The Hedgehog Signaling Pathway in Cancer

Page created by Travis Harmon
 
CONTINUE READING
The Hedgehog Signaling Pathway in Cancer
Molecular Pathways

The Hedgehog Signaling Pathway in Cancer
Marie Evangelista, Hua Tian, and Frederic J. de Sauvage

     Abstract          The Hedgehog (Hh) pathway is a signaling cascade that directs patterning in most animals and is
                       crucial for proper development. At the molecular level, Hh ligands drive cell proliferation in some
                       cell types while causing others to undergo differentiation. Hh signaling is most active during
                       embryogenesis, and aberrant reactivation of the pathway in adult tissue can lead to the develop-
                       ment of cancer. A comprehensive understanding of Hh signaling during development will un-
                       doubtedly shed light into the mechanism of Hh in cancer progression and identify potential
                       targets for therapeutic intervention.

 Pathway Overview                                                                 repressor (15). For the purposes of this review, the three
                                                                                  proteins will be referred to as Gli proteins. In the absence of Hh
   Hedgehog (Hh) is a morphogen that acts in a short- or long-                    signaling, protein kinases, such as protein kinase A, glycogen
range fashion on various tissue types (refer to refs. 1 – 3 for a                 synthase kinase 3h, and casein kinase 1a, phosphorylate Gli,
comprehensive review of Hh signaling). In mammals, there are                      leading to proteosome-mediated cleavage of Gli into an NH2-
three Hh proteins: Sonic Hh, Indian Hh, and Desert Hh. In                         terminal truncated form, which acts as a repressor of Hh target
cells that make Hh, newly made protein enters the secretory                       gene expression (Fig. 1A; refs. 16 – 19). Suppressor of fused
pathway and undergoes autoprocessing and lipid modifica-                          (Sufu) acts as another negative regulator of the pathway by
tions, resulting in the addition of a palmitoyl group to its NH2                  binding to Gli, both in the cytoplasm and in the nucleus, to
terminus and cholesterol to its COOH terminus (4). Subse-                         prevent it from activating Hh target genes (20 – 23).
quent release of Hh depends on Dispatched (Disp), a multi-                           In the presence of Hh ligand, the precise mechanism by
transmembrane protein, whereas diffusion of Hh requires                           which the Hh signal is transmitted from Smo to Gli is not well
Tout-velu-dependent synthesis of heparan sulfate proteogly-                       understood in vertebrates. In flies, Hh binding to Ptc leads to
cans (5, 6).                                                                      the surface localization of Smo along with a complex
   In cells receiving the Hh signal, pathway transmission is                      involving the kinase Fused (Fu) and the kinesin-like molecule
regulated at multiple levels. In the absence of Hh, Patched1                      Costal 2 (Cos2; refs. 24 – 27). Thus, Fu and Cos2 relay the
(Ptch1), a 12-transmembrane receptor, acts catalytically to sup-                  signal from Smo to cubitus interruptus (Ci), the fly homo-
press the activity of Smoothened (Smo), a seven-transmembrane                     logue of Gli. However, in vertebrates, this mechanism does not
protein, by preventing its localization to the cell surface                       seem to be conserved, as a Cos2 orthologue has not been
(Fig. 1A; refs. 7, 8). Hh pathway activation is initiated when                    identified and Fu is not required for transducing the Hh signal
Hh ligand binds to Ptch1, relieving its inhibition on Smo                         (28, 29). In contrast, recent studies have shown that Hh
(Fig. 1B; refs. 1, 2, 9). The amount of Hh available to bind                      signaling in vertebrates requires the presence of a nonmotile
Ptch1 is tightly regulated by Hh-binding proteins, such as Hh-                    cilium (Fig. 1B; refs. 30 – 32). Genetic studies in mice revealed
interacting protein (Hip; ref. 10) and growth arrest-specific                     that disruption of several components of anterograde or
gene (Gas1; ref. 11), which sequester Hh. In contrast, two                        retrograde intraflagellar transport leads to neural tube pattern-
newly identified single-transmembrane proteins, cell adhesion                     ing and limb development defects caused by impaired Hh
molecule-related/down-regulated by oncogenes (Cdon/Cdo)                           signaling between Smo and Gli (30 – 32). How Smo signals to
and brother of Cdo (Boc), may facilitate Hh ligand binding to                     the Gli proteins is currently an active area of research in the
Ptch1 (12 – 14).                                                                  Hh field today.
   Surface localization of Smo is thought to initiate a signaling
cascade, leading to the activation of the glioma-associated (Gli)
                                                                                     Hh Signaling in Cancer
family of zinc finger transcription factors. In vertebrates, there
are three Gli proteins, with Gli1 and Gli2 thought to activate                       Aberrant activation of the Hh pathway in cancers is caused
Hh target genes whereas Gli3 is thought to act mainly as a                        either by mutations in the pathway (ligand independent) or
                                                                                  through Hh overexpression (ligand dependent). Connections
                                                                                  between the former and cancer were established from observa-
Authors’ Affiliation: Department of Molecular Biology, Genentech, Inc., South     tions that mutations in the negative regulator, PTCH1, led to
San Francisco, California                                                         Gorlin syndrome (33, 34), a condition where patients develop
Received 7/14/06; revised 8/15/06; accepted 8/23/06.                              numerous basal cell carcinomas (BCC) and are predisposed to
Requests for reprints: Frederic J. de Sauvage, Department of Molecular Biology,   medulloblastoma (a rare tumor of cerebellar granule neuron
Genentech, Inc., 460 Point San Bruno Boulevard, South San Francisco, CA 94080.
Phone: 650-225-5841; Fax: 650-225-6497; E-mail: sauvage@ gene.com.
                                                                                  progenitor cells) and rhabdomyosarcoma (a muscle tumor).
  F 2006 American Association for Cancer Research.                                Inactivating mutations in PTCH1 have been detected in the
  doi:10.1158/1078-0432.CCR-06-1736                                               majority of sporadic BCC, although a small number (f10%) are

Clin Cancer Res 2006;12(20) October 15, 2006                                  5924                                       www.aacrjournals.org
The Hedgehog Signaling Pathway in Cancer
Hh Signaling Pathway in Cancer

Fig. 1. Model of Hh signaling in
vertebrates. A, in the absence of Hh ligand,
Ptch1inhibits surface localization of Smo
and protein kinases phosphorylate Gli
proteins, leading to an NH2-terminal
truncated form, which acts as a repressor of
Hh target gene expression. Gli3 is the
predominant repressor. Sufu also regulates
the pathway by binding to Gli, both in the
cytoplasm and in the nucleus, to prevent it
from activating Hh target genes. Table,
summary of genes implicated in cancer and
potential treatment using Hh antagonists or
Hh antibodies. B, Hh-mediated (Sonic,
Indian, or Desert) inactivation of Ptch1
allows relocation of Smo to the tip of cilia,
leading to downstream signaling events and
the activation of the Gli proteins. Mutations
in the pathway (*) or overexpression of
the Hh ligand are highlighted (**) in (A)
and (B) along with possible avenues of
treatment using antibodies or small
molecules (red arrows).

caused by a specific SMO mutation that make tumors less               Although mutations in components of the Hh pathway have
sensitive to PTCH1 repression (35, 36). In almost all BCC cases,   not yet been identified in tumors other than those mentioned
the Hh pathway is hyperactivated as indicated by up-regulation     above, activation of the Hh pathway via ligand overexpression
of Hh target genes (37). Mutations in PTCH1, SMO, or SUFU          is being documented in a growing number of cancers. Several
have also been identified in sporadic medulloblastoma, where       reports have shown that Hh overexpression, sometimes
one third of the tumors show increased Hh signaling (38, 39).      accompanied by increased expression of Hh target genes, is

www.aacrjournals.org                                           5925            Clin Cancer Res 2006;12(20) October 15, 2006
Molecular Pathways

detected in a broad spectrum of human tumor biopsies and cell        platelet-derived growth factor (53), fibroblast growth factor
lines, including small cell lung cancer (40), gastric and upper      (54), bone morphogenic protein (55), Notch (56), and Wnt
gastrointestinal track cancer (41), pancreatic cancer (42), and      (57, 58), which have been identified as Hh target genes in
prostate cancer (43, 44).                                            various models. However, to date, few universal target
                                                                     genes have been identified across different systems and much
                                                                     work still needs to be done to determine how Hh over-
 Role of the Hh Pathway in Ligand-Overexpressing
                                                                     expression contributes to tumorigenesis. Undoubtedly, to
 Tumors                                                              successfully target Hh-overexpressing cancers, it will be
   The mechanism by which Hh acts in ligand-overexpressing           important to understand the precise mechanism of how Hh
cancers is still unclear. One model suggests that these tumors       exerts its effects.
rely on autocrine signaling, where Hh ligand produced by                However, results derived from cell lines grown in vitro
tumor cells acts on neighboring tumor cells to stimulate             should be interpreted with caution. In some of these experi-
their growth or survival. This model is supported by in vitro        ments, high concentrations of Hh antagonist, 100- to 1,000-
data showing that proliferation of tumor cell lines is               fold higher than what is needed to block Hh signaling, were
accelerated by addition of Hh ligand (41) and inhibited by           necessary to cause growth inhibition. Interestingly, cell lines
a Hh neutralizing antibody (40) or by addition of cyclop-            established from the Ptch1 +/ P53 / medulloblastoma model
amine, a Hh pathway antagonist. In this model, Hh ligand             also become insensitive to Hh antagonists, whereas tumors
may directly affect the survival or proliferation of the entire      directly passaged s.c. in mice (i.e., direct allografts) main-
tumor cell (40 – 44). However, in prostate, Hh pathway               tained their molecular signature and were reproducibly
activity seems to correlate with higher-grade tumors, and            responsive to Hh antagonists (59). This suggests that cell
treatment of an aggressive prostate xenograft model with             lines grown in vitro may lose their dependence on the Hh
cyclopamine, a pathway antagonist (see below), prevents              pathway for survival and that the high concentrations of Hh
metastasis to the lung. Furthermore, constitutive activation of      antagonist required to inhibit their growth may be due to
the Hh pathway by overexpressing Gli1 in a rarely metasta-           nonspecific effects.
sizing clone, AT2.1, drove tumors to metastasize to the
lung through induction of genes involved in epithelial-                 Clinical-Translational Advances
mesenchymal transition (43). Thus, it has been suggested
that overexpression of Hh ligand leading to pathway                     The clear link between Hh pathway and human cancers, such
activation may be important for tumor proliferation, survival,       as BCC and medulloblastoma, drove the effort to identify small-
and/or metastasis (40 – 44).                                         molecule Hh antagonists to block the pathway. In addition to
   Alternatively, Hh may be acting on a small fraction of cancer     cyclopamine, a natural Hh antagonist isolated from corn lilies
stem cells that are capable of self-renewal and differentiation      that targets Smo (60), different classes of small-molecule Hh
among multiple lineages. Hh signaling is required for the            antagonists have been identified through cell-based screens using
normal growth and regeneration of organs, such as lung,              Hh reporter assays (61, 62). Using cyclopamine or small-
gastrointestinal track, prostate, from stem cells, and inappro-      molecule Hh antagonists as tools, experiments have shown the
priate and constitutive activation of Hh pathway during tissue       potential use of targeting this pathway both in tumors where the
repair and regeneration could promote tumorigenesis. Hh              pathway is mutated and in tumors where Hh ligand is
signaling has been indicated in proliferation of tissue stem cells   overexpressed. Elegantly illustrating the former, small-molecule
from central nervous system (45 – 47), mammary gland (48),           Hh antagonists have been used to treat endogenous medullo-
and hematopoietic stem cell (49). Genetic inhibition of Sonic        blastoma in the Ptch1 +/ P53 / mouse model where tumors
Hh signaling results in the loss of neural stem cell forming         develop with 100% incidence (63). In addition, studies have
potential from embryonic cortex (50), and activation of Hh           shown that cyclopamine can inhibit growth of medulloblastoma
signaling was shown to increase the number of mammary stem           tumors in vivo and in vitro (64, 65). Surprisingly, most human
cell in vitro (51). In addition, pharmacologic inhibition of Hh      medulloblastoma tumors tested seemed to be responsive,
signaling with cyclopamine in adult mice leads to reduced            suggesting potential broad application of Hh antagonist in
proliferation of stem cell in the subventricular zone (45)           treating this particular brain tumor, of which only f25% harbor
Expression of Hh pathway components has also been detected           mutations in the Hh pathway.
in mammary gland stem cells and in human breast "cancer                 Hh antagonists have also shown promises for treatment of
stem cells" characterized as CD44+CD24 /lowLin (51). In vitro,       BCC. In ex vivo models of BCC, CUR-61414, an aminoproline
cyclopamine was able to decrease mammary gland stem cells as         Hh antagonist, eliminated BCC-like lesions in various mouse
measured by their capacity to form mammospheres (48, 51).            models where tumor formation was driven by exposure of
Therefore, inhibition of the Hh pathway may provide a way to         skin explant cultures to recombinant Hh or by PTCH1
directly target cell populations that ultimately cause tumors.       mutations. This compound effectively down-regulates Hh
   Finally, Hh may act through a paracrine mechanism in              reporter gene expression and induces apoptosis specifically
ligand-dependent tumors. This model is more reminiscent of its       within BCC-like lesions while leaving normal adjacent cell
role in development and organogenesis, where Hh ligand is            intact (66).
secreted from the epithelium and signals to the underlying              In xenograft models of Hh-overexpressing tumors, including
myofibroblast/mesenchyme/stroma compartment. The stromal             small cell lung cancer, gastric and upper gastrointestinal track
compartment then signals back to the epithelium to regulate          cancer, pancreatic cancer, and prostate cancer, tumor growth
epithelial proliferation and differentiation through the pro-        inhibition on treatment with cyclopamine has also been shown
duction of factors, such as insulin-like growth factor (52),         (40 – 44). However, the activity of other pharmacologic agents

Clin Cancer Res 2006;12(20) October 15, 2006                     5926                                      www.aacrjournals.org
Hh Signaling Pathway in Cancer

has yet to be reported in these models, and the exact role of the                           inhibition of downstream effector molecules, such as Smo,
Hh pathway in these types of tumors still needs to be fully                                 with small-molecule antagonists. The latter has broader appli-
investigated to better assess the potential use of Hh pathway                               cations in treating both ligand-dependent and mutation-driven
antagonist in the clinic.                                                                   cancers.
                                                                                              An important issue that remains to be addressed is the
 Conclusion and Outlook                                                                     potential toxicity of blocking a critical developmental pathway,
                                                                                            such as Hh. Thus far, treatment with Hh antagonists, such as
  Aberrant Hh pathway activation, driven by either mutation                                 cyclopamine, seems to be well tolerated in animals. However,
or ligand overexpression, is often associated with human                                    because the pathway is reactivated during tissue repair and
cancers. Inhibition of the Hh pathway may provide a thera-                                  regeneration through stimulation of endogenous stem cell
peutic opportunity for treatment of tumors, such as pancreatic                              populations, the effect of systemic administration of Hh
cancer, where few options are available. Effective intervention                             antagonists on stem cell populations within the bone marrow,
of the Hh pathway can be achieved at the level of ligand                                    gut, muscle, liver, skin, brain, and others will have to be
binding to its receptor using anti-Hh antibodies or through                                 carefully monitored.

References
1. Huangfu D, Anderson KV. Signaling from Smo to            phorylation by glycogen synthase kinase 3 and casein           ia and prevents the expression of both activator and
  Ci/Gli: conservation and divergence of Hedgehog           kinase 1. Cell 2002;108:823 ^ 35.                              repressor functions of Gli. Dev Biol 2005;287:378 ^ 89.
  pathways from Drosophila to vertebrates. Develop-        18. Price MA, Kalderon D. Proteolysis of cubitus inter-        33. Hahn H,Wicking C, Zaphiropoulous PG, et al. Muta-
  ment 2006;133:3 ^ 14.                                     ruptus in Drosophila requires phosphorylation by               tion of the human homolog of Drosophila patched in
2. Jia J, Jiang J. Decoding the Hedgehog signal in          protein kinase A. Development 1999;126:4331 ^ 9.               the nevoid basal cell carcinoma syndrome. Cell 1996;
  animal development. Cell Mol Life Sci 2006;63:           19. Jia J, Amanai K, Wang G, Tang J, Wang B, Jiang J.           85:841 ^ 51.
  1249 ^ 65.                                                Shaggy/GSK3 antagonizes Hedgehog signaling by                 34. Johnson RL, Rothman AL, Xie J, et al. Human
3. Nybakken K, Perrimon N. Hedgehog signal transduc-        regulating Cubitus interruptus. Nature 2002;416:               homolog of patched, a candidate gene for the basal
  tion: recent findings. Curr Opin Genet Dev 2002;12:       548 ^ 52.                                                      cell nevus syndrome. Science 1996;272:1668 ^ 71.
  503 ^ 11.                                                20. Cheng SY, Bishop JM. Suppressor of Fused                   35. Reifenberger J,Wolter M,Weber RG, et al. Missense
4. Mann RK, Beachy PA. Novel lipid modifications of         represses Gli-mediated transcription by recruiting the         mutations in SMOH in sporadic basal cell carcinomas
  secreted protein signals. Annu Rev Biochem 2004;          SAP18-mSin3 corepressor complex. Proc Natl Acad                of the skin and primitive neuroectodermal tumors of
  73:891 ^ 923.                                             Sci U S A 2002;99:5442 ^ 7.                                    the central nervous system. Cancer Res 1998;58:
5. Burke R, Nellen D, Bellotto M, et al. Dispatched, a     21. Kogerman P, GrimmT, Kogerman L, et al. Mammali-             1798 ^ 803.
  novel sterol-sensing domain protein dedicated to the      an suppressor-of-fused modulates nuclear-cytoplastic          36. Xie J, Murone M, Luoh SM, et al. Activating
  release of cholesterol-modified hedgehog from sig-        shuttling of Gli-1. Nat Cell Biol 1999;1:312 ^ 9.              Smoothened mutations in sporadic basal-cell carcino-
  naling cells. Cell 1999;99:803 ^ 15.                     22. Ding Q, Fukami S, Meng X, et al. Mouse suppressor           ma. Nature 1998;391:90 ^ 2.
6. Bellaiche Y, The I, Perrimon N. Tout-velu is a           of fused is a negative regulator of sonic hedgehog sig-       37. Dahmane N, Lee J, Robins P, Heller P, Ruiz i Altaba
  Drosophila homologue of the putative tumour sup-          naling and alters the sucellular distribution of Gli1. Curr    A. Activation of the transcription factor Gli1 and
  pressor EXT-1 and is needed for Hh diffusion. Nature      Biol 1999;9:1119 ^ 22.                                         hedgehog signaling pathway in skin tumours. Nature
  1998;394:85 ^ 8.                                         23. Pearse RV II, Collier LS, Scott MP, Tabin CJ. Verte-        1997;389:876 ^ 81.
7. Taipale J, Cooper MK, Maiti T, Beachy PA. Patched        brate homologs of Drosophila suppressor of fused              38. Taylor MD, Liu L, Raffel C, et al. Mutations in SUFU
  acts catalytically to suppress the activity of Smooth-    interact with the gli family of transcriptional regulators.    predispose to medulloblastoma. Nat Genet 2002;31:
  ened. Nature 2002;418:892 ^ 7.                            Dev Biol 1999;212:323 ^ 36.                                    306 ^ 10.
8. Denef N, Neubuser D, Perez L, Cohen SM. Hedge-          24. Lum L, Zhang C, Oh S, et al. Hedgehog signal trans-        39. Raffel C, Jenkins RB, Frederick L, et al. Sporadic
  hog induces opposite changes in turnover and subcel-      duction via Smooothened association with a cyto-               medulloblastomas contain PTCH mutations. Cancer
  lular localization of patched and smoothened. Cell        plasmic complex scaffolded by the atypical kinesin,            Res 1997;57:842 ^ 5.
  2000;102:521 ^ 31.                                        Costal-2. Mol Cell 2003;12:1261 ^ 74.                         40. Watkins DN, Berman DM, Burkholder SG, Wang B,
9. Stone DM, Hynes M, Armanini M, et al. The tumour-       25. Jia J, Tong C, Jiang J. Smoothened transduces               Beachy PA, Baylin SB. Hedgehog signaling within
  suppressor gene patched encodes a candidate               Hedgehog signal by physically interacting with Cos-            airway epithelial progenitors and in small-cell lung
  receptor for Sonic hedgehog. Nature 1996;384:             tal2/Fused complex through its C-terminal tail. Genes          cancer. Nature 2003;422:313 ^ 7.
  129 ^ 34.                                                 Dev 2003;17:2709 ^ 20.                                        41. Berman DM, Karhadkar SS, Maitra A, et al. Wide-
10. Chuang PT, McMahon AP. Vertebrate Hedgehog             26. Ruel L, Rodriguez R, Gallet A, Lavenant-Staccini L,         spread requirement for Hedgehog ligand stimulation
  signaling modulated by induction of a Hedgehog-           Therond PP. Stability and association of Smoothened,           in growth of digestive tract tumours. Nature 2003;
  binding protein. Nature 1999;397:617 ^ 21.                Costal2, and Fused with Cubitus interruptus are regu-          425:846 ^ 51.
11. Lee CS, Buttitta L, Fan CM. Evidence that the WNT-      lated by Hedgehog. Nat Cell Biol 2003;5:907 ^ 13.             42. Thayer SP, di Magliano MP, Heiser PW, et al. Hedge-
  inducible growth arrest-specific gene 1 encodes an       27. Ogden SK, Ascano M, Jr., Stegman MA, Suber LM,              hog is and early and late mediator of pancreatic cancer
  antagonists of sonic hedgehog signaling in the somite.    Hooper JE, Robbins DJ. Identification of a functional          tumorigenesis. Nature 2003;425:851 ^ 6.
  Proc Natl Acad Sci U S A 2001;98:11347 ^ 52.              interaction between the transmembrane protein                 43. Karhadkar SS, Bova GS, Abdallah N, et al. Hedge-
12. Tenzen T, Allen BL, Cole F, Kang JS, Krauss RS,         Smoothened and the kinesin-related protein Costal2.            hog signaling in prostate regeneration, neoplasia, and
  McMahon AP. The cell surface membrane proteins            Curr Biol 2003;13:1998 ^ 2003.                                 metastasis. Nature 2004;431:707 ^ 12.
  Cdo and Boc are components and targets of the            28. Merchant M, Evangelista M, Luoh SM, et al. Loss of         44. Sanchez P, Hernandez AM, Stecca B, et al. Inhibi-
  Hedgehog signaling pathway and feedback network           the serine/threonine kinase fused results in postnatal         tion of prostate cancer proliferation by interference
  in mice. Dev Cell 2006;10:647 ^ 56.                       growth defects and lethality due to progressive hydro-         with SONIC HEDGEHOG-GLI1 signaling. Proc Natl
13. Yao S, Lum L, Beachy P. The ihog cell-surface pro-      cephalus. Mol Cell Biol 2005;25:7054 ^ 68.                     Acad Sci U S A 2004;101:12561 ^ 6.
  teins bind hedgehog and mediate pathway activation.      29. Chen MH, Gao N, Kawakami T, Chuang PT. Mice                45. Palma V, Lim DA, Dahmane N, et al. Sonic hedge-
  Cell 2006;125:343 ^ 57.                                   deficient in the fused homolog do not exhibit pheno-           hog controls stem cell behavior in the postnatal and
14. Zhang W, Kang JS, Cole F, Yi MJ, Krauss RS. Cdo         types indicative of perturbed hedgehog signaling               adult brain. Development 2005;132:335 ^ 44.
  functions at multiple points in the Sonic Hedgehog        during embryonic development. Mol Cell Biol 2005;             46. Ahn S, Joyner AL. In vivo analysis of quiescent
  pathway, and Cdo-deficient mice accurately model hu-      25:7042 ^ 53.                                                  adult neural stem cells responding to Sonic hedgehog.
  man holoprosencephaly. Dev Cell 2006;10:657 ^ 65.        30. Huangfu D, Liu A, Rakeman AS, Murcia NS,                    Nature 2005;437:894 ^ 7.
15. Ruiz i Altaba A. Catching a Gli-mpse of Hedgehog.       Niswander L, Anderson KV. Hedgehog signaling in               47. Machold R, Hayashi S, Rutlin M, et al. Sonic hedge-
  Cell 1997;90:193 ^ 6.                                     the mouse requires intraflagellar transport proteins.          hog is required for progenerator cell maintenance in tel-
16. Chen Y, Gallaher N, Goodman RH, Smolik SM.              Nature 2003;426:83 ^ 7.                                        encephalic stemcellniches. Neuron 2003;39:937 ^ 50.
  Protein kinase A directly regulates the activity and     31. Huangfu D, Anderson KV. Cilia and Hedgehog                 48. Liu S, Dontu G, Wicha MS. Mammary stem cells,
  proteolysis of cubitus interruptus. Proc Natl Acad Sci    responsiveness in the mouse. Proc Natl Acad Sci                self renewal pathways, and carcinogenesis. Breast
  U S A 1998;95:2349 ^ 54.                                  U S A 2005;102:11325 ^ 30.                                     Cancer Res 2005;7:86 ^ 95.
17. Price MA, Kalderon D. Proteolysis of the Hedgehog      32. May SR, AshiqueAM, Karlen M, et al. Loss of the ret-       49. Bhardwaj G, Murdoch B, Wu D, et al. Sonic hedge-
  signaling effector Cubitus interruptus requires phos-     rograde motor for IFTdisrupts localization of Smo to cil-      hog induces the proliferation of premative human

www.aacrjournals.org                                                                 5927                     Clin Cancer Res 2006;12(20) October 15, 2006
Molecular Pathways

 hematopoietic cells via BMP regulation. Nat Immunol         ulates proliferation and differentation of mesenchymal     61. Frank-Kamenetsky M, Zhang XM, Bottega S, et al.
 2001;2:172 ^ 80.                                            cells in the mouse metanephric kidney. Development          Small-molecule modulators of Hedgehog signaling:
50. Palma V, Ruiz i Altaba A. Hedgehog-GLI signaling         2002;129:5301 ^ 12.                                         identification and characterization of Smoothened ac-
 regulates the behavior of the cells with stem cell prop-   56. Hallahan AR, Pritchard JI, Hansen S, et al. The          tivity. J Biol 2002;1:10.
 erties in the developing neocortex. Development             SmoA1 mouse model reveals that notch signaling is          62. Chen JK, Taipale J, Young KE, Maiti T, Beachy PA.
 2004;131:337 ^ 45.                                          critical for the growth and survival of sonic hedge-        Small molecule modulation of Smoothened activity.
51. Liu S, Dontu G, Mantle ID, et al. Hedgehog signaling     hog-induced medulloblastomas. Cancer Res 2004;              Proc Natl Acad Sci U S A 2002;99:14071 ^ 6.
 and Bmi-1 regulate self-renewal of normal and malig-        64:7794 ^ 800.                                             63. Romer JT, Kimura H, Magdaleno S, et al. Suppres-
 nant human mammary stem cells. Cancer Res 2006;            57. Boras-Granic K, Chang H, Grosschedl R, Hamel PA.         sion of the Shh pathway using a small molecule inhi-
 66:6063 ^ 71.                                               Lef1is required for the transition of Wnt signaling from    bitor eliminates medulloblastoma in Ptc1(+/ )
52. Lipinski RJ, Cook CH, Barnett DH, Gipp JJ,               mesenchymal to epithelial cells in the mouse embry-         p53( / ) mice. Cancer Cell 2004;6:229 ^ 40.
 Peterson RE, Bushman W. Sonic hedgehog signaling            onic mammary gland. Dev Biol 2006;295:219 ^ 31.            64. Berman DM, Karhadkar SS, Hallahan AR, et al.
 regulates the expression of insulin-like growth factor     58. Madison BB, Braunstein K, Kuizon E, Portman K,           Medulloblastoma growth inhibition by hedgehog
 binding protein-6 during fetal prostate development.        Qiao XT, Gumucio DL. Epithelial hedgehog signals            pathway blockade. Science 2002;297:1559 ^ 61.
 Dev Dyn 2005;233:829 ^ 36.                                  pattern the intestinal crypt-villus axis. Development      65. Sanchez P, Ruiz i Altaba A. In vivo inhibition of
53. Xie J, Aszterbaum M, Zhang X, et al. A role of           2005;132:279 ^ 89.                                          endogenous brain tumors through systematic interfer-
 PDGFRa in basal cell carcinoma proliferation. Proc         59. Sasai K, Romer JT, LeeY, et al. Shh pathway activity     ence of Hedgehog signaling in mice. Mech Dev 2005;
 Natl Acad Sci U S A 2001;98:9255 ^ 9.                       is down-regulated in cultured medulloblastoma cells:        122:223 ^ 30.
54. Sun X, Lewandoski M, Meyers EN, LiuYH, Maxson            implications for preclinical studies. Cancer Res 2006;     66. Williams JA, Guicherit OM, Zaharian BI, et al. Identi-
 RE, Jr., Martin GR. Conditional inactivation of Fgf4        66:4215 ^ 22.                                               cation of a small molecule inhibitor of the hedgehog
 reveals complexity of signaling during limb bud devel-     60. ChenJK,TaipaleJ, Cooper MK, Beachy PA. Inhibition        signaling pathway: effects on basal cell carcinoma-
 opment. Nat Genet 2000;25:83 ^ 6.                           of Hedgehog signaling by direct binding of cyclop-          like lesions. Proc Natl Acad Sci U S A 2003;100:
55. Yu J, CarrollTJ, McMahon AP. Sonic hedgehog reg-         amine to Smoothened. Genes Dev 2002;16:2743 ^ 8.            4616 ^ 21.

Clin Cancer Res 2006;12(20) October 15, 2006                                        5928                                                       www.aacrjournals.org
You can also read