251
|
Pietanza MC, Litvak AM, Varghese AM, Krug LM, Fleisher M, Teitcher JB, Holodny AI, Sima CS, Woo KM, Ng KK, Won HH, Berger MF, Kris MG, Rudin CM. A phase I trial of the Hedgehog inhibitor, sonidegib (LDE225), in combination with etoposide and cisplatin for the initial treatment of extensive stage small cell lung cancer. Lung Cancer 2016; 99:23-30. [PMID: 27565909 DOI: 10.1016/j.lungcan.2016.04.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/15/2016] [Accepted: 04/23/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVES The Hedgehog pathway has been implicated in small cell lung cancer (SCLC) tumor initiation and progression. Pharmacologic blockade of the key Hedgehog regulator, Smoothened, may inhibit these processes. We performed a phase I study to determine the maximum tolerated dose (MTD) of sonidegib (LDE225), a selective, oral Smoothened antagonist, in combination with etoposide/cisplatin in newly diagnosed patients with extensive stage SCLC. MATERIALS AND METHODS Patients received 4-6 21-day cycles of etoposide/cisplatin with daily sonidegib. Patients with response or stable disease were continued on sonidegib until disease progression or unacceptable toxicity. Two dose levels of sonidegib were planned: 400mg and 800mg daily, with 200mg daily de-escalation if necessary. Next generation sequencing was performed on available specimens. Circulating tumor cells (CTCs) were quantified at baseline and with disease evaluation. RESULTS Fifteen patients were enrolled. 800mg was established as the recommended phase II dose of sonidegib in combination with etoposide/cisplatin. Grade 3 or greater toxicities included: anemia (n=5), neutropenia (n=8), CPK elevation (n=2), fatigue (n=2), and nausea (n=2). Toxicity led to removal of one patient from study. Partial responses were confirmed in 79% (11/14; 95% CI: 49-95%). One patient with SOX2 amplification remains progression-free on maintenance sonidegib after 27 months. CTC count, at baseline, was associated with the presence of liver metastases and after 1 cycle of therapy, with overall survival. CONCLUSIONS Sonidegib 800mg daily was the MTD when administered with EP. Further genomic characterization of exceptional responders may reveal clinically relevant predictive biomarkers that could tailor use in patients most likely to benefit.
Collapse
Affiliation(s)
- M Catherine Pietanza
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States.
| | - Anya M Litvak
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States
| | - Anna M Varghese
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Lee M Krug
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States
| | - Martin Fleisher
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jerrold B Teitcher
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Andrei I Holodny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Cami S Sima
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kaitlin M Woo
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kenneth K Ng
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States
| | - Helen H Won
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Michael F Berger
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Mark G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States
| | - Charles M Rudin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
252
|
Cohen PR, Tomson BN, Elkin SK, Marchlik E, Carter JL, Kurzrock R. Genomic portfolio of Merkel cell carcinoma as determined by comprehensive genomic profiling: implications for targeted therapeutics. Oncotarget 2016; 7:23454-67. [PMID: 26981779 PMCID: PMC5029639 DOI: 10.18632/oncotarget.8032] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/28/2016] [Indexed: 12/22/2022] Open
Abstract
Merkel cell carcinoma is an ultra-rare cutaneous neuroendocrine cancer for which approved treatment options are lacking. To better understand potential actionability, the genomic landscape of Merkel cell cancers was assessed. The molecular aberrations in 17 patients with Merkel cell carcinoma were, on physician request, tested in a Clinical Laboratory Improvement Amendments (CLIA) laboratory (Foundation Medicine, Cambridge, MA) using next-generation sequencing (182 or 236 genes) and analyzed by N-of-One, Inc. (Lexington, MA). There were 30 genes harboring aberrations and 60 distinct molecular alterations identified in this patient population. The most common abnormalities involved the TP53 gene (12/17 [71% of patients]) and the cell cycle pathway (CDKN2A/B, CDKN2C or RB1) (12/17 [71%]). Abnormalities also were observed in the PI3K/AKT/mTOR pathway (AKT2, FBXW7, NF1, PIK3CA, PIK3R1, PTEN or RICTOR) (9/17 [53%]) and DNA repair genes (ATM, BAP1, BRCA1/2, CHEK2, FANCA or MLH1) (5/17 [29%]). Possible cognate targeted therapies, including FDA-approved drugs, could be identified in most of the patients (16/17 [94%]). In summary, Merkel cell carcinomas were characterized by multiple distinct aberrations that were unique in the majority of analyzed cases. Most patients had theoretically actionable alterations. These results provide a framework for investigating tailored combinations of matched therapies in Merkel cell carcinoma patients.
Collapse
Affiliation(s)
- Philip R. Cohen
- Department of Dermatology, University of California San Diego, San Diego, CA, USA
| | | | | | | | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, San Diego, CA, USA
| |
Collapse
|
253
|
Hempel JE, Cadar AG, Hong CC. Development of thieno- and benzopyrimidinone inhibitors of the Hedgehog signaling pathway reveals PDE4-dependent and PDE4-independent mechanisms of action. Bioorg Med Chem Lett 2016; 26:1947-53. [PMID: 26976215 PMCID: PMC5147493 DOI: 10.1016/j.bmcl.2016.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/04/2016] [Accepted: 03/05/2016] [Indexed: 10/22/2022]
Abstract
From a high content in vivo screen for modulators of developmental patterning in embryonic zebrafish, we previously identified eggmanone (EGM1, 3) as a Hedgehog (Hh) signaling inhibitor functioning downstream of Smoothened. Phenotypic optimization studies for in vitro probe development utilizing a Gli transcription-linked stable luciferase reporter cell line identified EGM1 analogs with improved potency and aqueous solubility. Mechanistic profiling of optimized analogs indicated two distinct scaffold clusters: PDE4 inhibitors able to inhibit downstream of Sufu, and PDE4-independent Hh inhibitors functioning between Smo and Sufu. Each class represents valuable in vitro probes for elucidating the complex mechanisms of Hh regulation.
Collapse
Affiliation(s)
- Jonathan E Hempel
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, PRB 383, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, 896 Preston Research Building, Nashville, TN 37232, USA
| | - Adrian G Cadar
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, PRB 383, Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
| | - Charles C Hong
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, PRB 383, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, 896 Preston Research Building, Nashville, TN 37232, USA; Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
254
|
Macdonald TJ. Hedgehog Pathway in Pediatric Cancers: They're Not Just for Brain Tumors Anymore. Am Soc Clin Oncol Educ Book 2016:605-9. [PMID: 24451804 DOI: 10.14694/edbook_am.2012.32.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Hedgehog (HH) pathway regulates fundamental processes in embryonic development, including stem cell maintenance, cell differentiation, tissue polarity, and cell proliferation. In the vertebrate pathway, Sonic hedgehog (SHH) binds to Patched1 (PTCH1), which relieves its inhibition of Smoothened (SMO), allowing the GLI family of transcription factors to translocate to the nucleus and activate HH target genes such as GLI1, GLI2, PTCH1, CYCLIN D1, BCL-2, and MYCN. The HH pathway is also an active participant in tumorigenesis. In 1996, loss-of-function mutation in PTCH1 was discovered to be the cause of nevoid basal cell carcinoma syndrome (NBCCS, or Gorlin syndrome), an autosomal dominant disease associated with increased rates of basal cell carcinoma (BCC), medulloblastoma (MB), and rarely, rhabdomyosarcoma. It is now estimated that 100% of sporadic BCC and up to 20% to 30% of MB also harbor activating HH pathway mutations. Together, these discoveries firmly established the linkage between HH pathway activation and cancer development. Intense research has since been focused on further defining the role of the HH pathway in BCC and MB and potential therapeutic strategies to inhibit HH signaling. Early clinical trials of SMO inhibitors have shown promising results in the treatment of adult BCC and SHH-driven MB. More recently, a number of other pediatric cancers have been reported to show HH activity, making these tumors potential candidates for HH inhibitor therapy. To date however, no HH pathway mutations have been identified in other pediatric cancers. This review will describe the HH pathway signaling in development and cancer with a focus on recent evidence for HH pathway activation in central nervous system (CNS) and non-CNS pediatric cancers.
Collapse
Affiliation(s)
- Tobey J Macdonald
- From the Pediatric Neuro-Oncology Program, Aflac Cancer Center and Blood Disorders Service, Children's Healthcare of Atlanta, and Emory University School of Medicine, Emory Children's Center, Atlanta, GA
| |
Collapse
|
255
|
Rehefeldt-Erne S, Nägeli MC, Winterton N, Felderer L, Weibel L, Hafner J, Dummer R. Nevoid Basal Cell Carcinoma Syndrome: Report from the Zurich Nevoid Basal Cell Carcinoma Syndrome Cohort. Dermatology 2016; 232:285-92. [DOI: 10.1159/000444792] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 02/05/2016] [Indexed: 11/19/2022] Open
|
256
|
Bonilla X, Parmentier L, King B, Bezrukov F, Kaya G, Zoete V, Seplyarskiy VB, Sharpe HJ, McKee T, Letourneau A, Ribaux PG, Popadin K, Basset-Seguin N, Ben Chaabene R, Santoni FA, Andrianova MA, Guipponi M, Garieri M, Verdan C, Grosdemange K, Sumara O, Eilers M, Aifantis I, Michielin O, de Sauvage FJ, Antonarakis SE, Nikolaev SI. Genomic analysis identifies new drivers and progression pathways in skin basal cell carcinoma. Nat Genet 2016; 48:398-406. [PMID: 26950094 DOI: 10.1038/ng.3525] [Citation(s) in RCA: 361] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/11/2016] [Indexed: 12/13/2022]
Abstract
Basal cell carcinoma (BCC) of the skin is the most common malignant neoplasm in humans. BCC is primarily driven by the Sonic Hedgehog (Hh) pathway. However, its phenotypic variation remains unexplained. Our genetic profiling of 293 BCCs found the highest mutation rate in cancer (65 mutations/Mb). Eighty-five percent of the BCCs harbored mutations in Hh pathway genes (PTCH1, 73% or SMO, 20% (P = 6.6 × 10(-8)) and SUFU, 8%) and in TP53 (61%). However, 85% of the BCCs also harbored additional driver mutations in other cancer-related genes. We observed recurrent mutations in MYCN (30%), PPP6C (15%), STK19 (10%), LATS1 (8%), ERBB2 (4%), PIK3CA (2%), and NRAS, KRAS or HRAS (2%), and loss-of-function and deleterious missense mutations were present in PTPN14 (23%), RB1 (8%) and FBXW7 (5%). Consistent with the mutational profiles, N-Myc and Hippo-YAP pathway target genes were upregulated. Functional analysis of the mutations in MYCN, PTPN14 and LATS1 suggested their potential relevance in BCC tumorigenesis.
Collapse
Affiliation(s)
- Ximena Bonilla
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | | | - Bryan King
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Fedor Bezrukov
- Department of Physics, University of Connecticut, Storrs, Connecticut, USA
- RIKEN BNL Research Center, Brookhaven National Laboratory, Upton, New York, USA
| | - Gürkan Kaya
- Department of Dermatology, University Hospitals of Geneva, Geneva, Switzerland
| | - Vincent Zoete
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vladimir B Seplyarskiy
- Institute of Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Hayley J Sharpe
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California, USA
| | - Thomas McKee
- Service of Clinical Pathology, University Hospitals of Geneva, Geneva, Switzerland
| | - Audrey Letourneau
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Pascale G Ribaux
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Konstantin Popadin
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Nicole Basset-Seguin
- Department of Dermatology, Saint Louis Hospital, Paris 7 University, Paris, France
| | - Rouaa Ben Chaabene
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Federico A Santoni
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Maria A Andrianova
- Institute of Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Michel Guipponi
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Marco Garieri
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Carole Verdan
- Service of Clinical Pathology, University Hospitals of Geneva, Geneva, Switzerland
| | - Kerstin Grosdemange
- Department of Dermatology, University Hospitals of Geneva, Geneva, Switzerland
| | - Olga Sumara
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Olivier Michielin
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Frederic J de Sauvage
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California, USA
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Sergey I Nikolaev
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
257
|
Filocamo G, Brunetti M, Colaceci F, Sasso R, Tanori M, Pasquali E, Alfonsi R, Mancuso M, Saran A, Lahm A, Di Marcotullio L, Steinkühler C, Pazzaglia S. MK-4101, a Potent Inhibitor of the Hedgehog Pathway, Is Highly Active against Medulloblastoma and Basal Cell Carcinoma. Mol Cancer Ther 2016; 15:1177-89. [DOI: 10.1158/1535-7163.mct-15-0371] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 02/27/2016] [Indexed: 11/16/2022]
|
258
|
Lim Y, Gondek L, Li L, Wang Q, Ma H, Ma H, Chang E, Huso DL, Foerster S, Marchionni L, McGovern K, Watkins DN, Peacock CD, Levis M, Smith BD, Merchant AA, Small D, Matsui W. Integration of Hedgehog and mutant FLT3 signaling in myeloid leukemia. Sci Transl Med 2016; 7:291ra96. [PMID: 26062848 DOI: 10.1126/scitranslmed.aaa5731] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations resulting in constitutive kinase activity are common in acute myeloid leukemia (AML) and carry a poor prognosis. Several agents targeting FLT3 have been developed, but their limited clinical activity suggests that the inhibition of other factors contributing to the malignant phenotype is required. We examined gene expression data sets as well as primary specimens and found that the expression of GLI2, a major effector of the Hedgehog (Hh) signaling pathway, was increased in FLT3-ITD compared to wild-type FLT3 AML. To examine the functional role of the Hh pathway, we studied mice in which Flt3-ITD expression results in an indolent myeloproliferative state and found that constitutive Hh signaling accelerated the development of AML by enhancing signal transducer and activator of transcription 5 (STAT5) signaling and the proliferation of bone marrow myeloid progenitors. Furthermore, combined FLT3 and Hh pathway inhibition limited leukemic growth in vitro and in vivo, and this approach may serve as a therapeutic strategy for FLT3-ITD AML.
Collapse
Affiliation(s)
- Yiting Lim
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lukasz Gondek
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Li Li
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Qiuju Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hayley Ma
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Emily Chang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David L Huso
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah Foerster
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Luigi Marchionni
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - David Neil Watkins
- Cancer Developmental Biology, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Craig D Peacock
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark Levis
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Bruce Douglas Smith
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Akil A Merchant
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Donald Small
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - William Matsui
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
259
|
Christ A, Herzog K, Willnow TE. LRP2, an auxiliary receptor that controls sonic hedgehog signaling in development and disease. Dev Dyn 2016; 245:569-79. [PMID: 26872844 DOI: 10.1002/dvdy.24394] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/31/2022] Open
Abstract
To fulfill their multiple roles in organ development and adult tissue homeostasis, hedgehog (HH) morphogens act through their receptor Patched (PTCH) on target cells. However, HH actions also require HH binding proteins, auxiliary cell surface receptors that agonize or antagonize morphogen signaling in a context-dependent manner. Here, we discuss recent findings on the LDL receptor-related protein 2 (LRP2), an exemplary HH binding protein that modulates sonic hedgehog activities in stem and progenitor cell niches in embryonic and adult tissues. LRP2 functions are crucial for developmental processes in a number of tissues, including the brain, the eye, and the heart, and defects in this receptor pathway are the cause of devastating congenital diseases in humans. Developmental Dynamics 245:569-579, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Katja Herzog
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| |
Collapse
|
260
|
Jung B, Messias AC, Schorpp K, Geerlof A, Schneider G, Saur D, Hadian K, Sattler M, Wanker EE, Hasenöder S, Lickert H. Novel small molecules targeting ciliary transport of Smoothened and oncogenic Hedgehog pathway activation. Sci Rep 2016; 6:22540. [PMID: 26931153 PMCID: PMC4773810 DOI: 10.1038/srep22540] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/15/2016] [Indexed: 01/04/2023] Open
Abstract
Trafficking of the G protein-coupled receptor (GPCR) Smoothened (Smo) to the primary cilium (PC) is a potential target to inhibit oncogenic Hh pathway activation in a large number of tumors. One drawback is the appearance of Smo mutations that resist drug treatment, which is a common reason for cancer treatment failure. Here, we undertook a high content screen with compounds in preclinical or clinical development and identified ten small molecules that prevent constitutive active mutant SmoM2 transport into PC for subsequent Hh pathway activation. Eight of the ten small molecules act through direct interference with the G protein-coupled receptor associated sorting protein 2 (Gprasp2)-SmoM2 ciliary targeting complex, whereas one antagonist of ionotropic receptors prevents intracellular trafficking of Smo to the PC. Together, these findings identify several compounds with the potential to treat drug-resistant SmoM2-driven cancer forms, but also reveal off-target effects of established drugs in the clinics.
Collapse
Affiliation(s)
- Bomi Jung
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Ana C Messias
- Institute of Structural Biology, Helmholtz Zentrum München, Germany.,Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemistry, Technische Universität München, 85747 Garching, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Helmholtz Zentrum München, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Helmholtz Zentrum München, Germany
| | - Günter Schneider
- Department of Internal Medicine II, Klinikum rechts der Isar, München, Germany.,Technische Universität München, München, Germany
| | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar, München, Germany.,Technische Universität München, München, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Helmholtz Zentrum München, Germany
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Germany.,Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemistry, Technische Universität München, 85747 Garching, Germany
| | - Erich E Wanker
- Neuroproteomics, Max Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany
| | - Stefan Hasenöder
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany.,Technische Universität München, München, Germany.,German Center for Diabetes Research (DZD), Germany
| |
Collapse
|
261
|
Crisan M, Solaimani Kartalaei P, Neagu A, Karkanpouna S, Yamada-Inagawa T, Purini C, Vink CS, van der Linden R, van Ijcken W, Chuva de Sousa Lopes SM, Monteiro R, Mummery C, Dzierzak E. BMP and Hedgehog Regulate Distinct AGM Hematopoietic Stem Cells Ex Vivo. Stem Cell Reports 2016; 6:383-95. [PMID: 26923823 PMCID: PMC4788785 DOI: 10.1016/j.stemcr.2016.01.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 11/19/2022] Open
Abstract
Hematopoietic stem cells (HSC), the self-renewing cells of the adult blood differentiation hierarchy, are generated during embryonic stages. The first HSCs are produced in the aorta-gonad-mesonephros (AGM) region of the embryo through endothelial to a hematopoietic transition. BMP4 and Hedgehog affect their production and expansion, but it is unknown whether they act to affect the same HSCs. In this study using the BRE GFP reporter mouse strain that identifies BMP/Smad-activated cells, we find that the AGM harbors two types of adult-repopulating HSCs upon explant culture: One type is BMP-activated and the other is a non-BMP-activated HSC type that is indirectly controlled by Hedgehog signaling through the VEGF pathway. Transcriptomic analyses demonstrate that the two HSC types express distinct but overlapping genetic programs. These results revealing the bifurcation in HSC types at early embryonic stages in the AGM explant model suggest that their development is dependent upon the signaling molecules in the microenvironment. AGM explants contain two HSC types, BMP-activated and non-BMP-activated Non-BMP-activated HSCs are dependent on Hedgehog/VEGF Changes in the microenvironment ex vivo contribute to novel HSC composition
Collapse
Affiliation(s)
- Mihaela Crisan
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; BHF Centre for Cardiovascular Science, Scottish Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Parham Solaimani Kartalaei
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Alex Neagu
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Sofia Karkanpouna
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Tomoko Yamada-Inagawa
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Caterina Purini
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Chris S Vink
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Reinier van der Linden
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Wilfred van Ijcken
- Center for Biomics, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | | | - Rui Monteiro
- Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Elaine Dzierzak
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
262
|
Tang S, Thompson S, Smee R. Metastatic basal cell carcinoma: case series and review of the literature. Australas J Dermatol 2016; 58:e40-e43. [PMID: 26916335 DOI: 10.1111/ajd.12459] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/19/2016] [Indexed: 11/29/2022]
Abstract
Metastatic basal cell carcinoma is exceedingly uncommon, with a poorly defined natural history, and its incidence, risk factors, patterns of spread, prognosis and potential treatment options are not well understood. This retrospective single institution case series aims to shed light on these issues. Aggressive local management in patients with locoregional disease may result in long-term survivors.
Collapse
Affiliation(s)
- Simon Tang
- Department of Radiation Oncology, Illawarra Cancer Care Centre, Wollongong New South Wales, Australia
| | - Stephen Thompson
- Department of Radiation Oncology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Robert Smee
- Department of Radiation Oncology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
263
|
Small J, Barton V, Peterson B, Alberg AJ. Keratinocyte Carcinoma as a Marker of a High Cancer-Risk Phenotype. Adv Cancer Res 2016; 130:257-91. [PMID: 27037755 DOI: 10.1016/bs.acr.2016.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Keratinocyte carcinoma (KC) (also referred to as nonmelanoma skin cancer) is by far the most common form of human cancer. A personal history of KC is well established to be associated with increased risk of recurrent KC and malignant melanoma, a less common yet more fatal form of skin cancer. More surprising is that a substantial body of epidemiologic evidence now indicates that a personal history of KC is significantly associated with an overall elevated risk of noncutaneous malignancies. This association is not limited to one or a few types of cancer but applies across many different types of malignancy. This association has been consistently observed in prospective studies across genders for both major histologic types of KC, basal cell carcinoma and squamous cell carcinoma. The risk of other cancers has been even stronger in those with younger compared with older age of onset of KC. A robust body of evidence lends support to the notion that KC may be a marker of a high cancer-risk phenotype. The underlying mechanisms for this association remain to be elucidated, but the cross-cutting nature of this association across numerous malignancies suggests that research to uncover these mechanisms is a promising line of inquiry that could potentially yield valuable insight into human carcinogenesis.
Collapse
Affiliation(s)
- J Small
- Medical University of South Carolina, Charleston, SC, United States
| | - V Barton
- Medical University of South Carolina, Charleston, SC, United States
| | - B Peterson
- Medical University of South Carolina, Charleston, SC, United States
| | - A J Alberg
- Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
264
|
Papadopoulos V, Tsapakidis K, Riobo Del Galdo NA, Papandreou CN, Del Galdo F, Anthoney A, Sakellaridis N, Dimas K, Kamposioras K. The Prognostic Significance of the Hedgehog Signaling Pathway in Colorectal Cancer. Clin Colorectal Cancer 2016; 15:116-27. [PMID: 27032873 DOI: 10.1016/j.clcc.2016.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/15/2016] [Accepted: 02/03/2016] [Indexed: 12/11/2022]
Abstract
Despite significant advances in the management of colorectal cancer (CRC) the identification of new prognostic biomarkers continues to be a challenge. Since its initial discovery, the role of the Hedgehog (Hh) signaling pathway in carcinogenesis has been extensively studied. We herein review and comment on the prognostic significance of the Hh signaling pathway in CRC. The differential expression of Hh pathway components between malignant and nonmalignant conditions as well as correlation of Hh activation markers with various clinicopathological parameters and the effect on disease-free survival, overall survival, and disease recurrence in patients with CRC is summarized and discussed. According to the studies reviewed herein the activation of the Hh pathway seems to be correlated with adverse clinicopathological features and worse survival. However, to date study results show significant variability with regard to the effect on outcomes. Such results need to be interpreted carefully and emphasize the need for further well designed studies to characterize the actual influence of the Hh pathway in CRC prognosis.
Collapse
Affiliation(s)
| | | | - Natalia A Riobo Del Galdo
- Department of Biochemistry and Molecular Biology and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Francesco Del Galdo
- Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine, LMBRU, University of Leeds, Leeds, United Kingdom
| | - Alan Anthoney
- Department of Medical Oncology, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Nikos Sakellaridis
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| | - Konstantinos Kamposioras
- Department of Medical Oncology, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; Department of Medical Oncology, The Mid Yorkshire Hospitals NHS Trust, Wakefield, United Kingdom.
| |
Collapse
|
265
|
Meani RE, Lim SW, Chang ALS, Kelly JW. Emergence of chemoresistance in a metastatic basal cell carcinoma patient after complete response to hedgehog pathway inhibitor vismodegib (GDC-0449). Australas J Dermatol 2016; 55:218-21. [PMID: 25117162 DOI: 10.1111/ajd.12196] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/08/2014] [Indexed: 11/28/2022]
Abstract
Vismodegib (GDC-0449, Genentech, USA), a small molecule inhibitor of the Hedgehog signalling pathway, has potent anti-tumour activity in advanced basal cell carcinoma (BCC). We report a case of a 67-year-old Australian man with metastatic BCC including pulmonary disease with malignant effusion who showed a dramatic complete response to vismodegib but subsequently experienced a recurrence of pulmonary disease, indicative of chemoresistance to vismodegib. This case is the first to illustrate chemoresistance in a patient with metastatic BCC, and demonstrates the need for closely monitoring metastatic BCC patients even after an apparently complete response.
Collapse
Affiliation(s)
- Rowena E Meani
- Victorian Melanoma Service, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
266
|
Calzavara Pinton P, Licitra L, Peris K, Santoro A, Ascierto PA. Vismodegib in the treatment of basal cell carcinoma: indications for clinical practice. Future Oncol 2016; 11:1429-35. [PMID: 25952787 DOI: 10.2217/fon.15.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Basal cell carcinoma (BCC) is a frequent skin cancer which can cause substantial morbidity due to its location on the face, its frequency of relapse and its capacity to invade local tissues. The primary treatment of BCC usually involves surgery or radiotherapy. In patients who have exhausted surgical and radiotherapy options or with metastatic BCC, guidelines recommend the use of the Hedgehog pathway inhibitor vismodegib. This molecule is indicated for the treatment of adults with metastatic BCC, or with locally advanced BCC which has recurred following surgery or who are not eligible to surgery or radiation. This paper aims to provide suggestions on the optimal management of BCC patients treated with vismodegib in clinical practice, according to the large experience gained by a group of Italian dermatologists and oncologists. In particular, the focus of this paper will be on the monitoring of patients and the management of adverse events.
Collapse
|
267
|
Wang A, Stefanick ML, Kapphahn K, Hedlin H, Desai M, Manson JAE, Strickler H, Martin L, Wactawski-Wende J, Simon M, Tang JY. Relation of statin use with non-melanoma skin cancer: prospective results from the Women's Health Initiative. Br J Cancer 2016; 114:314-20. [PMID: 26742009 PMCID: PMC4742576 DOI: 10.1038/bjc.2015.376] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 09/24/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The relationship between statin use and non-melanoma skin cancer (NMSC) is unclear with conflicting findings in literature. Data from the Women's Health Initiative (WHI) Observational Study and WHI Clinical Trial were used to investigate the prospective relationship between statin use and NMSC in non-Hispanic white (NHW) postmenopausal women. METHODS The WHI study enrolled women aged 50-79 years at 40 US centres. Among 133,541 NHW participants, 118,357 with no cancer history at baseline and complete medication/covariate data comprised the analytic cohort. The association of statin use (baseline, overall as a time-varying variable, duration, type, potency, lipophilicity) and NMSC incidence was determined using random-effects logistic regression models. RESULTS Over a mean of 10.5 years of follow-up, we identified 11,555 NMSC cases. Compared with participants with no statin use, use of any statin at baseline was associated with significantly increased NMSC incidence (adjusted odds ratio (ORadj) 1.21; 95% confidence interval (CI): 1.07-1.35)). In particular, lovastatin (OR 1.52; 95% CI: 1.08-2.16), simvastatin (OR 1.38; 95% CI: 1.12-1.69), and lipophilic statins (OR 1.39; 95% CI: 1.18-1.64) were associated with higher NMSC risk. Low and high, but not medium, potency statins were associated with higher NMSC risk. No significant effect modification of the statin-NMSC relationship was found for age, BMI, smoking, solar irradiation, vitamin D use, and skin cancer history. CONCLUSIONS Use of statins, particularly lipophilic statins, was associated with increased NMSC risk in postmenopausal white women in the WHI cohort. The lack of duration-effect relationship points to possible residual confounding. Additional prospective research should further investigate this relationship.
Collapse
Affiliation(s)
- Ange Wang
- Department of Dermatology, Stanford University School of Medicine, 450 Broadway Street, Pavilion B, 4th Floor MC 5338, Redwood City, CA 94063, USA
| | - Marcia L Stefanick
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristopher Kapphahn
- Quantitative Sciences Unit, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Haley Hedlin
- Quantitative Sciences Unit, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Manisha Desai
- Quantitative Sciences Unit, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jo Ann E Manson
- Department of Epidemiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lisa Martin
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Jean Wactawski-Wende
- Department of Social and Preventive Medicine, University at Buffalo, Buffalo, NY, USA
| | - Michael Simon
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Jean Y Tang
- Department of Dermatology, Stanford University School of Medicine, 450 Broadway Street, Pavilion B, 4th Floor MC 5338, Redwood City, CA 94063, USA
| |
Collapse
|
268
|
Vismodegib, itraconazole and sonidegib as hedgehog pathway inhibitors and their relative competencies in the treatment of basal cell carcinomas. Crit Rev Oncol Hematol 2016; 98:235-41. [DOI: 10.1016/j.critrevonc.2015.11.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 10/06/2015] [Accepted: 11/11/2015] [Indexed: 12/17/2022] Open
|
269
|
|
270
|
Arnhold V, Boos J, Lanvers-Kaminsky C. Targeting hedgehog signaling pathway in pediatric tumors: in vitro evaluation of SMO and GLI inhibitors. Cancer Chemother Pharmacol 2016; 77:495-505. [DOI: 10.1007/s00280-016-2962-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/05/2016] [Indexed: 12/23/2022]
|
271
|
Mimeault M, Rachagani S, Muniyan S, Seshacharyulu P, Johansson SL, Datta K, Lin MF, Batra SK. Inhibition of hedgehog signaling improves the anti-carcinogenic effects of docetaxel in prostate cancer. Oncotarget 2016; 6:3887-903. [PMID: 25682877 PMCID: PMC4414161 DOI: 10.18632/oncotarget.2932] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022] Open
Abstract
The establishment of docetaxel-based chemotherapeutic treatments has improved the survival of castration-resistant prostate cancer (CRPC) patients. However, most patients develop resistance supporting the development of therapy. The current study was undertaken to establish the therapeutic benefit to target hedgehog signaling cascade using GDC-0449 to improve the efficacy of chemotherapeutic drug, docetaxel. Here, we show that the combination of GDC-0449 plus docetaxel inhibited the proliferation of WPE1-NB26 cells and PC3 cells via a blockade of G1 and G2M phases. The combined treatment significantly inhibited PC cell migration in vitro. Moreover, the apoptotic effect induced by GDC-0449 plus docetaxel on PC3 cells was mediated, at least partly, via the mitochondrial membrane depolarization, H2O2 production and caspase cascade activation. Interestingly, GDC-0449 was effective at inhibiting the prostasphere formation, inducing the prostasphere disintegration and apoptotic death of side population (SP) from PC3 cells and reversing the resistance of SP cells to docetaxel. In addition, GDC-0449 plus docetaxel also have shown a greater anti-tumoral growth inhibitory effect on PC3 cell xenografts. These findings support the use of the hedgehog inhibitor GDC-0449, which is currently in clinical trials, for improving the anticarcinogenic efficacy of docetaxel-based chemotherapeutic treatments against locally advanced, AI and metastatic PC.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Sonny L Johansson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
272
|
Srinath S, Iyengar AR, Mysorekar V. Sonic hedgehog in oral squamous cell carcinoma: An immunohistochemical study. J Oral Maxillofac Pathol 2016; 20:377-383. [PMID: 27721600 PMCID: PMC5051283 DOI: 10.4103/0973-029x.190906] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Recent studies have revealed the involvement of hedgehog (Hh) signaling component in proliferation and invasive behavior of many carcinomas. AIM This study aims to identify the expression of sonic Hh (SHH) protein of SHH pathway in oral epithelial dysplasia and oral squamous cell carcinoma (OSCC) using SHH (H-160) (Santa Cruz, sc-9042) which could have therapeutic implication in future. MATERIALS AND METHODS A total of 250 cases comprising 50 normal oral mucosa, 50 cases of oral epithelial dysplasia, 50 well, 50 moderate and 50 poorly differentiated OSCCs were included in the study. Immunohistochemical evaluation of SHH protein expression was conducted using monoclonal antibody. Interpretation of the expression was done by immunoreactive score of Remmele and Stegner (IRS) scoring method. STATISTICAL ANALYSIS Chi-Square test was used to analyze the results. RESULTS The study showed that SHH signaling molecules are highly expressed in OSCC, and their expression was mainly in the cytoplasm of epithelial cells. CONCLUSION The SHH signaling component is associated with the pathological parameter in OSCC and oral epithelial dysplasia.
Collapse
Affiliation(s)
- Sahana Srinath
- Department of Oral Pathology, GDCRI, Bengaluru, Karnataka, India
| | - Asha R Iyengar
- Department of Oral Medicine and Radiology, DAPMRV, Bengaluru, Karnataka, India
| | | |
Collapse
|
273
|
Inhibition of Hedgehog-Signaling Driven Genes in Prostate Cancer Cells by Sutherlandia frutescens Extract. PLoS One 2015; 10:e0145507. [PMID: 26710108 PMCID: PMC4694108 DOI: 10.1371/journal.pone.0145507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/04/2015] [Indexed: 11/19/2022] Open
Abstract
Sutherlandia frutescens (L) R. Br. (Sutherlandia) is a South African botanical that is traditionally used to treat a variety of health conditions, infections and diseases, including cancer. We hypothesized Sutherlandia might act through Gli/ Hedgehog (Hh)-signaling in prostate cancer cells and used RNA-Seq transcription profiling to profile gene expression in TRAMPC2 murine prostate cancer cells with or without Sutherlandia extracts. We found 50% of Hh-responsive genes can be repressed by Sutherlandia ethanol extract, including the canonical Hh-responsive genes Gli1 and Ptch1 as well as newly distinguished Hh-responsive genes Hsd11b1 and Penk.
Collapse
|
274
|
Catenacci DVT, Junttila MR, Karrison T, Bahary N, Horiba MN, Nattam SR, Marsh R, Wallace J, Kozloff M, Rajdev L, Cohen D, Wade J, Sleckman B, Lenz HJ, Stiff P, Kumar P, Xu P, Henderson L, Takebe N, Salgia R, Wang X, Stadler WM, de Sauvage FJ, Kindler HL. Randomized Phase Ib/II Study of Gemcitabine Plus Placebo or Vismodegib, a Hedgehog Pathway Inhibitor, in Patients With Metastatic Pancreatic Cancer. J Clin Oncol 2015; 33:4284-92. [PMID: 26527777 PMCID: PMC4678179 DOI: 10.1200/jco.2015.62.8719] [Citation(s) in RCA: 422] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Sonic hedgehog (SHH), an activating ligand of smoothened (SMO), is overexpressed in > 70% of pancreatic cancers (PCs). We investigated the impact of vismodegib, an SHH antagonist, plus gemcitabine (GV) or gemcitabine plus placebo (GP) in a multicenter phase Ib/randomized phase II trial and preclinical PC models. PATIENTS AND METHODS Patients with PC not amenable to curative therapy who had received no prior therapy for metastatic disease and had Karnofsky performance score ≥ 80 were enrolled. Patients were randomly assigned in a one-to-one ratio to GV or GP. The primary end point was progression-free-survival (PFS). Exploratory correlative studies included serial SHH serum levels and contrast perfusion computed tomography imaging. To further investigate putative biologic mechanisms of SMO inhibition, two autochthonous pancreatic cancer models (Kras(G12D); p16/p19(fl/fl); Pdx1-Cre and Kras(G12D); p53(R270H/wt); Pdx1-Cre) were studied. RESULTS No safety issues were identified in the phase Ib portion (n = 7), and the phase II study enrolled 106 evaluable patients (n = 53 in each arm). Median PFS was 4.0 and 2.5 months for GV and GP arms, respectively (95% CI, 2.5 to 5.3 and 1.9 to 3.8, respectively; adjusted hazard ratio, 0.81; 95% CI, 0.54 to 1.21; P = .30). Median overall survival (OS) was 6.9 and 6.1 months for GV and GP arms, respectively (95% CI, 5.8 to 8.0 and 5.0 to 8.0, respectively; adjusted hazard ratio, 1.04; 95% CI, 0.69 to 1.58; P = .84). Response rates were not significantly different. There were no significant associations between correlative markers and overall response rate, PFS, or OS. Preclinical trials revealed no significant differences with vismodegib in drug delivery, tumor growth rate, or OS in either model. CONCLUSION The addition of vismodegib to gemcitabine in an unselected cohort did not improve overall response rate, PFS, or OS in patients with metastatic PC. Our preclinical and clinical results revealed no statistically significant differences with respect to drug delivery or treatment efficacy using vismodegib.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Melissa R Junttila
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Theodore Karrison
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Nathan Bahary
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Margit N Horiba
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sreenivasa R Nattam
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Robert Marsh
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James Wallace
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mark Kozloff
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lakshmi Rajdev
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Deirdre Cohen
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James Wade
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Bethany Sleckman
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Heinz-Josef Lenz
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Patrick Stiff
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Pankaj Kumar
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Peng Xu
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Les Henderson
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Naoko Takebe
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ravi Salgia
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Xi Wang
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Walter M Stadler
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Frederic J de Sauvage
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hedy L Kindler
- Daniel V.T. Catenacci, Theodore Karrison, James Wallace, Mark Kozloff, Peng Xu, Les Henderson, Ravi Salgia, Walter M. Stadler, Hedy L. Kindler, University of Chicago Medical Center; Patrick Stiff, Loyola University Medical Center, Chicago; Robert Marsh, Northshore University Health System, Evanston; James Wallace, Mark Kozloff, Ingalls Hospital, Harvey; James Wade, Decatur Memorial Hospital, Decatur; Pankaj Kumar, Oncology/Hematology Associates, Peoria, IL; Melissa R. Junttila, Xi Wang, and Frederic J. de Sauvage, Genentech, South San Francisco; Heinz-Josef Lenz, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Nathan Bahary, University of Pittsburgh Cancer Institute, Pittsburgh, PA; Margit N. Horiba, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Sreenivasa R. Nattam, Ft Wayne Medical Oncology/Hematology, Ft Wayne, IN; Lakshmi Rajdev, Montefiore Medical Center, Bronx; Deirdre Cohen, New York University Cancer Center, New York, NY; Bethany Sleckman, St John's Mercy Medical Center, St Louis, MO; and Naoko Takebe, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
275
|
Leon G, MacDonagh L, Finn SP, Cuffe S, Barr MP. Cancer stem cells in drug resistant lung cancer: Targeting cell surface markers and signaling pathways. Pharmacol Ther 2015; 158:71-90. [PMID: 26706243 DOI: 10.1016/j.pharmthera.2015.12.001] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Despite advances in anti-cancer therapies such as chemotherapy, radiotherapy and targeted therapies, five-year survival rates remain poor (<15%). Inherent and acquired resistance has been identified as a key factor in reducing the efficacy of current cytotoxic therapies in the management of non-small cell lung cancer (NSCLC). There is growing evidence suggesting that cancer stem cells (CSCs) play a critical role in tumor progression, metastasis and drug resistance. Similar to normal tissue stem cells, CSCs exhibit significant phenotypic and functional heterogeneity. While CSCs have been reported in a wide spectrum of human tumors, the biology of CSCs in NSCLC remain elusive. Current anti-cancer therapies fail to eradicate CSC clones and instead, favor the expansion of the CSC pool and select for resistant CSC clones thereby resulting in treatment resistance and subsequent relapse in these patients. The identification of CSC-specific marker subsets and the targeted therapeutic destruction of CSCs remains a significant challenge. Strategies aimed at efficient targeting of CSCs are becoming increasingly important for monitoring the progress of cancer therapy and for evaluating new therapeutic approaches. This review focuses on the current knowledge of cancer stem cell markers in treatment-resistant lung cancer cells and the signaling cascades activated by these cells to maintain their stem-like properties. Recent progress in CSC-targeted drug development and the current status of novel agents in clinical trials are also reviewed.
Collapse
Affiliation(s)
- Gemma Leon
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Dublin 8, Ireland
| | - Lauren MacDonagh
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Dublin 8, Ireland
| | - Stephen P Finn
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Dublin 8, Ireland; Department of Histopathology, St James's Hospital, Dublin 8, Ireland
| | - Sinead Cuffe
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Dublin 8, Ireland
| | - Martin P Barr
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Dublin 8, Ireland.
| |
Collapse
|
276
|
Abstract
Basal cell carcinomas are the most common primary cutaneous malignant neoplasms. The diagnosis of basal cell carcinoma represents a common and routine task for pathologists and dermatopathologists. The aim of this review is the clinical and histopathological presentation of the most common subtypes of basal cell carcinoma. Furthermore, the rare variants of basal cell carcinoma and their differential diagnoses are also discussed.
Collapse
Affiliation(s)
- J Liersch
- Dermatopathologie Duisburg, An der Abtei 7-11, 47166, Duisburg, Deutschland
| | | |
Collapse
|
277
|
John AM, Schwartz RA. Basal cell naevus syndrome: an update on genetics and treatment. Br J Dermatol 2015; 174:68-76. [PMID: 26409035 DOI: 10.1111/bjd.14206] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2015] [Indexed: 12/14/2022]
Abstract
Basal cell naevus syndrome is an autosomal dominant disorder that stems from mutations in multiple genes, most commonly patched 1 (PTCH1). The classic triad of symptoms consists of basal cell carcinomas, jaw keratocysts and cerebral calcifications, although there are many other systemic manifestations. Because of the broad range of symptoms and development of several types of tumours, early diagnosis and close monitoring are essential to preserve quality of life. Targeting treatment is often difficult because of tumour prevalence. Newer inhibitors of the hedgehog signalling pathway and proteins involved in proliferative growth have shown therapeutic promise. In addition, preventive medications are being devised. We propose a method for determining appropriate treatment for cutaneous tumours.
Collapse
Affiliation(s)
- A M John
- Department of Dermatology, Rutgers New Jersey Medical School, Newark, NJ, 07103, U.S.A
| | - R A Schwartz
- Department of Dermatology, Rutgers New Jersey Medical School, Newark, NJ, 07103, U.S.A.,Rutgers University School of Public Affairs and Administration, Newark, NJ, U.S.A
| |
Collapse
|
278
|
Zhang N, Liu S, Wang N, Deng S, Song L, Wu Q, Liu L, Su W, Wei Y, Xie Y, Gong C. Biodegradable polymeric micelles encapsulated JK184 suppress tumor growth through inhibiting Hedgehog signaling pathway. NANOSCALE 2015; 7:2609-24. [PMID: 25581613 DOI: 10.1039/c4nr06300g] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
JK184 can specially inhibit Gli in the Hedgehog (Hh) pathway, which showed great promise for cancer therapeutics. For developing aqueous formulation and improving anti-tumor activity of JK184, we prepared JK184 encapsulated MPEG-PCL micelles by the solid dispersion method without using surfactants or toxic organic solvents. The cytotoxicity and cellular uptake of JK184 micelles were both increased compared with the free drug. JK184 micelles induced more apoptosis and blocked proliferation of Panc-1 and BxPC-3 tumor cells. In addition, JK184 micelles exerted a sustained in vitro release behavior and had a stronger inhibitory effect on proliferation, migration and invasion of HUVECs than free JK184. Furthermore, JK184 micelles had stronger tumor growth inhibiting effects in subcutaneous Panc-1 and BxPC-3 tumor models. Histological analysis showed that JK184 micelles improved anti-tumor activity by inducing more apoptosis, decreasing microvessel density and reducing expression of CD31, Ki67, and VEGF in tumor tissues. JK184 micelles showed a stronger inhibition of Gli expression in Hh signaling, which played an important role in pancreatic carcinoma. Furthermore, circulation time of JK184 in blood was prolonged after entrapment in polymeric micelles. Our results suggested that JK184 micelles are a promising drug candidate for treating pancreatic tumors with a highly inhibitory effect on Hh activity.
Collapse
Affiliation(s)
- Nannan Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Rivers JK, Mistry BD, Hung T, Vostretsova K, Mistry N. A 13-Year Retrospective Study of Basal Cell Carcinoma in a Canadian Dermatology Practice: A Comparison Between Anatomical Location and Histopathologic Subtypes. J Cutan Med Surg 2015; 20:233-40. [PMID: 26631769 DOI: 10.1177/1203475415620396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND It is unknown whether the histologic subtypes of basal cell carcinoma (BCC) arise from a common progenitor cell or whether other factors play a role in their development. OBJECTIVE To investigate the relationship between the different BCC histopathologic subtypes and anatomical distribution of BCCs in a Canadian dermatology practice. METHODS The charts of all patients diagnosed with BCC between 1993 and 2005 from a single private dermatology practice in Vancouver, Canada, were reviewed. Descriptive data analysis was undertaken to look at the distribution of histologic subtypes based on age, gender, and anatomical location. RESULTS Nodular BCCs accounted for 58% of all tumors. Sixty-six percent of these were situated on the head/neck (odds ratio [OR] = 3.0, 95% confidence interval [CI] = 2.1-4.3, P < .0001). Infiltrative (OR = 2.4, 95% CI = 1.5-4.1, P = .0003) and superficial BCCs were more common in women (OR = 3.7, 95% CI = 2.5-5.7, P < .0001), affected the trunk (OR = 3.2, 95% CI = 2.1-4.9, P < .0001), and appeared in younger individuals (OR = 1.8, 95% CI = 1.2-2.7, P = .004). CONCLUSION Our results show a preference of distinct BCC subtypes for certain anatomical locations.
Collapse
Affiliation(s)
- Jason K Rivers
- Department of Dermatology and Skin Science, University of British Columbia and Pacific Dermaesthetics, Vancouver, BC, Canada
| | - Bhavik D Mistry
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tawny Hung
- DynalifeDx, University of Alberta, Departments of Pathology and Dermatology, University of Alberta, Edmonton, AB, Canada
| | | | - Nisha Mistry
- Department of Medicine (Dermatology), University of Toronto, Toronto, ON, Canada
| |
Collapse
|
280
|
Borah A, Raveendran S, Rochani A, Maekawa T, Kumar DS. Targeting self-renewal pathways in cancer stem cells: clinical implications for cancer therapy. Oncogenesis 2015; 4:e177. [PMID: 26619402 PMCID: PMC4670961 DOI: 10.1038/oncsis.2015.35] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 12/21/2022] Open
Abstract
Extensive cancer research in the past few decades has identified the existence of a rare subpopulation of stem cells in the grove of cancer cells. These cells are known as the cancer stem cells marked by the presence of surface biomarkers, multi-drug resistance pumps and deregulated self-renewal pathways (SRPs). They have a crucial role in provoking cancer cells leading to tumorigenesis and its progressive metastasis. Cancer stem cells (CSCs) are much alike to normal stem cells in their self-renewal mechanisms. However, deregulations in the SRPs are seen in CSCs, making them resistant to conventional chemotherapeutic agents resulting in the tumor recurrence. Current treatment strategies in cancer fail to detect and differentiate the CSCs from their non-tumorigenic progenies owing to absence of specific biomarkers. Now, it has become imperative to understand complex functional biology of CSCs, especially the signaling pathways to design improved treatment strategies to target them. It is hopeful that the SRPs in CSCs offer a promising target to alter their survival strategies and impede their tumorigenic potential. However, there are many perils associated with the direct targeting method by conventional therapeutic agents such as off targets, poor bioavailability and poor cellular distribution. Recent evidences have shown an increased use of small molecule antagonists directly to target these SRPs may lead to severe side-effects. An alternative to solve these issues could be an appropriate nanoformulation. Nanoformulations of these molecules could provide an added advantage for the selective targeting of the pathways especially Hedgehog, Wnt, Notch and B-cell-specific moloney murine leukemia virus integration site 1 in the CSCs while sparing the normal stem cells. Hence, to achieve this goal a complete understanding of the molecular pathways corroborate with the use of holistic nanosystem (nanomaterial inhibition molecule) could possibly be an encouraging direction for future cancer therapy.
Collapse
Affiliation(s)
- A Borah
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - S Raveendran
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - A Rochani
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - T Maekawa
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - D S Kumar
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
281
|
Rudnick EW, Thareja S, Cherpelis B. Oral therapy for nonmelanoma skin cancer in patients with advanced disease and large tumor burden: a review of the literature with focus on a new generation of targeted therapies. Int J Dermatol 2015; 55:249-58; quiz 256, 258. [PMID: 26566923 DOI: 10.1111/ijd.12961] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/26/2014] [Accepted: 12/31/2014] [Indexed: 12/20/2022]
Abstract
Nonmelanoma skin cancer (NMSC) is the most common cancer in patients and includes basal cell carcinoma (BCC) and squamous cell carcinoma (SCC). Treatments useful for SCC and BCC include surgical, topical, and in advanced cases systemic chemo-radiation. This review of the literature aims to describe previous and current treatment options for oral therapy in locally advanced and metastatic NMSC otherwise unamenable to standard treatment. Oral Smoothened (Smo) inhibitors Vismodegib, Sonidegib, and Taladegib have shown to be effective in several trials. Oral tyrosine kinase inhibitors Erlotinib and Gefitinib, which target epidermal growth factor receptor (EGFR), have early supporting data and are currently undergoing large multicenter trials. Other less studied oral therapies which have shown at least partial efficacy include 5-Fluorouracil, capecitabine, and picropodophyllin. In vitro studies have elucidated new targets for dual combination oral therapy targeting both EGFR and insulin-like growth factor 1 receptor (IGF-1R). It is important to stratify treatment options based on patient risk of advanced disease, failure of conservative treatment, and ill-tolerated intravenous chemotherapy adverse events. Oral therapy in NMSC is useful in high risk patients with recurrent and aggressive disease who may not tolerate other systemic therapies.
Collapse
Affiliation(s)
- Eric W Rudnick
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sumeet Thareja
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Basil Cherpelis
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
282
|
Lu T, Wang B, Gao Y, Dresser M, Graham RA, Jin JY. Semi-Mechanism-Based Population Pharmacokinetic Modeling of the Hedgehog Pathway Inhibitor Vismodegib. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:680-9. [PMID: 26783504 PMCID: PMC4716579 DOI: 10.1002/psp4.12039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
Vismodegib, approved for the treatment of advanced basal cell carcinoma, has shown unique pharmacokinetic (PK) nonlinearity and binding to α1-acid glycoprotein (AAG) in humans. A semi-mechanism-based population pharmacokinetic (PopPK) model was developed from a meta-dataset of 225 subjects enrolled in five clinical studies to quantitatively describe the clinical PK of vismodegib and identify sources of interindividual variability. Total and unbound vismodegib were analyzed simultaneously, together with time-varying AAG data. The PK of vismodegib was adequately described by a one-compartment model with first-order absorption, first-order elimination of unbound drug, and saturable binding to AAG with fast-equilibrium. The variability of total vismodegib concentration at steady-state was predominantly explained by the range of AAG level. The impact of AAG on unbound concentration was clinically insignificant. Various approaches were evaluated for model validation. The semi-mechanism-based PopPK model described herein provided insightful information on the nonlinear PK and has been utilized for various clinical applications.
Collapse
Affiliation(s)
- T Lu
- Genentech, Inc. South San Francisco California USA
| | - B Wang
- Genentech, Inc. South San Francisco California USA
| | - Y Gao
- Quantitative Solutions, Inc. Menlo Park California USA
| | - M Dresser
- Genentech, Inc. South San Francisco California USA
| | - R A Graham
- Genentech, Inc. South San Francisco California USA
| | - J Y Jin
- Genentech, Inc. South San Francisco California USA
| |
Collapse
|
283
|
Fecher LA, Sharfman WH. Advanced basal cell carcinoma, the hedgehog pathway, and treatment options - role of smoothened inhibitors. Biologics 2015; 9:129-40. [PMID: 26604681 PMCID: PMC4642804 DOI: 10.2147/btt.s54179] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Cutaneous basal cell carcinoma (BCC) is the most common human cancer and its incidence is rising worldwide. Ultraviolet radiation exposure, including tanning bed use, as well as host factors play a role in its development. The majority of cases are treated and cured with local therapies including surgery. Yet, the health care costs of diagnosis and treatment of BCCs in the US is substantial. In the United States, the cost of nonmelanoma skin cancer care in the Medicare population is estimated to be US$426 million per year. While rare, locally advanced BCCs that can no longer be controlled with surgery and/or radiation, and metastatic BCCs do occur and can be associated with significant morbidity and mortality. Vismodegib (GDC-0449), a smoothened inhibitor targeted at the hedgehog pathway, is the first US Food and Drug Association (FDA)-approved agent in the treatment of locally advanced, unresectable, and metastatic BCCs. This class of agents appears to be changing the survival rates in advanced BCC patients, but appropriate patient selection and monitoring are important. Multidisciplinary assessments are essential for the optimal care and management of these patients. For some patients with locally advanced BCC, treatment with a hedgehog inhibitor may eliminate the need for an excessively disfiguring or morbid surgery.
Collapse
Affiliation(s)
- Leslie A Fecher
- Department of Internal Medicine and Dermatology, Indiana University Health Simon Cancer Center, Indianapolis, IN, USA
- Department of Internal Medicine and Dermatology, University of Michigan, MI, USA
| | - William H Sharfman
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
284
|
Roy Chaudhuri T, Straubinger NL, Pitoniak RF, Hylander BL, Repasky EA, Ma WW, Straubinger RM. Tumor-Priming Smoothened Inhibitor Enhances Deposition and Efficacy of Cytotoxic Nanoparticles in a Pancreatic Cancer Model. Mol Cancer Ther 2015; 15:84-93. [PMID: 26516158 DOI: 10.1158/1535-7163.mct-15-0602] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/01/2015] [Indexed: 12/13/2022]
Abstract
Most pancreatic adenocarcinoma patients present with unresectable disease and benefit little from chemotherapy. Poor tumor perfusion and vascular permeability limit drug deposition. Previous work showed that Smoothened inhibitors of hedgehog signaling (sHHI) promote neovascularization in spontaneous mouse models of pancreatic cancer (PaCA) and enhance tumor permeability to low-molecular weight compounds. Here, we tested the hypothesis that sHHI can enhance tumor deposition and efficacy of drug-containing nanoparticles consisting of 80 to 100 nm sterically-stabilized liposomes (SSL) containing doxorubicin (SSL-DXR). SCID mice bearing low-passage patient-derived PaCA xenografts (PDX) were pretreated p.o. for 10 days with 40 mg/kg/d NVP-LDE225 (erismodegib), followed by i.v. SSL-DXR. Microvessel density, permeability, perfusion, and morphology were compared with untreated controls, as was SSL deposition and therapeutic efficacy. The sHHI alone affected tumor growth minimally, but markedly increased extravasation of nanoparticles into adenocarcinoma cell-enriched regions of the tumor. Immunostaining showed that sHHI treatment decreased pericyte coverage (α-SMA(+)) of CD31(+) vascular endothelium structures, and increased the abundance of endothelium-poor (CD31(-)) basement membrane structures (collagen IV(+)), suggesting increased immature microvessels. SSL-DXR (15 mg/kg) administered after sHHI pretreatment arrested tumor volume progression and decreased tumor perfusion/permeability, suggesting an initial vascular pruning response. Compared with controls, one cycle of 10-day sHHI pretreatment followed by 6 mg/kg SSL-DXR doubled median tumor progression time. Three cycles of treatment with sHHI and SSL-DXR, with a 10-day between-cycle drug holiday, nearly tripled median tumor progression time. Based upon these data, short-term sHHI treatment sequenced with nanoparticulate drug carriers constitutes a potential strategy to enhance efficacy of pancreatic cancer therapy.
Collapse
Affiliation(s)
- Tista Roy Chaudhuri
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York. Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, New York
| | - Ninfa L Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York
| | | | - Bonnie L Hylander
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | | | - Wen Wee Ma
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York. Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, New York. Department of Cancer Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York. New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York.
| |
Collapse
|
285
|
Advances in small-molecule drug discovery for triple-negative breast cancer. Future Med Chem 2015; 7:2019-39. [PMID: 26495746 DOI: 10.4155/fmc.15.129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of poor prognosis, highly invasive and difficult-to-treat breast cancers accounting for approximately 15% of clinical cases. Given the poor outlook and lack of sustained response to conventional therapies, TNBC has been the subject of intense studies on new therapeutic approaches in recent years. The development of targeted cancer therapies, often in combination with established chemotherapy, has been applied to a number of new clinical studies in this setting in recent years. This review will highlight recent therapeutic advances in TNBC, focusing on small-molecule drugs and their associated biological mechanisms of action, and offering the possibility of improved prospects for this patient group in the near future.
Collapse
|
286
|
Tibes R, Al-Kali A, Oliver GR, Delman DH, Hansen N, Bhagavatula K, Mohan J, Rakhshan F, Wood T, Foran JM, Mesa RA, Bogenberger JM. The Hedgehog pathway as targetable vulnerability with 5-azacytidine in myelodysplastic syndrome and acute myeloid leukemia. J Hematol Oncol 2015; 8:114. [PMID: 26483188 PMCID: PMC4615363 DOI: 10.1186/s13045-015-0211-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 09/28/2015] [Indexed: 02/04/2023] Open
Abstract
Background Therapy and outcome for elderly acute myeloid leukemia (AML) patients has not improved for many years. Similarly, there remains a clinical need to improve response rates in advanced myelodysplastic syndrome (MDS) patients treated with hypomethylating agents, and few combination regimens have shown clinical benefit. We conducted a 5-azacytidine (5-Aza) RNA-interference (RNAi) sensitizer screen to identify gene targets within the commonly deleted regions (CDRs) of chromosomes 5 and 7, whose silencing enhances the activity of 5-Aza. Methods and results An RNAi silencing screen of 270 genes from the CDRs of chromosomes 5 and 7 was performed in combination with 5-Aza treatment in four AML cell lines (TF-1, THP-1, MDS-L, and HEL). Several genes within the hedgehog pathway (HhP), specifically SHH, SMO, and GLI3, were identified as 5-Aza sensitizing hits. The smoothened (SMO) inhibitors LDE225 (erismodegib) and GDC0449 (vismodegib) showed moderate single-agent activity in AML cell lines. Further studies with erismodegib in combination with 5-Aza demonstrated synergistic activity with combination index (CI) values of 0.48 to 0.71 in seven AML lines. Clonogenic growth of primary patient samples was inhibited to a greater extent in the combination than with single-agent erismodegib or 5-Aza in 55 % (6 of 11) primary patient samples examined. There was no association of the 5-Aza/erismodegib sensitization potential to clinical-cytogenetic features or common myeloid mutations. Activation of the HhP, as determined by greater expression of HhP-related genes, showed less responsiveness to single-agent SMO inhibition, while synergy between both agents was similar regardless of HhP gene expression. In vitro experiments suggested that concurrent dosing showed stronger synergy than sequential dosing. Conclusions Inhibition of the HhP with SMO inhibitors in combination with the hypomethylating agent 5-Aza demonstrates synergy in vitro and inhibits long-term repopulation capacity ex vivo in AML and MDS. A clinical trial combining 5-Aza with LDE225 (erismodegib) in MDS and AML is ongoing based on these results as well as additional publications suggesting a role for HhP signaling in myeloid disease. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0211-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raoul Tibes
- Division of Hematology and Medical Oncology, Mayo Clinic/Mayo Clinic Cancer Center, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| | - Aref Al-Kali
- Mayo Clinic's Campus in Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Gavin R Oliver
- Mayo Clinic's Campus in Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Devora H Delman
- Division of Hematology and Medical Oncology, Mayo Clinic/Mayo Clinic Cancer Center, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| | - Nanna Hansen
- Division of Hematology and Medical Oncology, Mayo Clinic/Mayo Clinic Cancer Center, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| | - Keerthi Bhagavatula
- Division of Hematology and Medical Oncology, Mayo Clinic/Mayo Clinic Cancer Center, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| | - Jayaram Mohan
- Washington University St. Louis, St. Louis, MO, 63130-4899, USA.
| | - Fariborz Rakhshan
- Mayo Clinic's Campus in Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Thomas Wood
- Mayo Clinic's Campus in Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| | - James M Foran
- Mayo Clinic's Campus in Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Ruben A Mesa
- Division of Hematology and Medical Oncology, Mayo Clinic/Mayo Clinic Cancer Center, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| | - James M Bogenberger
- Division of Hematology and Medical Oncology, Mayo Clinic/Mayo Clinic Cancer Center, 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA.
| |
Collapse
|
287
|
Lin H, Jackson GA, Lu Y, Drenkhahn SK, Brownstein KJ, Starkey NJ, Lamberson WR, Fritsche KL, Mossine VV, Besch-Williford CL, Folk WR, Zhang Y, Lubahn DB. Inhibition of Gli/hedgehog signaling in prostate cancer cells by "cancer bush" Sutherlandia frutescens extract. Cell Biol Int 2015; 40:131-42. [PMID: 26377232 DOI: 10.1002/cbin.10544] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/03/2015] [Indexed: 12/21/2022]
Abstract
Sutherlandia frutescens is a medicinal plant, traditionally used to treat various types of human diseases, including cancer. Previous studies of several botanicals link suppression of prostate cancer growth with inhibition of the Gli/hedgehog (Gli/Hh) signaling pathway. Here we hypothesized the anti-cancer effect of S. frutescens was linked to its inhibition of the Gli/Hh signaling in prostate cancer. We found a dose- and time-dependent growth inhibition in human prostate cancer cells, PC3 and LNCaP, and mouse prostate cancer cell, TRAMP-C2, treated with S. frutescens methanol extract (SLE). We also observed a dose-dependent inhibition of the Gli-reporter activity in Shh Light II and TRAMP-C2QGli cells treated with SLE. In addition, SLE can inhibit Gli/Hh signaling by blocking Gli1 and Ptched1 gene expression in the presence of a Gli/Hh signaling agonist (SAG). A diet supplemented with S. frutescens suppressed the formation of poorly differentiated carcinoma in prostates of TRAMP mice. Finally, we found Sutherlandioside D was the most potent compound in the crude extract that could suppress Gli-reporter in Shh Light II cells. Together, this suggests that the S. frutescens extract may exert anti-cancer effect by targeting Gli/Hh signaling, and Sutherlandioside D is one of the active compounds.
Collapse
Affiliation(s)
- Hui Lin
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Reproductive Physiology & Embryo Technology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China.,Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| | - Glenn A Jackson
- MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA.,Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, 65211, USA.,Department of Veterinary Technology, Nebraska College of Technical Agriculture, Curtis, Nebraska, 69025, USA
| | - Yuan Lu
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| | - Sara K Drenkhahn
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| | - Korey J Brownstein
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA.,Institute of Biological Chemistry, Washington State University, Pullman, Washington, 99164, USA
| | - Nicholas J Starkey
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| | - William R Lamberson
- MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA.,Department of Animal Sciences, University of Missouri, Columbia, Missouri, 65211, USA
| | - Kevin L Fritsche
- MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA.,Department of Animal Sciences, University of Missouri, Columbia, Missouri, 65211, USA
| | - Valeri V Mossine
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| | - Cynthia L Besch-Williford
- MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA.,Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, 65211, USA
| | - William R Folk
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Reproductive Physiology & Embryo Technology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Dennis B Lubahn
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211, USA.,MU Center for Botanical Interaction Studies, University of Missouri, Columbia, Missouri, 65211, USA
| |
Collapse
|
288
|
Combination Trimodality Therapy Using Vismodegib for Basal Cell Carcinoma of the Face. Case Rep Oncol Med 2015; 2015:827608. [PMID: 26504605 PMCID: PMC4609426 DOI: 10.1155/2015/827608] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/17/2015] [Indexed: 11/24/2022] Open
Abstract
Background. For large basal cell carcinomas (BCCs) of the head and neck, definitive surgery often requires extensive resection and reconstruction that may result in prolonged recovery and limited cosmesis. Vismodegib, a small-molecule inhibitor of the hedgehog pathway, is approved for advanced and metastatic BCCs. We present a case of advanced BCC treated with combination of vismodegib, radiotherapy, and local excision resulting in excellent response and cosmesis. Case Presentation. A 64-year-old gentleman presented with a 5-year history of a 7 cm enlarging right cheek mass, with extensive vascularization, central ulceration, and skin, soft tissue, and buccal mucosa involvement. Biopsy revealed BCC, nodular type. Up-front surgical option involved a large resection and reconstruction. After multidisciplinary discussion, we recommended and he opted for combined modality of vismodegib, radiotherapy, and local excision. The patient tolerated vismodegib well and his right cheek lesion decreased significantly in size. He was then treated with radiotherapy followed by local excision that revealed only focal residual BCC. Currently, he is without evidence of disease and has excellent cosmesis. Conclusions. We report a case of locally advanced BCC treated with trimodality therapy with vismodegib, radiotherapy, and local excision, resulting in excellent outcome and facial cosmesis, without requiring extensive resection or reconstructive surgery.
Collapse
|
289
|
Brown NA, Betz BL. Ameloblastoma: A Review of Recent Molecular Pathogenetic Discoveries. BIOMARKERS IN CANCER 2015; 7:19-24. [PMID: 26483612 PMCID: PMC4597444 DOI: 10.4137/bic.s29329] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 12/30/2022]
Abstract
Ameloblastoma is an odontogenic neoplasm whose molecular pathogenesis has only recently been elucidated. The discovery of recurrent activating mutations in FGFR2, BRAF, and RAS in a large majority of ameloblastomas has implicated dysregulation of MAPK pathway signaling as a critical step in the pathogenesis of this tumor. Some degree of controversy exists regarding the role of mutations affecting the sonic hedgehog (SHH) pathway, specifically Smoothened (SMO), which have been postulated to serve as either an alternative pathogenetic mechanism or secondary mutations. Here, we review recent advances in our understanding of the molecular pathogenesis of ameloblastoma as well as the diagnostic, prognostic, and therapeutic implications of these discoveries.
Collapse
Affiliation(s)
- Noah A Brown
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bryan L Betz
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
290
|
Justilien V, Fields AP. Molecular pathways: novel approaches for improved therapeutic targeting of Hedgehog signaling in cancer stem cells. Clin Cancer Res 2015; 21:505-13. [PMID: 25646180 DOI: 10.1158/1078-0432.ccr-14-0507] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Hedgehog (Hh) signaling pathway is critical for embryonic development. In adult tissues, Hh signaling is relatively quiescent with the exception of roles in tissue maintenance and repair. Aberrant activation of Hh signaling is implicated in multiple aspects of transformation, including the maintenance of the cancer stem cell (CSC) phenotype. Preclinical studies indicate that CSCs from many tumor types are sensitive to Hh pathway inhibition and that Hh-targeted therapeutics block many aspects of transformation attributed to CSCs, including drug resistance, relapse, and metastasis. However, to date, Hh inhibitors, specifically those targeting Smoothened [such as vismodegib, BMS-833923, saridegib (IPI-926), sonidegib/erismodegib (LDE225), PF-04449913, LY2940680, LEQ 506, and TAK-441], have demonstrated good efficacy as monotherapy in patients with basal cell carcinoma and medulloblastoma, but have shown limited activity in other tumor types. This lack of success is likely due to many factors, including a lack of patient stratification in early trials, cross-talk between Hh and other oncogenic signaling pathways that can modulate therapeutic response, and a limited knowledge of Hh pathway activation mechanisms in CSCs from most tumor types. Here, we discuss Hh signaling mechanisms in the context of human cancer, particularly in the maintenance of the CSC phenotype, and consider new therapeutic strategies that hold the potential to expand considerably the scope and therapeutic efficacy of Hh-directed anticancer therapy.
Collapse
Affiliation(s)
- Verline Justilien
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Alan P Fields
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida.
| |
Collapse
|
291
|
Booms P, Harth M, Sader R, Ghanaati S. Vismodegib hedgehog-signaling inhibition and treatment of basal cell carcinomas as well as keratocystic odontogenic tumors in Gorlin syndrome. Ann Maxillofac Surg 2015; 5:14-9. [PMID: 26389028 PMCID: PMC4555941 DOI: 10.4103/2231-0746.161049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Vismodegib hedgehog signaling inhibition treatment has potential for reducing the burden of multiple skin basal cell carcinomas and jaw keratocystic odontogenic tumors. They are major criteria for the diagnosis of Gorlin syndrome, also called nevoid basal cell carcinoma syndrome. Clinical features of Gorlin syndrome are reported, and the relevance of hedgehog signaling pathway inhibition by oral vismodegib for maxillofacial surgeons is highlighted. In summary, progressed basal cell carcinoma lesions are virtually inoperable. Keratocystic odontogenic tumors have an aggressive behavior including rapid growth and extension into adjacent tissues. Interestingly, nearly complete regression of multiple Gorlin syndrome-associated keratocystic odontogenic tumors following treatment with vismodegib. Due to radio-hypersensitivity in Gorlin syndrome, avoidance of treatment by radiotherapy is strongly recommended for all affected individuals. Vismodegib can help in those instances where radiation is contra-indicated, or the lesions are inoperable. The effect of vismodegib on basal cell carcinomas was associated with a significant decrease in hedgehog-signaling and tumor proliferation. Vismodegib, a new and approved drug for the treatment of advanced basal cell carcinoma, is a specific oncogene inhibitor. It also seems to be effective for treatment of keratocystic odontogenic tumors and basal cell carcinomas in Gorlin syndrome, rendering the surgical resections less challenging.
Collapse
Affiliation(s)
- Patrick Booms
- Frankfurt Orofacial Regenerative Medicine, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Marc Harth
- Center for Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Robert Sader
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Shahram Ghanaati
- Frankfurt Orofacial Regenerative Medicine, University Hospital Frankfurt, Frankfurt am Main, Germany ; Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
292
|
Gonzalez AC, Ferreira M, Ariel T, Reis SR, Andrade Z, Peixoto Medrado A. Immunohistochemical evaluation of hedgehog signalling in epithelial/mesenchymal interactions in squamous cell carcinoma transformation: a pilot study. J Oral Pathol Med 2015; 45:173-9. [PMID: 26947270 DOI: 10.1111/jop.12346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2015] [Indexed: 11/28/2022]
Abstract
Precancerous lesions have been studied because of their carcinogenic potential and their association with squamous cell carcinoma (SCC) has been reported. In the tumour microenvironment, the processes of angiogenesis and tissue remodelling are regulated by a family of proteins (Hedgehog) described as being able to modulate epithelial/mesenchymal interactions. The objective of this study was to perform a comparative study of precancerous lesions and SCCs by immunohistochemistry for the presence of Sonic, Gli2, SMO and Patched proteins, members of the Hedgehog pathway. Sixteen cases diagnosed as actinic cheilitis associated with SCC were compared to normal oral mucosa. The sections were subjected to immunohistochemistry and the positively stained cells were counted by morphometric analysis. There was a significant progressive increase in expression of all proteins of the Hedgehog pathway, both in the epithelium and in the connective tissue, when sections of normal mucosa, dysplasia and carcinoma were compared (P < 0.05). Thus, one may suggest that the Hedgehog pathway in tumour transformation influences SCC, and more studies should be conducted to expand the understanding of the role of these proteins in neoplastic transformation.
Collapse
Affiliation(s)
- Ana Cristina Gonzalez
- Laboratory of Experimental Pathology, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Maira Ferreira
- Basic Science, Bahiana Schoool of Medicine and Public Health, Salvador, Bahia, Brazil
| | - Tamires Ariel
- Laboratory of Experimental Pathology, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Sílvia Regina Reis
- Basic Science, Bahiana Schoool of Medicine and Public Health, Salvador, Bahia, Brazil
| | - Zilton Andrade
- Laboratory of Experimental Pathology, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Alena Peixoto Medrado
- Basic Science, Bahiana Schoool of Medicine and Public Health, Salvador, Bahia, Brazil
| |
Collapse
|
293
|
Ma Z, Liu W, Zeng J, Zhou J, Guo P, Xie H, Yang Z, Zheng L, Xu S, Wang X, Chang LS, He D, Li L. Silibinin induces apoptosis through inhibition of the mTOR-GLI1-BCL2 pathway in renal cell carcinoma. Oncol Rep 2015; 34:2461-8. [PMID: 26323996 DOI: 10.3892/or.2015.4224] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 11/05/2022] Open
Abstract
The downstream transcriptional factor of the hedgehog (Hh) pathway, GLI family zinc finger 1 (GLI1), plays a crucial role in regulating tumor progression. In the present study, we demonstrated that silibinin, a natural flavonoid antioxidant isolated from extracts of the milk thistle herb, exerts its anticancer capabilities by restraining GLI1 function in renal cell carcinoma (RCC) cells in vitro and in vivo. In the present study, we confirmed that silibinin induced growth inhibition of RCC through caspase-dependent apoptosis and downregulation of GLI1 and BCL2, which could be partially reversed by GLI1 overexpression. Moreover, we determined that the decreased GLI1 expression by silibinin was mediated by the mammalian target of rapamycin (mTOR) pathway. The in vivo mouse xenograft study also showed that silibinin significantly reduced RCC tumor growth and specifically targeted the mTOR-GLI1-BCL2 signaling pathway. In conclusion, our findings demonstrated for the first time that silibinin induces apoptosis of RCC cells through inhibition of the mTOR-GLI1‑BCL2 pathway. These findings also indicate that GLI1 is a novel regulator for the potential therapeutic application of silibinin against RCC.
Collapse
Affiliation(s)
- Zhenkun Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiancheng Zhou
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hongjun Xie
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zhao Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Long Zheng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Luke S Chang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
294
|
Rovida E, Stecca B. Mitogen-activated protein kinases and Hedgehog-GLI signaling in cancer: A crosstalk providing therapeutic opportunities? Semin Cancer Biol 2015; 35:154-67. [PMID: 26292171 DOI: 10.1016/j.semcancer.2015.08.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 01/07/2023]
Abstract
The Hedgehog-GLI (HH-GLI) signaling is of critical importance during embryonic development, where it regulates a number of cellular processes, including patterning, proliferation and differentiation. Its aberrant activation has been linked to several types of cancer. HH-GLI signaling is triggered by binding of ligands to the transmembrane receptor patched and is subsequently mediated by transcriptional effectors belonging to the GLI family, whose function is fine tuned by a series of molecular interactions and modifications. Several HH-GLI inhibitors have been developed and are in clinical trials. Similarly, the mitogen-activated protein kinases (MAPK) are involved in a number of biological processes and play an important role in many diseases including cancer. Inhibiting molecules targeting MAPK signaling, especially those elicited by the MEK1/2-ERK1/2 pathway, have been developed and are moving into clinical trials. ERK1/2 may be activated as a consequence of aberrant activation of upstream signaling molecules or during development of drug resistance following treatment with kinase inhibitors such as those for PI3K or BRAF. Evidence of a crosstalk between HH-GLI and other oncogenic signaling pathways has been reported in many tumor types, as shown by recent reviews. Here we will focus on the interaction between HH-GLI and the final MAPK effectors ERK1/2, p38 and JNK in cancer in view of its possible implications for cancer therapy. Several reports highlight the existence of a consistent crosstalk between HH signaling and MAPK, especially with the MEK1/2-ERK1/2 pathway, and this fact should be taken into consideration for designing optimal treatment and prevent tumor relapse.
Collapse
Affiliation(s)
- Elisabetta Rovida
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Sezione di Patologia, Università degli Studi di Firenze, Firenze, Italy
| | - Barbara Stecca
- Laboratory of Tumor Cell Biology, Core Research Laboratory-Istituto Toscano Tumori (CRL-ITT), Florence, Italy; Department of Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| |
Collapse
|
295
|
Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX, Ivy SP. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol 2015; 12:445-64. [PMID: 25850553 PMCID: PMC4520755 DOI: 10.1038/nrclinonc.2015.61] [Citation(s) in RCA: 984] [Impact Index Per Article: 98.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During the past decade, cancer stem cells (CSCs) have been increasingly identified in many malignancies. Although the origin and plasticity of these cells remain controversial, tumour heterogeneity and the presence of small populations of cells with stem-like characteristics is established in most malignancies. CSCs display many features of embryonic or tissue stem cells, and typically demonstrate persistent activation of one or more highly conserved signal transduction pathways involved in development and tissue homeostasis, including the Notch, Hedgehog (HH), and Wnt pathways. CSCs generally have slow growth rates and are resistant to chemotherapy and/or radiotherapy. Thus, new treatment strategies targeting these pathways to control stem-cell replication, survival and differentiation are under development. Herein, we provide an update on the latest advances in the clinical development of such approaches, and discuss strategies for overcoming CSC-associated primary or acquired resistance to cancer treatment. Given the crosstalk between the different embryonic developmental signalling pathways, as well as other pathways, designing clinical trials that target CSCs with rational combinations of agents to inhibit possible compensatory escape mechanisms could be of particular importance. We also share our views on the future directions for targeting CSCs to advance the clinical development of these classes of agents.
Collapse
Affiliation(s)
- Naoko Takebe
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - Lucio Miele
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - Pamela Jo Harris
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - Woondong Jeong
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - Hideaki Bando
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - Michael Kahn
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - Sherry X. Yang
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| | - S. Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive MSC9739, Bethesda, MD 20852, USA (N.T., P.J.H., S.P.I.). Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, USA (L.M.). Cancer Therapy and Research Center, University of Texas, USA (W.J.). Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan (H.B.). Norris Comprehensive Cancer Research Center, University of Southern California, USA (M.K.). National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, USA (S.X.Y.)
| |
Collapse
|
296
|
Ozgur OK, Yin V, Chou E, Ball S, Kies M, William WN, Migden M, Thuro BA, Esmaeli B. Hedgehog Pathway Inhibition for Locally Advanced Periocular Basal Cell Carcinoma and Basal Cell Nevus Syndrome. Am J Ophthalmol 2015; 160:220-227.e2. [PMID: 25935097 DOI: 10.1016/j.ajo.2015.04.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/23/2015] [Accepted: 04/27/2015] [Indexed: 11/19/2022]
Abstract
PURPOSE To review our experience treating patients with the Hedgehog pathway inhibitor, vismodegib, in patients with orbital or periocular locally advanced or metastatic basal cell carcinoma (BCC) or basal cell nevus syndrome. DESIGN Retrospective interventional case series. METHODS We reviewed all patients with locally advanced or metastatic orbital or periocular BCC or basal cell nevus syndrome treated with the Hedgehog pathway inhibitor, vismodegib, at a comprehensive cancer center from 2009 through 2015. Reviewed data included age; sex; American Joint Commission on Cancer tumor, node, metastasis staging system designation; type and grade of drug-related side effects; response to treatment; duration of follow-up, and status at last follow-up. RESULTS The study included 10 white men and 2 white women; the median age was 64.5 years. Ten patients had locally advanced BCC; 2 had basal cell nevus syndrome. Among the patients with locally advanced BCC, 5 had T3bN0M0 disease at presentation; 1 each had T3aN0M0, T3bN1M0, T2N1M1, T4N1M1, and T4N2cM1 disease. Overall, 3 patients had a complete response, 6 had a partial response, and 3 had stable disease at last follow-up. Two patients developed progressive disease after a complete response for 38 months and stable disease for 16 months, respectively. All patients developed grade I drug-related adverse effects, most commonly muscle spasms (12 patients), weight loss (10), dysgeusia (9), alopecia (9), decreased appetite (5), and fatigue (4). Five patients developed grade II adverse effects. At last follow-up, none of the 5 patients presenting with T3bN0M0, nor the patient with T3bN1M0 disease, had required orbital exenteration. CONCLUSION Hedgehog pathway inhibition produces a significant clinical response in most patients with locally advanced or metastatic orbital or periocular BCC or basal cell nevus syndrome and can obviate orbital exenteration in some patients. Drug-related adverse effects are manageable in most patients.
Collapse
Affiliation(s)
- Omar K Ozgur
- Department of Plastic Surgery, Orbital Oncology and Ophthalmic Plastic Surgery Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivian Yin
- Department of Plastic Surgery, Orbital Oncology and Ophthalmic Plastic Surgery Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eva Chou
- Department of Plastic Surgery, Orbital Oncology and Ophthalmic Plastic Surgery Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sharon Ball
- Department of Plastic Surgery, Orbital Oncology and Ophthalmic Plastic Surgery Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Merrill Kies
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William N William
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Migden
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bradley A Thuro
- Department of Plastic Surgery, Orbital Oncology and Ophthalmic Plastic Surgery Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bita Esmaeli
- Department of Plastic Surgery, Orbital Oncology and Ophthalmic Plastic Surgery Program, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
297
|
Verma RK, Yu W, Singh SP, Shankar S, Srivastava RK. Anthothecol-encapsulated PLGA nanoparticles inhibit pancreatic cancer stem cell growth by modulating sonic hedgehog pathway. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015. [PMID: 26199979 DOI: 10.1016/j.nano.2015.07.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
UNLABELLED Anthothecol, a limonoid isolated from plant Khaya anthotheca (Meliaceae), is an antimalarial compound. The objectives of this study were to examine the molecular mechanisms by which anthothecol-encapsulated PLGA-nanoparticles (Antho-NPs) regulate the behavior of pancreatic cancer stem cells (CSCs). Antho-NPs inhibited cell proliferation and colony formation, and induced apoptosis in pancreatic CSCs and cancer cell lines, but had no effects on human normal pancreatic ductal epithelial cells. Antho-NPs inhibited self-renewal capacity of pancreatic CSCs isolated from human and Kras(G12D) mice. Furthermore, antho-NPs suppressed cell motility, migration and invasion by up-regulating E-cadherin and inhibiting N-cadherin and Zeb1. In addition, Antho-NPs inhibited pluripotency maintaining factors and stem cell markers, suggesting their inhibitory role on CSC population. Anthothecol disrupted binding of Gli to DNA, and inhibited Gli transcription and Gli target genes. Our studies establish preclinical significance of Antho-NPs for the treatment and/or prevention of pancreatic cancer. FROM THE CLINICAL EDITOR Despite medical advances, the prognosis of pancreatic cancer remains poor. The search for an effective treatment has been under intensive research for some time. In this article, the authors investigated the efficacy and mechanism of anthothecol (an antimalarial compound), encapsulated by PLGA nanoparticles (Antho-NPs), against pancreatic cancer cell lines. It was found that Antho-NPs acted via the Sonic hedgehog signaling pathway and inhibited cancer stem cell growth. These results have provided important basis for further clinical trials.
Collapse
Affiliation(s)
| | - Wei Yu
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Surya Pratap Singh
- Centre of Bioinformatics (IIDS), University of Allahabad, Allahabad, UP, India
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, MO, USA; Department of Pathology, University of Missouri-School of Medicine, Kansas City, MO, USA
| | | |
Collapse
|
298
|
Consensus for nonmelanoma skin cancer treatment: basal cell carcinoma, including a cost analysis of treatment methods. Dermatol Surg 2015; 41:550-71. [PMID: 25868035 DOI: 10.1097/dss.0000000000000296] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Basal cell carcinoma (BCC) is the most common cancer in the US population affecting approximately 2.8 million people per year. Basal cell carcinomas are usually slow-growing and rarely metastasize, but they do cause localized tissue destruction, compromised function, and cosmetic disfigurement. OBJECTIVE To provide clinicians with guidelines for the management of BCC based on evidence from a comprehensive literature review, and consensus among the authors. MATERIALS AND METHODS An extensive review of the medical literature was conducted to evaluate the optimal treatment methods for cutaneous BCC, taking into consideration cure rates, recurrence rates, aesthetic and functional outcomes, and cost-effectiveness of the procedures. RESULTS Surgical approaches provide the best outcomes for BCCs. Mohs micrographic surgery provides the highest cure rates while maximizing tissue preservation, maintenance of function, and cosmesis. CONCLUSION Mohs micrographic surgery is an efficient and cost-effective procedure and remains the treatment of choice for high-risk BCCs and for those in cosmetically sensitive locations. Nonsurgical modalities may be used for low-risk BCCs when surgery is contraindicated or impractical, but the cure rates are lower.
Collapse
|
299
|
Yang JJ, Wu YL. Insight into early-phase trials for lung cancer in the United States. CHINESE JOURNAL OF CANCER 2015; 34:288-94. [PMID: 26162603 PMCID: PMC4593356 DOI: 10.1186/s40880-015-0027-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/15/2015] [Indexed: 01/28/2023]
Abstract
Introduction Few data have been published comparing early-phase trials for lung cancer between China and the United States (US). This study was to investigate the differences of phase 1 trials for lung cancer between these two countries. Methods In 2014, a cross-sectional survey was conducted to compare phase 1 trials for lung cancer between the Guangdong Lung Cancer Institute (GLCI), the University of Wisconsin Carbone Cancer Center (UWCCC), and the University of Texas MD Anderson Cancer Center (MDACC). Results We found that the GLCI had a lower percentage of phase 1 lung cancer trials than the MDACC in December 2014 (23.8% [5/21] vs. 59.8% [28/47], P = 0.006) and the UWCCC in September 2014 (16.7% [3/18] vs. 34.8% [8/23], P = 0.345). Descriptive analyses were performed for early-phase trials conducted by the Cancer Therapy Evaluation Program at the National Cancer Institute (CTEP/NCI), the MDACC, and the Chinese Thoracic Oncology Group (CTONG). There were 149 ongoing early-phase trials in the Department of Investigational Cancer Therapeutics (Phase 1 program) at the MDACC in October 2014. In contrast, no phase 1 trials had been initiated by the CTONG since its establishment in 2007. Conclusions These data suggest that a significantly higher percentage of phase 1 trials for lung cancer were conducted in the US than in China. Early-phase oncology trials with robust preclinical data had a higher chance of being approved by the Investigational Drug Branch at the CTEP/NCI. Given the importance of early-phase oncology trials in developing innovative cancer medicines, such studies should be highly encouraged and strategically funded in China.
Collapse
Affiliation(s)
- Jin-Ji Yang
- Guangdong Lung Cancer Institute (GLCI), Guangdong General Hospital (GGH), Guangdong Academy of Medical Sciences (GAMS), Guangzhou, Guangdong, 510080, P.R. China.
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute (GLCI), Guangdong General Hospital (GGH), Guangdong Academy of Medical Sciences (GAMS), Guangzhou, Guangdong, 510080, P.R. China.
| |
Collapse
|
300
|
Maugeri-Saccà M, Vici P, Di Lauro L, Barba M, Amoreo CA, Gallo E, Mottolese M, De Maria R. Cancer stem cells: are they responsible for treatment failure? Future Oncol 2015; 10:2033-44. [PMID: 25396775 DOI: 10.2217/fon.14.126] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Overcoming resistance to standard anticancer treatments represents a significant challenge. The interest regarding cancer stem cells, a cellular population that has the ability to self-renew and to propagate the tumor, was prompted by experimental evidence delineating the molecular mechanisms that are selectively activated in this cellular subset in order to survive chemotherapy. This has also stimulated combination strategies aimed at rendering cancer stem cells vulnerable to anticancer agents. Moreover, cancer stem cells offer a unique opportunity for modeling human cancers in mice, thus emerging as a powerful tool for testing novel drugs and combinations in a simulation of human disease. These novel animal models may lay the foundation for a new generation of clinical trials aimed at anticipating the benefit to patients of anticancer therapies.
Collapse
Affiliation(s)
- Marcello Maugeri-Saccà
- Division of Medical Oncology B, 'Regina Elena' National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|