Ovarian cancer stem cells express ROR1, which can be targeted for anti–cancer-stem-cell therapy

Although initially responsive to chemotherapy, many patients with ovarian cancer subsequently develop relapsed and potentially fatal metastatic disease, which is thought to develop from cancer stem cells (CSCs) that are relatively resistant to conventional therapy. Here, we show that CSCs express a...

Full description

Saved in:
Bibliographic Details
Published inProceedings of the National Academy of Sciences - PNAS Vol. 111; no. 48; pp. 17266 - 17271
Main Authors Zhang, Suping, Cui, Bing, Lai, Hsien, Liu, Grace, Ghia, Emanuela M., Widhopf, George F., Zhang, Zhuhong, Wu, Christina C. N., Chen, Liguang, Wu, Rongrong, Schwab, Richard, Carson, Dennis A., Kipps, Thomas J.
Format Journal Article
LanguageEnglish
Published United States National Academy of Sciences 02.12.2014
National Acad Sciences
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Although initially responsive to chemotherapy, many patients with ovarian cancer subsequently develop relapsed and potentially fatal metastatic disease, which is thought to develop from cancer stem cells (CSCs) that are relatively resistant to conventional therapy. Here, we show that CSCs express a type I receptor tyrosine kinase-like orphan receptor (ROR1), which is expressed during embryogenesis and by many different cancers, but not normal postpartum tissues. Ovarian cancers with high levels of ROR1 had stem cell-like gene-expression signatures. Furthermore, patients with ovarian cancers with high levels of ROR1 had higher rates of relapse and a shorter median survival than patients with ovarian cancers that expressed low-to-negligible amounts of ROR1 . We found that ROR1-positive (ROR1 ⁺) cells isolated from primary tumor-derived xenografts (PDXs) also expressed aldehyde dehydrogenase 1 (ALDH1) and had a greater capacity to form spheroids and to engraft immune-deficient mice than did ROR1-negative (ROR1 ᴺᵉᵍ) ovarian cancer cells isolated from the same tumor population. Treatment with UC-961, an anti-ROR1 mAb, or shRNA silencing of ROR1 inhibited expression of the polycomb ring-finger oncogene, Bmi-1, and other genes associated with the epithelial–mesenchymal transition. Moreover, shRNA silencing of ROR1, depletion of ROR1 ⁺ cells, or treatment with UC-961 impaired the capacity of ovarian cancer cells to form spheroids or tumor xenografts. More importantly, treatment with anti-ROR1 affected the capacity of the xenograft to reseed a virgin mouse, indicating that targeting ROR1 may affect CSC self-renewal. Collectively, these studies indicate that ovarian CSCs express ROR1, which contributes to their capacity to form tumors, making ROR1 a potential target for the therapy of patients with ovarian cancer. Significance This study demonstrates that the oncoembryonic surface antigen, receptor tyrosine kinase-like orphan receptor 1 (ROR1), is expressed on human ovarian cancer stem cells (CSCs), on which it seems to play a functional role in promoting migration/invasion or spheroid formation in vitro and tumor engraftment in immune-deficient mice. Treatment with a humanized mAb specific for ROR1 (UC-961) could inhibit the capacity of ovarian cancer cells to migrate, form spheroids, or engraft immune-deficient mice. Moreover, such treatment inhibited the growth of tumor xenografts, which in turn had a reduced capacity to engraft immune-deficient mice and were relatively depleted of cells with features of CSC, suggesting that treatment with UC-961 could impair CSC renewal. Collectively, these studies indicate that ovarian CSCs express ROR1, which may be targeted for anti-CSC therapy.
Bibliography:http://dx.doi.org/10.1073/pnas.1419599111
ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Author contributions: S.Z. and T.J.K. designed research; S.Z., B.C., H.L., G.L., Z.Z., C.C.N.W., L.C., and R.W. performed research; G.F.W. and R.S. contributed new reagents/analytic tools; S.Z., B.C., H.L., G.L., E.M.G., D.A.C., and T.J.K. analyzed data; and S.Z., H.L., and T.J.K. wrote the paper.
Contributed by Dennis A. Carson, October 22, 2014 (sent for review September 15, 2014; reviewed by Brunhilde H. Felding)
Reviewers included: B.H.F., The Scripps Research Institute.
1S.Z. and B.C. contributed equally to this work.
ISSN:0027-8424
1091-6490
DOI:10.1073/pnas.1419599111