701
|
Onishi H, Kai M, Odate S, Iwasaki H, Morifuji Y, Ogino T, Morisaki T, Nakashima Y, Katano M. Hypoxia activates the hedgehog signaling pathway in a ligand-independent manner by upregulation of Smo transcription in pancreatic cancer. Cancer Sci 2011; 102:1144-50. [DOI: 10.1111/j.1349-7006.2011.01912.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
702
|
|
703
|
Majewski IJ, Bernards R. Taming the dragon: genomic biomarkers to individualize the treatment of cancer. Nat Med 2011; 17:304-12. [DOI: 10.1038/nm.2311] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
704
|
Abstract
The odontogenic keratocyst (OKC, currently designated by the World Health Organization as a keratocystic odontogenic tumor) is a locally aggressive, cystic jaw lesion with a putative high growth potential and a propensity for recurrence. Although it is generally agreed that some features of OKCs are those of a neoplasia, notably the relatively high proliferative rate of epithelial cells, controversies over the behavior and management of OKCs still exist. This article is intended to review this intriguing entity and to summarize the findings of recent studies related to the nature of OKCs and their clinical and therapeutic implications. Recent advances in genetic and molecular research, i.e., PTCH1 mutations and involvement of the Hedgehog signaling pathway, have led to increased knowledge of OKC pathogenesis which hints at potential new treatment options, although the question of whether the OKC is a cyst or a cystic neoplasm is yet to be answered with certainty. Since some advocate a more conservative treatment for OKCs, notably marsupialization and decompression, future treatment strategies may focus on molecular approaches and eventually reduce or eliminate the need for aggressive surgeries.
Collapse
Affiliation(s)
- T-J Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, 22 South Zhongguancun Avenue, Haidian District, Beijing 100081, PR China.
| |
Collapse
|
705
|
Ailles L, Siu LL. Targeting the Hedgehog pathway in cancer: can the spines be smoothened? Clin Cancer Res 2011; 17:2071-3. [PMID: 21367749 DOI: 10.1158/1078-0432.ccr-11-0211] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aberrant Hedgehog (Hh) pathway signaling has been suggested to play a role in the development of multiple solid tumors and hematologic malignancies. GDC-0449 is a novel first-in-human, first-in-class smoothened (SMO) inhibitor, which has completed its phase I evaluation and achieved proof of concept in tumors with Hh pathway mutations.
Collapse
Affiliation(s)
- Laurie Ailles
- Princess Margaret Hospital, Ontario Cancer Institute, University of Toronto, Toronto, Canada
| | | |
Collapse
|
706
|
Singh MK, Brewer JD. Current Approaches to Skin Cancer Management in Organ Transplant Recipients. ACTA ACUST UNITED AC 2011; 30:35-47. [DOI: 10.1016/j.sder.2011.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
707
|
Roudaut H, Traiffort E, Gorojankina T, Vincent L, Faure H, Schoenfelder A, Mann A, Manetti F, Solinas A, Taddei M, Ruat M. Identification and mechanism of action of the acylguanidine MRT-83, a novel potent Smoothened antagonist. Mol Pharmacol 2011; 79:453-60. [PMID: 21177415 DOI: 10.1124/mol.110.069708] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
There is a clear need to develop novel pharmacological tools to improve our understanding of Smoothened (Smo) function in normal and pathological states. Here, we report the discovery, the mechanism of action, and the in vivo activity of N-(2-methyl-5-(3-(3,4,5-trimethoxybenzoyl)guanidino)phenyl)biphenyl-4-carboxamide (MRT-83), a novel potent antagonist of Smo that belongs to the acylguanidine family of molecules. MRT-83 fits to a proposed pharmacophoric model for Smo antagonists with three hydrogen bond acceptor groups and three hydrophobic regions. MRT-83 blocks Hedgehog (Hh) signaling in various assays with an IC50 in the nanomolar range, showing greater potency than the reference Smo antagonist cyclopamine. MRT-83 inhibits Bodipy-cyclopamine binding to human and mouse Smo but does not modify Wnt signaling in human embryonic kidney 293 transiently transfected with a Tcf/Lef-dependent Firefly luciferase reporter together with a Renilla reniformis luciferase control reporter. MRT-83 abrogates the agonist-induced trafficking of endogenous mouse or human Smo to the primary cilium of C3H10T1/2 or NT2 cells that derive from a pluripotent testicular carcinoma. Stereotaxic injection into the lateral ventricle of adult mice of MRT-83 but not of a structurally related compound inactive at Smo abolished up-regulation of Patched transcription induced by Sonic Hedgehog in the neighboring subventricular zone. These data demonstrate that MRT-83 efficiently antagonizes Hh signaling in vivo. All together, these molecular, functional and biochemical studies provide evidence that MRT-83 interacts with Smo. Thus, this novel Smo antagonist will be useful for manipulating Hh signaling and may help develop new therapies against Hh-pathway related diseases.
Collapse
Affiliation(s)
- Hermine Roudaut
- Laboratoire de Neurobiologie et Développement, Institut de Neurobiologie Alfred Fessard IFR2118, Centre National de la Recherche Scientifique, UPR-3294, Gif-sur-Yvette, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
708
|
Camp WL, Turnham JW, Athar M, Elmets CA. New agents for prevention of ultraviolet-induced nonmelanoma skin cancer. SEMINARS IN CUTANEOUS MEDICINE AND SURGERY 2011; 30:6-13. [PMID: 21540016 PMCID: PMC3488433 DOI: 10.1016/j.sder.2011.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the incidence of nonmelanoma skin cancer on the rise, current prevention methods, such as the use of sunscreens, have yet to prove adequate to reverse this trend. There has been considerable interest in identifying compounds that will inhibit or reverse the biochemical changes required for skin cancers to develop, either by pharmacologic intervention or by dietary manipulation. By targeting different pathways identified as important in the pathogenesis of nonmelanoma skin cancers, a combination approach with multiple agents or the addition of chemopreventative agents to topical sunscreens may offer the potential for novel and synergistic therapies in treating nonmelanoma skin cancer.
Collapse
MESH Headings
- Anilides/therapeutic use
- Carcinoma, Basal Cell/drug therapy
- Carcinoma, Basal Cell/etiology
- Carcinoma, Basal Cell/prevention & control
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/etiology
- Carcinoma, Squamous Cell/prevention & control
- Carotenoids/therapeutic use
- Cell Transformation, Neoplastic/radiation effects
- Cyclooxygenase 2 Inhibitors/therapeutic use
- DNA Repair Enzymes/therapeutic use
- Diet, Fat-Restricted
- Eflornithine/therapeutic use
- Flavonoids/therapeutic use
- Genistein/therapeutic use
- Humans
- Keratosis, Actinic/drug therapy
- Keratosis, Actinic/etiology
- Keratosis, Actinic/prevention & control
- Lycopene
- Phenols/therapeutic use
- Photochemotherapy
- Polyphenols
- Pyridines/therapeutic use
- Retinoids/therapeutic use
- Skin Neoplasms/drug therapy
- Skin Neoplasms/etiology
- Skin Neoplasms/prevention & control
- Sunscreening Agents/therapeutic use
- Ultraviolet Rays/adverse effects
Collapse
Affiliation(s)
- William L. Camp
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer W. Turnham
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL
- UAB Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Craig A. Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL
- UAB Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
709
|
Chung CH, Dignam JJ, Hammond ME, Klimowicz AC, Petrillo SK, Magliocco A, Jordan R, Trotti A, Spencer S, Cooper JS, Le QT, Ang KK. Glioma-associated oncogene family zinc finger 1 expression and metastasis in patients with head and neck squamous cell carcinoma treated with radiation therapy (RTOG 9003). J Clin Oncol 2011; 29:1326-34. [PMID: 21357786 DOI: 10.1200/jco.2010.32.3295] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Glioma-associated oncogene family zinc finger 1 (GLI1) expression was assessed to determine a potential role of hedgehog (Hh) signaling in head and neck squamous cell carcinoma (HNSCC). Additional proteins known to be modulated by Hh signaling, including beta-catenin (CTNNB1) and epidermal growth factor receptor (EGFR), were also assessed to determine the correlation among these distinct signaling pathways. PATIENTS AND METHODS Nuclear GLI1 and CTNNB1 expression levels were determined in tumors from patients enrolled on Radiation Therapy Oncology Group (RTOG) 9003, a radiation fractionation trial. The results were also correlated with previously determined EGFR expression. The expression levels were evaluated in relation to three end points: time to metastasis (TTM), time to disease progression (TDP), and overall survival (OS). RESULTS Among 1,068 eligible patients, data on GLI1, CTNNB1, and EGFR were available in 339, 164, and 300 patients, respectively. Although CTNNB1 expression did not differentiate prognosis, GLI1 was associated with poorer outcomes, adjusted for age, TNM stages, and Karnofsky performance score, and the significant influence persisted in a multivariable analysis (quartile 4 [Q4] v Q1 to Q3: TTM hazard ratio [HR], 2.7; 95% CI, 1.5 to 4.9; TDP HR, 1.6; 95% CI, 1.1 to 2.5; OS HR, 1.9; 95% CI, 1.4 to 2.7). The significance of GLI1 persisted in a multivariable analysis that included EGFR expression levels. CONCLUSION These data suggest that Hh signaling may play an important role in metastasis and that GLI1 could serve as a marker in HNSCC, but the regulatory mechanisms and oncogenic significance need further investigation. Risk classification based on this analysis needs a validation in independent cohorts.
Collapse
Affiliation(s)
- Christine H Chung
- Department of Oncology, Johns Hopkins University School of Medicine, 1650 Orleans St, CRB-1 Room 344, Baltimore, MD 21231-1000, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
710
|
Li F, Shi W, Capurro M, Filmus J. Glypican-5 stimulates rhabdomyosarcoma cell proliferation by activating Hedgehog signaling. J Cell Biol 2011; 192:691-704. [PMID: 21339334 PMCID: PMC3044117 DOI: 10.1083/jcb.201008087] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 01/24/2011] [Indexed: 12/11/2022] Open
Abstract
Glypican-5 (GPC5) is one of the six members of the glypican family. It has been previously reported that GPC5 stimulates the proliferation of rhabdomyosarcoma cells. In this study, we show that this stimulatory activity of GPC5 is a result of its ability to promote Hedgehog (Hh) signaling. We have previously shown that GPC3, another member of the glypican family, inhibits Hh signaling by competing with Patched 1 (Ptc1) for Hh binding. Furthermore, we showed that GPC3 binds to Hh through its core protein but not to Ptc1. In this paper, we demonstrate that GPC5 increases the binding of Sonic Hh to Ptc1. We also show that GPC5 binds to both Hh and Ptc1 through its glycosaminoglycan chains and that, unlike GPC3, GPC5 localizes to the primary cilia. Interestingly, we found that the heparan sulfate chains of GPC5 display a significantly higher degree of sulfation than those of GPC3. Based on these results, we propose that GPC5 stimulates Hh signaling by facilitating/stabilizing the interaction between Hh and Ptc1.
Collapse
Affiliation(s)
- Fuchuan Li
- Division of Molecular and Cell Biology, Sunnybrook Research Institute, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N3M5, Canada
| | - Wen Shi
- Division of Molecular and Cell Biology, Sunnybrook Research Institute, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N3M5, Canada
| | - Mariana Capurro
- Division of Molecular and Cell Biology, Sunnybrook Research Institute, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N3M5, Canada
| | - Jorge Filmus
- Division of Molecular and Cell Biology, Sunnybrook Research Institute, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N3M5, Canada
| |
Collapse
|
711
|
Graham RA, Lum BL, Cheeti S, Jin JY, Jorga K, Von Hoff DD, Rudin CM, Reddy JC, Low JA, Lorusso PM. Pharmacokinetics of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with locally advanced or metastatic solid tumors: the role of alpha-1-acid glycoprotein binding. Clin Cancer Res 2011; 17:2512-20. [PMID: 21300760 DOI: 10.1158/1078-0432.ccr-10-2736] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE In a phase I trial for patients with refractory solid tumors, hedgehog pathway inhibitor vismodegib (GDC-0449) showed little decline in plasma concentrations over 7 days after a single oral dose and nonlinearity with respect to dose and time after single and multiple dosing. We studied the role of GDC-0449 binding to plasma protein alpha-1-acid glycoprotein (AAG) to better understand these unusual pharmacokinetics. EXPERIMENTAL DESIGN Sixty-eight patients received GDC-0449 at 150 (n = 41), 270 (n = 23), or 540 (n = 4) mg/d, with pharmacokinetic (PK) sampling at multiple time points. Total and unbound (dialyzed) GDC-0449 plasma concentrations were assessed by liquid chromatography/tandem mass spectrometry, binding kinetics by surface plasmon resonance-based microsensor, and AAG levels by ELISA. RESULTS A linear relationship between total GDC-0449 and AAG plasma concentrations was observed across dose groups (R(2) = 0.73). In several patients, GDC-0449 levels varied with fluctuations in AAG levels over time. Steady-state, unbound GDC-0449 levels were less than 1% of total, independent of dose or total plasma concentration. In vitro, GDC-0449 binds AAG strongly and reversibly (K(D) = 13 μmol/L) and human serum albumin less strongly (K(D) = 120 μmol/L). Simulations from a derived mechanistic PK model suggest that GDC-0449 pharmacokinetics are mediated by AAG binding, solubility-limited absorption, and slow metabolic elimination. CONCLUSIONS GDC-0449 levels strongly correlated with AAG levels, showing parallel fluctuations of AAG and total drug over time and consistently low, unbound drug levels, different from previously reported AAG-binding drugs. This PK profile is due to high-affinity, reversible binding to AAG and binding to albumin, in addition to solubility-limited absorption and slow metabolic elimination properties.
Collapse
|
712
|
LoRusso PM, Rudin CM, Reddy JC, Tibes R, Weiss GJ, Borad MJ, Hann CL, Brahmer JR, Chang I, Darbonne WC, Graham RA, Zerivitz KL, Low JA, Von Hoff DD. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res 2011; 17:2502-11. [PMID: 21300762 DOI: 10.1158/1078-0432.ccr-10-2745] [Citation(s) in RCA: 415] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE The hedgehog (Hh) signaling pathway, a key regulator of cell growth and differentiation during development is implicated in pathogenesis of certain cancers. Vismodegib (GDC-0449) is a small-molecule inhibitor of smoothened, a key component of Hh signaling. This phase I trial assessed GDC-0449 treatment in patients with solid tumors refractory to current therapies or for which no standard therapy existed. EXPERIMENTAL DESIGN Sixty-eight patients received GDC-0449 at 150 mg/d (n = 41), 270 mg/d (n = 23), or 540 mg/d (n = 4). Adverse events, tumor responses, pharmacokinetics, and pharmacodynamic down-modulation of GLI1 expression in noninvolved skin were assessed. RESULTS Thirty-three of 68 patients had advanced basal cell carcinoma (BCC), 8 had pancreatic cancer, 1 had medulloblastoma; 17 other types of cancer were also represented. GDC-0449 was generally well-tolerated. Six patients (8.8%) experienced 7 grade 4 events (hyponatremia, fatigue, pyelonephritis, presyncope, resectable pancreatic adenocarcinoma, and paranoia with hyperglycemia), and 27.9% of patients experienced a grade 3 event [most commonly hyponatremia (10.3%), abdominal pain (7.4%), and fatigue (5.9%)]. No maximum tolerated dose was reached. The recommended phase II dose was 150 mg/d, based on achievement of maximal plasma concentration and pharmacodynamic response at this dose. Tumor responses were observed in 20 patients (19 with BCC and 1 unconfirmed response in medulloblastoma), 14 patients had stable disease as best response, and 28 had progressive disease. Evidence of GLI1 down-modulation was observed in noninvolved skin. CONCLUSIONS GDC-0449 has an acceptable safety profile and encouraging anti-tumor activity in advanced BCC and medulloblastoma. Further study in these and other cancer types is warranted.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Karmanos Cancer Institute, Detroit, Michigan; Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
713
|
Yamanaka H, Oue T, Uehara S, Fukuzawa M. Hedgehog signal inhibitor forskolin suppresses cell proliferation and tumor growth of human rhabdomyosarcoma xenograft. J Pediatr Surg 2011; 46:320-5. [PMID: 21292081 DOI: 10.1016/j.jpedsurg.2010.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 11/04/2010] [Indexed: 01/22/2023]
Abstract
BACKGROUND/PURPOSE We have previously reported that the Hedgehog (Hh) signaling pathway is activated in pediatric malignancies. In this study, we examined the effect of the Hh signal inhibitor forskolin on the growth of rhabdomyosarcoma (RMS) in vivo and in vitro and thereby elucidated the possibility of considering Hh signaling pathway as a therapeutic target for RMS. METHODS We evaluated the messenger RNA expressions of Hh signal mediators in 3 human RMS cell lines using reverse transcriptase-polymerase chain reaction method. The effect of forskolin on the tumor cell proliferation was investigated using WST-1 assay (Dojindo Co, Kumamoto, Japan). We inoculated 10(7) tumor cells into the back of nude mice to create RMS xenograft tumor models. Forskolin was subcutaneously administered in the region around the tumor, and the effect on the tumor growth was evaluated. RESULTS The messenger RNA expression of glioma-associated oncogene homolog 1, the marker of Hh signaling activation, was expressed at various levels in RMS cell lines. The proliferation of RMS cells was inhibited in a dose-dependent fashion by forskolin. Similarly, in the xenograft model, tumor growth was also significantly reduced by forskolin treatment. CONCLUSIONS Our findings suggest that the Hh signaling pathway plays an important role in the tumorigenesis of RMS and that this pathway can be considered to be a potential molecular target of new treatment strategies for RMS.
Collapse
Affiliation(s)
- Hiroaki Yamanaka
- Division of Pediatric Surgery, Department of Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
714
|
Mazumdar T, DeVecchio J, Shi T, Jones J, Agyeman A, Houghton JA. Hedgehog signaling drives cellular survival in human colon carcinoma cells. Cancer Res 2011; 71:1092-102. [PMID: 21135115 PMCID: PMC3032813 DOI: 10.1158/0008-5472.can-10-2315] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aberrant activation of Hedgehog (HH) signaling is implicated in many human cancers. Classical HH signaling is characterized by Smoothened (Smo)-dependent activation of Gli1 and Gli2, which transcriptionally regulate target genes. A small molecule inhibitor of Gli1 and Gli2, GANT61, was used to block HH signaling in human colon carcinoma cell lines that express HH signaling components. GANT61 administration induced robust cytotoxicity in 5 of 6 cell lines and moderate cytotoxicity in the remaining 1 cell line. In comparison, the classical Smo inhibitor, cyclopamine, induced modest cytotoxicity. Further, GANT61 treatment abolished the clonogenicity of all six human colon carcinoma cell lines. Analysis of the molecular mechanisms of GANT61-induced cytotoxicity in HT29 cells showed increased Fas expression and decreased expression of PDGFRα, which also regulates Fas. Furthermore, DR5 expression was increased whereas Bcl-2 (direct target of Gli2) was downregulated following GANT61 treatment. Suppression of Gli1 by shRNA mimicked the changes in gene expression observed in GANT61-treated cells. Overexpression of dominant-negative FADD (to abrogate Fas/DR5-mediated death receptor signaling) and/or Bcl-2 (to block mitochondria-mediated apoptosis) partially rescued GANT61-induced cytotoxicity in HT29 cells. Thus, activated GLI genes repress DR5 and Fas expressions while upregulating Bcl-2 and PDGFRα expressions to inhibit Fas and facilitate cell survival. Collectively, these results highlight the importance of Gli activation downstream of Smo as a therapeutic target in models of human colon carcinoma.
Collapse
Affiliation(s)
- Tapati Mazumdar
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jennifer DeVecchio
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ting Shi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Janay Jones
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Akwasi Agyeman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Janet A. Houghton
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
715
|
Amin SH, Tibes R, Kim JE, Hybarger CP. Hedgehog antagonist GDC-0449 is effective in the treatment of advanced basal cell carcinoma. Laryngoscope 2011; 120:2456-9. [PMID: 20927781 DOI: 10.1002/lary.21145] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVES/HYPOTHESIS To demonstrate the efficacy of the hedgehog pathway inhibitor GDC-0449 in the treatment of advanced basal cell carcinoma. DESIGN STUDY Case series. METHODS Three patients treated in a referral center for locally advanced basal cell carcinoma, one with metastases, were referred for treatment in a GDC-0449 phase I clinical trial. The treatment was once per day continuous therapy with oral GDC-0449. RESULTS Two patients showed complete clinical and radiologic resolution of disease, whereas one patient had significant reduction in tumor burden with radiologic evidence of slowly progressive local disease. Side effects were taste changes, mild to moderate hair loss, and muscle cramps in one patient. CONCLUSIONS GDC-0449 showed significant inhibitory activity in the treatment of advanced basal cell carcinoma.
Collapse
Affiliation(s)
- Shivan H Amin
- Department of Head and Neck Surgery, Kaiser Permanente Medical Center, Oakland, California, USA.
| | | | | | | |
Collapse
|
716
|
Hehlmann R, Jung-Munkwitz S, Saußele S. Treatment of chronic myeloid leukemia when imatinib fails. Expert Opin Pharmacother 2011; 12:269-83. [DOI: 10.1517/14656566.2011.533169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
717
|
Ganti AK, Kessinger A. Systemic therapy for disseminated basal cell carcinoma: an uncommon manifestation of a common cancer. Cancer Treat Rev 2011; 37:440-3. [PMID: 21216106 DOI: 10.1016/j.ctrv.2010.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/30/2010] [Accepted: 12/07/2010] [Indexed: 12/28/2022]
Abstract
While basal cell carcinoma (BCC) is the most common human malignancy, distant metastases from this are rare. Current therapy for disseminated BCC is based on anecdotal reports in the absence of clinical trials to guide management. For many years, platinum based cytotoxic chemotherapy was the mainstay of treatment. Advances in the understanding of the biology of BCC have led to the development of targeted therapies (e.g. inhibitors of the hedgehog and the epidermal growth factor receptor pathways) that are currently being investigated in this disease. This review summarizes the available data on the epidemiology and management of disseminated BCC.
Collapse
Affiliation(s)
- Apar Kishor Ganti
- Department of Internal Medicine, Division of Oncology/Hematology, University of Nebraska Medical Center, Omaha, USA.
| | | |
Collapse
|
718
|
Karlou M, Tzelepi V, Efstathiou E. Therapeutic targeting of the prostate cancer microenvironment. Nat Rev Urol 2011; 7:494-509. [PMID: 20818327 DOI: 10.1038/nrurol.2010.134] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Solid tumors can be thought of as multicellular 'organs' that consist of a variety of cells as well as a scaffold of noncellular matrix. Stromal-epithelial crosstalk is integral to prostate cancer progression and metastasis, and androgen signaling is an important component of this crosstalk at both the primary and metastatic sites. Intratumoral production of androgen is an important mechanism of castration resistance and has been the focus of novel therapeutic approaches with promising results. Various other pathways are important for stromal-epithelial crosstalk and represent attractive candidate therapeutic targets. Hedgehog signaling has been associated with tumor progression, growth and survival, while Src family kinases have been implicated in tumor progression and in regulation of cancer cell migration. Fibroblast growth factors and transforming growth factor beta signaling regulate cell proliferation, apoptosis and angiogenesis in the prostate cancer microenvironment. Integrins mediate communication between the cell and the extracellular matrix, enhancing growth, migration, invasion and metastasis of cancer cells. The contribution of stromal-epithelial crosstalk to prostate cancer initiation and progression provides the impetus for combinatorial microenvironment-targeting strategies.
Collapse
Affiliation(s)
- Maria Karlou
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | |
Collapse
|
719
|
[What's new in dermatological research?]. Ann Dermatol Venereol 2011; 137 Suppl 4:S137-44. [PMID: 21193117 DOI: 10.1016/s0151-9638(10)70040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dermatology research has been very rich this year, once again. The physiopathological mechanisms of paradoxical reactions to anti-TNF are better understood and new therapeutic targets for psoriasis have been evidenced. Targeted therapy in oncodermatology have shown their potential usefulness clinically but fundamental data have also clarified their mechanisms of action as well as their limits. The key role played by the immune system in nonsegmental vitiligo has also been clearly demonstrated. Fibroblasts as well as visible light seem to play a key role that has been poorly understood to date within the complex mechanisms of cutaneous pigmentation. Specific receptors of pruritus have been reported and foster hope for the development of more effective antipruriginous treatments in the near future. Other studies report new potential targets for diseases such as fungoid mycosis, atopic dermatitis, or scleroderma. Finally, physiopathological explanations have contributed to a variety of domains such as greying hair, axillary odors, HIV and herpes virus interrelations, and the teratogenicity of thalidomide.
Collapse
|
720
|
Liu H, Gu D, Xie J. Clinical implications of hedgehog signaling pathway inhibitors. CHINESE JOURNAL OF CANCER 2011; 30:13-26. [PMID: 21192841 PMCID: PMC3137255 DOI: 10.5732/cjc.010.10540] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 11/25/2010] [Accepted: 11/25/2010] [Indexed: 12/21/2022]
Abstract
Hedgehog was first described in Drosophila melanogaster by the Nobel laureates Eric Wieschaus and Christiane Nüsslein-Volhard. The hedgehog (Hh) pathway is a major regulator of cell differentiation, proliferation, tissue polarity, stem cell maintenance, and carcinogenesis. The first link of Hh signaling to cancer was established through studies of a rare familial disease, Gorlin syndrome, in 1996. Follow-up studies revealed activation of this pathway in basal cell carcinoma, medulloblastoma and, leukemia as well as in gastrointestinal, lung, ovarian, breast, and prostate cancer. Targeted inhibition of Hh signaling is now believed to be effective in the treatment and prevention of human cancer. The discovery and synthesis of specific inhibitors for this pathway are even more exciting. In this review, we summarize major advances in the understanding of Hh signaling pathway activation in human cancer, mouse models for studying Hh-mediated carcinogenesis, the roles of Hh signaling in tumor development and metastasis, antagonists for Hh signaling and their clinical implications.
Collapse
Affiliation(s)
- Hailan Liu
- Wells Center for Pediatric Research, Division of Hematology and Oncology, Department of Pediatrics, Indiana University Simon Cancer Center, Indiana University, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
721
|
Abstract
Functional chemicals are very useful tools for molecular biology studies. Due to its small size, large progeny clutch, and embryonic transparency, zebrafish serves as a superb in vivo animal model for chemical compound screens and characterization. During zebrafish embryogenesis, multiple developmental phenotypes can be easily examined under the microscope, therefore allowing a more comprehensive evaluation for identifying novel functional chemicals than cell-based assays. Ever since the first zebrafish-based chemical screen was conducted in the year 2000, many functional chemicals have been discovered using this strategy. In this chapter, we describe how to perform a typical zebrafish-based chemical screen and discuss the details of the protocol by using the example of the identification and characterization of two new Smo inhibitors with a Gli:GFP transgenic line.
Collapse
Affiliation(s)
- Hanbing Zhong
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen University Town, Shenzhen, China.
| | | |
Collapse
|
722
|
Abstract
Hedgehog is a ligand-activated signaling pathway that regulates Gli-mediated transcription. Although most noted for its role as an embryonic morphogen, hyperactive hedgehog also causes human skin and brain malignancies. The hedgehog-related gene anomalies found in these tumors are rarely found in prostate cancer. Yet surveys of human prostate tumors show concordance of high expression of hedgehog ligands and Gli2 that correlate with the potential for metastasis and therapy-resistant behavior. Likewise, prostate cancer cell lines express hedgehog target genes, and their growth and survival is affected by hedgehog/Gli inhibitors. To date, the preponderance of data supports the idea that prostate tumors benefit from a paracrine hedgehog microenvironment similar to the developing prostate. Uncertainty remains as to whether hedgehog's influence in prostate cancer also includes aspects of tumor cell autocrine-like signaling. The recent findings that Gli proteins interact with the androgen receptor and affect its transcriptional output have helped to identify a novel pathway through which hedgehog/Gli might affect prostate tumor behavior and raises questions as to whether hedgehog signaling in prostate cancer cells is suitably measured by the expression of Gli target genes alone.
Collapse
Affiliation(s)
- Mengqian Chen
- Ordway Research Institute, 150 New Scotland Avenue, Albany, NY 12208, USA
| | - Richard Carkner
- Ordway Research Institute, 150 New Scotland Avenue, Albany, NY 12208, USA
| | - Ralph Buttyan
- Ordway Research Institute, 150 New Scotland Avenue, Albany, NY 12208, USA
- Division of Urology, Albany Medical College, New York, NY, USA
| |
Collapse
|
723
|
Matthaei H, Maitra A. Precursor Lesions of Pancreatic Cancer. PRE-INVASIVE DISEASE: PATHOGENESIS AND CLINICAL MANAGEMENT 2011:395-420. [DOI: 10.1007/978-1-4419-6694-0_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
724
|
Basal cell carcinoma: from the molecular understanding of the pathogenesis to targeted therapy of progressive disease. J Skin Cancer 2010; 2011:650258. [PMID: 21253551 PMCID: PMC3021865 DOI: 10.1155/2011/650258] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 09/21/2010] [Indexed: 12/21/2022] Open
Abstract
Due to intensified research over the past decade, the Hedgehog (HH) pathway has been identified as a pivotal defect implicated in roughly 25% of all cancers. As one of the most frequent cancer worldwide, the development of Basal cell carcinoma (BCC) due to activation of the HH pathway has been convincingly demonstrated. Thus the discovery of this central tumor-promoting signalling pathway has not only revolutionized the understanding of BCC carcinogenesis but has also enabled the development of a completely novel therapeutic approach. Targeting just a few of several potential mutations, HH inhibitors such as GDC-0449 achieved already the first promising results in metastatic or locally advanced BCC. This paper summarizes the current understanding of BCC carcinogenesis and describes the current “mechanism-based” therapeutic strategies.
Collapse
|
725
|
Lin TL, Wang QH, Brown P, Peacock C, Merchant AA, Brennan S, Jones E, McGovern K, Watkins DN, Sakamoto KM, Matsui W. Self-renewal of acute lymphocytic leukemia cells is limited by the Hedgehog pathway inhibitors cyclopamine and IPI-926. PLoS One 2010; 5:e15262. [PMID: 21203400 PMCID: PMC3011010 DOI: 10.1371/journal.pone.0015262] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 11/03/2010] [Indexed: 01/16/2023] Open
Abstract
Conserved embryonic signaling pathways such as Hedgehog (Hh), Wingless and Notch have been implicated in the pathogenesis of several malignancies. Recent data suggests that Hh signaling plays a role in normal B-cell development, and we hypothesized that Hh signaling may be important in precursor B-cell acute lymphocytic leukemia (B-ALL). We found that the expression of Hh pathway components was common in human B-ALL cell lines and clinical samples. Moreover, pathway activity could be modulated by Hh ligand or several pathway inhibitors including cyclopamine and the novel SMOOTHENED (SMO) inhibitor IPI-926. The inhibition of pathway activity primarily impacted highly clonogenic B-ALL cells expressing aldehyde dehydrogenase (ALDH) by limiting their self-renewal potential both in vitro and in vivo. These data demonstrate that Hh pathway activation is common in B-ALL and represents a novel therapeutic target regulating self-renewal and persistence of the malignant clone.
Collapse
Affiliation(s)
- Tara L. Lin
- Section of Hematology and Oncology, Department of Internal Medicine, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Qiuju H. Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Patrick Brown
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Craig Peacock
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Akil A. Merchant
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sarah Brennan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Evan Jones
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Karen McGovern
- Infinity Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - D. Neil Watkins
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kathleen M. Sakamoto
- Gwynne Hazen Cherry Memorial Laboratories, Department of Pediatrics, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - William Matsui
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
726
|
Abstract
OBJECTIVE To conduct a systematic review of the role that the hedgehog signaling pathway has in pancreatic cancer tumorigenesis. METHOD PubMed search (2000-2010) and literature based references. RESULTS Firstly, in 2009 a genetic analysis of pancreatic cancers found that a core set of 12 cellular signaling pathways including hedgehog were genetically altered in 67-100% of cases. Secondly, in vitro and in vivo studies of treatment with cyclopamine (a naturally occurring antagonist of the hedgehog signaling pathway component; Smoothened) has shown that inhibition of hedgehog can abrogate pancreatic cancer metastasis. Thirdly, experimental evidence has demonstrated that sonic hedgehog (Shh) is correlated with desmoplasia in pancreatic cancer. This is important because targeting the Shh pathway potentially may facilitate chemotherapeutic drug delivery as pancreatic cancers tend to have a dense fibrotic stroma that extrinsically compresses the tumor vasculature leading to a hypoperfusing intratumoral circulation. It is probable that patients with locally advanced pancreatic cancer will derive the greatest benefit from treatment with Smoothened antagonists. Fourthly, it has been found that ligand dependent activation by hedgehog occurs in the tumor stromal microenvironment in pancreatic cancer, a paracrine effect on tumorigenesis. Finally, in pancreatic cancer, cells with the CD44+CD24+ESA+ immunophenotype select a population enriched for cancer initiating stem cells. Shh is increased 46-fold in CD44+CD24+ESA+ cells compared with normal pancreatic epithelial cells. Medications that destruct pancreatic cancer initiating stem cells are a potentially novel strategy in cancer treatment. CONCLUSIONS Aberrant hedgehog signaling occurs in pancreatic cancer tumorigenesis and therapeutics that target the transmembrane receptor Smoothened abrogate hedgehog signaling and may improve the outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Fergal C Kelleher
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland.
| |
Collapse
|
727
|
Beauchamp EM, Ringer L, Bulut G, Sajwan KP, Hall MD, Lee YC, Peaceman D, Ozdemirli M, Rodriguez O, Macdonald TJ, Albanese C, Toretsky JA, Uren A. Arsenic trioxide inhibits human cancer cell growth and tumor development in mice by blocking Hedgehog/GLI pathway. J Clin Invest 2010; 121:148-60. [PMID: 21183792 DOI: 10.1172/jci42874] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 10/13/2010] [Indexed: 12/19/2022] Open
Abstract
The Hedgehog (Hh) pathway is activated in some human cancers, including medulloblastoma. The glioma-associated oncogene homolog (GLI) transcription factors are critical mediators of the activated Hh pathway, and their expression may be elevated in some tumors independent of upstream Hh signaling. Thus, therapies targeting GLI transcription factors may benefit a wide spectrum of patients with mutations at different nodal points of the Hh pathway. In this study, we present evidence that arsenic trioxide (ATO) suppresses human cancer cell growth and tumor development in mice by inhibiting GLI1. Mechanistically, ATO directly bound to GLI1 protein, inhibited its transcriptional activity, and decreased expression of endogenous GLI target genes. Consistent with this, ATO inhibited the growth of human cancer cell lines that depended on upregulated GLI expression in vitro and in vivo in a xenograft model of Ewing sarcoma. Furthermore, ATO improved survival of a clinically relevant spontaneous mouse model of medulloblastoma with activated Hh pathway signaling. Our results establish ATO as a Hh pathway inhibitor acting at the level of GLI1 both in vitro and in vivo. These results warrant the clinical investigation of ATO for tumors with activated Hh/GLI signaling, in particular patients who develop resistance to current therapies targeting the Hh pathway upstream of GLI.
Collapse
Affiliation(s)
- Elspeth M Beauchamp
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
728
|
Basal cell carcinoma of the head and neck. J Skin Cancer 2010; 2011:496910. [PMID: 21209728 PMCID: PMC3010650 DOI: 10.1155/2011/496910] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/17/2010] [Accepted: 10/19/2010] [Indexed: 11/18/2022] Open
Abstract
Basal cell carcinoma (BCC) is a malignant neoplasm derived from nonkeratinizing cells that originate from the basal layer of the epidermis and is the most frequent type of skin cancer in humans, with cumulative exposure to ultraviolet radiation as an important risk factor. BCC occurs most frequently at sun-exposed sites, with the head and neck being common areas. Tumors can be classified as nodular, superficial, morpheaform, infiltrating, metatypic, and fibroepithelioma of Pinkus. Several treatment options such as surgical excision and nonsurgical procedures are available. The choice of treatment should be determined based on the histological subtype of a lesion, cost, its size and location, patient age, medical condition of the patient, treatment availability, and the patient's wishes. The aim of any therapy selected for BCC treatment involving the head and neck is to ensure complete removal, the preservation of function, and a good cosmetic outcome.
Collapse
|
729
|
Roussel MF. New concepts in organ site research on medulloblastoma: genetics and genomics. Future Oncol 2010; 6:1229-31. [PMID: 20799868 DOI: 10.2217/fon.10.86] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Medulloblastoma is the most common solid tumor in childhood, developing in the cerebellum, a hindbrain structure with origins from two distinct germinal zones. Therefore, medulloblastoma tumors show a diversity of signaling abnormalities and stem cell biologies. Sonic Hedgehog and Wnt pathways drive a substantial fraction of tumors; however, genetic pathways underlying the most aggressive subset of these tumors remain uncharted. The speakers of this session discussed the genetic diversity of both tumors and metastases, including forward genetic screens, molecular classifications and stem cell biology. They also addressed novel signaling pathways, mouse models and biomarkers, and clinical progress in targeting inappropriate activation of Shh signaling.
Collapse
Affiliation(s)
- Martine F Roussel
- St Jude Children's Research Hospital, Department of Genetics & Tumor Cell Biology, Memphis, TN 38105, USA.
| |
Collapse
|
730
|
Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol 2010; 8:97-106. [PMID: 21151206 DOI: 10.1038/nrclinonc.2010.196] [Citation(s) in RCA: 743] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor relapse and metastasis remain major obstacles for improving overall cancer survival, which may be due at least in part to the existence of cancer stem cells (CSCs). CSCs are characterized by tumorigenic properties and the ability to self-renew, form differentiated progeny, and develop resistance to therapy. CSCs use many of the same signaling pathways that are found in normal stem cells, such as Wnt, Notch, and Hedgehog (Hh). The origin of CSCs is not fully understood, but data suggest that they originate from normal stem or progenitor cells, or possibly other cancer cells. Therapeutic targeting of both CSCs and bulk tumor populations may provide a strategy to suppress tumor regrowth. Development of agents that target critical steps in the Wnt, Notch, and Hh pathways will be complicated by signaling cross-talk. The role that embryonic signaling pathways play in the function of CSCs, the development of new anti-CSC therapeutic agents, and the complexity of potential CSC signaling cross-talk are described in this Review.
Collapse
Affiliation(s)
- Naoko Takebe
- National Cancer Institute, Division of Cancer Treatment and Diagnosis, Cancer Therapy Evaluation Program, Investigational Drug Branch, EPN7131, 6130 Executive Boulevard, Rockville, Bethesda, MD 20852, USA
| | | | | | | |
Collapse
|
731
|
Lea DH, Skirton H, Read CY, Williams JK. Implications for Educating the Next Generation of Nurses on Genetics and Genomics in the 21st Century. J Nurs Scholarsh 2010; 43:3-12. [DOI: 10.1111/j.1547-5069.2010.01373.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
732
|
Neal JW, Sequist LV. Exciting new targets in lung cancer therapy: ALK, IGF-1R, HDAC, and Hh. Curr Treat Options Oncol 2010; 11:36-44. [PMID: 20676809 DOI: 10.1007/s11864-010-0120-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The anaplastic lymphoma kinase (ALK) inhibitor crizotinib will become an integral addition to the treatment of patients with non-small cell lung cancer (NSCLC) harboring genetic ALK translocations. The insulin-like growth factor receptor (IGF-1R) monoclonal antibody figitumumab, while initially promising, appears to increase toxicity and death in combination with chemotherapy in the treatment of patients with NSCLC of squamous histology; therefore, clinical development of this class of agents will need to proceed with caution. The histone deacetylation (HDAC) inhibitor vorinostat did not demonstrate an improvement in overall survival (OS) compared with placebo in a large randomized trial, but other agents in this class may have greater selectivity and efficacy. Inhibitors of the hedgehog (Hh) signaling pathways have some early clinical promise in both NSCLC and small cell lung cancer (SCLC), and larger studies using these agents are eagerly anticipated.
Collapse
Affiliation(s)
- Joel W Neal
- Stanford Cancer Center, 875 Blake Wilbur Drive, Stanford, CA 94305, USA,
| | | |
Collapse
|
733
|
Cancer stem cells: repair gone awry? JOURNAL OF ONCOLOGY 2010; 2011:465343. [PMID: 21188169 PMCID: PMC3003969 DOI: 10.1155/2011/465343] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 10/23/2010] [Indexed: 12/17/2022]
Abstract
Because cell turnover occurs in all adult organs, stem/progenitor cells within the stem-cell niche of each tissue must be appropriately mobilized and differentiated to maintain normal organ structure and function. Tissue injury increases the demands on this process, and thus may unmask defective regulation of pathways, such as Hedgehog (Hh), that modulate progenitor cell fate. Hh pathway dysregulation has been demonstrated in many types of cancer, including pancreatic and liver cancers, in which defective Hh signaling has been linked to outgrowth of Hh-responsive cancer stem-initiating cells and stromal elements. Hence, the Hh pathway might be a therapeutic target in such tumors.
Collapse
|
734
|
Chia YH, Ma CX. Hedgehog Pathway Inhibitors: Potential Applications in Breast Cancer. CURRENT BREAST CANCER REPORTS 2010. [DOI: 10.1007/s12609-010-0031-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
735
|
Dijkgraaf GJP, Alicke B, Weinmann L, Januario T, West K, Modrusan Z, Burdick D, Goldsmith R, Robarge K, Sutherlin D, Scales SJ, Gould SE, Yauch RL, de Sauvage FJ. Small molecule inhibition of GDC-0449 refractory smoothened mutants and downstream mechanisms of drug resistance. Cancer Res 2010; 71:435-44. [PMID: 21123452 DOI: 10.1158/0008-5472.can-10-2876] [Citation(s) in RCA: 299] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inappropriate Hedgehog (Hh) signaling has been directly linked to medulloblastoma (MB), a common malignant brain tumor in children. GDC-0449 is an Hh pathway inhibitor (HPI) currently under clinical investigation as an anticancer agent. Treatment of a MB patient with GDC-0449 initially regressed tumors, but this individual ultimately relapsed with a D473H resistance mutation in Smoothened (SMO), the molecular target of GDC-0449. To explore the role of the mutated aspartic acid residue in SMO function, we substituted D473 with every amino acid and found that all functional mutants were resistant to GDC-0449, with positively charged residues conferring potential oncogenic properties. Alanine scan mutagenesis of SMO further identified E518 as a novel prospective mutation site for GDC-0449 resistance. To overcome this form of acquired resistance, we screened a panel of chemically diverse HPIs and identified several antagonists with potent in vitro activity against these GDC-0449-resistant SMO mutants. The bis-amide compound 5 was of particular interest, as it was able to inhibit tumor growth mediated by drug resistant SMO in a murine allograft model of MB. However, focal amplifications of the Hh pathway transcription factor Gli2 and the Hh target gene cyclin D1 (Ccnd1) were observed in two additional resistant models, indicating that resistance may also occur downstream of SMO. Importantly, these HPI resistant MB allografts retained their sensitivity to PI3K inhibition, presenting additional opportunities for the treatment of such tumors.
Collapse
Affiliation(s)
- Gerrit J P Dijkgraaf
- Department of Molecular Biology, Genentech Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
736
|
Nehmé R, Mus-Veteau I. Proteins of the Hedgehog signaling pathway as therapeutic targets against cancer. Expert Rev Proteomics 2010; 7:601-12. [PMID: 20653513 DOI: 10.1586/epr.10.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Hedgehog pathway plays a crucial role in growth and patterning during embryonic development and is involved in stem cell maintenance and proliferation in adult tissues. Mutations that increase the overall activity of the pathway are often associated with a higher incidence of cancers. This article focuses on the mutations, misfoldings and deregulations of the Hedgehog pathway proteins that have been reported to be involved in different tumors, and on small molecules targeting these proteins shown to slow down the growth of certain tumors in various animal models. We propose that proteomics could be a powerful tool to identify new targets of the Hedgehog pathway, enabling the discovery of effective and novel treatments for cancers.
Collapse
Affiliation(s)
- Rony Nehmé
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
737
|
Von Hoff DD, Stephenson JJ, Rosen P, Loesch DM, Borad MJ, Anthony S, Jameson G, Brown S, Cantafio N, Richards DA, Fitch TR, Wasserman E, Fernandez C, Green S, Sutherland W, Bittner M, Alarcon A, Mallery D, Penny R. Pilot Study Using Molecular Profiling of Patients' Tumors to Find Potential Targets and Select Treatments for Their Refractory Cancers. J Clin Oncol 2010; 28:4877-83. [PMID: 20921468 DOI: 10.1200/jco.2009.26.5983] [Citation(s) in RCA: 489] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PurposeTo compare the progression-free survival (PFS) using a treatment regimen selected by molecular profiling (MP) of a patient's tumor with the PFS for the most recent regimen on which the patient had experienced progression (ie, patient as his own control).Patients and MethodsPatients with refractory metastatic cancer had tissue samples submitted for MP in two formats including formalin-fixed tissue for immunohistochemistry and fluorescent in situ hybridization assays and immediately frozen tissue for oligonucleotide microarray (MA) gene expression assays (all performed in a Clinical Laboratory Improvement Amendments [ CLIA ] –certified laboratory). The MP approach was deemed of clinical benefit for the individual patient who had a PFS ratio (PFS on MP-selected therapy/PFS on prior therapy) of ≥ 1.3.ResultsIn 86 patients who had MP attempted, there was a molecular target detected in 84 (98%). Sixty-six of the 84 patients were treated according to MP results. Eighteen (27%) of 66 patients had a PFS ratio of ≥ 1.3 (95% CI, 17% to 38%; one-sided, one-sample P = .007). Therefore, the null hypothesis (that ≤ 15% of this patient population would have a PFS ratio of ≥ 1.3) was rejected.ConclusionIt is possible to identify molecular targets in patients' tumors from nine different centers across the United States. In 27% of patients, the MP approach resulted in a longer PFS on an MP-suggested regimen than on the regimen on which the patient had just experienced progression. Issues to be considered in interpretation of this study include limited prior experience with patients as their own controls as a study end point and overall patient attrition.
Collapse
Affiliation(s)
- Daniel D. Von Hoff
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Joseph J. Stephenson
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Peter Rosen
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - David M. Loesch
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Mitesh J. Borad
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Stephen Anthony
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Gayle Jameson
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Susan Brown
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Nina Cantafio
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Donald A. Richards
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Tom R. Fitch
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Ernesto Wasserman
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Cristian Fernandez
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Sylvan Green
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - William Sutherland
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Michael Bittner
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Arlet Alarcon
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - David Mallery
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| | - Robert Penny
- From the Translational Genomics Research Institute (TGen); Caris Life Sciences Phoenix; TGen Clinical Research Service at Scottsdale Healthcare; Mayo Clinic Arizona, Scottsdale; University of Arizona Cancer Center, Tucson, AZ; Cancer Centers of the Carolinas, Greenville, SC; Tower Cancer Research Foundation, Beverly Hills, CA; Central Indiana Cancer Centers, Indianapolis, IN; Texas Oncology at Tyler, Tyler, TX; and AAIPharma, Wilmington, NC
| |
Collapse
|
738
|
Abstract
In vertebrate hedgehog signaling, hedgehog ligands are processed to become bilipidated and then multimerize, which allows them to leave the signaling cell via Dispatched 1 and become transported via glypicans and megalin to the responding cells. Hedgehog then interacts with a complex of Patched 1 and Cdo/Boc, which activates endocytic Smoothened to the cilium. Patched 1 regulates the activity of Smoothened (1) via Vitamin D3, which inhibits Smoothened in the absence of hedgehog ligand or (2) via oxysterols, which activate Smoothened in the presence of hedgehog ligand. Hedgehog ligands also interact with Hip1, Patched 2, and Gas1, which regulate the range as well as the level of hedgehog signaling. In vertebrates, Smoothened is shortened at its C-terminal end and lacks most of the phosphorylation sites of importance in Drosophila. Cos2, also of importance in Drosophila, plays no role in mammalian transduction, nor do its homologs Kif7 and Kif27. The cilium may provide a function analogous to that of Cos2 by linking Smoothened to the modulation of Gli transcription factors. Disorders associated with the hedgehog signaling network follow, including nevoid basal cell carcinoma syndrome, holoprosencephaly, Smith-Lemli-Opitz syndrome, Greig cephalopolysyndactyly syndrome, Pallister-Hall syndrome, Carpenter syndrome, and Rubinstein-Taybi syndrome.
Collapse
Affiliation(s)
- M Michael Cohen
- Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
739
|
Ortonne N. [Hereditary skin cancer: the pathologist's point of view]. Ann Pathol 2010; 30:93-6. [PMID: 21055558 DOI: 10.1016/j.annpat.2010.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 08/02/2010] [Indexed: 10/19/2022]
Affiliation(s)
- Nicolas Ortonne
- Département de pathologie, hôpital Henri-Mondor, groupe hospitalier Henri-Mondor-Albert-Chenevier, 51 avenue du Maréchal-Lattre-de-Tassigny, Créteil, France.
| |
Collapse
|
740
|
|
741
|
Castanedo GM, Wang S, Robarge KD, Blackwood E, Burdick D, Chang C, Dijkgraaf GJ, Gould S, Gunzner J, Guichert O, Halladay J, Khojasteh C, Lee L, Marsters JC, Murray L, Peterson D, Plise E, Salphati L, de Sauvage FJ, Wong S, Sutherlin DP. Second generation 2-pyridyl biphenyl amide inhibitors of the hedgehog pathway. Bioorg Med Chem Lett 2010; 20:6748-53. [DOI: 10.1016/j.bmcl.2010.08.134] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Revised: 08/24/2010] [Accepted: 08/27/2010] [Indexed: 11/17/2022]
|
742
|
Curtin JC, Lorenzi MV. Drug discovery approaches to target Wnt signaling in cancer stem cells. Oncotarget 2010; 1:563-577. [PMID: 21317452 PMCID: PMC3248130 DOI: 10.18632/oncotarget.191] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Accepted: 10/27/2010] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells (CSCs) represent a unique subset of cells within a tumor that possess self-renewal capacity and pluripotency, and can drive tumor initiation and maintenance. First identified in hematological malignancies, CSCs are now thought to play an important role in a wide variety of solid tumors such as NSCLC, breast and colorectal cancer. The role of CSCs in driving tumor formation illustrates the dysregulation of differentiation in tumorigenesis. The Wnt, Notch and Hedgehog (HH) pathways are developmental pathways that are commonly activated in many types of cancer. While substantial progress has been made in developing therapeutics targeting Notch and HH, the Wnt pathway has remained an elusive therapeutic target. This review will focus on the clinical relevance of the Wnt pathway in CSCs and tumor cell biology, as well as points of therapeutic intervention and recent advances in targeting Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Joshua C Curtin
- Oncology Drug Discovery, Research and Development, Bristol-Myers Squibb, Princeton, NJ, USA
| | | |
Collapse
|
743
|
Seidel K, Ahn CP, Lyons D, Nee A, Ting K, Brownell I, Cao T, Carano RAD, Curran T, Schober M, Fuchs E, Joyner A, Martin GR, de Sauvage FJ, Klein OD. Hedgehog signaling regulates the generation of ameloblast progenitors in the continuously growing mouse incisor. Development 2010; 137:3753-61. [PMID: 20978073 PMCID: PMC3049275 DOI: 10.1242/dev.056358] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2010] [Indexed: 01/22/2023]
Abstract
In many organ systems such as the skin, gastrointestinal tract and hematopoietic system, homeostasis is dependent on the continuous generation of differentiated progeny from stem cells. The rodent incisor, unlike human teeth, grows throughout the life of the animal and provides a prime example of an organ that rapidly deteriorates if newly differentiated cells cease to form from adult stem cells. Hedgehog (Hh) signaling has been proposed to regulate self-renewal, survival, proliferation and/or differentiation of stem cells in several systems, but to date there is little evidence supporting a role for Hh signaling in adult stem cells. We used in vivo genetic lineage tracing to identify Hh-responsive stem cells in the mouse incisor and we show that sonic hedgehog (SHH), which is produced by the differentiating progeny of the stem cells, signals to several regions of the incisor. Using a hedgehog pathway inhibitor (HPI), we demonstrate that Hh signaling is not required for stem cell survival but is essential for the generation of ameloblasts, one of the major differentiated cell types in the tooth, from the stem cells. These results therefore reveal the existence of a positive-feedback loop in which differentiating progeny produce the signal that in turn allows them to be generated from stem cells.
Collapse
Affiliation(s)
- Kerstin Seidel
- Departments of Orofacial Sciences and Pediatrics and Program in Craniofacial and Mesenchymal Biology, UCSF, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| | - Christina P. Ahn
- Department of Molecular Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - David Lyons
- Departments of Orofacial Sciences and Pediatrics and Program in Craniofacial and Mesenchymal Biology, UCSF, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| | - Alexander Nee
- Departments of Orofacial Sciences and Pediatrics and Program in Craniofacial and Mesenchymal Biology, UCSF, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| | - Kevin Ting
- Departments of Orofacial Sciences and Pediatrics and Program in Craniofacial and Mesenchymal Biology, UCSF, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| | - Isaac Brownell
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10021, USA
| | - Tim Cao
- Biomedical Imaging Group, Department of Tumor Biology and Angiogenesis, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Richard A. D. Carano
- Biomedical Imaging Group, Department of Tumor Biology and Angiogenesis, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tom Curran
- Department of Pathology and Laboratory Medicine, The Joseph Stokes Jr Research Institute, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Markus Schober
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Alexandra Joyner
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10021, USA
| | - Gail R. Martin
- Department of Anatomy and Program in Developmental Biology, UCSF, 1550 4th Street, San Francisco, CA 94143-2711, USA
| | - Frederic J. de Sauvage
- Department of Molecular Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ophir D. Klein
- Departments of Orofacial Sciences and Pediatrics and Program in Craniofacial and Mesenchymal Biology, UCSF, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| |
Collapse
|
744
|
Abstract
The Hedgehog (Hh) signaling pathway is critical for cell growth and differentiation during embryogenesis and early development. While it is mostly quiescent in adults, inappropriate reactivation of the Hh pathway has been shown to be involved in the development of cancer. A number of tumor types rely on overexpression of Hh ligands to activate the pathway in a paracrine manner from the tumor to the surrounding stroma. Alternatively, Hh ligands may act on cancer stem cells in some hematopoietic cancers, such as chronic myelogenous leukemia. However, the role of the Hh pathway is best established in tumors, such as basal cell carcinoma and medulloblastoma, where the pathway is activated via mutations. Understanding the contribution of Hh signaling in these various tumor types will be critical to the development and use of agents targeting this pathway in the clinic. We review here the activity of clinical inhibitors of the Hh pathway, including GDC-0449, a small molecule inhibitor of Smoothened (SMO).
Collapse
Affiliation(s)
- Jennifer A Low
- Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
745
|
Moulder S. Intrinsic Resistance to Chemotherapy in Breast Cancer. WOMENS HEALTH 2010; 6:821-30. [DOI: 10.2217/whe.10.60] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Systemic therapy improves disease-free survival in patients with breast cancer, but does not cure patients with advanced or metastatic disease, and fails to benefit the majority of patients with localized breast cancer. Intrinsic resistance to chemotherapy is emerging as a significant cause of treatment failure and evolving research has identified several potential causes of resistance, such as drug efflux pumps, disregulation of apoptosis and cancer stem cells. Building upon preclinical models, drugs designed to reverse resistance to therapy are currently under investigation in clinical trials for the treatment of breast cancer.
Collapse
Affiliation(s)
- Stacy Moulder
- Breast Medical Oncology, Unit 1354, The University of Texas MD Anderson Cancer Center, PO Box 301438, Houston, TX 77030, USA, Tel.: +1 713 792 2817, Fax: +1 713 794 4385,
| |
Collapse
|
746
|
Affiliation(s)
- Hui K. Gan
- Princess Margaret Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
747
|
Alison MR, Lim SML, Nicholson LJ. Cancer stem cells: problems for therapy? J Pathol 2010; 223:147-61. [PMID: 21125672 DOI: 10.1002/path.2793] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 09/13/2010] [Accepted: 09/21/2010] [Indexed: 12/20/2022]
Abstract
Many, if not all, tumours contain a sub-population of self-renewing and expanding stem cells known as cancer stem cells (CSCs). The symmetric division of CSCs is one mechanism enabling expansion in their numbers as tumours grow, while epithelial-mesenchymal transition (EMT) is an increasingly recognized mechanism to generate further CSCs endowed with a more invasive and metastatic phenotype. Putative CSCs are prospectively isolated using methods based on either a surface marker or an intracellular enzyme activity and then assessed by a 'sphere-forming' assay in non-adherent culture and/or by their ability to initiate new tumour growth when xenotransplanted into immunocompromised mice-hence, these cells are often referred to as tumour-propagating cells (TPCs). Cell sub-populations enriched for tumour-initiating ability have also been found in murine tumours, countering the argument that xenografting human cells merely select human cells with an ability to grow in mice. Cancer progression can be viewed as an evolutionary process that generates new/multiple clones with a fresh identity; this may be a major obstacle to successful cancer stem cell eradication if treatment targets only a single type of stem cell. In this review, we first briefly discuss evidence that cancer can originate from normal stem cells or closely related descendants. We then outline the attributes of CSCs and review studies in which they have been identified in various cancers. Finally, we discuss the implications of these findings for successful cancer therapies, concentrating on the self-renewal pathways (Wnt, Notch, and Hedgehog), aldehyde dehydrogenase activity, EMT, miRNAs, and other epigenetic modifiers as potential targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Malcolm R Alison
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | | | |
Collapse
|
748
|
Garvalov BK, Acker T. Cancer stem cells: a new framework for the design of tumor therapies. J Mol Med (Berl) 2010; 89:95-107. [DOI: 10.1007/s00109-010-0685-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 08/27/2010] [Accepted: 09/16/2010] [Indexed: 12/18/2022]
|
749
|
Stoff B, Salisbury C, Parker D, O'Reilly Zwald F. Dermatopathology of skin cancer in solid organ transplant recipients. Transplant Rev (Orlando) 2010; 24:172-89. [DOI: 10.1016/j.trre.2010.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Accepted: 05/17/2010] [Indexed: 12/21/2022]
|
750
|
|