author_facet Zhang, Mo
Kobayashi, Naoko
Zettlitz, Kirstin A.
Kono, Evelyn A.
Yamashiro, Joyce M.
Tsai, Wen-Ting K.
Jiang, Ziyue K.
Tran, Chau P.
Wang, Chung
Guan, Johnny
Wu, Anna M.
Reiter, Robert E.
Zhang, Mo
Kobayashi, Naoko
Zettlitz, Kirstin A.
Kono, Evelyn A.
Yamashiro, Joyce M.
Tsai, Wen-Ting K.
Jiang, Ziyue K.
Tran, Chau P.
Wang, Chung
Guan, Johnny
Wu, Anna M.
Reiter, Robert E.
author Zhang, Mo
Kobayashi, Naoko
Zettlitz, Kirstin A.
Kono, Evelyn A.
Yamashiro, Joyce M.
Tsai, Wen-Ting K.
Jiang, Ziyue K.
Tran, Chau P.
Wang, Chung
Guan, Johnny
Wu, Anna M.
Reiter, Robert E.
spellingShingle Zhang, Mo
Kobayashi, Naoko
Zettlitz, Kirstin A.
Kono, Evelyn A.
Yamashiro, Joyce M.
Tsai, Wen-Ting K.
Jiang, Ziyue K.
Tran, Chau P.
Wang, Chung
Guan, Johnny
Wu, Anna M.
Reiter, Robert E.
Clinical Cancer Research
Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
Cancer Research
Oncology
author_sort zhang, mo
spelling Zhang, Mo Kobayashi, Naoko Zettlitz, Kirstin A. Kono, Evelyn A. Yamashiro, Joyce M. Tsai, Wen-Ting K. Jiang, Ziyue K. Tran, Chau P. Wang, Chung Guan, Johnny Wu, Anna M. Reiter, Robert E. 1078-0432 1557-3265 American Association for Cancer Research (AACR) Cancer Research Oncology http://dx.doi.org/10.1158/1078-0432.ccr-18-1382 <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>The inability to intraoperatively distinguish primary tumor, as well as lymphatic spread, increases the probability of positive surgical margins, tumor recurrence, and surgical toxicity. The goal of this study was to develop a tumor-specific optical probe for real-time fluorescence-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>A humanized antibody fragment against PSCA (A11 minibody, A11 Mb) was conjugated with a near-infrared fluorophore, IRDye800CW. The integrity and binding of the probe to PSCA were confirmed by gel electrophoresis, size-exclusion chromatography, and flow cytometry, respectively. The ability of the probe to detect tumor-infiltrated lymph nodes and metastatic lesions was evaluated in 2 xenograft models, as well as in transgenic mice expressing human PSCA (hPSCA). An invasive intramuscular model was utilized to evaluate the efficacy of the A11 Mb-IRDye800CW-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>A11 Mb was successfully conjugated with IRDye800CW and retained specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 48 hours. The A11 Mb-IRDye800CW specifically detected PSCA-positive primary tumors, tumor-infiltrated lymph nodes, and distant metastases with high contrast. Fluorescence guidance facilitated more complete tumor resection, reduced tumor recurrence, and improved overall survival, compared with conventional white light surgery. The probe successfully identified primary orthotopic tumors and metastatic lesions in hPSCA transgenic mice.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>Real-time fluorescence image–guided surgery with A11 Mb-IRDye800CW enabled detection of lymph node metastases and positive surgical margins, facilitated more complete tumor removal, and improved survival, compared with white light surgery. These results may be translatable into clinical practice to improve surgical and patient outcomes.</jats:p> </jats:sec> Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models Clinical Cancer Research
doi_str_mv 10.1158/1078-0432.ccr-18-1382
facet_avail Online
Free
finc_class_facet Medizin
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xMDc4LTA0MzIuY2NyLTE4LTEzODI
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xMDc4LTA0MzIuY2NyLTE4LTEzODI
institution DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
imprint American Association for Cancer Research (AACR), 2019
imprint_str_mv American Association for Cancer Research (AACR), 2019
issn 1078-0432
1557-3265
issn_str_mv 1078-0432
1557-3265
language English
mega_collection American Association for Cancer Research (AACR) (CrossRef)
match_str zhang2019nearinfrareddyelabeledantiprostatestemcellantigenminibodyenablesrealtimefluorescenceimagingandtargetedsurgeryintranslationalmousemodels
publishDateSort 2019
publisher American Association for Cancer Research (AACR)
recordtype ai
record_format ai
series Clinical Cancer Research
source_id 49
title Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_unstemmed Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_full Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_fullStr Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_full_unstemmed Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_short Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_sort near-infrared dye-labeled anti-prostate stem cell antigen minibody enables real-time fluorescence imaging and targeted surgery in translational mouse models
topic Cancer Research
Oncology
url http://dx.doi.org/10.1158/1078-0432.ccr-18-1382
publishDate 2019
physical 188-200
description <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>The inability to intraoperatively distinguish primary tumor, as well as lymphatic spread, increases the probability of positive surgical margins, tumor recurrence, and surgical toxicity. The goal of this study was to develop a tumor-specific optical probe for real-time fluorescence-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>A humanized antibody fragment against PSCA (A11 minibody, A11 Mb) was conjugated with a near-infrared fluorophore, IRDye800CW. The integrity and binding of the probe to PSCA were confirmed by gel electrophoresis, size-exclusion chromatography, and flow cytometry, respectively. The ability of the probe to detect tumor-infiltrated lymph nodes and metastatic lesions was evaluated in 2 xenograft models, as well as in transgenic mice expressing human PSCA (hPSCA). An invasive intramuscular model was utilized to evaluate the efficacy of the A11 Mb-IRDye800CW-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>A11 Mb was successfully conjugated with IRDye800CW and retained specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 48 hours. The A11 Mb-IRDye800CW specifically detected PSCA-positive primary tumors, tumor-infiltrated lymph nodes, and distant metastases with high contrast. Fluorescence guidance facilitated more complete tumor resection, reduced tumor recurrence, and improved overall survival, compared with conventional white light surgery. The probe successfully identified primary orthotopic tumors and metastatic lesions in hPSCA transgenic mice.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>Real-time fluorescence image–guided surgery with A11 Mb-IRDye800CW enabled detection of lymph node metastases and positive surgical margins, facilitated more complete tumor removal, and improved survival, compared with white light surgery. These results may be translatable into clinical practice to improve surgical and patient outcomes.</jats:p> </jats:sec>
container_issue 1
container_start_page 188
container_title Clinical Cancer Research
container_volume 25
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792348127511969796
geogr_code not assigned
last_indexed 2024-03-01T18:06:03.493Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Near-Infrared+Dye-Labeled+Anti-Prostate+Stem+Cell+Antigen+Minibody+Enables+Real-Time+Fluorescence+Imaging+and+Targeted+Surgery+in+Translational+Mouse+Models&rft.date=2019-01-01&genre=article&issn=1557-3265&volume=25&issue=1&spage=188&epage=200&pages=188-200&jtitle=Clinical+Cancer+Research&atitle=Near-Infrared+Dye-Labeled+Anti-Prostate+Stem+Cell+Antigen+Minibody+Enables+Real-Time+Fluorescence+Imaging+and+Targeted+Surgery+in+Translational+Mouse+Models&aulast=Reiter&aufirst=Robert+E.&rft_id=info%3Adoi%2F10.1158%2F1078-0432.ccr-18-1382&rft.language%5B0%5D=eng
SOLR
_version_ 1792348127511969796
author Zhang, Mo, Kobayashi, Naoko, Zettlitz, Kirstin A., Kono, Evelyn A., Yamashiro, Joyce M., Tsai, Wen-Ting K., Jiang, Ziyue K., Tran, Chau P., Wang, Chung, Guan, Johnny, Wu, Anna M., Reiter, Robert E.
author_facet Zhang, Mo, Kobayashi, Naoko, Zettlitz, Kirstin A., Kono, Evelyn A., Yamashiro, Joyce M., Tsai, Wen-Ting K., Jiang, Ziyue K., Tran, Chau P., Wang, Chung, Guan, Johnny, Wu, Anna M., Reiter, Robert E., Zhang, Mo, Kobayashi, Naoko, Zettlitz, Kirstin A., Kono, Evelyn A., Yamashiro, Joyce M., Tsai, Wen-Ting K., Jiang, Ziyue K., Tran, Chau P., Wang, Chung, Guan, Johnny, Wu, Anna M., Reiter, Robert E.
author_sort zhang, mo
container_issue 1
container_start_page 188
container_title Clinical Cancer Research
container_volume 25
description <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>The inability to intraoperatively distinguish primary tumor, as well as lymphatic spread, increases the probability of positive surgical margins, tumor recurrence, and surgical toxicity. The goal of this study was to develop a tumor-specific optical probe for real-time fluorescence-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>A humanized antibody fragment against PSCA (A11 minibody, A11 Mb) was conjugated with a near-infrared fluorophore, IRDye800CW. The integrity and binding of the probe to PSCA were confirmed by gel electrophoresis, size-exclusion chromatography, and flow cytometry, respectively. The ability of the probe to detect tumor-infiltrated lymph nodes and metastatic lesions was evaluated in 2 xenograft models, as well as in transgenic mice expressing human PSCA (hPSCA). An invasive intramuscular model was utilized to evaluate the efficacy of the A11 Mb-IRDye800CW-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>A11 Mb was successfully conjugated with IRDye800CW and retained specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 48 hours. The A11 Mb-IRDye800CW specifically detected PSCA-positive primary tumors, tumor-infiltrated lymph nodes, and distant metastases with high contrast. Fluorescence guidance facilitated more complete tumor resection, reduced tumor recurrence, and improved overall survival, compared with conventional white light surgery. The probe successfully identified primary orthotopic tumors and metastatic lesions in hPSCA transgenic mice.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>Real-time fluorescence image–guided surgery with A11 Mb-IRDye800CW enabled detection of lymph node metastases and positive surgical margins, facilitated more complete tumor removal, and improved survival, compared with white light surgery. These results may be translatable into clinical practice to improve surgical and patient outcomes.</jats:p> </jats:sec>
doi_str_mv 10.1158/1078-0432.ccr-18-1382
facet_avail Online, Free
finc_class_facet Medizin
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xMDc4LTA0MzIuY2NyLTE4LTEzODI
imprint American Association for Cancer Research (AACR), 2019
imprint_str_mv American Association for Cancer Research (AACR), 2019
institution DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229, DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161
issn 1078-0432, 1557-3265
issn_str_mv 1078-0432, 1557-3265
language English
last_indexed 2024-03-01T18:06:03.493Z
match_str zhang2019nearinfrareddyelabeledantiprostatestemcellantigenminibodyenablesrealtimefluorescenceimagingandtargetedsurgeryintranslationalmousemodels
mega_collection American Association for Cancer Research (AACR) (CrossRef)
physical 188-200
publishDate 2019
publishDateSort 2019
publisher American Association for Cancer Research (AACR)
record_format ai
recordtype ai
series Clinical Cancer Research
source_id 49
spelling Zhang, Mo Kobayashi, Naoko Zettlitz, Kirstin A. Kono, Evelyn A. Yamashiro, Joyce M. Tsai, Wen-Ting K. Jiang, Ziyue K. Tran, Chau P. Wang, Chung Guan, Johnny Wu, Anna M. Reiter, Robert E. 1078-0432 1557-3265 American Association for Cancer Research (AACR) Cancer Research Oncology http://dx.doi.org/10.1158/1078-0432.ccr-18-1382 <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>The inability to intraoperatively distinguish primary tumor, as well as lymphatic spread, increases the probability of positive surgical margins, tumor recurrence, and surgical toxicity. The goal of this study was to develop a tumor-specific optical probe for real-time fluorescence-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>A humanized antibody fragment against PSCA (A11 minibody, A11 Mb) was conjugated with a near-infrared fluorophore, IRDye800CW. The integrity and binding of the probe to PSCA were confirmed by gel electrophoresis, size-exclusion chromatography, and flow cytometry, respectively. The ability of the probe to detect tumor-infiltrated lymph nodes and metastatic lesions was evaluated in 2 xenograft models, as well as in transgenic mice expressing human PSCA (hPSCA). An invasive intramuscular model was utilized to evaluate the efficacy of the A11 Mb-IRDye800CW-guided surgery.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>A11 Mb was successfully conjugated with IRDye800CW and retained specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 48 hours. The A11 Mb-IRDye800CW specifically detected PSCA-positive primary tumors, tumor-infiltrated lymph nodes, and distant metastases with high contrast. Fluorescence guidance facilitated more complete tumor resection, reduced tumor recurrence, and improved overall survival, compared with conventional white light surgery. The probe successfully identified primary orthotopic tumors and metastatic lesions in hPSCA transgenic mice.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>Real-time fluorescence image–guided surgery with A11 Mb-IRDye800CW enabled detection of lymph node metastases and positive surgical margins, facilitated more complete tumor removal, and improved survival, compared with white light surgery. These results may be translatable into clinical practice to improve surgical and patient outcomes.</jats:p> </jats:sec> Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models Clinical Cancer Research
spellingShingle Zhang, Mo, Kobayashi, Naoko, Zettlitz, Kirstin A., Kono, Evelyn A., Yamashiro, Joyce M., Tsai, Wen-Ting K., Jiang, Ziyue K., Tran, Chau P., Wang, Chung, Guan, Johnny, Wu, Anna M., Reiter, Robert E., Clinical Cancer Research, Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models, Cancer Research, Oncology
title Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_full Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_fullStr Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_full_unstemmed Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_short Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
title_sort near-infrared dye-labeled anti-prostate stem cell antigen minibody enables real-time fluorescence imaging and targeted surgery in translational mouse models
title_unstemmed Near-Infrared Dye-Labeled Anti-Prostate Stem Cell Antigen Minibody Enables Real-Time Fluorescence Imaging and Targeted Surgery in Translational Mouse Models
topic Cancer Research, Oncology
url http://dx.doi.org/10.1158/1078-0432.ccr-18-1382