Review

Targeting Medulloblastoma: Small-Molecule Inhibitors of the Sonic Hedgehog Pathway as Potential Cancer Therapeutics

Justyna Romer and Tom Curran

Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee

Abstract develop and test new targeted therapies. We propose here a three- step approach for preclinical development of such drugs: (a) Medulloblastoma is the most common malignant pediatric Identify the control pathways driving tumor growth. (b) Create a brain tumor for which no satisfactory treatments exist. The genetically equivalent, high-incidence animal model, which reca- Sonic Hedgehog signaling pathway seems to play an impor- pitulates the specific subset of human disease with respect to tant role in the pathology of this disease. Here we review our developmental time frame and anatomy. (c) Design specific recent demonstration that a small-molecule inhibitor of this inhibitors of the growth control pathway impaired in tumors pathway can regress tumors that arise in a transgenic mouse and test them in the animal model. Efforts to identify new cancer model of medulloblastoma. These and other findings suggest modalities have increasingly relied upon advances in realizing the that inhibitors of Sonic Hedgehog signaling may offer an three elements of this strategy. effective way to target some malignancies. (Cancer Res 2005; Our laboratory studies the molecular causes underlying 65(12): 4975-8) medulloblastoma, the most common malignant pediatric brain tumor. Whereas there are several genetic subtypes of medullo- Introduction blastoma, the study described here is concerned specifically with Current strategies to develop better cancer therapies rely to a tumors arising due to disruption of the Sonic Hedgehog (SHH) great extent on targets identified from studying the genetics of pathway. The first indication that mutations in the SHH pathway human cancers. It is hoped that understanding the molecular contribute to medulloblastoma was the finding that Gorlin pathways disrupted in cancer will yield a better understanding of syndrome patients, who inherit germ line mutations in the molecular targets for therapy and thus lead to more specific and PTCH1 gene, are predisposed to medulloblastoma (2, 3). PTCH1 is therefore less toxic treatments. Brain tumors offer a particularly a trans-membrane protein that functions as a receptor for Shh. In difficult challenge not only because of their less accessible the absence of Shh, PTCH1 maintains the Shh pathway in the off anatomic location but also because they are relatively rare, state by acting on (SMO). Upon Shh binding to especially in the case of pediatric brain tumors. PTCH1, this repression is lifted and a signal is transduced Traditional drug development approaches for brain tumors were through Smo to the nucleus increasing expression of target genes optimized for cytotoxic therapies involving radiation and chemo- including GLI1 and PTCH1 itself (Fig. 1). This pathway is crucial therapy. Although these treatments do show some efficacy, the for normal development of the cerebellum, because it governs the drawback, especially for treatment of pediatric brain tumors, are proliferation of granule neuron precursor (GNP) cells, the most the severe and life-long side effects (1). As yet, no specific abundant neuronal population in the brain. GNP cells are ‘‘designer’’ drug has been made for brain tumors. One of the chief believed to be the cells of origin for medulloblastoma, although obstacles in the field is that testing new drugs relies on tissue definitive proof is still lacking. It has been suggested that culture and xenograft models, which recapitulate some but not all medulloblastoma arises as an aberration of normal developmental the features of malignancy. In particular, medulloblastoma tumor processes; a GNP cell fails to exit the cell cycle at the appropriate cells are notoriously difficult to grow ex vivo, and due to the time and remains in the EGL, eventually expanding to form a selective pressures imposed by cell culture, there are significant tumor (Fig. 2; ref. 4). The finding that over 30% of sporadic concerns whether the cell lines used are representative of the medulloblastomas show elevated expression of GLI1 supports this original tumor. Another consideration is that drugs are often hypothesis (5). tested in heterogeneous populations of tumor cells that are not The contribution of an inappropriately active SHH pathway to profiled genetically, impeding development of targeted therapies medulloblastoma formation makes it an ideal target to develop that are specific for a molecular pathway and that may only be new therapies. In our recent study (6), we set out to investigate disrupted in a subset of tumors. Some of these considerations may whether suppressing the overactive SHH signaling in medulloblas- explain why previous strategies for drug development have brought toma could cause tumor regression in vivo. To achieve this aim, we only modest improvements to the treatment of brain tumors. At used an animal model of medulloblastoma (7), in which mice are the current time, most advances in the clinic are still based on the heterozygous for the Ptc1 gene and null for the p53 gene. All +/À À/À stratification of patients into risk groups and optimizing the dose Ptc1 p53 mice spontaneously develop medulloblastoma as of radiation and chemotherapy accordingly. Therefore, there early as at 2 weeks of age and they die from the disease by week 16. remains a great need for more relevant models in which to The 100% incidence and short latency of tumor development makes this mouse strain well-suited to validate the therapeutic potential of Shh pathway inhibitors. The naturally occurring Requests for reprints: Justyna Romer, Department of Developmental inhibitor of Shh signaling, (8, 9), which binds to Smo Neurobiology, St. Jude Children’s Research Hospital, 332 North Lauderdale Street, (10), has been shown to inhibit the growth of medulloblastoma Memphis, TN 38105. Phone: 901-495-4801; Fax: 901-495-2270; E-mail: [email protected]. cells in cell culture and in murine allografts and xenografts (11). I2005 American Association for Cancer Research. However, although these studies are encouraging, extrapolation of www.aacrjournals.org 4975 Cancer Res 2005; 65: (12). June 15, 2005

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2005 American Association for Cancer Research. Cancer Research

Figure 1. Developing targeted therapies for medulloblastoma. The Shh pathway is critical for the normal development of the cerebellum. Shh released from the Purkinje cells acts on overlying GNP cells in the EGL, leading to proliferation. After this period of Shh-dependent proliferation, granule neurons exit the cell cycle, begin to differentiate, and migrate inwards, past the Purkinje cell layer to reside in the IGL in the mature cerebellum. these results to appropriate tissue settings would be desired to expression indicated that HhAntag-691 is capable of reaching its address shortcomings of the artificial environments presented by target in vivo, in the cerebellum of tumor-bearing mice. Further tissue culture and xenograft studies. This is especially pertinent in analysis of tumors isolated from treated mice showed that the case of Shh pathway–dependent tumors, as we have shown HhAntag-691 leads to a drastic decrease in proliferation as well that this pathway is down-regulated in cultured medulloblastoma as a modest increase in tumor apoptosis. Additionally, we found cells, rendering them ineffective for testing therapies based on that the treated brains became infiltrated with macrophages and suppression of the Shh pathway (6). reactive astrocytes, indicating that treatment incites the host To avoid these problems we relied on in vivo studies using a immune response and tissue repair mechanisms. When mice were novel, small-molecule inhibitor of the Shh pathway, HhAntag-691 treated for a longer period, a dramatic loss of tumor volume was (12), that readily enters the brain and suppresses Shh signaling by observed. In mice treated at intermediate doses, a 7-fold loss of binding to Smo (Fig. 3). This compound, a benzimidazole tumor mass was noted, and no tumor mass remained after derivative, inhibits the Shh pathway at lower concentrations than treatment with the highest dose. Finally, long-term treatment with cyclopamine. We treated Ptc1+/Àp53À/À mice with different doses HhAntag-691 prolonged tumor-free survival of Ptc1+/Àp53À/À of HhAntag-691, as well as with vehicle alone, and found that the mice, when compared with vehicle-treated mice. Importantly, daily treatment led to a dose-dependent down-regulation of several monitoring of the treated mice did not reveal any weight loss or genes that are overexpressed in medulloblastoma (Gli1, Ptc2, Sfrp1, other obvious deleterious side effects. These results offer strong and Math1). In contrast, Reln, a gene highly expressed in hope that targeting the Shh pathway in a subset of human medulloblastoma but not associated with the Shh pathway, was medulloblastomas may provide a feasible, nontoxic therapy for this not inhibited. We also saw a reduction of Gli2 expression.1 This was disease. important, because Gli2 can to some extent compensate for loss of Traditional approaches to testing new therapies, which are Gli1 in medulloblastoma formation (13). Suppression of Gli1 based on tissue culture and murine xenograft studies, might actually hinder development of meaningful therapies, because they rely on artificial systems that do not fully recapitulate the 1 Unpublished data. disease in situ. Whereas it is also possible to fault some features

Cancer Res 2005; 65: (12). June 15, 2005 4976 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2005 American Association for Cancer Research. Targeting Medulloblastoma of transgenic mouse models of cancer, the perspective provided patients. For this reason, the hope for clinical trials will likely by the study described above illustrates a new way to develop start with tests on adult tumors that rely on Shh signaling, drugs for medulloblastoma and other solid tumors that are thereby paving the way for clinical trials for medulloblastoma in driven by deregulation of the Shh pathway. By directly assaying children. Basal cell carcinoma (BCC) is one example of a tumor the molecular status of the pathway being targeted, the model in which the initial genetic lesion (PTCH1 mutation) is the same offers an invaluable tool for preclinical pharmacodynamic and as in medulloblastoma, making BCC suitable for evaluating the pharmacokinetic experiments. With molecular targeted therapeu- efficacy of Shh suppression as a therapy (14). Additionally, recent tics, the traditional strategy of testing drugs at the maximum reports point to other solid tumors that are dependent on Shh tolerated dose is clearly not appropriate. Instead, the specific signaling for growth, although genetic lesion in the Shh pathway nature of such agents dictates that they should be used at doses have yet to be identified in these cases (15–17). Such tumors that can effectively suppress the target; going beyond the offer additional candidates for initial clinical trials of Shh biologically relevant level may lead to deleterious effects not inhibitors. We hope that the efforts to produce animal models associated with target inhibition. Caution needs to be exercised which recapitulate specific human diseases will offer an in applying results from mouse trials to human disease; the improved chance of developing nontoxic, targeted cancer animal model system described here should be used in therapies, each one designed for a specific subtype of tumor, conjunction with studies done on human tumor material, if based on its genetic profile. This would be especially beneficial in possible, before moving forward to clinical trials. However, in the the area of pediatric brain tumors, where the currently available case of medulloblastoma, human systems which reliably recapit- cytotoxic treatments leave patients to cope with sometimes very ulate the in situ disease are not yet available. If the mouse model severe, neurologic and neuroendocrine consequences for the rest recapitulates the genetic signature of the human disease, as well of their lives. as its anatomic and temporal characteristics, targeting the same pathway in human disease may also be effective. One issue for medulloblastoma is that it is predominantly a pediatric tumor, Acknowledgments and new treatments for cancer are usually tested first in adult Received 2/11/2005; revised 4/5/2005; accepted 4/22/2005.

Figure 2. Medulloblastoma can arise as an aberration of cerebellar development, when a GNP cell fails to exit the cell cycle and remains proliferating in the EGL. Somatic mutations in different components of Shh pathway, including PTCH1 and SMOH may contribute to sporadic medulloblastoma (18–21) and germ line mutations of PTCH1 gene underlie tumors arising in patients with Gorlin syndrome (22, 23). www.aacrjournals.org 4977 Cancer Res 2005; 65: (12). June 15, 2005

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2005 American Association for Cancer Research. Cancer Research

Figure 3. The link between the Shh pathway and medulloblastoma led us to investigate whether suppressing Shh signaling in these tumors in vivo could be therapeutic. We used a small molecule inhibitor of the Shh pathway (HhAntag-691, a Smo inhibitor) in a mouse model of medulloblastoma. We found that treating mice with the Smo inhibitor suppressed the Shh pathway and led to the elimination of tumors. This was accompanied by a decrease in tumor cell proliferation and an increase in apoptosis. Importantly, long-term treatment prolonged tumor-free survival of the mice. This experiment underlines the importance of developing relevant animal models in which to test new cancer therapies.

References cyclopian effects of steroidal alkaloids from the plant 17. Watkins DN, Berman DM, Burkholder SG, Wang B, and structurally related compounds from other sources. Beachy PA, Baylin SB. Hedgehog signalling within airway 1. Gilbertson RJ. Medulloblastoma: signalling a change in Teratology 1968;1:5–10. epithelial progenitors and in small-cell lung cancer. treatment. Lancet Oncol 2004;5:209–18. 10. Chen JK, Taipale J, Cooper MK, Beachy PA. Inhibition Nature 2003;422:313–7. 2. Goodrich LV, Scott MP. Hedgehog and patched in of hedgehog signaling by direct binding of cyclopamine 18. Raffel C, Jenkins RB, Frederick L, et al. Sporadic neural development and disease. Neuron 1998;21: to smoothened. Genes Dev 2002;16:2743–8. medulloblastomas contain PTCH mutations. Cancer Res 1243–57. 11. Berman DM, Karhadkar SS, Hallahan AR, et al. 1997;57:842–5. 3. Kimonis VE, Goldstein AM, Pastakia B, et al. Clinical Medulloblastoma growth inhibition by hedgehog path- 19. Pietsch T, Waha A, Koch A, et al. Medulloblastomas manifestations in 105 persons with nevoid basal cell way blockade. Science 2002;297:1559–61. of the desmoplastic variant carry mutations of the carcinoma syndrome. Am J Med Genet 1997;69:299–308. 12. Gabay L, Lowell S, Rubin LL, Anderson DJ. Deregu- human homologue of Drosophila patched. Cancer Res 4. Wechsler-Reya R, Scott MP. The developmental biology lation of dorsoventral patterning by FGF confers 1997;57:2085–8. of brain tumors. Annu Rev Neurosci 2001;24:385–428. trilineage differentiation capacity on CNS stem cells 20. Vorechovsky I, Tingby O, Hartman M, et al. Somatic 5. Lee Y, Miller HL, Jensen P, et al. A molecular fingerprint in vitro. Neuron 2003;40:485–99. mutations in the human homologue of Drosophila for medulloblastoma. Cancer Res 2003;63:5428–37. 13. Kimura H, Stephen D, Joyner A, Curran T. Gli1 is patched in primitive neuroectodermal tumours. Onco- 6. Romer JT, Kimura H, Magdaleno S, et al. Suppression important for medulloblastoma formation in Ptc1(+/À) gene 1997;15:361–6. of the Shh pathway using a small molecule inhibitor mice. Oncogene 2005. 21. Wolter M, Reifenberger J, Sommer C, Ruzicka T, eliminates medulloblastoma in Ptc1(+/À)p53(À/À) 14. Williams JA, Guicherit OM, Zaharian BI, et al. Reifenberger G. Mutations in the human homologue of mice. Cancer Cell 2004;6:229–40. Identification of a small molecule inhibitor of the the Drosophila segment polarity gene patched (PTCH) 7. Wetmore C, Eberhart DE, Curran T. Loss of p53 but hedgehog signaling pathway: effects on basal cell in sporadic basal cell carcinomas of the skin and not ADP ribosylation factor accelerates medulloblasto- carcinoma-like lesions. Proc Natl Acad Sci U S A 2003; primitive neuroectodermal tumors of the central ma in mice heterozygous for patched. Cancer Res 2001; 100:4616–21. nervous system. Cancer Res 1997;57:2581–5. 61:513–6. 15. Berman DM, Karhadkar SS, Maitra A, et al. 22. Hahn H, Wicking C, Zaphiropoulous PG, et al. 8. Binns W, James LF, Shupe JL, Everett G. A congenital Widespread requirement for hedgehog ligand stimula- Mutations of the human homolog of Drosophila patched cyclopian-type malformation in lambs induced by tion in growth of digestive tract tumours. Nature 2003; in the nevoid basal cell carcinoma syndrome. Cell 1996; maternal ingestion of a range plant, Veratrum californi- 425:846–51. 85:841–51. cum. Am J Vet Res 1963;24:1164–75. 16. Thayer SP, di Magliano MP, Heiser PW, et al. 23. Johnson RL, Rothman AL, Xie J, et al. Human 9. Keeler RF, Binns W. Teratogenic compounds of Hedgehog is an early and late mediator of pancreatic homolog of patched, a candidate gene for the basal cell Veratrum californicum (Durand). V. Comparison of cancer tumorigenesis. Nature 2003;425:851–6. nevus syndrome. Science 1996;272:1668–71.

Cancer Res 2005; 65: (12). June 15, 2005 4978 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2005 American Association for Cancer Research. Targeting Medulloblastoma: Small-Molecule Inhibitors of the Sonic Hedgehog Pathway as Potential Cancer Therapeutics

Justyna Romer and Tom Curran

Cancer Res 2005;65:4975-4978.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/65/12/4975

Cited articles This article cites 22 articles, 9 of which you can access for free at: http://cancerres.aacrjournals.org/content/65/12/4975.full#ref-list-1

Citing articles This article has been cited by 25 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/65/12/4975.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/65/12/4975. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2005 American Association for Cancer Research.