)
- Mason J Belue
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Stephanie A Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Dong Yang
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Julie Y An
- Department of Radiology, University of California, San Diego, California, USA (J.Y.A.)
| | - Sonia Gaur
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA (S.G.)
| | - Yan Mee Law
- Department of Radiology, Singapore General Hospital, Singapore (Y.M.L.)
| | - Evrim Turkbey
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA (E.T., B.J.W.)
| | - Ziyue Xu
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Jesse Tetreault
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Nathan S Lay
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Enis C Yilmaz
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Tim E Phelps
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Benjamin Simon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Liza Lindenberg
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Esther Mena
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (P.A.P.)
| | - Ulas Bagci
- Radiology and Biomedical Engineering Department, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA (U.B.)
| | - Bradford J Wood
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA (E.T., B.J.W.); Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (B.J.W.)
| | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (D.E.C.)
| | - William L Dahut
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (W.L.D., R.A.M.)
| | - Ravi A Madan
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (W.L.D., R.A.M.)
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (J.L.G.)
| | - Daguang Xu
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Peter L Choyke
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.).
| |
Collapse
|
6
|
Paindelli C, Parietti V, Barrios S, Shepherd P, Pan T, Wang WL, Satcher RL, Logothetis CJ, Navone N, Campbell MT, Mikos AG, Dondossola E. Bone mimetic environments support engineering, propagation, and analysis of therapeutic response of patient-derived cells, ex vivo and in vivo. Acta Biomater 2024; 178:83-92. [PMID: 38387748 PMCID: PMC12016311 DOI: 10.1016/j.actbio.2024.02.025] [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: 11/21/2023] [Revised: 01/22/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Bone metastases are the most common milestone in the lethal progression of prostate cancer and prominent in a substantial portion of renal malignancies. Interactions between cancer and bone host cells have emerged as drivers of both disease progression and therapeutic resistance. To best understand these central host-epithelial cell interactions, biologically relevant preclinical models are required. To achieve this goal, we here established and characterized tissue-engineered bone mimetic environments (BME) capable of supporting the growth of patient-derived xenograft (PDX) cells, ex vivo and in vivo. The BME consisted of a polycaprolactone (PCL) scaffold colonized by human mesenchymal stem cells (hMSCs) differentiated into osteoblasts. PDX-derived cells were isolated from bone metastatic prostate or renal tumors, engineered to express GFP or luciferase and seeded onto the BMEs. BMEs supported the growth and therapy response of PDX-derived cells, ex vivo. Additionally, BMEs survived after in vivo implantation and further sustained the growth of PDX-derived cells, their serial transplant, and their application to study the response to treatment. Taken together, this demonstrates the utility of BMEs in combination with patient-derived cells, both ex vivo and in vivo. STATEMENT OF SIGNIFICANCE: Our tissue-engineered BME supported the growth of patient-derived cells and proved useful to monitor the therapy response, both ex vivo and in vivo. This approach has the potential to enable co-clinical strategies to monitor bone metastatic tumor progression and therapy response, including identification and prioritization of new targets for patient treatment.
Collapse
Affiliation(s)
- Claudia Paindelli
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Vanessa Parietti
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Sergio Barrios
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States; Rice University, Department of Bioengineering, Houston, TX, 77030, United States
| | - Peter Shepherd
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Tianhong Pan
- Department of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Robert L Satcher
- Department of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Nora Navone
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Antonios G Mikos
- Rice University, Department of Bioengineering, Houston, TX, 77030, United States
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States.
| |
Collapse
|
7
|
Bi W, Guo W, Fan G, Xie L, Jiang C. Identification and validation of a novel overall survival prediction model for immune-related genes in bone metastases of prostate cancer. Aging (Albany NY) 2023; 15:7161-7186. [PMID: 37494663 PMCID: PMC10415549 DOI: 10.18632/aging.204900] [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: 04/14/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023]
Abstract
Immunotherapy has become a revolutionary treatment for cancer and brought new vitality to tumor immunity. Bone metastases are the most prevalent metastatic site for advanced prostate cancer (PCa). Therefore, finding new immunotherapy targets in PCa patients with bone metastasis is urgently needed. We conducted an elaborative bioinformatics study of immune-related genes (IRGs) and tumor-infiltrating immune cells (TIICs) in PCa bone metastases. Databases were integrated to obtain RNA-sequencing data and clinical prognostic information. Univariate and multivariate Cox regression analyses were conducted to construct an overall survival (OS) prediction model. GSE32269 was analyzed to acquire differentially expressed IRGs. The OS prediction model was established by employing six IRGs (MAVS, HSP90AA1, FCGR3A, CTSB, FCER1G, and CD4). The CIBERSORT algorithm was adopted to assess the proportion of TIICs in each group. Furthermore, Transwell, MTT, and wound healing assays were employed to determine the effect of MAVS on PCa cells. High-risk patients had worse OS compared to the low-risk patients in the training and validation cohorts. Meanwhile, clinically practical nomograms were generated using these identified IRGs to predict the 3- and 5-year survival rates of patients. The infiltration percentages of some TIICs were closely linked to the risk score of the OS prediction model. Some tumor-infiltrating immune cells were related to the OS. FCGR3A was closely correlated with some TIICs. In vitro experiments verified that up-regulation of MAVS suppressed the proliferation and metastatic abilities of PCa cells. Our work presented a thorough interpretation of TIICs and IRGs for illustrating and discovering new potential immune checkpoints in bone metastases of PCa.
Collapse
Affiliation(s)
- Wen Bi
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Weiming Guo
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Gang Fan
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Lei Xie
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Changqing Jiang
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
8
|
He M, Cao Y, Chi C, Yang X, Ramin R, Wang S, Yang G, Mukhtorov O, Zhang L, Kazantsev A, Enikeev M, Hu K. Research progress on deep learning in magnetic resonance imaging-based diagnosis and treatment of prostate cancer: a review on the current status and perspectives. Front Oncol 2023; 13:1189370. [PMID: 37546423 PMCID: PMC10400334 DOI: 10.3389/fonc.2023.1189370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/30/2023] [Indexed: 08/08/2023] Open
Abstract
Multiparametric magnetic resonance imaging (mpMRI) has emerged as a first-line screening and diagnostic tool for prostate cancer, aiding in treatment selection and noninvasive radiotherapy guidance. However, the manual interpretation of MRI data is challenging and time-consuming, which may impact sensitivity and specificity. With recent technological advances, artificial intelligence (AI) in the form of computer-aided diagnosis (CAD) based on MRI data has been applied to prostate cancer diagnosis and treatment. Among AI techniques, deep learning involving convolutional neural networks contributes to detection, segmentation, scoring, grading, and prognostic evaluation of prostate cancer. CAD systems have automatic operation, rapid processing, and accuracy, incorporating multiple sequences of multiparametric MRI data of the prostate gland into the deep learning model. Thus, they have become a research direction of great interest, especially in smart healthcare. This review highlights the current progress of deep learning technology in MRI-based diagnosis and treatment of prostate cancer. The key elements of deep learning-based MRI image processing in CAD systems and radiotherapy of prostate cancer are briefly described, making it understandable not only for radiologists but also for general physicians without specialized imaging interpretation training. Deep learning technology enables lesion identification, detection, and segmentation, grading and scoring of prostate cancer, and prediction of postoperative recurrence and prognostic outcomes. The diagnostic accuracy of deep learning can be improved by optimizing models and algorithms, expanding medical database resources, and combining multi-omics data and comprehensive analysis of various morphological data. Deep learning has the potential to become the key diagnostic method in prostate cancer diagnosis and treatment in the future.
Collapse
Affiliation(s)
- Mingze He
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Changliang Chi
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), Changchun, Jilin, China
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Rzayev Ramin
- Department of Radiology, The Second University Clinic, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Shuowen Wang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Guodong Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Otabek Mukhtorov
- Regional State Budgetary Health Care Institution, Kostroma Regional Clinical Hospital named after Korolev E.I. Avenue Mira, Kostroma, Russia
| | - Liqun Zhang
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, China
| | - Anton Kazantsev
- Regional State Budgetary Health Care Institution, Kostroma Regional Clinical Hospital named after Korolev E.I. Avenue Mira, Kostroma, Russia
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Kebang Hu
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), Changchun, Jilin, China
| |
Collapse
|
9
|
Somanath PR, Chernoff J, Cummings BS, Prasad SM, Homan HD. Targeting P21-Activated Kinase-1 for Metastatic Prostate Cancer. Cancers (Basel) 2023; 15:2236. [PMID: 37190165 PMCID: PMC10137274 DOI: 10.3390/cancers15082236] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic prostate cancer (mPCa) has limited therapeutic options and a high mortality rate. The p21-activated kinase (PAK) family of proteins is important in cell survival, proliferation, and motility in physiology, and pathologies such as infectious, inflammatory, vascular, and neurological diseases as well as cancers. Group-I PAKs (PAK1, PAK2, and PAK3) are involved in the regulation of actin dynamics and thus are integral for cell morphology, adhesion to the extracellular matrix, and cell motility. They also play prominent roles in cell survival and proliferation. These properties make group-I PAKs a potentially important target for cancer therapy. In contrast to normal prostate and prostatic epithelial cells, group-I PAKs are highly expressed in mPCA and PCa tissue. Importantly, the expression of group-I PAKs is proportional to the Gleason score of the patients. While several compounds have been identified that target group-I PAKs and these are active in cells and mice, and while some inhibitors have entered human trials, as of yet, none have been FDA-approved. Probable reasons for this lack of translation include issues related to selectivity, specificity, stability, and efficacy resulting in side effects and/or lack of efficacy. In the current review, we describe the pathophysiology and current treatment guidelines of PCa, present group-I PAKs as a potential druggable target to treat mPCa patients, and discuss the various ATP-competitive and allosteric inhibitors of PAKs. We also discuss the development and testing of a nanotechnology-based therapeutic formulation of group-I PAK inhibitors and its significant potential advantages as a novel, selective, stable, and efficacious mPCa therapeutic over other PCa therapeutics in the pipeline.
Collapse
Affiliation(s)
- Payaningal R. Somanath
- Department of Clinical & Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- MetasTx LLC, Basking Ridge, NJ 07920, USA
| | - Jonathan Chernoff
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Brian S. Cummings
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Sandip M. Prasad
- Morristown Medical Center, Atlantic Health System, Morristown, NJ 07960, USA
| | | |
Collapse
|
10
|
Pan J, Ma Z, Liu B, Qian H, Shao X, Liu J, Wang Q, Xue W. Identification of cancer-associated fibroblasts subtypes in prostate cancer. Front Immunol 2023; 14:1133160. [PMID: 37033924 PMCID: PMC10080037 DOI: 10.3389/fimmu.2023.1133160] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Cancer-associated fibroblasts (CAFs) are one of the most abundant cell types in tumor microenvironment. However, the phenotypic and functional heterogeneities among CAFs have not been sufficiently investigated in prostate cancer. Methods We obtained and analyzed the single-cell RNA-sequencing data from 26 hormone-sensitive prostate cancer samples and 8 castration-resistant prostate cancer samples, along with the analysis of bulk-sequencing datasets. Furthermore, we performed multicolor immunofluorescence staining to verify the findings from the data analysis. Results We identified two major CAFs subtypes with distinct molecular characteristics and biological functions in prostate cancer microenvironment, namely αSMA+ CAV1+ CAFs-C0 and FN1+ FAP+ CAFs-C1. Another single-cell RNA-sequencing dataset containing 7 bone metastatic prostate cancer samples demonstrated that osteoblasts in the bone metastatic lesions comprised two subtypes with molecular characteristics and biological functions similar to CAFs-C0 and CAFs-C1 in the primary tumor sites. In addition, we discovered a transcriptional factor regulatory network depending on CAFs-C1. CAFs-C1, but not CAFs-C0, was associated with castration resistance and poor prognosis. We also found that CAFs-C1 signature was involved in treatment resistance to immune checkpoint inhibitors. Discussion In summary, our results identified the presence of heterogeneous CAFs subtypes in prostate cancer microenvironment and the potential of specific CAFs subtype as therapeutic target for castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jiahua Pan
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zehua Ma
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyang Qian
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoguang Shao
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiazhou Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Qi Wang, ; Wei Xue,
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Qi Wang, ; Wei Xue,
| |
Collapse
|
11
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
12
|
Osteoblast Secretome Modulated by Abiraterone Treatment Affects Castration Resistant Prostate Cancer Cell Proliferation. Biomedicines 2022; 10:biomedicines10092154. [PMID: 36140255 PMCID: PMC9496251 DOI: 10.3390/biomedicines10092154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/02/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Abiraterone is a selective inhibitor of androgen biosynthesis approved for the treatment of metastatic patients affected by castration-resistant or castration-sensitive prostate cancer. Intriguingly, clinical data revealed that abiraterone also delayed disease progression in bone improving bone-related endpoints. Our group has previously demonstrated in vitro a direct effect of abiraterone on osteoclast and osteoblast function suggesting its ability to modulate bone microenvironment. Here, we performed an extensive proteomic analysis to investigate how abiraterone influences osteoblast cell secretome and, consequently, osteoblast/prostate cancer cells interaction. A panel of 507 soluble molecules were analyzed in osteoblast conditioned media (OCM) obtained from osteoblast treated or not with abiraterone. Subsequently, OCM was added to prostate cancer cells to investigate its potential effect on prostate cancer cell proliferation and androgen receptor (AR) activation status. Out of 507 screened molecules, 39 of them were differentially expressed in OCM from osteoblasts treated with abiraterone (OCM ABI) compared to OCM obtained from untreated OBs (OCM CTRL). Pathway enrichment analysis revealed that abiraterone down-modulated the release of specific osteoblast soluble factors, positively associated with cell proliferation pathways (false discovery rate adjusted p-value = 0.0019). In vitro validation data showed that OCM ABI treatment significantly reduced cancer proliferation in C4-2B cells (p = 0.022), but not in AR- negative PC-3 cells. Moreover, we also found a reduction in AR activation in C4-2B cells (p = 0.017) confirming the “indirect” anti-tumor AR-dependent effect of abiraterone mediated by osteoblasts. This study provides the first evidence of an additional antitumor effect of abiraterone through the modulation of multiple osteoblast proliferative signals.
Collapse
|
13
|
Foster BM, Shi L, Harris KS, Patel C, Surratt VE, Langsten KL, Kerr BA. Bone Marrow-Derived Stem Cell Factor Regulates Prostate Cancer-Induced Shifts in Pre-Metastatic Niche Composition. Front Oncol 2022; 12:855188. [PMID: 35515124 PMCID: PMC9063312 DOI: 10.3389/fonc.2022.855188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Skeletal metastasis is the leading cause of morbidity and mortality in prostate cancer, with 80% of advanced prostate cancer patients developing bone metastases. Before metastasis, bone remodeling occurs, stimulating pre-metastatic niche formation and bone turnover, and platelets govern this process. Stem cell factor (SCF, Kit Ligand) is increased in advanced prostate cancer patient platelet releasates. Further, SCF and its receptor, CD117/c-kit, correlate with metastatic prostate cancer severity. We hypothesized that bone-derived SCF plays an important role in prostate cancer tumor communication with the bone inducing pre-metastatic niche formation. We generated two cell-specific SCF knockout mouse models deleting SCF in either mature osteoblasts or megakaryocytes and platelets. Using two syngeneic androgen-insensitive murine prostate cancer cell lines, RM1 (Ras and Myc co-activation) and mPC3 (Pten and Trp53 deletion), we examined the role of bone marrow-derived SCF in primary tumor growth and bone microenvironment alterations. Platelet-derived SCF was required for mPC3, but not RM1, tumor growth, while osteoblast-derived SCF played no role in tumor size in either cell line. While exogenous SCF induced proangiogenic protein secretion by RM1 and mPC3 prostate cancer cells, no significant changes in tumor angiogenesis were measured by immunohistochemistry. Like our previous studies, tumor-induced bone formation occurred in mice bearing RM1 or mPC3 neoplasms, demonstrated by bone histomorphometry. RM1 tumor-bearing osteoblast SCF knockout mice did not display tumor-induced bone formation. Bone stromal cell composition analysis by flow cytometry showed significant shifts in hematopoietic stem cell (HSC), mesenchymal stem cell (MSC), and osteoblast cell percentages in mice bearing RM1 or mPC3 tumors. There were no significant changes in the percentage of macrophages, osteoclasts, or osteocytes. Our study demonstrates that megakaryocyte/platelet-derived SCF regulates primary mPC3 tumor growth, while SCF originating from osteoblasts plays a role in bone marrow-derived progenitor cell composition and pre-metastatic niche formation. Further, we show that both the source of SCF and the genetic profile of prostate cancer determine the effects of SCF. Thus, targeting the SCF/CD117 signaling axis with tyrosine kinase inhibitors could affect primary prostate carcinomas or play a role in reducing bone metastasis dependent on the gene deletions or mutations driving the patients' prostate cancer.
Collapse
Affiliation(s)
- Brittni M. Foster
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Koran S. Harris
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Chirayu Patel
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Victoria E. Surratt
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Kendall L. Langsten
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Bethany A. Kerr
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, United States
| |
Collapse
|
14
|
Ribelli G, Simonetti S, Iuliani M, Rossi E, Vincenzi B, Tonini G, Pantano F, Santini D. Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms. Front Oncol 2021; 11:789885. [PMID: 34966687 PMCID: PMC8711264 DOI: 10.3389/fonc.2021.789885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Patients with metastatic prostate cancer frequently develop bone metastases that elicit significant skeletal morbidity and increased mortality. The high tropism of prostate cancer cells for bone and their tendency to induce the osteoblastic-like phenotype are a result of a complex interplay between tumor cells and osteoblasts. Although the role of osteoblasts in supporting prostate cancer cell proliferation has been reported by previous studies, their precise contribution in tumor growth remains to be fully elucidated. Here, we tried to dissect the molecular signaling underlining the interactions between castration-resistant prostate cancer (CRPC) cells and osteoblasts using in vitro co-culture models. Transcriptomic analysis showed that osteoblast-conditioned media (OCM) induced the overexpression of genes related to cell cycle in the CRPC cell line C4-2B but, surprisingly, reduced androgen receptor (AR) transcript levels. In-depth analysis of AR expression in C4-2B cells after OCM treatment showed an AR reduction at the mRNA (p = 0.0047), protein (p = 0.0247), and functional level (p = 0.0029) and, concomitantly, an increase of C4-2B cells in S-G2-M cell cycle phases (p = 0.0185). An extensive proteomic analysis revealed in OCM the presence of some molecules that reduced AR activation, and among these, Matrix metalloproteinase-1 (MMP-1) was the only one able to block AR function (0.1 ng/ml p = 0.006; 1 ng/ml p = 0.002; 10 ng/ml p = 0.0001) and, at the same time, enhance CRPC proliferation (1 ng/ml p = 0.009; 10 ng/ml p = 0.033). Although the increase of C4-2B cell growth induced by MMP-1 did not reach the proliferation levels observed after OCM treatment, the addition of Vorapaxar, an MMP-1 receptor inhibitor (Protease-activated receptor-1, PAR-1), significantly reduced C4-2B cell cycle (0.1 μM p = 0.014; 1 μM p = 0.0087). Overall, our results provide a novel AR-independent mechanism of CRPC proliferation and suggest that MMP-1/PAR-1 could be one of the potential pathways involved in this process.
Collapse
Affiliation(s)
- Giulia Ribelli
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Sonia Simonetti
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Michele Iuliani
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Elisabetta Rossi
- Department of Immunology and Molecular Oncology, Istituto Oncologico Veneto (IOV) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Francesco Pantano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| |
Collapse
|
15
|
Liu X, Han C, Cui Y, Xie T, Zhang X, Wang X. Detection and Segmentation of Pelvic Bones Metastases in MRI Images for Patients With Prostate Cancer Based on Deep Learning. Front Oncol 2021; 11:773299. [PMID: 34912716 PMCID: PMC8666439 DOI: 10.3389/fonc.2021.773299] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022] Open
Abstract
Objective To establish and evaluate the 3D U-Net model for automated segmentation and detection of pelvic bone metastases in patients with prostate cancer (PCa) using diffusion-weighted imaging (DWI) and T1 weighted imaging (T1WI) images. Methods The model consisted of two 3D U-Net algorithms. A total of 859 patients with clinically suspected or confirmed PCa between January 2017 and December 2020 were enrolled for the first 3D U-Net development of pelvic bony structure segmentation. Then, 334 PCa patients were selected for the model development of bone metastases segmentation. Additionally, 63 patients from January to May 2021 were recruited for the external evaluation of the network. The network was developed using DWI and T1WI images as input. Dice similarity coefficient (DSC), volumetric similarity (VS), and Hausdorff distance (HD) were used to evaluate the segmentation performance. Sensitivity, specificity, and area under the curve (AUC) were used to evaluate the detection performance at the patient level; recall, precision, and F1-score were assessed at the lesion level. Results The pelvic bony structures segmentation on DWI and T1WI images had mean DSC and VS values above 0.85, and the HD values were <15 mm. In the testing set, the AUC of the metastases detection at the patient level were 0.85 and 0.80 on DWI and T1WI images. At the lesion level, the F1-score achieved 87.6% and 87.8% concerning metastases detection on DWI and T1WI images, respectively. In the external dataset, the AUC of the model for M-staging was 0.94 and 0.89 on DWI and T1WI images. Conclusion The deep learning-based 3D U-Net network yields accurate detection and segmentation of pelvic bone metastases for PCa patients on DWI and T1WI images, which lays a foundation for the whole-body skeletal metastases assessment.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Chao Han
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Yingpu Cui
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Tingting Xie
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaodong Zhang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Xiaoying Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| |
Collapse
|
16
|
Intracellular Signaling Responses Induced by Radiation within an In Vitro Bone Metastasis Model after Pre-Treatment with an Estrone Analogue. Cells 2021; 10:cells10082105. [PMID: 34440874 PMCID: PMC8394480 DOI: 10.3390/cells10082105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022] Open
Abstract
2-Ethyl-3-O-sulfamoyl-estra-1,3,5(10)16-tetraene (ESE-16) is an in silico-designed estradiol analogue which has improved the parent compound’s efficacy in anti-cancer studies. In this proof-of-concept study, the potential radiosensitizing effects of ESE-16 were investigated in an in vitro deconstructed bone metastasis model. Prostate (DU 145) and breast (MDA-MB-231) tumor cells, osteoblastic (MC3T3-E1) and osteoclastic (RAW 264.7) bone cells and human umbilical vein endothelial cells (HUVECs) were representative components of such a lesion. Cells were exposed to a low-dose ESE-16 for 24 hours prior to radiation at non-lethal doses to determine early signaling and molecular responses of this combination treatment. Tartrate-resistant acid phosphatase activity and actin ring formation were investigated in osteoclasts, while cell cycle progression, reactive oxygen species generation and angiogenic protein expression were investigated in HUVECs. Increased cytotoxicity was evident in tumor and endothelial cells while bone cells appeared to be spared. Increased mitotic indices were calculated, and evidence of increased deoxyribonucleic acid damage with retarded repair, together with reduced metastatic signaling was observed in tumor cells. RAW 264.7 macrophages retained their ability to differentiate into osteoclasts. Anti-angiogenic effects were observed in HUVECs, and expression of hypoxia-inducible factor 1-α was decreased. Through preferentially inducing tumor cell death and potentially inhibiting neovascularization whilst preserving bone physiology, this low-dose combination regimen warrants further investigation for its promising therapeutic application in bone metastases management, with the additional potential of limited treatment side effects.
Collapse
|
17
|
Fernández R, Eppard E, Lehnert W, Jiménez-Franco LD, Soza-Ried C, Ceballos M, Ribbeck J, Kluge A, Rösch F, Meckel M, Zhernosekov K, Kramer V, Amaral H. Evaluation of Safety and Dosimetry of 177Lu-DOTA-ZOL for Therapy of Bone Metastases. J Nucl Med 2021; 62:1126-1132. [PMID: 33419945 PMCID: PMC8833870 DOI: 10.2967/jnumed.120.255851] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022] Open
Abstract
Palliative treatment of bone metastasis using radiolabeled bisphosphonates is a well-known concept proven to be safe and effective. A new therapeutic radiopharmaceutical for bone metastasis is 177Lu-DOTA-zoledronic acid (177Lu-DOTA-ZOL). In this study, the safety and dosimetry of a single therapeutic dose of 177Lu-DOTA-ZOL were evaluated on the basis of a series of SPECT/CT images and blood samples. Methods: Nine patients with exclusive bone metastases from metastatic castration-resistant prostate cancer (mCRPC) (70.8 ± 8.4 y) and progression under conventional therapies participated in this prospective study. After receiving 5,780 ± 329 MBq 177Lu-DOTA-ZOL, patients underwent 3-dimensional whole-body SPECT/CT imaging and venous blood sampling over 7 d. Dosimetric evaluation was performed for main organs and tumor lesions. Safety was assessed by blood biomarkers. Results:177Lu-DOTA-ZOL showed fast uptake and high retention in bone lesions and fast clearance from the bloodstream in all patients. The average retention in tumor lesions was 0.02% injected activity per gram at 6 h after injection and approximately 0.01% at 170 h after injection. In this cohort, the average absorbed doses in bone tumor lesions, kidneys, red bone marrow, and bone surfaces were 4.21, 0.17, 0.36, and 1.19 Gy/GBq, respectively. The red marrow was found to be the dose-limiting organ for all patients. A median maximum tolerated injected activity of 6.0 GBq may exceed the defined threshold of 2 Gy for the red bone marrow in individual patients (4/8). Conclusion:177Lu-DOTA-ZOL is safe and has a favorable therapeutic index compared with other radiopharmaceuticals used in the treatment of osteoblastic bone metastases. Personalized dosimetry, however, should be considered to avoid severe hematotoxicity for individual patients.
Collapse
Affiliation(s)
- René Fernández
- Center for Nuclear Medicine and PET/CT, PositronMed, Santiago, Chile
| | | | - Wencke Lehnert
- ABX-CRO, Dresden, Germany
- Department of Nuclear Medicine, University Medical Center, Hamburg, Germany
| | | | | | - Matías Ceballos
- Center for Nuclear Medicine and PET/CT, PositronMed, Santiago, Chile
| | | | | | - Frank Rösch
- Institute of Nuclear Chemistry, Johannes Gutenberg-University, Mainz, Germany; and
| | | | | | - Vasko Kramer
- Center for Nuclear Medicine and PET/CT, PositronMed, Santiago, Chile
- Positronpharma SA, Santiago, Chile
| | - Horacio Amaral
- Center for Nuclear Medicine and PET/CT, PositronMed, Santiago, Chile
- Positronpharma SA, Santiago, Chile
| |
Collapse
|
18
|
Vahidfar N, Eppard E, Farzanehfar S, Yordanova A, Fallahpoor M, Ahmadzadehfar H. An Impressive Approach in Nuclear Medicine: Theranostics. PET Clin 2021; 16:327-340. [PMID: 34053577 DOI: 10.1016/j.cpet.2021.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Radiometal-based theranostics or theragnostics, first used in the early 2000s, is the combined application of diagnostic and therapeutic agents that target the same molecule, and represents a considerable advancement in nuclear medicine. One of the promising fields related to theranostics is radioligand therapy. For instance, the concepts of targeting the prostate-specific membrane antigen (PSMA) for imaging and therapy in prostate cancer, or somatostatin receptor targeted imaging and therapy in neuroendocrine tumors (NETs) are part of the field of theranostics. Combining targeted imaging and therapy can improve prognostication, therapeutic decision-making, and monitoring of the therapy.
Collapse
Affiliation(s)
- Nasim Vahidfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elisabeth Eppard
- Positronpharma SA, Santiago, Chile; Department of Nuclear Medicine, University Hospital Magdeburg, Germany
| | - Saeed Farzanehfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Fallahpoor
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
19
|
Bhadresha KP, Jain NK, Rawal RM. Assessing the Protective Effect of Moringa oleifera Extract against Bone Metastasis: An In Vitro Simulated Digestion Approach. Nutr Cancer 2021; 74:1023-1036. [PMID: 34170200 DOI: 10.1080/01635581.2021.1933099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Moringa oleifera possesses numerous advantageous effects like anti-microbial, antioxidant, and anti-inflammatory, leaves contain a high multiplicity of the bioactive compound; however, little is identified about its bioaccessibility. The objective of this study was to assess the bioefficacy, bioaccessible and anticancer activity of Moringa oleifera in a PC3 cell line before and after simulated in vitro digestion. Digested and non-digested extracts were prepared and evaluated for total polyphenols, flavonoids, and total antioxidant capacity by spectrophotometric analysis and LCMS analysis. Cell viability, apoptosis, colony formation, cell cycle, Glutathione level, and gene expression study were tested with Moringa oleifera (MO) and digested Moringa oleifera (DMO). Results revealed that total polyphenols, total flavonoids, and TAC were significantly (P < 0.05) reduced after in vitro digestion. Furthermore, biological activity against the PC3 cell line showed that DMO extracts significant cytotoxic and reduced cell vitality compared to the MO. In addition, DMO extract had a noteworthy effect in apoptosis and inhibiting the colony formation ability; while cell cycle was blocked in S phase by both extracts but significant effect showed in DMO. These studies have increased understanding of the influence of in vitro simulation digestion on the biological activity effect of M. oleifera against prostate cancer bone metastasis.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1933099 .
Collapse
Affiliation(s)
- Kinjal P Bhadresha
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Nayan K Jain
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Rakesh M Rawal
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| |
Collapse
|
20
|
Mitrofanov YA, Larenkov AA, Kodina GE. Complexation of Scandium with
Oxabis(ethylenenitrilo)tetramethylenephosphonic Acid and Applicability of Its
44Sc-Labelled Analogue as Bone-Seeking
Agent. RUSS J GEN CHEM+ 2021. [DOI: 10.1134/s1070363221020080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
21
|
Zdravkovic A, Hasenoehrl T, Crevenna R. Resistance Exercise in Prostate Cancer Patients: a Short Review. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2021. [DOI: 10.1007/s40141-021-00307-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Abstract
Purpose of Review
The aim of this paper is to provide an overview of recent findings concerning the utilization of resistance exercise (RE) in prostate cancer (PCa), in particular as pertaining to the management of cancer therapy side effects.
Recent Findings
As of late, studies investigating the effects of RE in PCa patients have found positive effects on muscle strength, body composition, physical functioning, quality of life, and fatigue. The combination of RE and impact training appears to decrease the loss of bone mineral density. RE seems to be well accepted and tolerated, even by patients with bone metastatic disease, although a modification of the RE prescription is often necessary.
Summary
In PCa patients, RE has been well-researched and the data are clear that it is beneficial in multiple ways. Future directions should look at the long-term effects of RE, including mortality and relapse, as well as implementation of exercise programs.
Collapse
|
22
|
Isaac A, Lecouvet F, Dalili D, Fayad L, Pasoglou V, Papakonstantinou O, Ahlawat S, Messiou C, Weber MA, Padhani AR. Detection and Characterization of Musculoskeletal Cancer Using Whole-Body Magnetic Resonance Imaging. Semin Musculoskelet Radiol 2020; 24:726-750. [PMID: 33307587 DOI: 10.1055/s-0040-1719018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Whole-body magnetic resonance imaging (WB-MRI) is gradually being integrated into clinical pathways for the detection, characterization, and staging of malignant tumors including those arising in the musculoskeletal (MSK) system. Although further developments and research are needed, it is now recognized that WB-MRI enables reliable, sensitive, and specific detection and quantification of disease burden, with clinical applications for a variety of disease types and a particular application for skeletal involvement. Advances in imaging techniques now allow the reliable incorporation of WB-MRI into clinical pathways, and guidelines recommending its use are emerging. This review assesses the benefits, clinical applications, limitations, and future capabilities of WB-MRI in the context of other next-generation imaging modalities, as a qualitative and quantitative tool for the detection and characterization of skeletal and soft tissue MSK malignancies.
Collapse
Affiliation(s)
- Amanda Isaac
- School of Biomedical Engineering & Imaging Sciences, Kings College London, United Kingdom.,Guy's & St Thomas' Hospitals, London, United Kingdom
| | - Frederic Lecouvet
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Danoob Dalili
- School of Biomedical Engineering & Imaging Sciences, Kings College London, United Kingdom.,Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Laura Fayad
- The Russell H. Morgan Department of Radiology and Radiological Science, John's Hopkins School of Medicine, Baltimore, Maryland
| | - Vasiliki Pasoglou
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Olympia Papakonstantinou
- 2nd Department of Radiology, National and Kapodistrian University of Athens, "Attikon" Hospital, Athens, Greece
| | - Shivani Ahlawat
- The Russell H. Morgan Department of Radiology and Radiological Science, John's Hopkins School of Medicine, Baltimore, Maryland
| | - Christina Messiou
- The Royal Marsden Hospital, London, United Kingdom.,The Institute of Cancer Research, London, United Kingdom
| | - Marc-André Weber
- Institute of Diagnostic and Interventional Radiology, Paediatric Radiology and Neuroradiology, University Medical Centre Rostock, Rostock, Germany
| | - Anwar R Padhani
- The Institute of Cancer Research, London, United Kingdom.,Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, United Kingdom
| |
Collapse
|
23
|
Downregulation of miRNA-205 Expression and Biological Mechanism in Prostate Cancer Tumorigenesis and Bone Metastasis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6037434. [PMID: 33178832 PMCID: PMC7646560 DOI: 10.1155/2020/6037434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/22/2020] [Accepted: 10/15/2020] [Indexed: 12/23/2022]
Abstract
Background The expression and mechanism of microRNA-205 (miRNA-205) in prostate cancer (PCa) and its bone metastasis remain controversial. Materials and Methods The expression and discriminating capability of miRNA-205 were assessed by drawing a forest plot and a summarized receiver operating characteristic (SROC) curve, using data available from 27 miRNA-array and miRNA-sequencing datasets. The miRNA-205 target genes were acquired from online prediction tools, differentially upregulated genes in PCa, and differentially expressed genes (DEGs) after miRNA-205 transfection into PCa cell lines. Functional enrichment analysis was conducted to explore the biological mechanism of miRNA-205 targets. Immunohistochemistry (IHC) was applied to verify the protein level of the hub gene. Results The expression of miRNA-205 in the PCa group (1,461 samples) was significantly lower than that in the noncancer group (510 samples), and the downregulation of miRNA-205 showed excellent sensitivity and specificity in differentiating between the two groups. In bone metastatic PCa, the miRNA-205 level was further reduced than in nonbone metastatic PCa, and it showed a good capability in distinguishing between the two groups. In total, 153 miRNA-205 targets were screened through the three aforementioned methods. Based on the results of functional enrichment analysis, the targets of miRNA-205 were mainly enriched during chromosome segregation and phospholipid-translocating ATPase activity and in the spindle microtubule and the p53 signaling pathway. CDK1 had the highest connectivity in the PPI network analysis and was screened as one of the hub genes. A statistically significant negative correlation between miRNA-205 and CDK1 was observed. The expression of CDK1 in PCa samples was pronouncedly upregulated in terms of both the mRNA level and the protein level when compared with noncancer samples. Conclusion miRNA-205 may play a vital role in PCa tumorigenesis and bone metastasis by targeting CDK1.
Collapse
|
24
|
Franko A, Berti L, Guirguis A, Hennenlotter J, Wagner R, Scharpf MO, de Angelis MH, Wißmiller K, Lickert H, Stenzl A, Birkenfeld AL, Peter A, Häring HU, Lutz SZ, Heni M. Characterization of Hormone-Dependent Pathways in Six Human Prostate-Cancer Cell Lines: A Gene-Expression Study. Genes (Basel) 2020; 11:E1174. [PMID: 33036464 PMCID: PMC7599530 DOI: 10.3390/genes11101174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa), the most incident cancer in men, is tightly regulated by endocrine signals. A number of different PCa cell lines are commonly used for in vitro experiments, but these are of diverse origin, and have very different cell-proliferation rates and hormone-response capacities. By analyzing the gene-expression pattern of main hormone pathways, we systematically compared six PCa cell lines and parental primary cells. We compared these cell lines (i) with each other and (ii) with PCa tissue samples from 11 patients. We found major differences in the gene-expression levels of androgen, insulin, estrogen, and oxysterol signaling between PCa tissue and cell lines, and between different cell lines. Our systematic characterization gives researchers a solid basis to choose the appropriate PCa cell model for the hormone pathway of interest.
Collapse
Affiliation(s)
- Andras Franko
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Lucia Berti
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Alke Guirguis
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (A.S.)
| | - Robert Wagner
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Marcus O. Scharpf
- Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Martin Hrabĕ de Angelis
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Katharina Wißmiller
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany;
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Medicine, Technical University of Munich, 81675 München, Germany
| | - Heiko Lickert
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany;
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Medicine, Technical University of Munich, 81675 München, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (A.S.)
| | - Andreas L. Birkenfeld
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Stefan Z. Lutz
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, 72116 Mössingen, Germany
| | - Martin Heni
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
25
|
Laranga R, Duchi S, Ibrahim T, Guerrieri AN, Donati DM, Lucarelli E. Trends in Bone Metastasis Modeling. Cancers (Basel) 2020; 12:E2315. [PMID: 32824479 PMCID: PMC7464021 DOI: 10.3390/cancers12082315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Bone is one of the most common sites for cancer metastasis. Bone tissue is composed by different kinds of cells that coexist in a coordinated balance. Due to the complexity of bone, it is impossible to capture the intricate interactions between cells under either physiological or pathological conditions. Hence, a variety of in vivo and in vitro approaches have been developed. Various models of tumor-bone diseases are routinely used to provide valuable information on the relationship between metastatic cancer cells and the bone tissue. Ideally, when modeling the metastasis of human cancers to bone, models would replicate the intra-tumor heterogeneity, as well as the genetic and phenotypic changes that occur with human cancers; such models would be scalable and reproducible to allow high-throughput investigation. Despite the continuous progress, there is still a lack of solid, amenable, and affordable models that are able to fully recapitulate the biological processes happening in vivo, permitting a correct interpretation of results. In the last decades, researchers have demonstrated that three-dimensional (3D) methods could be an innovative approach that lies between bi-dimensional (2D) models and animal models. Scientific evidence supports that the tumor microenvironment can be better reproduced in a 3D system than a 2D cell culture, and the 3D systems can be scaled up for drug screening in the same way as the 2D systems thanks to the current technologies developed. However, 3D models cannot completely recapitulate the inter- and intra-tumor heterogeneity found in patients. In contrast, ex vivo cultures of fragments of bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Moreover, ex vivo bone organ cultures could be a better model to resemble the human pathogenic metastasis condition and useful tools to predict in vivo response to therapies. The aim of our review is to provide an overview of the current trends in bone metastasis modeling. By showing the existing in vitro and ex vivo systems, we aspire to contribute to broaden the knowledge on bone metastasis models and make these tools more appealing for further translational studies.
Collapse
Affiliation(s)
- Roberta Laranga
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Serena Duchi
- BioFab3D@ACMD, St Vincent’s Hospital, Melbourne, VIC 3065, Australia;
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Ania Naila Guerrieri
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Davide Maria Donati
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
- Rizzoli Laboratory Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Via di Barbiano 1/10, 40136 Bologna, Italy
- 3rd Orthopaedic and Traumatologic Clinic Prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy
| | - Enrico Lucarelli
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| |
Collapse
|
26
|
Borel M, Lollo G, Magne D, Buchet R, Brizuela L, Mebarek S. Prostate cancer-derived exosomes promote osteoblast differentiation and activity through phospholipase D2. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165919. [PMID: 32800947 DOI: 10.1016/j.bbadis.2020.165919] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) is the most frequent cancer in men aged 65 and over. PCa mainly metastasizes in the bone, forming osteosclerotic lesions, inducing pain, fractures, and nerve compression. Cancer cell-derived exosomes participate in the metastatic spread, ranging from oncogenic reprogramming to the formation of pre-metastatic niches. Moreover, exosomes were recently involved in the dialog between PCa cells and the bone metastasis microenvironment. Phospholipase D (PLD) isoforms PLD1/2 catalyze the hydrolysis of phosphatidylcholine to yield phosphatidic acid (PA), regulating tumor progression and metastasis. PLD is suspected to play a role in exosomes biogenesis. We aimed to determine whether PCa-derived exosomes, through PLD, interact with the bone microenvironment, especially osteoblasts, during the metastatic process. Here we demonstrate for the first time that PLD2 is present in exosomes of C4-2B and PC-3 cells. C4-2B-derived exosomes activate proliferation and differentiation of osteoblasts models, by stimulating ERK 1/2 phosphorylation, by increasing the tissue-nonspecific alkaline phosphatase activity and the expression of osteogenic differentiation markers. Contrariwise, when C4-2B exosomes are generated in the presence of halopemide, a PLD pan-inhibitor, they lose their ability to stimulate osteoblasts. Furthermore, the number of released exosomes diminishes significantly (-40%). When the PLD product PA is combined with halopemide, exosome secretion is fully restored. Taken together, our results indicate that PLD2 stimulates exosome secretion in PCa cell models as well as their ability to increase osteoblast activity. Thus, PLD2 could be considered as a potent player in the establishment of PCa bone metastasis acting through tumor cell derived-exosomes.
Collapse
Affiliation(s)
- Mathieu Borel
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, UMR 5246, ICBMS, F-69622 Lyon, France
| | - Giovanna Lollo
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, UMR 5007, LAGEPP, F-69622 Lyon, France
| | - David Magne
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, UMR 5246, ICBMS, F-69622 Lyon, France
| | - René Buchet
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, UMR 5246, ICBMS, F-69622 Lyon, France
| | - Leyre Brizuela
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, UMR 5246, ICBMS, F-69622 Lyon, France
| | - Saida Mebarek
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, UMR 5246, ICBMS, F-69622 Lyon, France.
| |
Collapse
|
27
|
Bonollo F, Thalmann GN, Kruithof-de Julio M, Karkampouna S. The Role of Cancer-Associated Fibroblasts in Prostate Cancer Tumorigenesis. Cancers (Basel) 2020; 12:E1887. [PMID: 32668821 PMCID: PMC7409163 DOI: 10.3390/cancers12071887] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Tumors strongly depend on their surrounding tumor microenvironment (TME) for growth and progression, since stromal elements are required to generate the optimal conditions for cancer cell proliferation, invasion, and possibly metastasis. Prostate cancer (PCa), though easily curable during primary stages, represents a clinical challenge in advanced stages because of the acquisition of resistance to anti-cancer treatments, especially androgen-deprivation therapies (ADT), which possibly lead to uncurable metastases such as those affecting the bone. An increasing number of studies is giving evidence that prostate TME components, especially cancer-associated fibroblasts (CAFs), which are the most abundant cell type, play a causal role in PCa since the very early disease stages, influencing therapy resistance and metastatic progression. This is highlighted by the prognostic value of the analysis of stromal markers, which may predict disease recurrence and metastasis. However, further investigations on the molecular mechanisms of tumor-stroma interactions are still needed to develop novel therapeutic approaches targeting stromal components. In this review, we report the current knowledge of the characteristics and functions of the stroma in prostate tumorigenesis, including relevant discussion of normal prostate homeostasis, chronic inflammatory conditions, pre-neoplastic lesions, and primary and metastatic tumors. Specifically, we focus on the role of CAFs, to point out their prognostic and therapeutic potential in PCa.
Collapse
Affiliation(s)
- Francesco Bonollo
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
| | - George N. Thalmann
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
- Department of Urology, Inselspital, Bern University Hospital, 3008 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
- Department of Urology, Inselspital, Bern University Hospital, 3008 Bern, Switzerland
| | - Sofia Karkampouna
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
| |
Collapse
|
28
|
DOTA-ZOL: A Promising Tool in Diagnosis and Palliative Therapy of Bone Metastasis-Challenges and Critical Points in Implementation into Clinical Routine. Molecules 2020; 25:molecules25132988. [PMID: 32629930 PMCID: PMC7412164 DOI: 10.3390/molecules25132988] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/17/2023] Open
Abstract
The novel compound 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-ZOL (DOTA-conjugated zoledronic acid) is a promising candidate for the diagnosis and therapy of bone metastasis. The combination of the published methodology for this bisphosphonate with pharmaceutical and regulatory requirements turned out to be unexpectedly challenging. The scope of this work is the presentation and discussion of problems encountered during this process. Briefly, the radiolabelling process and purification, as well as the quality control published, did not meet the expectations. The constant effort setting up an automated radiolabelling procedure resulted in (a) an enhanced manual method using coated glass reactors, (b) a combination of three different reliable radio thin-layer chromatography (TLC) methods instead of the published and (c) a preliminary radio high-pressure liquid chromatography (HPLC) method for identification of the compound. Additionally, an automated radiolabelling process was developed, but it requires further improvement, e.g., in terms of a reactor vessel or purification of the crude product. The published purification method was found to be unsuitable for clinical routine, and an intense screening did not lead to a satisfactory result; here, more research is necessary. To sum up, implementation of DOTA-ZOL was possible but revealed a lot of critical points, of which not all could be resolved completely yet.
Collapse
|
29
|
Lin SR, Mokgautsi N, Liu YN. Ras and Wnt Interaction Contribute in Prostate Cancer Bone Metastasis. Molecules 2020; 25:E2380. [PMID: 32443915 PMCID: PMC7287876 DOI: 10.3390/molecules25102380] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent and malignant cancer types in men, which causes more than three-hundred thousand cancer death each year. At late stage of PCa progression, bone marrow is the most often metastatic site that constitutes almost 70% of metastatic cases of the PCa population. However, the characteristic for the osteo-philic property of PCa is still puzzling. Recent studies reported that the Wnt and Ras signaling pathways are pivotal in bone metastasis and that take parts in different cytological changes, but their crosstalk is not well studied. In this review, we focused on interactions between the Wnt and Ras signaling pathways during each stage of bone metastasis and present the fate of those interactions. This review contributes insights that can guide other researchers by unveiling more details with regard to bone metastasis and might also help in finding potential therapeutic regimens for preventing PCa bone metastasis.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Graduate Institute of Cancer Biology and Drug Discovery, Collage of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
| | - Ntlotlang Mokgautsi
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, Collage of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
| |
Collapse
|
30
|
Verma A, Artham S, Alwhaibi A, Adil MS, Cummings BS, Somanath PR. PAK1 inhibitor IPA-3 mitigates metastatic prostate cancer-induced bone remodeling. Biochem Pharmacol 2020; 177:113943. [PMID: 32240651 DOI: 10.1016/j.bcp.2020.113943] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Metastatic prostate cancer (PCa) has high mortality and a poor 5-year survival rate primarily due to the lack of effective treatments. Bone is the primary site of PCa metastasis in humans and the development of reliable therapeutic options for bone metastatic PCa will make a huge impact in reducing the mortality among these patients. Although P21 activated kinases (PAKs) have been studied in the past for their role in cancer, the efficacy of targeting PAKs to treat lung and bone metastatic PCa has not been tested yet. In the current study, we report that targeting PAK1 using IPA-3, an allosteric inhibitor of PAK1 kinase activity, significantly inhibits the murine metastatic PCa (RM1) cell proliferation and motility in vitro, and metastasis to the lungs in vivo. More importantly, we demonstrate for the first time that treatment with IPA-3 can blunt metastatic PCa-induced bone remodeling in vivo as analyzed by the 3-dimensional microcomputer tomography analysis. Our study has identified IPA-3 as a potential drug to treat bone metastatic PCa.
Collapse
Affiliation(s)
- Arti Verma
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Abdulrahman Alwhaibi
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Mir S Adil
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Brian S Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States; Department of Medicine and Vascular Biology Center, Augusta University, Augusta, GA, United States.
| |
Collapse
|
31
|
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
|
32
|
Ashraf MAB, Zahid A, Ashraf S, Waquar S, Iqbal S, Malik A. Implication of Prophetic Variables and their Impulsive Interplay in CA Prostate Patients Experiencing Osteo-Metastasis. Anticancer Agents Med Chem 2020; 20:2106-2113. [PMID: 32718298 DOI: 10.2174/1871520620666200727094430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/06/2020] [Accepted: 06/11/2020] [Indexed: 12/30/2022]
Abstract
AIMS To identify variables having a critical role in prostate cancer patients experiencing osteometastasis. BACKGROUND Prostatic carcinoma is a multifactorial complex disorder that exhibits an increased propensity to develop bone metastasis. An interplay of inflammatory and bone remodeling parameters promotes the formation of pre-metastatic niches in bones of patients, which could render them more vulnerable to skeletal disabilities. OBJECTIVE To evaluate the multi-dynamic inter-relationship of circulating variables in prostate cancer patients experiencing osteo-metastasis. MATERIALS AND METHODS Fifty-seven (n=57) men with clinically confirmed prostate cancer, fifty-nine (n=59) with skeletal metastases, and one hundred (n=100) healthy subjects i.e., men aging from 53-84 years with no clinical evidence of prostate were recruited from the Jinnah Hospital Lahore, Pakistan. Informed consent was obtained, and a venous blood sample was drawn and stored at -70oC until assayed. Levels of variables were evaluated using appropriate methods. Levels of Matrix Metalloproteinases (MMPs), Osteopontin (OPN), TGH- β, and sRANKL were estimated by the ELISA method. Each sample was suspended and the given protocol was employed. ELISA readings were obtained for the estimation of all variables. RESULTS Highly significant (P˂0.05) differential expression of oxidative stress, inflammatory cytokines, and bone remodeling variables were observed in localized and osteo-metastatic CA prostate patients. A strong positive correlation was revealed among OPN, sRANKL, MMP-7, MMP-9, PSA, and TGF-β (OPN vs. MMP-7, r=0.698* and OPN vs. MMP-9, r=0.765**, OPN vs. RANKL, =0.856*, sRANKL vs. MMP-9, r=0.825**, TGF- β vs. RANKL, r=0.868* and PSA vs. TGF- β, r=0.752*); lower levels of OPG were estimated in metastasized patients, showing that both osteolytic and osteoblastic phases of bone remodeling occur simultaneously. CONCLUSION The altered oxidative and inflammatory responses endorse Matrix Metalloproteinases (MMPs) increased activity, RANKL/OPG imbalance, and enhanced bone matrix proteins turnover, which can foster the process of osteo-metastasis. The perturbed RANKL/OPG drift and enhanced PSA levels are associated with increased TGF-β activity to aggravate Epithelial Mesenchymal transition (EM) and osteo-tropism of prostate cancer. Thus, designing novel targets of these major variables can minimize the incidence of prostate cancer patients.
Collapse
Affiliation(s)
- Muhammad A B Ashraf
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Ayesha Zahid
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Shazia Ashraf
- Department of Pathology, Independent Medical College, Faisalabad, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Saima Iqbal
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| |
Collapse
|
33
|
Han Z, Zhan R, Chen S, Deng J, Shi J, Wang W. miR-181b/Oncostatin m axis inhibits prostate cancer bone metastasis via modulating osteoclast differentiation. J Cell Biochem 2019; 121:1664-1674. [PMID: 31680294 DOI: 10.1002/jcb.29401] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
The activation of osteoblasts is significantly correlated to prostate tumor bone metastasis and bone loss. Oncostatin M (OSM) could promote breast cancer metastasis to bone. However, its role and mechanism in prostate cancer bone metastasis remain unclear. MicroRNAs (miRNAs) could play important roles in cancers via post-transcriptionally regulating target genes via binding to specific sequences in the 3' UTR of downstream target genes. In the present study, we performed microarray profiling analyses to identify differentially-expressed miRNAs in preosteoclast before and after osteoclast differentiation that could target OSM. miR-181b-5p was downregulated during Raw264.7 cells differentiation into osteoclast. By direct targeting OSM 3' UTR, miR-181b-5p inhibited OSM messenger RNA expression and protein levels, subsequently decreasing IL-6 and AREG and increasing OPG, while OSM overexpression exerted an opposing effect. More importantly, co-culture with miR-181b-5p-overexpressing differentiated Raw264.7 cells suppressed proliferation, migration, and invasion of mouse prostate cancer RM-1 cells, while co-culture with OSM-overexpressing Raw264.7 cells led to opposing cellular effects. More importantly, the effects of miR-181b-5p on osteoclastogenic factors and RM-1 cells could be significantly reversed by OSM overexpression. In summary, miR-181b-5p/OSM axis could be a viable therapeutic target for patients with surgically removed primary tumors to reduce bone metastasis and prevent bone loss.
Collapse
Affiliation(s)
- Ziwei Han
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruisen Zhan
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shijie Chen
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Deng
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Shi
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiguo Wang
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
34
|
González Díaz EC, Sinha S, Avedian RS, Yang F. Tissue-engineered 3D models for elucidating primary and metastatic bone cancer progression. Acta Biomater 2019; 99:18-32. [PMID: 31419564 DOI: 10.1016/j.actbio.2019.08.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/12/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Malignant bone tumors are aggressive neoplasms which arise from bone tissue or as a result of metastasis. The most prevalent types of cancer, such as breast, prostate, and lung cancer, all preferentially metastasize to bone, yet the role of the bone niche in promoting cancer progression remains poorly understood. Tissue engineering has the potential to bridge this knowledge gap by providing 3D in vitro systems that can be specifically designed to mimic key properties of the bone niche in a more physiologically relevant context than standard 2D culture. Elucidating the crucial components of the bone niche that recruit metastatic cells, support tumor growth, and promote cancer-induced destruction of bone tissue would support efforts for preventing and treating these devastating malignancies. In this review, we summarize recent efforts focused on developing in vitro 3D models of primary bone cancer and bone metastasis using tissue engineering approaches. Such 3D in vitro models can enable the identification of effective therapeutic targets and facilitate high-throughput drug screening to effectively treat bone cancers. STATEMENT OF SIGNIFICANCE: Biomaterials-based 3D culture have been traditionally used for tissue regeneration. Recent research harnessed biomaterials to create 3D in vitro cancer models, with demonstrated advantages over conventional 2D culture in recapitulating tumor progression and drug response in vivo. However, previous work has been largely limited to modeling soft tissue cancer, such as breast cancer and brain cancer. Unlike soft tissues, bone is characterized with high stiffness and mineral content. Primary bone cancer affects mostly children with poor treatment outcomes, and bone is the most common site of cancer metastasis. Here we summarize emerging efforts on engineering 3D bone cancer models using tissue engineering approaches, and future directions needed to further advance this relatively new research area.
Collapse
|
35
|
Ham J, Lever L, Fox M, Reagan MR. In Vitro 3D Cultures to Reproduce the Bone Marrow Niche. JBMR Plus 2019; 3:e10228. [PMID: 31687654 PMCID: PMC6820578 DOI: 10.1002/jbm4.10228] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/23/2019] [Accepted: 07/29/2019] [Indexed: 12/30/2022] Open
Abstract
Over the past century, the study of biological processes in the human body has progressed from tissue culture on glass plates to complex 3D models of tissues, organs, and body systems. These dynamic 3D systems have allowed for more accurate recapitulation of human physiology and pathology, which has yielded a platform for disease study with a greater capacity to understand pathophysiology and to assess pharmaceutical treatments. Specifically, by increasing the accuracy with which the microenvironments of disease processes are modeled, the clinical manifestation of disease has been more accurately reproduced in vitro. The application of these models is crucial in all realms of medicine, but they find particular utility in diseases related to the complex bone marrow niche. Osteoblast, osteoclasts, bone marrow adipocytes, mesenchymal stem cells, and red and white blood cells represent some of cells that call the bone marrow microenvironment home. During states of malignant marrow disease, neoplastic cells migrate to and join this niche. These cancer cells both exploit and alter the niche to their benefit and to the patient's detriment. Malignant disease of the bone marrow, both primary and secondary, is a significant cause of morbidity and mortality today. Innovative study methods are necessary to improve patient outcomes. In this review, we discuss the evolution of 3D models and compare them to the preceding 2D models. With a specific focus on malignant bone marrow disease, we examine 3D models currently in use, their observed efficacy, and their potential in developing improved treatments and eventual cures. Finally, we comment on the aspects of 3D models that must be critically examined as systems continue to be optimized so that they can exert greater clinical impact in the future. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Justin Ham
- Center for Molecular MedicineMaine Medical Center Research InstituteScarboroughMEUSA,University of New EnglandBiddefordMEUSA
| | - Lauren Lever
- Center for Molecular MedicineMaine Medical Center Research InstituteScarboroughMEUSA,University of New EnglandBiddefordMEUSA
| | - Maura Fox
- University of New EnglandBiddefordMEUSA
| | - Michaela R Reagan
- Center for Molecular MedicineMaine Medical Center Research InstituteScarboroughMEUSA,University of Maine Graduate School of Biomedical Science and EngineeringOronoMEUSA,Sackler School of Graduate Biomedical SciencesTufts UniversityBostonMAUSA
| |
Collapse
|
36
|
The Mode-of-Action of Targeted Alpha Therapy Radium-223 as an Enabler for Novel Combinations to Treat Patients with Bone Metastasis. Int J Mol Sci 2019; 20:ijms20163899. [PMID: 31405099 PMCID: PMC6720648 DOI: 10.3390/ijms20163899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022] Open
Abstract
Bone metastasis is a common clinical complication in several cancer types, and it causes a severe reduction in quality of life as well as lowering survival time. Bone metastases proceed through a vicious self-reinforcing cycle that can be osteolytic or osteoblastic in nature. The vicious cycle is characterized by cancer cells residing in bone releasing signal molecules that promote the differentiation of osteoclasts and osteoblasts either directly or indirectly. The increased activity of osteoclasts and osteoblasts then increases bone turnover, which releases growth factors that benefit metastatic cancer cells. In order to improve the prognosis of patients with bone metastases this cycle must be broken. Radium-223 dichloride (radium-223), the first targeted alpha therapy (TAT) approved, is an osteomimetic radionuclide that is incorporated into bone metastases where its high-linear energy transfer alpha radiation disrupts both the activity of bone cells and cancer cells. Therefore, radium-223 treatment has been shown preclinically to directly affect cancer cells in both osteolytic breast cancer and osteoblastic prostate cancer bone metastases as well as to inhibit the differentiation of osteoblasts and osteoclasts. Clinical studies have demonstrated an increase in survival in patients with metastatic castration-resistant prostate cancer. Due to the effectiveness and low toxicity of radium-223, several novel combination treatment strategies are currently eliciting considerable research interest.
Collapse
|
37
|
Kolb AD, Bussard KM. The Bone Extracellular Matrix as an Ideal Milieu for Cancer Cell Metastases. Cancers (Basel) 2019; 11:cancers11071020. [PMID: 31330786 PMCID: PMC6678871 DOI: 10.3390/cancers11071020] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/01/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Bone is a preferential site for cancer metastases, including multiple myeloma, prostate, and breast cancers.The composition of bone, especially the extracellular matrix (ECM), make it an attractive site for cancer cell colonization and survival. The bone ECM is composed of living cells embedded within a matrix composed of both organic and inorganic components. Among the organic components, type I collagen provides the tensile strength of bone. Inorganic components, including hydroxyapatite crystals, are an integral component of bone and provide bone with its rigidity. Under normal circumstances, two of the main cell types in bone, the osteoblasts and osteoclasts, help to maintain bone homeostasis and remodeling through cellular communication and response to biophysical signals from the ECM. However, under pathological conditions, including osteoporosis and cancer, bone remodeling is dysregulated. Once in the bone matrix, disseminated tumor cells utilize normal products of bone remodeling, such as collagen type I, to fuel cancer cell proliferation and lesion outgrowth. Models to study the complex interactions between the bone matrix and metastatic cancer cells are limited. Advances in understanding the interactions between the bone ECM and bone metastatic cancer cells are necessary in order to both regulate and prevent metastatic cancer cell growth in bone.
Collapse
Affiliation(s)
- Alexus D Kolb
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Karen M Bussard
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
38
|
Liu X, Chen L, Fan Y, Hong Y, Yang X, Li Y, Lu J, Lv J, Pan X, Qu F, Cui X, Gao Y, Xu D. IFITM3 promotes bone metastasis of prostate cancer cells by mediating activation of the TGF-β signaling pathway. Cell Death Dis 2019; 10:517. [PMID: 31273201 PMCID: PMC6609682 DOI: 10.1038/s41419-019-1750-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/15/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022]
Abstract
Advanced-stage prostate cancer (PCa) is often diagnosed with bone metastasis, for which there are limited therapies. Transforming growth factor β (TGF-β) is known to induce epithelial–mesenchymal transition (EMT), and abundance of TGF-β in the bone matrix is one of the important growth factors contributing to bone metastasis. TGF-β is reported as a key mediator of bone metastasis, but the underlying mechanism has not been elucidated. It was found in our study that Interferon-inducible Transmembrane Protein 3 (IFITM3) played a key role in the regulation of malignant tumor cell proliferation, invasion, and bone migration by binding to Smad4, thus activating the TGF-β-Smads Signaling Pathway. Lentivirus-mediated short hairpin RNA (shRNA) knockdown of IFITM3 inhibited cell proliferation and colony formation, induced apoptosis and inhibited migration by reversing EMT and downregulating the expression of metastasis-related molecules including FGFs and PTHrP. Microarray analysis showed that IFITM3 knockdown could alter the MAPK pathway associated with TGF-β-Smads signaling. By knocking down and overexpressing IFITM3, we demonstrated that IFITM3 expression level had an effect on MAPK pathway activation, and this change was more pronounced upon exogenous TGF-β stimulation. These results suggest that IFITM3 played an oncogenic role in PCa progression and bone metastasis via a novel TGF-β-Smads-MAPK pathway.
Collapse
Affiliation(s)
- Xi Liu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lu Chen
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yinghui Fan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Hong
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoqun Yang
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yao Li
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jianlei Lu
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jianmin Lv
- Department of Urology, Third Affiliated Hospital, Second Military Medical University, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, Third Affiliated Hospital, Second Military Medical University, Shanghai, China
| | - Fajun Qu
- Department of Urology, Third Affiliated Hospital, Second Military Medical University, Shanghai, China
| | - Xingang Cui
- Department of Urology, Third Affiliated Hospital, Second Military Medical University, Shanghai, China
| | - Yi Gao
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Danfeng Xu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
39
|
Stress-induced tunneling nanotubes support treatment adaptation in prostate cancer. Sci Rep 2019; 9:7826. [PMID: 31127190 PMCID: PMC6534589 DOI: 10.1038/s41598-019-44346-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
Tunneling nanotubes (TNTs) are actin-based membranous structures bridging distant cells for intercellular communication. We define roles for TNTs in stress adaptation and treatment resistance in prostate cancer (PCa). Androgen receptor (AR) blockade and metabolic stress induce TNTs, but not in normal prostatic epithelial or osteoblast cells. Co-culture assays reveal enhanced TNT formation between stressed and unstressed PCa cells as well as from stressed PCa to osteoblasts. Stress-induced chaperones clusterin and YB-1 localize within TNTs, are transported bi-directionally via TNTs and facilitate TNT formation in PI3K/AKT and Eps8-dependent manner. AR variants, induced by AR antagonism to mediate resistance to AR pathway inhibition, also enhance TNT production and rescue loss of clusterin- or YB-1-repressed TNT formation. TNT disruption sensitizes PCa to treatment-induced cell death. These data define a mechanistic network involving stress induction of chaperone and AR variants, PI3K/AKT signaling, actin remodeling and TNT-mediated intercellular communication that confer stress adaptative cell survival.
Collapse
|
40
|
Manna FL, Karkampouna S, Zoni E, De Menna M, Hensel J, Thalmann GN, Kruithof-de Julio M. Metastases in Prostate Cancer. Cold Spring Harb Perspect Med 2019; 9:a033688. [PMID: 29661810 PMCID: PMC6396340 DOI: 10.1101/cshperspect.a033688] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Prostate cancer (PCa) prognosis and clinical outcome is directly dependent on metastatic occurrence. The bone microenvironment is a favorable metastatic niche. Different biological processes have been suggested to contribute to the osteotropism of PCa such as hemodynamics, bone-specific signaling interactions, and the "seed and soil" hypothesis. However, prevalence of disseminating tumor cells in the bone is not proportional to the actual occurrence of metastases, as not all patients will develop bone metastases. The fate and tumor-reforming ability of a metastatic cell is greatly influenced by the microenvironment. In this review, the molecular mechanisms of bone and soft-tissue metastasis in PCa are discussed. Specific attention is dedicated to the residual disease, novel approaches, and animal models used in oncological translational research are illustrated.
Collapse
Affiliation(s)
- Federico La Manna
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Sofia Karkampouna
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Eugenio Zoni
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Marta De Menna
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Janine Hensel
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - George N Thalmann
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
41
|
Prostate cancer promotes a vicious cycle of bone metastasis progression through inducing osteocytes to secrete GDF15 that stimulates prostate cancer growth and invasion. Oncogene 2019; 38:4540-4559. [PMID: 30755731 DOI: 10.1038/s41388-019-0736-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/15/2019] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
Abstract
Bone is the most frequent site of prostate cancer (PCa) metastasis; however, little is known about the role of the most common cell in bone, the osteocyte (OCy), in cancer biology. In this study we explored the crosstalk between PCa cells and OCys to determine if it contributes to PCa progression. PCa cells induced OCys to promote PCa proliferation, migration and invasion. A chemokine screen revealed that PCa cell induced OCys to produce growth-derived factor 15 (GDF15). Knockdown of GDF15 in OCys demonstrated that PCa cells conferred the ability on OCys to promote PCa proliferation, migration and invasion through GDF15. Consistent with this finding was the observation that the GDF15 receptor, GFRAL, was expressed on multiple PCa cell lines. Transcription factor array screening of PCa cells exposed to OCys with or without knockdown of GDF15 revealed that GDF15 in OCys promoted early growth response 1 (EGR1) expression in the PCa cells. Knockdown of EGR1 expression in PCa cells revealed it was required for the OCy-derived GDF15-mediated induction of in vitro PCa cell proliferation, migration and invasion. Subcutaneous co-injection of PCa cells and OCys into mice revealed that OCys promoted tumor growth in vivo, which was diminished by knockdown of GDF15 in the OCys. Knockdown of GDF15 in the tibiae diminished growth of PCa cancer cells injected into the tibiae, which was accompanied by decreased tumor cell proliferation and EGR1 expression. These results shed light on a novel mechanism through which PCa cells educate OCys to promote progression of PCa bone metastasis. They also suggest that targeting of GDF15-based and EGR1-based signaling pathways should be further explored for their potential to diminish progression of PCa bone metastasis.
Collapse
|
42
|
Zoni E, Astrologo L, Ng CKY, Piscuoglio S, Melsen J, Grosjean J, Klima I, Chen L, Snaar-Jagalska EB, Flanagan K, van der Pluijm G, Kloen P, Cecchini MG, Kruithof-de Julio M, Thalmann GN. Therapeutic Targeting of CD146/MCAM Reduces Bone Metastasis in Prostate Cancer. Mol Cancer Res 2019; 17:1049-1062. [PMID: 30745464 DOI: 10.1158/1541-7786.mcr-18-1220] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/06/2018] [Accepted: 02/06/2019] [Indexed: 11/16/2022]
Abstract
Prostate Cancer is the most common cancer and the second leading cause of cancer-related death in males. When prostate cancer acquires castration resistance, incurable metastases, primarily in the bone, occur. The aim of this study is to test the applicability of targeting melanoma cell adhesion molecule (MCAM; CD146) with a mAb for the treatment of lytic prostate cancer bone metastasis. We evaluated the effect of targeting MCAM using in vivo preclinical bone metastasis models and an in vitro bone niche coculture system. We utilized FACS, cell proliferation assays, and gene expression profiling to study the phenotype and function of MCAM knockdown in vitro and in vivo. To demonstrate the impact of MCAM targeting and therapeutic applicability, we employed an anti-MCAM mAb in vivo. MCAM is elevated in prostate cancer metastases resistant to androgen ablation. Treatment with DHT showed MCAM upregulation upon castration. We investigated the function of MCAM in a direct coculture model of human prostate cancer cells with human osteoblasts and found that there is a reduced influence of human osteoblasts on human prostate cancer cells in which MCAM has been knocked down. Furthermore, we observed a strongly reduced formation of osteolytic lesions upon bone inoculation of MCAM-depleted human prostate cancer cells in animal model of prostate cancer bone metastasis. This phenotype is supported by RNA sequencing (RNA-seq) analysis. Importantly, in vivo administration of an anti-MCAM human mAb reduced the tumor growth and lytic lesions. These results highlight the functional role for MCAM in the development of lytic bone metastasis and suggest that MCAM is a potential therapeutic target in prostate cancer bone metastasis. IMPLICATIONS: This study highlights the functional application of an anti-MCAM mAb to target prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Eugenio Zoni
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Letizia Astrologo
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Charlotte K Y Ng
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Janine Melsen
- Department of Urology, Urology Research Laboratory Leiden University Medical Center, Leiden, the Netherlands
| | - Joël Grosjean
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Irena Klima
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Lanpeng Chen
- Institue of Biology, University of Leiden, Leiden, the Netherlands
| | | | - Kenneth Flanagan
- Prothena Biosciences, 331 Oyster Point Blvd, South San Francisco, California
| | - Gabri van der Pluijm
- Department of Urology, Urology Research Laboratory Leiden University Medical Center, Leiden, the Netherlands
| | - Peter Kloen
- Department of Orthopedic Trauma Surgery, Academic Medical Center, Amsterdam, the Netherlands
| | - Marco G Cecchini
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | | | - George N Thalmann
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Prostate cancer bone metastasis is the lethal progression of the disease. The disease frequently presents with osteoblastic lesions in bone. The tumor-induced bone can cause complications that significantly hamper the quality of life of patients. A better understanding of how prostate cancer induces aberrant bone formation and how the aberrant bone affects the progression and treatment of the disease may improve the therapies for this disease. RECENT FINDINGS Prostate cancer-induced bone was shown to enhance tumor growth and confer therapeutic resistance in bone metastasis. Clinically, Radium-223, an alpha emitter that selectively targets bone, was shown to improve overall survival in patients, supporting a role of tumor-induced bone in prostate cancer progression in bone. Recently, it was discovered that PCa-induced aberrant bone formation is due, in part, from tumor-associated endothelial cells that were converted into osteoblasts through endothelial-to-osteoblast (EC-to-OSB) conversion by tumor-secreted BMP4. The unique bone-forming phenotype of prostate cancer bone metastasis plays a role in prostate cancer progression in bone and therapy resistance. Therapies that incorporate targeting the tumor-induced osteoblasts or EC-to-OSB conversion mechanism may reduce tumor-induced bone formation and improve therapy outcomes.
Collapse
Affiliation(s)
- Song-Chang Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
44
|
Matsugaki A, Harada T, Kimura Y, Sekita A, Nakano T. Dynamic Collision Behavior Between Osteoblasts and Tumor Cells Regulates the Disordered Arrangement of Collagen Fiber/Apatite Crystals in Metastasized Bone. Int J Mol Sci 2018; 19:ijms19113474. [PMID: 30400633 PMCID: PMC6274720 DOI: 10.3390/ijms19113474] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/25/2018] [Accepted: 11/02/2018] [Indexed: 01/09/2023] Open
Abstract
Bone metastasis is one of the most intractable bone diseases; it is accompanied with a severe mechanical dysfunction of bone tissue. We recently discovered that the disorganized collagen/apatite microstructure in cancer-bearing bone is a dominant determinant of the disruption of bone mechanical function; disordered osteoblast arrangement was found to be one of the principal determinants of the deteriorated collagen/apatite microstructure. However, the precise molecular mechanisms regulating the disordered osteoblast arrangement triggered by cancer invasion are not yet understood. Herein, we demonstrate a significant disorganization of bone tissue anisotropy in metastasized bone in our novel ex vivo metastasis model. Further, we propose a novel mechanism underlying the disorganization of a metastasized bone matrix: A dynamic collision behavior between tumor cells and osteoblasts disturbs the osteoblast arrangement along the collagen substrate.
Collapse
Affiliation(s)
- Aira Matsugaki
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Suita 5650871, Japan.
| | - Tatsuki Harada
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Suita 5650871, Japan.
| | - Yumi Kimura
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Suita 5650871, Japan.
| | - Aiko Sekita
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Suita 5650871, Japan.
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Suita 5650871, Japan.
| |
Collapse
|
45
|
Burke M, Akens M, Kiss A, Willett T, Whyne C. Mechanical behavior of metastatic vertebrae are influenced by tissue architecture, mineral content, and organic feature alterations. J Orthop Res 2018; 36:3013-3022. [PMID: 29978906 DOI: 10.1002/jor.24105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/01/2018] [Indexed: 02/04/2023]
Abstract
Diminished vertebral mechanical behavior with metastatic involvement is typically attributed to modified architecture and trabecular bone content. Previous work has identified organic and mineral phase bone quality changes in the presence of metastases, yet limited work exists on the potential influence of such tissue level modifications on vertebral mechanical characteristics. This work seeks to determine correlations between features of bone (structural and tissue level) and mechanical behavior in metastatically involved vertebral bone. It is hypothesized that tissue level properties (mineral and organic) will improve these correlations beyond architectural properties and BMD alone. Twenty-four female athymic rats were inoculated with HeLa or Ace-1 cancer cells lines producing osteolytic (N = 8) or mixed (osteolytic/osteoblastic, N = 7) metastases, respectively. Twenty-one days post-inoculation L1-L3 pathologic vertebral motion segments were excised and μCT imaged. 3D morphometric parameters and axial rigidity of the L2 vertebrae were quantified. Sequential loading and μCT imaging measured progression of failure, stiffness and peak force. Relationships between mechanical testing (whole bone and tissue-level) and tissue-level material property modifications with metastatic involvement were evaluated utilizing linear regression models. Osteolytic involvement reduced vertebral trabecular bone volume, structure, CT-derived axial rigidity, stiffness and failure force compared to healthy controls (N = 9). Mixed metastases demonstrated similar trends. Previously assessed collagen cross-linking and proline-based residues were correlated to mechanical behavior and improved the predictive ability of the regression models. Similarly, collagen organization improved predictive regression models for metastatic bone hardness. This work highlights the importance of both bone content/architecture and organic tissue-level features in characterizing metastatic vertebral mechanics. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:3013-3022, 2018.
Collapse
Affiliation(s)
- Mikhail Burke
- Orthopaedics Biomechanics Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave., Room S620, Toronto, Ontario,. M4N 3M5.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario
| | - Margarete Akens
- Department of Surgery, University of Toronto, Toronto, Ontario.,Techna, University Health Network, Toronto, Ontario
| | - Alex Kiss
- Evaluative Clinical Sciences, Hurvitz Brain Science Program, Sunnybrook Research Institute, Toronto, Ontario
| | - Thomas Willett
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, Ontario
| | - Cari Whyne
- Orthopaedics Biomechanics Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave., Room S620, Toronto, Ontario,. M4N 3M5.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario.,Department of Surgery, University of Toronto, Toronto, Ontario
| |
Collapse
|
46
|
Gosewisch A, Delker A, Tattenberg S, Ilhan H, Todica A, Brosch J, Vomacka L, Brunegraf A, Gildehaus FJ, Ziegler S, Bartenstein P, Böning G. Patient-specific image-based bone marrow dosimetry in Lu-177-[DOTA 0,Tyr 3]-Octreotate and Lu-177-DKFZ-PSMA-617 therapy: investigation of a new hybrid image approach. EJNMMI Res 2018; 8:76. [PMID: 30076556 PMCID: PMC6081875 DOI: 10.1186/s13550-018-0427-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The bone marrow (BM) is a main organ at risk in Lu-177-PSMA-617 therapy of prostate cancer and Lu-177-Octreotate therapy of neuroendocrine tumours. BM dosimetry is challenging and time-consuming, as different sequential quantitative measurements must be combined. The BM absorbed dose from the remainder of the body (ROB) can be determined from sequential whole-body planar (WB-P) imaging, while quantitative Lu-177-SPECT allows for more robust tumour and organ absorbed doses. The aim was to investigate a time-efficient and patient-friendly hybrid protocol (HP) for the ROB absorbed dose to the BM. It combines three abdominal quantitative SPECT (QSPECT) scans with a single WB-P acquisition and was compared with a reference protocol (RP) using sequential WB-P in combination with sequential QSPECT images. We investigated five patients receiving 7.4 GBq Lu-177-Octreotate and five patients treated with 3.7 GBq Lu-177-PSMA-617. Each patient had WB-P and abdominal SPECT acquisitions 24 (+ CT), 48, and 72 h post-injection. Blood samples were drawn 30 min, 80 min, 24 h, 48 h, and 72 h post-injection. BM absorbed doses from the ROB were estimated from sequential WB-P images (RP), via a mono-exponential fit and mass-scaled organ-level S values. For the HP, a mono-exponential fit on the QSPECT data was scaled with the activity of one WB-P image acquired either 24, 48, or 72 h post-injection (HP24, HP48, HP72). Total BM absorbed doses were determined as a sum of ROB, blood, major organ, and tumour contributions. RESULTS Compared with the RP and for Lu-177-Octreotate therapy, median differences of the total BM absorbed doses were 13% (9-17%), 8% (4-15%), and 1% (0-5%) for the HP24, HP48, and HP72, respectively. For Lu-177-PSMA-617 therapy, total BM absorbed doses deviated 10% (2-20%), 3% (0-6%), and 2% (0-6%). CONCLUSION For both Lu-177-Octreotate and Lu-177-PSMA-617 therapy, BM dosimetry via sequential QSPECT imaging and a single WB-P acquisition is feasible, if this WB-P image is acquired at a late time point (48 or 72 h post-injection). The reliability of the HP can be well accepted considering the uncertainties of quantitative Lu-177 imaging and BM dosimetry using standardised organ-level S values.
Collapse
Affiliation(s)
- Astrid Gosewisch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Andreas Delker
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Sebastian Tattenberg
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Andrei Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Julia Brosch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Lena Vomacka
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Anika Brunegraf
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Franz Josef Gildehaus
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
| |
Collapse
|
47
|
The p21-activated kinase 4-Slug transcription factor axis promotes epithelial-mesenchymal transition and worsens prognosis in prostate cancer. Oncogene 2018; 37:5147-5159. [PMID: 29849120 DOI: 10.1038/s41388-018-0327-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/22/2018] [Accepted: 04/17/2018] [Indexed: 01/01/2023]
Abstract
Epithelial-mesenchymal transition (EMT) facilitates cancer invasion and metastasis and thus accelerates cancer progression. p21-activated kinase 4 (PAK4) is a critical regulator of prostate cancer (PC) progression. Here, we report that PAK4 activation promotes PC progression through the EMT regulator Slug. We find that phosphorylated PAK4S474 (pPAK4) levels, an index of PAK4 activation, were tightly associated with Gleason score (p < 0.001), a clinical indicator of PC progression, but not with prostate serum antigen levels or tumor stage. Stable silencing of PAK4 in PC cells reduced their potential for EMT, cellular invasion, and metastasis in vivo. PAK4 bound and directly phosphorylated Slug at two previously unknown sites, S158 and S254, which resulted in its stabilization. The non-phosphorylatable form SlugS158A/S254A upregulated transcription of CDH1, which encodes E-cadherin, and thus suppressed EMT and invasion, to a greater extent than did wild-type Slug. The strong EMT inducer TGF-β elevated pPAK4 and pSlugS158 levels; PAK4 knockdown or introduction of a dominant-negative form of PAK4 inhibited both TGF-β-stimulated EMT and an increase in pSlugS158 levels. Finally, immunohistochemistry revealed a positive correlation between pPAK4 and pSlugS158 but an inverse correlation between pSlugS158 and E-cadherin. The results suggest that the PAK4-Slug axis represents a novel pathway that promotes PC progression.
Collapse
|
48
|
Targeted alpha therapy using Radium-223: From physics to biological effects. Cancer Treat Rev 2018; 68:47-54. [PMID: 29859504 DOI: 10.1016/j.ctrv.2018.05.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
With the advance of the use of ionizing radiation in therapy, targeted alpha therapy (TAT) has assumed an important role around the world. This kind of therapy can potentially reduce side effects caused by radiation in normal tissues and increased destructive radiobiological effects in tumor cells. However, in many countries, the use of this therapy is still in a pioneering phase. Radium-223 (223Ra), an alpha-emitting radionuclide, has been the first of its kind to be approved for the treatment of bone metastasis in metastatic castration-resistant prostate cancer. Nevertheless, the interaction mechanism and the direct effects of this radiopharmaceutical in tumor cells are not fully understood neither characterized at a molecular level. In fact, the ways how TAT is linked to radiobiological effects in cancer is not yet revised. Therefore, this review introduces some physical properties of TAT that leads to biological effects and links this information to the hallmarks of cancer. The authors also collected the studies developed with 223Ra to correlate with the three categories reviewed - properties of TAT, 5 R's of radiobiology and hallmarks of cancer- and with the promising future to this radiopharmaceutical.
Collapse
|
49
|
Karlsson T, Sundar R, Widmark A, Landström M, Persson E. Osteoblast-derived factors promote metastatic potential in human prostate cancer cells, in part via non-canonical transforming growth factor β (TGFβ) signaling. Prostate 2018; 78:446-456. [PMID: 29383751 DOI: 10.1002/pros.23489] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Transforming growth factor β (TGFβ) functions as a double-edged sword in prostate cancer tumorigenesis. In initial stages of the disease, TGFβ acts as a growth inhibitor upon tumor cells, whereas it in later stages of disease rather promotes invasion and metastatic potential. One well-known cellular source of TGFβ in the bone metastatic site is the bone-forming osteoblasts. Here we have studied the effects by osteoblast-derived factors on metastatic potential in several human prostate cancer cell lines. METHODS Effects on metastatic potential in prostate cancer cells by osteoblast-derived factors were studied in vitro using several methods, including Transwell migration and evaluation of formation of pro-migratory protrusions. Confocal microscopy was used to evaluate possible changes in differentiation state in tumor cells by analysis of markers for epithelial-to-mesenchymal transition (EMT). The Matrigel-on-top 3D culture method was used for further assessment of metastatic characteristics in tumor cells by analysis of formation of filopodium-like protrusions (FLPs). RESULTS Osteoblast-derived factors increased migration of PC-3U cells, an effect less prominent in cells overexpressing a mutated type I TGFβ receptor (TβRI) preventing non-canonical TRAF6-dependent TGFβ signaling. Osteoblast-derived factors also increased the formation of long protrusions and loss of cell-cell contacts in PC-3U cells, suggesting induction of a more aggressive phenotype. In addition, treatment with TGFβ or osteoblast-derived factors of PC-3U cells in Matrigel-on-top 3D cultures promoted formation of FLPs, previously shown to be essential for metastatic establishment. CONCLUSIONS These findings suggests that factors secreted from osteoblasts, including TGFβ, can induce several cellular traits involved in metastatic potential of PC-3U cells, further strengthening the role for bone cells to promote metastatic tumor cell behavior.
Collapse
Affiliation(s)
- Terese Karlsson
- Department of Radiation Sciences, Section for Oncology, Umeå University, Umeå, Sweden
| | - Reshma Sundar
- Department of Medical Biosciences, Section for Pathology, Umeå University, Umeå, Sweden
| | - Anders Widmark
- Department of Radiation Sciences, Section for Oncology, Umeå University, Umeå, Sweden
| | - Maréne Landström
- Department of Medical Biosciences, Section for Pathology, Umeå University, Umeå, Sweden
| | - Emma Persson
- Department of Radiation Sciences, Section for Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
50
|
Self-assembling nanoparticles encapsulating zoledronic acid inhibit mesenchymal stromal cells differentiation, migration and secretion of proangiogenic factors and their interactions with prostate cancer cells. Oncotarget 2018; 8:42926-42938. [PMID: 28477013 PMCID: PMC5522116 DOI: 10.18632/oncotarget.17216] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/22/2017] [Indexed: 12/16/2022] Open
Abstract
Zoledronic Acid (ZA) rapidly concentrates into the bone and reduces skeletal-related events and pain in bone metastatic prostate cancer (PCa), but exerts only a limited or absent impact as anti-cancer activity. Recently, we developed self-assembling nanoparticles (NPS) encapsulating zoledronic acid (NZ) that allowed a higher intratumor delivery of the drug compared with free zoledronic acid (ZA) in in vivo cancer models of PCa. Increasing evidence suggests that Bone Marrow (BM) Mesenchymal stromal cells (BM-MSCs) are recruited into the stroma of developing tumors where they contribute to progression by enhancing tumor growth and metastasis. We demonstrated that treatment with NZ decreased migration and differentiation into adipocytes and osteoblasts of MSCs and inhibited osteoclastogenesis. Treatment with NZ reduced the capability of MSCs to promote the migration and the clonogenic growth of the prostate cancer cell lines PC3 and DU145. The levels of Interleukin-6 and of the pro-angiogenic factors VEGF and FGF-2 were significantly reduced in MSC-CM derived from MSCs treated with NZ, and CCL5 secretion was almost totally abolished. Moreover, treatment of MSCs with supernatants from PC3 cells, leading to tumor-educated MSCs (TE-MSCs), increased the secretion of IL-6, CCL5, VEGF and FGF-2 by MSCs and increased their capability to increase PC3 cells clonogenic growth. Treatment with NZ decreased cytokine secretion and the pro-tumorigenic effects also of TE-MSCS. In conclusion, demonstrating that NZ is capable to inhibit the cross talk between MSCs and PCa, this study provides a novel insight to explain the powerful anticancer activity of NZ on PCa.
Collapse
|