151
|
Moench R, Grimmig T, Kannen V, Tripathi S, Faber M, Moll EM, Chandraker A, Lissner R, Germer CT, Waaga-Gasser AM, Gasser M. Exclusive inhibition of PI3K/Akt/mTOR signaling is not sufficient to prevent PDGF-mediated effects on glycolysis and proliferation in colorectal cancer. Oncotarget 2018; 7:68749-68767. [PMID: 27626684 PMCID: PMC5356587 DOI: 10.18632/oncotarget.11899] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022] Open
Abstract
Platelet-derived growth factor (PDGF) and signaling via its receptors plays a crucial role in tumor cell proliferation and thus may represent an attractive target besides VEGF/EGFR-based antibody therapies. In this study we analyzed the influence of PDGF in colorectal cancer. PDGF was expressed intensively in early and even more intensively in late stage primary CRCs. Like VEGF, PDGF enhanced human colon cancer proliferation, and increased oxidative glycolytic activity, and activated HIF1α and c-Myc in vitro. PDGF activated the PI3K/Akt/mTOR pathway while leaving MAPK signaling untouched. Further dissection showed that inhibition of Akt strongly impeded cancer cell growth while inhibition of PI3K did not. MAPK analysis suggested an inhibitory crosstalk between both pathways, thus explaining the different effects of the Akt and PI3K inhibitors on cancer cell proliferation. PDGF stimulates colon cancer cell proliferation, and prevents inhibitor induced apoptosis, resulting in tumor growth. Therefore inhibition of PDGF signaling seems to be a promising target in colorectal cancer therapy. However, due to the multifaceted nature of the intracellular PDGF signaling, careful intervention strategies are needed when looking into specific signaling pathways like PI3K/Akt/mTOR and MAPK.
Collapse
Affiliation(s)
- Romana Moench
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Tanja Grimmig
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Vinicius Kannen
- Ribeirao Preto Pharmaceutical Sciences School, Department of Toxicology, Bromatology, and Clinical Analysis, University of Sao Paulo, Sao Paulo, Brazil
| | - Sudipta Tripathi
- Brigham and Women's Hospital, Transplant Research Center, Harvard Medical School, Boston, MA, USA
| | - Marc Faber
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Eva-Maria Moll
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Anil Chandraker
- Brigham and Women's Hospital, Transplant Research Center, Harvard Medical School, Boston, MA, USA
| | - Reinhard Lissner
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Ana Maria Waaga-Gasser
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany.,Brigham and Women's Hospital, Transplant Research Center, Harvard Medical School, Boston, MA, USA
| | - Martin Gasser
- Department of Surgery I, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
152
|
Muscatello LV, Avallone G, Serra F, Seuberlich T, Mandara MT, Sisó S, Brunetti B, Oevermann A. Glomeruloid Microvascular Proliferation, Desmoplasia, and High Proliferative Index as Potential Indicators of High Grade Canine Choroid Plexus Tumors. Vet Pathol 2018; 55:391-401. [PMID: 29402204 DOI: 10.1177/0300985817754124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Choroid plexus tumors (CPT) are intraventricular neoplasms accounting for 10% of all primary central nervous system tumors in dogs. They are frequently classified according to the human WHO classification into choroid plexus papilloma (CPP, grade I), atypical CPP (aCPP, grade II), and choroid plexus carcinoma (CPC, grade III). Histological features observed in canine CPT such as increased vascular density (IVD) and glomeruloid microvascular proliferation (GMVP) are not part of the WHO classification. This multi-centric study aimed to investigate tumor-associated vascular hyperplasia in dogs by determining the prevalence of GMVP and IVD in 52 canine CPT and their association with tumor grade. In addition, the expression of angiogenic factors was assessed by immunohistochemistry in 25 tumors to investigate the pathogenesis of tumor-associated vascular hyperplasia. Based on the classical histological hallmarks, this study of 52 CPT identified 22 (42%) CPP (grade I) and 30 of (58%) CPC (grade III). GMVP was more prevalent in CPC (13/30; 43%) than CPP (1/22; 4%), whereas IVD occurred to a similar extent in CPP and CPC. Desmoplasia was more common in CPC (19/30; 63%) than CPP (2/22; 9%), and similarly, the proliferative index (PI) of neoplastic epithelium was significantly higher in CPC (5.14%) than CPP (0.94%). The majority of CPT expressed platelet-derived growth factor (PDGF), PDGFRα, PDGFRβ, and vascular endothelial growth factor (VEGF) irrespective of tumor grade or tumor-associated vascular hyperplasia. These results suggest that tumor-associated GMVP, desmoplasia, and PI may serve as histological indicators of malignancy in CPT.
Collapse
Affiliation(s)
- Luisa Vera Muscatello
- 1 Department of Veterinary Medical Science (DIMEVET), University of Bologna, Bologna, Italy
| | - Giancarlo Avallone
- 1 Department of Veterinary Medical Science (DIMEVET), University of Bologna, Bologna, Italy
| | - Fabienne Serra
- 2 Division of Neurological Sciences, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Torsten Seuberlich
- 2 Division of Neurological Sciences, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Silvia Sisó
- 4 UC Davis School of Veterinary Medicine, Davis, CA, USA.,5 Biomarin Pharmaceuticals Inc., Novato, CA, USA
| | - Barbara Brunetti
- 1 Department of Veterinary Medical Science (DIMEVET), University of Bologna, Bologna, Italy
| | - Anna Oevermann
- 2 Division of Neurological Sciences, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
153
|
Salim MN, Masyitha D, Harris A, Balqis U, Iskandar CD, Hambal M, Darmawi. Anti-inflammatory activity of Jatropha curcas Linn. latex in cream formulation on CD68 expression in mice skin wound. Vet World 2018; 11:99-103. [PMID: 29657387 PMCID: PMC5891885 DOI: 10.14202/vetworld.2018.99-103] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/19/2017] [Indexed: 11/16/2022] Open
Abstract
Aim: The purpose of the present study was to determine the potential of Jatropha curcas latex in the cream formulation on CD68 immune expression (macrophages) during inflammatory phase wound healing process in mice skin. Materials and Methods: Amount of 12 two-months-old male mice were used between 30 and 40 g. To surgical procedures, wound skin incision was performed 2.0 cm in length until subcutaneous on the paravertebral of each animal. The treatment was carried under locally anesthetized with procaine cream. The mice were allotted into four groups of each, entire surface of each group wound covered by base cream control, sulfadiazine 0.1% cream, J. curcas latex cream 10% and, 15%, respectively. All experiments were performed twice a day for 3 days. The wound healing was assayed in stained histological sections in immunohistochemical of the wounds. CD68 expression was investigated under a microscope. Results: The results showed that the cream from the 10% and 15% latex of J. curcas revealed moderate immune reaction to CD68 on wound healing. Conclusion: We concluded that the latex cream of J. curcas possesses anti-inflammatory activity in wound healing process of mice skin.
Collapse
Affiliation(s)
- Muhammad Nur Salim
- Laboratory of Pathology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Dian Masyitha
- Laboratory of Histology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Abdul Harris
- Laboratory of Pharmacology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Ummu Balqis
- Laboratory of Pathology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Cut Dahlia Iskandar
- Laboratory of Histology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Muhammad Hambal
- Laboratory of Parasitology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Darmawi
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia.,Laboratory of Research, Faculty of Veterinary Medicine, Syiah Kuala University, Darussalam, Banda Aceh 23111, Indonesia
| |
Collapse
|
154
|
Sugg KB, Markworth JF, Disser NP, Rizzi AM, Talarek JR, Sarver DC, Brooks SV, Mendias CL. Postnatal tendon growth and remodeling require platelet-derived growth factor receptor signaling. Am J Physiol Cell Physiol 2017; 314:C389-C403. [PMID: 29341790 DOI: 10.1152/ajpcell.00258.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platelet-derived growth factor receptor (PDGFR) signaling plays an important role in the fundamental biological activities of many cells that compose musculoskeletal tissues. However, little is known about the role of PDGFR signaling during tendon growth and remodeling in adult animals. Using the hindlimb synergist ablation model of tendon growth, our objectives were to determine the role of PDGFR signaling in the adaptation of tendons subjected to a mechanical growth stimulus, as well as to investigate the biological mechanisms behind this response. We demonstrate that both PDGFRs, PDGFRα and PDGFRβ, are expressed in tendon fibroblasts and that the inhibition of PDGFR signaling suppresses the normal growth of tendon tissue in response to mechanical growth cues due to defects in fibroblast proliferation and migration. We also identify membrane type-1 matrix metalloproteinase (MT1-MMP) as an essential proteinase for the migration of tendon fibroblasts through their extracellular matrix. Furthermore, we report that MT1-MMP translation is regulated by phosphoinositide 3-kinase/Akt signaling, while ERK1/2 controls posttranslational trafficking of MT1-MMP to the plasma membrane of tendon fibroblasts. Taken together, these findings demonstrate that PDGFR signaling is necessary for postnatal tendon growth and remodeling and that MT1-MMP is a critical mediator of tendon fibroblast migration and a potential target for the treatment of tendon injuries and diseases.
Collapse
Affiliation(s)
- Kristoffer B Sugg
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - James F Markworth
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | - Nathaniel P Disser
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | - Andrew M Rizzi
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | - Jeffrey R Talarek
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School , Ann Arbor, Michigan
| | - Dylan C Sarver
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | - Susan V Brooks
- Department of Molecular and Integrative Physiology, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan Medical School , Ann Arbor, Michigan
| | - Christopher L Mendias
- Department of Orthopaedic Surgery, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School , Ann Arbor, Michigan.,Hospital for Special Surgery , New York, New York
| |
Collapse
|
155
|
Dihydroartemisinin selectively inhibits PDGFRα-positive ovarian cancer growth and metastasis through inducing degradation of PDGFRα protein. Cell Discov 2017; 3:17042. [PMID: 29387451 PMCID: PMC5787695 DOI: 10.1038/celldisc.2017.42] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023] Open
Abstract
To develop traditional medicines as modern pharmacotherapies, understanding their molecular mechanisms of action can be very helpful. We have recently reported that Artemisinin and its derivatives, which are clinically used anti-malarial drugs, have significant effects against ovarian cancer, but the direct molecular targets and related combination therapy have been unclear. Herein, we report that dihydroartemisinin, one of the most active derivatives of Artemisinin, directly targets platelet-derived growth factor receptor-alpha (PDGFRα) to inhibit ovarian cancer cell growth and metastasis. Dihydroartemisinin directly binds to the intercellular domain of PDGFRα, reducing its protein stability by accelerating its ubiquitin-mediated degradation, which further inactivates downstream phosphoinositide 3-Kinase and mitogen-activated protein kinase pathways and subsequently represses epithelial–mesenchymal transition, inhibiting cell growth and metastasis of PDGFRα-positive ovarian cancer in vitro and in vivo. A combinational treatment reveals that dihydroartemisinin sensitizes ovarian cancer cells to PDGFR inhibitors. Our clinical study also finds that PDGFRα is overexpressed and positively correlated with high grade and metastasis in human ovarian cancer. Considering that Artemisinin compounds are currently clinically used drugs with favorable safety profiles, the results from this study will potentiate their use in combination with clinically used PDGFRα inhibitors, leading to maximal therapeutic efficacy with minimal adverse effects in PDGFRα-positive cancer patients. These findings also shed high light on future development of novel Artemisinin-based targeted therapy.
Collapse
|
156
|
Al-Aqtash RA, Zihlif MA, Hammad H, Nassar ZD, Meliti JA, Taha MO. Ligand-based computational modelling of platelet-derived growth factor beta receptor leading to new angiogenesis inhibitory leads. Comput Biol Chem 2017; 71:170-179. [PMID: 29101826 DOI: 10.1016/j.compbiolchem.2017.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 01/03/2023]
Abstract
Platelet derived growth factor beta receptor (PDGFR- β) plays an important role in angiogenesis. PDGFR-β expression is correlated with increased vascularity and maturation of blood vessels in cancer. Pharmacophore modeling and quantitative structure-activity relationship (QSAR) analysis were combined to explore the structural requirements for ligand-PDGFR-β recognition using 107 known PDGFR-β inhibitors. Genetic function algorithm (GFA) coupled to k nearest neighbor (kNN) and multiple linear regression (MLR) analysis were employed to generate predictive QSAR models based on optimal combinations of pharmacophores and physicochemical descriptors. The successful pharmacophores were complemented with exclusion spheres to optimize their receiver operating characteristic curve (ROC) profiles. The QSAR models and their associated pharmacophore hypotheses were validated by identification and experimental evaluation of new angiogenesis inhibitory leads retrieved from the National Cancer Institute (NCI) structural database. Two hits illustrated low micromolar IC50 values in two distinct anti-angiogenesis bioassays.
Collapse
Affiliation(s)
- Rua'a A Al-Aqtash
- Department of Pharmacology, Faculty of Medicine, University of Jordan, Amman, Jordan
| | - Malek A Zihlif
- Department of Pharmacology, Faculty of Medicine, University of Jordan, Amman, Jordan
| | - Hana Hammad
- Department of Biology, University of Jordan, Amman, Jordan
| | - Zeyad D Nassar
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Jehad Al Meliti
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Mutasem O Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan.
| |
Collapse
|
157
|
Schachtschneider KM, Schwind RM, Darfour-Oduro KA, De AK, Rund LA, Singh K, Principe DR, Guzman G, Ray CE, Ozer H, Gaba RC, Schook LB. A validated, transitional and translational porcine model of hepatocellular carcinoma. Oncotarget 2017; 8:63620-63634. [PMID: 28969016 PMCID: PMC5609948 DOI: 10.18632/oncotarget.18872] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 06/05/2017] [Indexed: 12/29/2022] Open
Abstract
Difficult questions are confronting clinicians attempting to improve hepatocellular carcinoma (HCC) outcomes. A large animal model with genetic, anatomical, and physiological similarities to humans is required to transition from mouse models to human clinical trials to address unmet clinical needs. To validate our previously reported inducible porcine cancer model (Oncopig) as a transitional HCC model, Oncopig hepatocyte cultures were transformed using Cre recombinase. The resulting porcine HCC cells (pHCC) expressed oncogenic TP53R167H and KRASG12D, and displayed nuclear pleomorphisms with pale to granular cytoplasm arranged in expanded plates similar to human HCC histopathology. Human HCC transcriptional hallmarks were detected in pHCC cells using RNA-seq, including TERT reactivation, apoptosis evasion, angiogenesis activation, and Wnt signaling activation. Master regulators of gene expression were conserved across Oncopig and 18 human HCC cell lines. pHCC injection into SCID mice resulted in tumors recapitulating human HCC characteristics, including thick trabeculae formation, pseudoacini patterning, and sheets of well-vascularized stroma. Finally, autologous injection of pHCC cells subcutaneously yielded a tumor histologically characterized as Edmondson Steiner (HCC nuclear grade assessment system) grade 2 HCC with trabecular patterning and T-lymphocyte infiltration. These data demonstrate the Oncopig HCC model's utility for improving detection, treatment, and biomarker discovery relevant to human HCC.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
- Animal Breeding and Genomics Centre, Wageningen University, Wageningen, The Netherlands
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Regina M. Schwind
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Arun K. De
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Lauretta A. Rund
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Kuldeep Singh
- Veterinary Diagnostic Laboratory, University of Illinois, Urbana, IL, USA
| | - Daniel R. Principe
- College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Charles E. Ray
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Howard Ozer
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ron C. Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Lawrence B. Schook
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
158
|
Siemann DW, Chaplin DJ, Horsman MR. Realizing the Potential of Vascular Targeted Therapy: The Rationale for Combining Vascular Disrupting Agents and Anti-Angiogenic Agents to Treat Cancer. Cancer Invest 2017; 35:519-534. [DOI: 10.1080/07357907.2017.1364745] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- D. W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | | - M. R. Horsman
- Department of Experimental Clinical Oncology, Aarhus University, Denmark
| |
Collapse
|
159
|
Jackson S, ElAli A, Virgintino D, Gilbert MR. Blood-brain barrier pericyte importance in malignant gliomas: what we can learn from stroke and Alzheimer's disease. Neuro Oncol 2017; 19:1173-1182. [PMID: 28541444 PMCID: PMC5570196 DOI: 10.1093/neuonc/nox058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pericyte, a constitutive component of the central nervous system, is a poorly understood cell type that envelops the endothelial cell with the intended purpose of regulating vascular flow and endothelial cell permeability. Previous studies of pericyte function have been limited to a small number of disease processes such as ischemic stroke and Alzheimer's disease. Recently, publications have postulated a link between glioma stem cell differentiation and pericyte function. These studies suggest that there may be an important interaction of pericytes with tumor cells and other components of the tumor microenvironment in malignant primary glial neoplasms, most notably glioblastoma. This potential cellular interaction underscores the need to pursue more investigations of pericytes in malignant brain tumor biology. In this review, we summarize the functional roles of pericytes, particularly focusing on changes in pericyte biology during response to immune cells, inflammation, and hypoxic conditions. The information presented is based on the available data from studies of pericyte function in other central nervous system diseases but will serve as a foundation for research investigations to further understand the role of pericytes in malignant gliomas.
Collapse
Affiliation(s)
- Sadhana Jackson
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Ayman ElAli
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Mark R Gilbert
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
160
|
Carrera M, Cañas B, Lopez-Ferrer D. Fast Global Phosphoproteome Profiling of Jurkat T Cells by HIFU-TiO 2-SCX-LC-MS/MS. Anal Chem 2017; 89:8853-8862. [PMID: 28787133 DOI: 10.1021/acs.analchem.7b01321] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We propose a new workflow for fast phosphoproteome profiling. The workflow is based on the use of accelerated in-solution trypsin digestion under an ultrasonic field provided by high-intensity focused ultrasound (HIFU) combined with an inverse strategy based on TiO2 selective phosphopeptide enrichment, fractionation by strong cation exchange chromatography (SCX) and analysis by liquid chromatography tandem mass spectrometry (LC-MS/MS) using a high-resolution mass spectrometer. The performance of the method was established for the global phosphoproteome analysis of unstimulated human Jurkat leukemia T cells (E6.1). Using this accelerated workflow, 15367 phosphorylation sites from 13029 different phosphopeptides belonging to 3163 different phosphoproteins were efficiently identified with high-throughput and reproducibility in less than 15 h. The functional analysis revealed significant phosphorylation-based networks that are implicated in immune function and tumor development pathways. The present strategy, HIFU-TiO2-SCX-LC-MS/MS, is the fastest analytical method reported to date for generating large-scale phosphoproteomics data sets (<15 h).
Collapse
Affiliation(s)
- Mónica Carrera
- Spanish National Research Council (CSIC), 36208, Vigo, Spain
| | - Benito Cañas
- Complutense University of Madrid (UCM) , 28040, Madrid, Spain
| | | |
Collapse
|
161
|
A Review of Anti-Angiogenic Targets for Monoclonal Antibody Cancer Therapy. Int J Mol Sci 2017; 18:ijms18081786. [PMID: 28817103 PMCID: PMC5578174 DOI: 10.3390/ijms18081786] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor angiogenesis is a key event that governs tumor progression and metastasis. It is controlled by the complicated and coordinated actions of pro-angiogenic factors and their receptors that become upregulated during tumorigenesis. Over the past several decades, vascular endothelial growth factor (VEGF) signaling has been identified as a central axis in tumor angiogenesis. The remarkable advent of recombinant antibody technology has led to the development of bevacizumab, a humanized antibody that targets VEGF and is a leading clinical therapy to suppress tumor angiogenesis. However, despite the clinical efficacy of bevacizumab, its significant side effects and drug resistance have raised concerns necessitating the identification of novel drug targets and development of novel therapeutics to combat tumor angiogenesis. This review will highlight the role and relevance of VEGF and other potential therapeutic targets and their receptors in angiogenesis. Simultaneously, we will also cover the current status of monoclonal antibodies being developed to target these candidates for cancer therapy.
Collapse
|
162
|
Determination of 16 serum angiogenic factors in stage I non-small cell lung cancer using a bead-based multiplex immunoassay. Biomed Pharmacother 2017; 88:1031-1037. [DOI: 10.1016/j.biopha.2017.01.141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/17/2022] Open
|
163
|
Rustenhoven J, Jansson D, Smyth LC, Dragunow M. Brain Pericytes As Mediators of Neuroinflammation. Trends Pharmacol Sci 2017; 38:291-304. [DOI: 10.1016/j.tips.2016.12.001] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 01/03/2023]
|
164
|
Corrales L, Nogueira A, Passiglia F, Listi A, Caglevic C, Giallombardo M, Raez L, Santos E, Rolfo C. Second-Line Treatment of Non-Small Cell Lung Cancer: Clinical, Pathological, and Molecular Aspects of Nintedanib. Front Med (Lausanne) 2017; 4:13. [PMID: 28293555 PMCID: PMC5329017 DOI: 10.3389/fmed.2017.00013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/06/2017] [Indexed: 12/11/2022] Open
Abstract
Lung carcinoma is the leading cause of death by cancer in the world. Nowadays, most patients will experience disease progression during or after first-line chemotherapy demonstrating the need for new, effective second-line treatments. The only approved second-line therapies for patients without targetable oncogenic drivers are docetaxel, gemcitabine, pemetrexed, and erlotinib and for patients with target-specific oncogenes afatinib, osimertinib, crizotinib, alectinib, and ceritinib. In recent years, evidence on the role of antiangiogenic agents have been established as important and effective therapeutic targets in non-small cell lung cancer (NSCLC). Nintedanib is a tyrosine kinase inhibitor targeting three angiogenesis-related transmembrane receptors (vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor). Several preclinical and clinical studies have proven the usefulness of nintedanib as an anticancer agent for NSCLC. The most important study was the phase III LUME-Lung 1 trial, which investigated the combination of nintedanib with docetaxel for second-line treatment in advanced NSCLC patients. The significant improvement in overall survival and the manageable safety profile led to the approval of this new treatment in Europe. This review focuses on the preclinical and clinical studies with nintedanib in NSCLC.
Collapse
Affiliation(s)
- Luis Corrales
- Clinical Oncology Department, Hospital San Juan de Dios, San José, Costa Rica
| | - Amanda Nogueira
- Phase I – Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Center for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
| | - Francesco Passiglia
- Phase I – Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Center for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Angela Listi
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Christian Caglevic
- Medical Oncology Department, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Marco Giallombardo
- Phase I – Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Center for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
| | - Luis Raez
- Thoracic Oncology Program, Memorial Cancer Institute, Memorial Health Care System, Pembroke Pines, FL, USA
| | - Edgardo Santos
- Oncology Department, Lynn Cancer Institute, Boca Raton, FL, USA
| | - Christian Rolfo
- Phase I – Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Center for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
| |
Collapse
|
165
|
Cimpean AM, Cobec IM, Ceaușu RA, Popescu R, Tudor A, Raica M. Platelet Derived Growth Factor BB: A "Must-have" Therapeutic Target "Redivivus" in Ovarian Cancer. Cancer Genomics Proteomics 2017; 13:511-517. [PMID: 27807074 DOI: 10.21873/cgp.20014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/22/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND We aimed to validate PDGF-BB protein expression by RNAscope, a sensitive method for PDGF-BB mRNA evaluation on paraffin embedded (FFPE) specimens of ovarian tumors. MATERIALS AND METHODS Seventy-five FFPE ovarian cancer biopsies were assessed by immunohistochemistry followed by PDGF-BB mRNA RNAscope validation. RESULTS AND CONCLUSION Dual PDGF-BB expression in tumor and stromal cells have been observed, being highly suggestive for PDGF-BB mediated stromal-tumor cells reciprocal interaction in ovarian cancer (p=0.008). It seems that the nuclear expression of the PDGF-BB represents a negative prognostic factor in ovarian tumors. Being a controversial issue in the literature, PDGF-BB nuclear expression detected by immunohistochemistry was validated by RNAscope in situ hybridization. More than 65% of cases had PDGF-BB mRNA amplification, confirming immunohistochemical results. We herein validated PDGF-BB as a potential therapeutic and prognostic tool of ovarian cancer aggressiveness.
Collapse
Affiliation(s)
- Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ionut Marcel Cobec
- Department of Obstetrics and Gynecology, Diakonie Klinikum, Academic Hospital of the Heidelberg University, Schwäbisch Hall, Germany
| | - Raluca Amalia Ceaușu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Roxana Popescu
- Department of Microscopic Morphology/ Cell and Molecular Biology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anca Tudor
- Department of Medical Informatics and Biostatistics, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
166
|
Corlan AS, Cîmpean AM, Jitariu AA, Melnic E, Raica M. Endocrine Gland-Derived Vascular Endothelial Growth Factor/Prokineticin-1 in Cancer Development and Tumor Angiogenesis. Int J Endocrinol 2017; 2017:3232905. [PMID: 28386275 PMCID: PMC5366234 DOI: 10.1155/2017/3232905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/15/2017] [Accepted: 01/23/2017] [Indexed: 12/26/2022] Open
Abstract
A lot of data suggests endocrine gland-derived vascular endothelial growth factor (EG-VEGF) to be restricted to endocrine glands and to some endocrine-dependent organs. Many evidences show that EG-VEGF stimulates angiogenesis and cell proliferation, although it is not a member of the VEGF family. At the time, a lot of data regarding the role of this growth factor in normal development are available. However, controversial results have been published in the case of pathological conditions and particularly in malignant tumors. Thus, our present paper has been focused on the role of EG-VEGF in normal tissues and various malignant tumors and their angiogenic processes.
Collapse
Affiliation(s)
- Ana Silvia Corlan
- Department of Endocrinology, “Vasile Goldis” University of Arad, Arad, Romania
| | - Anca Maria Cîmpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara, Romania
- *Anca Maria Cîmpean:
| | - Adriana-Andreea Jitariu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara, Romania
| | - Eugen Melnic
- Department of Pathology, “Nicolae Testemitanu” University of Medicine and Pharmacy, Chișinău, Moldova
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara, Romania
| |
Collapse
|
167
|
Nemaysh V, Luthra PM. Computational analysis revealing that K634 and T681 mutations modulate the 3D-structure of PDGFR-β and lead to sunitinib resistance. RSC Adv 2017. [DOI: 10.1039/c7ra01305a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Platelet-derived growth factor receptor-beta (PDGFR-β) is expressed by endothelial cells (ECs) of tumor-associated blood vessels and regulates primarily early hematopoiesis.
Collapse
Affiliation(s)
- Vishal Nemaysh
- Neuropharmaceutical Chemistry Research Laboratory
- Dr B. R. Ambedkar Center for Biomedical Research
- University of Delhi
- Delhi-110007
- India
| | - Pratibha Mehta Luthra
- Neuropharmaceutical Chemistry Research Laboratory
- Dr B. R. Ambedkar Center for Biomedical Research
- University of Delhi
- Delhi-110007
- India
| |
Collapse
|
168
|
Mendonça F, Soares R. Obesity and cancer phenotype: Is angiogenesis a missed link? Life Sci 2015; 139:16-23. [PMID: 26297445 DOI: 10.1016/j.lfs.2015.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/28/2015] [Accepted: 08/14/2015] [Indexed: 12/13/2022]
Abstract
Obesity remains nowadays one of the main threats to human health, being a problem of worldwide proportions. It is characterized by augmented storage of fatty acids in an enlarged adipose tissue. This process is possible thanks to a rich capillary network, supported by a mechanism that has also a crucial role on cancer: angiogenesis. Given that several studies point obesity as a risk factor for cancer development, angiogenesis may be approached as the missed link between these two pathologies. Understanding the different pathways behind angiogenesis becomes essential to break this link by developing new anti-angiogenic therapies or improving the actual ones. In the first phase, this paper will focus the structural and cellular changes that adipose tissue suffers in obesity. Then, the main pro-angiogenic players will be reviewed, taking into account the pathways that explain their putative role in obesity-cancer link. Finally, the clinical implications of the presented mechanisms will also be regarded, being the main focus on the anti-angiogenic therapies.
Collapse
Affiliation(s)
- Fernando Mendonça
- Department of Biochemistry, Faculty of Medicine, University of Porto, Portugal
| | - Raquel Soares
- Department of Biochemistry, Faculty of Medicine, University of Porto, Portugal; Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Portugal.
| |
Collapse
|
169
|
The Effect of Electroporation of a Lyotroic Liquid Crystal Genistein-Based Formulation in the Recovery of Murine Melanoma Lesions. Int J Mol Sci 2015; 16:15425-41. [PMID: 26184156 PMCID: PMC4519906 DOI: 10.3390/ijms160715425] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/11/2015] [Accepted: 06/25/2015] [Indexed: 12/15/2022] Open
Abstract
A lamellar lyotropic liquid crystal genistein-based formulation (LLC-Gen) was prepared in order to increase the aqueous solubility of the lipophilic phytocompound genistein. The formulation was applied locally, in a murine model of melanoma, with or without electroporation. The results demonstrated that, when the formulation was applied by electroporation, the tumors appeared later. During the 21 days of the experiment, the LLC-Gen formulation decreased the tumor volume, the amount of melanin and the degree of erythema, but when electroporation was applied, all these parameters indicated a better prognosis even (lower tumor volume, amount of melanin and degree of erythema). Although hematoxylin-eosin (HE) staining confirmed the above events, application of the LLC-Gen formulation by electroporation did not lead to a significant effect in terms of the serum concentrations of the protein S100B and serum neuron specific enolase (NSE), or the tissue expression of the platelet-derived growth factor receptor β (PDGFRβ) antibody.
Collapse
|
170
|
Rajabzadeh AR, Eimani H, Mohseni Koochesfahani H, Shahvardi AH, Fathi R. Morphological study of isolated ovarian preantral follicles using fibrin gel plus platelet lysate after subcutaneous transplantation. CELL JOURNAL 2015; 17:145-52. [PMID: 25870844 PMCID: PMC4393662 DOI: 10.22074/cellj.2015.521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 03/10/2014] [Indexed: 11/20/2022]
Abstract
Objective Ovarian and follicle transplantation may preserve fertility in young cancer survivors. In this study, we have transplanted preantral follicles using fibrin gel as a carrier
and fibrin gel supplemented with platelet lysate (PL) as a rich source of angiogenic and
growth factors. The purpose of this study was to evaluate the role of fibrin gel and PL in
follicle transplantation. Materials and Methods In this experimental study, ovaries were taken from 14-day-
old Naval Medical Research Institute (NMRI) mice. Preantral follicles were dissected
from the ovaries and encapsulated into fibrin gel supplemented with 5, 10, 15 or 20%
PL, then transplanted back into the same donor mice. Fibrin gels supplemented with
PL that contained preantral follicles were placed in a subcutaneous pocket in the
back of the neck of the recipient, donor mouse (the same mouse that follicles were
collected). After 14 days the grafts were processed and embedded in paraffin blocks,
then serially sectioned for histological evaluation. We counted the follicles and classified them according to stage (preantral or antral). Data were presented as mean ±
standard error of mean (SEM) and analysed by analysis of variance (ANOVA) and the
Kruskal-Wallistest.
Results The mean percentage of recovered follicles encapsulated and transplanted
in each group were 33.30 ± 2.47 (fibrin gel), 31.96 ± 1.90 (fibrin gel+5% PL), 34.02
± 2.44 (fibrin gel+10% PL), 48.31 ± 2.06 (fibrin gel+15% PL) and 17.60 ± 2.79 (fibrin
gel+20% PL). There was a significant increase in the recovery rate of grafted follicles with fibrin gel+15% PL (48.31%; p<0.001). The percentage of preantral follicles
showed no significant difference in all groups (p<0.05). The percentage of antral follicles showed a significant decrease in follicles grafted with fibrin gel+20% PL when
compared to the other groups (11.77%; p<0.005) but no significant difference was
observed in the other groups.
Conclusion The use of PL in follicle transplantation can improve ovarian follicular
survival rate.
Collapse
Affiliation(s)
- Ali Reza Rajabzadeh
- Department of Embryology at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran ; Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Hossein Eimani
- Department of Embryology at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran ; Department of Anatomy, Faculty of Medicine, Baqiyatallah (a.s.) University of Medical Sciences, Tehran, Iran
| | - Homa Mohseni Koochesfahani
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Abdol-Hossein Shahvardi
- Department of Embryology at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
171
|
Jitariu AA, Cimpean AM, Kundnani NR, Raica M. Platelet-derived growth factors induced lymphangiogenesis: evidence, unanswered questions and upcoming challenges. Arch Med Sci 2015; 11:57-66. [PMID: 25861290 PMCID: PMC4379379 DOI: 10.5114/aoms.2015.49217] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 11/20/2013] [Accepted: 12/04/2013] [Indexed: 01/03/2023] Open
Abstract
Crosstalk between angiogenesis and lymphangiogenesis in embryonic development continues during postnatal life and has specific mechanisms involving factors that initiate activation of the intracellular cascade for their specific receptors. Platelet-derived growth factors (PDGFs) and their corresponding receptors (PDGFRs) are known as important regulators of blood vessel development in both normal and pathologic angiogenesis. Despite some recent papers which reported a potential role of the PDGF/PDGFR axis in lymphatic spread of tumor cells, a few papers have suggested the potential role of PDGFs in tumor lymphangiogenesis development. The present paper summarizes the potential lymphangiogenic role of the PDGF/PDGFR axis, underlying upcoming challenges in the field.
Collapse
Affiliation(s)
| | | | | | - Marius Raica
- Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
172
|
Tolerability of Nintedanib (BIBF 1120) in Combination with Docetaxel: A Phase 1 Study in Japanese Patients with Previously Treated Non–Small-Cell Lung Cancer. J Thorac Oncol 2015; 10:346-52. [DOI: 10.1097/jto.0000000000000395] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
173
|
Gacche RN, Meshram RJ. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim Biophys Acta Rev Cancer 2014; 1846:161-79. [DOI: 10.1016/j.bbcan.2014.05.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/17/2022]
|
174
|
Mross K, Büchert M, Frost A, Medinger M, Stopfer P, Studeny M, Kaiser R. Vascular effects, efficacy and safety of nintedanib in patients with advanced, refractory colorectal cancer: a prospective phase I subanalysis. BMC Cancer 2014; 14:510. [PMID: 25012508 PMCID: PMC4105047 DOI: 10.1186/1471-2407-14-510] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 07/04/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Nintedanib is a potent, oral angiokinase inhibitor that targets VEGF, PDGF and FGF signalling, as well as RET and Flt3. The maximum tolerated dose of nintedanib was evaluated in a phase I study of treatment-refractory patients with advanced solid tumours. In this preplanned subanalysis, the effect of nintedanib on the tumour vasculature, along with efficacy and safety, was assessed in 30 patients with colorectal cancer (CRC). METHODS Patients with advanced CRC who had failed conventional treatment, or for whom no therapy of proven efficacy existed, were treated with nintedanib ranging from 50-450 mg once-daily (n = 14) or 150-250 mg twice-daily (n = 16) for 28 days. After a 1-week rest, further courses were permitted in the absence of progression or undue toxicity. The primary objective was the effect on the tumour vasculature using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and expressed as the initial area under the DCE-MRI contrast agent concentration-time curve after 60 seconds (iAUC60) or the volume transfer constant between blood plasma and extravascular extracellular space (Ktrans). RESULTS Patients received a median of 4.0 courses (range: 1-13). Among 21 evaluable patients, 14 (67%) had a ≥40% reduction from baseline in Ktrans and 13 (62%) had a ≥40% decrease from baseline in iAUC60, representing clinically relevant effects on tumour blood flow and permeability, respectively. A ≥40% reduction from baseline in Ktrans was positively associated with non-progressive tumour status (Fisher's exact: p = 0.0032). One patient achieved a partial response at 250 mg twice-daily and 24 (80%) achieved stable disease lasting ≥8 weeks. Time to tumour progression (TTP) at 4 months was 26% and median TTP was 72.5 days (95% confidence interval: 65-114). Common drug-related adverse events (AEs) included nausea (67%), vomiting (53%) and diarrhoea (40%); three patients experienced drug-related AEs ≥ grade 3. Four patients treated with nintedanib once-daily had an alanine aminotransferase and/or aspartate aminotransferase increase ≥ grade 3. No increases > grade 2 were seen in the twice-daily group. CONCLUSIONS Nintedanib modulates tumour blood flow and permeability in patients with advanced, refractory CRC, while achieving antitumour activity and maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Klaus Mross
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Martin Büchert
- Magnetic Resonance Development and Application Center, Department of Radiology, University Medical Center Freiburg, Freiburg, Germany
| | - Annette Frost
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Michael Medinger
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Matus Studeny
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Rolf Kaiser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
175
|
Dai J, Kong Y, Si L, Chi Z, Cui C, Sheng X, Mao L, Li S, Lian B, Yang R, Liu S, Xu X, Guo J. Large-scale analysis of PDGFRA mutations in melanomas and evaluation of their sensitivity to tyrosine kinase inhibitors imatinib and crenolanib. Clin Cancer Res 2013; 19:6935-42. [PMID: 24132921 DOI: 10.1158/1078-0432.ccr-13-1266] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Platelet-derived growth factor receptor α (PDGFRA) is a target for tyrosine kinase inhibitor (TKI)-based targeted therapy. Dysregulation of PDGFRA has been reported in many cancers. However, PDGFRA mutations in melanomas have not been well studied. We analyzed the genetic mutations of PDGFRA in Chinese patients with melanoma and determined the inhibitory potency of TKIs, such as imatinib and crenolanib, on mutant PDGFRA. EXPERIMENTAL DESIGN Of note, 351 melanoma tissue samples were examined for genetic mutations in exons 12, 14, and 18 of PDGFRA. Activities of mutations in response to imatinib and crenolanib were analyzed by Western blotting of tyrosine-phosphorylated PDGFRA and cell proliferation assays. RESULTS PDGFRA mutations were observed in 4.6% (16 of 351) of melanomas, and these mutations were mainly detected in acral and mucosal melanomas. PDGFRA mutations seem to be mutually exclusive with KIT mutations, but may coexist with BRAF and NRAS mutations. The genetic mutations of PDGFRA were unrelated to the age, thickness, and ulceration status of primary melanomas. Thirteen mutations were not reported before, and five (P577S, V658A, R841K, H845Y, and G853D) of them resulted in strong autophosphorylation of PDGFRA. Crenolanib showed higher potency than imatinib in inhibiting the kinase activity of PDGFRA. Except that V658A mutation was imatinib-resistant, all the other mutations were sensitive to both imatinib and crenolanib. CONCLUSIONS PDGFRA mutations are detected in a small population of melanoma patients. Our study suggests that patients with melanoma harboring certain PDGFRA mutations may benefit from imatinib and crenolanib treatment.
Collapse
Affiliation(s)
- Jie Dai
- Authors' Affiliations: Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China; and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Proliferative vitreoretinopathy after eye injuries: an overexpression of growth factors and cytokines leading to a retinal keloid. Mediators Inflamm 2013; 2013:269787. [PMID: 24198445 PMCID: PMC3806231 DOI: 10.1155/2013/269787] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 08/26/2013] [Indexed: 01/16/2023] Open
Abstract
Eye injury is a significant disabling worldwide health problem. Proliferative Vitreoretinopathy (PVR) is a common complication that develops in up to 40–60% of patients with an open-globe injury. Our knowledge about the pathogenesis of PVR has improved in the last decades. It seems that the introduction of immune cells into the vitreous, like in penetrating ocular trauma, triggers the production of growth factors and cytokines that come in contact with intra-retinal cells, like Müller cells and RPE cells. Growth factors and cytokines drive the cellular responses leading to PVR's development. Knowledge of the pathobiological and pathophysiological mechanisms involved in posttraumatic PVR is increasing the possibilities of management, and it is hoped that in the future our treatment strategies will evolve, in particular adopting a multidrug approach, and become even more effective in vision recovery. This paper reviews the current literature and clinical trial data on the pathogenesis of PVR and its correlation with ocular trauma and describes the biochemical/molecular events that will be fundamental for the development of novel treatment strategies. This literature review included PubMed articles published from 1979 through 2013. Only studies written in English were included.
Collapse
|
177
|
Inaba Y, Kanai F, Aramaki T, Yamamoto T, Tanaka T, Yamakado K, Kaneko S, Kudo M, Imanaka K, Kora S, Nishida N, Kawai N, Seki H, Matsui O, Arioka H, Arai Y. A randomised phase II study of TSU-68 in patients with hepatocellular carcinoma treated by transarterial chemoembolisation. Eur J Cancer 2013; 49:2832-40. [PMID: 23764238 DOI: 10.1016/j.ejca.2013.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/11/2013] [Accepted: 05/14/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND TSU-68 is an antitumour drug that acts by inhibiting angiogenesis. We evaluated the efficacy and safety of TSU-68 in combination with transarterial chemoembolisation (TACE) in patients with intermediate-stage hepatocellular carcinoma (HCC). PATIENTS AND METHODS In this multicenter, open-label phase II study, we randomised patients with HCC who had been treated with a single session of TACE to receive either 200mg TSU-68 twice daily or no medication. The primary end-point was progression-free survival (PFS). RESULTS A total of 103 patients were enrolled. Median PFS was 157.0days (95% confidence interval [CI], 124.0-230.0days) in the TSU-68 group and 122.0days (95% CI, 73.0-170.0days) in the control group. The hazard ratio was 0.699 (95% CI, 0.450-1.088). Fatigue, elevated aspartate aminotransferase (AST), elevated alkaline phosphatase, oedema and anorexia were more frequent in the TSU-68 group than in the control group. The most frequent grade 3/4 adverse events were AST elevation (46% of patients in the TSU-68 group and 12% of controls) and alanine aminotransferase elevation (26% of patients in the TSU-68 group and 8% of controls). Two deaths, grade 5 hepatic failure and melena were noted in the TSU-68 group. CONCLUSION This exploratory study shows a trend towards prolonged PFS with TSU-68 treatment after a single session of TACE, but this observation was not statistically significant. The two deaths were related to the study treatment. These results suggest that further examination of the study design is necessary to determine whether TSU-68 has any clinical benefits when combined with TACE.
Collapse
Affiliation(s)
- Yoshitaka Inaba
- Department of Diagnostic and Interventional Radiology, Aichi Cancer Center Hospital, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Sathish V, Babu E, Ramdass A, Lu ZZ, Chang TT, Velayudham M, Thanasekaran P, Lu KL, Li WS, Rajagopal S. Photoswitchable alkoxy-bridged binuclear rhenium(i) complexes – a potential probe for biomolecules and optical cell imaging. RSC Adv 2013. [DOI: 10.1039/c3ra42627k] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
179
|
Verstraete K, Savvides SN. Extracellular assembly and activation principles of oncogenic class III receptor tyrosine kinases. Nat Rev Cancer 2012; 12:753-66. [PMID: 23076159 DOI: 10.1038/nrc3371] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intracellular signalling cascades initiated by class III receptor tyrosine kinases (RTK-IIIs) and their cytokine ligands contribute to haematopoiesis and mesenchymal tissue development. They are also implicated in a wide range of inflammatory disorders and cancers. Recent snapshots of RTK-III ectodomains in complex with cognate cytokines have revealed timely insights into the structural determinants of RTK-III activation, evolution and pathology. Importantly, candidate 'driver' and 'passenger' mutations that have been identified in RTK-IIIs can now be collectively mapped for the first time to structural scaffolds of the corresponding RTK-III ectodomains. Such insights will generate a renewed interest in dissecting the mechanistic effects of such mutations and their therapeutic relevance.
Collapse
Affiliation(s)
- Kenneth Verstraete
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering, Ghent University, K.L. Ledeganckstraat 35, 9000 Ghent, Belgium.
| | | |
Collapse
|
180
|
Plé PA, Jung F, Ashton S, Hennequin L, Laine R, Lambert-van der Brempt C, Morgentin R, Pasquet G, Taylor S. Discovery of new quinoline ether inhibitors with high affinity and selectivity for PDGFR tyrosine kinases. Bioorg Med Chem Lett 2012; 22:3050-5. [DOI: 10.1016/j.bmcl.2012.03.074] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 03/19/2012] [Accepted: 03/21/2012] [Indexed: 10/28/2022]
|
181
|
Wang H, Yin Y, Li W, Zhao X, Yu Y, Zhu J, Qin Z, Wang Q, Wang K, Lu W, Liu J, Huang L. Over-expression of PDGFR-β promotes PDGF-induced proliferation, migration, and angiogenesis of EPCs through PI3K/Akt signaling pathway. PLoS One 2012; 7:e30503. [PMID: 22355314 PMCID: PMC3280261 DOI: 10.1371/journal.pone.0030503] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Accepted: 12/16/2011] [Indexed: 12/04/2022] Open
Abstract
The proliferation, migration, and angiogenesis of endothelial progenitor cells (EPCs) play critical roles in postnatal neovascularization and re-endothelialization following vascular injury. Here we evaluated whether the over-expression of platelet-derived growth factor receptor-β (PDGFR-β) can enhance the PDGF-BB-stimulated biological functions of EPCs through the PDGFR-β/phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. We first confirmed the expression of endogenous PDGFR-β and its plasma membrane localization in spleen-derived EPCs. We then demonstrated that the PDGFR-β over-expression in EPCs enhanced the PDGF-BB-induced proliferation, migration, and angiogenesis of EPCs. Using AG1295 (a PDGFR kinase inhibitor), LY294002 (a PI3K inhibitor), and sc-221226 (an Akt inhibitor), we further showed that the PI3K/Akt signaling pathway participates in the PDGF-BB-induced proliferation, migration, and angiogenesis of EPCs. In addition, the PI3K/Akt signaling pathway is required for PDGFR-β over-expression to enhance these PDGF-BB-induced phenotypes.
Collapse
Affiliation(s)
- Hang Wang
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Yangguang Yin
- Emergency Department, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Wei Li
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Xiaohui Zhao
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Yang Yu
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Jinkun Zhu
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Zhexue Qin
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Qiang Wang
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Kui Wang
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Wei Lu
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Jie Liu
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
| | - Lan Huang
- Institute of Cardiovascular Science Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, People's Republic of China
- * E-mail:
| |
Collapse
|
182
|
Kim M, Bae M, Na H, Yang M. Environmental toxicants--induced epigenetic alterations and their reversers. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2012; 30:323-367. [PMID: 23167630 DOI: 10.1080/10590501.2012.731959] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Epigenetics has been emphasized in the postgenome era to clarify obscure health risks of environmental toxicants including endocrine disrupting chemicals (EDCs). In addition, mixed exposure in real life can modify health consequences of the toxicants. Particularly, some nutritional and dietary materials modify individual susceptibility through changes in the epigenome. Therefore, we focused on some environmental toxicants that induce epigenetic alterations, and introduced chemopreventive materials to reverse the toxicants-induced epigenetic alterations. Methodologically, we used global and specific DNA methylation as epigenetic end points and searched epigenetic modulators in food. We reviewed various epigenetic end points induced by environmental toxicants including alcohol, asbestos, nanomaterials, benzene, EDCs, metals, and ionizing radiation. The epigenetic end points can be summarized into global hypomethylation and specific hypermethylation at diverse tumor suppress genes. Exposure timing, dose, sex, or organ specificity should be considered to use the epigenetic end points as biomarkers for exposure to the epimutagenic toxicants. Particularly, neonatal exposure to the epimutagens can influence their future adult health because of characteristics of the epimutagens, which disrupt epigenetic regulation in imprinting, organogenesis, development, etc. Considering interaction between epimutagenic toxicants and their reversers in food, we suggest that multiple exposures to them can alleviate or mask epigenetic toxicity in real life. Our present review provides useful information to find new end points of environmental toxicants and to prevention from environment-related diseases.
Collapse
Affiliation(s)
- Minju Kim
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Yongsan-gu, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
183
|
Clavreul A, Etcheverry A, Chassevent A, Quillien V, Avril T, Jourdan ML, Michalak S, François P, Carré JL, Mosser J, Menei P. Isolation of a new cell population in the glioblastoma microenvironment. J Neurooncol 2011; 106:493-504. [DOI: 10.1007/s11060-011-0701-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 08/16/2011] [Indexed: 01/14/2023]
|
184
|
Shan H, Takahashi T, Bando Y, Izumi K, Uehara H. Inhibitory effect of soluble platelet-derived growth factor receptor β on intraosseous growth of breast cancer cells in nude mice. Cancer Sci 2011; 102:1904-10. [PMID: 21733044 DOI: 10.1111/j.1349-7006.2011.02026.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Bone metastasis is a frequent complication of advanced breast cancer. On the basis of functional and molecular evidence, signaling mediated by the binding of platelet-derived growth factor (PDGF)-BB and -DD to PDGF receptor β (PDGFRβ) is critical for the survival and growth of metastatic breast cancer cells within the bone microenvironment. In this study, we propose a new approach to blocking PDGFRβ signaling using soluble PDGFRβ (sPDGFRβ) as a decoy receptor for PDGF-BB and -DD secreted from tumor cells and bone marrow stromal cells. A bone-seeking TNBCT/Bo cell line was established by in vivo selection from TNBCT human breast cancer cells, which are negative for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 protein expression. The TNBCT/Bo cells were transfected with a mammalian expression vector encoding the extracellular domain of PDGFRβ. A stable transfectant (TNBCT/Bo-sPDGFRβ) grew at a similar rate to that of control cells under normal culture conditions, although growth stimulation of human fibroblasts with PDGF-BB was neutralized by the culture medium from TNBCT/Bo-sPDGFRβ cells. Intratibial injection of TNBCT/Bo-sPDGFRβ cells into athymic nude mice resulted in a significant decrease in tumor incidence compared with control mice (P < 0.01). This attenuated growth correlated with decreased cancer cell proliferation, angiogenesis, and recruitment of stromal cells, and with an increase in the number of apoptotic cells. These findings suggest that sPDGFRβ is useful for the treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Hongchao Shan
- Department of Molecular and Environmental Pathology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | | | |
Collapse
|
185
|
Abstract
Breast cancer frequently metastasizes to the skeleton, interrupting the normal bone remodeling process and causing bone degradation. Osteolytic lesions are the end result of osteoclast activity; however, osteoclast differentiation and activation are mediated by osteoblast production of RANKL (receptor activator for NFκB ligand) and several osteoclastogenic cytokines. Osteoblasts themselves are negatively affected by cancer cells as evidenced by an increase in apoptosis and a decrease in proteins required for new bone formation. Thus, bone loss is due to both increased activation of osteoclasts and suppression of osteoblasts. This review summarizes the current understanding of the osteolytic mechanisms of bone metastases, including a discussion of current therapies.
Collapse
|