701
|
Caro I, Low JA. The role of the hedgehog signaling pathway in the development of basal cell carcinoma and opportunities for treatment. Clin Cancer Res 2010; 16:3335-9. [PMID: 20439455 DOI: 10.1158/1078-0432.ccr-09-2570] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The hedgehog (Hh) signaling pathway plays an important role in embryogenesis across multiple species. Its activity is reduced or absent in adult organisms. However, activation of the pathway has been shown to be a factor in the development of a number of human malignancies and inhibition of the pathway is being investigated as a potential treatment for multiple cancers. The most extensively investigated and best characterized is basal cell carcinoma (BCC), which occurs in both an inherited form (basal cell nevus syndrome or Gorlin's syndrome) and a sporadic form. Sporadic BCCs are the most common human malignancy. There is recent data available on the use of a small molecule inhibitor of the pathway in BCC.
Collapse
Affiliation(s)
- Ivor Caro
- Genentech BioOncology, 1 DNA Way, South San Francisco, California 94080, USA.
| | | |
Collapse
|
702
|
Yamasaki A, Kameda C, Xu R, Tanaka H, Tasaka T, Chikazawa N, Suzuki H, Morisaki T, Kubo M, Onishi H, Tanaka M, Katano M. Nuclear factor kappaB-activated monocytes contribute to pancreatic cancer progression through the production of Shh. Cancer Immunol Immunother 2010; 59:675-86. [PMID: 19862523 PMCID: PMC11030800 DOI: 10.1007/s00262-009-0783-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 10/09/2009] [Indexed: 12/26/2022]
Abstract
Recently, it was reported that Hh signaling is activated in tumor stromal cells but not in tumor cells themselves and that stromal cells may play a role in the proliferation of cancer cells. This suggests the possibility that stromal cells have an important role in the proliferation of tumor cells that may be mediated through Hh signaling. In this report, we present for the first time that inflammation-stimulated monocytes produce Shh through activation of the NF-kappaB signaling pathway, and that the Shh produced promotes the proliferation of pancreatic cancer cells in a paracrine manner through Hh signaling.
Collapse
Affiliation(s)
- Akio Yamasaki
- Department of Cancer Therapy and Research, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
703
|
Abstract
OBJECTIVE Current studies indicate that Hedgehog (Hh) signaling must be excluded during early stages of pancreas formation. However, conflicting evidence suggests that Hh signaling may be active later during pancreas formation and that it is required for insulin production and secretion in cultured beta-cell lines. The objective of this study was to address these discrepancies by assessing the in vivo role of epithelial Hh signaling in the pancreas. RESEARCH DESIGN AND METHODS To identify Hh-active cells in the developing and adult pancreas epithelium, we characterized transgenic reporter Patched1-LacZ mice. To determine the requirement for epithelial Hh signaling in the pancreas, we eliminated an essential Hh signaling component, Smoothened (Smo), in the pancreatic epithelium, and assessed pancreatic development and adult beta-cell physiology phenotypes. RESULTS Characterization of Patched1-LacZ reporter mice revealed low-level LacZ expression in pancreatic epithelial cells throughout development until birth, when LacZ activity increases in intensity specifically in endocrine and ductal cells. In the absence of Hh signaling, Smo-deficient mice have delayed pancreas formation leading to a temporary reduction in pancreatic epithelium and beta-cell numbers. Although beta-cell numbers recover by birth, adult Smo-deficient mice display glucose intolerance, increased insulin sensitivity, and reduced total insulin production. CONCLUSIONS These data show that Hh signaling functions early during pancreas morphogenesis to regulate epithelial and beta-cell expansion and to modulate glucose metabolism by regulating insulin production in adult mice.
Collapse
Affiliation(s)
- Janet Lau
- From the Diabetes Center, University of California, San Francisco, San Francisco, California
| | - Matthias Hebrok
- From the Diabetes Center, University of California, San Francisco, San Francisco, California
- Corresponding author: Matthias Hebrok,
| |
Collapse
|
704
|
Slusarz A, Shenouda NS, Sakla MS, Drenkhahn SK, Narula AS, MacDonald RS, Besch-Williford CL, Lubahn DB. Common botanical compounds inhibit the hedgehog signaling pathway in prostate cancer. Cancer Res 2010; 70:3382-90. [PMID: 20395211 DOI: 10.1158/0008-5472.can-09-3012] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Many botanical compounds have been proposed to prevent cancer. We investigated the cancer treatment and prevention abilities of apigenin, baicalein, curcumin, epigallocatechin 3-gallate (EGCG), genistein, quercetin, and resveratrol both in vivo in transgenic adenocarcinoma of the mouse prostate (TRAMP) mice as well as in vitro in prostate cancer cell lines. In our experiments, these seven compounds act similarly to the Hedgehog antagonist cyclopamine, a teratogenic plant alkaloid, which had been previously shown to "cure" prostate cancer in a mouse xenograft model. With IC(50) values ranging from <1 to 25 mumol/L, these compounds can inhibit Gli1 mRNA concentration by up to 95% and downregulate Gli reporter activity by 80%. We show that four compounds, genistein, curcumin, EGCG, and resveratrol, inhibit Hedgehog signaling as monitored by real-time reverse transcription-PCR analysis of Gli1 mRNA concentration or by Gli reporter activity. Three compounds, apigenin, baicalein, and quercetin, decreased Gli1 mRNA concentration but not Gli reporter activity. Our results show that these compounds are also able to reduce or delay prostate cancer in vivo in TRAMP mice. All seven compounds, when fed in combination as pure compounds or as crude plant extracts, inhibit well-differentiated carcinoma of the prostate by 58% and 81%, respectively. In vitro, we show that all seven compounds also inhibit growth in human and mouse prostate cancer cell lines. Mechanistically, we propose the Hedgehog signaling pathway to be a direct or indirect target of these compounds. These botanicals at pharmacologic concentrations are potentially safer and less expensive alternatives to cyclopamine and its pharmaceutical analogues for cancer therapy.
Collapse
Affiliation(s)
- Anna Slusarz
- Department of Biochemistry, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | | | | | | | | | |
Collapse
|
705
|
|
706
|
Mas C, Ruiz i Altaba A. Small molecule modulation of HH-GLI signaling: current leads, trials and tribulations. Biochem Pharmacol 2010; 80:712-23. [PMID: 20412786 DOI: 10.1016/j.bcp.2010.04.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 04/12/2010] [Accepted: 04/13/2010] [Indexed: 01/07/2023]
Abstract
Many human sporadic cancers have been recently shown to require the activity of the Hedgehog-GLI pathway for sustained growth. The survival and expansion of cancer stem cells is also HH-GLI dependent. Here we review the advances on the modulation of HH-GLI signaling by small molecules. We focus on both natural compounds and synthetic molecules that target upstream pathway components, mostly SMOOTHENED, and those that target the last steps of the pathway, the GLI transcription factors. In this review we have sought to provide some bases for useful comparisons, listing original assays used and sources to facilitate comparisons of IC50 values. This area is a rapidly expanding field where biology, medicine and chemistry intersect, both in academia and industry. We also highlight current clinical trials, with positive results in early stages. While we have tried to be exhaustive regarding the molecules, not all data is in the public domain yet. Indeed, we have opted to avoid listing chemical structures but these can be easily found in the references given. Finally, we are hopeful that the best molecules will soon reach the patients but caution about the lack of investment on compounds that lack tight IP positions. While the market in developed nations is expected to compensate the investment and risk of making HH-GLI modulators, other sources or plans must be available for developing nations and poor patient populations. The promise of curing cancer recalls the once revered dream of El Dorado, which taught us that not everything that GLI-tters is gold.
Collapse
Affiliation(s)
- Christophe Mas
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1 rue Michel Servet, CH-1211 Geneva, Switzerland.
| | | |
Collapse
|
707
|
Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS, Fereshteh M, Reya T, Liu JO, Epstein EH, Stevens DA, Beachy PA. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell 2010; 17:388-99. [PMID: 20385363 PMCID: PMC4039177 DOI: 10.1016/j.ccr.2010.02.027] [Citation(s) in RCA: 409] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 12/03/2009] [Accepted: 02/12/2010] [Indexed: 12/21/2022]
Abstract
In a screen of drugs previously tested in humans we identified itraconazole, a systemic antifungal, as a potent antagonist of the Hedgehog (Hh) signaling pathway that acts by a mechanism distinct from its inhibitory effect on fungal sterol biosynthesis. Systemically administered itraconazole, like other Hh pathway antagonists, can suppress Hh pathway activity and the growth of medulloblastoma in a mouse allograft model and does so at serum levels comparable to those in patients undergoing antifungal therapy. Mechanistically, itraconazole appears to act on the essential Hh pathway component Smoothened (SMO) by a mechanism distinct from that of cyclopamine and other known SMO antagonists, and prevents the ciliary accumulation of SMO normally caused by Hh stimulation.
Collapse
Affiliation(s)
- James Kim
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Jean Y Tang
- Department of Dermatology, Stanford University, Stanford, CA 94305, USA
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Ruoyu Gong
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Jynho Kim
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - John J. Lee
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Karl V. Clemons
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA 94305, USA
- California Institute for Medical Research, San Jose, CA 95128, USA
| | - Curtis R. Chong
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- The Johns Hopkins Clinical Compound Screening Initiative, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Kris S. Chang
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Mark Fereshteh
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Tannishtha Reya
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- The Johns Hopkins Clinical Compound Screening Initiative, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Ervin H. Epstein
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - David A. Stevens
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA 94305, USA
- California Institute for Medical Research, San Jose, CA 95128, USA
| | - Philip A. Beachy
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
- Institute for Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
708
|
Raaijmakers MHGP, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, Ebert BL, Al-Shahrour F, Hasserjian RP, Scadden EO, Aung Z, Matza M, Merkenschlager M, Lin C, Rommens JM, Scadden DT. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 2010; 464:852-7. [PMID: 20305640 PMCID: PMC3422863 DOI: 10.1038/nature08851] [Citation(s) in RCA: 842] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 01/19/2010] [Indexed: 12/17/2022]
Abstract
Mesenchymal cells contribute to the 'stroma' of most normal and malignant tissues, with specific mesenchymal cells participating in the regulatory niches of stem cells. By examining how mesenchymal osteolineage cells modulate haematopoiesis, here we show that deletion of Dicer1 specifically in mouse osteoprogenitors, but not in mature osteoblasts, disrupts the integrity of haematopoiesis. Myelodysplasia resulted and acute myelogenous leukaemia emerged that had acquired several genetic abnormalities while having intact Dicer1. Examining gene expression altered in osteoprogenitors as a result of Dicer1 deletion showed reduced expression of Sbds, the gene mutated in Schwachman-Bodian-Diamond syndrome-a human bone marrow failure and leukaemia pre-disposition condition. Deletion of Sbds in mouse osteoprogenitors induced bone marrow dysfunction with myelodysplasia. Therefore, perturbation of specific mesenchymal subsets of stromal cells can disorder differentiation, proliferation and apoptosis of heterologous cells, and disrupt tissue homeostasis. Furthermore, primary stromal dysfunction can result in secondary neoplastic disease, supporting the concept of niche-induced oncogenesis.
Collapse
Affiliation(s)
- Marc H G P Raaijmakers
- Center for Regenerative Medicine, Massachusetts General Hospital and Harvard Medical School CPZN, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
709
|
Hedgehog regulates distinct vascular patterning events through VEGF-dependent and -independent mechanisms. Blood 2010; 116:653-60. [PMID: 20339091 DOI: 10.1182/blood-2009-12-256644] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Despite the clear importance of Hedgehog (Hh) signaling in blood vascular development as shown by genetic analysis, its mechanism of action is still uncertain. To better understand the role of Hh in vascular development, we further characterized its roles in vascular development in mouse embryos and examined its interaction with vascular endothelial growth factor (VEGF), a well-known signaling pathway essential to blood vascular development. We found that VEGF expression in the mouse embryo depended on Hh signaling, and by using genetic rescue approaches, we demonstrated that the role of Hh both in endothelial tube formation and Notch-dependent arterial identity was solely dependent on its regulation of VEGF. In contrast, overactivation of the Hh pathway through deletion of Patched1 (Ptch1), a negative regulator of Hh signaling, resulted in reduced vascular density and increased Delta-like ligand 4 expression. The Ptch1 phenotype was independent of VEGF pathway dysregulation and was not rescued when Delta-like ligand 4 levels were restored to normal. These findings establish that Hh uses both VEGF- and Notch-dependent and -independent mechanisms to pattern specific events in early blood vascular development.
Collapse
|
710
|
ABC Transporters as Molecular Effectors of Pancreatic Oncogenic Pathways: The Hedgehog-GLI Model. J Gastrointest Cancer 2010; 41:153-8. [DOI: 10.1007/s12029-010-9144-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
711
|
Lin Y, Huang G, Huang Y, Tzeng TRJ, Chrisey DB. Process-Induced Cell Injury in Laser Direct Writing of Human Colon Cancer Cells. Tissue Eng Part C Methods 2010:110525193859077. [PMID: 20151815 DOI: 10.1089/ten.tec.2009.0606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Matrix-assisted pulsed-laser evaporation direct-write has emerged as a promising technique for biological construct fabrication. The posttransfer cell viability in matrix-assisted pulsed-laser evaporation direct-write depends on various operating conditions such as the applied laser fluence. To date, the effects of operating conditions such as laser fluence, direct-writing height, and cell density on the posttransfer cell viability have not been well elucidated. This study investigates the effects of operating conditions on the posttransfer cell viability in laser direct writing of human colon cancer HT-29 cells. It has been observed that (1) the HT-29 cell viability decreases from 95% to 78% as the laser fluence increases from 258 to 1482 mJ/cm(2), and the posttransfer cell proliferation capacity does not vary significantly as the laser fluence changes; (2) the direct-writing height does not have noticeable effect on the posttransfer cell viability under low laser fluences (258 and 869 mJ/cm(2)). However, a larger height (such as 29.3 mm) led to an almost 8% viability improvement compared with that of 16.6 mm under a high laser fluence (1482 mJ/cm(2)); and (3) the posttransfer cell viability is not dependent on the cell density for a range from 1 × 10(6) to 1 × 10(7) cells/mL.
Collapse
Affiliation(s)
- Yafu Lin
- 1 Department of Mechanical Engineering, Clemson University , Clemson, South Carolina
| | | | | | | | | |
Collapse
|
712
|
Saqui-Salces M, Merchant JL. Hedgehog signaling and gastrointestinal cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:786-95. [PMID: 20307590 DOI: 10.1016/j.bbamcr.2010.03.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 03/12/2010] [Accepted: 03/15/2010] [Indexed: 12/23/2022]
Abstract
Hedgehog (Hh) signaling is critical for embryonic development and in differentiation, proliferation, and maintenance of multiple adult tissues. De-regulation of the Hh pathway is associated with birth defects and cancer. In the gastrointestinal tract, Hh ligands Sonic (Shh) and Indian (Ihh), as well as the receptor Patched (Ptch1), and transcription factors of Glioblastoma family (Gli) are all expressed during development. In the adult, Shh expression is restricted to the stomach and colon, while Ihh expression occurs throughout the luminal gastrointestinal tract, its expression being highest in the proximal duodenum. Several studies have demonstrated a requirement for Hh signaling during gastrointestinal tract development. However to date, the specific role of the Hh pathway in the adult stomach and intestine is not completely understood. The current review will place into context the implications of recent published data related to the biochemistry and cell biology of Hh signaling on the luminal gastrointestinal tract during development, normal physiology and subsequently carcinogenesis.
Collapse
Affiliation(s)
- Milena Saqui-Salces
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
713
|
Saito RA, Micke P, Paulsson J, Augsten M, Peña C, Jönsson P, Botling J, Edlund K, Johansson L, Carlsson P, Jirström K, Miyazono K, Ostman A. Forkhead box F1 regulates tumor-promoting properties of cancer-associated fibroblasts in lung cancer. Cancer Res 2010; 70:2644-54. [PMID: 20233876 DOI: 10.1158/0008-5472.can-09-3644] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer-associated fibroblasts (CAF) attract increasing attention as potential cancer drug targets due to their ability to stimulate, for example, tumor growth, invasion, angiogenesis, and metastasis. However, the molecular mechanisms causing the tumor-promoting properties of CAFs remain poorly understood. Forkhead box F1 (FoxF1) is a mesenchymal target of hedgehog signaling, known to regulate mesenchymal-epithelial interactions during lung development. Studies with FoxF1 gain- and loss-of-function fibroblasts revealed that FoxF1 regulates the contractility of fibroblasts, their production of hepatocyte growth factor and fibroblast growth factor-2, and their stimulation of lung cancer cell migration. FoxF1 status of fibroblasts was also shown to control the ability of fibroblasts to stimulate xenografted tumor growth. FoxF1 was expressed in CAFs of human lung cancer and associated with activation of hedgehog signaling. These observations suggest that hedgehog-dependent FoxF1 is a clinically relevant lung CAF-inducing factor, and support experimentally the general concept that CAF properties can be induced by activation of developmentally important transcription factors.
Collapse
Affiliation(s)
- Roy-Akira Saito
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
714
|
Walter K, Omura N, Hong SM, Griffith M, Vincent A, Borges M, Goggins M. Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clin Cancer Res 2010; 16:1781-9. [PMID: 20215540 PMCID: PMC2846609 DOI: 10.1158/1078-0432.ccr-09-1913] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Accumulating evidence suggests that cancer-associated stromal fibroblasts (CAF) contribute to tumor growth by actively communicating with cancer cells. Our aim is to identify signaling pathways involved in tumor-stromal cell interactions in human pancreatic cancer. EXPERIMENTAL DESIGN We established primary fibroblast cultures from human pancreatic adenocarcinomas and nonneoplastic pancreas tissues. To identify differentially expressed genes in CAFs, we did gene expression profiling of human pancreatic CAFs and nonneoplastic pancreatic fibroblasts. RESULTS The Hedgehog receptor Smoothened (SMO) was upregulated in CAFs relative to control fibroblasts. CAFs expressing SMO could transduce the Sonic hedgehog signal to activate Gli1 expression, and small interfering RNA knockdown of SMO blocked the induction of Gli1 in these cells. Stromal fibroblasts of human primary pancreatic adenocarcinomas overexpressed Smo compared with normal pancreatic fibroblasts. CONCLUSIONS These findings implicate overexpression of Smo as a mechanism for the activation of Hedgehog signaling in human pancreatic CAFs and suggest that stromal cells may be a therapeutic target for Smo antagonists in pancreatic cancer.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line
- Female
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- Immunoenzyme Techniques
- Male
- Middle Aged
- Oligonucleotide Array Sequence Analysis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Smoothened Receptor
- Stromal Cells/metabolism
- Stromal Cells/pathology
- Tissue Array Analysis
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Zinc Finger Protein GLI1
Collapse
Affiliation(s)
- Kimberly Walter
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Noriyuki Omura
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Seung-Mo Hong
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Margaret Griffith
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Audrey Vincent
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
715
|
Abstract
The discovery of a Sonic Hedgehog (Shh) signaling pathway in the mature vertebrate CNS has paved the way to the characterization of the functional roles of Shh signals in normal and diseased brain. Shh is proposed to participate in the establishment and maintenance of adult neurogenic niches and to regulate the proliferation of neuronal or glial precursors in several brain areas. Consistent with its role during brain development, misregulation of Shh signaling is associated with tumorigenesis while its recruitement in damaged neural tissue might be part of the regenerating process. This review focuses on the most recent data of the Hedgehog pathway in the adult brain and its relevance as a novel therapeutic approach for brain diseases including brain tumors.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- CNRS, Alfred Fessard Institute of Neurobiology, Laboratory of Neurobiology and Development, UPR-3294, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France.
| | | | | |
Collapse
|
716
|
Varnat F, Duquet A, Malerba M, Zbinden M, Mas C, Gervaz P, Ruiz i Altaba A. Human colon cancer epithelial cells harbour active HEDGEHOG-GLI signalling that is essential for tumour growth, recurrence, metastasis and stem cell survival and expansion. EMBO Mol Med 2010; 1:338-51. [PMID: 20049737 PMCID: PMC3378144 DOI: 10.1002/emmm.200900039] [Citation(s) in RCA: 373] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human colon cancers often start as benign adenomas through loss of APC, leading to enhanced βCATENIN (βCAT)/TCF function. These early lesions are efficiently managed but often progress to invasive carcinomas and incurable metastases through additional changes, the nature of which is unclear. We find that epithelial cells of human colon carcinomas (CCs) and their stem cells of all stages harbour an active HH-GLI pathway. Unexpectedly, they acquire a high HEDGEHOG-GLI (HH-GLI) signature coincident with the development of metastases. We show that the growth of CC xenografts, their recurrence and metastases require HH-GLI function, which induces a robust epithelial-to-mesenchymal transition (EMT). Moreover, using a novel tumour cell competition assay we show that the self-renewal of CC stem cells in vivo relies on HH-GLI activity. Our results indicate a key and essential role of the HH-GLI1 pathway in promoting CC growth, stem cell self-renewal and metastatic behavior in advanced cancers. Targeting HH-GLI1, directly or indirectly, is thus predicted to decrease tumour bulk and eradicate CC stem cells and metastases.
Collapse
Affiliation(s)
- Frédéric Varnat
- Department Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
717
|
A digest on the role of the tumor microenvironment in gastrointestinal cancers. CANCER MICROENVIRONMENT 2010; 3:167-76. [PMID: 21209782 DOI: 10.1007/s12307-010-0040-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 01/27/2010] [Indexed: 12/12/2022]
Abstract
Experimental studies and analyses of clinical material have convincingly demonstrated that tumor formation and progression occurs through a concerted action of malignant cells and the surrounding microenvironment of the tumor stroma. The tumor microenvironment is comprised of various cell types like fibroblasts, immune cells, vascular cells and bone-marrow-derived cells embedded in the extracellular matrix. This review, focusing on recent findings in the context of gastrointestinal tumors, introduces the different stromal cell types and delineates their contributions to cancer initiation, growth and metastasis. By selected examples we also present how the tumor microenvironment is emerging as a promising target for therapeutic intervention.
Collapse
|
718
|
Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res 2010; 316:1324-31. [PMID: 20211171 DOI: 10.1016/j.yexcr.2010.02.045] [Citation(s) in RCA: 855] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 02/28/2010] [Indexed: 01/08/2023]
Abstract
Ten years ago, Hanahan and Weinberg delineated six "Hallmarks of cancer" which summarize several decades of intense cancer research. However, tumor cells do not act in isolation, but rather subsist in a rich microenvironment provided by resident fibroblasts, endothelial cells, pericytes, leukocytes, and extra-cellular matrix. It is increasingly appreciated that the tumor stroma is an integral part of cancer initiation, growth and progression. The stromal elements of tumors hold prognostic, as well as response-predictive, information, and abundant targeting opportunities within the tumor microenvironment are continually identified. Herein we review the current understanding of tumor cell interactions with the tumor stroma with a particular focus on cancer-associated fibroblasts and pericytes. Moreover, we discuss emerging fields of research which need to be further explored in order to fulfil the promise of stroma-targeted therapies for cancer.
Collapse
Affiliation(s)
- Kristian Pietras
- Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Scheeles väg 2, SE-171 77 Stockholm.
| | | |
Collapse
|
719
|
Singh RR, Kim JE, Davuluri Y, Drakos E, Cho-Vega JH, Amin HM, Vega F. Hedgehog signaling pathway is activated in diffuse large B-cell lymphoma and contributes to tumor cell survival and proliferation. Leukemia 2010; 24:1025-36. [PMID: 20200556 DOI: 10.1038/leu.2010.35] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hedgehog (HH) signaling is important in the pathogenesis of several malignancies. Recently, we described that HH signaling proteins are commonly expressed in diffuse large B-cell lymphoma (DLBCL); however, the functional role of HH pathway in DLBCL has not been explored. Here, we assessed the possibility that HH pathway activation contributes to the survival of DLBCL. We found that HH signaling inhibition induces predominantly cell-cycle arrest in DLBCL cells of germinal center (GC) B-cell type, and apoptosis in DLBCL cells of activated B-cell (ABC) type. Apoptosis after HH signaling inhibition in DLBCL cells of ABC type was associated with downregulation of BCL2; however HH inhibition was not associated with BCL2 downregulation in DLBCL of GC type. Functional inhibition of BCL2 significantly increased apoptosis induced by HH inhibition in DLBCL cells of both types. We also showed that DLBCL cells synthesize, secrete and respond to endogenous HH ligands, providing support for the existence of an autocrine HH signaling loop. Our findings provide novel evidence that dysregulation of HH pathway is involved in the biology of DLBCL and have significant therapeutic implications as they identify HH signaling as a potential therapeutic target in DLBCL, in particular for those lymphomas expressing the HH receptor smoothened.
Collapse
Affiliation(s)
- R R Singh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
720
|
STROBEL OLIVER, ROSOW DAVIDE, RAKHLIN ELENAY, LAUWERS GREGORYY, TRAINOR AMANDAG, ALSINA JANIVETTE, CASTILLO CARLOSFERNÁNDEZDEL, WARSHAW ANDREWL, THAYER SARAHP. Pancreatic duct glands are distinct ductal compartments that react to chronic injury and mediate Shh-induced metaplasia. Gastroenterology 2010; 138:1166-77. [PMID: 20026066 PMCID: PMC3806111 DOI: 10.1053/j.gastro.2009.12.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 10/25/2009] [Accepted: 12/02/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Pancreatic intraepithelial neoplasia (PanIN) are pancreatic cancer precursor lesions of unclear origin and significance. PanIN aberrantly express sonic hedgehog (Shh), an initiator of pancreatic cancer, and gastrointestinal mucins. A majority of PanIN are thought to arise from ducts. We identified a novel ductal compartment that is gathered in gland-like outpouches (pancreatic duct glands [PDG]) of major ducts and characterized its role in injury and metaplasia. METHODS The ductal system was analyzed in normal pancreata and chronic pancreatitis in humans and mice. Anatomy was assessed by serial hematoxylin and eosin sections and scanning electron microscopy of corrosion casts. Expression of mucins and developmental genes and proliferation were assessed by immunohistochemistry or real-time quantitative polymerase chain reaction. Effects of Shh on ductal cells were investigated by exposure to Shh in vitro and transgenic misexpression in vivo. RESULTS Three-dimensional analysis revealed blind-ending outpouches of ducts in murine and human pancreata. These PDG are morphologically and molecularly distinct from normal ducts; even in normal pancreata they display PanIN and metaplastic features, such as expression of Shh and gastric mucins. They express other developmental genes, such as Pdx-1 and Hes-1. In injury, Shh is up-regulated along with gastric mucins. Expansion of the PDG compartment results in a mucinous metaplasia. Shh promotes this transformation in vitro and in vivo. CONCLUSIONS PDG are distinct gland-like mucinous compartments with a distinct molecular signature. In response to injury, PDG undergo an Shh-mediated mucinous gastrointestinal metaplasia with PanIN-like features. PDG may provide a link between Shh, mucinous metaplasia, and neoplasia.
Collapse
Affiliation(s)
- OLIVER STROBEL
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts,Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - DAVID E. ROSOW
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - ELENA Y. RAKHLIN
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - GREGORY Y. LAUWERS
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - AMANDA G. TRAINOR
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - JANIVETTE ALSINA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - ANDREW L. WARSHAW
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - SARAH P. THAYER
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
721
|
Fei DL, Li H, Kozul CD, Black KE, Singh S, Gosse JA, DiRenzo J, Martin KA, Wang B, Hamilton JW, Karagas MR, Robbins DJ. Activation of Hedgehog signaling by the environmental toxicant arsenic may contribute to the etiology of arsenic-induced tumors. Cancer Res 2010; 70:1981-8. [PMID: 20179202 PMCID: PMC2831120 DOI: 10.1158/0008-5472.can-09-2898] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exposure to the environmental toxicant arsenic, through both contaminated water and food, contributes to significant health problems worldwide. In particular, arsenic exposure is thought to function as a carcinogen for lung, skin, and bladder cancer via mechanisms that remain largely unknown. More recently, the Hedgehog signaling pathway has also been implicated in the progression and maintenance of these same cancers. Based on these similarities, we tested the hypothesis that arsenic may act in part through activating Hedgehog signaling. Here, we show that arsenic is able to activate Hedgehog signaling in several primary and established tissue culture cells as well as in vivo. Arsenic activates Hedgehog signaling by decreasing the stability of the repressor form of GLI3, one of the transcription factors that ultimately regulate Hedgehog activity. We also show, using tumor samples from a cohort of bladder cancer patients, that high levels of arsenic exposure are associated with high levels of Hedgehog activity. Given the important role Hedgehog signaling plays in the maintenance and progression of a variety of tumors, including bladder cancer, these results suggest that arsenic exposure may in part promote cancer through the activation of Hedgehog signaling. Thus, we provide an important insight into the etiology of arsenic-induced human carcinogenesis, which may be relevant to millions of people exposed to high levels of arsenic worldwide.
Collapse
Affiliation(s)
- Dennis Liang Fei
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - Hua Li
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - Courtney D. Kozul
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - Kendall E. Black
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - Samer Singh
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - Julie A. Gosse
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - James DiRenzo
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
| | - Kathleen A. Martin
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
- Department of Surgery, Dartmouth Medical School, Hanover, NH 03755
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY 10021
| | - Joshua W. Hamilton
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
- Bay Paul Center in Comparative Molecular Biology & Evolution, Marine Biological Laboratory, Woods Hole MA 02543
| | - Margaret R. Karagas
- Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Dartmouth Medical School, Lebanon, NH 03756
| | - David J. Robbins
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755
- Norris Cotton Cancer Center, Lebanon, NH 03756
| |
Collapse
|
722
|
Affiliation(s)
- ANIRBAN MAITRA
- Departments of Pathology and Oncology, The Sol Goldman Pancreatic Cancer, Research Center, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| |
Collapse
|
723
|
Gao S, Wang Q, Huang LJS, Lum L, Chen C. Chemical and biological studies of nakiterpiosin and nakiterpiosinone. J Am Chem Soc 2010; 132:371-83. [PMID: 20000429 DOI: 10.1021/ja908626k] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nakiterpiosin and nakiterpiosinone are two related C-nor-D-homosteroids isolated from the sponge Terpios hoshinota that show promise as anticancer agents. We have previously described the asymmetric synthesis and revision of the relative configuration of nakiterpiosin. We now provide detailed information on the stereochemical analysis that supports our structure revision and the synthesis of the originally proposed and revised nakiterpiosin. In addition, we herein describe a refined approach for the synthesis of nakiterpiosin, the first synthesis of nakiterpiosinone, and preliminary mechanistic studies of nakiterpiosin's action in mammalian cells. Cells treated with nakiterpiosin exhibit compromised formation of the primary cilium, an organelle that functions as an assembly point for components of the Hedgehog signal transduction pathway. We provide evidence that the biological effects exhibited by nakiterpiosin are mechanistically distinct from those of well-established antimitotic agents such as taxol. Nakiterpiosin may be useful as an anticancer agent in those tumors resistant to existing antimitotic agents and those dependent on Hedgehog pathway responses for growth.
Collapse
Affiliation(s)
- Shuanhu Gao
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, USA
| | | | | | | | | |
Collapse
|
724
|
Kinetics of hedgehog-dependent full-length Gli3 accumulation in primary cilia and subsequent degradation. Mol Cell Biol 2010; 30:1910-22. [PMID: 20154143 DOI: 10.1128/mcb.01089-09] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hedgehog (Hh) signaling in vertebrates depends on intraflagellar transport (IFT) within primary cilia. The Hh receptor Patched is found in cilia in the absence of Hh and is replaced by the signal transducer Smoothened within an hour of Hh stimulation. By generating antibodies capable of detecting endogenous pathway transcription factors Gli2 and Gli3, we monitored their kinetics of accumulation in cilia upon Hh stimulation. Localization occurs within minutes of Hh addition, making it the fastest reported readout of pathway activity, which permits more precise temporal and spatial localization of Hh signaling events. We show that the species of Gli3 that accumulates at cilium tips is full-length and likely not protein kinase A phosphorylated. We also confirmed that phosphorylation and betaTrCP/Cul1 are required for endogenous Gli3 processing and that this is inhibited by Hh. Surprisingly, however, Hh-dependent inhibition of processing does not lead to accumulation of full-length Gli3, but instead renders it labile, leading to its proteasomal degradation via the SPOP/Cul3 complex. In fact, full-length Gli3 disappears with faster kinetics than the Gli3 repressor, the latter not requiring SPOP/Cul3 or betaTrCP/Cul1. This may contribute to the increased Gli3 activator/repressor ratios found in IFT mutants.
Collapse
|
725
|
Valin A, Avril MF, Magnaldo T. [Dermal fibroblasts exert a key influence in the development of basal-cell skin cancers: the model of Gorlin syndrome]. Med Sci (Paris) 2010; 26:22-5. [PMID: 20132767 DOI: 10.1051/medsci/201026122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
726
|
Determination of GDC-0449, a small-molecule inhibitor of the Hedgehog signaling pathway, in human plasma by solid phase extraction-liquid chromatographic-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878:785-90. [PMID: 20172765 DOI: 10.1016/j.jchromb.2010.01.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 01/20/2010] [Accepted: 01/24/2010] [Indexed: 11/24/2022]
Abstract
To support clinical development, a solid phase extraction (SPE) liquid chromatographic-tandem mass spectrometry (LC-MS/MS) method for the determination of GDC-0449 concentrations in human plasma has been developed and validated. Samples (200 microl) were extracted using an Oasis MCX 10 mg 96-well SPE plate and the resulting extracts were analyzed using reverse-phase chromatography coupled with a turbo-ionspray interface. The method was validated over calibration curve range 5-5000 ng/mL. Quadratic regression and 1/x(2) weighing were used. Within-run relative standard deviation (%RSD) was within 10.1% and accuracy ranged from 88.6% to 109.0% of nominal. Between-run %RSD was within 8.6% and accuracy ranged from 92.4% to 105.3% of nominal. Extraction recovery of GDC-0449 was between 88.3% and 91.2% as assessed using quality control sample concentrations. GDC-0449 was stable in plasma for 315 days when stored at -70 degrees C and stable in reconstituted sample extracts for 117 h when stored at room temperature. Quantitative matrix effect/ion suppression experiment was performed and no significant matrix ion suppression was observed. This assay allows for the determination of GDC-0449 plasma concentrations over a sufficient time period to determine pharmacokinetic parameters at relevant clinical doses.
Collapse
|
727
|
Agarwal JR, Matsui W. Multiple myeloma: a paradigm for translation of the cancer stem cell hypothesis. Anticancer Agents Med Chem 2010; 10:116-20. [PMID: 20184542 PMCID: PMC3033115 DOI: 10.2174/187152010790909344] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 12/15/2009] [Indexed: 01/28/2023]
Abstract
Despite recent advances in drug development, multiple myeloma (MM) remains incurable for the majority of patients due to relapse and disease progression. The cancer stem cell (CSC) hypothesis may provide an explanation for these clinical findings. It suggests that the long-term proliferative potential responsible for disease initiation, maintenance, and relapse is contained within specific subpopulations of biologically distinct tumor cells. Data in MM suggest that CSCs represent a rare cell population phenotypically resembling normal memory B cells. Compared to MM plasma cells, MM CSCs also appear to be relatively resistant to a wide variety of standard anti-cancer agents suggesting they may persist following treatment and mediate tumor re-growth and relapse. A unique property CSCs share with their normal counterparts is the potential for self-renewal that likely maintains the malignant clone over time. The development of therapeutic strategies targeting the signaling elements contributing to cancer cell self-renewal has been limited primarily because the cellular processes involved are poorly understood. However, it is common that the signaling pathway components regulating normal stem cell self-renewal are aberrantly activated in human cancers and may serve as potential therapeutic targets. One class of shared regulatory pathways are those active during normal embryonic patterning and organ formation such as Hedgehog (Hh), Notch and Wingless (Wnt), and emerging data suggest that these may play a role in CSCs. Here we review the identification and characterization of MM CSCs, the role of Hh in MM, and issues to be considered during the early clinical testing of CSC targeting agents.
Collapse
Affiliation(s)
- Jasmin Roya Agarwal
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William Matsui
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
728
|
Nakamura K, Sasajima J, Mizukami Y, Sugiyama Y, Yamazaki M, Fujii R, Kawamoto T, Koizumi K, Sato K, Fujiya M, Sasaki K, Tanno S, Okumura T, Shimizu N, Kawabe JI, Karasaki H, Kono T, Ii M, Bardeesy N, Chung DC, Kohgo Y. Hedgehog promotes neovascularization in pancreatic cancers by regulating Ang-1 and IGF-1 expression in bone-marrow derived pro-angiogenic cells. PLoS One 2010; 5:e8824. [PMID: 20098680 PMCID: PMC2809097 DOI: 10.1371/journal.pone.0008824] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2009] [Accepted: 01/01/2010] [Indexed: 01/11/2023] Open
Abstract
Background The hedgehog (Hh) pathway has been implicated in the pathogenesis of cancer including pancreatic ductal adenocarcinoma (PDAC). Recent studies have suggested that the oncogenic function of Hh in PDAC involves signaling in the stromal cells rather than cell autonomous effects on the tumor cells. However, the origin and nature of the stromal cell type(s) that are responsive to Hh signaling remained unknown. Since Hh signaling plays a crucial role during embryonic and postnatal vasculogenesis, we speculated that Hh ligand may act on tumor vasculature specifically focusing on bone marrow (BM)-derived cells. Methodology/Principal Findings Cyclopamine was utilized to inhibit the Hh pathway in human PDAC cell lines and their xenografts. BM transplants, co-culture systems of tumor cells and BM-derived pro-angiogenic cells (BMPCs) were employed to assess the role of tumor-derived Hh in regulating the BM compartment and the contribution of BM-derived cells to angiogenesis in PDAC. Cyclopamine administration attenuated Hh signaling in the stroma rather than in the cancer cells as reflected by decreased expression of full length Gli2 protein and Gli1 mRNA specifically in the compartment. Cyclopamine inhibited the growth of PDAC xenografts in association with regression of the tumor vasculature and reduced homing of BM-derived cells to the tumor. Host-derived Ang-1 and IGF-1 mRNA levels were downregulated by cyclopamine in the tumor xenografts. In vitro co-culture and matrigel plug assays demonstrated that PDAC cell-derived Shh induced Ang-1 and IGF-1 production in BMPCs, resulting in their enhanced migration and capillary morphogenesis activity. Conclusions/Significance We identified the BMPCs as alternative stromal targets of Hh-ligand in PDAC suggesting that the tumor vasculature is an attractive therapeutic target of Hh blockade. Our data is consistent with the emerging concept that BM-derived cells make important contributions to epithelial tumorigenesis.
Collapse
Affiliation(s)
- Kazumasa Nakamura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Junpei Sasajima
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Yusuke Mizukami
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
- * E-mail:
| | - Yoshiaki Sugiyama
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Madoka Yamazaki
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Rie Fujii
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Toru Kawamoto
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Kazuya Koizumi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Kazuya Sato
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Katsunori Sasaki
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Satoshi Tanno
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
- Department of General Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Toshikatsu Okumura
- Department of General Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Norihiko Shimizu
- Department of Animal Facility, Asahikawa Medical College, Asahikawa, Japan
| | - Jun-ichi Kawabe
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical College, Asahikawa, Japan
| | - Hidenori Karasaki
- Division of Gastroenterological and General Surgery, Department of Surgery, Asahikawa Medical College, Asahikawa, Japan
| | - Toru Kono
- Division of Gastroenterological and General Surgery, Department of Surgery, Asahikawa Medical College, Asahikawa, Japan
| | - Masaaki Ii
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel C. Chung
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, United States of America
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yutaka Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| |
Collapse
|
729
|
Stecca B, Ruiz i Altaba A. Context-dependent regulation of the GLI code in cancer by HEDGEHOG and non-HEDGEHOG signals. J Mol Cell Biol 2010; 2:84-95. [PMID: 20083481 DOI: 10.1093/jmcb/mjp052] [Citation(s) in RCA: 204] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A surprisingly large and unrelated number of human tumors depend on sustained HEDGEHOG-GLI (HH-GLI) signaling for growth. This includes cancers of the skin, brain, colon, lungs, prostate, blood and pancreas among others. The basis of such commonality is not obvious. HH-GLI signaling has also been shown to be active in and required for cancer stem cell survival and expansion in different cancer types, and its activity is essential not only for tumor growth but also for recurrence and metastatic growth, two key medical problems. Here we review recent data on the role of HH-GLI signaling in cancer focusing on the role of the GLI code, the regulated combinatorial and cooperative function of repressive and activating forms of all Gli transcription factors, as a signaling nexus that integrates not only HH signals but also those of multiple tumor suppressors and oncogenes. Recent data support the view that the context-dependent regulation of the GLI code by oncogenes and tumor suppressors constitutes a basis for the widespread involvement of GLI1 in human cancers, representing a perversion of its normal role in the control of stem cell lineages during normal development and homeostasis.
Collapse
Affiliation(s)
- Barbara Stecca
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva CH-1211, Switzerland
| | | |
Collapse
|
730
|
Firestone AJ, Chen JK. Controlling destiny through chemistry: small-molecule regulators of cell fate. ACS Chem Biol 2010; 5:15-34. [PMID: 20000447 PMCID: PMC2807212 DOI: 10.1021/cb900249y] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
Controlling cell fate is essential for embryonic development, tissue regeneration, and the prevention of human disease. With each cell in the human body sharing a common genome, achieving the appropriate spectrum of stem cells and their differentiated lineages requires the selective activation of developmental signaling pathways, the expression of specific target genes, and the maintenance of these cellular states through epigenetic mechanisms. Small molecules that target these regulatory processes are therefore valuable tools for probing and manipulating the molecular mechanisms by which stem cells self-renew, differentiate, and arise from somatic cell reprogramming. Pharmacological modulators of cell fate could also help remediate human diseases caused by dysregulated cell proliferation or differentiation, heralding a new era in molecular therapeutics.
Collapse
Affiliation(s)
- Ari J. Firestone
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California 94305
| | - James K. Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
731
|
Hirotsu M, Setoguchi T, Sasaki H, Matsunoshita Y, Gao H, Nagao H, Kunigou O, Komiya S. Smoothened as a new therapeutic target for human osteosarcoma. Mol Cancer 2010; 9:5. [PMID: 20067614 PMCID: PMC2818696 DOI: 10.1186/1476-4598-9-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 01/12/2010] [Indexed: 12/26/2022] Open
Abstract
Background The Hedgehog signaling pathway functions as an organizer in embryonic development. Recent studies have demonstrated constitutive activation of Hedgehog pathway in various types of malignancies. However, it remains unclear how Hedgehog pathway is involved in the pathogenesis of osteosarcoma. To explore the involvement of aberrant Hedgehog pathway in the pathogenesis of osteosarcoma, we investigated the expression and activation of Hedgehog pathway in osteosarcoma and examined the effect of SMOOTHENED (SMO) inhibition. Results To evaluate the expression of genes of Hedgehog pathway, we performed real-time PCR and immunohistochemistry using osteosarcoma cell lines and osteosarcoma biopsy specimens. To evaluate the effect of SMO inhibition, we did cell viability, colony formation, cell cycle in vitro and xenograft model in vivo. Real-time PCR revealed that osteosarcoma cell lines over-expressed Sonic hedgehog, Indian hedgehog, PTCH1, SMO, and GLI. Real-time PCR revealed over-expression of SMO, PTCH1, and GLI2 in osteosarcoma biopsy specimens. These findings showed that Hedgehog pathway is activated in osteosarcomas. Inhibition of SMO by cyclopamine, a specific inhibitor of SMO, slowed the growth of osteosarcoma in vitro. Cell cycle analysis revealed that cyclopamine promoted G1 arrest. Cyclopamine reduced the expression of accelerators of the cell cycle including cyclin D1, cyclin E1, SKP2, and pRb. On the other hand, p21cip1 wprotein was up-regulated by cyclopamine treatment. In addition, knockdown of SMO by SMO shRNA prevents osteosarcoma growth in vitro and in vivo. Conclusions These findings suggest that inactivation of SMO may be a useful approach to the treatment of patients with osteosarcoma.
Collapse
Affiliation(s)
- Masataka Hirotsu
- Department of Orthopaedic Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
732
|
D'Angelo RC, Wicha MS. Stem cells in normal development and cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 95:113-58. [PMID: 21075331 DOI: 10.1016/b978-0-12-385071-3.00006-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this chapter we provide an overview of stem cells in normal tissues as well as in many different types of cancers. All tissues in the body are derived from organ-specific stem cells that retain the ability to self-renew and differentiate into specific cell types. The cancer stem cell hypothesis suggests that tumors arise from cell populations with dysregulated self-renewal. This may be tissue stem cells or more differentiated cells that acquire self-renewal capabilities. In addition, we outline some useful assays for purification and isolation of cancer stem cells including the dye exclusion side population assay, flow cytometry sorting techniques for identification of putative cancer stem cell markers, tumorspheres assay, aldehyde dehydrogenase activity assay, PKH, and other membrane staining used to label the cancer stem cells, as well as in vivo xenograft transplantation assays. We also examine some of the cell signaling pathways that regulate stem cell self-renewal including the Notch, Hedgehog, HER2/PI3K/Akt/PTEN, and p53 pathways. We also review information demonstrating the involvement of the microenvironment or stem cell niche and its effects on the growth and maintenance of cancer stem cells. Finally, we highlight the therapeutic implications of targeting stem cells by inhibiting these pathways for the treatment and prevention of cancer.
Collapse
Affiliation(s)
- Rosemarie Chirco D'Angelo
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
733
|
Abstract
The Hedgehog (Hh) signal transduction pathway is essential for the development and patterning of numerous organ systems, and has important roles in a variety of human cancers. Genetic screens for mouse embryonic patterning mutants first showed a connection between mammalian Hh signaling and intraflagellar transport (IFT), a process required for construction of the primary cilium, a small cellular projection found on most vertebrate cells. Additional genetic and cell biological studies have provided very strong evidence that mammalian Hh signaling depends on the primary cilium. Here, we review the evidence that defines the integral roles that IFT proteins and cilia play in the regulation of the Hh signal transduction pathway in vertebrates. We discuss the mechanisms that control localization of Hh pathway proteins to the cilium, focusing on the transmembrane protein Smoothened (Smo), which moves into the cilium in response to Hh ligand. The phenotypes caused by loss of cilia-associated proteins are complex, which suggests that cilia and IFT play active roles in mediating Hh signaling rather than serving simply as a compartment in which pathway components are concentrated. Hh signaling in Drosophila does not depend on cilia, but there appear to be ancient links between cilia and components of the Hh pathway that may reveal how this fundamental difference between the Drosophila and mammalian Hh pathways arose in evolution.
Collapse
Affiliation(s)
- Sarah C. Goetz
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065
| | - Polloneal J.R. Ocbina
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065
- Neuroscience Program, Weill Graduate School of Medical Sciences, Cornell University, New York, New York
| | - Kathryn V. Anderson
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065
| |
Collapse
|
734
|
Activation of hedgehog signaling is not a frequent event in ovarian cancers. Mol Cancer 2009; 8:112. [PMID: 19943941 PMCID: PMC2787497 DOI: 10.1186/1476-4598-8-112] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2009] [Accepted: 11/27/2009] [Indexed: 01/05/2023] Open
Abstract
The hedgehog (Hh) signaling pathway regulates many processes of development and tissue homeostasis. Activation of hedgehog signaling has been reported in about 30% of human cancer including ovarian cancer. Inhibition of hedgehog signaling has been pursued as an effective strategy for cancer treatment including an ongoing phase II clinical trial in ovarian cancer. However, the rate of hedgehog signaling activation in ovarian cancer was reported differently by different groups. To predict the successful for future clinical trials of hedgehog signaling inhibitors in ovarian cancer, we assessed hedgehog pathway activation in 34 ovarian epithelial tumor specimens through analyses of target gene expression by in-situ hybridization, immunohistochemistry, RT-PCR and real-time PCR. In contrast to previous reports, we only detected a small proportion of ovarian cancers with hedgehog target gene expression, suggesting that identification of the tumors with activated hedgehog signaling activation will facilitate chemotherapy with hedgehog signaling inhibitors.
Collapse
|
735
|
Activation of the hedgehog-signaling pathway in human cancer and the clinical implications. Oncogene 2009; 29:469-81. [PMID: 19935712 DOI: 10.1038/onc.2009.392] [Citation(s) in RCA: 253] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The hedgehog pathway, initially discovered by two Nobel laureates Drs E Wieschaus and C Nusslein-Volhard in Drosophila, is a major regulator for cell differentiation, tissue polarity and cell proliferation. Studies from many laboratories reveal activation of this pathway in a variety of human cancer, including basal cell carcinomas (BCCs), medulloblastomas, leukemia, gastrointestinal, lung, ovarian, breast and prostate cancers. It is thus believed that targeted inhibition of hedgehog signaling may be effective in treatment and prevention of human cancer. Even more exciting is the discovery and synthesis of specific signaling antagonists for the hedgehog pathway, which have significant clinical implications in novel cancer therapeutics. In this review, we will summarize major advances in the last 2 years in our understanding of hedgehog signaling activation in human cancer, interactions between hedgehog signaling and other pathways in carcinogenesis, potential antagonists for hedgehog signaling inhibition and their clinical implications for human cancer treatment.
Collapse
|
736
|
Translational advances and novel therapies for pancreatic ductal adenocarcinoma: hope or hype? Expert Rev Mol Med 2009; 11:e34. [PMID: 19919723 DOI: 10.1017/s1462399409001240] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biological complexity, inaccessible anatomical location, nonspecific symptoms, lack of a screening biomarker, advanced disease at presentation and drug resistance epitomise pancreatic ductal adenocarcinoma (PDA) as a poor-prognosis, lethal disease. Twenty-five years of research (basic, translational and clinical) have barely made strides to improve survival, mainly because of a fundamental lack of knowledge of the biological processes initiating and propagating PDA. However, isolation of pancreas cancer stem cells or progenitors, whole-genome sequencing for driver mutations, advances in functional imaging, mechanistic dissection of the desmoplastic reaction and novel targeted therapies are likely to shed light on how best to treat PDA. Here we summarise current knowledge and areas where the field is advancing, and give our opinion on the research direction the field should be focusing on to better deliver promising therapies for our patients.
Collapse
|
737
|
Cancer stem cells. J Thorac Oncol 2009; 4:S1079-81. [PMID: 19861925 DOI: 10.1097/01.jto.0000361758.17413.7c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
738
|
Noubissi FK, Goswami S, Sanek NA, Kawakami K, Minamoto T, Moser A, Grinblat Y, Spiegelman VS. Wnt signaling stimulates transcriptional outcome of the Hedgehog pathway by stabilizing GLI1 mRNA. Cancer Res 2009; 69:8572-8. [PMID: 19887615 DOI: 10.1158/0008-5472.can-09-1500] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wnt and Hedgehog signaling pathways play central roles in embryogenesis, stem cell maintenance, and tumorigenesis. However, the mechanisms by which these two pathways interact are not well understood. Here, we identified a novel mechanism by which Wnt signaling pathway stimulates the transcriptional output of Hedgehog signaling. Wnt/beta-catenin signaling induces expression of an RNA-binding protein, CRD-BP, which in turn binds and stabilizes GLI1 mRNA, causing an elevation of GLI1 expression and transcriptional activity. The newly described mode of regulation of GLI1 seems to be important to several functions of Wnt, including survival and proliferation of colorectal cancer cells.
Collapse
Affiliation(s)
- Felicite K Noubissi
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|
739
|
Tremblay MR, Nesler M, Weatherhead R, Castro AC. Recent patents for Hedgehog pathway inhibitors for the treatment of malignancy. Expert Opin Ther Pat 2009; 19:1039-56. [PMID: 19505195 DOI: 10.1517/13543770903008551] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND There is increasing evidence suggesting that blocking aberrant Hedgehog (Hh) signaling can be a novel therapeutic avenue for the treatment of cancer. During the past decade, efforts from academic and industrial groups have led to the discovery of a variety of Hh pathway inhibitors. OBJECTIVE This review covers the patent literature related to Hh pathway inhibitors for the treatment of proliferative diseases, regardless of their modes of action. METHODS A comprehensive survey of the patent literature since 1999 is presented. RESULTS/CONCLUSION Most reported Hh pathway inhibitors act on the key signaling transducer Smoothened (SMO). Screening of compound libraries using reporter and binding assays have identified a broad diversity of chemical structures that interact with SMO. These screening approaches, followed by conventional medicinal chemistry, have delivered important clinical drug candidates, such as GDC-0449 and XL-139. In addition, modification of the naturally occurring Veratrum alkaloid cyclopamine has resulted in various active analogues, including clinical drug candidate IPI-926. Although there are recent scientific literature reports of small molecules acting downstream of SMO, there is limited patent literature on this mode of Hh pathway inhibition.
Collapse
|
740
|
Stroma in breast development and disease. Semin Cell Dev Biol 2009; 21:11-8. [PMID: 19857593 DOI: 10.1016/j.semcdb.2009.10.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Accepted: 10/12/2009] [Indexed: 12/21/2022]
Abstract
It is increasingly apparent that normal and malignant breast tissues require complex local and systemic stromal interactions for development and progression. During development, mammary cell fate specification and differentiation require highly regulated contextual signals derived from the stroma. Likewise, during breast carcinoma development, the tissue stroma can provide tumor suppressing and tumor-promoting environments that serve to regulate neoplastic growth of the epithelium. This review focuses on the role of the stroma as a mediator of normal mammary development, as well as a critical regulator of malignant conversion and progression in breast cancer. Recognition of the important role of the stroma during the progression of breast cancers leads to the possibility of new targets for treatment of the initial breast cancer lesion as well as prevention of recurrence.
Collapse
|
741
|
Gonda TA, Varro A, Wang TC, Tycko B. Molecular biology of cancer-associated fibroblasts: can these cells be targeted in anti-cancer therapy? Semin Cell Dev Biol 2009; 21:2-10. [PMID: 19840860 DOI: 10.1016/j.semcdb.2009.10.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 09/30/2009] [Accepted: 10/06/2009] [Indexed: 12/17/2022]
Abstract
It is increasingly recognized that the non-neoplastic stromal compartment in most solid cancers plays an active role in tumor proliferation, invasion and metastasis. Cancer-associated fibroblasts (CAFs) are one of the most abundant cell types in the tumor stroma, and these cells are pro-tumorigenic. Evidence that CAFs are epigenetically and possibly also genetically distinct from normal fibroblasts is beginning to define these cells as potential targets of anti-cancer therapy. Here, we review the cell-of-origin and molecular biology of CAFs, arguing that such knowledge provides a rational basis for designing therapeutic strategies to coordinately and synergistically target both the stromal and malignant epithelial component of human cancers.
Collapse
Affiliation(s)
- Tamas A Gonda
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, United States.
| | | | | | | |
Collapse
|
742
|
Zhou BBS, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov 2009; 8:806-23. [PMID: 19794444 DOI: 10.1038/nrd2137] [Citation(s) in RCA: 651] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The hypothesis that cancer is driven by tumour-initiating cells (popularly known as cancer stem cells) has recently attracted a great deal of attention, owing to the promise of a novel cellular target for the treatment of haematopoietic and solid malignancies. Furthermore, it seems that tumour-initiating cells might be resistant to many conventional cancer therapies, which might explain the limitations of these agents in curing human malignancies. Although much work is still needed to identify and characterize tumour-initiating cells, efforts are now being directed towards identifying therapeutic strategies that could target these cells. This Review considers recent advances in the cancer stem cell field, focusing on the challenges and opportunities for anticancer drug discovery.
Collapse
Affiliation(s)
- Bin-Bing S Zhou
- Oncology Discovery, Wyeth Research, 401 North Middletown Road, Pearl River, New York 10965, USA.
| | | | | | | | | | | |
Collapse
|
743
|
O'Toole SA, Swarbrick A, Sutherland RL. The Hedgehog signalling pathway as a therapeutic target in early breast cancer development. Expert Opin Ther Targets 2009; 13:1095-103. [PMID: 19659449 DOI: 10.1517/14728220903130612] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The Hedgehog (Hh) signalling pathway is a highly conserved developmental pathway, which plays critical roles in patterning of the embryo through epithelial to mesenchymal signalling and the maintenance of stem cells in the adult organism. There is increasing evidence that this pathway is dysregulated in many malignancies, including breast cancer. While there has been a significant decrease in mortality from breast cancer, a number of treatment challenges remain, particularly in those tumours which develop resistance to endocrine-based therapy, or which lack expression of hormone or c-erbB2/HER2 receptors. Therapeutic manipulation of the Hh pathway as a potential cancer therapy is attracting great interest, with preclinical studies and clinical trials underway in a range of malignancies. This review highlights important recent developments that affect the potential of the Hh pathway as a novel therapeutic target in early breast cancer.
Collapse
Affiliation(s)
- Sandra A O'Toole
- Garvan Institute of Medical Research, Cancer Research Program, Darlinghurst 2010, NSW, Australia
| | | | | |
Collapse
|
744
|
Bailey JM, Mohr AM, Hollingsworth MA. Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer. Oncogene 2009; 28:3513-25. [PMID: 19633682 PMCID: PMC2910592 DOI: 10.1038/onc.2009.220] [Citation(s) in RCA: 248] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Revised: 05/03/2009] [Accepted: 05/03/2009] [Indexed: 12/24/2022]
Abstract
Sonic hedgehog (SHH) expression is tightly regulated throughout development. In the adult, aberrant expression of SHH is associated with the onset and progression of pancreatic cancer, as evidenced by increased levels of expression in premalignant and malignant lesions of the pancreas. We investigated the hypothesis that SHH, secreted from pancreatic tumors, functions in a paracrine manner to influence the biological condition of mesenchymal and endothelial cells. Orthotopic implantation of a pancreatic tumor cell line expressing SHH (Capan-2) and a transformed primary cell line that overexpresses SHH (T-HPNE.SHH) were used to show that overexpression of SHH increased primary tumor size and metastasis. Treatment with a neutralizing antibody, 5E1, decreased primary tumor volume and inhibited metastasis. Lyve-1+ vessels and stromal fibroblasts in tumors expressed primary cilium and showed localization of the receptor Smoothened to the primary cilium, providing evidence of active SHH signaling through this pathway. Although primary cilia are present on normal ductal cells of the pancreas, we did not observe primary cilium on the ductal tumor cells, suggesting decreased autocrine signaling through pathways mediated by the primary cilium in pancreatic cancer. These data support the hypothesis that SHH, secreted from pancreatic epithelia, is critical in establishing and regulating the tumor microenvironment and thereby contributes to progression of pancreatic cancer.
Collapse
Affiliation(s)
- Jennifer M. Bailey
- Eppley Institute, University of Nebraska Medical Center. Omaha, NE 68198-6805
| | - Ashley M. Mohr
- Eppley Institute, University of Nebraska Medical Center. Omaha, NE 68198-6805
| | | |
Collapse
|
745
|
Hedgehog pathway responsiveness correlates with the presence of primary cilia on prostate stromal cells. BMC DEVELOPMENTAL BIOLOGY 2009; 9:50. [PMID: 19811645 PMCID: PMC2767347 DOI: 10.1186/1471-213x-9-50] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 10/07/2009] [Indexed: 11/30/2022]
Abstract
Background Hedgehog (Hh) signaling from the urogenital sinus (UGS) epithelium to the surrounding mesenchyme plays a critical role in regulating ductal formation and growth during prostate development. The primary cilium, a feature of most interphase vertebrate cell types, serves as a required localization domain for Hh signaling transducing proteins. Results Immunostaining revealed the presence of primary cilia in mesenchymal cells of the developing prostate. Cell-based assays of a urongenital sinus mesenchymal cell line (UGSM-2) revealed that proliferation-limiting (serum starvation and/or confluence) growth conditions promoted cilia formation and correlated with pathway activation associated with accumulation of Smoothened in primary cilia. The prostate cancer cell lines PC-3, LNCaP, and 22RV1, previously shown to lack demonstrable autocrine Hh signaling capacity, did not exhibit primary cilia even under proliferation-limiting growth conditions. Conclusion We conclude that paracrine Hedgehog signaling activity in the prostate is associated with the presence of primary cilia on stromal cells but that a role in autocrine Hh signaling remains speculative.
Collapse
|
746
|
Sarangi A, Valadez JG, Zieber S, Abel TW, Thompson RC, Cooper MK. Targeted inhibition of the Hedgehog pathway in established malignant glioma xenografts enhances survival. Oncogene 2009; 28:3468-76. [PMID: 19617900 PMCID: PMC2756306 DOI: 10.1038/onc.2009.208] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 05/03/2009] [Accepted: 06/18/2009] [Indexed: 11/08/2022]
Abstract
Hedgehog pathway activity has been demonstrated in malignant glioma. However, its role in tumor growth has not been determined. Here we demonstrate that pharmacological inhibition of the Hedgehog pathway in established orthotopic malignant glioma xenografts confers a survival advantage. Pathway inhibition is measured in transplanted human tumor cells and not in host mouse brain. Correspondingly, survival benefit is observed only in tumors with an operational Hedgehog pathway. These data indicate that Hedgehog signaling regulates the growth of select malignant gliomas. We also demonstrate that Hedgehog pathway component and gene target expression segregate to CD133(+) tumor initiating cells. Treated mice eventually succumb to disease, thus, targeting the Hedgehog pathway in CD133(+) cells produces significant, but incomplete tumor regression. Therefore, our studies suggest that more complete tumor regression may require the inclusion of other therapeutic targets, including CD133(-) cells.
Collapse
Affiliation(s)
- A Sarangi
- Department of Neurology, Vanderbilt Medical Center, Nashville, TN 37232
- Vanderbilt Neuroscience Graduate Program, Vanderbilt Medical Center, Nashville, TN 37232
| | - J G Valadez
- Department of Neurology, Vanderbilt Medical Center, Nashville, TN 37232
| | - S Zieber
- Department of Pediatrics, Vanderbilt Medical Center, Nashville, TN 37232
| | - T W Abel
- Department of Pathology, Vanderbilt Medical Center, Nashville, TN 37232
| | - R C Thompson
- Department of Neurological Surgery, Vanderbilt Medical Center, Nashville, TN 37232
- Vanderbilt Ingram Cancer Center, Vanderbilt Medical Center, Nashville, TN 37232
| | - M K Cooper
- Department of Neurology, Vanderbilt Medical Center, Nashville, TN 37232
- Vanderbilt Neuroscience Graduate Program, Vanderbilt Medical Center, Nashville, TN 37232
| |
Collapse
|
747
|
Affiliation(s)
- Mats Ljungman
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA.
| |
Collapse
|
748
|
The Sonic Hedgehog pathway stimulates prostate tumor growth by paracrine signaling and recapitulates embryonic gene expression in tumor myofibroblasts. Oncogene 2009; 28:4480-90. [PMID: 19784071 PMCID: PMC2795794 DOI: 10.1038/onc.2009.294] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Hedgehog (Hh) pathway contributes to prostate cancer growth and progression. The presence of robust Sonic Hedgehog (Shh) expression in both normal prostate and localized cancer challenged us to explain the unique growth-promoting effect in cancer. We show here that paracrine Hh signaling exerts a non-cell autonomous effect on xenograft tumor growth and that Hh pathway activation in myofibroblasts alone is sufficient to stimulate tumor growth. Nine genes regulated by Hh in the mesenchyme of the developing prostate were found to be regulated in the stroma of Hh overexpressing xenograft tumors. Correlation analysis of gene expression in matched specimens of benign and malignant human prostate tissue revealed a partial five-gene fingerprint of Hh-regulated expression in stroma of all cancers and the complete nine-gene fingerprint in the subset of tumors exhibiting a reactive stroma. No expression fingerprint was observed in benign tissues. We conclude that changes in the prostate stroma due to association with cancer result in an altered transcriptional response to Hh that mimics the growth-promoting actions of the fetal mesenchyme. Patients with an abundance of myofibroblasts in biopsy tissue may comprise a subgroup that will exhibit a particularly good response to anti-Hh therapy.
Collapse
|
749
|
Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, Tibes R, Weiss GJ, Borad MJ, Hann CL, Brahmer JR, Mackey HM, Lum BL, Darbonne WC, Marsters JC, de Sauvage FJ, Low JA. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med 2009; 361:1164-72. [PMID: 19726763 DOI: 10.1056/nejmoa0905360] [Citation(s) in RCA: 821] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mutations in hedgehog pathway genes, primarily genes encoding patched homologue 1 (PTCH1) and smoothened homologue (SMO), occur in basal-cell carcinoma. In a phase 1 clinical trial, we assessed the safety and pharmacokinetics of GDC-0449, a small-molecule inhibitor of SMO, and responses of metastatic or locally advanced basal-cell carcinoma to the drug. METHODS We selected 33 patients with metastatic or locally advanced basal-cell carcinoma to receive oral GDC-0449 at one of three doses; 17 patients received 150 mg per day, 15 patients received 270 mg per day, and 1 patient received 540 mg per day. We assessed tumor responses with the use of Response Evaluation Criteria in Solid Tumors (RECIST), physical examination, or both. Molecular aspects of the tumors were examined. RESULTS The median duration of the study treatment was 9.8 months. Of the 33 patients, 18 had an objective response to GDC-0449, according to assessment on imaging (7 patients), physical examination (10 patients), or both (1 patient). Of the patients who had a response, 2 had a complete response and 16 had a partial response. The other 15 patients had either stable disease (11 patients) or progressive disease (4 patients). Eight grade 3 adverse events that were deemed to be possibly related to the study drug were reported in six patients, including four with fatigue, two with hyponatremia, one with muscle spasm, and one with atrial fibrillation. One grade 4 event, asymptomatic hyponatremia, was judged to be unrelated to GDC-0449. One patient withdrew from the study because of adverse events. We found evidence of hedgehog signaling in tumors that responded to the treatment. CONCLUSIONS GDC-0449, an orally active small molecule that targets the hedgehog pathway, appears to have antitumor activity in locally advanced or metastatic basal-cell carcinoma. (ClinicalTrials.gov number, NCT00607724.)
Collapse
Affiliation(s)
- Daniel D Von Hoff
- Translational Genomics Research Institute and Scottsdale Healthcare, Scottsdale, AZ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
750
|
|