101
|
Shih S, Dai C, Ansari A, Urso BA, Laughlin AI, Solomon JA. Advances in genetic understanding of gorlin syndrome and emerging treatment options. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1483233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shawn Shih
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Christina Dai
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Ahmed Ansari
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Brittany A Urso
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Amy I Laughlin
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James A Solomon
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
- University of Illinois College of Medicine, Urbana, Illinois
- Ameriderm Research, Ormond Beach, FL
- Department of Dermatology, Florida State University College of Medicine, Tallahassee, FL
| |
Collapse
|
102
|
Latuske EM, Stamm H, Klokow M, Vohwinkel G, Muschhammer J, Bokemeyer C, Jücker M, Kebenko M, Fiedler W, Wellbrock J. Combined inhibition of GLI and FLT3 signaling leads to effective anti-leukemic effects in human acute myeloid leukemia. Oncotarget 2018; 8:29187-29201. [PMID: 28418873 PMCID: PMC5438723 DOI: 10.18632/oncotarget.16304] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/20/2017] [Indexed: 12/13/2022] Open
Abstract
Activation of the Hedgehog pathway has been implicated in the pathogenesis of several tumor types including myeloid leukemia. Previously we demonstrated that overexpression of Hedgehog downstream mediators GLI1/2 confers an adverse prognosis to patients with acute myeloid leukemia (AML) and is correlated with a FLT3 mutated status. To analyze a possible non-canonical activation of the Hedgehog pathway via FLT3 and PI3K, we performed blocking experiments utilizing inhibitors for FLT3 (sunitinib), PI3K (PF-04691502) and GLI1/2 (GANT61) in FLT3-mutated and FLT3 wildtype AML cell lines and primary blasts. Combination of all three compounds had stronger anti-leukemic effects in FLT3-mutated compared to FLT3 wildtype AML cells in vitro. Interestingly, the colony growth of normal CD34+ cells from healthy donors was not impeded by the triple inhibitor combination possibly opening a therapeutic window for the clinical use of inhibitor combinations. Besides, combined treatment with sunitinib, PF-04691502 and GANT61 significantly prolonged the survival of mice transplanted with FLT3-mutated MV4-11 cells compared to the single agent treatments. Furthermore, the inhibition of FLT3 and PI3K resulted in reduced GLI protein expression and promotor activity in FLT3-mutated but not in FLT3 wildtype AML cell lines in western blotting and GLI1/2 promoter assays supporting our hypothesis of non-canonical GLI activation via FLT3. In summary, FLT3-mutated in contrast to FLT3 wildtype cells or normal human hematopoietic progenitor cells are exquisitely sensitive to combined inhibition by FLT3, PI3K and GLI1/2 overcoming some of the limitations of current FLT3 directed therapy in AML. The development of GLI1/2 inhibitors is highly desirable.
Collapse
Affiliation(s)
- Emily-Marie Latuske
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hauke Stamm
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Klokow
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabi Vohwinkel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Muschhammer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maxim Kebenko
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
103
|
Wang J, Garancher A, Ramaswamy V, Wechsler-Reya RJ. Medulloblastoma: From Molecular Subgroups to Molecular Targeted Therapies. Annu Rev Neurosci 2018; 41:207-232. [PMID: 29641939 DOI: 10.1146/annurev-neuro-070815-013838] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain tumors are the leading cause of cancer-related death in children, and medulloblastoma (MB) is the most common malignant pediatric brain tumor. Advances in surgery, radiation, and chemotherapy have improved the survival of MB patients. But despite these advances, 25-30% of patients still die from the disease, and survivors suffer severe long-term side effects from the aggressive therapies they receive. Although MB is often considered a single disease, molecular profiling has revealed a significant degree of heterogeneity, and there is a growing consensus that MB consists of multiple subgroups with distinct driver mutations, cells of origin, and prognosis. Here, we review recent progress in MB research, with a focus on the genes and pathways that drive tumorigenesis, the animal models that have been developed to study tumor biology, and the advances in conventional and targeted therapy.
Collapse
Affiliation(s)
- Jun Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| | - Alexandra Garancher
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| | - Vijay Ramaswamy
- Division of Haematology/Oncology and Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| |
Collapse
|
104
|
Palacios-Álvarez I, González-Sarmiento R, Fernández-López E. Gorlin Syndrome. ACTAS DERMO-SIFILIOGRAFICAS 2018. [DOI: 10.1016/j.adengl.2018.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
105
|
Byrne EF, Luchetti G, Rohatgi R, Siebold C. Multiple ligand binding sites regulate the Hedgehog signal transducer Smoothened in vertebrates. Curr Opin Cell Biol 2018; 51:81-88. [PMID: 29268141 PMCID: PMC5949240 DOI: 10.1016/j.ceb.2017.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 12/31/2022]
Abstract
The Hedgehog (Hh) pathway plays a central role in the development of multicellular organisms, guiding cell differentiation, proliferation and survival. While many components of the vertebrate pathway were discovered two decades ago, the mechanism by which the Hh signal is transmitted across the plasma membrane remains mysterious. This fundamental task in signalling is carried out by Smoothened (SMO), a human oncoprotein and validated cancer drug target that is a member of the G-protein coupled receptor protein family. Recent structural and functional studies have advanced our mechanistic understanding of SMO activation, revealing its unique regulation by two separable but allosterically-linked ligand-binding sites. Unexpectedly, these studies have nominated cellular cholesterol as having an instructive role in SMO signalling.
Collapse
Affiliation(s)
- Eamon Fx Byrne
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Giovanni Luchetti
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, United States.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
106
|
Danhof R, Lewis K, Brown M. Small Molecule Inhibitors of the Hedgehog Pathway in the Treatment of Basal Cell Carcinoma of the Skin. Am J Clin Dermatol 2018; 19:195-207. [PMID: 28887802 DOI: 10.1007/s40257-017-0319-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Basal cell carcinoma (BCC) is the most common type of skin cancer, with rising incidence rates primarily attributed to an aging population and ultraviolet radiation exposure. While the majority of BCCs are localized and respond to standard therapies, a very small minority of these tumors become locally destructive or metastasize. These advanced BCCs may not be amenable to localized treatment with surgery and/or radiation therapy. Most BCCs result from mutations in key receptors in the Hedgehog (HH) signaling pathway. As a result, identification of drugs that inhibit the receptor Smoothened (SMO) in the HH pathway has resulted in novel therapeutic approaches to treating patients with advanced BCC. These HH-pathway inhibiting medications have shown efficacy in clinical trials, and two medications, vismodegib and sonidegib, have received FDA approval. However, several limitations of these drugs have been identified, including treatment-limiting adverse events, drug resistance, and the formation of additional malignancies. This paper aims to summarize the clinical trials leading to the approval of SMO inhibitors, as well as reviewing potential mechanisms driving tumor resistance and the formation of cutaneous squamous cell carcinomas. Strategies to overcome some of these challenges, including the development of drugs that inhibit other downstream targets in the HH pathway, are the subject of ongoing clinical trials.
Collapse
Affiliation(s)
- Rebecca Danhof
- Department of Dermatology, Mayo Clinic College of Medicine and Science, Onalaska, WI, USA
| | - Karl Lewis
- Division of Hematology and Oncology, Department of Internal Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mariah Brown
- Department of Dermatology, University of Colorado School of Medicine, Mail Stop F703, 1665, North Aurora Court, Aurora, CO, 80045, USA.
| |
Collapse
|
107
|
Chaturvedi NK, Kling MJ, Coulter DW, McGuire TR, Ray S, Kesherwani V, Joshi SS, Sharp JG. Improved therapy for medulloblastoma: targeting hedgehog and PI3K-mTOR signaling pathways in combination with chemotherapy. Oncotarget 2018; 9:16619-16633. [PMID: 29682173 PMCID: PMC5908274 DOI: 10.18632/oncotarget.24618] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/21/2018] [Indexed: 02/07/2023] Open
Abstract
Aberrant activation and interactions of hedgehog (HH) and PI3K/AKT/mTOR signaling pathways are frequently associated with high-risk medulloblastoma (MB). Thus, combined targeting of the HH and PI3K/AKT/mTOR pathways could be a viable therapeutic strategy to treat high-risk patients. Therefore, we investigated the anti-MB efficacies of combined HH inhibitor Vismodegib and PI3K-mTOR dual-inhibitor BEZ235 together or combined individually with cisplatin against high-risk MB. Using non-MYC- and MYC-amplified cell lines, and a xenograft mouse model, the in vitro and in vivo efficacies of these therapies on cell growth/survival and associated molecular mechanism(s) were investigated. Results showed that combined treatment of Vismodegib and BEZ235 together, or with cisplatin, significantly decreased MB cell growth/survival in a dose-dependent-fashion. Corresponding changes in the expression of targeted molecules following therapy were observed. Results demonstrated that inhibitors not only suppressed MB cell growth/survival when combined, but also significantly enhanced cisplatin-mediated cytotoxicity. Of these combinations, BEZ235 exhibited a significantly greater efficacy in enhancing cisplatin-mediated MB cytotoxicity. Results also demonstrated that the MYC-amplified MB lines showed a higher sensitivity to combined therapies compared to non-MYC-amplified cell lines. Therefore, we tested the efficacy of combined approaches against MYC-amplified MB growing in NSG mice. In vivo results showed that combination of Vismodegib and BEZ235 or their combination with cisplatin, significantly delayed MB tumor growth and increased survival of xenografted mice by targeting HH and mTOR pathways. Thus, our studies lay a foundation for translating these combined therapeutic strategies to the clinical setting to determine their efficacies in high-risk MB patients.
Collapse
Affiliation(s)
- Nagendra K Chaturvedi
- Departments of Pediatrics, Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Matthew J Kling
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Don W Coulter
- Departments of Pediatrics, Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Timothy R McGuire
- Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Sutapa Ray
- Departments of Pediatrics, Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Varun Kesherwani
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Shantaram S Joshi
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - J Graham Sharp
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 69198, USA
| |
Collapse
|
108
|
Carballo GB, Honorato JR, de Lopes GPF, Spohr TCLDSE. A highlight on Sonic hedgehog pathway. Cell Commun Signal 2018; 16:11. [PMID: 29558958 PMCID: PMC5861627 DOI: 10.1186/s12964-018-0220-7] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/16/2018] [Indexed: 12/25/2022] Open
Abstract
Hedgehog (Hh) signaling pathway plays an essential role during vertebrate embryonic development and tumorigenesis. It is already known that Sonic hedgehog (Shh) pathway is important for the evolution of radio and chemo-resistance of several types of tumors. Most of the brain tumors are resistant to chemotherapeutic drugs, consequently, they have a poor prognosis. So, a better knowledge of the Shh pathway opens an opportunity for targeted therapies against brain tumors considering a multi-factorial molecular overview. Therefore, emerging studies are being conducted in order to find new inhibitors for Shh signaling pathway, which could be safely used in clinical trials. Shh can signal through a canonical and non-canonical way, and it also has important points of interaction with other pathways during brain tumorigenesis. So, a better knowledge of Shh signaling pathway opens an avenue of possibilities for the treatment of not only for brain tumors but also for other types of cancers. In this review, we will also highlight some clinical trials that use the Shh pathway as a target for treating brain cancer.
Collapse
Affiliation(s)
- Gabriela Basile Carballo
- Laboratorio de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rua do Rezende 156, Centro, Rio de Janeiro, CEP: 20230-024, Brazil.,Programa de Pós-Gradução em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jéssica Ribeiro Honorato
- Laboratorio de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rua do Rezende 156, Centro, Rio de Janeiro, CEP: 20230-024, Brazil.,Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Pesquisa em Hemato-Oncologia Molecular, Coordenação de Pesquisa, Instituto Nacional de Câncer (INCA), RJ, Brazil.,Programa de Pós-Gradução em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle Pinto Farias de Lopes
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Pesquisa em Hemato-Oncologia Molecular, Coordenação de Pesquisa, Instituto Nacional de Câncer (INCA), RJ, Brazil
| | - Tania Cristina Leite de Sampaio E Spohr
- Laboratorio de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rua do Rezende 156, Centro, Rio de Janeiro, CEP: 20230-024, Brazil.
| |
Collapse
|
109
|
Presutti D, Ceccarelli M, Micheli L, Papoff G, Santini S, Samperna S, Lalli C, Zentilin L, Ruberti G, Tirone F. Tis21-gene therapy inhibits medulloblastoma growth in a murine allograft model. PLoS One 2018. [PMID: 29538458 PMCID: PMC5851620 DOI: 10.1371/journal.pone.0194206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Medulloblastoma (MB), the tumor of the cerebellum, is the most frequent brain cancer in childhood and a major cause of pediatric mortality. Based on gene profiling, four MB subgroups have been identified, i.e., Wnt or Sonic Hedgehog (Shh) types, and subgroup 3 or 4. The Shh-type MB has been shown to arise from the cerebellar precursors of granule neurons (GCPs), where a hyperactivation of the Shh pathway leads to their neoplastic transformation. We have previously shown that the gene Tis21 (PC3/Btg2) inhibits the proliferation and promotes the differentiation and migration of GCPs. Moreover, the overexpression or the deletion of Tis21 in Patched1 heterozygous mice, a model of spontaneous Shh-type MB, highly reduces or increases, respectively, the frequency of MB. Here we tested whether Tis21 can inhibit MB allografts. Athymic nude mice were subcutaneously grafted with MB cells explanted from Patched1 heterozygous mice. MB allografts were then injected with adeno-associated viruses either carrying Tis21 (AAV-Tis21) or empty (AAV-CBA). We observed that the treatment with AAV-Tis21 significantly inhibited the growth of tumor nodules, as judged by their volume, and reduced the number of proliferating tumor cells (labeled with Ki67 or BrdU), relative to AAV-CBA-treated control mice. In parallel, AAV-Tis21 increased significantly tumor cells labeled with early and late neural differentiation markers. Overall the results suggest that Tis21-gene therapy slows down MB tumor growth through inhibition of proliferation and enhancement of neural differentiation. These results validate Tis21 as a relevant target for MB therapy.
Collapse
Affiliation(s)
- Dario Presutti
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Monterotondo, Rome, Italy
| | - Manuela Ceccarelli
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Fondazione Santa Lucia, Rome, Italy
| | - Laura Micheli
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Fondazione Santa Lucia, Rome, Italy
| | - Giuliana Papoff
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Monterotondo, Rome, Italy
| | - Simonetta Santini
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Monterotondo, Rome, Italy
| | - Simone Samperna
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Monterotondo, Rome, Italy
| | - Cristiana Lalli
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Monterotondo, Rome, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, Trieste, Italy
| | - Giovina Ruberti
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Monterotondo, Rome, Italy
- * E-mail: (GR); (FT)
| | - Felice Tirone
- Institute of Cell Biology and Neurobiology, National Research Council (IBCN-CNR), Fondazione Santa Lucia, Rome, Italy
- * E-mail: (GR); (FT)
| |
Collapse
|
110
|
Dong X, Wang C, Chen Z, Zhao W. Overcoming the resistance mechanisms of Smoothened inhibitors. Drug Discov Today 2018; 23:704-710. [DOI: 10.1016/j.drudis.2018.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/15/2017] [Accepted: 01/04/2018] [Indexed: 12/31/2022]
|
111
|
Li H, Yue D, Jin JQ, Woodard GA, Tolani B, Luh TM, Giroux-Leprieur E, Mo M, Chen Z, Che J, Zhang Z, Zhou Y, Wang L, Hao X, Jablons D, Wang C, He B. Gli promotes epithelial-mesenchymal transition in human lung adenocarcinomas. Oncotarget 2018; 7:80415-80425. [PMID: 27533453 PMCID: PMC5348330 DOI: 10.18632/oncotarget.11246] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/28/2016] [Indexed: 01/18/2023] Open
Abstract
Adenocarcinoma is the most common type of lung cancer. Epithelial-mesenchymal transition (EMT) is required for tumor invasion/metastasis and the components that control this process are potential therapeutic targets. This study we examined the role of Gli in lung adenocarcinoma and whether its activation regulates metastasis through EMT in lung adenocarcinoma. We found that tumors with high Gli expression had significantly lower E-Cadherin expression in two independent cohorts of patients with lung adenocarcinoma that we studied. In vitro up-regulation of SHh resulted in increased cell migration while small molecule inhibitors of Smo or Gli significantly reduced cell mobility both in a wound healing assay and in a 3D cell invasion assay. Inhibition of Gli in vivo decreased tumor growth and induced an increase in E-Cadherin expression. Our results indicate that Gli may be critical for lung adenocarcinoma metastasis and that a novel Gli inhibitor shows promise as a therapeutic agent by preventing cell migration and invasion in vitro and significantly reducing tumor growth and increasing E-Cadherin expression in vivo.
Collapse
Affiliation(s)
- Hui Li
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Dongsheng Yue
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA.,Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Joy Q Jin
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Gavitt A Woodard
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Bhairavi Tolani
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Thomas M Luh
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Etienne Giroux-Leprieur
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Minli Mo
- Beijing ACCB Biotech Ltd., Beijing 100084, China
| | - Zhao Chen
- Beijing ACCB Biotech Ltd., Beijing 100084, China
| | - Juanjuan Che
- Department of Oncology, Beijing Friendship Hospital of Capital Medical University, Beijing 100050, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yong Zhou
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Lei Wang
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA.,Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xishan Hao
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - David Jablons
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| | - Changli Wang
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Biao He
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
| |
Collapse
|
112
|
Pusapati GV, Kong JH, Patel BB, Gouti M, Sagner A, Sircar R, Luchetti G, Ingham PW, Briscoe J, Rohatgi R. G protein-coupled receptors control the sensitivity of cells to the morphogen Sonic Hedgehog. Sci Signal 2018; 11:eaao5749. [PMID: 29438014 PMCID: PMC5828112 DOI: 10.1126/scisignal.aao5749] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The morphogen Sonic Hedgehog (SHH) patterns tissues during development by directing cell fates in a concentration-dependent manner. The SHH signal is transmitted across the membrane of target cells by the heptahelical transmembrane protein Smoothened (SMO), which activates the GLI family of transcription factors through a mechanism that is undefined in vertebrates. Using CRISPR-edited null alleles and small-molecule inhibitors, we systematically analyzed the epistatic interactions between SMO and three proteins implicated in SMO signaling: the heterotrimeric G protein subunit GαS, the G protein-coupled receptor kinase 2 (GRK2), and the GαS-coupled receptor GPR161. Our experiments uncovered a signaling mechanism that modifies the sensitivity of target cells to SHH and consequently changes the shape of the SHH dose-response curve. In both fibroblasts and spinal neural progenitors, the loss of GPR161, previously implicated as an inhibitor of basal SHH signaling, increased the sensitivity of target cells across the entire spectrum of SHH concentrations. Even in cells lacking GPR161, GRK2 was required for SHH signaling, and Gαs, which promotes the activation of protein Kinase A (PKA), antagonized SHH signaling. We propose that the sensitivity of target cells to Hedgehog morphogens, and the consequent effects on gene expression and differentiation outcomes, can be controlled by signals from G protein-coupled receptors that converge on Gαs and PKA.
Collapse
Affiliation(s)
- Ganesh V Pusapati
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer H Kong
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bhaven B Patel
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mina Gouti
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Andreas Sagner
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Ria Sircar
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Giovanni Luchetti
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798, Singapore
- Living Systems Institute, University of Exeter, Exeter EX4 4RJ, UK
| | - James Briscoe
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Rajat Rohatgi
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
113
|
Pietrobono S, Santini R, Gagliardi S, Dapporto F, Colecchia D, Chiariello M, Leone C, Valoti M, Manetti F, Petricci E, Taddei M, Stecca B. Targeted inhibition of Hedgehog-GLI signaling by novel acylguanidine derivatives inhibits melanoma cell growth by inducing replication stress and mitotic catastrophe. Cell Death Dis 2018; 9:142. [PMID: 29396391 PMCID: PMC5833413 DOI: 10.1038/s41419-017-0142-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 12/21/2022]
Abstract
Aberrant activation of the Hedgehog (HH) signaling is a critical driver in tumorigenesis. The Smoothened (SMO) receptor is one of the major upstream transducers of the HH pathway and a target for the development of anticancer agents. The SMO inhibitor Vismodegib (GDC-0449/Erivedge) has been approved for treatment of basal cell carcinoma. However, the emergence of resistance during Vismodegib treatment and the occurrence of numerous side effects limit its use. Our group has recently discovered and developed novel and potent SMO inhibitors based on acylguanidine or acylthiourea scaffolds. Here, we show that the two acylguanidine analogs, compound (1) and its novel fluoride derivative (2), strongly reduce growth and self-renewal of melanoma cells, inhibiting the level of the HH signaling target GLI1 in a dose-dependent manner. Both compounds induce apoptosis and DNA damage through the ATR/CHK1 axis. Mechanistically, they prevent G2 to M cell cycle transition, and induce signs of mitotic aberrations ultimately leading to mitotic catastrophe. In a melanoma xenograft mouse model, systemic treatment with 1 produced a remarkable inhibition of tumor growth without body weight loss in mice. Our data highlight a novel route for cell death induction by SMO inhibitors and support their use in therapeutic approaches for melanoma and, possibly, other types of cancer with active HH signaling.
Collapse
Affiliation(s)
| | - Roberta Santini
- Core Research Laboratory, Istituto Toscano Tumori, Florence, Italy
| | | | - Francesca Dapporto
- Consiglio Nazionale delle Ricerche, Istituto di Fisiologia Clinica and Core Research Laboratory, Istituto Toscano Tumori, AOU Senese, Siena, Italy
| | - David Colecchia
- Consiglio Nazionale delle Ricerche, Istituto di Fisiologia Clinica and Core Research Laboratory, Istituto Toscano Tumori, AOU Senese, Siena, Italy
| | - Mario Chiariello
- Consiglio Nazionale delle Ricerche, Istituto di Fisiologia Clinica and Core Research Laboratory, Istituto Toscano Tumori, AOU Senese, Siena, Italy
| | - Cosima Leone
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Massimo Valoti
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Elena Petricci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Maurizio Taddei
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Barbara Stecca
- Core Research Laboratory, Istituto Toscano Tumori, Florence, Italy. .,Department of Oncology, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
114
|
Magistri P, Battistelli C, Strippoli R, Petrucciani N, Pellinen T, Rossi L, Mangogna L, Aurello P, D'Angelo F, Tripodi M, Ramacciato G, Nigri G. SMO Inhibition Modulates Cellular Plasticity and Invasiveness in Colorectal Cancer. Front Pharmacol 2018; 8:956. [PMID: 29456503 PMCID: PMC5801594 DOI: 10.3389/fphar.2017.00956] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/15/2017] [Indexed: 12/17/2022] Open
Abstract
HIGHLIGHTS Preliminary results of this work were presented at the 2016 Academic Surgical Congress, Jacksonville (FL), February 2-4 2016 (Original title: Selective Smo-Inhibition Interferes With Cellular Energetic Metabolism In Colorectal Cancer)This study was funded by "Sapienza-University of Rome" (Funds for young researchers) and "AIRC" (Italian Association for Cancer Research)Hedgehog inhibitor was kindly provided by Genentech, Inc.®. Colon Cancer (CC) is the fourth most frequently diagnosed tumor and the second leading cause of death in the USA. Abnormalities of Hedgehog pathway have been demonstrated in several types of human cancers, however the role of Hedgehog (Hh) in CC remain controversial. In this study, we analyzed the association between increased mRNA expression of GLI1 and GLI2, two Hh target genes, and CC survival and recurrence by gene expression microarray from a cohort of 382 CC patients. We found that patients with increased expression of GLI1 showed a statistically significant reduction in survival. In order to demonstrate a causal role of Hh pathway activation in the pathogenesis of CC, we treated HCT 116, SW480 and SW620 CC cells lines with GDC-0449, a pharmacological inhibitor of Smoothened (SMO). Treatment with GDC-0449 markedly reduced expression of Hh target genes GLI1, PTCH1, HIP1, MUC5AC, thus indicating that this pathway is constitutively active in CC cell lines. Moreover, GDC-0449 partially reduced cell proliferation, which was associated with upregulation of p21 and downregulation of CycD1. Finally, treatment with the same drug reduced migration and three-dimensional invasion, which were associated with downregulation of Snail1, the EMT master gene, and with induction of the epithelial markers Cytokeratin-18 and E-cadherin. These results were confirmed by SMO genetic silencing. Notably, treatment with 5E1, a Sonic Hedgehog-specific mAb, markedly reduced the expression of Hedgehog target genes, as well as inhibited cell proliferation and mediated reversion toward an epithelial phenotype. This suggests the existence of a Hedgehog autocrine signaling loop affecting cell plasticity and fostering cell proliferation and migration/invasion in CC cell lines. These discoveries encourage future investigations to better characterize the role of Hedgehog in cellular plasticity and invasion during the different steps of CC pathogenesis.
Collapse
Affiliation(s)
- Paolo Magistri
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Cecilia Battistelli
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Raffaele Strippoli
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Niccolò Petrucciani
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Teijo Pellinen
- FIMM Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Lucia Rossi
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Livia Mangogna
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Paolo Aurello
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Francesco D'Angelo
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Marco Tripodi
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Giovanni Ramacciato
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Giuseppe Nigri
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| |
Collapse
|
115
|
Palacios-Álvarez I, González-Sarmiento R, Fernández-López E. Gorlin Syndrome. ACTAS DERMO-SIFILIOGRAFICAS 2018; 109:207-217. [PMID: 29373110 DOI: 10.1016/j.ad.2017.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/30/2017] [Accepted: 07/01/2017] [Indexed: 01/08/2023] Open
Abstract
Gorlin syndrome is a rare autosomal dominant disease caused by mutations in the sonic hedgehog signaling pathway. Of particular importance is the PTCH1 gene. The disease is characterized by the development of multiple basal cell carcinomas at young ages. These tumors may present with other skin manifestations such as palmoplantar pits and with extracutaneous manifestations such as odontogenic keratocysts and medulloblastoma. Although the dermatologist may be key for recognizing clinical suspicion of the syndrome, a multidisciplinary team is usually necessary for diagnosis, treatment, and follow-up. Skin treatment may be complicated due to the large number of basal cell carcinomas and the extent of involvement. In recent years, new drugs that inhibit targets in the sonic hedgehog pathway have been developed. Although these agents appear promising options for patients with Gorlin syndrome, their efficacy is limited by adverse effects and the development of resistance.
Collapse
Affiliation(s)
- I Palacios-Álvarez
- Departamento de Dermatología, Clínica Universidad de Navarra, Pamplona, España.
| | - R González-Sarmiento
- Unidad de Medicina Molecular, Facultad de Medicina, Universidad de Salamanca, Salamanca, España; Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Salamanca, España
| | - E Fernández-López
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Salamanca, España; Departamento de Dermatología, Hospital Clínico Universitario, Salamanca, España
| |
Collapse
|
116
|
A synthetic combinatorial approach to disabling deviant Hedgehog signaling. Sci Rep 2018; 8:1133. [PMID: 29348431 PMCID: PMC5773580 DOI: 10.1038/s41598-018-19408-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/29/2017] [Indexed: 12/20/2022] Open
Abstract
Mutations in components of the Hedgehog (HH) signal transduction pathway are found in the majority of basal cell carcinoma (BCC) and medulloblastoma incidents. Cancerous cells with intrinsic or acquired resistance to antagonists targeting the seven transmembrane effector Smoothened (SMO) frequently invoke alternative mechanisms for maintaining deviant activity of the GLI DNA binding proteins. Here we introduce a chemical agent that simultaneously achieves inhibition of SMO and GLI activity by direct targeting of the SMO heptahelical domain and the GLI-modifying enzymes belonging to the histone deacetylase (HDAC) family. We demonstrate a small molecule SMO-HDAC antagonist (IHR-SAHA) retains inhibitory activity for GLI transcription induced by SMO-dependent and -independent mechanisms frequently associated with cancer biogenesis. Synthetic combinatorial therapeutic agents such as IHR-SAHA that a priori disable cancer drivers and anticipated mechanisms of drug resistance could extend the duration of disease remission, and provide an alternative clinical development path for realizing combinatorial therapy modalities.
Collapse
|
117
|
Xin M, Ji X, De La Cruz LK, Thareja S, Wang B. Strategies to target the Hedgehog signaling pathway for cancer therapy. Med Res Rev 2018; 38:870-913. [PMID: 29315702 DOI: 10.1002/med.21482] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/09/2017] [Accepted: 12/13/2017] [Indexed: 01/10/2023]
Abstract
Hedgehog (Hh) signaling is an essential pathway in the human body, and plays a major role in embryo development and tissue patterning. Constitutive activation of the Hh signaling pathway through sporadic mutations or other mechanisms is explicitly associated with cancer development and progression in various solid malignancies. Therefore, targeted inhibition of the Hh signaling pathway has emerged as an attractive and validated therapeutic strategy for the treatment of a wide range of cancers. Vismodegib, a first-in-class Hh signaling pathway inhibitor was approved by the US Food and Drug Administration in 2012, and sonidegib, another potent Hh pathway inhibitor, received FDA's approval in 2015 as a new treatment of locally advanced or metastatic basal cell carcinoma. The clinical success of vismodegib and sonidegib provided strong support for the development of Hh signaling pathway inhibitors via targeting the smoothened (Smo) receptor. Moreover, Hh signaling pathway inhibitors aimed to target proteins, which are downstream or upstream of Smo, have also been pursued based on the identification of additional therapeutic benefits. Recently, much progress has been made in Hh singling and inhibitors of this pathway. Herein, medicinal chemistry strategies, especially the structural optimization process of different classes of Hh inhibitors, are comprehensively summarized. Further therapeutic potentials and challenges are also discussed.
Collapse
Affiliation(s)
- Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, P.R. China.,Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Xinyue Ji
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Ladie Kimberly De La Cruz
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Suresh Thareja
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
118
|
Recurrent extraneural sonic hedgehog medulloblastoma exhibiting sustained response to vismodegib and temozolomide monotherapies and inter-metastatic molecular heterogeneity at progression. Oncotarget 2018. [PMID: 29515801 PMCID: PMC5839382 DOI: 10.18632/oncotarget.23699] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Response to targeting and non-targeting agents is variable and molecular information remains poorly described in patients with recurrent sonic-hedgehog-driven medulloblastoma (SHH-MB). Materials and Methods Clinical and PET/CT findings during treatment with successive hedgehog antagonists and temozolomide monotherapies are described in a heavily pre-treated patient with recurrent extraneural metastases from PTCH1 mutated/ wild type smoothened (SMO) CNS SHH-MB. Molecular tests were prospectively performed in tissue from two extraneural sites at progression. Results Sustained clinical/metabolic response was obtained to vismodegib. At progression, itraconazole was ineffective, but salvage temozolomide treatment results in a response similar to vismodegib. At further progression, acquired SMO and PIK3CA mutations were identified in bone (G477L and H1047A, respectively) and epidural (L412P and H1065L, respectively) metastases. No response was observed with subsequent sonidegib treatment. Conclusions This is the first clinical report of recurrent extraneural PTCH1 mutated SHH-MB exhibiting: 1) a sustained response to vismodegib and temozolomide, and 2) inter-metastatic molecular heterogeneity and acquired SMO-G477L, SMO-L412P, and PIK3CA-H1065L mutations at progression, highlighting the need for a multitarget treatment approach.
Collapse
|
119
|
Wu CC, Hou S, Orr BA, Kuo BR, Youn YH, Ong T, Roth F, Eberhart CG, Robinson GW, Solecki DJ, Taketo MM, Gilbertson RJ, Roussel MF, Han YG. mTORC1-Mediated Inhibition of 4EBP1 Is Essential for Hedgehog Signaling-Driven Translation and Medulloblastoma. Dev Cell 2017; 43:673-688.e5. [PMID: 29103956 PMCID: PMC5736446 DOI: 10.1016/j.devcel.2017.10.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 08/29/2017] [Accepted: 10/07/2017] [Indexed: 12/13/2022]
Abstract
Mechanistic target of rapamycin (MTOR) cooperates with Hedgehog (HH) signaling, but the underlying mechanisms are incompletely understood. Here we provide genetic, biochemical, and pharmacologic evidence that MTOR complex 1 (mTORC1)-dependent translation is a prerequisite for HH signaling. The genetic loss of mTORC1 function inhibited HH signaling-driven growth of the cerebellum and medulloblastoma. Inhibiting translation or mTORC1 blocked HH signaling. Depleting 4EBP1, an mTORC1 target that inhibits translation, alleviated the dependence of HH signaling on mTORC1. Consistent with this, phosphorylated 4EBP1 levels were elevated in HH signaling-driven medulloblastomas in mice and humans. In mice, an mTORC1 inhibitor suppressed medulloblastoma driven by a mutant SMO that is inherently resistant to existing SMO inhibitors, prolonging the survival of the mice. Our study reveals that mTORC1-mediated translation is a key component of HH signaling and an important target for treating medulloblastoma and other cancers driven by HH signaling.
Collapse
Affiliation(s)
- Chang-Chih Wu
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Shirui Hou
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Bryan R Kuo
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yong Ha Youn
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Taren Ong
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Fanny Roth
- Sorbonne Universités, UPMC Paris 06, INSERM, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 47 Boulevard de l'hôpital, Paris 13, Paris, France
| | - Charles G Eberhart
- Department of Pathology, The Johns Hopkins University School of Medicine, Ross Building 558, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Giles W Robinson
- Department of Oncology, Division of Neuro-Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David J Solecki
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-Konoé-cho, Sakyo, Kyoto 606-8501, Japan
| | - Richard J Gilbertson
- Department of Oncology and CRUK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, England
| | - Martine F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
120
|
Abstract
Signaling pathways direct organogenesis, often through concentration-dependent effects on cells. The hedgehog pathway enables cells to sense and respond to hedgehog ligands, of which the best studied is sonic hedgehog. Hedgehog signaling is essential for development, proliferation, and stem cell maintenance, and it is a driver of certain cancers. Lipid metabolism has a profound influence on both hedgehog signal transduction and the properties of the ligands themselves, leading to changes in the strength of hedgehog signaling and cellular functions. Here we review the evolving understanding of the relationship between lipids and hedgehog signaling.
Collapse
Affiliation(s)
- Robert Blassberg
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - John Jacob
- Nuffield Department of Clinical Neurosciences (NDCN), Level 6, West Wing, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK. .,Department of Neurology, West Wing, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK. .,Milton Keynes University Hospital, Standing Way, Eaglestone, Milton Keynes, MK6 5LD, UK.
| |
Collapse
|
121
|
Zhou Q, Zhou Y, Liu X, Shen Y. GDC-0449 improves the antitumor activity of nano-doxorubicin in pancreatic cancer in a fibroblast-enriched microenvironment. Sci Rep 2017; 7:13379. [PMID: 29042665 PMCID: PMC5645386 DOI: 10.1038/s41598-017-13869-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/03/2017] [Indexed: 01/04/2023] Open
Abstract
Pancreatic cancer is one of the most lethal human cancers that currently does not have effective therapies. Novel treatments including nanomedicines and combination therapies are thus urgently needed for these types of deadly diseases. A key feature of pancreatic cancer is its tumor protective dense stroma, which is generated by cancer-associated fibroblasts (CAFs). The interaction between CAFs and pancreatic cancer cells abnormally activates sonic hedgehog (SHH) signaling and facilitates tumor growth, metastasis, and drug resistance. Here, we report that the commercial SHH inhibitor GDC-0449 reverses fibroblast-induced resistance to doxorubicin in Smoothened (SMO)-positive pancreatic cancer cells by downregulating SHH signaling proteins. Importantly, the synergistic combination of GDC-0449 with PEG-PCL-Dox exhibited potent antitumor efficacy in a BxPC-3 tumor xenograft model, whereas single treatments did not significantly inhibit tumor growth. Our findings reveal a potential treatment strategy for fibroblast-enriched pancreatic cancer.
Collapse
Affiliation(s)
- Quan Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Yongcun Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Xiangrui Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
| | - Youqing Shen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
122
|
Liu Y, Yuelling LW, Wang Y, Du F, Gordon RE, O'Brien JA, Ng JMY, Robins S, Lee EH, Liu H, Curran T, Yang ZJ. Astrocytes Promote Medulloblastoma Progression through Hedgehog Secretion. Cancer Res 2017; 77:6692-6703. [PMID: 28986380 DOI: 10.1158/0008-5472.can-17-1463] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/10/2017] [Accepted: 09/26/2017] [Indexed: 12/30/2022]
Abstract
Astrocytes, the most abundant type of glial cells in the brain, play critical roles in supporting neuronal development and brain function. Although astrocytes have been frequently detected in brain tumors, including medulloblastoma (MB), their functions in tumorigenesis are not clear. Here, we demonstrate that astrocytes are essential components of the MB tumor microenvironment. Tumor-associated astrocytes (TAA) secrete the ligand sonic hedgehog (Shh), which is required for maintaining MB cell proliferation despite the absence of its primary receptor Patched-1 (Ptch1). Shh drives expression of Nestin in MB cells through a smoothened-dependent, but Gli1-independent mechanism. Ablation of TAA dramatically suppresses Nestin expression and blocks tumor growth. These findings demonstrate an indispensable role for astrocytes in MB tumorigenesis and reveal a novel Ptch1-independent Shh pathway involved in MB progression. Cancer Res; 77(23); 6692-703. ©2017 AACR.
Collapse
Affiliation(s)
- Yongqiang Liu
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Larra W Yuelling
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Yuan Wang
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Fang Du
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Renata E Gordon
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Jenny A O'Brien
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Jessica M Y Ng
- Children's Research Institute, Children's Mercy Kansas City, Kansas City, Missouri
| | - Shannon Robins
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Eric H Lee
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Hailong Liu
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Tom Curran
- Children's Research Institute, Children's Mercy Kansas City, Kansas City, Missouri
| | - Zeng-Jie Yang
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania.
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
123
|
Pharmacological targeting of GLI1 inhibits proliferation, tumor emboli formation and in vivo tumor growth of inflammatory breast cancer cells. Cancer Lett 2017; 411:136-149. [PMID: 28965853 DOI: 10.1016/j.canlet.2017.09.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/15/2017] [Accepted: 09/21/2017] [Indexed: 01/01/2023]
Abstract
Activation of the Hedgehog (Hh) pathway effector GLI1 is linked to tumorigenesis and invasiveness in a number of cancers, with targeting of GLI1 by small molecule antagonists shown to be effective. We profiled a collection of GLI antagonists possessing distinct mechanisms of action for efficacy in phenotypic models of inflammatory and non-inflammatory breast cancer (IBC and non-IBC) that we showed expressed varying levels of Hh pathway mediators. Compounds GANT61, HPI-1, and JK184 decreased cell proliferation, inhibited GLI1 mRNA expression and decreased the number of colonies formed in TN-IBC (SUM149) and TNBC (MDA-MB-231 and SUM159) cell lines. In addition, GANT61 and JK184 significantly down-regulated GLI1 targets that regulate cell cycle (cyclin D and E) and apoptosis (Bcl2). GANT61 reduced SUM149 spheroid growth and emboli formation, and in orthotopic SUM149 tumor models significantly decreased tumor growth. We successfully utilized phenotypic profiling to identify a subset of GLI1 antagonists that were prioritized for testing in in vivo models. Our results indicated that GLI1 activation in TN-IBC as in TNBC, plays a vital role in promoting cell proliferation, motility, tumor growth, and formation of tumor emboli.
Collapse
|
124
|
Liu G, Huang W, Wang J, Liu X, Yang J, Zhang Y, Geng Y, Tan W, Zhang A. Discovery of Novel Macrocyclic Hedgehog Pathway Inhibitors Acting by Suppressing the Gli-Mediated Transcription. J Med Chem 2017; 60:8218-8245. [PMID: 28873303 DOI: 10.1021/acs.jmedchem.7b01185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A systemic medicinal chemistry campaign was conducted based on a literature hit compound 5 bearing the 4,5-dihydro-2H-benzo[b][1,5]oxazocin-6(3H)-one core through cyclization of two side substituents of the bicyclic skeleton combined with N-atom walking or ring walking and the central ring expansion or extraction approaches, leading to several series of structurally unique tricyclic compounds. Among these, compound 29a was identified as the most potent against the Hedgehog (Hh) signaling pathway showing an IC50 value of 23 nM. Mechanism studies indicated that compound 29a inhibited the Hh signaling pathway by suppressing the expression of the transcriptional factors Gli rather than by interrupting the binding of Gli with DNA. We further observed that 29a was equally potent against both Smo wild type and the two major resistant mutants (Smo D473H and Smo W535L). It potently inhibited the proliferation of medulloblastoma cells and showed significant tumor growth inhibition in the ptch± ;p53-/- medulloblastoma allograft mice model. Though more studies are needed to clarify the precise interaction pattern of 29a with Gli, its promising in vitro and in vivo properties encourage further profiling as a new-generation Hh signaling inhibitor to treat tumors primarily or secondarily resistant to current Smo inhibitors.
Collapse
Affiliation(s)
- Gang Liu
- CAS Key Laboratory of Receptor Research, and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM) , Shanghai 201203, China.,University of Chinese Academy of Sciences , Beijing 100049, China
| | - Wenjing Huang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Juan Wang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Xiaohua Liu
- CAS Key Laboratory of Receptor Research, and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM) , Shanghai 201203, China
| | - Jun Yang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Yong Geng
- CAS Key Laboratory of Receptor Research, and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM) , Shanghai 201203, China.,University of Chinese Academy of Sciences , Beijing 100049, China
| | - Wenfu Tan
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM) , Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, China.,University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
125
|
Kuzan-Fischer CM, Guerreiro Stucklin AS, Taylor MD. Advances in Genomics Explain Medulloblastoma Behavior at the Bedside. Neurosurgery 2017; 64:21-26. [PMID: 28899050 DOI: 10.1093/neuros/nyx248] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/18/2017] [Indexed: 12/27/2022] Open
Affiliation(s)
- Claudia M Kuzan-Fischer
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ana S Guerreiro Stucklin
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Surgery and the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
126
|
Bakshi A, Chaudhary SC, Rana M, Elmets CA, Athar M. Basal cell carcinoma pathogenesis and therapy involving hedgehog signaling and beyond. Mol Carcinog 2017; 56:2543-2557. [PMID: 28574612 DOI: 10.1002/mc.22690] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/23/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023]
Abstract
Basal cell carcinoma (BCC) of the skin is driven by aberrant hedgehog signaling. Thus blocking this signaling pathway by small molecules such as vismodegib inhibits tumor growth. Primary cilium in the epidermal cells plays an integral role in the processing of hedgehog signaling-related proteins. Recent genomic studies point to the involvement of additional genetic mutations that might be associated with the development of BCCs, suggesting significance of other signaling pathways, such as WNT, NOTCH, mTOR, and Hippo, aside from hedgehog in the pathogenesis of this human neoplasm. Some of these pathways could be regulated by noncoding microRNA. Altered microRNA expression profile is recognized with the progression of these lesions. Stopping treatment with Smoothened (SMO) inhibitors often leads to tumor reoccurrence in the patients with basal cell nevus syndrome, who develop 10-100 of BCCs. In addition, the initial effectiveness of these SMO inhibitors is impaired due to the onset of mutations in the drug-binding domain of SMO. These data point to a need to develop strategies to overcome tumor recurrence and resistance and to enhance efficacy by developing novel single agent-based or multiple agents-based combinatorial approaches. Immunotherapy and photodynamic therapy could be additional successful approaches particularly if developed in combination with chemotherapy for inoperable and metastatic BCCs.
Collapse
Affiliation(s)
- Anshika Bakshi
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama.,Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Sandeep C Chaudhary
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mehtab Rana
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Craig A Elmets
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
127
|
Han Y, Xiong Y, Shi X, Wu J, Zhao Y, Jiang J. Regulation of Gli ciliary localization and Hedgehog signaling by the PY-NLS/karyopherin-β2 nuclear import system. PLoS Biol 2017; 15:e2002063. [PMID: 28777795 PMCID: PMC5544186 DOI: 10.1371/journal.pbio.2002063] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022] Open
Abstract
Hedgehog (Hh) signaling in vertebrates depends on primary cilia. Upon stimulation, Hh pathway components, including Gli transcription factors, accumulate at primary cilia to transduce the Hh signal, but the mechanisms underlying their ciliary targeting remains largely unknown. Here, we show that the PY-type nuclear localization signal (PY-NLS)/karyopherinβ2 (Kapβ2) nuclear import system regulates Gli ciliary localization and Hh pathway activation. Mutating the PY-NLS in Gli or knockdown of Kapβ2 diminished Gli ciliary localization. Kapβ2 is required for the formation of Gli activator (GliA) in wild-type but not in Sufu mutant cells. Knockdown of Kapβ2 affected Hh signaling in zebrafish embryos, as well as in vitro cultured cerebellum granule neuron progenitors (CGNPs) and SmoM2-driven medulloblastoma cells. Furthermore, Kapβ2 depletion impaired the growth of cultured medulloblastoma cells, which was rescued by Gli overexpression. Interestingly, Kapβ2 is a transcriptional target of the Hh pathway, thus forming a positive feedback loop for Gli activation. Our study unravels the molecular mechanism and cellular machinery regulating Gli ciliary localization and identifies Kapβ2 as a critical regulator of the Hh pathway and a potential drug target for Hh-driven cancers. The secreted Hedgehog (Hh) protein plays an evolutionarily conserved role in both embryonic development and adult tissue homeostasis. Malfunction of Hh signaling activity contributes to a wide range of human diseases, including birth defects and cancer. Hh signaling in vertebrates critically depends on the primary cilium, a microtubule-based plasma membrane protrusion present on the surface of most mammalian cells. Upon ligand stimulation, Hh pathway components, including the seven-transmembrane protein Smoothened (Smo) and Gli transcription factors, accumulate at primary cilia to transduce the Hh signal, but the mechanisms underlying their ciliary targeting are still poorly understood. Here, we discover that the PY-type nuclear localization signal (PY-NLS) and the nuclear import factor karyopherinβ2 (Kapβ2) regulate Gli ciliary localization and Hh pathway activity. Mutating the PY-NLS in Gli or knockdown of Kapβ2 diminished Gli ciliary localization without affecting Smo ciliary accumulation in response to Hh. Kapβ2 regulates the formation of the active form of Gli, which is required for proper Hh signaling in zebrafish embryos and cultured cerebellum granule neuron progenitors (CGNPs). Kapβ2 depletion impaired the growth of medulloblastoma cells driven by an oncogenic form of Smo. Finally, Kapβ2 is a transcriptional target of the Hh pathway, forming a positive feedback loop to promote Gli activation. Our study reveals the molecular mechanism underlying the regulation of Gli ciliary targeting and identifies Kapβ2 as a potential cancer drug target.
Collapse
Affiliation(s)
- Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Yue Xiong
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institute of Life Sciences, CAS, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xuanming Shi
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Jiang Wu
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Yun Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institute of Life Sciences, CAS, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- * E-mail: (JJ); (YZ)
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- * E-mail: (JJ); (YZ)
| |
Collapse
|
128
|
Mpekris F, Papageorgis P, Polydorou C, Voutouri C, Kalli M, Pirentis AP, Stylianopoulos T. Sonic-hedgehog pathway inhibition normalizes desmoplastic tumor microenvironment to improve chemo- and nanotherapy. J Control Release 2017; 261:105-112. [PMID: 28662901 DOI: 10.1016/j.jconrel.2017.06.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/21/2017] [Accepted: 06/24/2017] [Indexed: 01/08/2023]
Abstract
Targeting the rich extracellular matrix of desmoplastic tumors has been successfully shown to normalize collagen and hyaluronan levels and re-engineer intratumoral mechanical forces, improving tumor perfusion and chemotherapy. As far as targeting the abundant cancer-associated fibroblasts (CAFs) in desmoplastic tumors is concerned, while both pharmacologic inhibition of the sonic-hedgehog pathway and genetic depletion of fibroblasts have been employed in pancreatic cancers, the results between the two methods have been contradictory. In this study, we employed vismodegib to inhibit the sonic-hedgehog pathway with the aim to i) elucidate the mechanism of how CAFs depletion improves drug delivery, ii) extent and evaluate the potential use of sonic-hedgehog inhibitors to breast cancers, and iii) investigate whether sonic-hedgehog inhibition improves not only chemotherapy, but also the efficacy of the most commonly used breast cancer nanomedicines, namely Abraxane® and Doxil®. We found that treatment with vismodegib normalizes the tumor microenvironment by reducing the proliferative CAFs and in cases the levels of collagen and hyaluronan. These modulations re-engineered the solid and fluid stresses in the tumors, improving blood vessel functionality. As a result, the delivery and efficacy of chemotherapy was improved in two models of pancreatic cancer. Additionally, vismodegib treatment significantly improved the efficacy of both Abraxane and Doxil in xenograft breast tumors. Our results suggest the use of vismodegib, and sonic hedgehog inhibitors in general, to enhance cancer chemo- and nanotherapy.
Collapse
Affiliation(s)
- Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Panagiotis Papageorgis
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus; Department of Life Sciences, Program in Biological Sciences, European University Cyprus, Nicosia, Cyprus
| | - Christiana Polydorou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Maria Kalli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Athanassios P Pirentis
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
129
|
Ye L, Ding K, Zhao F, Liu X, Wu Y, Liu Y, Xue D, Zhou F, Zhang X, Stevens RC, Xu F, Zhao S, Tao H. A structurally guided dissection-then-evolution strategy for ligand optimization of smoothened receptor. MEDCHEMCOMM 2017; 8:1332-1336. [PMID: 30108845 PMCID: PMC6072208 DOI: 10.1039/c7md00104e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/20/2017] [Indexed: 01/18/2023]
Abstract
We present herein a novel dissection-then-evolution strategy for ligand optimization. Using the co-crystal structure of the smoothened receptor (SMO) as a guide, we studied the modular contribution of LY2940680 by systematically "silencing" the specific interaction between the individual residue(s) and the fragment in the ligand. Following evolution by focusing on the benzoyl part finally yielded an improved ligand 21.
Collapse
Affiliation(s)
- Lintao Ye
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 555 Zuchongzhi Road, Building 3, Room 426 , Shanghai , 201203 , China
- University of Chinese Academy of Sciences , No. 19A, Yuquan Road , Beijing 100049 , China
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Kang Ding
- Key Laboratory of Computational Biology , CAS-MPG Partner Institute for Computational Biology , Shanghai Institutes for Biological Sciences , Chinese Academy of Sciences , Shanghai , 200031 , China
- University of Chinese Academy of Sciences , No. 19A, Yuquan Road , Beijing 100049 , China
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Fei Zhao
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
| | - Xiaoyan Liu
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
| | - Yiran Wu
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
| | - Yang Liu
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
| | - Dongxiang Xue
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 555 Zuchongzhi Road, Building 3, Room 426 , Shanghai , 201203 , China
- University of Chinese Academy of Sciences , No. 19A, Yuquan Road , Beijing 100049 , China
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Fang Zhou
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 555 Zuchongzhi Road, Building 3, Room 426 , Shanghai , 201203 , China
- University of Chinese Academy of Sciences , No. 19A, Yuquan Road , Beijing 100049 , China
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Xianjun Zhang
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- Institute of Biochemistry and Cell Biology , Shanghai Institutes for Biological Sciences , Chinese Academy of Sciences , Shanghai , 200031 , China
- University of Chinese Academy of Sciences , No. 19A, Yuquan Road , Beijing 100049 , China
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Raymond C Stevens
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Fei Xu
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Suwen Zhao
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
- School of Life Science and Technology , ShanghaiTech University , Shanghai 201210 , China
| | - Houchao Tao
- iHuman Institute , ShanghaiTech University , 2F Building 6, 99 Haike Road, Pudong New District , Shanghai 201210 , China . ;
| |
Collapse
|
130
|
Design, Synthesis and Biological Evaluation of novel Hedgehog Inhibitors for treating Pancreatic Cancer. Sci Rep 2017; 7:1665. [PMID: 28490735 PMCID: PMC5431907 DOI: 10.1038/s41598-017-01942-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
Hedgehog (Hh) pathway is involved in epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) maintenance resulting in tumor progression. GDC-0449, an inhibitor of Hh pathway component smoothened (Smo) has shown promise in the treatment of various cancers including pancreatic cancer. However, the emergence of resistance during GDC-0449 treatment with numerous side effects limits its use. Therefore, here we report the design, synthesis and evaluation of novel GDC-0449 analogs using N-[3-(2-pyridinyl) phenyl] benzamide scaffold. Cell-based screening followed by molecular simulation revealed 2-chloro-N1-[4-chloro-3-(2-pyridinyl)phenyl]-N4,N4-bis(2-pyridinylmethyl)-1,4-benzenedicarboxamide (MDB5) as most potent analog, binding with an extra interactions in seven-transmembrane (7-TM) domain of Smo due to an additional 2-pyridylmethyl group than GDC-0449. Moreover, MDB5 was more efficient in inhibiting Hh pathway components as measured by Gli-1 and Shh at transcriptional and translational levels. Additionally, a significant reduction of ALDH1, CD44 and Oct-3/4, key markers of pancreatic CSC was observed when MIA PaCa-2 cells were treated with MDB5 compared to GDC-0449. In a pancreatic tumor mouse model, MDB5 containing nanoparticles treated group showed significant inhibition of tumor growth without loss in body weight. These evidence highlight the enhanced Hh pathway inhibition and anticancer properties of MDB5 leaving a platform for mono and/or combination therapy.
Collapse
|
131
|
Ingallina C, Costa PM, Ghirga F, Klippstein R, Wang JT, Berardozzi S, Hodgins N, Infante P, Pollard SM, Botta B, Al-Jamal KT. Polymeric glabrescione B nanocapsules for passive targeting of Hedgehog-dependent tumor therapy in vitro. Nanomedicine (Lond) 2017; 12:711-728. [PMID: 28322108 PMCID: PMC5986025 DOI: 10.2217/nnm-2016-0388] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/26/2017] [Indexed: 01/09/2023] Open
Abstract
AIM With the purpose of delivering high doses of glabrescione B (GlaB) to solid tumors after systemic administration, long-circulating GlaB-loaded oil-cored polymeric nanocapsules (NC-GlaB) were formulated. MATERIALS & METHODS Synthesis of GlaB and its encapsulation in nanocapsules (NCs) was performed. Empty and GlaB-loaded NCs were assessed for their physico-chemical properties, in vitro cytotoxicity and in vivo biodistribution. RESULTS GlaB was efficiently loaded into NCs (∽90%), which were small (∽160 nm), homogeneous and stable upon storage. Further, GlaB and NC-GlaB demonstrated specific activities against the cancer stem cells. Preliminary studies in tumor-bearing mice supported the ability of NC to accumulate in pancreatic tumors. CONCLUSION This study provides early evidence that NC-GlaB has the potential to be utilized in a preclinical setting and justifies the need to perform therapeutic experiments in mice.
Collapse
Affiliation(s)
- Cinzia Ingallina
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
| | - Pedro M Costa
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Francesca Ghirga
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Rebecca Klippstein
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Julie T Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Simone Berardozzi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Naomi Hodgins
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Paola Infante
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Steven M Pollard
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| |
Collapse
|
132
|
Liu KW, Pajtler KW, Worst BC, Pfister SM, Wechsler-Reya RJ. Molecular mechanisms and therapeutic targets in pediatric brain tumors. Sci Signal 2017; 10:10/470/eaaf7593. [PMID: 28292958 DOI: 10.1126/scisignal.aaf7593] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Brain tumors are among the leading causes of cancer-related deaths in children. Although surgery, aggressive radiation, and chemotherapy have improved outcomes, many patients still die of their disease. Moreover, those who survive often suffer devastating long-term side effects from the therapies. A greater understanding of the molecular underpinnings of these diseases will drive the development of new therapeutic approaches. Advances in genomics and epigenomics have provided unprecedented insight into the molecular diversity of these diseases and, in several cases, have revealed key genes and signaling pathways that drive tumor growth. These not only serve as potential therapeutic targets but also have facilitated the creation of animal models that faithfully recapitulate the human disease for preclinical studies. In this Review, we discuss recent progress in understanding the molecular basis of the three most common malignant pediatric brain tumors-medulloblastoma, ependymoma, and high-grade glioma-and the implications for development of safer and more effective therapies.
Collapse
Affiliation(s)
- Kun-Wei Liu
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Kristian W Pajtler
- Division of Pediatric Neurooncology, German Cancer Research Centre (Deutsches Krebsforschungszentrum, DKFZ) and Heidelberg University Hospital, D-69120 Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University Hospital, D-69120 Heidelberg, Germany.,German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), Core Center Heidelberg, D-69120 Heidelberg, Germany
| | - Barbara C Worst
- Division of Pediatric Neurooncology, German Cancer Research Centre (Deutsches Krebsforschungszentrum, DKFZ) and Heidelberg University Hospital, D-69120 Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University Hospital, D-69120 Heidelberg, Germany.,German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), Core Center Heidelberg, D-69120 Heidelberg, Germany
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Centre (Deutsches Krebsforschungszentrum, DKFZ) and Heidelberg University Hospital, D-69120 Heidelberg, Germany. .,Department of Pediatric Oncology, Hematology and Immunology, University Hospital, D-69120 Heidelberg, Germany.,German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), Core Center Heidelberg, D-69120 Heidelberg, Germany
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
133
|
Wu F, Zhang Y, Sun B, McMahon AP, Wang Y. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem Biol 2017; 24:252-280. [PMID: 28286127 DOI: 10.1016/j.chembiol.2017.02.010] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/29/2016] [Accepted: 02/10/2017] [Indexed: 02/07/2023]
Abstract
The Hedgehog (HH) signaling pathway was discovered originally as a key pathway in embryonic patterning and development. Since its discovery, it has become increasingly clear that the HH pathway also plays important roles in a multitude of cancers. Therefore, HH signaling has emerged as a therapeutic target of interest for cancer therapy. In this review, we provide a brief overview of HH signaling and the key molecular players involved and offer an up-to-date summary of our current knowledge of endogenous and exogenous small molecules that modulate HH signaling. We discuss experiences and lessons learned from the decades-long efforts toward the development of cancer therapies targeting the HH pathway. Challenges to develop next-generation cancer therapies are highlighted.
Collapse
Affiliation(s)
- Fujia Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
134
|
Alfonsi R, Botta B, Cacchi S, Di Marcotullio L, Fabrizi G, Faedda R, Goggiamani A, Iazzetti A, Mori M. Design, Palladium-Catalyzed Synthesis, and Biological Investigation of 2-Substituted 3-Aroylquinolin-4(1H)-ones as Inhibitors of the Hedgehog Signaling Pathway. J Med Chem 2017; 60:1469-1477. [DOI: 10.1021/acs.jmedchem.6b01135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Romina Alfonsi
- Department of Molecular
Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Sandro Cacchi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular
Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
- Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Giancarlo Fabrizi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Roberta Faedda
- Department of Molecular
Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Antonella Goggiamani
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Antonia Iazzetti
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Mattia Mori
- Center for Life Nano
Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
135
|
Wahid M, Jawed A, Dar SA, Mandal RK, Haque S. Differential pharmacology and clinical utility of sonidegib in advanced basal cell carcinoma. Onco Targets Ther 2017; 10:515-520. [PMID: 28182134 PMCID: PMC5279825 DOI: 10.2147/ott.s97713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Patients suffering from advanced basal cell carcinoma (BCC) have very limited treatment options. Sonidegib selectively inhibits the growth of Hedgehog pathway-dependent tumors and can treat locally advanced BCC patients who are not candidates for surgery or radiation therapy. The BOLT clinical trials were conducted to evaluate the efficacy/potency of sonidegib in the treatment of advanced BCC or metastatic BCC. The patients were randomized in 1:2 ratios to receive 200 or 800 mg oral sonidegib daily, stratified by disease, histological subtype and geographical region. The primary efficacy analyses showed that 18 patients in the 200 mg group and 35 patients in the 800 mg group show an objective response (Central Review Committee) that corresponds to 43% (95% confidence interval [CI]: 28–59) and 38% (95% CI: 28–48) in their respective categories. Disease control was found in 93% (39 patients) and 80% (74 patients) of the patients administered 200 and 800 mg sonidegib, respectively. The adverse events were assessed by the Central Review Committee as well as the investigator review team as per the guidelines of National Cancer Institute Common Terminology Criteria for Adverse Events version 4.03. The most frequently found adverse events reported in BOLT trials were muscle spasms, alopecia, dysgeusia (taste disturbance), nausea, elevated blood creatine kinase and fatigue. Comparatively, the patients administered 200 mg sonidegib showed fewer adverse events than those in the 800 mg sonidegib category. Thus, the benefit of using the 200 mg dose of sonidegib outweighs the associated risks and it can be inferred that it would be judicious to choose doses of lesser strength.
Collapse
Affiliation(s)
- Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Sajad Ahmad Dar
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Raju K Mandal
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
136
|
Benvenuto M, Masuelli L, De Smaele E, Fantini M, Mattera R, Cucchi D, Bonanno E, Di Stefano E, Frajese GV, Orlandi A, Screpanti I, Gulino A, Modesti A, Bei R. In vitro and in vivo inhibition of breast cancer cell growth by targeting the Hedgehog/GLI pathway with SMO (GDC-0449) or GLI (GANT-61) inhibitors. Oncotarget 2017; 7:9250-70. [PMID: 26843616 PMCID: PMC4891038 DOI: 10.18632/oncotarget.7062] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/18/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant Hedgehog (Hh)/glioma-associated oncogene (GLI) signaling has been implicated in cancer progression. Here, we analyzed GLI1, Sonic Hedgehog (Shh) and NF-κB expression in 51 breast cancer (ductal carcinoma) tissues using immunohistochemistry. We found a positive correlation between nuclear GLI1 expression and tumor grade in ductal carcinoma cases. Cytoplasmic Shh staining significantly correlated with a lower tumor grade. Next, the in vitro effects of two Hh signaling pathway inhibitors on breast cancer cell lines were evaluated using the Smoothened (SMO) antagonist GDC-0449 and the direct GLI1 inhibitor GANT-61. GDC-0449 and GANT-61 exhibited the following effects: a) inhibited breast cancer cell survival; b) induced apoptosis; c) inhibited Hh pathway activity by decreasing the mRNA expression levels of GLI1 and Ptch and inhibiting the nuclear translocation of GLI1; d) increased/decreased EGFR and ErbB2 protein expression, reduced p21-Ras and ERK1/ERK2 MAPK activities and inhibited AKT activation; and e) decreased the nuclear translocation of NF-κB. However, GANT-61 exerted these effects more effectively than GDC-0449. The in vivo antitumor activities of GDC-0449 and GANT-61 were analyzed in BALB/c mice that were subcutaneously inoculated with mouse breast cancer (TUBO) cells. GDC-0449 and GANT-61 suppressed tumor growth of TUBO cells in BALB/c mice to different extents. These findings suggest that targeting the Hh pathway using antagonists that act downstream of SMO is a more efficient strategy than using antagonists that act upstream of SMO for interrupting Hh signaling in breast cancer.
Collapse
Affiliation(s)
- Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Massimo Fantini
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Rosanna Mattera
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy.,Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | - Danilo Cucchi
- Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | - Elena Bonanno
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Enrica Di Stefano
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Giovanni Vanni Frajese
- Department of Physical Education, Human Sciences and Health, University of Rome "Foro Italico", Rome, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | - Alberto Gulino
- Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
137
|
Trinh TN, McLaughlin EA, Gordon CP, Bernstein IR, Pye VJ, Redgrove KA, McCluskey A. Small molecule Hedgehog pathway antagonists. Org Biomol Chem 2017; 15:3046-3059. [DOI: 10.1039/c6ob01959e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Leveraging our quinolone-1-(2H)-one based Hedgehog signalling pathway (HSP) inhibitors we have developed two new classes of HSP inhibitors based on: l-tryptophan and benzo[1,3]dioxol-5-ylmethyl-[2-(1H-indol-3-yl)-ethyl]-amine.
Collapse
Affiliation(s)
- Trieu N. Trinh
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Eileen A. McLaughlin
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Christopher P. Gordon
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
- Nanoscale Organization and Dynamics Group
| | - Ilana R. Bernstein
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Victoria J. Pye
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Kate A. Redgrove
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Adam McCluskey
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| |
Collapse
|
138
|
Dash RC, Maschinot CR, Hadden MK. A molecular dynamics approach to identify an oxysterol-based hedgehog pathway inhibitor. Biochim Biophys Acta Gen Subj 2016; 1861:168-177. [PMID: 27825830 DOI: 10.1016/j.bbagen.2016.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/26/2016] [Accepted: 11/03/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Multiple oxysterols (OHCs) have demonstrated the ability to act as agonists or antagonists of the hedgehog (Hh) signaling pathway, a developmental signaling pathway that has been implicated as a potential therapeutic target in a variety of human diseases. These OHCs are known to modulate Hh signaling through direct binding interactions with the N-terminal cysteine rich domain (CRD) of Smoothened, a key regulator of Hh signal transduction. METHODS Homology modeling, molecular dynamics simulations, and MM/GBSA energy calculations were utilized to explore binding interactions between the OHC scaffold and the human Smoothened CRD. Follow-up cellular assays explored the in vitro activity of potential Hh pathway modulators. RESULTS Structural features that govern key molecular interactions between the Smoothened CRD and the OHC scaffold were identified. Orientation of the iso-octyl side chain as well as the overall entropy of the OHC-CRD complex are important for determining activity against the Hh pathway. OHC 9, which was previously thought to be inactive because it was not an Hh agonist, was identified as an inhibitor of Hh signal transmission. CONCLUSIONS Calculated MM/GBSA binding energies for OHCs in complex with the CRD of Smoothened correlate well with in vitro Hh modulatory activity. Compounds with high affinity stabilize Smoothened and are antagonists, whereas compounds with reduced affinity allow a conformational change in Smoothened that results in pathway activation. GENERAL SIGNIFICANCE Computational modeling and molecular dynamics simulations can be used to predict whether a small molecule that binds the Smoothened CRD will be an agonist or antagonist of the pathway.
Collapse
Affiliation(s)
- Radha Charan Dash
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd., Unit 3092, Storrs, CT, 06269-3092, United States
| | - Chad R Maschinot
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd., Unit 3092, Storrs, CT, 06269-3092, United States
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd., Unit 3092, Storrs, CT, 06269-3092, United States.
| |
Collapse
|
139
|
Gracia-Cazaña T, Salazar N, Zamarrón A, Mascaraque M, Lucena S, Juarranz Á. Resistance of Nonmelanoma Skin Cancer to Nonsurgical Treatments. Part II: Photodynamic Therapy, Vismodegib, Cetuximab, Intralesional Methotrexate, and Radiotherapy. ACTAS DERMO-SIFILIOGRAFICAS 2016. [DOI: 10.1016/j.adengl.2016.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
140
|
Liu G, Xue D, Yang J, Wang J, Liu X, Huang W, Li J, Long YQ, Tan W, Zhang A. Design, Synthesis, and Pharmacological Evaluation of 2-(2,5-Dimethyl-5,6,7,8-tetrahydroquinolin-8-yl)-N-aryl Propanamides as Novel Smoothened (Smo) Antagonists. J Med Chem 2016; 59:11050-11068. [PMID: 27736063 DOI: 10.1021/acs.jmedchem.6b01247] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A series of novel Smo antagonists were developed either by directly incorporating the basic skeleton of the natural product artemisinin or by first breaking artemisinin into structurally simpler and stable intermediates and then reconstructing into diversified heterocyclic derivatives, equipped with a Smo-targeting bullet. 2-(2,5-Dimethyl-5,6,7,8-tetrahydroquinolin-8-yl)-N-arylpropanamide 65 was identified as the most potent, with an IC50 value of 9.53 nM against the Hh signaling pathway. Complementary mechanism studies confirmed that 65 inhibits Hh signaling pathway by targeting Smo and shares the same binding site as that of the tool drug cyclopamine. Meanwhile, 65 has a good plasma exposure and an acceptable oral bioavailability. Dose-dependent antiproliferative effects were observed in ptch+/-;p53-/- medulloblastoma cells, and significant tumor growth inhibitions were achieved for 65 in the ptch+/-;p53-/- medulloblastoma allograft model.
Collapse
Affiliation(s)
- Gang Liu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), University of Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Shanghai 201203, China
| | - Ding Xue
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), University of Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Shanghai 201203, China
| | - Jun Yang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Juan Wang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Xiaohua Liu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), University of Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Shanghai 201203, China
| | - Wenjing Huang
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Jie Li
- School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, China
| | - Ya-Qiu Long
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), University of Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Shanghai 201203, China
| | - Wenfu Tan
- Department of Pharmacology, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), University of Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, China
| |
Collapse
|
141
|
Luchetti G, Sircar R, Kong JH, Nachtergaele S, Sagner A, Byrne EFX, Covey DF, Siebold C, Rohatgi R. Cholesterol activates the G-protein coupled receptor Smoothened to promote Hedgehog signaling. eLife 2016; 5:e20304. [PMID: 27705744 PMCID: PMC5123864 DOI: 10.7554/elife.20304] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/03/2016] [Indexed: 12/12/2022] Open
Abstract
Cholesterol is necessary for the function of many G-protein coupled receptors (GPCRs). We find that cholesterol is not just necessary but also sufficient to activate signaling by the Hedgehog (Hh) pathway, a prominent cell-cell communication system in development. Cholesterol influences Hh signaling by directly activating Smoothened (SMO), an orphan GPCR that transmits the Hh signal across the membrane in all animals. Unlike many GPCRs, which are regulated by cholesterol through their heptahelical transmembrane domains, SMO is activated by cholesterol through its extracellular cysteine-rich domain (CRD). Residues shown to mediate cholesterol binding to the CRD in a recent structural analysis also dictate SMO activation, both in response to cholesterol and to native Hh ligands. Our results show that cholesterol can initiate signaling from the cell surface by engaging the extracellular domain of a GPCR and suggest that SMO activity may be regulated by local changes in cholesterol abundance or accessibility.
Collapse
Affiliation(s)
- Giovanni Luchetti
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, United States
| | - Ria Sircar
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, United States
| | - Jennifer H Kong
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, United States
| | - Sigrid Nachtergaele
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, United States
| | - Andreas Sagner
- Mill Hill Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Eamon FX Byrne
- Division of Structural Biology, University of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Christian Siebold
- Division of Structural Biology, University of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Rajat Rohatgi
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
142
|
Chen J, Lv H, Hu J, Ji M, Xue N, Li C, Ma S, Zhou Q, Lin B, Li Y, Yu S, Chen X. CAT3, a novel agent for medulloblastoma and glioblastoma treatment, inhibits tumor growth by disrupting the Hedgehog signaling pathway. Cancer Lett 2016; 381:391-403. [DOI: 10.1016/j.canlet.2016.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/31/2016] [Accepted: 07/21/2016] [Indexed: 01/20/2023]
|
143
|
Infante P, Alfonsi R, Ingallina C, Quaglio D, Ghirga F, D'Acquarica I, Bernardi F, Di Magno L, Canettieri G, Screpanti I, Gulino A, Botta B, Mori M, Di Marcotullio L. Inhibition of Hedgehog-dependent tumors and cancer stem cells by a newly identified naturally occurring chemotype. Cell Death Dis 2016; 7:e2376. [PMID: 27899820 PMCID: PMC5059851 DOI: 10.1038/cddis.2016.195] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/24/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Hedgehog (Hh) inhibitors have emerged as valid tools in the treatment of a wide range of cancers. Indeed, aberrant activation of the Hh pathway occurring either by ligand-dependent or -independent mechanisms is a key driver in tumorigenesis. The smoothened (Smo) receptor is one of the main upstream transducers of the Hh signaling and is a validated target for the development of anticancer compounds, as underlined by the FDA-approved Smo antagonist Vismodegib (GDC-0449/Erivedge) for the treatment of basal cell carcinoma. However, Smo mutations that confer constitutive activity and drug resistance have emerged during treatment with Vismodegib. For this reason, the development of new effective Hh inhibitors represents a major challenge for cancer therapy. Natural products have always represented a unique source of lead structures in drug discovery, and in recent years have been used to modulate the Hh pathway at multiple levels. Here, starting from an in house library of natural compounds and their derivatives, we discovered novel chemotypes of Hh inhibitors by mean of virtual screening against the crystallographic structure of Smo. Hh functional based assay identified the chalcone derivative 12 as the most effective Hh inhibitor within the test set. The chalcone 12 binds the Smo receptor and promotes the displacement of Bodipy-Cyclopamine in both Smo WT and drug-resistant Smo mutant. Our molecule stands as a promising Smo antagonist able to specifically impair the growth of Hh-dependent tumor cells in vitro and in vivo and medulloblastoma stem-like cells and potentially overcome the associated drug resistance.
Collapse
Affiliation(s)
- Paola Infante
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Romina Alfonsi
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Cinzia Ingallina
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Deborah Quaglio
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, Italy
| | - Francesca Ghirga
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Ilaria D'Acquarica
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, Italy
| | - Flavia Bernardi
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Laura Di Magno
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Alberto Gulino
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, Italy
| | - Mattia Mori
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| |
Collapse
|
144
|
Liu X, Pitarresi JR, Cuitiño MC, Kladney RD, Woelke SA, Sizemore GM, Nayak SG, Egriboz O, Schweickert PG, Yu L, Trela S, Schilling DJ, Halloran SK, Li M, Dutta S, Fernandez SA, Rosol TJ, Lesinski GB, Shakya R, Ludwig T, Konieczny SF, Leone G, Wu J, Ostrowski MC. Genetic ablation of Smoothened in pancreatic fibroblasts increases acinar-ductal metaplasia. Genes Dev 2016; 30:1943-55. [PMID: 27633013 PMCID: PMC5066238 DOI: 10.1101/gad.283499.116] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/08/2016] [Indexed: 12/17/2022]
Abstract
Liu et al. show that disruption of paracrine Hedgehog signaling via genetic ablation of Smoothened (Smo) in stromal fibroblasts in a KrasG12D mouse model increased acinar-to-ductal metaplasia (ADM). Smo-deleted fibroblasts had higher expression of transforming growth factor-α (Tgfα) mRNA and secreted higher levels of TGFα, leading to activation of EGFR signaling in acinar cells and increased ADM. The contribution of the microenvironment to pancreatic acinar-to-ductal metaplasia (ADM), a preneoplastic transition in oncogenic Kras-driven pancreatic cancer progression, is currently unclear. Here we show that disruption of paracrine Hedgehog signaling via genetic ablation of Smoothened (Smo) in stromal fibroblasts in a KrasG12D mouse model increased ADM. Smo-deleted fibroblasts had higher expression of transforming growth factor-α (Tgfa) mRNA and secreted higher levels of TGFα, leading to activation of EGFR signaling in acinar cells and increased ADM. The mechanism involved activation of AKT and noncanonical activation of the GLI family transcription factor GLI2. GLI2 was phosphorylated at Ser230 in an AKT-dependent fashion and directly regulated Tgfa expression in fibroblasts lacking Smo. Additionally, Smo-deleted fibroblasts stimulated the growth of KrasG12D/Tp53R172H pancreatic tumor cells in vivo and in vitro. These results define a non-cell-autonomous mechanism modulating KrasG12D-driven ADM that is balanced by cross-talk between Hedgehog/SMO and AKT/GLI2 pathways in stromal fibroblasts.
Collapse
Affiliation(s)
- Xin Liu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Jason R Pitarresi
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Maria C Cuitiño
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Raleigh D Kladney
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Sarah A Woelke
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Gina M Sizemore
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Sunayana G Nayak
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Onur Egriboz
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Patrick G Schweickert
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA; the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA; the Bindley Bioscience Center, Purdue University, West Lafayette, Indiana 47907, USA
| | - Lianbo Yu
- Department of Biomedical Informatics' Center for Biostatistics, The Ohio State University, Columbus, Ohio 43210, USA
| | - Stefan Trela
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Daniel J Schilling
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Shannon K Halloran
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Maokun Li
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Shourik Dutta
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Soledad A Fernandez
- Department of Biomedical Informatics' Center for Biostatistics, The Ohio State University, Columbus, Ohio 43210, USA
| | - Thomas J Rosol
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio 43210, USA
| | - Gregory B Lesinski
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Department of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Reena Shakya
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Thomas Ludwig
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Stephen F Konieczny
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA; the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA; the Bindley Bioscience Center, Purdue University, West Lafayette, Indiana 47907, USA
| | - Gustavo Leone
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Jinghai Wu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| | - Michael C Ostrowski
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA; Cancer Biology and Genetics Department, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
145
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
146
|
Huang P, Nedelcu D, Watanabe M, Jao C, Kim Y, Liu J, Salic A. Cellular Cholesterol Directly Activates Smoothened in Hedgehog Signaling. Cell 2016; 166:1176-1187.e14. [PMID: 27545348 DOI: 10.1016/j.cell.2016.08.003] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/14/2016] [Accepted: 07/30/2016] [Indexed: 12/28/2022]
Abstract
In vertebrates, sterols are necessary for Hedgehog signaling, a pathway critical in embryogenesis and cancer. Sterols activate the membrane protein Smoothened by binding its extracellular, cysteine-rich domain (CRD). Major unanswered questions concern the nature of the endogenous, activating sterol and the mechanism by which it regulates Smoothened. We report crystal structures of CRD complexed with sterols and alone, revealing that sterols induce a dramatic conformational change of the binding site, which is sufficient for Smoothened activation and is unique among CRD-containing receptors. We demonstrate that Hedgehog signaling requires sterol binding to Smoothened and define key residues for sterol recognition and activity. We also show that cholesterol itself binds and activates Smoothened. Furthermore, the effect of oxysterols is abolished in Smoothened mutants that retain activation by cholesterol and Hedgehog. We propose that the endogenous Smoothened activator is cholesterol, not oxysterols, and that vertebrate Hedgehog signaling controls Smoothened by regulating its access to cholesterol.
Collapse
Affiliation(s)
- Pengxiang Huang
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Daniel Nedelcu
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Miyako Watanabe
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Cindy Jao
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Youngchang Kim
- Structural Biology Center, Biosciences Division, Argonne National Laboratory, Argonne, Illinois 60439, USA
| | - Jing Liu
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
147
|
Lacroix C, Fish I, Torosyan H, Parathaman P, Irwin JJ, Shoichet BK, Angers S. Identification of Novel Smoothened Ligands Using Structure-Based Docking. PLoS One 2016; 11:e0160365. [PMID: 27490099 PMCID: PMC4973902 DOI: 10.1371/journal.pone.0160365] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022] Open
Abstract
The seven transmembrane protein Smoothened is required for Hedgehog signaling during embryonic development and adult tissue homeostasis. Inappropriate activation of the Hedgehog signalling pathway leads to cancers such as basal cell carcinoma and medulloblastoma, and Smoothened inhibitors are now available clinically to treat these diseases. However, resistance to these inhibitors rapidly develops thereby limiting their efficacy. The determination of Smoothened crystal structures enables structure-based discovery of new ligands with new chemotypes that will be critical to combat resistance. In this study, we docked 3.2 million available, lead-like molecules against Smoothened, looking for those with high physical complementarity to its structure; this represents the first such campaign against the class Frizzled G-protein coupled receptor family. Twenty-one high-ranking compounds were selected for experimental testing, and four, representing three different chemotypes, were identified to antagonize Smoothened with IC50 values better than 50 μM. A screen for analogs revealed another six molecules, with IC50 values in the low micromolar range. Importantly, one of the most active of the new antagonists continued to be efficacious at the D473H mutant of Smoothened, which confers clinical resistance to the antagonist vismodegib in cancer treatment.
Collapse
Affiliation(s)
- Celine Lacroix
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Inbar Fish
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
| | - Hayarpi Torosyan
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Pranavan Parathaman
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (BS); (SA)
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (BS); (SA)
| |
Collapse
|
148
|
Gracia-Cazaña T, Salazar N, Zamarrón A, Mascaraque M, Lucena SR, Juarranz Á. Resistance of Nonmelanoma Skin Cancer to Nonsurgical Treatments. Part II: Photodynamic Therapy, Vismodegib, Cetuximab, Intralesional Methotrexate, and Radiotherapy. ACTAS DERMO-SIFILIOGRAFICAS 2016; 107:740-750. [PMID: 27436804 DOI: 10.1016/j.ad.2016.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 04/22/2016] [Accepted: 04/30/2016] [Indexed: 12/18/2022] Open
Abstract
A wide range of treatments is now available for nonmelanoma skin cancer, including 5-fluorouracil, ingenol mebutate, imiquimod, diclofenac, photodynamic therapy, methotrexate, cetuximab, vismodegib, and radiotherapy. All are associated with high clinical and histologic response rates. However, some tumors do not respond due to resistance, which may be primary or acquired. Study of the resistance processes is a broad area of research that aims to increase our understanding of the nature of each tumor and the biologic features that make it resistant, as well as to facilitate the design of new therapies directed against these tumors. In this second article, having covered the topical treatments of nonmelanoma skin cancer, we review resistance to other nonsurgical treatments, such as monoclonal antibodies against basal and squamous cell carcinomas, intralesional chemotherapy, photodynamic therapy, and radiotherapy.
Collapse
Affiliation(s)
- T Gracia-Cazaña
- Unidad de Dermatología, Hospital de Barbastro, Barbastro, Huesca, España; Instituto Aragonés de Ciencias de la Salud, Zaragoza, España.
| | - N Salazar
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, España
| | - A Zamarrón
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, España
| | - M Mascaraque
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, España
| | - S R Lucena
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, España
| | - Á Juarranz
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, España
| |
Collapse
|
149
|
Maschinot CA, Pace JR, Hadden MK. Synthetic Small Molecule Inhibitors of Hh Signaling As Anti-Cancer Chemotherapeutics. Curr Med Chem 2016; 22:4033-57. [PMID: 26310919 DOI: 10.2174/0929867322666150827093904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 12/11/2022]
Abstract
The hedgehog (Hh) pathway is a developmental signaling pathway that is essential to the proper embryonic development of many vertebrate systems. Dysregulation of Hh signaling has been implicated as a causative factor in the development and progression of several forms of human cancer. As such, the development of small molecule inhibitors of Hh signaling as potential anti-cancer chemotherapeutics has been a major area of research interest in both academics and industry over the past ten years. Through these efforts, synthetic small molecules that target multiple components of the Hh pathway have been identified and advanced to preclinical or clinical development. The goal of this review is to provide an update on the current status of several synthetic small molecule Hh pathway inhibitors and explore the potential of several recently disclosed inhibitory scaffolds.
Collapse
Affiliation(s)
| | | | - M K Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA.
| |
Collapse
|
150
|
Gonnissen A, Isebaert S, Haustermans K. Targeting the Hedgehog signaling pathway in cancer: beyond Smoothened. Oncotarget 2016; 6:13899-913. [PMID: 26053182 PMCID: PMC4546439 DOI: 10.18632/oncotarget.4224] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
An essential role for Hedgehog (Hh) signaling in human cancer has been established beyond doubt. At present, targeting Hh signaling has mainly been investigated with SMO inhibitors. Unfortunately, resistance against currently used SMO inhibitors has already been observed in basal cell carcinoma (BCC) patients. Therefore, the use of Hh inhibitors targeting the signaling cascade more downstream of SMO could represent a more promising strategy. Furthermore, besides the classical canonical way of Hh signaling activation, non-canonical activation of the GLI transcription factors by multiple important signaling pathways (e.g. MAPK, PI3K, TGFβ) has also been described, pinpointing the importance of targeting the transcription factors GLI1/2. The most promising agent in this context is probably the GLI1/2 inhibitor GANT61 which has been investigated preclinically in numerous tumor types in the last few years. In this review, the emerging role of Hh signaling in cancer is critically evaluated focusing on the potential of targeting Hh signaling more downstream of SMO, i.e. at the level of the GLI transcription factors. Furthermore, the working mechanism and therapeutic potential of the most extensively studied GLI inhibitor in human cancer, i.e. GANT61, is discussed in detail. In conclusion, GANT61 appears to be highly effective against human cancer cells and in xenograft mouse models, targeting almost all of the classical hallmarks of cancer and could hence represent a promising treatment option for human cancer.
Collapse
Affiliation(s)
- Annelies Gonnissen
- University of Leuven (KU Leuven), Department of Oncology, Laboratory of Experimental Radiotherapy, Leuven, Belgium
| | - Sofie Isebaert
- University of Leuven (KU Leuven), Department of Oncology, Laboratory of Experimental Radiotherapy, Leuven, Belgium
| | - Karin Haustermans
- University of Leuven (KU Leuven), Department of Oncology, Laboratory of Experimental Radiotherapy, Leuven, Belgium.,University Hospitals Leuven, Department of Radiation Oncology, Leuven, Belgium
| |
Collapse
|