Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.

MUC1 is associated with cellular transformation and tumorigenicity and is considered as an important tumor-associated antigen (TAA) for cancer therapy. We previously reported that anti-MUC1 monoclonal antibody C595 (MAb C595) plus docetaxel (DTX) increased efficacy of DTX alone and caused cultured h...

Full description

Bibliographic Details
Main Authors: Li Wang, Hongmin Chen, Mohammad H Pourgholami, Julia Beretov, Jingli Hao, Hongtu Chao, Alan C Perkins, John H Kearsley, Yong Li
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2011-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3170300?pdf=render
id doaj-347bd2e49d014941885779c10d1cbad2
record_format Article
spelling doaj-347bd2e49d014941885779c10d1cbad22020-11-24T21:26:37ZengPublic Library of Science (PLoS)PLoS ONE1932-62032011-01-0169e2440510.1371/journal.pone.0024405Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.Li WangHongmin ChenMohammad H PourgholamiJulia BeretovJingli HaoHongtu ChaoAlan C PerkinsJohn H KearsleyYong LiMUC1 is associated with cellular transformation and tumorigenicity and is considered as an important tumor-associated antigen (TAA) for cancer therapy. We previously reported that anti-MUC1 monoclonal antibody C595 (MAb C595) plus docetaxel (DTX) increased efficacy of DTX alone and caused cultured human epithelial ovarian cancer (EOC) cells to undergo apoptosis. To further study the mechanisms of this combination-mediated apoptosis, we investigated the effectiveness of this combination therapy in vivo in an intraperitoneal (i.p.) EOC mouse model. OVCAR-3 cells were implanted intraperitoneally in female athymic nude mice and allowed to grow tumor and ascites. Mice were then treated with single MAb C595, DTX, combination test (MAb C595 and DTX), combination control (negative MAb IgG(3) and DTX) or vehicle control i.p. for 3 weeks. Treated mice were killed 4 weeks post-treatment. Ascites volume, tumor weight, CA125 levels from ascites and survival of animals were assessed. The expression of MUC1, CD31, Ki-67, TUNEL and apoptotic proteins in tumor xenografts was evaluated by immunohistochemistry. MAb C595 alone inhibited i.p. tumor growth and ascites production in a dose-dependent manner but did not obviously prevent tumor development. However, combination test significantly reduced ascites volume, tumor growth and metastases, CA125 levels in ascites and improved survival of treated mice compared with single agent-treated mice, combination control or vehicle control-treated mice (P<0.05). The data was in a good agreement with that from cultured cells in vitro. The mechanisms behind the observed effects could be through targeting MUC1 antigens, inhibition of tumor angiogenesis, and induction of apoptosis. Our results suggest that this combination approach can effectively reduce tumor burden and ascites, prolong survival of animals through induction of tumor apoptosis and necrosis, and may provide a potential therapy for advanced metastatic EOC.http://europepmc.org/articles/PMC3170300?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Li Wang
Hongmin Chen
Mohammad H Pourgholami
Julia Beretov
Jingli Hao
Hongtu Chao
Alan C Perkins
John H Kearsley
Yong Li
spellingShingle Li Wang
Hongmin Chen
Mohammad H Pourgholami
Julia Beretov
Jingli Hao
Hongtu Chao
Alan C Perkins
John H Kearsley
Yong Li
Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
PLoS ONE
author_facet Li Wang
Hongmin Chen
Mohammad H Pourgholami
Julia Beretov
Jingli Hao
Hongtu Chao
Alan C Perkins
John H Kearsley
Yong Li
author_sort Li Wang
title Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
title_short Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
title_full Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
title_fullStr Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
title_full_unstemmed Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
title_sort anti-muc1 monoclonal antibody (c595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2011-01-01
description MUC1 is associated with cellular transformation and tumorigenicity and is considered as an important tumor-associated antigen (TAA) for cancer therapy. We previously reported that anti-MUC1 monoclonal antibody C595 (MAb C595) plus docetaxel (DTX) increased efficacy of DTX alone and caused cultured human epithelial ovarian cancer (EOC) cells to undergo apoptosis. To further study the mechanisms of this combination-mediated apoptosis, we investigated the effectiveness of this combination therapy in vivo in an intraperitoneal (i.p.) EOC mouse model. OVCAR-3 cells were implanted intraperitoneally in female athymic nude mice and allowed to grow tumor and ascites. Mice were then treated with single MAb C595, DTX, combination test (MAb C595 and DTX), combination control (negative MAb IgG(3) and DTX) or vehicle control i.p. for 3 weeks. Treated mice were killed 4 weeks post-treatment. Ascites volume, tumor weight, CA125 levels from ascites and survival of animals were assessed. The expression of MUC1, CD31, Ki-67, TUNEL and apoptotic proteins in tumor xenografts was evaluated by immunohistochemistry. MAb C595 alone inhibited i.p. tumor growth and ascites production in a dose-dependent manner but did not obviously prevent tumor development. However, combination test significantly reduced ascites volume, tumor growth and metastases, CA125 levels in ascites and improved survival of treated mice compared with single agent-treated mice, combination control or vehicle control-treated mice (P<0.05). The data was in a good agreement with that from cultured cells in vitro. The mechanisms behind the observed effects could be through targeting MUC1 antigens, inhibition of tumor angiogenesis, and induction of apoptosis. Our results suggest that this combination approach can effectively reduce tumor burden and ascites, prolong survival of animals through induction of tumor apoptosis and necrosis, and may provide a potential therapy for advanced metastatic EOC.
url http://europepmc.org/articles/PMC3170300?pdf=render
work_keys_str_mv AT liwang antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT hongminchen antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT mohammadhpourgholami antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT juliaberetov antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT jinglihao antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT hongtuchao antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT alancperkins antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT johnhkearsley antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
AT yongli antimuc1monoclonalantibodyc595anddocetaxelmarkedlyreducetumorburdenandascitesandprolongsurvivalinaninvivoovariancancermodel
_version_ 1725978569317810176