501
|
Kieran MW, Roberts CW, Chi SN, Ligon KL, Rich BE, MacConaill LE, Garraway LA, Biegel JA. Absence of oncogenic canonical pathway mutations in aggressive pediatric rhabdoid tumors. Pediatr Blood Cancer 2012; 59:1155-7. [PMID: 22997201 PMCID: PMC3538080 DOI: 10.1002/pbc.24315] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 08/14/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rhabdoid tumors (also called atypical teratoid/rhabdoid tumor (AT/RT) in the brain), are highly malignant, poor prognosis lesions arising in the kidneys, soft tissues, and central nervous system. Targeted therapy in this disease would benefit from advanced technologies detecting relevant actionable mutations. PROCEDURE Here we report on the evaluation of 25 tumors, all with known SMARCB1/INI1 alterations, for the presence of 983 different mutations in 115 oncogenes and tumor-suppressor genes using OncoMap, a mass spectrometric method of allele detection. RESULTS Other than mutations in SMARCB1, our results identified a single activating mutation in NRAS and complete absence of oncogenic mutations in all other genes tested. CONCLUSION The absence of mutations in canonical pathways critical for development and progression of adult cancers suggests that distinct mechanisms drive these highly malignant pediatric tumors. This may limit the therapeutic utility of available targeted therapies and require a refocusing toward developmental and epigenetic pathways.
Collapse
Affiliation(s)
- Mark W. Kieran
- Director, Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute and Boston Children’s Hospital, Pediatric Hematology/ Oncology, 450 Brookline Avenue, Rm SW331, Boston, MA 02215 Tel (617) 632-4907 Fax (617) 632-4897 ,Address reprint requests to, Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute and Boston Children’s Hospital, Pediatric Hematology/Oncology, 450 Brookline Avenue, Rm SW331, Boston, MA 02215
| | - Charles W.M. Roberts
- Dana-Farber Cancer Institute and Boston Children’s Hospital, Pediatric Hematology/Oncology, 450 Brookline Avenue, Boston, MA 02215
| | - Susan N. Chi
- Dana-Farber Cancer Institute and Boston Children’s Hospital, Pediatric Hematology/Oncology, 450 Brookline Avenue, Boston, MA 02215
| | - Keith L. Ligon
- Dana-Farber Cancer Institute, Department of Medical Oncology, and Center for Molecular Oncologic Pathology, 450 Brookline Avenue, Rm JF200, Boston, MA 02115 Department of Pathology, Harvard Medical School, Brigham and Women’s Hospital and Boston Children’s Hospital
| | - Benjamin E. Rich
- Dana-Farber Cancer Institute, Department of Medical Oncology, and Center for Molecular Oncologic Pathology, 450 Brookline Avenue, Rm JF200, Boston, MA 02115 Department of Pathology, Harvard Medical School, Brigham and Women’s Hospital and Boston Children’s Hospital
| | - Laura E. MacConaill
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Levi A. Garraway
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Jaclyn A. Biegel
- Department of Pediatrics, The Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, 1002 Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104
| |
Collapse
|
502
|
Cucchi D, Occhione MA, Gulino A, De Smaele E. Hedgehog signaling pathway and its targets for treatment in basal cell carcinoma. J Exp Pharmacol 2012; 4:173-85. [PMID: 27186130 PMCID: PMC4863577 DOI: 10.2147/jep.s28553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Basal cell carcinoma (BCC) of the skin is the most common type of cancer and accounts for up to 40% of all cancers in the US, with a growing incidence rate over recent decades in all developed countries. Surgery is curative for most patients, although it leaves unaesthetic scars, but those that develop locally advanced or metastatic BCC require different therapeutic approaches. Furthermore, patients with BCC present a high risk of developing additional tumors. The increasing economic burden and the morbidity of BCC render primary interest in the development of targeted treatments for this disease. Among the molecular signals involved in the development of BCC, the critical role of the morphogenetic Hedgehog (Hh) pathway has become evident. This pathway is found altered and activated in almost all BCCs, both sporadic and inherited. Given the centrality of the Hh pathway in the pathophysiology of BCC, the primary efforts to identify molecular targets for the topical or systemic treatment of this cancer have focused on the Hh components. Several Hh inhibitors have been so far identified - from the first identified natural cyclopamine to the recently Food and Drug Administration-approved synthetic vismodegib - most of which target the Hh receptor Smoothened (either its function or its translocation to the primary cilium). Other molecules await further characterization (bisamide compounds), while drugs currently approved for other diseases such as itraconazole (an antimicotic agent) and vitamin D3 have been tested on BCC with encouraging results. The outcomes of the numerous ongoing clinical trials are expected to expand the field in the very near future. Further research is needed to obtain drugs targeting downstream components of the Hh pathway (eg, Gli) or to exploit combinatorial therapies (eg, with phosphatidylinositol 3-kinase inhibitors or retinoids) in order to overcome potential drug resistance.
Collapse
Affiliation(s)
- Danilo Cucchi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Alberto Gulino
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Center of Life NanoScience @ La Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
503
|
Abstract
Abstract
After more than a decade of treatment of chronic myeloid leukemia (CML) patients with the BCR-ABL tyrosine kinase inhibitor imatinib, and despite the impressive clinical results of this targeted therapeutic, many questions remain unresolved. One major question is how to cure CML, and the next step for the future will be to address this key issue. CML is a good model of cancer. The fact that the majority of CML patients who respond very well but discontinue tyrosine kinase inhibitors later show evidence of molecular recurrence focuses attention on the need for further research on leukemic stem cells. The challenge now is to understand why, after stopping treatment, the leukemia recurs in some patients but not in others. If we win this battle, this progress will certainly benefit the treatment and management of other leukemias and solid tumors and will validate this new topic.
Collapse
|
504
|
Abstract
Vismodegib is the first Hedgehog pathway inhibitor to be approved in the US, where it is indicated for the treatment of adults with metastatic basal cell carcinoma (BCC), or with locally advanced BCC that has recurred following surgery or who are not candidates for surgery, and who are not candidates for radiation. Vismodegib selectively and potently inhibits the Hedgehog signalling pathway by binding to Smoothened, thereby inhibiting the activation of Hedgehog target genes. Oral vismodegib was effective in the treatment of patients with locally advanced (n = 63) or metastatic (n = 33) BCC, according to the results of an ongoing, noncomparative, multinational, pivotal, phase II trial (ERIVANCE BCC). In this trial (using a clinical cutoff date of 26 November 2010), the independent review facility overall response rate was 42.9% in patients with locally advanced BCC and 30.3% in patients with metastatic BCC. In both patients with locally advanced BCC and those with metastatic BCC, the median duration of response was 7.6 months and median progression-free survival was 9.5 months. Oral vismodegib had an acceptable tolerability profile in patients with advanced BCC.
Collapse
|
505
|
The Cancer Stem-Cell Hypothesis: Its Emerging Role in Lung Cancer Biology and Its Relevance for Future Therapy. J Thorac Oncol 2012; 7:1880-1890. [DOI: 10.1097/jto.0b013e31826bfbc6] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
506
|
Abstract
The use of imatinib in chronic myelogenous leukemia (CML) transformed the disease, rapidly changing the median survival from 4 years to at least 20 years. In this review, we outline the causes of this revolution, including the identification of a critical driving molecular aberration, BCR-ABL, and the development of a potent and specific inhibitor, imatinib. Equally important was the timing of the targeted therapy, specifically its administration to patients with newly diagnosed disease. In solid tumors, targeted therapies are often both developed and used in metastatic malignancies after conventional approaches have failed. We postulate that this strategy is similar to using imatinib in blast-crisis CML, in which response rates are less than 15%, all patients relapse, and median survival remains only about 1 year. We hypothesize that the imatinib-led revolution in CML, including the critically important factor of timing, may be applicable to other cancers as well. Therefore, it will be important to use promising targeted therapies in the earliest phases of biomarker-defined solid tumors, before metastatic progression, to determine if outcomes can be significantly improved and, thus, establish if the success of imatinib in CML is an anomaly or a paradigm.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Molecular Targeted Therapy
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Jason R Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston TX 77030, USA.
| | | |
Collapse
|
507
|
Fu J, Rodova M, Roy SK, Sharma J, Singh KP, Srivastava RK, Shankar S. GANT-61 inhibits pancreatic cancer stem cell growth in vitro and in NOD/SCID/IL2R gamma null mice xenograft. Cancer Lett 2012. [PMID: 23200667 DOI: 10.1016/j.canlet.2012.11.018] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple lines of evidence suggest that the Sonic Hedgehog (Shh) signaling pathway is aberrantly reactivated in pancreatic cancer stem cells (CSCs). The objectives of this study were to examine the molecular mechanisms by which GANT-61 (Gli transcription factor inhibitor) regulates stem cell characteristics and tumor growth. Effects of GANT-61 on CSC's viability, spheroid formation, apoptosis, DNA-binding and transcriptional activities, and epithelial-mesenchymal transition (EMT) were measured. Humanized NOD/SCID/IL2R gamma(null) mice were used to examine the effects of GANT-61 on CSC's tumor growth. GANT-61 inhibited cell viability, spheroid formation, and Gli-DNA binding and transcriptional activities, and induced apoptosis by activation of caspase-3 and cleavage of Poly-ADP ribose Polymerase (PARP). GANT-61 increased the expression of TRAIL-R1/DR4, TRAIL-R2/DR5 and Fas, and decreased expression of PDGFRα and Bcl-2. GANT-61 also suppressed EMT by up-regulating E-cadherin and inhibiting N-cadherin and transcription factors Snail, Slug and Zeb1. In addition, GANT-61 inhibited pluripotency maintaining factors Nanog, Oct4, Sox-2 and cMyc. Suppression of both Gli1 plus Gli2 by shRNA mimicked the changes in cell viability, spheroid formation, apoptosis and gene expression observed in GANT-61-treated pancreatic CSCs. Furthermore, GANT-61 inhibited CSC tumor growth which was associated with up-regulation of DR4 and DR5 expression, and suppression of Gli1, Gli2, Bcl-2, CCND2 and Zeb1 expression in tumor tissues derived from NOD/SCID IL2Rγ null mice. Our data highlight the importance of Shh pathway for self-renewal and metastasis of pancreatic CSCs, and also suggest Gli as a therapeutic target for pancreatic cancer in eliminating CSCs.
Collapse
Affiliation(s)
- Junsheng Fu
- Department of Pathology and Laboratory Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | | | | | | | | | | | |
Collapse
|
508
|
|
509
|
Zeng J, Aziz K, Chettiar ST, Aftab BT, Armour M, Gajula R, Gandhi N, Salih T, Herman JM, Wong J, Rudin CM, Tran PT, Hales RK. Hedgehog pathway inhibition radiosensitizes non-small cell lung cancers. Int J Radiat Oncol Biol Phys 2012. [PMID: 23182391 DOI: 10.1016/j.ijrobp.2012.10.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE Despite improvements in chemoradiation, local control remains a major clinical problem in locally advanced non-small cell lung cancer. The Hedgehog pathway has been implicated in tumor recurrence by promoting survival of tumorigenic precursors and through effects on tumor-associated stroma. Whether Hedgehog inhibition can affect radiation efficacy in vivo has not been reported. METHODS AND MATERIALS We evaluated the effects of a targeted Hedgehog inhibitor (HhAntag) and radiation on clonogenic survival of human non-small cell lung cancer lines in vitro. Using an A549 cell line xenograft model, we examined tumor growth, proliferation, apoptosis, and gene expression changes after concomitant HhAntag and radiation. In a transgenic mouse model of Kras(G12D)-induced and Twist1-induced lung adenocarcinoma, we assessed tumor response to radiation and HhAntag by serial micro-computed tomography (CT) scanning. RESULTS In 4 human lung cancer lines in vitro, HhAntag showed little or no effect on radiosensitivity. By contrast, in both the human tumor xenograft and murine inducible transgenic models, HhAntag enhanced radiation efficacy and delayed tumor growth. By use of the human xenograft model to differentiate tumor and stromal effects, mouse stromal cells, but not human tumor cells, showed significant and consistent downregulation of Hedgehog pathway gene expression. This was associated with increased tumor cell apoptosis. CONCLUSIONS Targeted Hedgehog pathway inhibition can increase in vivo radiation efficacy in lung cancer preclinical models. This effect is associated with pathway suppression in tumor-associated stroma. These data support clinical testing of Hedgehog inhibitors as a component of multimodality therapy for locally advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21231-2410, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
510
|
Gamba CA, Wysong A, Million L, Aasi S, Kim J, Tang JY. Is tanning bed exposure associated with aggressive basal cell carcinoma? J Clin Oncol 2012; 30:e333-6. [PMID: 23008324 DOI: 10.1200/jco.2012.42.1008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Christina A Gamba
- Department of Dermatology, Stanford University School of Medicine, 450 Broadway Street, Pavilion C, Room 232, Redwood City, CA 94063, USA
| | | | | | | | | | | |
Collapse
|
511
|
Ermertcan AT, Hellings PW, Cingi C. Nonmelanoma Skin Cancer of the Head and Neck. Facial Plast Surg Clin North Am 2012; 20:445-54. [DOI: 10.1016/j.fsc.2012.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
512
|
Karlou M, Tzelepi V, Maity S, Navone NM, Yang J, Hoang A, Lu JF, Logothetis CJ, Efstathiou E. Hedgehog signaling inhibition by the small molecule smoothened inhibitor GDC-0449 in the bone forming prostate cancer xenograft MDA PCa 118b. Prostate 2012; 72:1638-47. [PMID: 22457212 PMCID: PMC4977841 DOI: 10.1002/pros.22517] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 02/24/2012] [Indexed: 11/06/2022]
Abstract
BACKGROUND Hedgehog signaling is a stromal-mesenchymal pathway central to the development and homeostasis of both the prostate and the bone. Aberrant Hedgehog signaling activation has been associated with prostate cancer aggressiveness. We hypothesize that Hedgehog pathway is a candidate therapeutic target in advanced prostate cancer. We confirm increased Hedgehog signaling in advanced and bone metastatic castrate resistant prostate cancer and examine the pharmacodynamic effect of Smoothened inhibition by the novel reagent GDC-0449 in an experimental prostate cancer model. METHODS Hedgehog signaling component expression was assessed in tissue microarrays of high grade locally advanced and bone metastatic disease. Male SCID mice subcutaneously injected with the bone forming xenograft MDA PCa 118b were treated with GDC-0449. Hedgehog signaling in the tumor microenvironment was assessed by proteomic and species specific RNA expression and compared between GDC-0449 treated and untreated animals. RESULTS We observe Hedgehog signaling in high grade locally advanced and bone marrow infiltrating disease. Evidence of paracrine activation of Hedgehog signaling in the tumor xenograft, was provided by increased Sonic Hedgehog expression in human tumor epithelial cells, coupled with increased Gli1 and Patched1 expression in the murine stromal compartment, while normal murine stroma did not exhibit Hh signaling expression. GDC-0449 treatment attenuated Hh signaling as evidenced by reduced expression of Gli1 and Ptch1. Reduction in proliferation (Ki67) was observed with no change in tumor volume. CONCLUSIONS GDC-0449 treatment is pharmacodynamically effective as evidenced by paracrine Hedgehog signaling inhibition and results in tumor cell proliferation reduction. Understanding these observations will inform the clinical development of therapy based on Hedgehog signaling inhibition.
Collapse
Affiliation(s)
- Maria Karlou
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Vassiliki Tzelepi
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
- Department of Pathology, Medical School, University of Patras, 26500, Patras, Greece
| | - Sankar Maity
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Nora M. Navone
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Jun Yang
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Anh Hoang
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Jing-Fang Lu
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Christopher J. Logothetis
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
| | - Eleni Efstathiou
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research of Genitourinary Cancers, The Stanford Alexander Tissue Derivatives Laboratory, The University of Texas MD Anderson Cancer Center, Unit 1374, 1550 Holcombe Boulevard, Houston, TX 77030-4009
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece
- Correspondence to: Eleni Efstathiou, Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas M. D. Anderson Cancer Center, 1550 Holcombe Boulevard, Houston, TX 77030-4009, Office: 7135630894 Fax: 7135639409,
| |
Collapse
|
513
|
Colmont CS, Harding KG, Piguet V, Patel GK. Human skin cancer stem cells: a tale of mice and men. Exp Dermatol 2012; 21:576-80. [PMID: 22775992 DOI: 10.1111/j.1600-0625.2012.01533.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Carcinomas, cancers of epithelial tissues, are the commonest malignancies and cause the greatest cancer mortality worldwide. Among these, the incidence of keratinocyte-derived non-melanoma skin cancers (NMSC), by far the greatest, is increasing rapidly. Yet despite access to tumor tissue, acceptance of human NMSC as a model carcinoma has been hindered by the lack of a reliable xenograft model. Instead, we have relied on the murine two-step carcinogenesis protocol as a reproducible squamous cell carcinoma (SCC) model, but this differs from their human counterpart in cause, site, genetic basis and biological behaviour. By xeno-engraftment of primary human SCC, we were recently successful in demonstrating the presence of primary human SCC cancer stem cells or tumor-initiating cells. These findings once more align the study human SCC as the archetypal carcinoma model. In this review, we describe the evidence for the existence of tumor-initiating cells, with emphasis on skin cancer, limiting our discussions to primary human cancer studies where possible.
Collapse
Affiliation(s)
- Chantal S Colmont
- Department of Dermatology and Wound Healing, School of Medicine Cardiff University, Cardiff, UK
| | | | | | | |
Collapse
|
514
|
Eimer S, Dugay F, Airiau K, Avril T, Quillien V, Belaud-Rotureau MA, Belloc F. Cyclopamine cooperates with EGFR inhibition to deplete stem-like cancer cells in glioblastoma-derived spheroid cultures. Neuro Oncol 2012; 14:1441-51. [PMID: 23104476 DOI: 10.1093/neuonc/nos266] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Putative cancer stem cells have been identified in glioblastoma (GBM), associated with resistance to conventional therapies. Overcoming this resistance is a major challenge to manage this deadly brain tumor. Epidermal growth factor receptor (EGFR) is commonly amplified, over-expressed, and/or mutated in GBM, making it a compelling target for therapy. This study investigates the behavior of 3 primary neurosphere (NS) cell lines and their adherent counterparts originated from human GBM resections, when treated with EGFR-tyrosine kinase inhibitor erlotinib, associated or not with cyclopamine, a hedgehog pathway inhibitor. Adherent cells cultured in the presence of serum expressed the glial fibrillary acidic protein, whereas NS-forming cells cultured in serum-free medium expressed CD133, nestin, and Oct-4, markers of neural stem and progenitor cells. For the 3 adherent cell lines, erlotinib has a moderate effect (50% inhibitory concentration [IC50], >10 µM). Conversely, erlotinib induced a strong cell growth inhibition (IC50, <1 µM) on NS-forming cells, related to the EGFR gene amplification and EGFR protein expression. A short exposure to erlotinib reduced nestin-positive cell proliferation, but NS-initiating activity and self-renewal were not altered. EGFR pathway seems essential for GBM progenitor cell proliferation but dispensable for cancer stem-like cell self-renewal. Inhibition of hedgehog pathway with cyclopamine was evaluated in association with erlotinib on NS growth. Although each drug separately had no effect on sphere initiation, their combination significantly decreased the sphere number (P < .001). Our findings show synergic efficiency for erlotinib-cyclopamine association and provide a suitable in vitro model to explore drug combinations on GBM cells.
Collapse
Affiliation(s)
- Sandrine Eimer
- EA 2406 Histology and Molecular Pathology of Tumors Laboratory, Léo Saignat Victor Segalen University, Bordeaux, France.
| | | | | | | | | | | | | |
Collapse
|
515
|
Cohen DJ. Targeting the hedgehog pathway: role in cancer and clinical implications of its inhibition. Hematol Oncol Clin North Am 2012; 26:565-88, viii. [PMID: 22520980 DOI: 10.1016/j.hoc.2012.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The Hedgehog (Hh) pathway is a signaling cascade that is evolutionally highly conserved and plays an important role in embryonic pattern formation and stem cell response to tissue damage. Given the pivotal role the Hh pathway plays in embryonic development in terms of proliferation and differentiation, it is not surprising that it has also been implicated in tumorigenesis and tumor growth acceleration in a vast variety of malignancies. This article summarizes the mechanism of Hh pathway signal transduction, discusses the models of pathway activation, reviews the clinical data using Hh inhibitors, and discusses challenges to the development of pathway inhibitors.
Collapse
Affiliation(s)
- Deirdre J Cohen
- Division of GI Oncology, NYU Cancer Institute, 160 East 34th Street, New York, NY 10016, USA.
| |
Collapse
|
516
|
Berlin J, Bendell JC, Hart LL, Firdaus I, Gore I, Hermann RC, Mulcahy MF, Zalupski MM, Mackey HM, Yauch RL, Graham RA, Bray GL, Low JA. A randomized phase II trial of vismodegib versus placebo with FOLFOX or FOLFIRI and bevacizumab in patients with previously untreated metastatic colorectal cancer. Clin Cancer Res 2012; 19:258-67. [PMID: 23082002 DOI: 10.1158/1078-0432.ccr-12-1800] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Vismodegib, a Hedgehog pathway inhibitor, has preclinical activity in colorectal cancer (CRC) models. This trial assessed the efficacy, safety, and pharmacokinetics of adding vismodegib to first-line treatment for metastatic CRC (mCRC). EXPERIMENTAL DESIGN Patients were randomized to receive vismodegib (150 mg/day orally) or placebo, in combination with FOLFOX or FOLFIRI chemotherapy plus bevacizumab (5 mg/kg) every 2 weeks until disease progression or intolerable toxicity. The primary endpoint was progression-free survival (PFS). Key secondary objectives included evaluation of predictive biomarkers and pharmacokinetic drug interactions. RESULTS A total of 199 patients with mCRC were treated on protocol (124 FOLFOX, 75 FOLFIRI). The median PFS hazard ratio (HR) for vismodegib treatment compared with placebo was 1.25 (90% CI: 0.89-1.76; P = 0.28). The overall response rates for placebo-treated and vismodegib-treated patients were 51% (90% CI: 43-60) and 46% (90% CI: 37-55), respectively. No vismodegib-associated benefit was observed in combination with either FOLFOX or FOLFIRI. Increased tumor tissue Hedgehog expression did not predict clinical benefit. Grade 3 to 5 adverse events reported for more than 5% of patients that occurred more frequently in the vismodegib-treated group were fatigue, nausea, asthenia, mucositis, peripheral sensory neuropathy, weight loss, decreased appetite, and dehydration. Vismodegib did not alter the pharmacokinetics of FOLFOX, FOLFIRI, or bevacizumab. CONCLUSIONS Vismodegib does not add to the efficacy of standard therapy for mCRC. Compared with placebo, treatment intensity was lower for all regimen components in vismodegib-treated patients, suggesting that combined toxicity may have contributed to lack of efficacy.
Collapse
Affiliation(s)
- Jordan Berlin
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
517
|
Abstract
Molecular diagnostic strategies are gaining wider acceptance and use in dermatology and dermatopathology as more practitioners in this field develop an understanding of the principles and applications of genomic technologies. Molecular testing is facilitating more accurate diagnosis, staging, and prognostication, in addition to guiding the selection of appropriate treatment, monitoring of therapy, and identification of novel therapeutic targets, for a wide variety of skin diseases.
Collapse
Affiliation(s)
- Zendee Elaba
- Department of Pathology, Hartford Hospital, Hartford, CT, USA
| | | | | |
Collapse
|
518
|
McKee CM, Xu D, Cao Y, Kabraji S, Allen D, Kersemans V, Beech J, Smart S, Hamdy F, Ishkanian A, Sykes J, Pintile M, Milosevic M, van der Kwast T, Zafarana G, Ramnarine VR, Jurisica I, Mallof C, Lam W, Bristow RG, Muschel RJ. Protease nexin 1 inhibits hedgehog signaling in prostate adenocarcinoma. J Clin Invest 2012; 122:4025-36. [PMID: 23041623 DOI: 10.1172/jci59348] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 08/14/2012] [Indexed: 01/08/2023] Open
Abstract
Prostate adenocarcinoma (CaP) patients are classified into low-, intermediate-, and high-risk groups that reflect relative survival categories. While there are accepted treatment regimens for low- and high-risk patients, intermediate-risk patients pose a clinical dilemma, as treatment outcomes are highly variable for these individuals. A better understanding of the factors that regulate the progression of CaP is required to delineate risk. For example, aberrant activation of the Hedgehog (Hh) pathway is implicated in CaP progression. Here, we identify the serine protease inhibitor protease nexin 1 (PN1) as a negative regulator of Hh signaling in prostate. Using human CaP cell lines and a mouse xenograft model of CaP, we demonstrate that PN1 regulates Hh signaling by decreasing protein levels of the Hh ligand Sonic (SHH) and its downstream effectors. Furthermore, we show that SHH expression enhanced tumor growth while overexpression of PN1 inhibited tumor growth and angiogenesis in mice. Finally, using comparative genome hybridization, we found that genetic alterations in Hh pathway genes correlated with worse clinical outcomes in intermediate-risk CaP patients, indicating the importance of this pathway in CaP.
Collapse
Affiliation(s)
- Chad M McKee
- Gray Institute of Radiation Oncology and Biology, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
519
|
Inhibition of hedgehog signaling for the treatment of murine sclerodermatous chronic graft-versus-host disease. Blood 2012; 120:2909-17. [DOI: 10.1182/blood-2012-01-403428] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Chronic graft-versus-host disease (cGVHD) is a prognosis limiting complication of allogeneic stem cell transplantation. The molecular mechanisms underlying cGVHD are incompletely understood, and targeted therapies are not yet established for clinical use. Here we examined the role of the hedgehog pathway in sclerodermatous cGVHD. Hedgehog signaling was activated in human and murine cGVHD with increased expression of sonic hedgehog and accumulation of the transcription factors Gli-1 and Gli-2. Treatment with LDE223, a highly selective small-molecule antagonist of the hedgehog coreceptor Smoothened (Smo), abrogated the activation of hedgehog signaling and protected against experimental cGVHD. Preventive therapy with LDE223 almost completely impeded the development of clinical and histologic features of sclerodermatous cGVHD. Treatment with LDE223 was also effective, when initiated after the onset of clinical manifestations of cGVHD. Hedgehog signaling stimulated the release of collagen from cultured fibroblasts but did not affect leukocyte influx in murine cGVHD, suggesting direct, leukocyte-independent stimulatory effects on fibroblasts as the pathomechanism of hedgehog signaling in cGVHD. Considering the high morbidity of cGVHD, the current lack of efficient molecular therapies for clinical use, and the availability of well-tolerated inhibitors of Smo, targeting hedgehog signaling might be a novel strategy for clinical trials in cGVHD.
Collapse
|
520
|
Kaye SB, Fehrenbacher L, Holloway R, Amit A, Karlan B, Slomovitz B, Sabbatini P, Fu L, Yauch RL, Chang I, Reddy JC. A phase II, randomized, placebo-controlled study of vismodegib as maintenance therapy in patients with ovarian cancer in second or third complete remission. Clin Cancer Res 2012; 18:6509-18. [PMID: 23032746 DOI: 10.1158/1078-0432.ccr-12-1796] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Hedgehog pathway inhibition has been suggested as a potential maintenance treatment approach in ovarian cancer through disruption of tumor-stromal interactions. Vismodegib is an orally available Hedgehog pathway inhibitor with clinical activity in advanced basal cell carcinoma and medulloblastoma. This phase II, randomized, double-blind, placebo-controlled trial was designed to provide a preliminary estimate of efficacy in patients with ovarian cancer in second or third complete remission (CR). EXPERIMENTAL DESIGN Patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in second or third CR were randomized 1:1 to vismodegib (GDC-0449; 150 mg daily) or placebo three to 14 weeks after completing chemotherapy. Treatment continued until radiographic progression or toxicity. The primary endpoint was investigator-assessed progression-free survival (PFS). RESULTS One hundred four patients were randomized to vismodegib (n = 52) or placebo (n = 52); median PFS was 7.5 months and 5.8 months, respectively [HR 0.79; 95% confidence interval (CI), 0.46-1.35]. The HR was 0.66 (95% CI, 0.36-1.20) for second CR patients (n = 84) and 1.79 (95% CI, 0.50-6.48) for third CR patients (n = 20). The most common adverse events in the vismodegib arm were dysgeusia/ageusia, muscle spasms, and alopecia. Grade 3/4 adverse events occurred in 12 patients (23.1%) with vismodegib and six (11.5%) with placebo. Hedgehog expression was detected in 13.5% of archival tissues. CONCLUSIONS In this study, the sought magnitude of increase in PFS was not achieved for vismodegib maintenance versus placebo in patients with ovarian cancer in second or third CR. The frequency of Hedgehog ligand expression was lower than expected.
Collapse
|
521
|
Alison MR, Lin WR, Lim SML, Nicholson LJ. Cancer stem cells: in the line of fire. Cancer Treat Rev 2012; 38:589-598. [PMID: 22469558 DOI: 10.1016/j.ctrv.2012.03.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/07/2012] [Accepted: 03/09/2012] [Indexed: 12/20/2022]
Abstract
Most tumours appear to contain a sub-population(s) of self-renewing and expanding stem cells known as cancer stem cells (CSCs). The CSC model proposes that CSCs are at the apex of a hierarchically organized cell population, somewhat akin to normal tissue organization. Selection pressures may also facilitate the stochastic clonal expansion of sub-sets of cancer cells that may co-exist with CSCs and their progeny, moreover the trait of stemness may be more fluid than hitherto expected, and cells may switch between the stem and non-stem cell state. A large body of evidence points to the fact that CSCs are particularly resistant to radiotherapy and chemotherapy. In this review we discuss the basis of such resistance that highlights the roles of ABC transporters, aldehyde dehydrogenase (ALDH) activity, intracellular signalling pathways, the DNA damage response, hypoxia and proliferative quiescence as being significant determinants. In the light of such observations, we outline strategies for the successful eradication of CSCs, including targeting the self-renewal controlling pathways (Wnt, Notch and Hedgehog), ALDH activity and ABC transporters, blocking epithelial mesenchymal transition (EMT), differentiation therapy and niche targeting.
Collapse
Affiliation(s)
- Malcolm R Alison
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | | | | | |
Collapse
|
522
|
|
523
|
Cirrone F, Harris CS. Vismodegib and the Hedgehog Pathway: A New Treatment for Basal Cell Carcinoma. Clin Ther 2012; 34:2039-50. [DOI: 10.1016/j.clinthera.2012.08.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 09/05/2012] [Indexed: 11/28/2022]
|
524
|
Wang J, Mook RA, Lu J, Gooden DM, Ribeiro A, Guo A, Barak LS, Lyerly HK, Chen W. Identification of a novel Smoothened antagonist that potently suppresses Hedgehog signaling. Bioorg Med Chem 2012; 20:6751-7. [PMID: 23063522 DOI: 10.1016/j.bmc.2012.09.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/31/2012] [Accepted: 09/10/2012] [Indexed: 12/21/2022]
Abstract
The Hedgehog signaling pathway plays an essential role in embryo development and adult tissue homeostasis, in regulating stem cells and is abnormally activated in many cancers. Given the importance of this signaling pathway, we developed a novel and versatile high-throughput, cell-based screening platform using confocal imaging, based on the role of β-arrestin in Hedgehog signal transduction, that can identify agonists or antagonist of the pathway by a simple change to the screening protocol. Here we report the use of this assay in the antagonist mode to identify novel antagonists of Smoothened, including a compound (A8) with low nanomolar activity against wild-type Smo also capable of binding the Smo point mutant D473H associated with clinical resistance in medulloblastoma. Our data validate this novel screening approach in the further development of A8 and related congeners to treat Hedgehog related diseases, including the treatment of basal cell carcinoma and medulloblastoma.
Collapse
Affiliation(s)
- Jiangbo Wang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
525
|
Thacker CA, Weiss GJ, Tibes R, Blaydorn L, Downhour M, White E, Baldwin J, Hoff DD, Korn RL. 18-FDG PET/CT assessment of basal cell carcinoma with vismodegib. Cancer Med 2012; 1:230-6. [PMID: 23342272 PMCID: PMC3544445 DOI: 10.1002/cam4.33] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/18/2012] [Accepted: 08/20/2012] [Indexed: 12/26/2022] Open
Abstract
The use of 18-fluorodeoxyglucose (FDG) positron emission tomography with computed tomography (PET/CT) in subjects with advanced basal cell carcinoma (BCC) has not been fully explored due to the rarity of disease presentation. This study evaluated PET/CTs from subjects with advanced BCC participating in a phase I dose-escalation clinical trial of vismodegib. Fourteen subjects with BCC were imaged with 18-FDG PET/CT for lesion identification and response categorizing (European Organisation for Research and Treatment for Cancer [EORTC] and PET response criteria in solid tumors [PERCIST] 1.0). Several parameters including metabolic activity of target lesions, site of disease presentation and spread, treatment response, and prognostic significance of metabolic activity following therapy were evaluated. All subjects exhibited at least one hypermetabolic lesion. Most subjects had only four organ systems involved at study enrollment: skin–muscle (93%), lung (57%), lymph nodes (29%), and bone (21%). SUVmax measured across all lesions decreased (median 33%, SD ± 45%) following therapy with metabolic activity normalizing or disappearing in 42% of lesions. No significant difference was observed between EORTC and PERCIST 1.0. Subjects that demonstrated at least a 33% reduction in SUVmax from baseline had a significantly longer progression-free survival (PFS) (median 17 months, 95% confidence interval [CI] ±4 months vs. 9 months, 95% CI ±5 months, P = 0.038) and overall survival (OS) (median 24 months, 95% CI ±4 months vs. 17 months, 95% CI ±13 months, P = 0.019). BCC lesions are hypermetabolic on 18-FDG PET/CT. A decrease in SUVmax was associated with improved PFS and OS. These results further support the incorporation of 18-FDG PET/CT scans in advanced BCC management.
Collapse
Affiliation(s)
- Curtis A Thacker
- Scottsdale Medical Imaging, Ltd. Scottsdale, AZ; Midwestern University Glendale, AZ, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
526
|
Kobune M, Iyama S, Kikuchi S, Horiguchi H, Sato T, Murase K, Kawano Y, Takada K, Ono K, Kamihara Y, Hayashi T, Miyanishi K, Sato Y, Takimoto R, Kato J. Stromal cells expressing hedgehog-interacting protein regulate the proliferation of myeloid neoplasms. Blood Cancer J 2012; 2:e87. [PMID: 22961059 PMCID: PMC3461706 DOI: 10.1038/bcj.2012.36] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aberrant reactivation of hedgehog (Hh) signaling has been described in a wide variety of human cancers including cancer stem cells. However, involvement of the Hh-signaling system in the bone marrow (BM) microenvironment during the development of myeloid neoplasms is unknown. In this study, we assessed the expression of Hh-related genes in primary human CD34+ cells, CD34+ blastic cells and BM stromal cells. Both Indian Hh (Ihh) and its signal transducer, smoothened (SMO), were expressed in CD34+ acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS)-derived cells. However, Ihh expression was relatively low in BM stromal cells. Remarkably, expression of the intrinsic Hh-signaling inhibitor, human Hh-interacting protein (HHIP) in AML/MDS-derived stromal cells was markedly lower than in healthy donor-derived stromal cells. Moreover, HHIP expression levels in BM stromal cells highly correlated with their supporting activity for SMO+ leukemic cells. Knockdown of HHIP gene in stromal cells increased their supporting activity although control cells marginally supported SMO+ leukemic cell proliferation. The demethylating agent, 5-aza-2′-deoxycytidine rescued HHIP expression via demethylation of HHIP gene and reduced the leukemic cell-supporting activity of AML/MDS-derived stromal cells. This indicates that suppression of stromal HHIP could be associated with the proliferation of AML/MDS cells.
Collapse
Affiliation(s)
- M Kobune
- Fourth Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
527
|
Yun JI, Kim HR, Park H, Kim SK, Lee J. Small molecule inhibitors of the hedgehog signaling pathway for the treatment of cancer. Arch Pharm Res 2012; 35:1317-33. [PMID: 22941475 DOI: 10.1007/s12272-012-0801-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 06/05/2012] [Accepted: 06/06/2012] [Indexed: 12/28/2022]
Abstract
Over the past decade, the Hedgehog signaling pathway has attracted considerable interest because the pathway plays important roles in the tumorigenesis of several types of cancer as well as developmental processes. It has also been observed that Hedgehog signaling regulates the proliferation and self-renewal of cancer stem cells. A great number of Hedgehog pathway inhibitors have been discovered through small molecule screens and subsequent medicinal chemistry efforts. Among the inhibitors, several Smo antagonists have reached the clinical trial phase. It has been proved that the inhibition of Hedgehog signaling with Smo antagonists is beneficial to cancer patients with basal cell carcinoma and medulloblastoma. In this review, we provide an overview of Hedgehog pathway inhibitors with focusing on the preclinical and/or clinical efficacy and molecular mechanisms of these inhibitors.
Collapse
Affiliation(s)
- Jeong In Yun
- Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Deajeon, 305-600, Korea
| | | | | | | | | |
Collapse
|
528
|
Galimberti F, Busch AM, Chinyengetere F, Ma T, Sekula D, Memoli VA, Dragnev KH, Liu F, Johnson KC, Guo Y, Freemantle SJ, Andrew AS, Greninger P, Robbins DJ, Settleman J, Benes C, Dmitrovsky E. Response to inhibition of smoothened in diverse epithelial cancer cells that lack smoothened or patched 1 mutations. Int J Oncol 2012; 41:1751-61. [PMID: 22923130 PMCID: PMC3583816 DOI: 10.3892/ijo.2012.1599] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/19/2012] [Indexed: 12/11/2022] Open
Abstract
Hedgehog (HH) pathway Smoothened (Smo) inhibitors are active against Gorlin syndrome-associated basal cell carcinoma (BCC) and medulloblastoma where Patched (Ptch) mutations occur. We interrogated 705 epithelial cancer cell lines for growth response to the Smo inhibitor cyclopamine and for expressed HH pathway-regulated species in a linked genetic database. Ptch and Smo mutations that respectively conferred Smo inhibitor response or resistance were undetected. Previous studies revealed HH pathway activation in lung cancers. Therefore, findings were validated using lung cancer cell lines, transgenic and transplantable murine lung cancer models, and human normal-malignant lung tissue arrays in addition to testing other Smo inhibitors. Cyclopamine sensitivity most significantly correlated with high cyclin E (P=0.000009) and low insulin-like growth factor binding protein 6 (IGFBP6) (P=0.000004) levels. Gli family members were associated with response. Cyclopamine resistance occurred with high GILZ (P=0.002) expression. Newer Smo inhibitors exhibited a pattern of sensitivity similar to cyclopamine. Gain of cyclin E or loss of IGFBP6 in lung cancer cells significantly increased Smo inhibitor response. Cyclin E-driven transgenic lung cancers expressed a gene profile implicating HH pathway activation. Cyclopamine treatment significantly reduced proliferation of murine and human lung cancers. Smo inhibition reduced lung cancer formation in a syngeneic mouse model. In human normal-malignant lung tissue arrays cyclin E, IGFBP6, Gli1 and GILZ were each differentially expressed. Together, these findings indicate that Smo inhibitors should be considered in cancers beyond those with activating HH pathway mutations. This includes tumors that express genes indicating basal HH pathway activation.
Collapse
Affiliation(s)
- Fabrizio Galimberti
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
529
|
Gropper AB, Girouard SD, Hojman LP, Huang SJ, Qian X, Murphy GF, Vleugels RA. Metastatic basal cell carcinoma of the posterior neck: case report and review of the literature. J Cutan Pathol 2012; 39:526-34. [PMID: 22515224 DOI: 10.1111/j.1600-0560.2012.01871.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although primary basal cell carcinoma (BCC) represents an extremely common malignancy, metastases derived from BCC are exceedingly rare. The prognosis for metastatic BCC is poor, and little consensus exists regarding predictive factors or optimal treatment strategies. Here, we present the case of a 63-year-old man with BCC of the neck who subsequently developed multiple metastases to subcutaneous tissue, lymph nodes, and the parotid gland. Risk factors and clinical features of metastatic BCC are reviewed, as is the relationship of histopathologic subtype to metastatic behavior. Current chemotherapeutic and targeted therapies also are discussed in the context of recent advances in molecular biology.
Collapse
Affiliation(s)
- Adrienne B Gropper
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
530
|
Agyeman A, Mazumdar T, Houghton JA. Regulation of DNA damage following termination of Hedgehog (HH) survival signaling at the level of the GLI genes in human colon cancer. Oncotarget 2012; 3:854-68. [PMID: 23097684 PMCID: PMC3478462 DOI: 10.18632/oncotarget.586] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/18/2012] [Indexed: 01/23/2023] Open
Abstract
Transcriptional regulation of the Hedgehog (HH) signaling response is mediated by GLI genes (GLI1, GLI2) downstream of SMO, that are also activated by oncogenic signaling pathways. We have demonstrated the importance of targeting GLI downstream of SMO in the induction of cell death in human colon carcinoma cells. In HT29 cells inhibition of GLI1/GLI2 by the small molecule inhibitor GANT61 induced DNA double strand breaks (DSBs) and activation of ATM, MDC1 and NBS1; γH2AX and MDC1, NBS1 and MDC1 co-localized in nuclear foci. Early activation of ATM was decreased by 24 hr, when p-NBS1Ser343, activated by ATM, was significantly reduced in cell extracts. Bound γH2AX was detected in isolated chromatin fractions or nuclei during DNA damage but not during DNA repair. MDC1 was tightly bound to chromatin at 32 hr as cells accumulated in early S-phase prior to becoming subG1, and during DNA repair. Limited binding of NBS1 was detected at all times during DNA damage but was strongly bound during DNA repair. Transient overexpression of NBS1 protected HT29 cells from GANT61-induced cell death, while knockdown of H2AX by H2AXshRNA delayed DNA damage signaling. Data demonstrate following GLI1/GLI2 inhibition: 1) induction of DNA damage in cells that are also resistant to SMO inhibitors, 2) dynamic interactions between γH2AX, MDC1 and NBS1 in single cell nuclei and in isolated chromatin fractions, 3) expression and chromatin binding properties of key mediator proteins that mark DNA damage or DNA repair, and 4) the importance of NBS1 in the DNA damage response mechanism.
Collapse
Affiliation(s)
- Akwasi Agyeman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | |
Collapse
|
531
|
Queiroz KCS, Spek CA, Peppelenbosch MP. Targeting Hedgehog signaling and understanding refractory response to treatment with Hedgehog pathway inhibitors. Drug Resist Updat 2012; 15:211-22. [PMID: 22910179 DOI: 10.1016/j.drup.2012.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/25/2012] [Accepted: 05/26/2012] [Indexed: 12/01/2022]
Abstract
Hedgehog (Hh) signaling is a principal component of the morphogenetic code best known to direct pattern formation during embryogenesis. The Hh pathway remains active in adulthood however where it guides tissue regeneration and remodeling and Hh production in the niche plays an important role in maintaining stem cell compartment size. Deregulated Hh signaling activity is associated, depending on the context, with both cancer initiation and progression. Interestingly, the Hh pathway is remarkably druggable, raising hopes that inhibition of the pathway could support anticancer therapy. Indeed, a large body of preclinical data supports such an action, but promising clinical data are still limited to basal cell carcinoma (BSC) and medulloblastoma. Nevertheless cancer resistance against Hh targeting has already emerged as a major problem. Here we shall review the current situation with respect to targeting the Hh pathway in cancer in general and in chemotolerance in particular with a focus on the problems associated with the emergence of tumors resistant to treatment with inhibitors targeting the Hh receptor Smoothened (SMO).
Collapse
Affiliation(s)
- Karla C S Queiroz
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | |
Collapse
|
532
|
Crews LA, Jamieson CHM. Selective elimination of leukemia stem cells: hitting a moving target. Cancer Lett 2012; 338:15-22. [PMID: 22906415 DOI: 10.1016/j.canlet.2012.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/27/2012] [Accepted: 08/07/2012] [Indexed: 01/02/2023]
Abstract
Despite the widespread use of chemotherapeutic cytotoxic agents that eradicate proliferating cell populations, patients suffering from a wide variety of malignancies continue to relapse as a consequence of resistance to standard therapies. In hematologic malignancies, leukemia stem cells (LSCs) represent a malignant reservoir of disease that is believed to drive relapse and resistance to chemotherapy and tyrosine kinase inhibitor (TKIs). Major research efforts in recent years have been aimed at identifying and characterizing the LSC population in leukemias, such as chronic myeloid leukemia (CML), which represents an important paradigm for understanding the molecular evolution of cancer. However, the precise molecular mechanisms that promote LSC-mediated therapeutic recalcitrance have remained elusive. It has become clear that the LSC population evolves during disease progression, thus presenting a serious challenge for development of effective therapeutic strategies. Multiple reports have demonstrated that LSC initiation and propagation occurs as a result of aberrant activation of pro-survival and self-renewal pathways regulated by stem-cell related signaling molecules including β-catenin and Sonic Hedgehog (Shh). Enhanced survival in LSC protective microenvironments, such as the bone marrow niche, as well as acquired dormancy of cells in these niches, also contributes to LSC persistence. Key components of these cell-intrinsic and cell-extrinsic pathways provide novel potential targets for therapies aimed at eradicating this dynamic and therapeutically recalcitrant LSC population. Furthermore, combination strategies that exploit LSC have the potential to dramatically improve the quality and quantity of life for patients that are resistant to current therapies.
Collapse
Affiliation(s)
- Leslie A Crews
- Department of Medicine, Stem Cell Program and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
533
|
Kelleher FC, Cain JE, Healy JM, Watkins DN, Thomas DM. Prevailing importance of the hedgehog signaling pathway and the potential for treatment advancement in sarcoma. Pharmacol Ther 2012; 136:153-68. [PMID: 22906929 DOI: 10.1016/j.pharmthera.2012.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 07/18/2012] [Indexed: 12/19/2022]
Abstract
The hedgehog signaling pathway is important in embryogenesis and post natal development. Constitutive activation of the pathway due to mutation of pathway components occurs in ~25% of medulloblastomas and also in basal cell carcinomas. In many other malignancies the therapeutic role for hedgehog inhibition though intriguing, based on preclinical data, is far from assured. Hedgehog inhibition is not an established part of the treatment paradigm of sarcoma but the scientific rationale for a possible benefit is compelling. In chondrosarcoma there is evidence of hedgehog pathway activation and an ontologic comparison between growth plate chondrocyte differentiation and different chondrosarcoma subtypes. Immunostaining epiphyseal growth plate for Indian hedgehog is particularly positive in the zone of pre-hypertrophic chondrocytes which correlates ontologically with conventional chondrosarcoma. In Ewing sarcoma/PNET tumors the Gli1 transcription factor is a direct target of the EWS-FLI1 oncoprotein present in 85% of cases. In many cases of rhabdomyosarcomas there is increased expression of Gli1 (Ragazzini et al., 2004). Additionally, a third of embryonal rhabdomyosarcomas have loss of Chr.9q22 that encompasses the patched locus (Bridge et al., 2000). The potential to treat osteosarcoma by inhibition of Gli2 and the role of the pathway in ovarian fibromas and other connective tissue tumors is also discussed (Nagao et al., 2011; Hirotsu et al., 2010). Emergence of acquired secondary resistance to targeted therapeutics is an important issue that is also relevant to hedgehog inhibition. In this context secondary resistance of medulloblastomas to treatment with a smoothened antagonist in two tumor mouse models is examined.
Collapse
Affiliation(s)
- Fergal C Kelleher
- Sarcoma Service, Peter MacCallum Cancer Centre, 12 St. Andrew's Place, A'Beckitt Street, Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|
534
|
Abstract
Basal cell carcinoma (BCC) is the most common tumor of the skin and can result in significant morbidity as well as costs. The discovery of aberrant Hedgehog (HH) signaling in patients with genetic propensity to develop BCCs has resulted in a better understanding of the genetic abnormalities leading to the development of BCC. The current state of knowledge with regard to the genetics of BCC is discussed. Existing therapies are reviewed, in particular new targeted therapies to the HH signaling pathway that have resulted in a landmark breakthrough for patients with refractory BCC. Data from recent and ongoing trials are presented.
Collapse
|
535
|
Abstract
The complexity of cancer chemotherapy requires pharmacists be familiar with the complicated regimens and highly toxic agents used. This column reviews various issues related to preparation, dispensing, and administration of antineoplastic therapy, and the agents, both commercially available and investigational, used to treat malignant diseases.
Collapse
Affiliation(s)
- Aubrey Waddell
- University of Tennessee College of Pharmacy; Oncology Pharmacist, Pharmacy Department, Blount Memorial Hospital, 907 E. Lamar Alexander Parkway, Maryville, TN 37804
| | - Dominic A. Solimando
- Oncology Pharmacy Services, Inc., 4201 Wilson Boulevard #110-545, Arlington, VA 22203
| |
Collapse
|
536
|
Abstract
The Hedgehog (Hh) signaling pathway has been implicated in tumor initiation and metastasis across different malignancies. Major mechanisms by which the Hh pathway is aberrantly activated can be attributed to mutations of members of Hh pathway or excessive/inappropriate expression of Hh pathway ligands. The Hh signaling pathway also affects the regulation of cancer stem cells, leading to their capabilities in tumor formation, disease progression, and metastasis. Preliminary results of early phase clinical trials of Hh inhibitors administered as monotherapy demonstrated promising results in patients with basal cell carcinoma and medulloblastoma, but clinically meaningful anticancer efficacy across other tumor types seems to be lacking. Additionally, cases of resistance have been already observed. Mutations of SMO, activation of Hh pathway components downstream to SMO, and upregulation of alternative signaling pathways are possible mechanisms of resistance development. Determination of effective Hh inhibitor-based combination regimens and development of correlative biomarkers relevant to this pathway should remain as clear priorities for future research.
Collapse
Affiliation(s)
- Solmaz Sahebjam
- Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
537
|
Okabe S, Tauchi T, Tanaka Y, Katagiri S, Ohyashiki K. Effects of the hedgehog inhibitor GDC-0449, alone or in combination with dasatinib, on BCR-ABL-positive leukemia cells. Stem Cells Dev 2012; 21:2939-48. [PMID: 22642671 DOI: 10.1089/scd.2012.0016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hedgehog (Hh)-glioma-associated oncogene homolog (Gli) signaling is implicated in a large number of human cancers such as leukemia. In this study, we investigated the effects of the potent Hh antagonist GDC-0449 on the BCR-ABL-positive cell line OM9;22 and primary samples when leukemia cells were protected by a feeder cell line (S9 cells). The numbers of OM9;22 cells significantly increased with S9 cells. Treatment of OM9;22 cells with GDC-0449 caused cell growth inhibition and induced apoptosis. Moreover, GDC-0449 inhibited the colony growth of Philadelphia chromosome (Ph)-positive primary samples. We next investigated the effects of a combination of GDC-0449 and dasatinib on these cell lines. The growth inhibition typically promoted by dasatinib was significantly reduced in the presence of S9 cells. Treatment of Ph-positive leukemia cells with GDC-0449 and dasatinib in the presence of S9 caused significantly more cytotoxicity than that caused by each drug alone. Inhibition of Gli1 or Gli2 by siRNA transfection reduced the growth of the Ph-positive cell line K562 and increased cytotoxicity of dasatinib. Moreover, colony formations of Gli1 or Gli2 knockdown cells were also reduced. Data from this study suggest that administration of the Hh inhibitor GDC-0449 inhibits BCR-ABL-positive cell growth and enhances the cytotoxic effects of dasatinib in the presence of feeder cells.
Collapse
Affiliation(s)
- Seiichi Okabe
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
538
|
Abstract
Primary brain tumours are difficult to manage clinically due to their abilities to invade adjacent tissue and infiltrate distant neuropil. These contribute to challenges in surgical management and also limit the effectiveness of radiotherapy. Despite initial responses to chemotherapy, most tumours become chemo-resistant, leading to relapse. Recent identification and isolation of brain cancer stem cells (BCSCs) have broadened our understanding of the molecular pathogenesis and potential Achilles' heel of brain tumours. BCSCs are thought to drive and propagate the tumour and therefore present an important target for further investigations. This review explores the history of the discovery of BCSCs and the evolving concept of "cancer stem cells" in neuro-oncology. We attempt to present a balanced view on the subject and also to update the readers on the molecular biology of BCSCs. Lastly, we outline the potential strategies to target BCSCs which will translate into specific and effective therapies for brain tumours.
Collapse
|
539
|
Topical delivery of siRNA-based spherical nucleic acid nanoparticle conjugates for gene regulation. Proc Natl Acad Sci U S A 2012; 109:11975-80. [PMID: 22773805 DOI: 10.1073/pnas.1118425109] [Citation(s) in RCA: 326] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Topical application of nucleic acids offers many potential therapeutic advantages for suppressing genes in the skin, and potentially for systemic gene delivery. However, the epidermal barrier typically precludes entry of gene-suppressing therapy unless the barrier is disrupted. We now show that spherical nucleic acid nanoparticle conjugates (SNA-NCs), gold cores surrounded by a dense shell of highly oriented, covalently immobilized siRNA, freely penetrate almost 100% of keratinocytes in vitro, mouse skin, and human epidermis within hours after application. Significantly, these structures can be delivered in a commercial moisturizer or phosphate-buffered saline, and do not require barrier disruption or transfection agents, such as liposomes, peptides, or viruses. SNA-NCs targeting epidermal growth factor receptor (EGFR), an important gene for epidermal homeostasis, are > 100-fold more potent and suppress longer than siRNA delivered with commercial lipid agents in cultured keratinocytes. Topical delivery of 1.5 uM EGFR siRNA (50 nM SNA-NCs) for 3 wk to hairless mouse skin almost completely abolishes EGFR expression, suppresses downstream ERK phosphorylation, and reduces epidermal thickness by almost 40%. Similarly, EGFR mRNA in human skin equivalents is reduced by 52% after 60 h of treatment with 25 nM EGFR SNA-NCs. Treated skin shows no clinical or histological evidence of toxicity. No cytokine activation in mouse blood or tissue samples is observed, and after 3 wk of topical skin treatment, the SNA structures are virtually undetectable in internal organs. SNA conjugates may be promising agents for personalized, topically delivered gene therapy of cutaneous tumors, skin inflammation, and dominant negative genetic skin disorders.
Collapse
|
540
|
Demeure MJ, Craig DW, Sinari S, Moses TM, Christoforides A, Dinh J, Izatt T, Aldrich J, Decker A, Baker A, Cherni I, Watanabe A, Koep L, Lake D, Hostetter G, Trent JM, Von Hoff DD, Carpten JD. Cancer of the ampulla of Vater: analysis of the whole genome sequence exposes a potential therapeutic vulnerability. Genome Med 2012; 4:56. [PMID: 22762308 PMCID: PMC3580412 DOI: 10.1186/gm357] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 05/29/2011] [Accepted: 07/04/2012] [Indexed: 12/21/2022] Open
Abstract
Background Recent advances in the treatment of cancer have focused on targeting genomic aberrations with selective therapeutic agents. In rare tumors, where large-scale clinical trials are daunting, this targeted genomic approach offers a new perspective and hope for improved treatments. Cancers of the ampulla of Vater are rare tumors that comprise only about 0.2% of gastrointestinal cancers. Consequently, they are often treated as either distal common bile duct or pancreatic cancers. Methods We analyzed DNA from a resected cancer of the ampulla of Vater and whole blood DNA from a 63 year-old man who underwent a pancreaticoduodenectomy by whole genome sequencing, achieving 37× and 40× coverage, respectively. We determined somatic mutations and structural alterations. Results We identified relevant aberrations, including deleterious mutations of KRAS and SMAD4 as well as a homozygous focal deletion of the PTEN tumor suppressor gene. These findings suggest that these tumors have a distinct oncogenesis from either common bile duct cancer or pancreatic cancer. Furthermore, this combination of genomic aberrations suggests a therapeutic context for dual mTOR/PI3K inhibition. Conclusions Whole genome sequencing can elucidate an oncogenic context and expose potential therapeutic vulnerabilities in rare cancers.
Collapse
Affiliation(s)
- Michael J Demeure
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA ; Virginia G Piper Cancer Center, 10290 N. 92nd St, Scottsdale, AZ 85258, USA
| | - David W Craig
- Neurogenomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Shripad Sinari
- Neurogenomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Tracy M Moses
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Alexis Christoforides
- Neurogenomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Jennifer Dinh
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Tyler Izatt
- Neurogenomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Jessica Aldrich
- Neurogenomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Ardis Decker
- Clinical Translational Research Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Angela Baker
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Irene Cherni
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - April Watanabe
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Lawrence Koep
- Department of Surgery, Banner Good Samaritan Medical Center, 1111 E. McDowell Rd, Phoenix, AZ 85006, USA
| | - Douglas Lake
- School of Life Sciences, Arizona State University, 427 E. Tyler Mall MC 5401 SOLS, Phoenix, AZ 85287, USA
| | - Galen Hostetter
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Jeffrey M Trent
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - Daniel D Von Hoff
- Virginia G Piper Cancer Center, 10290 N. 92nd St, Scottsdale, AZ 85258, USA ; Clinical Translational Research Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| | - John D Carpten
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Ave, Phoenix, AZ 85004, USA
| |
Collapse
|
541
|
Sotiropoulou PA, Blanpain C. Development and homeostasis of the skin epidermis. Cold Spring Harb Perspect Biol 2012; 4:a008383. [PMID: 22751151 DOI: 10.1101/cshperspect.a008383] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The skin epidermis is a stratified epithelium that forms a barrier that protects animals from dehydration, mechanical stress, and infections. The epidermis encompasses different appendages, such as the hair follicle (HF), the sebaceous gland (SG), the sweat gland, and the touch dome, that are essential for thermoregulation, sensing the environment, and influencing social behavior. The epidermis undergoes a constant turnover and distinct stem cells (SCs) are responsible for the homeostasis of the different epidermal compartments. Deregulation of the signaling pathways controlling the balance between renewal and differentiation often leads to cancer formation.
Collapse
|
542
|
Seke Etet PF, Vecchio L, Nwabo Kamdje AH. Interactions between bone marrow stromal microenvironment and B-chronic lymphocytic leukemia cells: any role for Notch, Wnt and Hh signaling pathways? Cell Signal 2012; 24:1433-1443. [PMID: 22446006 DOI: 10.1016/j.cellsig.2012.03.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 03/05/2012] [Indexed: 01/02/2023]
Abstract
B-cell chronic lymphocytic leukemia (CLL), which is the most common lymphoproliferative disorder, displays characteristics consistent with a defect in programmed cell death and exhibit prolonged survival of affected cells in vivo. When recovered from peripheral blood or lymphoid tissues of patients and cultured in vitro, CLL malignant cells rapidly undergo spontaneous apoptosis. CLL B-cells co-culture with different adherent cell types, collectively referred to as stromal cells, induces leukemia cell survival, migration, and drug resistance. In addition, such survival-promoting microenvironments can rescue leukemia cells from cytotoxic therapy, giving way to disease relapse. Quite surprisingly considering that many anti-cancer drugs, including γ-secretase inhibitors, Cyclopamine and Quercetin, were reported to block Notch, Wnt, and Hedgehog anti-apoptotic signaling pathways respectively, the link between the latter anti-apoptotic pathways and bone marrow stromal cells in CLL has been pointed out only recently. Data concerning the pathogenesis of CLL have been critically reviewed in regards to the growing body of evidence indicating deregulations of Notch, Wnt and Hedgehog anti-apoptotic signaling pathways in the stromal microenvironment of affected cells.
Collapse
MESH Headings
- Bone Marrow/metabolism
- Cellular Microenvironment
- Gene Expression Regulation, Leukemic/genetics
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Molecular Targeted Therapy
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/metabolism
- Smoothened Receptor
- Stromal Cells/cytology
- Stromal Cells/metabolism
- Wnt Signaling Pathway
Collapse
|
543
|
Discovery of novel hedgehog antagonists from cell-based screening: Isosteric modification of p38 bisamides as potent inhibitors of SMO. Bioorg Med Chem Lett 2012; 22:4907-11. [DOI: 10.1016/j.bmcl.2012.04.104] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 04/19/2012] [Accepted: 04/23/2012] [Indexed: 01/06/2023]
|
544
|
Rodriguez-Blanco J, Schilling NS, Tokhunts R, Giambelli C, Long J, Liang Fei D, Singh S, Black KE, Wang Z, Galimberti F, Bejarano PA, Elliot S, Glassberg MK, Nguyen DM, Lockwood WW, Lam WL, Dmitrovsky E, Capobianco AJ, Robbins DJ. The hedgehog processing pathway is required for NSCLC growth and survival. Oncogene 2012; 32:2335-45. [PMID: 22733134 DOI: 10.1038/onc.2012.243] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Considerable interest has been generated from the results of recent clinical trials using smoothened (SMO) antagonists to inhibit the growth of hedgehog (HH) signaling-dependent tumors. This interest is tempered by the discovery of SMO mutations mediating resistance, underscoring the rationale for developing therapeutic strategies that interrupt HH signaling at levels distinct from those inhibiting SMO function. Here, we demonstrate that HH-dependent non-small cell lung carcinoma (NSCLC) growth is sensitive to blockade of the HH pathway upstream of SMO, at the level of HH ligand processing. Individually, the use of different lentivirally delivered shRNA constructs targeting two functionally distinct HH-processing proteins, skinny hedgehog (SKN) or dispatched-1 (DISP-1), in NSCLC cell lines produced similar decreases in cell proliferation and increased cell death. Further, providing either an exogenous source of processed HH or a SMO agonist reverses these effects. The attenuation of HH processing, by knocking down either of these gene products, also abrogated tumor growth in mouse xenografts. Finally, we extended these findings to primary clinical specimens, showing that SKN is frequently overexpressed in NSCLC and that higher DISP-1 expression is associated with an unfavorable clinical outcome. Our results show a critical role for HH processing in HH-dependent tumors, identifies two potential druggable targets in the HH pathway, and suggest that similar therapeutic strategies could be explored to treat patients harboring HH ligand-dependent cancers.
Collapse
Affiliation(s)
- J Rodriguez-Blanco
- Molecular Oncology Program, Department of Surgery, University of Miami, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
545
|
Abstract
PURPOSE OF REVIEW Drug development in oncology finds itself at the crossroad of unique opportunities and major challenges. The old paradigms should and can be replaced by a system that better matches the right patients to the right compounds and puts much more emphasis on the early stages of drug development. RECENT FINDINGS The clinical phases of drug development will no longer be split into phase I, II, and III studies, but rather into 'functional target pharmacology studies', followed by 'proof of concept studies'. The resulting development flow becomes Apollo-capsule shaped. SUMMARY Although randomized studies will still be needed for drugs using targets in the tumor environment, or for combinations of agents, drug registration might proceed without these if all of the following criteria are met in early development: availability of preclinical convincing evidence that the drug's target is the functional driver behind the disease phenotype, availability of a predictive biomarker that enables appropriate and actual patient selection in early pharmacology studies, a Response Evaluation Criteria In Solid Tumors (RECIST)-based single agent response rate of at least 50%, and/or a progression at first tumor assessment rate of 15% or less, a duration of absence of progression (stable disease) beyond doubt and considered clinically relevant, and no major safety concern. This set is not yet mature, but may be adapted over time. The concerns related to registering agents on the basis of small datasets can be adequately addressed by obligatory postmarketing hypothesis driven studies.
Collapse
|
546
|
Abstract
The Hedgehog (Hh) signaling pathway regulates embryonic development and may be aberrantly activated in a wide variety of human cancers. Efforts to target pathogenic Hh signaling have steadily progressed from the laboratory to the clinic, and the recent approval of the Hh pathway inhibitor vismodegib for patients with advanced basal cell carcinoma represents an important milestone. On the other hand, Hh pathway antagonists have failed to show significant clinical activity in other solid tumors. The reasons for these negative results are not precisely understood, but it is possible that the impact of Hh pathway inhibition has not been adequately measured by the clinical endpoints used thus far or that aberrancies in Hh signal transduction limits the activity of currently available pathway antagonists. Further basic and correlative studies to better understand Hh signaling in human tumors and validate putative antitumor mechanisms in the clinical setting may ultimately improve the success of Hh pathway inhibition to other tumor types.
Collapse
Affiliation(s)
- Ross McMillan
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, CRB245, 1650 Orleans Street, Baltimore, MD 21287, USA
| | | |
Collapse
|
547
|
A paradigm shift in tumour response evaluation of targeted therapy: the assessment of novel drugs in exploratory clinical trials. Curr Opin Oncol 2012; 24:338-44. [PMID: 22418613 DOI: 10.1097/cco.0b013e3283528b73] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To describe the difficulty in assessing the biological activity of a novel agent in phase II trials. RECENT FINDINGS Two major fields of research provide interesting new potential endpoints: endpoints based on new imaging techniques (e.g. PET or spectral imaging that explore tumour metabolism, dynamic contrast enhanced (DCE) ultrasonography or DCE-MRI that explore tumour vascularization and tumour growth inhibition) and endpoints integrating assessment of tumour burden across time, such as the growth modulation index. SUMMARY Most of the recently described techniques appear attractive, but require formal validation.
Collapse
|
548
|
Hedgehog signaling: from the cuirass to the heart of pancreatic cancer. Pancreatology 2012; 12:388-93. [PMID: 22898642 DOI: 10.1016/j.pan.2012.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 05/28/2012] [Accepted: 06/08/2012] [Indexed: 12/11/2022]
Abstract
Exocrine pancreatic cancer is the fifth cause of cancer-related death in Europe and carries a very poor prognosis for all disease stages. To date no medical treatment has significantly increased patients' survival. One of the reasons for pancreatic cancer's chemoresistence is the complex tumor architecture: cancer cells are surrounded by a dense desmoplastic stroma that blocks drug delivery. Moreover, pancreatic cancer is characterized by a marked heterogeneity of cells, including cancer stem cells (CSCs) that act as tumor-initiating cells and hierarchically control the differentiated cancer cells. In particular, this subpopulation is resistant to classic cytotoxic therapies, and seems to be responsible for disease renewal. Hedgehog signaling (HH) is implicated in pancreatic gland development during embryogenesis and is reactivated during tumorigenesis and the maintenance of pancreatic cancer. Some studies demonstrated that the Hedgehog-secreted signaling proteins are overexpressed in both the stromal and CSCs pools, implying an abnormal activation of HH in the main compartment of pancreatic cancer. For this reason, the Hedgehog pathway could be an interesting target for clinical trials to increase drug concentration in neoplastic cells and hence deplete the stroma and directly kill tumor-initiating cells.
Collapse
|
549
|
Wang Y, Arvanites AC, Davidow L, Blanchard J, Lam K, Yoo JW, Coy S, Rubin LL, McMahon AP. Selective identification of hedgehog pathway antagonists by direct analysis of smoothened ciliary translocation. ACS Chem Biol 2012; 7:1040-8. [PMID: 22554036 DOI: 10.1021/cb300028a] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hedgehog (Hh) signaling promotes tumorigenesis. The accumulation of the membrane protein Smoothened (Smo) within the primary cilium (PC) is a key event in Hh signal transduction, and many pharmacological inhibitors identified to date target Smo's actions. Smo ciliary translocation is inhibited by some pathway antagonists, while others promote ciliary accumulation, an outcome that can lead to a hypersensitive state on renewal of Hh signaling. To identify novel inhibitory compounds acting on the critical mechanistic transition of Smo accumulation, we established a high content screen to directly analyze Smo ciliary translocation. Screening thousands of compounds from annotated libraries of approved drugs and other agents, we identified several new classes of compounds that block Sonic hedgehog-driven Smo localization within the PC. Selective analysis was conducted on two classes of Smo antagonists. One of these, DY131, appears to inhibit Smo signaling through a common binding site shared by previously reported Smo agonists and antagonists. Antagonism by this class of compound is competed by high doses of Smo-binding agonists such as SAG and impaired by a mutation that generates a ligand-independent, oncogenic form of Smo (SmoM2). In contrast, a second antagonist of Smo accumulation within the PC, SMANT, was less sensitive to SAG-mediated competition and inhibited SmoM2 at concentrations similar to those that inhibit wild-type Smo. Our observations identify important differences among Hh antagonists and the potential for development of novel therapeutic approaches against mutant forms of Smo that are resistant to current therapeutic strategies.
Collapse
Affiliation(s)
- Yu Wang
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Anthony C. Arvanites
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Lance Davidow
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Joel Blanchard
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Kelvin Lam
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Jin Woo Yoo
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Shannon Coy
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Lee L. Rubin
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| | - Andrew P. McMahon
- Department
of Stem Cell and Regenerative Biology, ‡Department of Molecular and Cellular
Biology, §Harvard Stem Cell Institute, ∥Department of Chemistry and Chemical Biology, and ⊥Harvard College, Harvard University, Cambridge, Massachusetts
02138, United States
| |
Collapse
|
550
|
Jain RK, Lee JJ, Ng C, Hong D, Gong J, Naing A, Wheler J, Kurzrock R. Change in tumor size by RECIST correlates linearly with overall survival in phase I oncology studies. J Clin Oncol 2012; 30:2684-90. [PMID: 22689801 DOI: 10.1200/jco.2011.36.4752] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE RECIST is used to quantify tumor changes during exposure to anticancer agents. Responses are categorized as complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD). Clinical trials dictate a patient's management options based on the category into which his or her response falls. However, the association between response and survival is not well studied in the early trial setting. PATIENTS AND METHODS To study the correlation between response as quantified by RECIST and overall survival (OS, the gold-standard survival outcome), we analyzed 570 participants of 24 phase I trials conducted between October 2004 and May 2009, of whom 468 had quantifiable changes in tumor size. Analyses of Kaplan-Meier estimates of OS by response and null Martingale residuals of Cox models were the primary outcome measures. All analyses are landmark analyses. RESULTS Kaplan-Meier analyses revealed strong associations between change in tumor size by RECIST and survival (P = 4.5 × 10(-6) to < 1 × 10(-8)). The relationship was found to be near-linear (R(2) = 0.75 to 0.92) and confirmed by the residual analyses. No clear inflection points were found to exist in the relationship between tumor size changes and survival. CONCLUSION RECIST quantification of response correlates with survival, validating RECIST's use in phase I trials. However, the lack of apparent boundary values in the relationship between change in tumor size and OS demonstrates the arbitrary nature of the CR/PR/SD/PD categories and questions emphasis placed on this categorization scheme. Describing tumor responses as a continuous variable may be more informative than reporting categoric responses when evaluating novel anticancer therapies.
Collapse
Affiliation(s)
- Rajul K Jain
- Amgen, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | | | | | |
Collapse
|