j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 4 ) 1 e1 0

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 Q14 17 18 19 20 21 22 23 24 25 26 Q1 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

Available online at www.sciencedirect.com

ScienceDirect journal homepage: www.JournalofSurgicalResearch.com

Fluorescence-guided surgery of prostate cancer bone metastasis Shinji Miwa, MD, PhD,a,b,c Yasunori Matsumoto, MD,b Yukihiko Hiroshima, MD, PhD,a,b Shuya Yano, MD,a,b Fuminari Uehara, MD,a,b Mako Yamamoto, PhD,a,b Yong Zhang, MD,a Hiroaki Kimura, MD, PhD,c Katsuhiro Hayashi, MD, PhD,c Norio Yamamoto, MD, PhD,c Michael Bouvet, MD,b,* Naotoshi Sugimoto, MD, PhD,d Hiroyuki Tsuchiya, MD, PhD,c and Robert M. Hoffman, PhDa,b a

AntiCancer, Inc, San Diego, California Department of Surgery, University of California, San Diego, San Diego, California c Department of Orthopedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan d Department of Physiology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan b

article info

abstract

Article history:

Background: The aim of this study is to investigate the effectiveness of fluorescence-guided

Received 12 February 2014

surgery (FGS) of prostate cancer experimental skeletal metastasis.

Received in revised form

Materials and methods: Green fluorescent protein-expressing PC-3 human prostate cancer

16 March 2014

cells (PC-3-green fluorescent protein) were injected into the intramedullary cavity of the

Accepted 16 May 2014

tibia in 32 nude mice. After 2 wk, 16 of the mice underwent FGS; the other 16 mice un-

Available online xxx

derwent bright-light surgery (BLS). Half of BLS and FGS mice (8 mice in each group) received zoledronic acid (ZOL). Weekly fluorescence imaging of the mice was performed. Six weeks

Keywords:

after surgery, metastases to lung and inguinal lymph node were evaluated by fluorescence

Prostate cancer

imaging.

Bone metastasis

Results: The percentage of residual tumor after BLS and FGS was 9.9  2.2% and 0.9  0.3%,

GFP

respectively (P < 0.001). FGS reduced recurrent cancer growth compared with BLS (P
36 wk. The median overall survival increased from 16 wk in the BLS group to 31 wk in the FGS group. FGS resulted in a cure in 67% of mice (alive without evidence of tumor at >6 mo after surgery) compared with only 37% of mice that underwent BLS [17]. In the present report, we determined the advantage of FGS of red fluorescent protein-labeled experimental prostate cancer bone metastasis with and without zoledronic acid (ZOL).

2.

Materials and methods

2.1.

Cell lines and cell culture

The wild-type PC-3 cells, originally isolated from a human prostate adenocarcinoma that had metastasized to the bone, were genetically engineered to express GFP using a retrovirus expression vector [3]. PC-3-GFP cells were cultured in DMEM supplemented with 1% penicillin/streptomycin (Invitrogen) and 10% fetal bovine serum (SigmaeAldrich) at 37 C with 95% air and 5% CO2.

2.2.

Animal care

Athymic NCr nude mice (nu/nu) (AntiCancer Inc) aged 6e8 wk, were used in this study. Mice were maintained in a barrier facility on high efficiency particulate air-filtered racks. The animals were fed with autoclaved laboratory rodent diet. Animal experiments were performed in accordance with the Guide-lines for the Care and Use of Laboratory Animals under National Institutes of Health assurance number A3873-01.

2.3.

Tumor implantation

Six to eight-week-old male mice were anesthetized by a ketamine mixture (10 mL ketamine HCL, 7.6 mL xylazine, 2.4 mL

5

Fig. 5 e Time course of tumor growth in the bone visualized by fluorescent area. (A) FGS significantly reduced the recurrent tumor growth of prostate cancer cells in the bone (P [ 0.006). (B) ZOL slightly inhibited recurrent tumor growth in the bone, but not significantly. The data are expressed mean ± standard error of the mean. Statistical analysis was performed using the Student t-test. **P < 0.01, compared with BLS group.

acepromazine maleate, and 10 mL H2O) via subcutaneous injection. The left leg was sterilized with alcohol and an w5 mm midline skin incision was made just below the knee joint to expose the tibial tuberosity (Fig. 1A). PC-3-GFP cells (5  105) in 5 mL Matrigel (BD Bioscience) per mouse were injected into the Q6 intramedullary cavity of the left tibia with a 0.5 mL 28 G latexfree insulin syringe (TYCO Health Group LP). The skin was closed with a 6-0 suture. Two weeks after injection, fluorescence imaging was performed to confirm the tumor growing with GFP expression using the OV100 Small Imaging System (Olympus Corp) (Fig. 1B) [25]. Q7

2.4.

Tumor resection

A total of 32 mice were used in the experiments: 16 mice underwent FGS, and the other 16 mice underwent bright-light surgery (BLS) (Fig. 2). Two weeks after the implantation of the tumor cells, tumor-bearing mice were randomly assigned to the BLS or FGS group. Before resection of the metastatic tumor, mice were anesthetized by ketamine mixture, and their limbs were sterilized with alcohol. An w1.5 cm incision was made above the tumor. Resection of the metastatic bone tumor was performed using Dino-Lite digital camera

5.2.0 DTD  YJSRE12751_proof  11 June 2014  1:55 am  ce

586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650

6

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 4 ) 1 e1 0

651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 Fig. 6 e Time lapse imaging of tumor growth after BLS and FGS. (For interpretation of the references to color in this figure, 674 the reader is referred to the web version of this article.) 675 676 677 (AM4113T-GFBW Dino-Lite Premier; AnMo Electronics Corp, intralesional or marginal tumor excision was performed in all 678 the mice. Postoperatively, the surgical resection bed was 679 Q8 Taiwan) under bright-light illumination for the BLS group and under fluorescence illumination through a GFP filter for the imaged with the OV100 imaging system under both standard 680 681 FGS group [26]. For metastatic bone tumor resection, bright field and fluorescence illumination to assess 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 Fig. 7 e KaplaneMeier curve for DFS. (A) KaplaneMeier curve for DFS of mice in each group. (B) KaplaneMeier curve for DFS 715 of BLS and FGS mice. FGS significantly improved DFS. 5.2.0 DTD  YJSRE12751_proof  11 June 2014  1:55 am  ce

716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 4 ) 1 e1 0

781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 Q9 827 828 829 830 831 832 833 834 835 836 Q10 837 838 839 840 841 842 843 844 845

7

Fig. 8 e Fluorescence imaging of metastasis to the inguinal lymph node. (A) Inguinal lymph node without fluorescence (solid arrow). Bone lesion (dotted arrow) comprising GFP-expressing cancer cells was detected by fluorescence imaging. (B) Inguinal lymph node with GFP fluorescence. (For interpretation of the references to color in this figure, the reader is referred to the web version of this article.)

completeness of the surgical resection. The percentage of residual tumor was defined as follows: residual tumor ð%Þ ¼ ðpostoperative fluorescent areaÞ= ðpreoperative fluorescent areaÞ  100

2.5.

Postoperative adjuvant therapy

Half of the BLS and FGS mice (8 mice in each group) were randomly selected to undergo 4 wk of adjuvant therapy using ZOL (Novartis Pharmaceuticals). Subcutaneous injection of 120 mg/kg of ZOL or saline (vehicle) was performed for 5 d weekly for 4 wk.

2.6.

Animal imaging

Secondary metastases to lung and inguinal lymph

Eight weeks after surgical treatment, the lungs and inguinal lymph nodes were observed with the OV100 in all the mice to evaluate the effect of FGS or BLS and ZOL on secondary metastases to lung and lymph nodes.

Statistical analysis

Data showing comparisons between the two groups were assessed using the Student t-test or c2 test. Disease-free survival (DFS), which was defined as the time from the surgical resection to local recurrence, was calculated using the KaplaneMeier method and log-rank test. Differences were considered significant when P < 0.05. Data are expressed as mean  standard error of the mean. Statistical analyses were performed with EZR (Saitama Medical Center, Jichi Medical University).

3.

To assess for recurrence and to follow postoperative tumor progression, weekly noninvasive whole-body imaging of the mice was performed and GFP-expressing areas were recorded every week using the iBox Scientia Small Animal Imaging System (UVP, LLC) [27].

2.7. nodes

2.8.

Results

3.1. Fluorescence imaging before and after tumor resection Mouse models of skeletal metastasis of prostate cancer using bright green-fluorescent PC-3 cells were established in 32 mice (Fig. 1B). Two weeks after implantation of PC-3-GFP cells, the mice were randomly divided into BLS and FGS groups. Before the surgical resection, the fluorescent areas of BLS and FGS groups were 15.0  0.93 mm2 and 14.6  0.98 mm2, respectively (Fig. 3). There was no significant difference in preoperative tumor burden between BLS and FGS groups. A significant improvement in visualization of metastatic bone lesions enhanced distinction of tumor from surrounding

5.2.0 DTD  YJSRE12751_proof  11 June 2014  1:55 am  ce

846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910

8

911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 4 ) 1 e1 0

Table 1 e Correlation between the incidence of inguinal lymph node metastases and treatment with FGS and/or ZOL. Q13

ZOL FGS

Yes, %

No, %

P value

6.3 (1/16) 18.8 (3/16)

37.5 (6/16) 25.0 (4/16)

0.033 0.669

normal bone and soft tissues in FGS mice (Fig. 4A). The significantly enhanced distinction during FGS permitted a more complete resection of tumor, resulting in a significant difference in fluorescent area of residual tumor between the mice in the two groups (Fig. 4B). Percentages of residual tumor after BLS and FGS were 9.9  2.2% and 0.9  0.3%, respectively (P < 0.001).

3.2.

Growth of residual tumor after BLS and FGS

The mice with BLS had rapid growth of tumors with GFP expression, whereas the mice with FGS had little growth of the tumors (Fig. 5). Six weeks after surgery, fluorescent areas of BLS and FGS mice were 175.3  35.8 mm2 and 48.6  22.6 mm2, respectively (P ¼ 0.005). Time-course fluorescence imaging demonstrated rapid growth of GFP-expressing areas of BLS mice after BLS (Fig. 6).

3.3.

DFS after FGS and BLS

Six weeks after surgery, DFS of the BLS-only, BLS þ ZOL, FGSonly, and FGS þ ZOL mice were 0, 12.5 (P ¼ 0.554), 25.0 (P ¼ 0.097), and 62.5% (P ¼ 0.004), respectively (Fig. 7A). DFS is defined as the time from surgical resection to local recurrence.

The combination of FGS and ZOL significantly increased DFS over the combination of BLS and ZOL (P ¼ 0.01).

3.4. Influence of ZOL and FGS on inguinal lymph node and lung metastases Six weeks after the surgery, the left inguinal lymph nodes and bilateral lungs of all mice were observed with the OV100. Metastases were detected by GFP expression (Fig. 8). Inguinal lymph node metastases were observed in four mice of BLSonly group, no mice with BLS þ ZOL, two mice in the FGSonly group, and one mouse in the FGS þ ZOL group. Although FGS showed no significant effect on lymph node metastases, ZOL significantly reduced inguinal lymph node metastases (Table 1, P ¼ 0.033). The lung metastases were observed as nodules with GFP expression (Fig. 9A and B). Lung metastases were observed in two mice in the BLS-only group, two mice in the BLS þ ZOL group, three mice in the FGS-only group, and one mouse in the FGS þ ZOL group. No significant effects of ZOL and FGS on lung metastases were found (Table 2).

4.

Discussion

In advanced prostate cancer, 70% of the patients have metastases in bone [28], and 25%e42% of the patients have metastasis in regional lymph nodes [29]. The present study demonstrates that FGS significantly reduced residual tumor after surgery and significantly improved DFS in a mouse model of prostate cancer experimental metastases in the bone. We have previously reported the effectiveness of FGS using tumor-specific fluorescent antibodies [20]. There are several tumor-specific antigens in prostate cancer such as

Fig. 9 e Fluorescence imaging of lung metastases. GFP-expressing cells enabled distinction between normal lung tissue and lung metastases. (A) Lung without metastasis. (B) Lung with metastases. (For interpretation of the references to color in this figure, the reader is referred to the web version of this article.) 5.2.0 DTD  YJSRE12751_proof  11 June 2014  1:55 am  ce

976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 4 ) 1 e1 0

1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105

Table 2 e Correlation between the incidence of lung metastases and treatment with FGS and/or ZOL.

ZOL FGS

Yes, %

No, %

P value

18.8 (3/16) 25.0 (4/16)

31.3 (5/16) 25.0 (4/16)

0.414 1.000

9

writing the article. M.B. and R.M.H. did the critical revision of the article. M.B. and R.M.H. obtained the funding.

Disclosure The authors reported no proprietary or commercial interest in any product mentioned or concept discussed in the article.

prostate-specific antigen [30] that could be targeted with a tumor-specific fluorescent antibody may enable FGS in patients with prostate cancer. FGS did not inhibit either lung or lymph-node metastasis. The results suggest lung and lymph node metastasis are early events of remetastasis from the bone and that FGS should be performed as early as possible on prostate cancer with bone metastasis. Prostate cancer frequently metastasizes to bone. As cancer patients live longer, bone metastasis frequency has increased. Destruction of bone by metastatic disease results initially in microfractures, which can cause intractable pain and eventually a complete fracture of the bone. Surgery is most commonly performed for an impending or existing fracture, or intractable pain. The main advantages of surgery are pain relief and restored function. Although surgery for bone metastasis is usually only a palliative treatment, patients can survive for years after surgery [31]. Patients with a solitary bone metastasis have better survival than patients with multiple bone metastases. Surgery can be considered for solitary bone metastases that are slowgrowing, in patients that have a relatively long survival prognosis. Although surgery is often carried out for metastatic bone disease, there are only a few reports on the outcomes [32]. The opportunity that FGS offers for bone metastasis, as shown in the present report in a clinically-relevant mouse model, should significantly improve outcomes when tested in clinical trials.

Q11

5.

Conclusions

Our study demonstrated that FGS inhibits recurrent tumor growth in a mouse model of prostate cancer metastasis to bone, and that ZOL has inhibitory effect on cancer metastasis to lymph nodes. The combination of FGS and ZOL has potential for patients with bone-metastatic prostate cancer.

Acknowledgment

Q12

This study was supported in part by National Cancer Institute grants CA132971 and CA142669. Dedication: This article is dedicated to the memory of A.R. Moossa, MD. Authors’ contributions: S.M., M.B., and R.M.H. contributed to the conception and design. S.M., Y.M., Y.H., S.Y., F.U., M.Y., Y.Z., H.K., K.H., N.Y., M.B., N.S., H.T., and R.M.H. did the analysis and interpretation. S.M., Y.M., Y.H., S.Y., F.U., M.Y., and Y.Z. did the data collection. S.M., M.B., and R.M.H. did the

references

[1] Hoffman RM. The multiple uses of fluorescent proteins to visualize cancer in vivo. Nat Rev Cancer 2005;5:796. [2] Yang M, Hasegawa S, Jiang P, et al. Widespread skeletal metastatic potential of human lung cancer revealed by green fluorescent protein expression. Cancer Res 1998;58:4217. [3] Yang M, Jiang P, An Z, et al. Genetically fluorescent melanoma bone and organ metastasis models. Clin Cancer Res 1999;5:3549. [4] Yang M, Baranov E, Jiang P, et al. Whole-body optical imaging of green fluorescent protein-expressing tumors and metastases. Proc Natl Acad Sci USA 2000;97:1206. [5] Yang M, Jiang P, Sun FX, et al. A fluorescent orthotopic bone metastasis model of human prostate cancer. Cancer Res 1999;59:781. [6] Burton DW, Geller J, Yang M, et al. Monitoring of skeletal progression of prostate cancer by GFP imaging, X-ray, and serum OPG and PTHrP. The Prostate 2005;62:275. [7] Yang M, Burton DW, Geller J, et al. The bisphosphonate olpadronate inhibits skeletal prostate cancer progression in a green fluorescent protein nude mouse model. Clin Cancer Res 2006;12:2602. [8] Ongkeko WM, Burton D, Kiang A, et al. Parathyroid hormone related-protein promotes epithelial-to-mesenchymal transition in prostate cancer. PLoS One 2014;9:e85803. [9] Bouvet M, Hoffman RM. Glowing tumors make for better detection and resection. Sci Transl Med 2011;3:110fs10. [10] Troyan SL, Kianzad V, Gibbs-Strauss SL, et al. The FLARE intraoperative near-infrared fluorescence imaging system: a first-in-human clinical trial in breast cancer sentinel lymph node mapping. Ann Surg Oncol 2009;16:2943. [11] Stummer W, Pichlmeier U, Meinel T, et al. Fluorescenceguided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol 2006;7:392. [12] van Dam GM, Themelis G, Crane LM, et al. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-a targeting: first in-human results. Nat Med 2011;17:1315. [13] McElroy M, Kaushal S, Luiken GA, et al. Imaging of primary and metastatic pancreatic cancer using a fluorophore conjugated anti-CA19-9 antibody for surgical navigation. World J Surg 2008;32:1057. [14] Metildi CA, Kaushal S, Lee C, et al. An LED light source and novel fluorophore combinations improve fluorescence laparoscopic detection of metastatic pancreatic cancer in orthotopic mouse models. J Am Coll Surg 2012;214:997. [15] Metildi CA, Kaushal S, Hardamon CR, et al. Fluorescenceguided surgery allows for more complete resection of pancreatic cancer, resulting in longer disease-free survival compared with standard surgery in orthotopic mouse models. J Am Coll Surg 2012;215:126. [16] Bouvet M, Metildi CA, Hoffman RM. Reply re: fluorescenceguided surgery allows for more complete resection of pancreatic cancer, resulting in longer disease-free survival

5.2.0 DTD  YJSRE12751_proof  11 June 2014  1:55 am  ce

1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170

10

1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200

[17]

[18]

[19]

[20]

[21]

[22]

[23]

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 4 ) 1 e1 0

compared with standard surgery in orthotopic mouse models. J Am Coll Surg 2012;215:592. Metildi CA, Kaushal S, Snyder CS, et al. Fluorescence-guided surgery of human colon cancer increases complete resection resulting in cures in an orthotopic nude mouse model. J Surg Res 2013;179:87. Metildi CA, Hoffman RM, Bouvet M. Fluorescence-guided surgery and fluorescence laporascopy for gastrointestinal cancers in clinically-relevant mouse models. Gastroenterol Res Pract; 2013. Article ID 290634:8 pages. Metildi C, Kaushal S, Pu M, et al. Fluorescence-guided surgery with a fluorophore-conjugated antibody to carcinoembryonic antigen (CEA) that highlights the tumor, improves surgical resection, and increases survival in orthotopic mouse models of human pancreatic cancer. Ann Surg Oncol; 2014. in press. Metildi CA, Kaushal S, Luiken GA, et al. Fluorescently labeled chimeric anti-CEA antibody improves detection and resection of human colon cancer in a patient-derived orthotopic xenograft (PDOX) nude mouse model. J Surg Oncol; 2013 (in press, Epub ahead of print). Stiles BM, Adusumilli PS, Bhargava A, et al. Minimally invasive localization of oncolytic herpes simplex viral therapy of metastatic pleural cancer. Cancer Gene Ther 2006;13:53. Kishimoto H, Zhao M, Hayashi K, et al. In vivo internal tumor illumination by telomerase-dependent adenoviral GFP for precise surgical navigation. Proc Natl Acad Sci USA 2009;106: 14514. Kishimoto H, Aki R, Urata Y, et al. Tumor-selective, adenoviral-mediated GFP genetic labeling of human cancer

[24]

[25]

[26]

[27]

[28]

[29] [30] [31]

[32]

in the live mouse reports future recurrence after resection. Cell Cycle 2011;10:2737. Urano Y, Sakabe M, Kosaka N, et al. Rapid cancer detection by topically spraying a Y-glutamyltranspeptidaseeactivated, fluorescent probe. Sci Transl Med 2011;3:110ra119. Yamauchi K, Yang M, Jiang P, et al. Development of real-time subcellular dynamic multicolor imaging of cancer-cell trafficking in live mice with a variable-magnification wholemouse imaging system. Cancer Res 2006;66:4208. Hiroshima Y, Maawy A, Sato S, et al. Hand-held highresolution fluorescence imaging system for fluorescenceguided surgery of patient and cell-line pancreatic tumors growing orthotopically in nude mice. J Surg Res; 2013 (in press, Epub ahead of print). Kimura H, Zhang L, Zhao M, et al. Targeted therapy of spinal cord glioma with a genetically-modified Salmonella typhimurium. Cell Prolif 2010;43:41. Rassweiler J, Schulze M, Teber D, et al. Laparoscopic radical prostatectomy with the Heilbronn technique: oncological results in the first 500 patients. J Urol 2005;173:761. Coleman RE. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 2006;12:6243s. Hessels D, Schalken JA. Urinary biomarkers for prostate cancer: a review. Asian J Androl 2013;15:333. Ratasvuori M, Wedin R, Keller J, et al. Insight opinion to surgically treated metastatic bone disease: Scandinavian Sarcoma Group Skeletal Metastasis Registry report of 1195 operated skeletal metastasis. Surg Oncol 2013;22:132. Bickels J, Dadia S, Lidar Z. Surgical management of metastatic bone disease. J Bone Joint Surg Am 2009;91:1503.

5.2.0 DTD  YJSRE12751_proof  11 June 2014  1:55 am  ce

1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230

Fluorescence-guided surgery of prostate cancer bone metastasis.

The aim of this study is to investigate the effectiveness of fluorescence-guided surgery (FGS) of prostate cancer experimental skeletal metastasis...
2MB Sizes 4 Downloads 3 Views