Please wait a minute...
Search Asian J Urol Advanced Search
Share 
Asian Journal of Urology, 2016, 3(4): 229-239    
  本期目录 | 过刊浏览 | 高级检索 |
Specific bone region localization of osteolytic versus osteoblastic lesions in a patient-derived xenograft model of bone metastatic prostate cancer
Takeshi Hirataa, Seung Chol Parkb, Michelle T. Muldongc,d,e, Christina N. Wuc,f, Tomonori Yamaguchig, Amy Strasnerc,d,e, Omer Raheemd,e, Hiromi Kumona, Robert L. Sahh, Nicholas A. Cacalanoi, Catriona H. M. Jamiesonc,f, Christopher J. Kanec,d,e, Koichi Masudag, Anna A. Kulidjianc,g, Christina A. M. Jamiesonc,d,e
a Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan;
b Department of Urology, Wonkwang University School of Medicine and Hospital, Iksan, South Korea;
c Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA;
d Department of Urology, University of California, San Diego, La Jolla, CA, USA;
e Department of Surgery, University of California, San Diego, La Jolla, CA, USA;
f Department of Medicine, University of California, San Diego, La Jolla, CA, USA;
g Department of Orthopaedic Surgery, School of Medicine, University of California, San Diego, La Jolla, CA, USA;
h Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA;
i Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, USA
Specific bone region localization of osteolytic versus osteoblastic lesions in a patient-derived xenograft model of bone metastatic prostate cancer
Takeshi Hirataa, Seung Chol Parkb, Michelle T. Muldongc,d,e, Christina N. Wuc,f, Tomonori Yamaguchig, Amy Strasnerc,d,e, Omer Raheemd,e, Hiromi Kumona, Robert L. Sahh, Nicholas A. Cacalanoi, Catriona H. M. Jamiesonc,f, Christopher J. Kanec,d,e, Koichi Masudag, Anna A. Kulidjianc,g, Christina A. M. Jamiesonc,d,e
a Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan;
b Department of Urology, Wonkwang University School of Medicine and Hospital, Iksan, South Korea;
c Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA;
d Department of Urology, University of California, San Diego, La Jolla, CA, USA;
e Department of Surgery, University of California, San Diego, La Jolla, CA, USA;
f Department of Medicine, University of California, San Diego, La Jolla, CA, USA;
g Department of Orthopaedic Surgery, School of Medicine, University of California, San Diego, La Jolla, CA, USA;
h Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA;
i Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, USA
下载:  PDF (9505KB) 
输出:  BibTeX | EndNote (RIS)      
摘要 Objective: Bone metastasis occurs in up to 90% of men with advanced prostate cancer and leads to fractures, severe pain and therapy-resistance. Bone metastases induce a spectrum of types of bone lesions which can respond differently to therapy even within individual prostate cancer patients. Thus, the special environment of the bone makes the disease more complicated and incurable. A model in which bone lesions are reproducibly induced that mirrors the complexity seen in patients would be invaluable for pre-clinical testing of novel treatments. The microstructural changes in the femurs of mice implanted with PCSD1, a new patient-derived xenograft from a surgical prostate cancer bone metastasis specimen, were determined. Methods: Quantitative micro-computed tomography (micro-CT) and histological analyses were performed to evaluate the effects of direct injection of PCSD1 cells or media alone (Control) into the right femurs of Rag2-/-γc-/- male mice. Results: Bone lesions formed only in femurs of mice injected with PCSD1 cells. Bone volume (BV) was significantly decreased at the proximal and distal ends of the femurs (p<0.01) whereas BV (p<0.05) and bone shaft diameter (p<0.01) were significantly increased along the femur shaft. Conclusion: PCSD1 cells reproducibly induced bone loss leading to osteolytic lesions at the ends of the femur, and, in contrast, induced aberrant bone formation leading to osteoblastic lesions along the femur shaft. Therefore, the interaction of PCSD1 cells with different bone region-specific microenvironments specified the type of bone lesion. Our approach can be used to determine if different bone regions support more therapy resistant tumor growth, thus, requiring novel treatments.
服务
把本文推荐给朋友
加入引用管理器
E-mail Alert
RSS
作者相关文章
Takeshi Hirata
Seung Chol Park
Michelle T. Muldong
Christina N. Wu
Tomonori Yamaguchi
Amy Strasner
Omer Raheem
Hiromi Kumon
Robert L. Sah
Nicholas A. Cacalano
Catriona H. M. Jamieson
Christopher J. Kane
Koichi Masuda
Anna A. Kulidjian
Christina A. M. Jamieson
关键词:  Bone metastatic prostate cancer  Patient-derived xenograft  Bone microenvironment  Microstructural CT  Osteolytic lesions  Osteoblastic lesions    
Abstract: Objective: Bone metastasis occurs in up to 90% of men with advanced prostate cancer and leads to fractures, severe pain and therapy-resistance. Bone metastases induce a spectrum of types of bone lesions which can respond differently to therapy even within individual prostate cancer patients. Thus, the special environment of the bone makes the disease more complicated and incurable. A model in which bone lesions are reproducibly induced that mirrors the complexity seen in patients would be invaluable for pre-clinical testing of novel treatments. The microstructural changes in the femurs of mice implanted with PCSD1, a new patient-derived xenograft from a surgical prostate cancer bone metastasis specimen, were determined. Methods: Quantitative micro-computed tomography (micro-CT) and histological analyses were performed to evaluate the effects of direct injection of PCSD1 cells or media alone (Control) into the right femurs of Rag2-/-γc-/- male mice. Results: Bone lesions formed only in femurs of mice injected with PCSD1 cells. Bone volume (BV) was significantly decreased at the proximal and distal ends of the femurs (p<0.01) whereas BV (p<0.05) and bone shaft diameter (p<0.01) were significantly increased along the femur shaft. Conclusion: PCSD1 cells reproducibly induced bone loss leading to osteolytic lesions at the ends of the femur, and, in contrast, induced aberrant bone formation leading to osteoblastic lesions along the femur shaft. Therefore, the interaction of PCSD1 cells with different bone region-specific microenvironments specified the type of bone lesion. Our approach can be used to determine if different bone regions support more therapy resistant tumor growth, thus, requiring novel treatments.
Key words:  Bone metastatic prostate cancer    Patient-derived xenograft    Bone microenvironment    Microstructural CT    Osteolytic lesions    Osteoblastic lesions
收稿日期:  2016-06-09      修回日期:  2016-08-26           出版日期:  2016-10-01      发布日期:  2016-11-02      整期出版日期:  2016-10-01
通讯作者:  Christina A.M. Jamieson,E-mail address: camjamieson@ucsd.edu    E-mail:  camjamieson@ucsd.edu
引用本文:    
Takeshi Hirata, Seung Chol Park, Michelle T. Muldong, Christina N. Wu, Tomonori Yamaguchi, Amy Strasner, Omer Raheem, Hiromi Kumon, Robert L. Sah, Nicholas A. Cacalano, Catriona H. M. Jamieson, Christopher J. Kane, Koichi Masuda, Anna A. Kulidjian, Christina A. M. Jamieson. Specific bone region localization of osteolytic versus osteoblastic lesions in a patient-derived xenograft model of bone metastatic prostate cancer[J]. Asian Journal of Urology, 2016, 3(4): 229-239.
Takeshi Hirata, Seung Chol Park, Michelle T. Muldong, Christina N. Wu, Tomonori Yamaguchi, Amy Strasner, Omer Raheem, Hiromi Kumon, Robert L. Sah, Nicholas A. Cacalano, Catriona H. M. Jamieson, Christopher J. Kane, Koichi Masuda, Anna A. Kulidjian, Christina A. M. Jamieson. Specific bone region localization of osteolytic versus osteoblastic lesions in a patient-derived xenograft model of bone metastatic prostate cancer. Asian Journal of Urology, 2016, 3(4): 229-239.
链接本文:  
http://www.ajurology.com/CN/  或          http://www.ajurology.com/CN/Y2016/V3/I4/229
[1] Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics. CA Cancer J Clin 2014;64:9-29.
[2] Oster G, Lamerato L, Glass AG, Richert-Boe KE, Lopez A, Chung K, et al. Natural history of skeletal-related events in patients with breast, lung, or prostate cancer and metastases to bone:a 15-year study in two large US health systems. Support Care Cancer 2013;21:3279-86.
[3] Garnero P, Buchs N, Zekri J, Rizzoli R, Coleman RE, Delmas PD. Markers of bone turnover for the management of patients with bone metastases from prostate cancer. Br J Cancer 2000;82:858-64.
[4] Brown JE, Neville-Webbe H, Coleman RE. The role of bisphosphonates in breast and prostate cancers. Endocr Relat Cancer 2004;11:207-24.
[5] Shiao SL, Chu GC, Chung LW. Regulation of prostate cancer progression by the tumor microenvironment. Cancer Lett 2016;380:340-8.
[6] Vargas HA, Wassberg C, Fox JJ, Wibmer A, Goldman DA, Kuk D, et al. Bone metastases in castration-resistant prostate cancer:associations between morphologic CT patterns, glycolytic activity, and androgen receptor expression on PET and overall survival. Radiology 2014;271:220-9.
[7] Roudier MP, Morrissey C, True LD, Higano CS, Vessella RL, Ott SM. Histopathological assessment of prostate cancer bone osteoblastic metastases. J Urol 2008;180:1154-60.
[8] Morrissey C, Roudier MP, Dowell A, True LD, Ketchanji M, Welty C, et al. Effects of androgen deprivation therapy and bisphosphonate treatment on bone in patients with metastatic castration-resistant prostate cancer:results from the University of Washington Rapid Autopsy Series. J Bone Min Res 2013;28:333-40.
[9] Larson SR, Zhang X, Dumpit R, Coleman I, Lakely B, Roudier M, et al. Characterization of osteoblastic and osteolytic proteins in prostate cancer bone metastases. Prostate 2013;73:932-40.
[10] Nandana S, Chung LW. Prostate cancer progression and metastasis:potential regulatory pathways for therapeutic targeting. Am J Clin Exp Urol 2014;2:92-101.
[11] Thalmann GN, Rhee H, Sikes RA, Pathak S, Multani A, Zhau HE, et al. Human prostate fibroblasts induce growth and confer castration resistance and metastatic potential in LNCaP Cells. Eur Urol 2010;58:162-71.
[12] Kaighn ME, Lechner JF, Narayan KS, Jones LW. Prostatic carcinoma:tissue culture cell lines. NatI Cancer Inst Monogr 1978;49:17-21.
[13] Kaighn ME, Narayan KS, Ohnukl Y, Lechner JF, Jones LW. Establishment and characterization of a human prostatic carcinoma cell line (PC-3). Invest Urol 1979;17:16-23.
[14] Lee Y, Schwarz E, Davies M, Jo M, Gates J, Wu J, et al. Differences in the cytokine profiles associated with prostate cancer cell induced osteoblastic and osteolytic lesions in bone. J Orthop Res 2003;21:62-72.
[15] Hsu WK, Virk MS, Feeley BT, Stout DB, Chatziioannou AF, Lieberman JR. Characterization of osteolytic, osteoblastic, and mixed lesions in a prostate cancer mouse model using 18F-FDG and 18F-fluoride PET/CT. J Nucl Med 2008;49:414-21.
[16] Li ZG, Mathew P, Yang J, Starbuck MW, Zurita AJ, Liu J, et al. Androgen receptor-negative human prostate cancer cells induce osteogenesis in mice through FGF9-mediated mechanisms. J Clin Invest 2008;118:2697-710.
[17] Nguyen HM, Ruppender N, Zhang X, Brown LG, Gross TS, Morrissey C, et al. Cabozantinib inhibits growth of androgensensitive and castration-resistant prostate cancer and affects bone remodeling. PLoS One 2013;8:e78881.
[18] Vignani F, Bertaglia V, Buttigliero C, Tucci M, Scagliotti GV, Di Maio M. Skeletal metastases and impact of anticancer and bone-targeted agents in patients with castration-resistant prostate cancer. Cancer Treat Rev 2016;44:61-73.
[19] Raheem O, Kulidjian AA, Wu C, Jeong YB, Yamaguchi T, Smith KM, et al. A novel patient-derived intra-femoral xenograft model of bone metastatic prostate cancer that recapitulates mixed osteolytic and osteoblastic lesions. J Transl Med 2011;9:185.
[20] Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Müller R. Guidelines for assessment of bone microstructure in rodents using micro-computed tomography. J Bone Min Res 2010;25:1468-86.
[21] http://www.skyscan.be/next/ctan_usermanual.pdf#searchZ%20CTan+manual%20.
[22] Otsu N. A threshold selection method from gray-level histograms. IEEE Trans Sys, Man, Cyber 1979;9:62-6.
[23] Dempster DW, Compston JE, Drezner MK, Glorieux FH, Kanis JA, Malluche H, et al. Standardized nomenclature, symbols, and units for bone histomorphometry:a 2012 update of the report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Min Res 2013;28:2-17.
[24] Cook MJ. The Anatomy of the Laboratory Mouse. London:Academic Press 1965. Adapted for the web by Mouse Genome Informatics:The Jackson Laboratory Bar Harbor, Maine April 2005, Revised February 2008.
[25] Guezguez B, Campbell CJ, Boyd AL, Karanu F, Casado FL, Di Cresce C, et al. Regional localization within the bone marrow influences the functional capacity of human HSCs. Cell Stem Cell 2013;13:175-89.
[26] Goff DJ, Court Recart A, Sadarangani A, Chun HJ, Barrett CL, Krajewska M, et al. A Pan-BCL2 inhibitor renders bonemarrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell 2013;12:316-28.
[27] Juárez P, Guise TA. TGF-β in cancer and bone:implications for treatment of bone metastases. Bone 2011;48:23-9.
[28] Casimiro S, Mohammad KS, Pires R, Tato-Costa J, Alho I, Teixeira R, et al. RANKL/RANK/MMP-1 molecular triad contributes to the metastatic phenotype of breast and prostate cancer cells in vitro. PLoS One 2013;8:e63153.
[29] Xu W, Zhang Z, Yang Y, Hu Z, Wang CH, Morgan M, et al. Ad5/48 hexon oncolytic virus expressing sTGFβRIIFc produces reduced hepatic and systemic toxicities and inhibits prostate cancer bone metastases. Mol Ther 2014;22:1504-17.
[30] Godebu E, Muldong M, Strasner A, Wu CN, Park SC, Woo JR, et al. PCSD1, a new patient-derived model of bone metastatic prostate cancer, is castrate-resistant in the bone-niche. J Transl Med 2014;12:275.
No related articles found!
[1] Zhixiang Wang, Bing Liu, Xiaofeng Gao, Yi Bao, Yang Wang, Huamao Ye, Yinghao Sun, Linhui Wang. Laparoscopic ureterolysis with simultaneous ureteroscopy and percutaneous nephroscopy for treating complex ureteral obstruction after failed endoscopic intervention: A technical report[J]. Asian Journal of Urology, 2015, 2(4): 238 -243 .
[2] Louis R. Kavoussi. News from leading international academic urology departments[J]. Asian Journal of Urology, 2017, 4(1): 1 -2 .
[3] Rikiya Taoka, Yoshiyuki Kakehi. The influence of asymptomatic inflammatory prostatitis on the onset and progression of lower urinary tract symptoms in men with histologic benign prostatic hyperplasia[J]. Asian Journal of Urology, 2017, 4(3): 158 -163 .
[4] Cheuk Fan Shum, Weida Lau, Chang Peng Colin Teo. Medical therapy for clinical benign prostatic hyperplasia:a1 Antagonists, 5a reductase inhibitors and their combination[J]. Asian Journal of Urology, 2017, 4(3): 185 -190 .
[5] Foo Keong Tatt. Current consensus and controversies on male LUTS/BPH (part two)[J]. Asian Journal of Urology, 2018, 5(1): 8 -9 .
[6] Rishi R. Sekar, Claire M. De La Calle, Dattatraya Patil, Sarah A. Holzman, Yoram Baum, Umer Sheikh, Jonathan H. Huang, Adeboye O. Osunkoya, Brian P. Pollack, Haydn T. Kissick, Kenneth Ogan, Viraj A. Master. Major histocompatibility complex I upregulation in clear cell renal cell carcinoma is associated with increased survival[J]. Asian Journal of Urology, 2016, 3(2): 75 -81 .
[7] Ryan Yu, Jefferson Terry, Mutaz Alnassar, Jorge Demaria. Pediatric fibrous pseudotumor of the tunica vaginalis testis[J]. Asian Journal of Urology, 2016, 3(2): 99 -102 .
[8] Aso Omer Rashid, Saman Salih Fakhulddin. Risk factors for fever and sepsis after percutaneous nephrolithotomy[J]. Asian Journal of Urology, 2016, 3(2): 82 -87 .
[9] Christopher Hartman, Nikhil Gupta, David Leavitt, David Hoenig, Zeph Okeke, Arthur Smith. Advances in percutaneous stone surgery[J]. Asian Journal of Urology, 2015, 2(1): 26 -32 .
[10] Aldamanhori Reem,I.Osman Nadir,R.Chapple Christopher. Underactive bladder: Pathophysiology and clinical significance[J]. Asian Journal of Urology, 2018, 5(1): 17 -21 .
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed