201
|
Harrell CR, Simovic Markovic B, Fellabaum C, Arsenijevic A, Djonov V, Volarevic V. Molecular mechanisms underlying therapeutic potential of pericytes. J Biomed Sci 2018; 25:21. [PMID: 29519245 PMCID: PMC5844098 DOI: 10.1186/s12929-018-0423-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/21/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pericytes are multipotent cells present in every vascularized tissue in the body. Despite the fact that they are well-known for more than a century, pericytes are still representing cells with intriguing properties. This is mainly because of their heterogeneity in terms of definition, tissue distribution, origin, phenotype and multi-functional properties. The body of knowledge illustrates importance of pericytes in the regulation of homeostatic and healing processes in the body. MAIN BODY In this review, we summarized current knowledge regarding identification, isolation, ontogeny and functional characteristics of pericytes and described molecular mechanisms involved in the crosstalk between pericytes and endothelial or immune cells. We highlighted the role of pericytes in the pathogenesis of fibrosis, diabetes-related complications (retinopathy, nephropathy, neuropathy and erectile dysfunction), ischemic organ failure, pulmonary hypertension, Alzheimer disease, tumor growth and metastasis with the focus on their therapeutic potential in the regenerative medicine. The functions and capabilities of pericytes are impressive and, as yet, incompletely understood. Molecular mechanisms responsible for pericyte-mediated regulation of vascular stability, angiogenesis and blood flow are well described while their regenerative and immunomodulatory characteristics are still not completely revealed. Strong evidence for pericytes' participation in physiological, as well as in pathological conditions reveals a broad potential for their therapeutic use. Recently published results obtained in animal studies showed that transplantation of pericytes could positively influence the healing of bone, muscle and skin and could support revascularization. However, the differences in their phenotype and function as well as the lack of standardized procedure for their isolation and characterization limit their use in clinical trials. CONCLUSION Critical to further progress in clinical application of pericytes will be identification of tissue specific pericyte phenotype and function, validation and standardization of the procedure for their isolation that will enable establishment of precise clinical settings in which pericyte-based therapy will be efficiently applied.
Collapse
Affiliation(s)
- C. Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida USA
| | - Bojana Simovic Markovic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| | - Crissy Fellabaum
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida USA
| | - Aleksandar Arsenijevic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| | - Valentin Djonov
- University of Bern, Institute of Anatomy, Baltzerstrasse 2, Bern, Switzerland
| | - Vladislav Volarevic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| |
Collapse
|
202
|
Flattened microvessel independently predicts poor prognosis of patients with non-small cell lung cancer. Oncotarget 2018; 8:30092-30099. [PMID: 28404911 PMCID: PMC5444728 DOI: 10.18632/oncotarget.15617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/27/2017] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis plays an essential role in improving tumor progression, whereas, its value in prognosis predicting remains controversial, especially in non-small cell lung cancer (NSCLC). Most recently, microvessel pattern has been raised as a novel prognosis factor. In this study, flattened microvessel, evaluated by tumor microvessel aspect ratio (TMAR), was conducted as a prognostic factor in NSCLC patients. A total of 100 patients with NSCLC were retrospectively reviewed. Microvessel in tumor was visualized by immunochemistry staining and then TMAR was determined. The prognostic role of TMAR was evaluated by univariate and multivariate analysis. Most of intratumor microvessels were flattened with a median TMAR of 3.65 (range, 2.43 - 6.28). Patients were stratified into high TMAR group (TMAR ≥ 3.6) and low TMAR group (TMAR < 3.6). Compared with subpopulation with low TMAR, high TMAR had significantly high risk of cancer-related death (univariate analysis: HR = 5.06, 95% CI: 2.44-10.47, p<0.001; multivariate analysis: HR = 4.53, 95% CI: 1.70-12.06, p=0.002). In conclusion, the results of our study demonstrate that flattened microvessel in tumor tissue is a promising prognosis predictor of NSCLC patients.
Collapse
|
203
|
Affiliation(s)
- G S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - P H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - A Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - A Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
204
|
Azevedo PO, Sena IFG, Andreotti JP, Carvalho-Tavares J, Alves-Filho JC, Cunha TM, Cunha FQ, Mintz A, Birbrair A. Pericytes modulate myelination in the central nervous system. J Cell Physiol 2018; 233:5523-5529. [PMID: 29215724 DOI: 10.1002/jcp.26348] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis is a highly prevalent chronic demyelinating disease of the central nervous system. Remyelination is the major therapeutic goal for this disorder. The lack of detailed knowledge about the cellular and molecular mechanisms involved in myelination restricts the design of effective treatments. A recent study by using [De La Fuente et al. (2017) Cell Reports, 20(8): 1755-1764] by using state-of-the-art techniques, including pericyte-deficient mice in combination with induced demyelination, reveal that pericytes participate in central nervous system regeneration. Strikingly, pericytes presence is essential for oligodendrocyte progenitors differentiation and myelin formation during remyelination in the brain. The emerging knowledge from this research will be important for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - Juliana Carvalho-Tavares
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - José C Alves-Filho
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, New York
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil.,Department of Radiology, Columbia University Medical Center, New York, New York
| |
Collapse
|
205
|
Chen L, Liu GQ, Wu HY, Jin J, Yin X, Li D, Lu PR. Monocyte chemoattractant protein 1 and fractalkine play opposite roles in angiogenesis via recruitment of different macrophage subtypes. Int J Ophthalmol 2018; 11:216-222. [PMID: 29487809 DOI: 10.18240/ijo.2018.02.06] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
AIM To explore the interaction between macrophages and chemokines [monocyte chemoattractant protein 1 (MCP-1/CCL2) and fractalkine/CX3CL1] and the effects of their interaction on neovascularization. METHODS Human peripheral blood mononuclear cells, donated by healthy volunteers, were separated and cultured in RPMI-1640 medium containing 10% fetal bovine serum, then induced into macrophages by stimulation with 30 µg/L granulocyte macrophage-colony stimulating factor (GM-CSF). The expression of CCR2 and/or CX3CR1 in the macrophages was examined using flow cytometry. Macrophages were then stimulated with recombinant human CCL2 (rh-CCL2) or recombinant human CX3CL1 (rh-CX3CL1). The expression of angiogenesis-related genes, including VEGF-A, THBS-1 and ADAMTS-1 were examined using real-time quantitative polymerase chain reaction (PCR). Supernatants from stimulated macrophages were used in an assay of human retinal endothelial cell (HREC) proliferation. Finally, stimulated macrophages were co-cultured with HREC in a migration assay. RESULTS The expression rate of CCR2 in macrophages stimulated by GM-CSF was 42%±1.9%. The expression rate of CX3CR1 was 71%±3.3%. Compared with vehicle-treated groups, gene expression of VEGF-A in the macrophages was greater in 150 mg/L CCL2-treated groups (P<0.05), while expression of THBS-1 and ADAMTS-1 was significantly lower (P<0.05). By contrast, compared with vehicle-treated groups, expression of VEGF-A in 150 mg/L CX3CL1-treated groups was significantly lower (P<0.05), while expression of THBS-1 and ADAMTS-1 was greater (P<0.05). Supernatants from CCL2 treated macrophages promoted proliferation of HREC (P<0.05), while supernatants from CX3CL1-treated macrophages inhibited the proliferation of HREC (P<0.05). HREC migration increased when co-cultured with CCL2-treated macrophages, but decreased with CX3CL1-treated macrophages (P<0.05). CONCLUSION CCL2 and CX3CL1 exert different effects in regulation of macrophage in expression of angiogenesis-related factors, including VEGF-A, THBS-1 and ADAMTS-1. Our findings suggest that CCL2 and CX3CL1 may be candidate proteins for further exploration of novel targets for treatment of ocular neovascularization.
Collapse
Affiliation(s)
- Lei Chen
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Gao-Qin Liu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Hong-Ya Wu
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ji Jin
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Xue Yin
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Dan Li
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Pei-Rong Lu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
206
|
Leszczynska A, Murphy JM. Vascular Calcification: Is it rather a Stem/Progenitor Cells Driven Phenomenon? Front Bioeng Biotechnol 2018; 6:10. [PMID: 29479528 PMCID: PMC5811524 DOI: 10.3389/fbioe.2018.00010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Vascular calcification (VC) has witnessed a surge of interest. Vasculature is virtually an omnipresent organ and has a notably high capacity for repair throughout embryonic and adult life. Of the vascular diseases, atherosclerosis is a leading cause of morbidity and mortality on account of ectopic cartilage and bone formation. Despite the identification of a number of risk factors, all the current theories explaining pathogenesis of VC in atherosclerosis are far from complete. The most widely accepted response to injury theory and smooth muscle transdifferentiation to explain the VC observed in atherosclerosis is being challenged. Recent focus on circulating and resident progenitor cells in the vasculature and their role in atherogenesis and VC has been the driving force behind this review. This review discusses intrinsic cellular players contributing to fate determination of cells and tissues to form ectopic cartilage and bone formation.
Collapse
Affiliation(s)
- Aleksandra Leszczynska
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - J Mary Murphy
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
207
|
Jindatip D, Fujiwara K, Sarachana T, Mutirangura A, Yashiro T. Characteristics of pericytes in diethylstilbestrol (DES)-induced pituitary prolactinoma in rats. Med Mol Morphol 2018; 51:147-155. [PMID: 29344720 DOI: 10.1007/s00795-018-0180-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/09/2018] [Indexed: 12/31/2022]
Abstract
Prolactinomas are the most common tumor of the human pituitary. They result in excessive prolactin secretion and important changes in the vasculature. Pericytes are perivascular cells associated with capillaries and have crucial roles in physiological and pathological neovascularization. We previously reported that pericytes produce type I and III collagens in the anterior pituitary of adult rats. In addition, pituitary pericytes contained well-developed cell organelles and actively synthesized collagens during early postnatal development. However, the characteristics of pericytes in pituitary tumors are unclear. In this study, we used diethylstilbestrol (DES)-treated rats as an animal model of prolactinoma. Using five common pericyte markers, more pericytes were observed in rats treated with DES for 3 months (prolactinoma) compared to the control. Transmission electron microscopy revealed that attached and semidetached pericytes exhibited active cell organelles. Moreover, we identified pericyte migration between capillaries. Although the fine structure of pituitary pericytes was active in prolactinoma, expressions of type I and III collagen mRNAs were greatly diminished. In sum, the characteristics and functions of pericytes were altered in pituitary tumors. This study is the first to clarify fine structural changes of pericytes in rat prolactinomas and improves our understanding of the function of pericytes under pathological conditions.
Collapse
Affiliation(s)
- Depicha Jindatip
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Wangmai, Pathumwan, Bangkok, 10330, Thailand.
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University, School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Ken Fujiwara
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University, School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Tewarit Sarachana
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Rama 1 Rd., Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Apiwat Mutirangura
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Takashi Yashiro
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University, School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
208
|
Wiggins JM, Opoku-Acheampong AB, Baumfalk DR, Siemann DW, Behnke BJ. Exercise and the Tumor Microenvironment: Potential Therapeutic Implications. Exerc Sport Sci Rev 2018; 46:56-64. [PMID: 29166299 DOI: 10.1249/jes.0000000000000137] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
An imbalance in oxygen delivery to demand in solid tumors results in local areas of hypoxia leading to poor prognosis for the patient. We hypothesize that aerobic exercise increases tumor blood flow, recruits previously nonperfused tumor blood vessels, and thereby augments blood-tumor O2 transport and diminishes tumor hypoxia. When combined with conventional anticancer treatments, aerobic exercise can significantly improve the outcomes for several types of cancers.
Collapse
Affiliation(s)
- Jennifer M Wiggins
- Department of Radiation Oncology, College of Medicine, University of Florida.,Department of Radiation Oncology, College of Medicine, University of Florida
| | | | - Dryden R Baumfalk
- Department of Radiation Oncology, College of Medicine, University of Florida
| | - Dietmar W Siemann
- Department of Radiation Oncology, College of Medicine, University of Florida.,Department of Radiation Oncology, College of Medicine, University of Florida
| | - Bradley J Behnke
- Department of Radiation Oncology, College of Medicine, University of Florida.,Department of Radiation Oncology, College of Medicine, University of Florida
| |
Collapse
|
209
|
Abstract
Solid tumors are often characterized by insufficient oxygen supply (hypoxia), as a result of inadequate vascularization, which cannot keep up with the rapid growth rate of the tumor. Tumor hypoxia is a negative prognostic and predictive factor and is associated with a more aggressive phenotype in various tumor entities. Activation of the hypoxic response in tumors, which is centered around the hypoxia-inducible transcription factors (HIFs), has been causally linked to neovascularization, increased radio- and chemoresistance, altered cell metabolism, genomic instability, increased metastatic potential, and tumor stem cell characteristics. Thus, the hypoxic tumor microenvironment represents a main driving force for tumor progression and a potential target for therapeutic interventions. Here, we describe several methods for the analysis of tumor hypoxia and the hypoxic response in vivo in tumor xenograft models. These methods can be applied to various tumor models, including brain tumor xenotransplants, and allow simultaneously determining the extent and distribution of hypoxia within the tumor, analyzing HIF levels by immunohistochemistry and immunoblot, and quantifying the expression of HIF target genes in tumor tissue. The combination of these approaches provides an important tool to assess the role of the hypoxic tumor microenvironment in vivo.
Collapse
|
210
|
Kim J, Kim E, Euceda LR, Meyer DE, Langseth K, Bathen TF, Moestue SA, Huuse EM. Multiparametric characterization of response to anti-angiogenic therapy using USPIO contrast-enhanced MRI in combination with dynamic contrast-enhanced MRI. J Magn Reson Imaging 2017; 47:1589-1600. [DOI: 10.1002/jmri.25898] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/03/2017] [Indexed: 12/28/2022] Open
Affiliation(s)
- Jana Kim
- Department of Circulation and Medical Imaging; NTNU - Norwegian University of Science and Technology; Trondheim Norway
- Department of Radiology and Nuclear Medicine; St. Olavs Hospital, Trondheim University Hospital; Trondheim Norway
| | - Eugene Kim
- Department of Circulation and Medical Imaging; NTNU - Norwegian University of Science and Technology; Trondheim Norway
- Department of Radiology and Nuclear Medicine; St. Olavs Hospital, Trondheim University Hospital; Trondheim Norway
| | - Leslie R. Euceda
- Department of Circulation and Medical Imaging; NTNU - Norwegian University of Science and Technology; Trondheim Norway
| | - Dan E. Meyer
- Biosciences Technology Organization, GE Global Research Center; Niskayuna NY United States
| | | | - Tone F. Bathen
- Department of Circulation and Medical Imaging; NTNU - Norwegian University of Science and Technology; Trondheim Norway
| | - Siver A. Moestue
- Department of Circulation and Medical Imaging; NTNU - Norwegian University of Science and Technology; Trondheim Norway
- Department of Laboratory Medicine, Women's and Children's Health; NTNU - Norwegian University of Science and Technology; Trondheim Norway
| | - Else Marie Huuse
- Department of Circulation and Medical Imaging; NTNU - Norwegian University of Science and Technology; Trondheim Norway
- Department of Radiology and Nuclear Medicine; St. Olavs Hospital, Trondheim University Hospital; Trondheim Norway
| |
Collapse
|
211
|
Evolving Significance and Future Relevance of Anti-Angiogenic Activity of mTOR Inhibitors in Cancer Therapy. Cancers (Basel) 2017; 9:cancers9110152. [PMID: 29104248 PMCID: PMC5704170 DOI: 10.3390/cancers9110152] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
mTOR inhibitors have demonstrated remarkable anti-tumor activity in experimental models, mainly by reducing cancer cell growth and tumor angiogenesis. Their use in cancer patients as monotherapy has, however, generated only limited benefits, increasing median overall survival by only a few months. Likewise, in other targeted therapies, cancer cells develop resistance mechanisms to overcome mTOR inhibition. Hence, novel therapeutic strategies have to be designed to increase the efficacy of mTOR inhibitors in cancer. In this review, we discuss the present and future relevance of mTOR inhibitors in cancer therapy by focusing on their effects on tumor angiogenesis.
Collapse
|
212
|
Overcoming obstacles in the tumor microenvironment: Recent advancements in nanoparticle delivery for cancer theranostics. Biomaterials 2017; 156:217-237. [PMID: 29207323 DOI: 10.1016/j.biomaterials.2017.10.024] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
Despite rapid advancements in the field of nanotechnology, there is mounting frustration in the scientific community regarding the translational impact of nanomedicine. Modest therapeutic performance of FDA-approved nanomedicines combined with multiple disappointing clinical trials (such as phase III HEAT trial) have raised questions about the future of nanomedicine. Encouraging breakthroughs, however, have been made in the last few years towards the development of new classes of nanoparticles that can respond to tumor microenvironmental conditions and successfully deliver therapeutic agents to cancer cells. Concurrently, a great deal of effort has also been devoted to alter various parameters of tumor pathophysiology to pre-treat tumors before nanoparticles are administered. Such 'priming' treatments improve access of the systemically administered agents to the tumor and promote drug penetration into the deeper layers of tumor tissue. This review will highlight recent advances in cancer nanomedicine exploiting both nanoparticle design and tumor microenvironment modification; and provide a critical perspective on the future development of nanomedicine delivery in oncology.
Collapse
|
213
|
Kropski JA, Richmond BW, Gaskill CF, Foronjy RF, Majka SM. Deregulated angiogenesis in chronic lung diseases: a possible role for lung mesenchymal progenitor cells (2017 Grover Conference Series). Pulm Circ 2017; 8:2045893217739807. [PMID: 29040010 PMCID: PMC5731726 DOI: 10.1177/2045893217739807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic lung disease (CLD), including pulmonary fibrosis (PF) and chronic obstructive pulmonary disease (COPD), is the fourth leading cause of mortality worldwide. Both are debilitating pathologies that impede overall tissue function. A common co-morbidity in CLD is vasculopathy, characterized by deregulated angiogenesis, remodeling, and loss of microvessels. This substantially worsens prognosis and limits survival, with most current therapeutic strategies being largely palliative. The relevance of angiogenesis, both capillary and lymph, to the pathophysiology of CLD has not been resolved as conflicting evidence depicts angiogenesis as both reparative or pathologic. Therefore, we must begin to understand and model the underlying pathobiology of pulmonary vascular deregulation, alone and in response to injury induced disease, to define cell interactions necessary to maintain normal function and promote repair. Capillary and lymphangiogenesis are deregulated in both PF and COPD, although the mechanisms by which they co-regulate and underlie early pathogenesis of disease are unknown. The cell-specific mechanisms that regulate lung vascular homeostasis, repair, and remodeling represent a significant gap in knowledge, which presents an opportunity to develop targeted therapies. We have shown that that ABCG2pos multipotent adult mesenchymal stem or progenitor cells (MPC) influence the function of the capillary microvasculature as well as lymphangiogenesis. A balance of both is required for normal tissue homeostasis and repair. Our current models suggest that when lymph and capillary angiogenesis are out of balance, the non-equivalence appears to support the progression of disease and tissue remodeling. The angiogenic regulatory mechanisms underlying CLD likely impact other interstitial lung diseases, tuberous sclerosis, and lymphangioleiomyomatosis.
Collapse
Affiliation(s)
- Jonathan A Kropski
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bradley W Richmond
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christa F Gaskill
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert F Foronjy
- 3 5718 Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Susan M Majka
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,2 74498 Department of Medicine, Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
214
|
Vilalta M, Hughes NP, Von Eyben R, Giaccia AJ, Graves EE. Patterns of Vasculature in Mouse Models of Lung Cancer Are Dependent on Location. Mol Imaging Biol 2017; 19:215-224. [PMID: 27709411 DOI: 10.1007/s11307-016-1010-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Preclinical studies of hypoxia are generally done using ectopic xenograft tumors, which behave differently from human tumors. Our previous findings have shown that subcutaneously implanted lung tumors exhibit more hypoxia than their orthotopic implanted or spontaneous K-ras-induced counterparts. We hypothesize that differences in hypoxia are due to site-specific differences in vascularity and perfusion. PROCEDURES To compare the presence and functionality of vessels in these tumor models, we studied vascular perfusion in vivo in real time. RESULTS Orthotopically implanted and spontaneous K-ras-induced lung tumors showed elevated perfusion, demonstrating vasculature functionality. Little contrast agent uptake was observed within the subcutaneously implanted tumors, indicating vascular dysfunction. These findings were corroborated at the microscopic level with Hoechst 33342 and Meca-32 staining. CONCLUSIONS From these observations, we concluded that differences in hypoxia in experimental models is related to vessel perfusion. Thus, appropriate selection of preclinical lung tumor models is essential for the study of hypoxia, angiogenesis and therapies targeting these phenomena.
Collapse
Affiliation(s)
- Marta Vilalta
- Department of Radiation Oncology, Molecular Imaging Program at Stanford and Bio-X Program, Stanford University, Stanford, CA, USA
| | - Nicholas P Hughes
- Department of Radiation Oncology, Molecular Imaging Program at Stanford and Bio-X Program, Stanford University, Stanford, CA, USA
| | - Rie Von Eyben
- Department of Radiation Oncology, Molecular Imaging Program at Stanford and Bio-X Program, Stanford University, Stanford, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Molecular Imaging Program at Stanford and Bio-X Program, Stanford University, Stanford, CA, USA
| | - Edward E Graves
- Department of Radiation Oncology, Molecular Imaging Program at Stanford and Bio-X Program, Stanford University, Stanford, CA, USA.
| |
Collapse
|
215
|
|
216
|
Almeida VM, Paiva AE, Sena IFG, Mintz A, Magno LAV, Birbrair A. Pericytes Make Spinal Cord Breathless after Injury. Neuroscientist 2017; 24:440-447. [PMID: 29283016 DOI: 10.1177/1073858417731522] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Traumatic spinal cord injury is a devastating condition that leads to significant neurological deficits and reduced quality of life. Therapeutic interventions after spinal cord lesions are designed to address multiple aspects of the secondary damage. However, the lack of detailed knowledge about the cellular and molecular changes that occur after spinal cord injury restricts the design of effective treatments. Li and colleagues using a rat model of spinal cord injury and in vivo microscopy reveal that pericytes play a key role in the regulation of capillary tone and blood flow in the spinal cord below the site of the lesion. Strikingly, inhibition of specific proteins expressed by pericytes after spinal cord injury diminished hypoxia and improved motor function and locomotion of the injured rats. This work highlights a novel central cellular population that might be pharmacologically targeted in patients with spinal cord trauma. The emerging knowledge from this research may provide new approaches for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Viviani M Almeida
- 1 Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- 1 Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F G Sena
- 1 Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- 2 Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Luiz Alexandre V Magno
- 3 Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- 1 Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,4 Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.,5 Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
217
|
Murgai M, Ju W, Eason M, Kline J, Beury DW, Kaczanowska S, Miettinen MM, Kruhlak M, Lei H, Shern JF, Cherepanova OA, Owens GK, Kaplan RN. KLF4-dependent perivascular cell plasticity mediates pre-metastatic niche formation and metastasis. Nat Med 2017; 23:1176-1190. [PMID: 28920957 PMCID: PMC5724390 DOI: 10.1038/nm.4400] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 08/10/2017] [Indexed: 12/13/2022]
Abstract
A deeper understanding of the metastatic process is required for the development of new therapies that improve patient survival. Metastatic tumor cell growth and survival in distant organs is facilitated by the formation of a pre-metastatic niche composed of hematopoietic cells, stromal cells, and extracellular matrix (ECM). Perivascular cells, including vascular smooth muscle cells (vSMCs) and pericytes, are involved in new vessel formation and in promoting stem cell maintenance and proliferation. Given the well-described plasticity of perivascular cells, we hypothesize that perivascular cells similarly regulate tumor cell fate at metastatic sites. Using perivascular cell-specific and pericyte-specific lineage-tracing models, we trace the fate of perivascular cells in the pre-metastatic and metastatic microenvironments. We show that perivascular cells lose the expression of traditional vSMC/pericyte markers in response to tumor-secreted factors and exhibit increased proliferation, migration, and ECM synthesis. Increased expression of the pluripotency gene Klf4 in these phenotypically-switched perivascular cells promotes a less differentiated state characterized by enhanced ECM production that establishes a pro-metastatic fibronectin-rich environment. Genetic inactivation of Klf4 in perivascular cells decreases pre-metastatic niche formation and metastasis. Our data reveal a previously unidentified role for perivascular cells in pre-metastatic niche formation and uncover novel strategies for limiting metastasis.
Collapse
Affiliation(s)
- Meera Murgai
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Wei Ju
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Matthew Eason
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jessica Kline
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Daniel W Beury
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sabina Kaczanowska
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Markku M Miettinen
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Kruhlak
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jack F Shern
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Olga A Cherepanova
- Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
218
|
Increased PD-L1 expression and IL-6 secretion characterize human lung tumor-derived perivascular-like cells that promote vascular leakage in a perfusable microvasculature model. Sci Rep 2017; 7:10636. [PMID: 28878242 PMCID: PMC5587684 DOI: 10.1038/s41598-017-09928-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 08/01/2017] [Indexed: 12/13/2022] Open
Abstract
Pericytes represent important support cells surrounding microvessels found in solid organs. Emerging evidence points to their involvement in tumor progression and metastasis. Although reported to be present in the human lung, their specific presence and functional orientation within the tumor microenvironment in non-small cell lung cancer (NSCLC) has not yet been adequately studied. Using a multiparameter approach, we prospectively identified, sorted and expanded mesenchymal cells from human primary NSCLC samples based on co-expression of CD73 and CD90 while lacking hematopoietic and endothelial lineage markers (CD45, CD31, CD14 and Gly-A) and the epithelial marker EpCAM. Compared to their normal counterpart, tumor-derived Lineage-EpCAM-CD73+CD90+ cells showed enhanced expression of the immunosuppressive ligand PD-L1, a higher constitutive secretion of IL-6 and increased basal αSMA levels. In an in vitro model of 3D microvessels, both tumor-derived and matched normal Lineage-EpCAM-CD73+CD90+ cells supported the assembly of perfusable vessels. However, tumor-derived Lineage-EpCAM-CD73+CD90+ cells led to the formation of vessels with significantly increased permeability. Together, our data show that perivascular-like cells present in NSCLC retain functional abnormalities in vitro. Perivascular-like cells as an eventual target in NSCLC warrants further investigation.
Collapse
|
219
|
Willrodt AH, Beffinger M, Vranova M, Protsyuk D, Schuler K, Jadhav M, Heikenwalder M, van den Broek M, Borsig L, Halin C. Stromal Expression of Activated Leukocyte Cell Adhesion Molecule Promotes Lung Tumor Growth and Metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2558-2569. [PMID: 28822802 DOI: 10.1016/j.ajpath.2017.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/28/2017] [Accepted: 07/26/2017] [Indexed: 01/07/2023]
Abstract
Activated leukocyte cell adhesion molecule (ALCAM) is expressed on various cell types, including leukocytes, endothelial cells, and certain tumor cells. Although ALCAM expression on tumor cells has been linked to tumor invasion and metastatic spread, the contribution of ALCAM expressed in cells forming the tumor stroma to cancer progression has not been investigated. In this study, ALCAM-deficient (ALCAM-/-) mice were used to evaluate the role of ALCAM in lung tumor growth and metastasis. ALCAM-/- mice displayed an altered blood vascular network in the lung and the diaphragm, indicative of an angiogenetic defect. The absence of ALCAM expression in cells forming the stromal tumor microenvironment profoundly affected lung tumor growth in three different i.v. metastasis models. In the case of Lewis lung carcinoma (LLC), an additional defect in tumor cell homing to the lungs and a resulting reduction in the number of lung tumor nodules were observed. Similarly, when LLC cells were implanted subcutaneously for the study of spontaneous tumor cell metastasis, the rate of LLC metastasis to the lungs was profoundly reduced in ALCAM-/- mice. Taken together, our work demonstrates for the first time the in vivo contribution of ALCAM to angiogenesis and reveals a novel role of stromally expressed ALCAM in supporting tumor growth and metastatic spread.
Collapse
Affiliation(s)
- Ann-Helen Willrodt
- Institute of Pharmaceutical Sciences, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland
| | - Michal Beffinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Martina Vranova
- Institute of Pharmaceutical Sciences, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland
| | - Darya Protsyuk
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Katja Schuler
- Institute of Pharmaceutical Sciences, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland
| | - Maria Jadhav
- Institute of Pharmaceutical Sciences, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | | | - Lubor Borsig
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland.
| |
Collapse
|
220
|
Just J, Lykkemark S, Nielsen CH, Roshenas AR, Drasbek KR, Petersen SV, Bek T, Kristensen P. Pericyte modulation by a functional antibody obtained by a novel single-cell selection strategy. Microcirculation 2017; 24. [DOI: 10.1111/micc.12365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/21/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Jesper Just
- Department of Molecular Biology and Genetics; Aarhus University; Aarhus C Denmark
- Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
| | - Simon Lykkemark
- Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
- Sino-Danish Centre for Education and Research (SDC); Aarhus C Denmark
| | - Charlotte H. Nielsen
- Department of Molecular Biology and Genetics; Aarhus University; Aarhus C Denmark
| | - Ali R. Roshenas
- Department of Engineering; Aarhus University; Aarhus C Denmark
| | - Kim R. Drasbek
- Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
| | | | - Toke Bek
- Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
| | | |
Collapse
|
221
|
Mondino A, Vella G, Icardi L. Targeting the tumor and its associated stroma: One and one can make three in adoptive T cell therapy of solid tumors. Cytokine Growth Factor Rev 2017. [DOI: 10.1016/j.cytogfr.2017.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
222
|
Abstract
Tumours display considerable variation in the patterning and properties of angiogenic blood vessels, as well as in their responses to anti-angiogenic therapy. Angiogenic programming of neoplastic tissue is a multidimensional process regulated by cancer cells in concert with a variety of tumour-associated stromal cells and their bioactive products, which encompass cytokines and growth factors, the extracellular matrix and secreted microvesicles. In this Review, we discuss the extrinsic regulation of angiogenesis by the tumour microenvironment, highlighting potential vulnerabilities that could be targeted to improve the applicability and reach of anti-angiogenic cancer therapies.
Collapse
Affiliation(s)
- Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Daniela Biziato
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Division of Experimental Pathology, University of Lausanne and University of Lausanne Hospital, 1066 Lausanne, Switzerland
| |
Collapse
|
223
|
Lu J, Shenoy AK. Epithelial-to-Pericyte Transition in Cancer. Cancers (Basel) 2017; 9:cancers9070077. [PMID: 28677655 PMCID: PMC5532613 DOI: 10.3390/cancers9070077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/23/2017] [Accepted: 06/30/2017] [Indexed: 01/05/2023] Open
Abstract
During epithelial-to-mesenchymal transition (EMT), cells lose epithelial characteristics and acquire mesenchymal properties. These two processes are genetically separable and governed by distinct transcriptional programs, rendering the EMT outputs highly heterogeneous. Our recent study shows that the mesenchymal products generated by EMT often express multiple pericyte markers, associate with and stabilize blood vessels to fuel tumor growth, thus phenotypically and functionally resembling pericytes. Therefore, some EMT events represent epithelial-to-pericyte transition (EPT). The serum response factor (SRF) plays key roles in both EMT and differentiation of pericytes, and may inherently confer the pericyte attributes on EMT cancer cells. By impacting their intratumoral location and cell surface receptor expression, EPT may enable cancer cells to receive and respond to angiocrine factors produced by the vascular niche, and develop therapy resistance.
Collapse
Affiliation(s)
- Jianrong Lu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610-3633, USA.
| | - Anitha K Shenoy
- Department of Pharmaceutics and Biomedical Sciences, California Health Sciences University, Clovis, CA 93612, USA.
| |
Collapse
|
224
|
Fine-Tuning Tumor Endothelial Cells to Selectively Kill Cancer. Int J Mol Sci 2017; 18:ijms18071401. [PMID: 28665313 PMCID: PMC5535894 DOI: 10.3390/ijms18071401] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/25/2017] [Accepted: 06/26/2017] [Indexed: 01/13/2023] Open
Abstract
Tumor endothelial cells regulate several aspects of tumor biology, from delivering oxygen and nutrients to shaping the immune response against a tumor and providing a barrier against tumor cell dissemination. Accordingly, targeting tumor endothelial cells represents an important modality in cancer therapy. Whereas initial anti-angiogenic treatments focused mainly on blocking the formation of new blood vessels in cancer, emerging strategies are specifically influencing certain aspects of tumor endothelial cells. For instance, efforts are generated to normalize tumor blood vessels in order to improve tumor perfusion and ameliorate the outcome of chemo-, radio-, and immunotherapy. In addition, treatment options that enhance the properties of tumor blood vessels that support a host’s anti-tumor immune response are being explored. Hence, upcoming anti-angiogenic strategies will shape some specific aspects of the tumor blood vessels that are no longer limited to abrogating angiogenesis. In this review, we enumerate approaches that target tumor endothelial cells to provide anti-cancer benefits and discuss their therapeutic potential.
Collapse
|
225
|
Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 2017; 20:409-426. [PMID: 28660302 DOI: 10.1007/s10456-017-9562-9] [Citation(s) in RCA: 951] [Impact Index Per Article: 135.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/21/2017] [Indexed: 12/27/2022]
Abstract
Tumor blood vessels are a key target for cancer therapeutic management. Tumor cells secrete high levels of pro-angiogenic factors which contribute to the creation of an abnormal vascular network characterized by disorganized, immature and permeable blood vessels, resulting in poorly perfused tumors. The hypoxic microenvironment created by impaired tumor perfusion can promote the selection of more invasive and aggressive tumor cells and can also impede the tumor-killing action of immune cells. Furthermore, abnormal tumor perfusion also reduces the diffusion of chemotherapeutic drugs and radiotherapy efficiency. To fight against this defective phenotype, the normalization of the tumor vasculature has emerged as a new therapeutic strategy. Vascular normalization, by restoring proper tumor perfusion and oxygenation, could limit tumor cell invasiveness and improve the effectiveness of anticancer treatments. In this review, we investigate the mechanisms involved in tumor angiogenesis and describe strategies used to achieve vascular normalization.
Collapse
|
226
|
Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V, Feyen K, Tawney J, Hanahan D, Michael IP, Bergers G. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med 2017; 9:9/385/eaak9679. [PMID: 28404866 DOI: 10.1126/scitranslmed.aak9679] [Citation(s) in RCA: 549] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/20/2017] [Indexed: 12/13/2022]
Abstract
Inhibitors of VEGF (vascular endothelial growth factor)/VEGFR2 (vascular endothelial growth factor receptor 2) are commonly used in the clinic, but their beneficial effects are only observed in a subset of patients and limited by induction of diverse relapse mechanisms. We describe the up-regulation of an adaptive immunosuppressive pathway during antiangiogenic therapy, by which PD-L1 (programmed cell death ligand 1), the ligand of the negative immune checkpoint regulator PD-1 (programmed cell death protein 1), is enhanced by interferon-γ-expressing T cells in distinct intratumoral cell types in refractory pancreatic, breast, and brain tumor mouse models. Successful treatment with a combination of anti-VEGFR2 and anti-PD-L1 antibodies induced high endothelial venules (HEVs) in PyMT (polyoma middle T oncoprotein) breast cancer and RT2-PNET (Rip1-Tag2 pancreatic neuroendocrine tumors), but not in glioblastoma (GBM). These HEVs promoted lymphocyte infiltration and activity through activation of lymphotoxin β receptor (LTβR) signaling. Further activation of LTβR signaling in tumor vessels using an agonistic antibody enhanced HEV formation, immunity, and subsequent apoptosis and necrosis in pancreatic and mammary tumors. Finally, LTβR agonists induced HEVs in recalcitrant GBM, enhanced cytotoxic T cell (CTL) activity, and thereby sensitized tumors to antiangiogenic/anti-PD-L1 therapy. Together, our preclinical studies provide evidence that anti-PD-L1 therapy can sensitize tumors to antiangiogenic therapy and prolong its efficacy, and conversely, antiangiogenic therapy can improve anti-PD-L1 treatment specifically when it generates intratumoral HEVs that facilitate enhanced CTL infiltration, activity, and tumor cell destruction.
Collapse
Affiliation(s)
- Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Arnaud Jabouille
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lee B Rivera
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Inge Lodewijckx
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Rindert Missiaen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Veronica Steri
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin Feyen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Jaime Tawney
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Iacovos P Michael
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium. .,Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
227
|
Hosono J, Morikawa S, Ezaki T, Kawamata T, Okada Y. Pericytes promote abnormal tumor angiogenesis in a rat RG2 glioma model. Brain Tumor Pathol 2017. [PMID: 28646266 DOI: 10.1007/s10014-017-0291-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In glioma angiogenesis, tumor vessels cause morphological and functional abnormalities associated with malignancy and tumor progression. We hypothesized that certain structural changes or scantiness of functional pericytes may be involved in the formation of dysfunctional blood vessels in gliomas. In this study, we performed morphological examinations to elucidate the possible involvement of pericytes in brain tumor vessel abnormalities using a rat RG2 glioma model. After implantation of RG2 glioma cells in the syngeneic rat brain, gliomas were formed as early as day 7. In immunohistochemical examinations, desmin-positive pericytes, characterized by morphological abnormalities, were abundantly found on leaky vessels, as assessed by extravasation of lectin and high-molecular-weight dextrans. Interestingly, desmin-positive pericytes seemed to be characteristic of gliomas in rats. These pericytes were also found to express heat-shock protein 47, which plays an important role in the formation of the basement membrane, suggesting that RG2 pericytes promoted angiogenesis by producing basement membrane as a scaffold for newly forming blood vessels and caused functional abnormalities. We concluded that RG2 pericytes may be responsible for abnormal tumor angiogenesis lacking the functional ability to maintain the blood-brain barrier.
Collapse
Affiliation(s)
- Junji Hosono
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan.,Department of Anatomy and Developmental Biology, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yoshikazu Okada
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1, Kawada-cho Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
228
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
229
|
Zhan K, Bai L, Wu Q, Lei D, Wang G. Fractal characteristics of the microvascular network: A useful index to assess vascularization level of porous silk fibroin biomaterial. J Biomed Mater Res A 2017; 105:2276-2290. [PMID: 28445607 DOI: 10.1002/jbm.a.36094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 04/05/2017] [Accepted: 04/20/2017] [Indexed: 02/04/2023]
Abstract
The neovascularization of biomaterials for tissue engineering is not only related to growth of capillaries but also involves appropriate hierarchy distribution of the microvessels. In this study, we proposed hierarchy distribution contrast method which can assess vascular transport capacity, in order to examine the hierarchy distribution of the neovessels during vascularization of the porous silk fibroin biomaterials implanted into rats and its evolution. The results showed that the fractal characteristics appeared toward the end of the vascularization stages, and the structure of the microvascular network after 3 weeks of implantation was similar to the fractal microvascular tree with bifurcation exponent x = 3 and fractal dimension D = 1.46, which became a sign of maturation of the regenerative vasculature. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2276-2290, 2017.
Collapse
Affiliation(s)
- Kuihua Zhan
- School of Mechanical and Electric Engineering, Soochow University, 178 Gan Jiang East Road, Suzhou, 215006, China.,College of Textile and Clothing Engineering, Soochow University, 178 Gan Jiang East Road, Suzhou, 215006, China
| | - Lun Bai
- College of Textile and Clothing Engineering, Soochow University, 178 Gan Jiang East Road, Suzhou, 215006, China
| | - Qinqin Wu
- School of Mechanical and Electric Engineering, Soochow University, 178 Gan Jiang East Road, Suzhou, 215006, China
| | - Derong Lei
- School of Mechanical and Electric Engineering, Soochow University, 178 Gan Jiang East Road, Suzhou, 215006, China
| | - Guangqian Wang
- College of Textile and Clothing Engineering, Soochow University, 178 Gan Jiang East Road, Suzhou, 215006, China
| |
Collapse
|
230
|
Salimi Sartakhti J, Manshaei MH, Basanta D, Sadeghi M. Evolutionary emergence of angiogenesis in avascular tumors using a spatial public goods game. PLoS One 2017; 12:e0175063. [PMID: 28399181 PMCID: PMC5388338 DOI: 10.1371/journal.pone.0175063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/20/2017] [Indexed: 11/18/2022] Open
Abstract
Natural selection in cancer often results in the emergence of increasingly malignant tumor cells that display many if not all of the hallmarks of cancer. One of the most important traits acquired during cancer progression is angiogenesis. Tumor cells capable of secreting pro-angiogenic factors can be seen as cooperators where the improved oxygenation, nutrient delivery and waste disposal resulting from angiogenesis could be seen as a public good. Under this view, the relatively costly secretion of molecular signals required to orchestrate angiogenesis would be undertaken exclusively by cooperating tumor cells but the benefits of angiogenesis would be felt by neighboring tumor cells regardless of their contribution to the process. In this work we detail a mathematical model to better understand how clones capable of secreting pro-angiogenic factors can emerge in a tumor made of non-cooperative tumor cells. Given the importance of the spatial configuration of the tumor in determining the efficacy of the secretion of pro-angiogenic factors as well as the benefits of angiogenesis we have developed a spatial game theoretic approach where interactions and public good diffusion are described by graphs. The results show that structure of the population affects the evolutionary dynamics of the pro-angiogenic clone. Specifically, when the benefit of angiogenesis is represented by sigmoid function with regards to the number of pro-angiogenic clones then the probability of the coexistence of pro-angiogenic and angiogenesis-neutral clones increases. Our results demonstrate that pro-angiogenic clone equilibrates into clusters that appear from surrounding vascular tissues towards the center of tumor. These clusters appear notably less dense after anti-angiogenic therapy.
Collapse
Affiliation(s)
- Javad Salimi Sartakhti
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Mohammad Hossein Manshaei
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
- * E-mail:
| | - David Basanta
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States of America
| | - Mehdi Sadeghi
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran
| |
Collapse
|
231
|
Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V, Feyen K, Tawney J, Hanahan D, Michael IP, Bergers G. Combined antiangiogenic and anti–PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med 2017. [DOI: 10.1126/scitranslmed.aak9679 pmid: 28404866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Arnaud Jabouille
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lee B. Rivera
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Inge Lodewijckx
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Rindert Missiaen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Veronica Steri
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin Feyen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Jaime Tawney
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Iacovos P. Michael
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB-Center for Cancer Biology, Department of Oncology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Brain Tumor Research Center, Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
232
|
van Hell AJ, Haimovitz-Friedman A, Fuks Z, Tap WD, Kolesnick R. Gemcitabine kills proliferating endothelial cells exclusively via acid sphingomyelinase activation. Cell Signal 2017; 34:86-91. [PMID: 28238856 DOI: 10.1016/j.cellsig.2017.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/15/2017] [Accepted: 02/22/2017] [Indexed: 12/20/2022]
Abstract
Gemcitabine is a widely-used anti-cancer drug with a well-defined mechanism of action in normal and transformed epithelial cells. However, its effect on endothelial cells is largely unknown. Acid sphingomyelinase (ASMase) is highly expressed in endothelial cells, converting plasma membrane sphingomyelin to pro-apoptotic ceramide upon activation by diverse stresses. In the current study, we investigated gemcitabine impact in primary cultures of endothelial cells. We find baseline ASMase increases markedly in bovine aortic endothelial cells (BAEC) as they transit from a proliferative to a confluent growth-arrested state. Further, gemcitabine activates ASMase and induces release of a secretory ASMase form into the media only in proliferating endothelial cells. Additionally, proliferative, but not growth-arrested BAEC, are sensitive to gemcitabine-induced apoptotic death, an effect blocked by inhibiting ASMase with imipramine or by binding ceramide on the cell surface with an anti-ceramide Ab. Confluent growth-arrested BAEC can be re-sensitized to gemcitabine-induced apoptosis by provision of exogenous sphingomyelinase. A highly similar phenotype was observed in primary cultures of human coronary artery endothelial cells. These findings reveal a previously-unrecognized mechanism of gemcitabine cytotoxicity in endothelium that may well contribute to its clinical benefit, and suggest the potential for further improvement of its clinical efficacy via pharmacologic modulation of ASMase/ceramide signaling in proliferative tumor endothelium.
Collapse
Affiliation(s)
- Albert J van Hell
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | - Zvi Fuks
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Richard Kolesnick
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
233
|
Pautu V, Leonetti D, Lepeltier E, Clere N, Passirani C. Nanomedicine as a potent strategy in melanoma tumor microenvironment. Pharmacol Res 2017; 126:31-53. [PMID: 28223185 DOI: 10.1016/j.phrs.2017.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 12/19/2022]
Abstract
Melanoma originated from melanocytes is the most aggressive type of skin cancer. Despite considerable progresses in clinical treatment with the discovery of BRAF or MEK inhibitors and monoclonal antibodies, the durability of response to treatment is often limited to the development of acquired resistance and systemic toxicity. The limited success of conventional treatment highlights the importance of understanding the role of melanoma tumor microenvironment in tumor developement and drug resistance. Nanoparticles represent a promising strategy for the development of new cancer treatments able to improve the bioavailability of drugs and increase their penetration by targeting specifically tumors cells and/or tumor environment. In this review, we will discuss the main influence of tumor microenvironment in melanoma growth and treatment outcome. Furthermore, third generation loaded nanotechnologies represent an exciting tool for detection, treatment, and escape from possible mechanism of resistance mediated by tumor microenvironment, and will be highlighted in this review.
Collapse
Affiliation(s)
- Vincent Pautu
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | | | - Elise Lepeltier
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | - Nicolas Clere
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | - Catherine Passirani
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| |
Collapse
|
234
|
Fabian KPL, Chi-Sabins N, Taylor JL, Fecek R, Weinstein A, Storkus WJ. Therapeutic efficacy of combined vaccination against tumor pericyte-associated antigens DLK1 and DLK2 in mice. Oncoimmunology 2017; 6:e1290035. [PMID: 28405524 DOI: 10.1080/2162402x.2017.1290035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/25/2017] [Accepted: 01/26/2017] [Indexed: 12/28/2022] Open
Abstract
When compared with vascular cells in normal tissues, pericytes and vascular endothelial cells (VEC) in tumor blood vessels exhibit altered morphology and epigenetic programming that leads to the expression of unique antigens that allow for differential recognition by CD8+ T cells. We have previously shown that the Notch antagonist delta-like homolog 1 (DLK1) is a tumor pericyte-associated antigen expressed in setting of melanoma and a range of carcinomas. In this report, we show that therapeutic vaccination against DLK1 in murine models results in slowed tumor growth, but also to the compensatory expression of the DLK1 homolog, DLK2, by tumor-associated pericytes. Vaccines targeting both DLK1 and DLK2 resulted in superior antitumor benefits in association with improved activation and recruitment of antigen-specific Type 1 CD8+ T cells, reduced presence of myeloid-derived suppressive cells, T regulatory cell and tumor vascular normalization. The antitumor efficacy of vaccines coordinately targeting DLK1 and DLK2 was further improved by inclusion of PD-L1 blockade, thus defining a combination immunotherapy theoretically suitable for the treatment of a broad range of solid (vascularized) cancers.
Collapse
Affiliation(s)
- Kellsye Paula L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Nina Chi-Sabins
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Ronald Fecek
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Aliyah Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
235
|
Martinovic V, Vukusic Pusic T, Restovic I, Bocina I, Filipovic N, Saraga-Babic M, Vukojevic K. Expression of Epithelial and Mesenchymal Differentiation Markers in the Early Human Gonadal Development. Anat Rec (Hoboken) 2017; 300:1315-1326. [PMID: 27981799 DOI: 10.1002/ar.23531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 07/23/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022]
Abstract
Expressions of cytokeratin 8 (CK8), vimentin, nestin, and alpha-smooth-muscle-actin (alpha-SMA) were analyzed in the developing gonads of 12, 5-9 week old (W) human conceptuses by immunohistochemistry and immunofluorescence. During the investigated period, the number of CK8 positive cells increased from 56% to 92% in the gonadal surface epithelium, from 50% to 60% in the stroma, and from 23% to 42% in the medulla. In the early fetal period, the cell expression of CK8 increased in all gonadal parts, whereas primordial germ cells (PGC) remained negative. The expression of vimentin increased in the gonad stroma (gs) from 73% to 88%, and in the surface epithelium from 18% to 97% until ninth W. The medulla had the highest expression of vimentin in the seventh to eighth W (93%). Vimentin and CK8 colocalized in the somatic cells, while some PGCs showed vimentin expression only. Initially, nestin was positive in the gonad surface epithelium (8%) and stroma (52%), however during further development it decreased to 1% and 33%, respectively. In the early fetal period, the nestin positive cells decreased from 44% to 31% in the gonad medulla. Alpha-SMA was positive only in the blood vessels and mesonephros. The described pattern of expression of intermediate filaments (IF) in developing human gonads suggests their role in the control of PGC apoptosis, early differentiation of gs cells and cell migration. Both epithelial and mesenchymal origins of follicular cells and possible epithelial-to-mesenchymal transition of somatic cells is proposed. Lastly, IF intensity expression varies depending on the cell type and developmental period analyzed. Anat Rec, 300:1315-1326, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vlatka Martinovic
- Department of Pediatric Surgery, University Hospital Mostar, Bosnia and Herzegovina
| | | | | | - Ivana Bocina
- Faculty of Science, University of Split, Croatia
| | - Natalija Filipovic
- Laboratory for Neurocardiology, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Croatia.,Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Croatia
| | - Mirna Saraga-Babic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Croatia
| | - Katarina Vukojevic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Croatia.,Department of Histology and Embryology, School of Medicine, University of Mostar, Bosnia and Herzegovina
| |
Collapse
|
236
|
Tahara Y, Yoshikawa T, Sato H, Mori Y, Zahangir MH, Kishimura A, Mori T, Katayama Y. Encapsulation of a nitric oxide donor into a liposome to boost the enhanced permeation and retention (EPR) effect. MEDCHEMCOMM 2017; 8:415-421. [PMID: 30108759 PMCID: PMC6072363 DOI: 10.1039/c6md00614k] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 12/12/2016] [Indexed: 02/05/2023]
Abstract
We propose a method to improve the enhanced permeability and retention (EPR) effect of nanomedicines based on tumor-specific vasodilation using a nitric oxide (NO) donor-containing liposome. NONOate, a typical NO donor, was incorporated into a PEGylated liposome to retard the protonation-induced release of NO from NONOate by the protecting lipid bilayer membrane. The NONOate-containing liposome (NONOate-LP) showed similar blood retention to an empty PEGylated liposome but almost twice the amount accumulated within the tumor. This improvement in the EPR effect is thought to have been caused by specific vasodilation in the tumor tissue by NO released from the NONOate-LP accumulated in the tumor. The improved EPR effect by NONOate-LP will be useful for the accumulation of co-administered nanomedicines.
Collapse
Affiliation(s)
- Yu Tahara
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Takuma Yoshikawa
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
| | - Hikari Sato
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Yukina Mori
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Md Hosain Zahangir
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
- International Research Center for Molecular Systems , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
- International Research Center for Molecular Systems , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
- Center for Advanced Medical Innovation , Kyushu University , 744 Motooka , Nishi-Ku , Fukuoka 819-0395 , Japan
- Department of Biomedical Engineering , Chung Yuan Christian University , 200 Chung Pei Rd. , Chung Li , Taiwan , 32023 ROC
| |
Collapse
|
237
|
Jain M, Gamage NDH, Alsulami M, Shankar A, Achyut BR, Angara K, Rashid MH, Iskander A, Borin TF, Wenbo Z, Ara R, Ali MM, Lebedyeva I, Chwang WB, Guo A, Bagher-Ebadian H, Arbab AS. Intravenous Formulation of HET0016 Decreased Human Glioblastoma Growth and Implicated Survival Benefit in Rat Xenograft Models. Sci Rep 2017; 7:41809. [PMID: 28139732 PMCID: PMC5282583 DOI: 10.1038/srep41809] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/28/2016] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma (GBM) is a hypervascular primary brain tumor with poor prognosis. HET0016 is a selective CYP450 inhibitor, which has been shown to inhibit angiogenesis and tumor growth. Therefore, to explore novel treatments, we have generated an improved intravenous (IV) formulation of HET0016 with HPßCD and tested in animal models of human and syngeneic GBM. Administration of a single IV dose resulted in 7-fold higher levels of HET0016 in plasma and 3.6-fold higher levels in tumor at 60 min than that in IP route. IV treatment with HPßCD-HET0016 decreased tumor growth, and altered vascular kinetics in early and late treatment groups (p < 0.05). Similar growth inhibition was observed in syngeneic GL261 GBM (p < 0.05). Survival studies using patient derived xenografts of GBM811, showed prolonged survival to 26 weeks in animals treated with focal radiation, in combination with HET0016 and TMZ (p < 0.05). We observed reduced expression of markers of cell proliferation (Ki-67), decreased neovascularization (laminin and αSMA), in addition to inflammation and angiogenesis markers in the treatment group (p < 0.05). Our results indicate that HPßCD-HET0016 is effective in inhibiting tumor growth through decreasing proliferation, and neovascularization. Furthermore, HPßCD-HET0016 significantly prolonged survival in PDX GBM811 model.
Collapse
Affiliation(s)
- Meenu Jain
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | | | - Meshal Alsulami
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Adarsh Shankar
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Bhagelu R. Achyut
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Kartik Angara
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Mohammad H. Rashid
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Asm Iskander
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Thaiz F. Borin
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Zhi Wenbo
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
| | - Roxan Ara
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Meser M. Ali
- Cellular and Molecular Imaging Laboratory, Henry Ford Health System, Detroit, MI, USA
| | - Iryna Lebedyeva
- Department of Chemistry and Physics, Augusta University, Augusta, GA, USA
| | - Wilson B. Chwang
- Cellular and Molecular Imaging Laboratory, Henry Ford Health System, Detroit, MI, USA
| | - Austin Guo
- Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Hassan Bagher-Ebadian
- Cellular and Molecular Imaging Laboratory, Henry Ford Health System, Detroit, MI, USA
| | - Ali S. Arbab
- Tumor Angiogenesis Laboratory, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
238
|
Brand C, Schliemann C, Ring J, Kessler T, Bäumer S, Angenendt L, Mantke V, Ross R, Hintelmann H, Spieker T, Wardelmann E, Mesters RM, Berdel WE, Schwöppe C. NG2 proteoglycan as a pericyte target for anticancer therapy by tumor vessel infarction with retargeted tissue factor. Oncotarget 2017; 7:6774-89. [PMID: 26735180 PMCID: PMC4872748 DOI: 10.18632/oncotarget.6725] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/25/2015] [Indexed: 12/16/2022] Open
Abstract
tTF-TAA and tTF-LTL are fusion proteins consisting of the extracellular domain of tissue factor (TF) and the peptides TAASGVRSMH and LTLRWVGLMS, respectively. These peptides represent ligands of NG2, a surface proteoglycan expressed on angiogenic pericytes and some tumor cells. Here we have expressed the model compound tTF-NGR, tTF-TAA, and tTF-LTL with different lengths in the TF domain in E. coli and used these fusion proteins for functional studies in anticancer therapy. We aimed to retarget TF to tumor vessels leading to tumor vessel infarction with two barriers of selectivity, a) the leaky endothelial lining in tumor vessels with the target NG2 being expressed on pericytes on the abluminal side of the endothelial cell barrier and b) the preferential expression of NG2 on angiogenic vessels such as in tumors. Chromatography-purified tTF-TAA showed identical Factor X (FX)-activating procoagulatory activity as the model compound tTF-NGR with Km values of approx. 0.15 nM in Michaelis-Menten kinetics. The procoagulatory activity of tTF-LTL varied with the chosen length of the TF part of the fusion protein. Flow cytometry revealed specific binding of tTF-TAA to NG2-expressing pericytes and tumor cells with low affinity and dissociation KD in the high nM range. In vivo and ex vivo fluorescence imaging of tumor xenograft-carrying animals and of the explanted tumors showed reduction of tumor blood flow upon tTF-TAA application. Therapeutic experiments showed a reproducible antitumor activity of tTF-TAA against NG2-expressing A549-tumor xenografts, however, with a rather small therapeutic window (active/toxic dose in mg/kg body weight).
Collapse
Affiliation(s)
- Caroline Brand
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Christoph Schliemann
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Janine Ring
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Torsten Kessler
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Sebastian Bäumer
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Linus Angenendt
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Verena Mantke
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Rebecca Ross
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Heike Hintelmann
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Tilmann Spieker
- Gerhard-Domagk Institute for Pathology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Eva Wardelmann
- Gerhard-Domagk Institute for Pathology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Rolf M Mesters
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| | - Christian Schwöppe
- Department of Medicine A, Hematology, Oncology and Pneumology, University of Muenster, Albert-Schweitzer-Campus 1, D-48129 Muenster, Germany
| |
Collapse
|
239
|
Pfister F, Hussain H, Belharazem D, Busch S, Simon-Keller K, Becker D, Pfister E, Rieker R, Ströbel P, Marx A. Vascular architecture as a diagnostic marker for differentiation of World Health Organization thymoma subtypes and thymic carcinoma. Histopathology 2017; 70:693-703. [DOI: 10.1111/his.13114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 10/26/2016] [Indexed: 01/27/2023]
Affiliation(s)
- Frederick Pfister
- Department of Nephropathology; Institute of Pathology; Universitätsklinikum Erlangen; Friedrich-Alexander-University Erlangen-Nürnberg; Erlangen Germany
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Hussam Hussain
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Djeda Belharazem
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Svenja Busch
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Katja Simon-Keller
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Dominic Becker
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Eva Pfister
- Department of Nephropathology; Institute of Pathology; Universitätsklinikum Erlangen; Friedrich-Alexander-University Erlangen-Nürnberg; Erlangen Germany
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| | - Ralf Rieker
- Institute of Pathology; Universitätsklinikum Erlangen; Friedrich-Alexander-University Erlangen-Nürnberg; Erlangen Germany
| | - Philipp Ströbel
- Institute of Pathology; University Medicine Göttingen; Göttingen Germany
| | - Alexander Marx
- Institute of Pathology; University Medical Centre Mannheim; University of Heidelberg; Mannheim Germany
| |
Collapse
|
240
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
241
|
Kennedy-Lydon T. Immune Functions and Properties of Resident Cells in the Heart and Cardiovascular System: Pericytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:93-103. [PMID: 28667555 DOI: 10.1007/978-3-319-57613-8_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This chapter provides an introduction to pericyte physiology. Pericytes are smooth muscle-like cells that wrap around vessels and arterioles. Here, we discuss their structure, function, contractility and interaction with other cells including immune cells and finally their role in pathological processes. Additionally, we discuss recent studies describing pericyte populations in the heart and their potential as targets for future cardiac therapeutic interventions.
Collapse
|
242
|
Tanaka M, Homme M, Yamazaki Y, Shimizu R, Takazawa Y, Nakamura T. Modeling Alveolar Soft Part Sarcoma Unveils Novel Mechanisms of Metastasis. Cancer Res 2016; 77:897-907. [DOI: 10.1158/0008-5472.can-16-2486] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/05/2016] [Accepted: 11/21/2016] [Indexed: 01/20/2023]
|
243
|
Endothelial cell transplantation in tumors restores normal vasculature, reduces tumor hypoxia, and suppresses tumor outgrowth. J Oral Biosci 2016; 58:150-157. [DOI: 10.1016/j.job.2016.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/17/2023]
|
244
|
Harbi S, Wang R, Gregory M, Hanson N, Kobylarz K, Ryan K, Deng Y, Lopez P, Chiriboga L, Mignatti P. Infantile Hemangioma Originates From A Dysregulated But Not Fully Transformed Multipotent Stem Cell. Sci Rep 2016; 6:35811. [PMID: 27786256 PMCID: PMC5081534 DOI: 10.1038/srep35811] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 10/05/2016] [Indexed: 12/11/2022] Open
Abstract
Infantile hemangioma (IH) is the most common tumor of infancy. Its cellular origin and biological signals for uncontrolled growth are poorly understood, and specific pharmacological treatment is unavailable. To understand the process of hemangioma-genesis we characterized the progenitor hemangioma-derived stem cell (HemSC) and its lineage and non-lineage derivatives. For this purpose we performed a high-throughput (HT) phenotypic and gene expression analysis of HemSCs, and analyzed HemSC-derived tumorspheres. We found that IH is characterized by high expression of genes involved in vasculogenesis, angiogenesis, tumorigenesis and associated signaling pathways. These results show that IH derives from a dysregulated stem cell that remains in an immature, arrested stage of development. The potential biomarkers we identified can afford the development of diagnostic tools and precision-medicine therapies to "rewire" or redirect cellular transitions at an early stage, such as signaling pathways or immune response modifiers.
Collapse
Affiliation(s)
- Shaghayegh Harbi
- Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- VasculoTox Inc., New York, NY 10001, USA
| | - Rong Wang
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Michael Gregory
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Nicole Hanson
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Keith Kobylarz
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- Pfizer Inc., Pearl River, NY 10965, USA
| | - Kamilah Ryan
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Yan Deng
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Peter Lopez
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Luis Chiriboga
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Paolo Mignatti
- Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- Department of Cell Biology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| |
Collapse
|
245
|
Shenoy AK, Jin Y, Luo H, Tang M, Pampo C, Shao R, Siemann DW, Wu L, Heldermon CD, Law BK, Chang LJ, Lu J. Epithelial-to-mesenchymal transition confers pericyte properties on cancer cells. J Clin Invest 2016; 126:4174-4186. [PMID: 27721239 DOI: 10.1172/jci86623] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 09/01/2016] [Indexed: 01/31/2023] Open
Abstract
Carcinoma cells can acquire increased motility and invasiveness through epithelial-to-mesenchymal transition (EMT). However, the significance of EMT in cancer metastasis has been controversial, and the exact fates and functions of EMT cancer cells in vivo remain inadequately understood. Here, we tracked epithelial cancer cells that underwent inducible or spontaneous EMT in various tumor transplantation models. Unlike epithelial cells, the majority of EMT cancer cells were specifically located in the perivascular space and closely associated with blood vessels. EMT markedly activated multiple pericyte markers in carcinoma cells, in particular PDGFR-β and N-cadherin, which enabled EMT cells to be chemoattracted towards and physically interact with endothelium. In tumor xenografts generated from carcinoma cells that were prone to spontaneous EMT, a substantial fraction of the pericytes associated with tumor vasculature were derived from EMT cancer cells. Depletion of such EMT cells in transplanted tumors diminished pericyte coverage, impaired vascular integrity, and attenuated tumor growth. These findings suggest that EMT confers key pericyte attributes on cancer cells. The resulting EMT cells phenotypically and functionally resemble pericytes and are indispensable for vascular stabilization and sustained tumor growth. This study thus proposes a previously unrecognized role for EMT in cancer.
Collapse
|
246
|
Gomis RR, Gawrzak S. Tumor cell dormancy. Mol Oncol 2016; 11:62-78. [PMID: 28017284 PMCID: PMC5423221 DOI: 10.1016/j.molonc.2016.09.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/13/2016] [Accepted: 09/30/2016] [Indexed: 12/22/2022] Open
Abstract
Metastasis is the primary cause of death in cancer patients and current treatments fail to provide durable responses. Efforts to treat metastatic disease are hindered by the fact that metastatic cells often remain dormant for prolonged intervals of years, or even decades. Tumor dormancy reflects the capability of disseminated tumor cells (DTCs), or micrometastases, to evade treatment and remain at low numbers after primary tumor resection. Unfortunately, dormant cells will eventually produce overt metastasis. Innovations are needed to understand metastatic dormancy and improve cancer detection and treatment. Currently, few models exist that faithfully recapitulate metastatic dormancy and metastasis to clinically relevant tissues, such as the bone. Herein, we discuss recent advances describing genetic cell‐autonomous and systemic or local changes in the microenvironment that have been shown to endow DTCs with properties to survive and eventually colonize distant organs.
Collapse
Affiliation(s)
- Roger R Gomis
- Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; ICREA Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain.
| | - Sylwia Gawrzak
- Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
247
|
Kitahara S, Desaki J, Yoshii A, Matsui A, Morikawa S, Ezaki T. Electron microscopic study of capillary network remodeling in the extensor digitorum longus muscle of normal adult rat. Microscopy (Oxf) 2016; 65:508-516. [PMID: 27655937 DOI: 10.1093/jmicro/dfw040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/22/2016] [Indexed: 12/25/2022] Open
Abstract
Capillary networks demonstrate structural changes during maturation, aging, vascular disease, and cancer. Their morphological structure and function have an important influence on each other. Understanding the process of morphological vascular changes in the capillary network with advancing age may help overcome fatal vascular diseases. Aging-related structural changes of the capillary segments may accompany degeneration and regeneration of muscle fibers and serve to remodel the capillary network as a means of adapting to the changing environment. However, difficulty in obtaining human samples has hampered clarification of these microstructural changes. Herein, we examined serial ultrathin sections of capillary segments in the extensor digitorum longus muscle of normal mature (12 months old) rats in an attempt to analyze their structural changes. After bifurcation, a minimum of one capillary segment was filled with erythrocytes and was found to have fenestrations and plural endothelial disruptions, or pores, at the fenestrated portions. Some of the stagnated erythrocytes demonstrated extended protrusions, and their processes appeared to penetrate the basal lamina through the pores. These findings can also show that capillary segments are involved in partial remodeling of the capillary network. A better understanding of age-related structural changes of the capillary networks will help in fine-tuning novel vascular therapy for not only several fatal vascular diseases but also malignant tumors.
Collapse
Affiliation(s)
- Shuji Kitahara
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan.,Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, 7910295 MA, USA
| | - Junzo Desaki
- Department of Integrated Basic Medical Research, School of Medicine, 02114 Ehime University , Ehime, Japan
| | - Asuka Yoshii
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan.,Departments of Pharmacology and Experimental Therapeutics and Neurology, Boston University School of Medicine, Boston, 02118 MA, USA
| | - Aya Matsui
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan
| |
Collapse
|
248
|
LaGory EL, Giaccia AJ. The ever-expanding role of HIF in tumour and stromal biology. Nat Cell Biol 2016; 18:356-65. [PMID: 27027486 DOI: 10.1038/ncb3330] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Low oxygen tension (hypoxia) is a hallmark of cancer that influences cancer cell function, but is also an important component of the tumour microenvironment as it alters the extracellular matrix, modulates the tumour immune response and increases angiogenesis. Here we discuss the regulation and role of hypoxia and its key transcriptional mediators, the hypoxia-inducible factor (HIF) family of transcription factors, in the tumour microenvironment and stromal compartments.
Collapse
Affiliation(s)
- Edward L LaGory
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, California 94305, USA
| | - Amato J Giaccia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
249
|
Verrienti A, Tallini G, Colato C, Boichard A, Checquolo S, Pecce V, Sponziello M, Rosignolo F, de Biase D, Rhoden K, Casadei GP, Russo D, Visani M, Acquaviva G, Ferdeghini M, Filetti S, Durante C. RET mutation and increased angiogenesis in medullary thyroid carcinomas. Endocr Relat Cancer 2016; 23:665-76. [PMID: 27402614 DOI: 10.1530/erc-16-0132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 07/07/2016] [Indexed: 02/05/2023]
Abstract
Advanced medullary thyroid cancers (MTCs) are now being treated with drugs that inhibit receptor tyrosine kinases, many of which involved in angiogenesis. Response rates vary widely, and toxic effects are common, so treatment should be reserved for MTCs likely to be responsive to these drugs. RET mutations are common in MTCs, but it is unclear how they influence the microvascularization of these tumors. We examined 45 MTCs with germ-line or somatic RET mutations (RETmut group) and 34 with wild-type RET (RETwt). Taqman Low-Density Arrays were used to assess proangiogenic gene expression. Immunohistochemistry was used to assess intratumoral, peritumoral and nontumoral expression levels of VEGFR1, R2, R3, PDGFRa, PDGFB and NOTCH3. We also assessed microvessel density (MVD) and lymphatic vessel density (LVD) based on CD31-positive and podoplanin-positive vessel counts, respectively, and vascular pericyte density based on staining for a-smooth muscle actin (a-SMA), a pericyte marker. Compared with RETwt tumors, RETmut tumors exhibited upregulated expression of proangiogenic genes (mRNA and protein), especially VEGFR1, PDGFB and NOTCH3. MVDs and LVDs were similar in the two groups. However, microvessels in RETmut tumors were more likely to be a-SMA positive, indicating enhanced coverage by pericytes, which play key roles in vessel sprouting, maturation and stabilization. These data suggest that angiogenesis in RETmut MTCs may be more intense and complete than that found in RETwt tumors, a feature that might increase their susceptibility to antiangiogenic therapy. Given their increased vascular pericyte density, RETmut MTCs might also benefit from combined or preliminary treatment with PDGF inhibitors.
Collapse
MESH Headings
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/metabolism
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Humans
- Microvessels
- Mutation
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Proto-Oncogene Proteins c-ret/genetics
- Proto-Oncogene Proteins c-ret/metabolism
- Receptor, Notch3/genetics
- Receptor, Notch3/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Signal Transduction
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Vascular Endothelial Growth Factor Receptor-1/genetics
- Vascular Endothelial Growth Factor Receptor-1/metabolism
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/metabolism
- Vascular Endothelial Growth Factor Receptor-3/genetics
- Vascular Endothelial Growth Factor Receptor-3/metabolism
Collapse
Affiliation(s)
- Antonella Verrienti
- Department of Internal Medicine and Medical Specialties'Sapienza' University of Rome, Rome, Italy
| | - Giovanni Tallini
- Department of Medicine (DIMES)Anatomic Pathology-Molecular Diagnostic Unit AUSL of Bologna, University of Bologna School of Medicine, Bologna, Italy
| | - Chiara Colato
- Department of Diagnostics and Public HealthUniversity and Hospital Trust of Verona, Verona, Italy
| | - Amélie Boichard
- Laboratoire de Recherche Translationnelle etCentre de Ressources Biologiques, AMMICA, INSERM US23/CNRS UMS3655, Gustave Roussy, Villejuif, France
| | - Saula Checquolo
- Laboratory of Molecular PathologyDepartment of Medico-Surgical and Biotechnology, 'Sapienza' University of Rome, Latin, Italy
| | - Valeria Pecce
- Department of Internal Medicine and Medical Specialties'Sapienza' University of Rome, Rome, Italy
| | - Marialuisa Sponziello
- Department of Internal Medicine and Medical Specialties'Sapienza' University of Rome, Rome, Italy
| | - Francesca Rosignolo
- Department of Internal Medicine and Medical Specialties'Sapienza' University of Rome, Rome, Italy
| | - Dario de Biase
- Department of Pharmacology and Biotechnology (FaBiT)University of Bologna, Bologna, Italy
| | - Kerry Rhoden
- Department of Medicine (DIMEC)Medical Genetics Unit, University of Bologna School of Medicine, Bologna, Italy
| | - Gian Piero Casadei
- Anatomic Pathology UnitAUSL di Bologna-Maggiore Hospital, Bologna, Italy
| | - Diego Russo
- Department of Health SciencesUniversity of Catanzaro 'Magna Graecia', Catanzaro, Italy
| | - Michela Visani
- Department of Medicine (DIMES)Anatomic Pathology-Molecular Diagnostic Unit AUSL of Bologna, University of Bologna School of Medicine, Bologna, Italy
| | - Giorgia Acquaviva
- Department of Medicine (DIMES)Anatomic Pathology-Molecular Diagnostic Unit AUSL of Bologna, University of Bologna School of Medicine, Bologna, Italy
| | - Marco Ferdeghini
- Department of Diagnostics and Public HealthUniversity and Hospital Trust of Verona, Verona, Italy
| | - Sebastiano Filetti
- Department of Internal Medicine and Medical Specialties'Sapienza' University of Rome, Rome, Italy
| | - Cosimo Durante
- Department of Internal Medicine and Medical Specialties'Sapienza' University of Rome, Rome, Italy
| |
Collapse
|
250
|
Di N, Mao N, Cheng W, Pang H, Ren Y, Wang N, Liu X, Wang B. Blood oxygenation level-dependent magnetic resonance imaging during carbogen breathing: differentiation between prostate cancer and benign prostate hyperplasia and correlation with vessel maturity. Onco Targets Ther 2016; 9:4143-50. [PMID: 27462169 PMCID: PMC4939976 DOI: 10.2147/ott.s105480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate whether the blood oxygenation level-dependent (BOLD) contrast magnetic resonance imaging (MRI) can evaluate tumor maturity and preoperatively differentiate prostate cancer (PCa) from benign prostate hyperplasia (BPH). PATIENTS AND METHODS BOLD MRI based on transverse relaxation time*-weighted echo planar imaging was performed to assess PCa (19) and BPH (22) responses to carbogen (95% O2 and 5% CO2). The average signal values of PCa and BPH before and after carbogen breathing and the relative increased signal values were computed, respectively. The endothelial-cell marker, CD31, and the pericyte marker, α-smooth muscle actin (mature vessels), were detected with immunofluorescence, and were assessed by microvessel density (MVD) and microvessel pericyte density (MPD). The microvessel pericyte coverage index (MPI) was used to evaluate the degree of vascular maturity. The changed signal from BOLD MRI was correlated with MVD, MPD, and MPI. RESULTS After inhaling carbogen, both PCa and BPH showed an increased signal, but a lower slope was found in PCa than that in BPH (P<0.05). PCa had a higher MPD and MVD but a lower MPI than BPH. The increased signal intensity was positively correlated with MPI in PCa and that in BPH (r=0.616, P=0.011; r=0.658, P=0.002); however, there was no correlation between the increased signal intensity and MPD or MVD in PCa than that in BPH (P>0.05). CONCLUSION Our results confirmed that the increased signal values induced by BOLD MRI well differentiated PCa from BPH and had a positive correlation with vessel maturity in both of them. BOLD MRI can be utilized as a surrogate marker for the noninvasive assessment of the degree of vessel maturity.
Collapse
Affiliation(s)
- Ningning Di
- Department of Radiology, Affiliated Huashan Hospital of Fudan University, Shanghai; Department of Radiology, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Ning Mao
- Department of Radiology, Yantai Yuhangding Hospital, Yantai
| | - Wenna Cheng
- Department of Pharmacy, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Haopeng Pang
- Department of Radiology, Affiliated Huashan Hospital of Fudan University, Shanghai
| | - Yan Ren
- Department of Radiology, Affiliated Huashan Hospital of Fudan University, Shanghai
| | - Ning Wang
- Department of Radiology, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Xinjiang Liu
- Department of Radiology, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Bin Wang
- Department of Medical Imaging and Nuclear Medicine, Binzhou Medical University, Yantai, People's Republic of China
| |
Collapse
|