E. Belloni, M. Muenke, E. Roessler, G. Traverso, J. Siegel-bartelt et al., Identification of Sonic hedgehog as a candidate gene responsible for holoprosencephaly, Nat Genet, vol.14, issue.3, pp.353-359, 1996.

J. P. Incardona, W. Gaffield, R. P. Kapur, and H. Roelink, The teratogenic Veratrum alkaloid cyclopamine inhibits sonic hedgehog signal transduction, Dev Camb Engl. sept, vol.125, issue.18, pp.3553-62, 1998.

A. Joutel, C. Corpechot, A. Ducros, K. Vahedi, H. Chabriat et al., Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia, Nature, vol.383, issue.6602, pp.707-717, 1996.

L. Li, I. D. Krantz, Y. Deng, A. Genin, A. B. Banta et al., Alagille syndrome is caused by mutations in human Jagged1, which encodes a ligand for Notch1, Nat Genet. juill, vol.16, issue.3, pp.243-51, 1997.

T. Oda, A. G. Elkahloun, B. L. Pike, K. Okajima, I. D. Krantz et al., Mutations in the human Jagged1 gene are responsible for Alagille syndrome, Nat Genet. juill, vol.16, issue.3, pp.235-277, 1997.

C. Raffel, R. B. Jenkins, L. Frederick, D. Hebrink, B. Alderete et al., Sporadic medulloblastomas contain PTCH mutations, Cancer Res. 1 mars, vol.57, issue.5, pp.842-847, 1997.

J. Xie, M. Murone, S. M. Luoh, A. Ryan, Q. Gu et al., Activating Smoothened mutations in sporadic basal-cell carcinoma, Nature. 1 janv, vol.391, issue.6662, pp.90-92, 1998.

U. Tostar, C. J. Malm, J. M. Meis-kindblom, L. Kindblom, R. Toftgård et al., Deregulation of the hedgehog signalling pathway: a possible role for the PTCH and SUFU genes in human rhabdomyoma and rhabdomyosarcoma development, J Pathol. janv, vol.208, issue.1, pp.17-25, 2006.

L. L. Rubin and F. J. De-sauvage, Targeting the Hedgehog pathway in cancer, Nat Rev Drug Discov. déc, vol.5, issue.12, pp.1026-1059, 2006.

L. W. Ellisen, J. Bird, D. C. West, A. L. Soreng, T. C. Reynolds et al., TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms, Cell. 23 août, vol.66, issue.4, pp.649-61, 1991.

P. Rizzo, C. Osipo, K. Foreman, T. Golde, B. Osborne et al., Rational targeting of Notch signaling in cancer, Oncogene. 1 sept, vol.27, issue.38, pp.5124-5155, 2008.

N. Stransky, A. M. Egloff, A. D. Tward, A. D. Kostic, K. Cibulskis et al., The mutational landscape of head and neck squamous cell carcinoma, Science. 26 août, vol.333, issue.6046, pp.1157-60, 2011.

V. Hoff, D. D. Lorusso, P. M. Rudin, C. M. Reddy, J. C. Yauch et al., Inhibition of the hedgehog pathway in advanced basal-cell carcinoma, N Engl J Med. 17 sept, vol.361, issue.12, pp.1164-72, 2009.

A. Sekulic, M. R. Migden, A. E. Oro, L. Dirix, K. D. Lewis et al., Efficacy and Safety of Vismodegib in Advanced Basal-Cell Carcinoma, N Engl J Med. 7 juin, vol.366, issue.23, pp.2171-2180, 2012.

C. B. Burness, Sonidegib: First Global Approval, Drugs. sept, vol.75, issue.13, pp.1559-66, 2015.

J. Goldman, S. G. Eckhardt, M. J. Borad, K. K. Curtis, M. Hidalgo et al., Phase I dose-escalation trial of the oral investigational Hedgehog signaling pathway inhibitor TAK-441 in patients with advanced solid tumors, Clin Cancer Res Off J Am Assoc Cancer Res. 1 mars, vol.21, issue.5, pp.1002-1011, 2015.

S. Peukert, F. He, M. Dai, R. Zhang, Y. Sun et al., Discovery of NVP-LEQ506, a second-generation inhibitor of smoothened, ChemMedChem. août, vol.8, issue.8, pp.1261-1266, 2013.

D. J. Kim, J. Kim, K. Spaunhurst, J. Montoya, R. Khodosh et al., Open-label, exploratory phase II trial of oral itraconazole for the treatment of basal cell carcinoma, J Clin Oncol Off J Am Soc Clin Oncol. 10 mars, vol.32, issue.8, pp.745-51, 2014.

M. R. Savona, D. A. Pollyea, W. Stock, V. G. Oehler, M. A. Schroeder et al., Phase Ib Study of Glasdegib, a Hedgehog Pathway Inhibitor, in Combination with Standard Chemotherapy in Patients with AML or High-Risk MDS, Clin Cancer Res Off J Am Assoc Cancer Res. 15 mai, vol.24, issue.10, pp.2294-303, 2018.

A. J. Wagner, W. A. Messersmith, M. N. Shaik, S. Li, X. Zheng et al., A phase I study of PF-04449913, an oral hedgehog inhibitor, in patients with advanced solid tumors, Clin Cancer Res Off J Am Assoc Cancer Res, vol.01, issue.5, pp.1044-51, 2015.

G. Martinelli, V. G. Oehler, C. Papayannidis, R. Courtney, M. N. Shaik et al., Treatment with PF-04449913, an oral smoothened antagonist, in patients with myeloid malignancies: a phase 1 safety and pharmacokinetics study, Lancet Haematol. août, vol.2, issue.8, pp.339-346, 2015.

, Abstract 2819: Identification and characterization of a novel smoothened antagonist for the treatment of cancer with deregulated hedgehog signaling | Cancer Research

K. Sasaki, J. R. Gotlib, R. A. Mesa, K. J. Newberry, F. Ravandi et al., Phase II evaluation of IPI-926, an oral Hedgehog inhibitor, in patients with myelofibrosis, Leuk Lymphoma. juill, vol.56, issue.7, pp.2092-2099, 2015.

A. Jimeno, G. J. Weiss, W. H. Miller, S. Gettinger, B. Eigl et al., Phase I study of the Hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors, Clin Cancer Res Off J Am Assoc Cancer Res. 15 mai, vol.19, issue.10, pp.2766-74, 2013.

A. H. Ko, N. Loconte, M. A. Tempero, E. J. Walker, K. Kelley et al., A Phase I Study of FOLFIRINOX Plus IPI-926, a Hedgehog Pathway Inhibitor, for Advanced Pancreatic Adenocarcinoma, Pancreas. mars, vol.45, issue.3, pp.370-375, 2016.

D. W. Bowles, S. B. Keysar, J. R. Eagles, G. Wang, M. J. Glogowska et al., A pilot study of cetuximab and the hedgehog inhibitor IPI-926 in recurrent/metastatic head and neck squamous cell carcinoma, Oral Oncol. févr, vol.53, pp.74-83, 2016.

A. Abstract, A first-in-human, phase I study of an oral hedgehog pathway antagonist, BMS-833923 (XL139), in subjects with advanced or metastatic solid tumors | Molecular Cancer Therapeutics

D. Sur,

N. P. Shah, J. E. Cortes, G. Martinelli, B. D. Smith, C. E. Copland et al., Dasatinib Plus Smoothened (SMO) Inhibitor BMS-833923 in Chronic Myeloid Leukemia (CML) with Resistance or Suboptimal Response to a Prior Tyrosine Kinase Inhibitor (TKI): Phase I Study CA180323, Blood. 6 déc, vol.124, issue.21, pp.4539-4539, 2014.

T. Brinkhuizen, K. Frencken, P. J. Nelemans, M. Hoff, N. Kelleners-smeets et al., The effect of topical diclofenac 3% and calcitriol 3 ?g/g on superficial basal cell carcinoma (sBCC) and nodular basal cell carcinoma (nBCC): A phase II, randomized controlled trial, J Am Acad Dermatol. juill, vol.75, issue.1, pp.126-160, 2016.

C. M. Rudin, C. L. Hann, J. Laterra, R. L. Yauch, C. A. Callahan et al., Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449, N Engl J Med. 17 sept, vol.361, issue.12, pp.1173-1181, 2009.

E. Peer, S. Tesanovic, and F. Aberger, Next-Generation Hedgehog/GLI Pathway Inhibitors for Cancer Therapy, Cancers. 15 avr, vol.11, issue.4, 2019.

S. X. Atwood, K. Y. Sarin, R. J. Whitson, J. R. Li, G. Kim et al., Smoothened variants explain the majority of drug resistance in basal cell carcinoma, Cancer Cell. 9 mars, vol.27, issue.3, pp.342-53, 2015.

S. Buonamici, J. Williams, M. Morrissey, A. Wang, R. Guo et al., Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma, Sci Transl Med. 29 sept, vol.2, issue.51, pp.51-70, 2010.

A. W. Tolcher, W. A. Messersmith, S. M. Mikulski, K. P. Papadopoulos, E. L. Kwak et al., Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors, J Clin Oncol Off J Am Soc Clin Oncol. 1 juill, vol.30, pp.2348-53, 2012.

I. Diaz-padilla, M. K. Wilson, B. A. Clarke, H. W. Hirte, S. A. Welch et al., A phase II study of single-agent RO4929097, a gamma-secretase inhibitor of Notch signaling, in patients with recurrent platinum-resistant epithelial ovarian cancer: A study of the Princess Margaret, Chicago and California phase II consortia, Gynecol Oncol. mai, vol.137, issue.2, pp.216-238, 2015.

S. M. Lee, J. Moon, B. G. Redman, T. Chidiac, L. E. Flaherty et al., Phase 2 study of RO4929097, a gammasecretase inhibitor, in metastatic melanoma: SWOG 0933, Cancer. 1 févr, vol.121, issue.3, pp.432-472, 2015.

E. Pan, J. G. Supko, T. J. Kaley, N. A. Butowski, T. Cloughesy et al., Phase I study of RO4929097 with bevacizumab in patients with recurrent malignant glioma, J Neurooncol, vol.130, issue.3, pp.571-580, 2016.

M. A. Locatelli, P. Aftimos, E. C. Dees, P. M. Lorusso, M. D. Pegram et al., Phase I study of the gamma secretase inhibitor PF-03084014 in combination with docetaxel in patients with advanced triple-negative breast cancer, Oncotarget. 10 janv, vol.8, issue.2, pp.2320-2328, 2017.

I. Krop, T. Demuth, T. Guthrie, P. Y. Wen, W. P. Mason et al., Phase I pharmacologic and pharmacodynamic study of the gamma secretase (Notch) inhibitor MK-0752 in adult patients with advanced solid tumors, J Clin Oncol Off J Am Soc Clin Oncol. 1 juill, vol.30, pp.2307-2320, 2012.

D. C. Smith, P. D. Eisenberg, G. Manikhas, R. Chugh, M. A. Gubens et al., A phase I dose escalation and expansion study of the anticancer stem cell agent demcizumab (anti-DLL4) in patients with previously treated solid tumors, Clin Cancer Res Off J Am Assoc Cancer Res. 15 déc, vol.20, issue.24, pp.6295-303, 2014.

E. G. Chiorean, P. Lorusso, R. M. Strother, J. R. Diamond, A. Younger et al., A Phase I First-in-Human Study of Enoticumab (REGN421), a Fully Human Delta-like Ligand 4 (Dll4) Monoclonal Antibody in Patients with Advanced Solid Tumors, Clin Cancer Res Off J Am Assoc Cancer Res. 15 juin, vol.21, issue.12, pp.2695-703, 2015.

R. P. Sharma, Wingless a new mutant in Drosophila melanogaster, Drosoph Inf Serv, vol.50, pp.134-134, 1973.

S. Angers and R. T. Moon, Proximal events in Wnt signal transduction, Nat Rev Mol Cell Biol. juill, vol.10, issue.7, pp.468-77, 2009.

R. Nusse and H. E. Varmus, Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome, Cell. nov, vol.31, issue.1, pp.99-109, 1982.

F. Rijsewijk, M. Schuermann, E. Wagenaar, P. Parren, D. Weigel et al., The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless, Cell. 14 août, vol.50, issue.4, pp.649-57, 1987.

B. Vogelstein, N. Papadopoulos, V. E. Velculescu, S. Zhou, L. A. Diaz et al., Cancer genome landscapes, Science. 29 mars, vol.339, issue.6127, pp.1546-58, 2013.

E. D. Pleasance, R. K. Cheetham, P. J. Stephens, D. J. Mcbride, S. J. Humphray et al., A comprehensive catalogue of somatic mutations from a human cancer genome, Nature. 14 janv, vol.463, issue.7278, pp.191-197, 2010.

R. P. Dahmen, A. Koch, D. Denkhaus, J. C. Tonn, N. Sörensen et al., Deletions of AXIN1, a component of the WNT/wingless pathway, in sporadic medulloblastomas, Cancer Res, vol.61, pp.7039-7082, 2001.

S. Satoh, Y. Daigo, Y. Furukawa, T. Kato, N. Miwa et al., AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1, Nat Genet. mars, vol.24, issue.3, pp.245-50, 2000.

W. M. Clements, J. Wang, A. Sarnaik, O. J. Kim, J. Macdonald et al., beta-Catenin mutation is a frequent cause of Wnt pathway activation in gastric cancer, Cancer Res. 15 juin, vol.62, issue.12, pp.3503-3509, 2002.

S. Segditsas and I. Tomlinson, Colorectal cancer and genetic alterations in the Wnt pathway, Oncogene. 4 déc, vol.25, issue.57, pp.7531-7538, 2006.

J. N. Anastas and R. T. Moon, WNT signalling pathways as therapeutic targets in cancer, Nat Rev Cancer. janv, vol.13, issue.1, pp.11-26, 2013.

M. Sebbagh and J. Borg, Insight into planar cell polarity, Exp Cell Res, vol.328, issue.2, pp.284-95, 2014.

D. W. Chan, C. Chan, J. Yam, Y. Ching, and I. Ng, Prickle-1 negatively regulates Wnt/beta-catenin pathway by promoting Dishevelled ubiquitination/degradation in liver cancer, Gastroenterology. oct, vol.131, issue.4, pp.1218-1245, 2006.

P. A. Lawrence and P. M. Shelton, The determination of polarity in the developing insect retina, J Embryol Exp Morphol. avr, vol.33, issue.2, pp.471-86, 1975.

Z. Kibar, K. J. Vogan, N. Groulx, M. J. Justice, D. A. Underhill et al., Ltap, a mammalian homolog of Drosophila Strabismus/Van Gogh, is altered in the mouse neural tube mutant Loop-tail, Nat Genet. juill, vol.28, issue.3, pp.251-256, 2001.

M. Park and R. T. Moon, The planar cell-polarity gene stbm regulates cell behaviour and cell fate in vertebrate embryos, Nat Cell Biol. janv, vol.4, issue.1, pp.20-25, 2002.

J. N. Murdoch, K. Doudney, C. Paternotte, A. J. Copp, and P. Stanier, Severe neural tube defects in the loop-tail mouse result from mutation of Lpp1, a novel gene involved in floor plate specification, Hum Mol Genet, vol.10, issue.22, pp.2593-601, 2001.

Y. Lei, T. Zhang, H. Li, B. Wu, J. L. Wang et al., VANGL2 mutations in human cranial neural-tube defects, N Engl J Med. 10 juin, vol.362, issue.23, pp.2232-2237, 2010.

Z. Kibar, E. Torban, J. R. Mcdearmid, A. Reynolds, J. Berghout et al., Mutations in VANGL1 associated with neural-tube defects, N Engl J Med. 5 avr, vol.356, issue.14, pp.1432-1439, 2007.

J. A. Zallen, Planar polarity and tissue morphogenesis, Cell. 15 juin, vol.129, issue.6, pp.1051-63, 2007.

Y. Yang and M. Mlodzik, Wnt-Frizzled/planar cell polarity signaling: cellular orientation by facing the wind (Wnt), Annu Rev Cell Dev Biol, vol.31, pp.623-669, 2015.

S. Lejeune, E. L. Huguet, A. Hamby, R. Poulsom, and A. L. Harris, Wnt5a cloning, expression, and up-regulation in human primary breast cancers, Clin Cancer Res Off J Am Assoc Cancer Res. févr, vol.1, issue.2, pp.215-237, 1995.

C. D. Macmillan, H. S. Leong, D. W. Dales, A. E. Robertson, J. D. Lewis et al., Stage of breast cancer progression influences cellular response to activation of the WNT/planar cell polarity pathway, Sci Rep. 10 sept, vol.4, p.6315, 2014.

V. Luga, L. Zhang, A. M. Viloria-petit, A. A. Ogunjimi, M. R. Inanlou et al., Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration, Cell. 21 déc, vol.151, issue.7, pp.1542-56, 2012.

A. Avgustinova, M. Iravani, D. Robertson, A. Fearns, Q. Gao et al., Tumour cell-derived Wnt7a recruits and activates fibroblasts to promote tumour aggressiveness, Nat Commun. 18 janv, vol.7, p.10305, 2016.

L. Yang, X. Wu, Y. Wang, K. Zhang, J. Wu et al., FZD7 has a critical role in cell proliferation in triple negative breast cancer, Oncogene, vol.30, issue.43, pp.4437-4483, 2011.

J. N. Anastas, T. L. Biechele, M. Robitaille, J. Muster, K. H. Allison et al., A protein complex of SCRIB, NOS1AP and VANGL1 regulates cell polarity and migration, and is associated with breast cancer progression, Oncogene. 9 août, vol.31, issue.32, pp.3696-708, 2012.

T. M. Puvirajesinghe, F. Bertucci, A. Jain, P. Scerbo, E. Belotti et al., Identification of p62/SQSTM1 as a component of non-canonical Wnt VANGL2-JNK signalling in breast cancer, Nat Commun. 12 janv, vol.7, p.10318, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01447570

A. M. Daulat, F. Bertucci, S. Audebert, A. Sergé, P. Finetti et al., PRICKLE1 Contributes to Cancer Cell Dissemination through Its Interaction with mTORC2, Dev Cell. 23 mai, vol.37, issue.4, pp.311-336, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01321061

B. Ataseven, R. Angerer, R. Kates, A. Gunesch, P. Knyazev et al., PTK7 expression in triple-negative breast cancer, Anticancer Res. sept, vol.33, issue.9, pp.3759-63, 2013.

C. Henry, A. Quadir, N. J. Hawkins, E. Jary, E. Llamosas et al., Expression of the novel Wnt receptor ROR2 is increased in breast cancer and may regulate both ?-catenin dependent and independent Wnt signalling, J Cancer Res Clin Oncol. févr, vol.141, issue.2, pp.243-54, 2015.

M. Asad, M. K. Wong, T. Z. Tan, M. Choolani, J. Low et al., FZD7 drives in vitro aggressiveness in Stem-A subtype of ovarian cancer via regulation of non-canonical Wnt/PCP pathway, Cell Death Dis. 17 juill, vol.5, p.1346, 2014.

M. Taki, N. Kamata, K. Yokoyama, R. Fujimoto, S. Tsutsumi et al., Down-regulation of Wnt-4 and upregulation of Wnt-5a expression by epithelial-mesenchymal transition in human squamous carcinoma cells, Cancer Sci. juill, vol.94, issue.7, pp.593-600, 2003.

H. Qin, X. Liao, Y. Chen, S. Huang, W. Xue et al., Genomic Characterization of Esophageal Squamous Cell Carcinoma Reveals Critical Genes Underlying Tumorigenesis and Poor Prognosis, Am J Hum Genet. 7 avr, vol.98, issue.4, pp.709-736, 2016.

C. Dyberg, P. Papachristou, B. H. Haug, H. Lagercrantz, P. Kogner et al., Planar cell polarity gene expression correlates with tumor cell viability and prognostic outcome in neuroblastoma, BMC Cancer. 31 mars, vol.16, p.259, 2016.

S. K. Dissanayake, M. Wade, C. E. Johnson, M. P. O'connell, P. D. Leotlela et al., The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition, J Biol Chem. 8 juin, vol.282, issue.23, pp.17259-71, 2007.

J. N. Anastas, R. M. Kulikauskas, T. Tamir, H. Rizos, G. V. Long et al., WNT5A enhances resistance of melanoma cells to targeted BRAF inhibitors, J Clin Invest. juill, vol.124, issue.7, pp.2877-90, 2014.

D. Forno, P. D. Pringle, J. H. Hutchinson, P. Osborn, J. Huang et al., WNT5A expression increases during melanoma progression and correlates with outcome, Clin Cancer Res Off J Am Assoc Cancer Res. 15 sept, vol.14, issue.18, pp.5825-5857, 2008.

A. T. Weeraratna, Y. Jiang, G. Hostetter, K. Rosenblatt, P. Duray et al., Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma, Cancer Cell. avr, vol.1, issue.3, pp.279-88, 2002.

S. Tiwary and L. Xu, FRIZZLED7 Is Required for Tumor Initiation and Metastatic Growth of Melanoma Cells, PloS One, vol.11, issue.1, p.147638, 2016.

M. P. O'connell, J. L. Fiori, M. Xu, A. D. Carter, B. P. Frank et al., The orphan tyrosine kinase receptor, ROR2, mediates Wnt5A signaling in metastatic melanoma, Oncogene. 7 janv, vol.29, issue.1, pp.34-44, 2010.

M. P. O'connell, K. Marchbank, M. R. Webster, A. A. Valiga, A. Kaur et al., Hypoxia induces phenotypic plasticity and therapy resistance in melanoma via the tyrosine kinase receptors ROR1 and ROR2, Cancer Discov. déc, vol.3, issue.12, pp.1378-93, 2013.

A. Lhoumeau, S. Martinez, J. Boher, G. Monges, R. Castellano et al., Overexpression of the Promigratory and Prometastatic PTK7 Receptor Is Associated with an Adverse Clinical Outcome in Colorectal Cancer, PloS One, vol.10, issue.5, p.123768, 2015.

H. Mei, S. Lian, S. Zhang, W. Wang, Q. Mao et al., High expression of ROR2 in cancer cell correlates with unfavorable prognosis in colorectal cancer, Biochem Biophys Res Commun, vol.453, issue.4, pp.703-712, 2014.

R. Cheng, B. Sun, Z. Liu, X. Zhao, L. Qi et al., Wnt5a suppresses colon cancer by inhibiting cell proliferation and epithelial-mesenchymal transition, J Cell Physiol. déc, vol.229, issue.12, pp.1908-1925, 2014.

E. Bakker, A. M. Das, W. Helvensteijn, P. F. Franken, S. Swagemakers et al., Wnt5a promotes human colon cancer cell migration and invasion but does not augment intestinal tumorigenesis in Apc1638N mice, Carcinogenesis. nov, vol.34, issue.11, pp.2629-2667, 2013.

M. Kurayoshi, N. Oue, H. Yamamoto, M. Kishida, A. Inoue et al., Expression of Wnt-5a is correlated with aggressiveness of gastric cancer by stimulating cell migration and invasion, Cancer Res, vol.66, issue.21, pp.10439-10487, 2006.

D. T. Miyamoto, Y. Zheng, B. S. Wittner, R. J. Lee, H. Zhu et al., RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance, Science. 18 sept, vol.349, issue.6254, pp.1351-1357, 2015.

N. Kremenevskaja, R. Von-wasielewski, A. S. Rao, C. Schöfl, T. Andersson et al., Wnt-5a has tumor suppressor activity in thyroid carcinoma, Oncogene. 24 mars, vol.24, issue.13, pp.2144-54, 2005.

M. Kaucká, K. Plevová, S. Pavlová, P. Janovská, A. Mishra et al., The planar cell polarity pathway drives pathogenesis of chronic lymphocytic leukemia by the regulation of B-lymphocyte migration, Cancer Res. 1 mars, vol.73, issue.5, pp.1491-501, 2013.

P. Janovska, L. Poppova, K. Plevova, H. Plesingerova, M. Behal et al., Autocrine Signaling by Wnt-5a

, Deregulates Chemotaxis of Leukemic Cells and Predicts Clinical Outcome in Chronic Lymphocytic Leukemia, Clin Cancer Res Off J Am Assoc Cancer Res. 15 janv, vol.22, issue.2, pp.459-69, 2016.

J. Yu, L. Chen, B. Cui, G. F. Widhopf, Z. Shen et al., Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation, J Clin Invest. févr, vol.126, issue.2, pp.585-98, 2016.

T. Prebet, A. Lhoumeau, C. Arnoulet, A. Aulas, S. Marchetto et al., The cell polarity PTK7 receptor acts as a modulator of the chemotherapeutic response in acute myeloid leukemia and impairs clinical outcome. Blood. 30 sept, vol.116, pp.2315-2338, 2010.

G. Jiang, M. Zhang, B. Yue, M. Yang, C. Carter et al., PTK7: a new biomarker for immunophenotypic characterization of maturing T cells and T cell acute lymphoblastic leukemia, Leuk Res, vol.36, issue.11, pp.1347-53, 2012.

P. Le, J. D. Mcdermott, and A. Jimeno, Targeting the Wnt pathway in human cancers: therapeutic targeting with a focus on OMP-54F28, Pharmacol Ther. févr, vol.0, pp.1-11, 2015.

M. M. Fischer, B. Cancilla, V. P. Yeung, F. Cattaruzza, C. Chartier et al., WNT antagonists exhibit unique combinatorial antitumor activity with taxanes by potentiating mitotic cell death, Sci Adv, vol.3, issue.6, p.1700090, 2017.

C. Fukukawa, H. Hanaoka, S. Nagayama, T. Tsunoda, J. Toguchida et al., Radioimmunotherapy of human synovial sarcoma using a monoclonal antibody against FZD10, Cancer Sci. févr, vol.99, issue.2, pp.432-472, 2008.

H. Lenz and M. Kahn, Safely targeting cancer stem cells via selective catenin coactivator antagonism, Cancer Sci. sept, vol.105, issue.9, pp.1087-92, 2014.

S. Pozzi, M. Fulciniti, H. Yan, S. Vallet, H. Eda et al., In vivo and in vitro effects of a novel anti-Dkk1 neutralizing antibody in multiple myeloma, Bone. avr, vol.53, issue.2, pp.487-96, 2013.

G. Canesin, S. Evans-axelsson, R. Hellsten, A. Krzyzanowska, C. P. Prasad et al., Treatment with the WNT5A-mimicking peptide Foxy-5 effectively reduces the metastatic spread of WNT5A-low prostate cancer cells in an orthotopic mouse model, PloS One, vol.12, issue.9, p.184418, 2017.

D. C. Smith, L. S. Rosen, R. Chugh, J. W. Goldman, L. Xu et al., First-in-human evaluation of the human monoclonal antibody vantictumab (OMP-18R5; anti-Frizzled) targeting the WNT pathway in a phase I study for patients with advanced solid tumors, J Clin Oncol. 20 mai, vol.31, pp.2540-2540, 2013.

M. Y. Choi, G. F. Widhopf, E. M. Ghia, R. L. Kidwell, M. K. Hasan et al., Phase I Trial: Cirmtuzumab Inhibits ROR1 Signaling and Stemness Signatures in Patients with Chronic Lymphocytic Leukemia, Cell Stem Cell. 1 juin, vol.22, issue.6, pp.951-959, 2018.

S. Naylor, M. J. Smalley, D. Robertson, B. A. Gusterson, P. A. Edwards et al., Retroviral expression of Wnt-1 and Wnt7b produces different effects in mouse mammary epithelium, J Cell Sci. juin, vol.113, pp.2129-2167, 2000.

E. Jost, D. Gezer, S. Wilop, H. Suzuki, J. G. Herman et al., Epigenetic dysregulation of secreted Frizzledrelated proteins in multiple myeloma, Cancer Lett. 18 août, vol.281, issue.1, pp.24-31, 2009.

T. Schlange, Y. Matsuda, S. Lienhard, A. Huber, and N. E. Hynes, Autocrine WNT signaling contributes to breast cancer cell proliferation via the canonical WNT pathway and EGFR transactivation, Breast Cancer Res BCR, vol.9, issue.5, p.63, 2007.

Y. Matsuda, T. Schlange, E. J. Oakeley, A. Boulay, and N. E. Hynes, WNT signaling enhances breast cancer cell motility and blockade of the WNT pathway by sFRP1 suppresses MDA-MB-231 xenograft growth, Breast Cancer Res BCR, vol.11, issue.3, p.32, 2009.

T. A. Dimeo, K. Anderson, P. Phadke, C. Fan, C. Feng et al., A novel lung metastasis signature links Wnt signaling with cancer cell self-renewal and epithelial-mesenchymal transition in basal-like breast cancer, Cancer Res. 1 juill, vol.69, issue.13, pp.5364-73, 2009.

N. Barker, R. A. Ridgway, J. H. Van-es, M. Van-de-wetering, H. Begthel et al., Crypt stem cells as the cells-of-origin of intestinal cancer, Nature. 29 janv, vol.457, issue.7229, pp.608-619, 2009.

D. M. Gonzalez and D. Medici, Signaling mechanisms of the epithelial-mesenchymal transition, Sci Signal. 23 sept, vol.7, issue.344, p.8, 2014.

J. I. Yook, X. Li, I. Ota, C. Hu, H. S. Kim et al., A Wnt-Axin2-GSK3beta cascade regulates Snail1 activity in breast cancer cells, Nat Cell Biol. déc, vol.8, issue.12, pp.1398-406, 2006.

L. Qi, B. Sun, Z. Liu, H. Li, J. Gao et al., Dickkopf-1 inhibits epithelial-mesenchymal transition of colon cancer cells and contributes to colon cancer suppression, Cancer Sci. avr, vol.103, issue.4, pp.828-863, 2012.

X. Zi, Y. Guo, A. R. Simoneau, C. Hope, J. Xie et al., Expression of Frzb/secreted Frizzled-related protein 3, a secreted Wnt antagonist, in human androgen-independent prostate cancer PC-3 cells suppresses tumor growth and cellular invasiveness, Cancer Res, vol.65, issue.21, pp.9762-70, 2005.

X. Ye, B. Zerlanko, A. Kennedy, G. Banumathy, R. Zhang et al., Downregulation of Wnt signaling is a trigger for formation of facultative heterochromatin and onset of cell senescence in primary human cells, Mol Cell. 20 juill, vol.27, issue.2, pp.183-96, 2007.

P. D. Adams and G. H. Enders, Wnt signaling and senescence: a tug of war in early neoplasia?, Cancer Biol Ther. nov, vol.7, issue.11, pp.1706-1717, 2008.

H. Zhang, X. Zhang, X. Wu, W. Li, P. Su et al., Interference of Frizzled 1 (FZD1) reverses multidrug resistance in breast cancer cells through the Wnt/?-catenin pathway, Cancer Lett, vol.323, issue.1, pp.106-119, 2012.

T. Noda, H. Nagano, I. Takemasa, S. Yoshioka, M. Murakami et al., Activation of Wnt/beta-catenin signalling pathway induces chemoresistance to interferon-alpha/5-fluorouracil combination therapy for hepatocellular carcinoma, Br J Cancer. 19 mai, vol.100, issue.10, pp.1647-58, 2009.

M. Flahaut, R. Meier, A. Coulon, K. A. Nardou, F. K. Niggli et al., The Wnt receptor FZD1 mediates chemoresistance in neuroblastoma through activation of the Wnt/beta-catenin pathway, Oncogene. 11 juin, vol.28, issue.23, pp.2245-56, 2009.

Z. Zhou, J. Wang, X. Han, J. Zhou, and S. Linder, Up-regulation of human secreted frizzled homolog in apoptosis and its down-regulation in breast tumors, Int J Cancer. 25 sept, vol.78, issue.1, pp.95-104, 1998.

J. Shou, F. Ali-osman, A. S. Multani, S. Pathak, P. Fedi et al., Human Dkk-1, a gene encoding a Wnt antagonist, responds to DNA damage and its overexpression sensitizes brain tumor cells to apoptosis following alkylation damage of DNA, Oncogene. 31 janv, vol.21, issue.6, pp.878-89, 2002.

J. Mazieres, L. You, B. He, Z. Xu, A. Y. Lee et al., Inhibition of Wnt16 in human acute lymphoblastoid leukemia cells containing the t(1;19) translocation induces apoptosis, Oncogene. 11 août, vol.24, issue.34, pp.5396-400, 2005.

H. Hirata, Y. Hinoda, K. Nakajima, K. Kawamoto, N. Kikuno et al., Wnt antagonist DKK1 acts as a tumor suppressor gene that induces apoptosis and inhibits proliferation in human renal cell carcinoma, Int J Cancer. 15 avr, vol.128, issue.8, pp.1793-803, 2011.

B. He, L. You, K. Uematsu, Z. Xu, A. Y. Lee et al., A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells, Neoplasia N Y N. févr, vol.6, issue.1, pp.7-14, 2004.

A. J. Mikels and R. Nusse, Purified Wnt5a protein activates or inhibits beta-catenin-TCF signaling depending on receptor context, PLoS Biol. avr, vol.4, issue.4, p.115, 2006.

E. Bakker, A. M. Das, W. Helvensteijn, P. F. Franken, S. Swagemakers et al., Wnt5a promotes human colon cancer cell migration and invasion but does not augment intestinal tumorigenesis in Apc1638N mice, Carcinogenesis. nov, vol.34, issue.11, pp.2629-2667, 2013.

E. Belotti, J. Polanowska, A. M. Daulat, S. Audebert, V. Thomé et al., The human PDZome: a gateway to PSD95-Disc large-zonula occludens (PDZ)-mediated functions, Mol Cell Proteomics MCP. sept, vol.12, issue.9, pp.2587-603, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00862192

D. Bilder, M. Li, and N. Perrimon, Cooperative regulation of cell polarity and growth by Drosophila tumor suppressors, Science. 7 juill, vol.289, issue.5476, pp.113-119, 2000.

M. Montcouquiol, R. A. Rachel, P. J. Lanford, N. G. Copeland, N. A. Jenkins et al., Identification of Vangl2 and Scrb1 as planar polarity genes in mammals, Nature. 8 mai, vol.423, issue.6936, pp.173-180, 2003.

H. B. Pearson, P. A. Perez-mancera, L. E. Dow, A. Ryan, P. Tennstedt et al., SCRIB expression is deregulated in human prostate cancer, and its deficiency in mice promotes prostate neoplasia, J Clin Invest, vol.121, issue.11, pp.4257-67, 2011.

L. Zhan, A. Rosenberg, K. C. Bergami, M. Yu, Z. Xuan et al., Deregulation of scribble promotes mammary tumorigenesis and reveals a role for cell polarity in carcinoma, Cell, vol.135, issue.5, pp.865-78, 2008.

M. Cordenonsi, F. Zanconato, L. Azzolin, M. Forcato, A. Rosato et al., The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells, Cell, vol.147, issue.4, pp.759-72, 2011.

N. J. Godde, J. M. Sheridan, L. K. Smith, H. B. Pearson, K. L. Britt et al., Scribble modulates the MAPK/Fra1 pathway to disrupt luminal and ductal integrity and suppress tumour formation in the mammary gland, PLoS Genet. mai, vol.10, issue.5, p.1004323, 2014.

A. Lhoumeau, M. Arcangeli, D. Grandis, M. Giordano, M. Orsoni et al., Ptk7-Deficient Mice Have Decreased Hematopoietic Stem Cell Pools as a Result of Deregulated Proliferation and Migration, J Immunol Baltim Md, vol.15, issue.10, pp.4367-77, 1950.
URL : https://hal.archives-ouvertes.fr/hal-01306950

P. Jung, C. Sommer, F. M. Barriga, S. J. Buczacki, X. Hernando-momblona et al., Isolation of Human Colon Stem Cells Using Surface Expression of PTK7, Stem Cell Rep. 8 déc, vol.5, issue.6, pp.979-87, 2015.

I. F. Yamben, R. A. Rachel, S. Shatadal, N. G. Copeland, N. A. Jenkins et al., Scrib is required for epithelial cell identity and prevents epithelial to mesenchymal transition in the mouse, Dev Biol. 1 déc, vol.384, issue.1, pp.41-52, 2013.

I. A. Elsum, C. Martin, and P. O. Humbert, Scribble regulates an EMT polarity pathway through modulation of MAPK-ERK signaling to mediate junction formation, J Cell Sci. 1 sept, vol.126, pp.3990-3999, 2013.

T. S. Gujral, M. Chan, L. Peshkin, P. K. Sorger, M. W. Kirschner et al., A noncanonical Frizzled2 pathway regulates epithelial-mesenchymal transition and metastasis, Cell, vol.159, issue.4, pp.844-56, 2014.

B. C. Lim, S. Matsumoto, H. Yamamoto, H. Mizuno, J. Kikuta et al., Prickle1 promotes focal adhesion disassembly in cooperation with the CLASP-LL5? complex in migrating cells, J Cell Sci, vol.15, issue.16, pp.3115-3144, 2016.

A. Jenny, R. S. Darken, P. A. Wilson, and M. Mlodzik, Prickle and Strabismus form a functional complex to generate a correct axis during planar cell polarity signaling, EMBO J. 1 sept, vol.22, issue.17, pp.4409-4429, 2003.

L. Zhang, V. Luga, S. K. Armitage, M. Musiol, A. Won et al., A lateral signalling pathway coordinates shape volatility during cell migration, Nat Commun, vol.26, p.11714, 2016.

E. K. Kim, S. J. Yun, J. M. Ha, Y. W. Kim, I. H. Jin et al., Selective activation of Akt1 by mammalian target of rapamycin complex 2 regulates cancer cell migration, invasion, and metastasis, Oncogene. 30 juin, vol.30, issue.26, pp.2954-63, 2011.

C. Navarro, S. Nola, S. Audebert, M. Santoni, J. Arsanto et al., Junctional recruitment of mammalian Scribble relies on E-cadherin engagement, Oncogene. 23 juin, vol.24, issue.27, pp.4330-4339, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00118710

V. A. Cantrell and J. R. Jessen, The planar cell polarity protein Van Gogh-Like 2 regulates tumor cell migration and matrix metalloproteinase-dependent invasion, Cancer Lett. 1 janv, vol.287, issue.1, pp.54-61, 2010.

L. M. Desrochers, M. A. Antonyak, and R. A. Cerione, Extracellular Vesicles: Satellites of Information Transfer in Cancer and Stem Cell Biology, Dev Cell. 23 mai, vol.37, issue.4, pp.301-310, 2016.

S. Zhang, L. Chen, B. Cui, H. Chuang, Y. J. Wang-rodriguez et al., ROR1 is expressed in human breast cancer and associated with enhanced tumor-cell growth, PloS One, vol.7, issue.3, p.31127, 2012.

J. P. Mackeigan, L. O. Murphy, and J. Blenis, Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance, Nat Cell Biol. juin, vol.7, issue.6, pp.591-600, 2005.

T. D. King, W. Zhang, M. J. Suto, and Y. Li, Frizzled7 as an emerging target for cancer therapy, Cell Signal. avr, vol.24, issue.4, pp.846-51, 2012.

K. Ueno, S. Hazama, S. Mitomori, M. Nishioka, Y. Suehiro et al., Down-regulation of frizzled-7 expression decreases survival, invasion and metastatic capabilities of colon cancer cells, Br J Cancer, vol.101, issue.8, pp.1374-81, 2009.

K. Ueno, M. Hiura, Y. Suehiro, S. Hazama, H. Hirata et al., Frizzled-7 as a potential therapeutic target in colorectal cancer, Neoplasia N Y N. juill, vol.10, issue.7, pp.697-705, 2008.

F. Bertucci, P. Finetti, and D. Birnbaum, Basal breast cancer: a complex and deadly molecular subtype, Curr Mol Med. janv, vol.12, issue.1, pp.96-110, 2012.

, Erbitux (cétuximab) : importance de la détermination du statut mutationnel des gènes RAS de type sauvage (exons 2, 3 et 4 des gènes KRAS et NRAS) avant d'instaurer un traitement par Erbitux -Lettre aux professionnels de santé -ANSM : Agence nationale de sécurité du médicament et des produits de santé

D. Sur,

. Vectibix, nouvelle recommandation concernant le statut mutationnel RAS -Lettre aux professionnels de santé -ANSM : Agence nationale de sécurité du médicament et des produits de santé

X. Lu, A. Borchers, C. Jolicoeur, H. Rayburn, J. C. Baker et al., PTK7/CCK-4 is a novel regulator of planar cell polarity in vertebrates, Nature. 1 juill, vol.430, issue.6995, pp.93-101, 2004.

F. Puppo, V. Thomé, A. Lhoumeau, M. Cibois, A. Gangar et al., Protein tyrosine kinase 7 has a conserved role in Wnt/?-catenin canonical signalling, EMBO Rep. janv, vol.12, issue.1, pp.43-52, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00566856

M. Hayes, M. Naito, A. Daulat, S. Angers, and B. Ciruna, Ptk7 promotes non-canonical Wnt/PCP-mediated morphogenesis and inhibits Wnt/?-catenin-dependent cell fate decisions during vertebrate development, Dev Camb Engl. avr, vol.140, issue.8, pp.1807-1825, 2013.

K. Mossie, B. Jallal, F. Alves, I. Sures, G. D. Plowman et al., Colon carcinoma kinase-4 defines a new subclass of the receptor tyrosine kinase family, Oncogene, vol.11, issue.10, pp.2179-84, 1995.

S. Martinez, P. Scerbo, M. Giordano, A. M. Daulat, A. Lhoumeau et al., The PTK7 and ROR2 Protein Receptors Interact in the Vertebrate WNT/Planar Cell Polarity (PCP) Pathway, J Biol Chem. 18 déc, vol.290, issue.51, pp.30562-72, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01306990

M. Podleschny, A. Grund, H. Berger, E. Rollwitz, and A. Borchers, A PTK7/Ror2 Co-Receptor Complex Affects Xenopus Neural Crest Migration, PLoS ONE, vol.17, issue.2015

A. Lhoumeau, F. Puppo, T. Prébet, L. Kodjabachian, and J. Borg, PTK7: a cell polarity receptor with multiple facets, Cell Cycle Georget Tex. 15 avr, vol.10, issue.8, pp.1233-1239, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00848894

L. Meng, K. Sefah, M. B. O'donoghue, G. Zhu, D. Shangguan et al., Silencing of PTK7 in colon cancer cells: caspase-10-dependent apoptosis via mitochondrial pathway, PloS One, vol.16, issue.11, p.14018, 2010.

J. Hatakeyama, J. H. Wald, I. Printsev, H. Ho, and K. L. Carraway, Vangl1 and Vangl2: planar cell polarity components with a developing role in cancer, Endocr Relat Cancer, vol.21, issue.5, pp.345-356, 2014.

M. N. Hayes, K. Mccarthy, J. A. Oliveira, M. L. Iyer, S. Garcia et al., Vangl2/RhoA Signaling Pathway Regulates Stem Cell Self-Renewal Programs and Growth in Rhabdomyosarcoma, Cell Stem Cell. 1 mars, vol.22, issue.3, pp.414-427, 2018.

M. Kaucká, K. Plevová, S. Pavlová, P. Janovská, A. Mishra et al., The planar cell polarity pathway drives pathogenesis of chronic lymphocytic leukemia by the regulation of B-lymphocyte migration, Cancer Res. 1 mars, vol.73, issue.5, pp.1491-501, 2013.

E. Belotti, T. M. Puvirajesinghe, S. Audebert, E. Baudelet, L. Camoin et al., Molecular characterisation of endogenous Vangl2/Vangl1 heteromeric protein complexes, PloS One, vol.7, issue.9, p.46213, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01447666

I. P. Ivanov, A. E. Firth, A. M. Michel, J. F. Atkins, and P. V. Baranov, Identification of evolutionarily conserved non-AUG-initiated N-terminal extensions in human coding sequences, Nucleic Acids Res. mai, vol.39, issue.10, pp.4220-4254, 2011.

J. Merte, D. Jensen, K. Wright, S. Sarsfield, Y. Wang et al., Sec24b selectively sorts Vangl2 to regulate planar cell polarity during neural tube closure, Nat Cell Biol. janv, vol.12, issue.1, pp.41-46, 2010.

C. Wansleeben, H. Feitsma, M. Montcouquiol, C. Kroon, E. Cuppen et al., Planar cell polarity defects and defective Vangl2 trafficking in mutants for the COPII gene Sec24b, Dev Camb Engl. avr, vol.137, issue.7, pp.1067-73, 2010.

G. Boncompain, S. Divoux, N. Gareil, H. De-forges, A. Lescure et al., Synchronization of secretory protein traffic in populations of cells, Nat Methods. 11 mars, vol.9, issue.5, pp.493-501, 2012.

H. Strutt, J. Gamage, and D. Strutt, Robust Asymmetric Localization of Planar Polarity Proteins Is Associated with Organization into Signalosome-like Domains of Variable Stoichiometry, Cell Rep, vol.06, issue.10, pp.2660-71, 2016.

A. Gurney, F. Axelrod, C. J. Bond, J. Cain, C. Chartier et al., Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors, Proc Natl Acad Sci U S A. 17 juill, vol.109, issue.29, pp.11717-11739, 2012.

M. M. Mita, C. Becerra, D. A. Richards, A. C. Mita, E. Shagisultanova et al., Phase 1b study of WNT inhibitor vantictumab (VAN, human monoclonal antibody) with paclitaxel (P) in patients (pts) with 1st-to 3rd-line metastatic HER2-negative breast cancer (BC), J Clin Oncol. 20 mai, vol.34, pp.2516-2516, 2016.

C. Zhang, P. Yeung, L. Beviglia, B. Cancilla, T. Tang et al., Abstract 2830: Predictive biomarker identification for response to vantictumab (OMP-18R5; anti-Frizzled) by mining gene expression data of human breast cancer xenografts, Cancer Res, vol.74, pp.2830-2830, 2014.

J. C. Sachdev, M. L. Maitland, M. Sharma, V. Moreno, V. Boni et al., PF-06647020 (PF-7020), an antibodydrug conjugate (ADC) targeting protein tyrosine kinase 7 (PTK7), in patients (pts) with advanced solid tumors: Results of a phase I dose escalation and expansion study, J Clin Oncol. 20 mai, vol.36, issue.15_suppl, pp.5565-5565, 2018.

R. Tabatabai, Y. Linhares, D. Bolos, M. Mita, and A. Mita, Targeting the Wnt Pathway in Cancer: A Review of Novel Therapeutics, Target Oncol, vol.12, issue.5, pp.623-664, 2017.

A. J. Mikels and R. Nusse, Wnts as ligands: processing, secretion and reception, Oncogene. 4 déc, vol.25, issue.57, pp.7461-7469, 2006.

B. Chen, M. E. Dodge, W. Tang, J. Lu, Z. Ma et al., Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer, Nat Chem Biol. févr, vol.5, issue.2, pp.100-107, 2009.

J. Liu, S. Pan, M. H. Hsieh, N. Ng, F. Sun et al., Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974, Proc Natl Acad Sci, vol.110, issue.50, pp.20224-20233, 2013.

B. Madan, Z. Ke, N. Harmston, S. Y. Ho, A. O. Frois et al., Wnt addiction of genetically defined cancers reversed by PORCN inhibition, Oncogene, vol.28, issue.17, pp.2197-207, 2016.

N. Krishnamurthy and R. Kurzrock, Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors, Cancer Treat Rev. janv, vol.62, pp.50-60, 2018.

A. Säfholm, J. Tuomela, J. Rosenkvist, J. Dejmek, P. Härkönen et al., The Wnt-5a-Derived Hexapeptide Foxy-5 Inhibits Breast Cancer Metastasis In vivo by Targeting Cell Motility, Clin Cancer Res, vol.14, issue.20, pp.6556-63, 2008.

L. M. Mehdawi, C. P. Prasad, R. Ehrnström, T. Andersson, and A. Sjölander, Non-canonical WNT5A signaling up-regulates the expression of the tumor suppressor 15-PGDH and induces differentiation of colon cancer cells, Mol Oncol, vol.10, issue.9, pp.1415-1444, 2016.

Q. Wang, M. Williamson, S. Bott, N. Brookman-amissah, A. Freeman et al., Hypomethylation of WNT5A, CRIP1 and S100P in prostate cancer, Oncogene, vol.26, issue.45, pp.6560-6565, 2007.

A. S. Syed-khaja, L. Helczynski, A. Edsjö, R. Ehrnström, A. Lindgren et al., Elevated level of Wnt5a protein in localized prostate cancer tissue is associated with better outcome, PloS One, vol.6, issue.10, p.26539, 2011.

A. Khaja, L. Egevad, L. Helczynski, P. Wiklund, T. Andersson et al., Emphasizing the role of Wnt5a protein expression to predict favorable outcome after radical prostatectomy in patients with low-grade prostate cancer, Cancer Med. août, vol.1, issue.1, pp.96-104, 2012.

A. B. El-khoueiry, Y. Ning, D. Yang, S. Cole, M. Kahn et al., A phase I first-in-human study of PRI-724 in patients (pts) with advanced solid tumors, J Clin Oncol. 20 mai, vol.31, pp.2501-2501, 2013.

K. M. Sakamoto, K. B. Kim, A. Kumagai, F. Mercurio, C. M. Crews et al., Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation, Proc Natl Acad Sci U S A. 17 juill, vol.98, issue.15, pp.8554-8563, 2001.

C. P. Tinworth, H. Lithgow, and I. Churcher, Small molecule-mediated protein knockdown as a new approach to drug discovery, MedChemComm. 7 déc, vol.7, issue.12, pp.2206-2222, 2016.

K. M. Sakamoto, K. B. Kim, R. Verma, A. Ransick, B. Stein et al., Development of Protacs to target cancerpromoting proteins for ubiquitination and degradation, Mol Cell Proteomics MCP. déc, vol.2, issue.12, pp.1350-1358, 2003.

R. Baron and M. Kneissel, WNT signaling in bone homeostasis and disease: from human mutations to treatments, Nat Med. févr, vol.19, issue.2, pp.179-92, 2013.

C. M. Laine, K. S. Joeng, P. M. Campeau, R. Kiviranta, K. Tarkkonen et al., WNT1 mutations in early-onset osteoporosis and osteogenesis imperfecta, N Engl J Med. 9 mai, vol.368, pp.1809-1825, 2013.

K. Iwatsuki, H. Liu, A. Grónder, M. A. Singer, T. F. Lane et al., Wnt signaling interacts with Shh to regulate taste papilla development, Proc Natl Acad Sci U S A. 13 févr, vol.104, issue.7, pp.2253-2261, 2007.

L. Zhang and L. Lum, Chemical Modulation of WNT Signaling in Cancer, Prog Mol Biol Transl Sci, vol.153, pp.245-69, 2018.

B. Biehs, G. Dijkgraaf, R. Piskol, B. Alicke, S. Boumahdi et al., A cell identity switch allows residual BCC to survive Hedgehog pathway inhibition, Nature, vol.562, issue.7727, pp.429-462, 2018.

B. Madan and D. M. Virshup, Targeting Wnts at the source--new mechanisms, new biomarkers, new drugs, Mol Cancer Ther. mai, vol.14, issue.5, pp.1087-94, 2015.

J. J. Luke, R. Bao, S. Spranger, R. F. Sweis, and T. Gajewski, Correlation of WNT/?-catenin pathway activation with immune exclusion across most human cancers, J Clin Oncol. 20 mai, vol.34, pp.3004-3004, 2016.

, New PCP-dependent cellular functions of Vangl2, Historical perspective of Vangl2 functions in PCP

.. .. Acknowledgements,

E. Bailly, Seminars in Cell & Developmental Biology, 2017.

E. Bailly, Seminars in Cell & Developmental Biology, 2017.

J. Taylor, N. Abramova, J. Charlton, and P. N. Adler, Van Gogh: a new Drosophila tissue polarity gene, Genetics, vol.150, pp.199-210, 1998.

T. Wolf and G. M. Rubin, Strabismus: a novel gene that regulates tissue polarity and cell fate decisions in Drosophila, Development, vol.125, pp.1149-1159, 1998.

J. A. Zallen, Planar polarity and tissue morphogenesis, Cell, vol.129, pp.1051-1063, 2007.

Q. Schenkelaars, L. Fierro-constain, E. Renard, and C. Borchiellini, Retracing the path of planar cell polarity, BMC Evol. Biol, vol.16, pp.69-81, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01444062

R. Hale and D. Strutt, Conservation of planar pathway function across the animal kingdom, Annu. Rev. Genet, vol.49, pp.529-551, 2015.

E. Torban, A. Patenaude, S. Leclerc, S. Rakowiecki, S. Gauthier et al., Genetic interaction between members of the Vangl family causes neural tube defects in mice, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.3449-3454, 2008.

H. Song, J. Hu, W. Chen, G. Elliott, P. Andre et al., Planar cell polarity breaks bilateral symmetry by controlling ciliary positioning, Nature, vol.466, pp.378-382, 2010.

A. Iliescu, M. Gravel, C. Horth, S. Apuzzo, and P. Gros, Transmembrane topology of mammalian planar cell polarity protein Vangl1, vol.50, pp.2274-2282, 2010.

B. Gao, H. Song, K. Bishop, G. Elliot, L. Garrett et al., Wnt signaling gradients establish planar cell polarity by inducing Vangl2 phosphorylation through Ror2, Dev. Cell, vol.20, pp.163-176, 2011.

E. Torban, H. J. Wang, N. Groulx, and P. Gros, Independent mutations in mouse Vangl2 that cause neural tube defects in looptail mice impair interaction with members of the Dishevelled family, J. Biol. Chem, vol.279, pp.52703-52713, 2004.

A. Jenny, R. S. Darken, P. A. Wilson, and M. Mlodzik, Prickle and Strabismus form a functional complex to generate a correct axis during planar cell polarity signaling, EMBO J, vol.22, pp.4409-4420, 2003.

Y. Guo, G. Zanetti, and R. Schekman, A novel GTP-binding protein-adaptor protein complex responsible for export of Vangl2 from the trans Golgi network, Elife, vol.2, p.160, 2013.

D. Devenport, The cell biology of planar cell polarity, J. Cell Biol, vol.207, pp.171-179, 2014.

P. N. Adler, The frizzled/Stan pathway and planar cell polarity in the Drosophila wing, Curr. Top. Dev. Biol, vol.101, pp.1-31, 2012.

H. Strutt and D. Strutt, Asymmetric localisation of planar polarity proteins: mechanisms and consequences, Semin. Cell Dev. Biol, vol.20, pp.957-963, 2009.

M. Montcouquiol, N. Sans, D. Huss, J. Kach, J. D. Dickman et al., Asymmetric localization of Vangl2 and Fz3 indicate novel mechanisms for planar cell polarity in mammals, J. Neurosci, vol.26, pp.5265-5275, 2006.

Y. Wang, N. Guo, and J. Nathans, The role of frizzled3 and frizzled6 in neural tube closure and in the planar polarity of inner-ear sensory hair cells, J. Neurosci, vol.26, pp.2147-2156, 2006.

D. Devenport and E. Fuchs, Planar polarization in embryonic epidermis orchestrates global asymmetric morphogenesis of hair follicles, Nat. Cell Biol, vol.10, pp.1257-1268, 2008.

E. K. Vladar, R. D. Bayly, A. M. Sangoram, M. P. Scott, and J. D. Axelrod, Microtubules enable the planar cell polarity of airway cilia, Curr. Biol, vol.22, pp.2203-2212, 2012.

M. T. Butler and J. B. Wallingford, Control of vertebrate core planar cell polarity protein localization and dynamics by Prickle 2, Development, vol.142, pp.3429-3439, 2015.

J. Wu and M. Mlodzik, A quest for the mechanism regulating global planar cell polarity of tissues, Trends Cell Biol, vol.19, pp.295-305, 2009.

J. Casal, P. A. Laurence, and G. Struhl, Two separate molecular systems, Dachsous/Fat and Starry Nnight/Frizzled, act independently to confer planar cell polarity, Development, vol.133, pp.4561-4572, 2006.

J. Wu, A. C. Roman, J. M. Carvajal-gonzalez, and M. Mlodzik, Wg and Wnt4 provide long-range directional input to planar cell polarity orientation in Drosophila, Nat. Cell Biol, vol.15, pp.1045-1055, 2013.

Y. Yang and M. Mlodzik, Wnt-Frizzled/planar cell polarity signaling: cellular orientation by facing the wind (Wnt), Annu. Rev. Cell Dev. Biol, vol.31, pp.623-646, 2015.

B. Gao, Wnt regulation of planar cell polarity (PCP), Curr. Top. Dev. Biol, vol.101, pp.263-295, 2012.

E. Torban, C. Kor, and P. Gros, Van Gogh-like2 (Strabismus) and its role in planar cell polarity and convergent extension in vertebrates, Trends Genet, vol.20, pp.570-577, 2004.

L. C. Strong and F. Hollander, Hereditary loop-tail in the house mouse: accompanied by imperforate vagina and with lethal craniorachisschisis when homozygous, J. Hered, vol.40, pp.329-334, 1949.

Z. Kibar, K. J. Vogan, N. Groux, M. J. Justice, D. A. Underhill et al., Ltap, a mammalian homolog of Drosophila Strabismus/Vang Gogh, is altered in the mouse neural tube mutant Loop-tail, Nat. Genet, vol.28, pp.251-255, 2001.

M. Montcouquiol, R. A. Rachel, P. J. Lanford, N. G. Copeland, N. A. Jenkins et al., Identification of Vangl2 and Scrb1 as planar polarity genes in mammals, Nature, vol.423, pp.173-177, 2003.

J. N. Murdoch, D. J. Henderson, K. Doudney, C. Gaston-massuet, H. M. Phillips et al., Disruption of scribble (Scrb1) causes severe neural tube defects in the circletail mouse, Hum. Mol. Genet, vol.112, pp.87-98, 2003.

Y. P. Lei, T. Zhang, H. Li, B. L. Wu, L. Jin et al., VANGL2 mutations in human cranial neural-tube defects, vol.362, pp.2232-2235, 2010.

A. Iliescu, M. Gravel, C. Horth, and P. Gros, Independent mutations at Arg181 and Arg274 of Vangl proteins that are associated with neural tube defects in humans decrease protein stability and impair membrane targeting, Biochemistry, vol.53, pp.5356-5364, 2014.

Z. Wu, D. Epasinghe, J. He, L. Li, D. W. Green et al., Altered tooth morphogenesis after silencing the planar cell polarity core component, Vangl2, Cell Tissue Res, vol.366, pp.617-621, 2016.

Y. Wang, J. Williams, A. Rattner, S. Wu, A. G. Bassuk et al., Patterning of papillae on the mouse tongue: a system for the quantitative assessment of planar cell polarity signaling, Dev. Biol, vol.419, pp.298-310, 2016.

Z. L. Hua, H. Chang, Y. Wang, P. M. Smallwood, and J. Nathans, Partial interchangeability of Fz3 and Fz6 in tissue polarity signaling for epithelial orientation and axon growth and guidance, Development, vol.141, pp.3944-3954, 2014.

E. Belotti, T. M. Puvirajesinghe, S. Audebert, E. Baudelet, L. Camoin et al., Molecular characterisation of endogenous Vangl2/Vangl1 heteromeric protein complexes, PLoS One, vol.7, issue.9, p.46213, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01447666

B. L. Rocque, S. Babayeva, J. Li, V. Leung, L. Nezvitsky et al., Deficiency of the planar cell polarity protein Vangl2 in podocytes affects glomerular morphogenesis and increases susceptibility to injury, J. Am. Soc. Nephrol, vol.26, pp.576-586, 2015.

T. Poobalasingam, L. L. Yates, S. A. Walker, M. Pereira, N. Y. Gross et al., Heterozygous Vangl2Looptail mice reveal novel roles for the planar cell polarity pathway in adult lung homeostasis and repair, Dis. Model Mech, vol.10, pp.409-423, 2017.

L. L. Yates, J. Papakrivopoulou, D. A. Long, P. Goggolidou, J. O. Connolly et al., The planar cell polarity gene Vangl2 is required for mammalian kidney-branching morphogenesis and glomerular maturation, Hum. Mol. Genet, vol.19, pp.4663-4676, 2010.

L. L. Yates, C. Schnatwinkel, J. N. Murdoch, D. Bogani, C. J. Formstone et al., The PCP genes Celsr1 and Vangl2 are required for normal lung branching morphogenesis, Hum. Mol. Genet, vol.11, pp.2251-2267, 2010.

A. S. Findlay, D. A. Panzica, P. Walczysko, A. B. Holt, D. J. Henderson et al., The core planar cell polarity gene, Vangl2, directs adult corneal epithelial cell alignment and migration, R. Soc. Open Sci, vol.3, 2016.

H. Chen, D. D. Mruk, W. M. Lee, and C. Y. Cheng, Planar cell polarity (PCP) protein Vangl2 regulates ectoplasmic specialization dynamics via its effects on actin microfilaments in the testes of male rats, Endocrinology, vol.157, pp.2140-2159, 2016.

J. Yuana, J. Chaa, W. Denga, A. Bartosa, X. Suna et al., Planar cell polarity signaling in the uterus directs appropriate positioning of the crypt for embryo implantation, Proc. Natl. Acad. Sci. U. S. A, vol.113, pp.8079-8088, 2016.

J. B. Wallingford, Planar cell polarity signaling, cilia and polarized ciliary beating, Curr. Opin. Cell Biol, vol.22, pp.597-604, 2010.

B. Guirao, A. Meunier, S. Mortaud, A. Aguilar, J. M. Corsi et al., Coupling between hydrodynamic forces and planar cell polarity orients mammalian motile cilia, Nat. Cell Biol, vol.12, pp.341-3150, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00555153

J. Merte, D. Jensen, K. Wright, S. Sarsfield, Y. Wang et al., Sec24b selectively sorts Vangl2 to regulate planar cell polarity during neural tube closure, Nat. Cell Biol, vol.12, pp.41-46, 2010.

C. Wansleeben, H. Feitsma, M. Montcouquiol, C. Kroon, E. Cuppen et al., Planar cell polarity defects and defective Vangl2 trafficking in mutants for the COPII gene Sec24b, Development, vol.137, pp.1067-1073, 2010.

J. G. D'arcangelo, K. R. Stahmer, and E. A. Miller, Vesicle-mediated export from the ER: COPII coat function and regulation, Biochim. Biophys. Acta, vol.1833, pp.2464-2472, 2013.

X. Y. Yang, X. Y. Zhou, Q. Q. Wang, H. Li, Y. Chen et al., Mutations in the COPII vesicle component gene SEC24B are associated with human neural tube defects, Hum. Mutat, vol.34, pp.1094-1101, 2013.

J. M. Carvajal-gonzalez, S. Balmer, M. Mendoza, A. Dussert, G. Collu et al.,

U. Roman, B. Weber, M. Ciruna, and . Mlodzik, The clathrin adaptor AP-1 complex and Arf1 regulate planar cell polarity in vivo, Nat. Commun, vol.7, p.6751, 2015.

H. Strutt, S. J. Warrington, and D. Strutt, Dynamics of core planar polarity protein turnover and stable assembly into discrete membrane subdomains, Dev. Cell, vol.20, pp.511-525, 2011.

D. Shi, F. Usami, K. Komatsu, S. Oka, T. Abe et al., Dynamics of planar cell polarity protein Vangl2 in the mouse oviduct epithelium, Mech. Dev, vol.141, pp.78-89, 2016.

I. Roszko, D. S. Sepich, J. R. Jessen, and A. Chandrasekhar, Lilianna Solnica-Krezel, A dynamic intracellular distribution of Vangl2 accompanies cell polarization during zebrafish gastrulation, Development, vol.142, pp.2508-2520, 2015.

O. Ossipova, K. Kyeongmi, and S. Y. Sokol, Planar polarization of Vangl2 in the vertebrate neural plate is controlled by Wnt and Myosin II signaling, Biol. Open, vol.4, pp.722-730, 2015.

T. Nagaoka, A. Inutsuka, K. Begum, K. M. Hafiz, and M. Kishi, Vangl2 regulates E-cadherin in epithelial cells, Sci. Rep, vol.4, p.6940, 2014.

A. P. Giese, J. Ezan, L. Wang, L. Lasvaux, F. Lembo et al., Gipc1 has a dual role in Vangl2 trafficking and hair bundle integrity in the inner ear, vol.139, pp.3775-3785, 2012.

E. Belotti, J. Polanowska, A. M. Daulat, S. Audebert, V. Thome et al., The human PDZome: a gateway to PSD95-Disc large-zonula occludens (PDZ)-mediated functions, vol.12, pp.2587-2603, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00862192

F. Steinberg, M. Gallon, M. Winfield, E. C. Thomas, A. J. Bell et al., A global analysis of SNX27-retromer assembly and cargo specificity reveals a function in glucose and metal ion transport, Nat. Cell Biol, vol.15, pp.461-471, 2013.

S. Li, R. Esterberg, V. Lachance, D. Ren, K. Radde-gallwitz et al., Rack1 is required for Vangl2 membrane localization and planar cell polarity signaling while attenuating canonical Wnt activity, Proc. Natl. Acad. Sci. U. S. A, vol.108, pp.2264-2269, 2011.

N. Escobedo, O. Contreras, R. Muñoz, M. Farías, H. Carrasco et al., Syndecan 4 interacts genetically with Vangl2 to regulate neural tube closure and planar cell polarity, Development, vol.140, pp.3008-3017, 2013.

Y. Wang, N. Baeyens, F. Corti, K. Tanaka, J. S. Fang et al., Syndecan 4 controls lymphatic vasculature remodeling during mouse embryonic development, Development, vol.143, pp.4441-4451, 2016.

L. M. Kallay, A. Mcnickle, P. J. Brennwald, A. L. Hubbard, and L. T. Braiterman, Scribble associates with two polarity proteins, Lgl2 and Vangl2, via distinct molecular domains, J. Cell. Biochem, vol.99, pp.647-664, 2006.

T. M. Puvirajesinghe, F. Bertucci, A. Jain, P. Scerbo, E. Belotti et al., Identification of p62/SQSTM1 as a component of non-canonical Wnt VANGL2-JNK signalling in breast cancer, Nat. Commun, vol.7, p.10318, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01447570

T. Young, Y. Poobalan, E. K. Tan, S. Tao, S. Ong et al., The PDZ domain protein Mcc is a novel effector of non-canonical Wnt signaling during convergence and extension in zebrafish, Development, vol.141, pp.3505-3516, 2014.

J. H. Wald, J. Hatakeyama, I. Printsev, A. Cuevas, W. H. Fry et al., Suppression of planar cell polarity signaling and migration in glioblastoma by Nrdp1-mediated Dvl polyubiquitination, Oncogene, vol.36, pp.5158-5167, 2017.

M. Park and R. T. Moon, The planar cell-polarity gene stbm regulates cell behaviour and cell fate in vertebrate embryos, Nat. Cell Biol, vol.4, pp.20-25, 2002.

A. M. Daulat, O. Luu, A. Sing, L. Zhang, J. L. Wrana et al., Mink1 regulates ?-catenin-independent Wnt signaling via prickle phosphorylation, Mol. Cell. Biol, vol.32, pp.173-185, 2012.

H. Yin, C. O. Copley, L. V. Goodrich, and M. R. Deans, Comparison of phenotypes between different vangl2 mutants demonstrates dominant effects of the looptail mutation during hair cell development, PLoS One, vol.7, issue.2, p.31988, 2012.

H. Strutt, J. Gamage, and D. Strutt, Robust asymmetric localization of planar polarity proteins is associated with organization into signalosome-like domains of variable stoichiometry, Cell Rep, vol.17, pp.2660-2671, 2016.

J. Courbard, A. Djiane, J. Wu, and M. Mlodzik, The apical/basal-polarity determinant acribble cooperates with the PCP core factor Stbm/Vang and functions as one of its effectors, Dev. Biol, vol.333, pp.67-77, 2009.

F. Kharfallah, M. C. Guyot, A. R. Hassan, R. Allache, E. Merello et al., Scribble1 plays an important role in the pathogenesis of neural tube defects through its mediating effect of Par-3 and Vangl1/2 localization, Hum. Mol. Genet, vol.26, pp.2307-2320, 2017.

R. Yao, Y. Natsume, and T. Noda, MAGI-3 is involved in the regulation of the JNK signaling pathway as a scaffold protein for frizzled and Ltap, Oncogene, vol.23, issue.36, pp.6023-6030, 2004.

O. K. Lee, K. K. Frese, J. S. James, D. Chadda, Z. H. Chen et al., Discs-Large and Strabismus are functionally linked to plasma membrane formation, Nat. Cell Biol, vol.5, pp.987-993, 2003.

T. Yoshioka, A. Hagiwara, Y. Hida, and T. Ohtsuka, Vangl2 the planar cell polarity protein, is complexed with postsynaptic density protein PSD-95, FEBS Lett, vol.587, pp.1453-1459, 2013.

T. Nagaoka, K. Tabuchi, and M. Kishi, PDZ interaction of Vangl2 links PSD-95 and Prickle2 but plays only a limited role in the synaptic localisation of Vangl2, Sci. Rep, vol.5, p.12916, 2015.

P. Andre, Q. Wang, N. Wang, B. Gao, A. Schilit et al., The Wnt coreceptor Ryk regulates Wnt/planar cell polarity by modulating the degradation of the core planar cell polarity component Vangl2, J. Biol. Chem, vol.287, pp.44518-44525, 2012.

R. Suriben, S. Kivimae, D. A. Fisher, R. T. Moon, and B. N. Cheyette, Posterior malformations in Dact1 mutant mice arise through misregulated Vangl2 at the primitive streak, Nat. Genet, vol.41, issue.9, pp.977-985, 2009.

X. Y. Yang and B. N. Cheyette, SEC14 and spectrin domains 1 (Sestd1) and Dapper antagonist of catenin 1 (Dact1) scaffold proteins cooperatively regulate the Van Gogh-like 2 (Vangl2) four-pass transmembrane protein and planar cell polarity (PCP) pathway during embryonic development in mice, J. Biol. Chem, vol.288, pp.20111-20120, 2013.

M. Lindqvist, Z. Horn, V. Bryja, G. Schulte, P. Papachristou et al., Vang-like protein 2 and Rac1 interact to regulate adherens junctions, J. Cell Sci, vol.123, pp.472-483, 2010.

B. Shafer, K. Onishi, C. Lo, G. Colakoglu, and Y. Zou, Vangl2 promotes Wnt/planar cell polarity-like signaling by antagonizing Dvl1-mediated feedback inhibition in growth cone guidance, Dev. Cell, vol.20, pp.177-191, 2011.

T. Nagaoka, R. Ohash, A. Inutsuka, S. Sakai, N. Fujisawa et al., The Wnt/planar cell polarity pathway component Vangl2 induces synapse formation through direct control of N-cadherin, Cell Rep, vol.6, pp.916-927, 2014.

S. Babayeva, B. Rocque, L. Aoudjit, Y. Zilber, J. Li et al., Planar cell polarity pathway regulates nephrin endocytosis in developing podocytes, J. Biol. Chem, vol.288, pp.24035-24048, 2013.

B. B. Williams, V. A. Cantrell, N. A. Mundell, A. C. Bennett, R. E. Quick et al., VANGL2 regulates membrane trafficking of MMP14 to control cell polarity and migration, J. Cell Sci, vol.125, pp.2141-2147, 2012.

E. Torban, A. Iliescu, and P. Gros, An expanding role of Vangl proteins in embryonic development, Curr. Top. Dev. Biol, vol.101, pp.237-261, 2012.

M. Sebbagh and J. Borg, Insight into planar cell polarity, Exp. Cell Res, vol.328, pp.284-295, 2014.

E. K. Vladar, J. V. Nayak, C. E. Milla, and J. D. Axelrod, Airway epithelial homeostasis and planar cell polarity signaling depend on multiciliated cell differentiation, JCI Insight, vol.1, p.88027, 2016.

J. Hatakeyama, J. H. Wald, I. Printsev, H. Ho, and K. L. Carraway, Vangl1 and Vangl2: planar cell polarity components with a developing role in cancer, Endocr. Relat. Cancer, vol.21, pp.345-356, 2014.

J. N. Anastas, T. L. Biechele, M. Robitaille, J. Muster, K. H. Allison et al., A protein complex of SCRIB NOS1AP and VANGL1 regulates cell polarity and migration, and is associated with breast cancer progression, Oncogene, vol.31, pp.3696-3708, 2012.

H. L. May-simera, R. S. Petralia, M. Montcouquiol, Y. X. Wang, K. B. Szarama et al., Ciliary proteins Bbs8 and Ift20 promote planar cell polarity in the cochlea, vol.142, pp.555-566, 2015.