1
|
Ou S, Lin Y, Zhang Y, Shi K, Wu H. Epidemiology and tumor microenvironment of ocular surface and orbital tumors on growth and malignant transformation. Front Oncol 2024; 14:1388156. [PMID: 39421442 PMCID: PMC11484446 DOI: 10.3389/fonc.2024.1388156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The ocular surface and orbit constitute unique microenvironments in the human body. Current advances in molecular research have deepened our understanding of tumor development in these regions. Tumors exhibit greater heterogeneity compared to normal tissues, as revealed by pathological and histological examinations. The tumor microenvironment (TME) plays a crucial role in the proliferation and progression of cancer cells. Factors from the external environment or the body's own inflammation and microcirculation interact within the TME, maintaining a delicate balance. Disruption of this balance, through uncontrolled signal pathway activation, can transform normal or benign tissues into malignant ones. In recent years, various systemic immunotherapies have been developed for cancer treatment. This study reviews the epidemiology of ocular surface and orbital tumors include squamous cell carcinoma, basal cell carcinoma, sebaceous carcinoma and lymphoma in conjunction with their occurrence, growth, and underlying mechanisms. We propose that by examining clinical histopathological images, we can identify specific and shared microscopic features of tumors. By collecting, classifying, and analyzing data from these clinical histopathological images, we can pinpoint independent diagnostic factors characteristic of tumors. We hope this study provides a basis for future exploration of the mechanisms underlying different ocular diseases.
Collapse
Affiliation(s)
- Shangkun Ou
- Xiamen Eye Center and Eye Institute of Xiamen University, School of Medicine, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Yuan Lin
- Xiamen Eye Center and Eye Institute of Xiamen University, School of Medicine, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Yujie Zhang
- Xiamen Eye Center and Eye Institute of Xiamen University, School of Medicine, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Ke Shi
- Xiamen Eye Center and Eye Institute of Xiamen University, School of Medicine, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Huping Wu
- Xiamen Eye Center and Eye Institute of Xiamen University, School of Medicine, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
2
|
Vázquez-Aristizabal P, Henriksen-Lacey M, García-Astrain C, Jimenez de Aberasturi D, Langer J, Epelde C, Litti L, Liz-Marzán LM, Izeta A. Biofabrication and Monitoring of a 3D Printed Skin Model for Melanoma. Adv Healthc Mater 2024; 13:e2401136. [PMID: 38992996 DOI: 10.1002/adhm.202401136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/27/2024] [Indexed: 07/13/2024]
Abstract
There is an unmet need for in vitro cancer models that emulate the complexity of human tissues. 3D-printed solid tumor micromodels based on decellularized extracellular matrices (dECMs) recreate the biomolecule-rich matrix of native tissue. Herein a 3D in vitro metastatic melanoma model that is amenable for drug screening purposes and recapitulates features of both the tumor and the skin microenvironment is described. Epidermal, basement membrane, and dermal biocompatible inks are prepared by means of combined chemical, mechanical, and enzymatic processes. Bioink printability is confirmed by rheological assessment and bioprinting, and bioinks are subsequently combined with melanoma cells and dermal fibroblasts to build complex 3D melanoma models. Cells are tracked by confocal microscopy and surface-enhanced Raman spectroscopy (SERS) mapping. Printed dECMs and cell tracking allow modeling of the initial steps of metastatic disease, and may be used to better understand melanoma cell behavior and response to drugs.
Collapse
Affiliation(s)
- Paula Vázquez-Aristizabal
- Stem Cells and Aging Group, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain s/n, Donostia-San Sebastián, 20014, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
| | - Malou Henriksen-Lacey
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
| | - Clara García-Astrain
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
| | - Dorleta Jimenez de Aberasturi
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, 48009, Spain
| | - Judith Langer
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
| | - Claudia Epelde
- Obstetrics and Gynaecology Service, Donostia University Hospital, Paseo Dr. Begiristain s/n, Donostia-San Sebastián, 20014, Spain
| | - Lucio Litti
- Department of Chemical Sciences, University of Padova, Via Marzolo, 1, Padova, 35131, Italy
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, 48009, Spain
- Cinbio, Universidade de Vigo, Campus Universitario, Vigo, 36310, Spain
| | - Ander Izeta
- Stem Cells and Aging Group, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain s/n, Donostia-San Sebastián, 20014, Spain
- School of Engineering, Tecnun-University of Navarra, Donostia-San Sebastián, 20009, Spain
| |
Collapse
|
3
|
Figueroa L, Rosas M, Alvarez M, Aguilar E, Mateu V, Bonilla E. Interaction of Purine and its Derivatives with A1, A2-Adenosine Receptors and Vascular Endothelial Growth Factor Receptor-1 (Vegf-R1) as a Therapeutic Alternative to Treat Cancer. Drug Res (Stuttg) 2024; 74:379-393. [PMID: 39173673 DOI: 10.1055/a-2376-5771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
BACKGROUND There are several studies that indicate that cancer development may be conditioned by the activation of some biological systems that involve the interaction of different biomolecules, such as adenosine and vascular endothelial growth factor. These biomolecules have been targeted of some drugs for treat of cancer; however, there is little information on the interaction of purine derivatives with adenosine and vascular endothelial growth factor receptor (VEGF-R1). OBJECTIVE The aim of this research was to determine the possible interaction of purine (1: ) and their derivatives (2-31: ) with A1, A2-adenosine receptors, and VEGF-R1. METHODS Theoretical interaction of purine and their derivatives with A1, A2-adenosine receptors and VEGF-R1 was carried out using the 5uen, 5mzj and 3hng proteins as theoretical tools. Besides, adenosine, cgs-15943, rolofylline, cvt-124, wrc-0571, luf-5834, cvt-6883, AZD-4635, cabozantinib, pazopanib, regorafenib, and sorafenib drugs were used as controls. RESULTS The results showed differences in the number of aminoacid residues involved in the interaction of purine and their derivatives with 5uen, 5mzj and 3hng proteins compared with the controls. Besides, the inhibition constants (Ki) values for purine and their derivatives 5: , 9: , 10: , 14: , 15: , 16: , and 20: were lower compared with the controls CONCLUSIONS: Theoretical data suggest that purine and their derivatives 5: , 9: , 10: , 14: , 15: , 16: , and 20: could produce changes in cancer cell growth through inhibition of A1, A2-adenosine receptors and VEGFR-1 inhibition. These data indicate that these purine derivatives could be a therapeutic alternative to treat some types of cancer.
Collapse
Affiliation(s)
- Lauro Figueroa
- Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Campeche, Camp., México
| | - Marcela Rosas
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Magdalena Alvarez
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Emilio Aguilar
- Facultad de Medicina, Universidad Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Virginia Mateu
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Enrique Bonilla
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| |
Collapse
|
4
|
Malekan M, Haass NK, Rokni GR, Gholizadeh N, Ebrahimzadeh MA, Kazeminejad A. VEGF/VEGFR axis and its signaling in melanoma: Current knowledge toward therapeutic targeting agents and future perspectives. Life Sci 2024; 345:122563. [PMID: 38508233 DOI: 10.1016/j.lfs.2024.122563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is responsible for most skin cancer-associated deaths globally. The progression of melanoma is influenced by a number of pathogenic processes. Understanding the VEGF/VEGFR axis, which includes VEGF-A, PlGF, VEGF-B, VEGF-C, and VEGF-D and their receptors, VEGFR-1, VEGFR-2, and VEGFR-3, is of great importance in melanoma due to its crucial role in angiogenesis. This axis generates multifactorial and complex cellular signaling, engaging the MAPK/ERK, PI3K/AKT, PKC, PLC-γ, and FAK signaling pathways. Melanoma cell growth and proliferation, migration and metastasis, survival, and acquired resistance to therapy are influenced by this axis. The VEGF/VEGFR axis was extensively examined for their potential as diagnostic/prognostic biomarkers in melanoma patients and results showed that VEGF overexpression can be associated with unfavorable prognosis, higher level of tumor invasion and poor response to therapy. MicroRNAs linking to the VEGF/VEGFR axis were identified and, in this review, divided into two categories according to their functions, some of them promote melanoma angiogenesis (promotive group) and some restrict melanoma angiogenesis (protective group). In addition, the approach of treating melanoma by targeting the VEGF/VEGFR axis has garnered significant interest among researchers. These agents can be divided into two main groups: anti-VEGF and VEGFR inhibitors. These therapeutic options may be a prominent step along with the modern targeting and immune therapies for better coverage of pathological processes leading to melanoma progression and therapy resistance.
Collapse
Affiliation(s)
- Mohammad Malekan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | - Ghasem Rahmatpour Rokni
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Armaghan Kazeminejad
- Department of Dermatology, Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences,Sari, Iran
| |
Collapse
|
5
|
Zhao Y, Yu B, Wang Y, Tan S, Xu Q, Wang Z, Zhou K, Liu H, Ren Z, Jiang Z. Ang-1 and VEGF: central regulators of angiogenesis. Mol Cell Biochem 2024:10.1007/s11010-024-05010-3. [PMID: 38652215 DOI: 10.1007/s11010-024-05010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Angiopoietin-1 (Ang-1) and Vascular Endothelial Growth Factor (VEGF) are central regulators of angiogenesis and are often inactivated in various cardiovascular diseases. VEGF forms complexes with ETS transcription factor family and exerts its action by downregulating multiple genes. Among the target genes of the VEGF-ETS complex, there are a significant number encoding key angiogenic regulators. Phosphorylation of the VEGF-ETS complex releases transcriptional repression on these angiogenic regulators, thereby promoting their expression. Ang-1 interacts with TEK, and this phosphorylation release can be modulated by the Ang-1-TEK signaling pathway. The Ang-1-TEK pathway participates in the transcriptional activation of VEGF genes. In summary, these elements constitute the Ang-1-TEK-VEGF signaling pathway. Additionally, Ang-1 is activated under hypoxic and inflammatory conditions, leading to an upregulation in the expression of TEK. Elevated TEK levels result in the formation of the VEGF-ETS complex, which, in turn, downregulates the expression of numerous angiogenic genes. Hence, the Ang-1-dependent transcriptional repression is indirect. Reduced expression of many target genes can lead to aberrant angiogenesis. A significant overlap exists between the target genes regulated by Ang-1-TEK-VEGF and those under the control of the Ang-1-TEK-TSP-1 signaling pathway. Mechanistically, this can be explained by the replacement of the VEGF-ETS complex with the TSP-1 transcriptional repression complex at the ETS sites on target gene promoters. Furthermore, VEGF possesses non-classical functions unrelated to ETS and DNA binding. Its supportive role in TSP-1 formation may be exerted through the VEGF-CRL5-VHL-HIF-1α-VH032-TGF-β-TSP-1 axis. This review assesses the regulatory mechanisms of the Ang-1-TEK-VEGF signaling pathway and explores its significant overlap with the Ang-1-TEK-TSP-1 signaling pathway.
Collapse
Affiliation(s)
- Yuanqin Zhao
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Bo Yu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Yanxia Wang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Shiming Tan
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Qian Xu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Zhaoyue Wang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Kun Zhou
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Huiting Liu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Zhong Ren
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Zhisheng Jiang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China.
| |
Collapse
|
6
|
Su DG, Djureinovic D, Schoenfeld D, Marquez-Nostra B, Olino K, Jilaveanu L, Kluger H. Melanocortin-1 Receptor Expression as a Marker of Progression in Melanoma. JCO Precis Oncol 2024; 8:e2300702. [PMID: 38662983 PMCID: PMC11513442 DOI: 10.1200/po.23.00702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 04/30/2024] Open
Abstract
PURPOSE Melanocortin-1 receptor (MC1R) plays a critical role in human pigmentation and DNA repair mechanisms. MC1R-targeting agents are being investigated in clinical trials in patients with melanoma, yet large studies investigating the rate and degree of MC1R expression in primary and metastatic human melanoma tissue are lacking. METHODS Using tissue microarrays containing three large cohorts of 225 cases of benign nevi, 189 with primary melanoma, and 271 with metastatic melanoma, we applied quantitative immunofluorescence and immunohistochemistry to comprehensively study MC1R protein expression. RESULTS We show a stepwise elevation of MC1R expression in different stages of melanoma progression (nevi, primary, metastasis). Higher MC1R expression was seen in deeper (>1 mm) primary lesions and ulcerated lesions and was associated with shorter survival in primary and metastatic tumors. On multivariable analysis, Breslow thickness, male sex, and chronic sun exposure were independent predictors of worse overall survival in the primary melanoma cohort. CONCLUSION Our data suggest that MC1R might be a valuable drug target in aggressive melanoma. Additional studies are warranted to determine its functional significance in melanoma progression and its utility as a predictive biomarker in patients receiving MC1R-directed therapies.
Collapse
Affiliation(s)
- David G. Su
- Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT
| | - Dijana Djureinovic
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - David Schoenfeld
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Bernadette Marquez-Nostra
- Department of Radiology, Division of Advanced Medical Imaging Research, University of Alabama at Birmingham, Birmingham, AL
| | - Kelly Olino
- Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT
| | - Lucia Jilaveanu
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Harriet Kluger
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
7
|
García-Hevia L, Soltani R, González J, Chaloin O, Ménard-Moyon C, Bianco A, L. Fanarraga M. Carbon nanotubes targeted to the tumor microenvironment inhibit metastasis in a preclinical model of melanoma. Bioact Mater 2024; 34:237-247. [PMID: 38223536 PMCID: PMC10787223 DOI: 10.1016/j.bioactmat.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024] Open
Abstract
Despite notable progress in cancer therapy, metastatic diseases continue to be the primary cause of cancer-related mortality. Multi-walled carbon nanotubes (MWCNTs) can enter tissues and cells and interfere with the dynamics of the cytoskeletal nanofilaments biomimetically. This endows them with intrinsic anti-tumoral effects comparable to those of microtubule-binding chemotherapies such as Taxol®. In this study, our focus was on exploring the potential of oxidized MWCNTs in selectively targeting the vascular endothelial growth factor receptor (VEGFR). Our objective was to evaluate their effectiveness in inhibiting metastatic growth by inducing anti-proliferative, anti-migratory, and cytotoxic effects on both cancer and tumor microenvironment cells. Our findings demonstrated a significant reduction of over 80 % in malignant melanoma lung metastases and a substantial enhancement in overall animal welfare following intravenous administration of the targeted biodegradable MWCNTs. Furthermore, the combination of these nanomaterials with the conventional chemotherapy agent Taxol® yielded a remarkable 90 % increase in the antimetastatic effect. These results highlight the promising potential of this combined therapeutic approach against metastatic disease and are of paramount importance as metastasis is responsible for nearly 60,000 deaths each year.
Collapse
Affiliation(s)
- Lorena García-Hevia
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| | - Rym Soltani
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Jesús González
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| | - Olivier Chaloin
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Cécilia Ménard-Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Mónica L. Fanarraga
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| |
Collapse
|
8
|
Baginska J, Nau A, Gomez Diaz I, Giobbie-Hurder A, Weirather J, Vergara J, Abrecht C, Hallisey M, Dennis J, Severgnini M, Huezo J, Marciello I, Rahma O, Manos M, Brohl AS, Bedard PL, Renouf DJ, Sharon E, Streicher H, Ott PA, Buchbinder EI, Hodi FS. Ziv-aflibercept plus pembrolizumab in patients with advanced melanoma resistant to anti-PD-1 treatment. Cancer Immunol Immunother 2024; 73:17. [PMID: 38236249 PMCID: PMC10796592 DOI: 10.1007/s00262-023-03593-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Vascular endothelial growth factor is associated with reduced immune response and impaired anti-tumor activity. Combining antiangiogenic agents with immune checkpoint inhibition can overcome this immune suppression and enhance treatment efficacy. METHODS This study investigated the combination of ziv-aflibercept anti-angiogenic therapy with pembrolizumab in patients with advanced melanoma resistant to anti-PD-1 treatment. Baseline and on-treatment plasma and PBMC samples were analyzed by multiplex protein assay and mass cytometry, respectively. RESULTS In this Phase 1B study (NCT02298959), ten patients with advanced PD-1-resistant melanoma were treated with a combination of ziv-aflibercept (at 2-4 mg/kg) plus pembrolizumab (at 2 mg/kg), administered intravenously every 2 weeks. Two patients (20%) achieved a partial response, and two patients (20%) experienced stable disease (SD) as the best response. The two responders had mucosal melanoma, while both patients with SD had ocular melanoma. The combination therapy demonstrated clinical activity and acceptable safety, despite the occurrence of adverse events. Changes in plasma analytes such as platelet-derived growth factor and PD-L1 were explored, indicating potential alterations in myeloid cell function. Higher levels of circulating CXCL10 in non-responding patients may reflect pro-tumor activity. Specific subsets of γδ T cells were associated with poor clinical outcomes, suggesting impaired γδ T-cell function in non-responding patients. CONCLUSIONS Although limited by sample size and follow-up, these findings highlight the potential of the combination of ziv-aflibercept antiangiogenic therapy with pembrolizumab in patients with advanced melanoma resistant to anti-PD-1 treatment and the need for further research to improve outcomes in anti-PD-1-resistant melanoma. TRIAL REGISTRATION NUMBER NCT02298959.
Collapse
Affiliation(s)
- Joanna Baginska
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allison Nau
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ilana Gomez Diaz
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Jason Weirather
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juliana Vergara
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Charlotte Abrecht
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Margaret Hallisey
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jenna Dennis
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mariano Severgnini
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Julia Huezo
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Isabella Marciello
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Osama Rahma
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Manos
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew S Brohl
- Sarcoma Department and Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Daniel J Renouf
- Cancer and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Elad Sharon
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Patrick A Ott
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elizabeth I Buchbinder
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
9
|
Huang J, Chan SC, Ko S, Lok V, Zhang L, Lin X, Lucero-Prisno Iii DE, Xu W, Zheng ZJ, Elcarte E, Withers M, Wong MCS. Global Incidence, Mortality, Risk Factors and Trends of Melanoma: A Systematic Analysis of Registries. Am J Clin Dermatol 2023; 24:965-975. [PMID: 37296344 DOI: 10.1007/s40257-023-00795-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Melanoma of the skin is the most dangerous skin cancer in the world, though the numbers of reported new cases and melanoma-related deaths are low. OBJECTIVE This study evaluated the global incidence, mortality, risk factors and temporal trends by age, sex and locations of melanoma skin cancer. PATIENTS AND METHODS Cancer Incidence in Five Continents (CI5) volumes I-XI; the Nordic Cancer Registries (NORDCAN); the Surveillance, Epidemiology and End Results (SEER) Program; and the World Health Organization (WHO) International Agency for Research on Cancer (IARC) mortality database were accessed for worldwide incidence and mortality rates. Average Annual Percentage Change (AAPC) was calculated using a Joinpoint regression to examine trends. RESULTS Age-standardized rates of cancer incidence and mortality were 3.4 and 0.55 per 100,000 worldwide in 2020. Australia and New Zealand reported the highest incidence and mortality rates. Associated risk factors included higher prevalence of smoking, alcohol consumption, unhealthy diet, obesity and metabolic diseases. Increasing incidence trends were observed mostly in European countries, whilst mortality displayed an overall decreasing trend. For both sexes in the age group 50 years and above, a significant increase in incidence trend was observed. CONCLUSIONS Although mortality rates and trends were found to decrease, global incidence has increased, especially in older age groups and males. Whilst incidence increase may be attributed to improved healthcare infrastructure and cancer detection methods, the growing prevalence of lifestyle and metabolic risk factors in developed countries should not be discounted. Future research should explore underlying variables behind epidemiological trends.
Collapse
Affiliation(s)
- Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sze Chai Chan
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Samantha Ko
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Veeleah Lok
- Department of Global Public Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Lin Zhang
- School of Population and Global Health, The University of Melbourne, Victoria, Australia
- School of Public Health, The Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Don Eliseo Lucero-Prisno Iii
- Department of Global Health and Development, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Wanghong Xu
- School of Public Health, Fudan University, Shanghai, China
| | - Zhi-Jie Zheng
- Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Edmar Elcarte
- University of the Philippines, Manila, The Philippines
| | - Mellissa Withers
- Department of Population and Health Sciences, Institute for Global Health, University of Southern California, Los Angeles, USA
| | - Martin C S Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- School of Public Health, The Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Global Health, School of Public Health, Peking University, Beijing, China.
| |
Collapse
|
10
|
García-Pérez O, Melgar-Vilaplana L, Sifaoui I, García-Bello MÁ, Córdoba-Lanús E, Fernández-de-Misa R. Expression of angiogenic and lymphangiogenic genes in primary cutaneous melanoma: relationship with angiolymphatic invasion and disease-free survival. Melanoma Res 2023; 33:375-387. [PMID: 37307530 PMCID: PMC10470437 DOI: 10.1097/cmr.0000000000000904] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/09/2023] [Indexed: 06/14/2023]
Abstract
Melanoma is one of the most common cancers in the world. The main routes of tumor progression are related to angiogenesis and lymphangiogenesis. These routes can occur by local invasion, which is called angiolymphatic invasion (ALI). In this study, we assess gene expression of relevant biomarkers of angiogenesis and lymphangiogenesis in 80 FFPE melanoma samples to determine a molecular profile that correlates with ALI, tumor progression, and disease-free survival. The results were enhanced by a posttranscriptional analysis by an immunofluorescence assay. Three SNPs in the VEGFR-2 gene were genotyped in 237 malignant melanoma (MM) blood DNA samples by qPCR. A significant correlation was found for LYVE -1 and ALI, qualitative ( P = 0.017) and quantitative ( P = 0.005). An increased expression of protein LIVE-1 in ALI samples supported these results ( P = 0.032). VEGFR2 was lower in patients who showed disease progression ( P = 0.005) and protein VEGFR2 posttranscriptional expression decreased ( P = 0.016). DFS curves showed differences ( P = 0.023) for VEGFR2 expression detected versus the absence of VEGFR2 expression. No significant influence on DFS was detected for the remaining analyzed genes. Cox regression analysis suggested that VEGFR2 expression has a protective role (HR = 0.728; 95% CI = 0.552-0.962; P = 0.025) on disease progression. No significant association was found between any of the studied SNPs of VEGFR2 and DFS or progression rate. Our main results suggest that LYVE-1 gene expression is closely related to ALI; the relationship with the development of metastases in MM deserves further studies. Low expression of VEGFR2 was associated with disease progression and the expression of VEGFR2 correlates with an increased DFS.
Collapse
Affiliation(s)
- Omar García-Pérez
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife
- Universidad de La Laguna, Calle Padre Herrera, s/n
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), San Cristóbal de La Laguna
| | | | - Ines Sifaoui
- Universidad de La Laguna, Calle Padre Herrera, s/n
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), San Cristóbal de La Laguna
| | | | - Elizabeth Córdoba-Lanús
- Universidad de La Laguna, Calle Padre Herrera, s/n
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), San Cristóbal de La Laguna
| | - Ricardo Fernández-de-Misa
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife
- Universidad de La Laguna, Calle Padre Herrera, s/n
- Dermatology Department, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| |
Collapse
|
11
|
Su D, Djureinovic D, Schoenfeld D, Marquez-Nostra B, Olino K, Jilaveanu L, Kluger H. Melanocortin 1 receptor (MC1R) expression as a marker of progression in melanoma. RESEARCH SQUARE 2023:rs.3.rs-3314825. [PMID: 37790306 PMCID: PMC10543287 DOI: 10.21203/rs.3.rs-3314825/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Melanocortin-1 receptor (MC1R) plays a critical role in human pigmentation and DNA repair mechanisms. MC1R-targeting agents are being investigated in clinical trials in melanoma patients, yet large studies investigating the rate and degree of MC1R expression in primary and metastatic human melanoma tissue are lacking. Using tissue microarrays containing three large cohorts of 225 cases of benign nevi, 189 with primary melanoma, and 271 with metastatic melanoma, we applied quantitative immunofluorescence and immunohistochemistry to comprehensively study MC1R protein expression. We show a stepwise elevation of MC1R expression in different stages of melanoma progression (nevi, primary, metastasis). Higher MC1R expression was seen in deeper (>1 mm) primary lesions, ulcerated lesions, and mucosal melanomas compared to cutaneous melanomas and was associated with shorter survival in primary and metastatic tumors. On multi-variable analysis, Breslow thickness, ulceration, male sex, and chronic sun exposure were independent predictors of worse overall survival in the primary melanoma cohort. In the metastatic melanoma cohort, MC1R expression and mucosal melanomas were independent predictors of inferior overall survival. Our data suggest that MC1R might be a valuable drug target in aggressive melanoma. Additional studies are warranted to determine its functional significance in melanoma progression and its utility as a predictive biomarker in patients receiving MC1R-directed therapies.
Collapse
Affiliation(s)
- David Su
- Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT
| | - Dijana Djureinovic
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - David Schoenfeld
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Bernadette Marquez-Nostra
- Department of Radiology, Division of Advanced Medical Imaging Research, University of Alabama at Birmingham, Birmingham, AL
| | - Kelly Olino
- Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT
| | - Lucia Jilaveanu
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Harriet Kluger
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
12
|
Zahedipour F, Zamani P, Mashreghi M, Astaneh M, Sankian M, Amiri A, Jamialahmadi K, Jaafari MR. Nanoliposomal VEGF-R2 peptide vaccine acts as an effective therapeutic vaccine in a murine B16F10 model of melanoma. Cancer Nanotechnol 2023; 14:62. [PMID: 37333490 PMCID: PMC10264216 DOI: 10.1186/s12645-023-00213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
Background The vascular endothelial growth factor receptor-2 (VEGFR-2) plays an important role in melanoma development and progression. Peptide vaccines have shown great potential in cancer immunotherapy by targeting VEGFR-2 as a tumor-associated antigen and boosting the immune response against both tumor cells and tumor endothelial cells. Despite this, the low efficiency of peptide vaccines has resulted in moderate therapeutic results in the majority of studies. Enhancing the delivery of peptide vaccines using nanoliposomes is an important strategy for improving the efficacy of peptide vaccines. In this regard, we designed VEGFR-2-derived peptides restricted to both mouse MHC I and human HLA-A*02:01 using immunoinformatic tools and selected three peptides representing the highest binding affinities. The peptides were encapsulated in nanoliposomal formulations using the film method plus bath sonication and characterized for their colloidal properties. Results The mean diameter of peptide-encapsulated liposomes was around 135 nm, zeta potential of - 17 mV, and encapsulation efficiency of approximately 70%. Then, vaccine formulations were injected subcutaneously in mice bearing B16F10-established melanoma tumors and their efficiency in triggering immunological, and anti-tumor responses was evaluated. Our results represented that one of our designed VEGFR-2 peptide nanoliposomal formulations (Lip-V1) substantially activated CD4+ (p < 0.0001) and CD8+ (P < 0.001) T cell responses and significantly boosted the production of IFN-γ (P < 0.0001) and IL-4 (P < 0.0001). Furthermore, this formulation led to a significant decrease in tumor volume (P < 0.0001) and enhanced survival (P < 0.05) in mice. Conclusion Our findings suggest that the nanoliposomal formulation containing VEGFR-2 peptides could be a promising therapeutic vaccination approach capable of eliciting strong antigen-specific immunologic and anti-tumor responses. Supplementary Information The online version contains supplementary material available at 10.1186/s12645-023-00213-7.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Astaneh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Santamaria-Barria JA, Matsuba C, Khader A, Scholar AJ, Garland-Kledzik M, Fischer TD, Essner R, Salomon MP, Mammen JMV, Goldfarb M. Age-related next-generation sequencing mutational analysis in 1196 melanomas. J Surg Oncol 2023; 127:1187-1195. [PMID: 36938777 DOI: 10.1002/jso.27239] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND AND OBJECTIVES Melanoma mutational burden is high and approximately 50% have oncogenic mutations in BRAF. We sought to evaluate age-related mutational differences in melanoma. METHODS We analyzed melanoma samples in the Genomics Evidence Neoplasia Information Exchange database. Targetable mutations were identified using the Precision Oncology Knowledge Base (OncoKB). RESULTS We found 1194 patients with a common set of 30 genes. The top mutated genes in patients <40 years old (y/o) (n = 98) were BRAF (59%), TP53 (31%), NRAS (17%), and PTEN (14%); in 40-59 y/o (n = 354) were BRAF (51%), NRAS (30%), TP53 (26%), and APC (13%); and in ≥60 y/o (n = 742) were BRAF (38%), NRAS (33%), TP53 (26%), and KDR (19%). BRAF mutations were almost mutually exclusive from NRAS mutations in <40 y/o (58/59). Mutational burden increased with age, with means of 2.39, 2.92, and 3.67 mutations per sample in patients <40, 40-59, and ≥60 y/o, respectively (p < 0.0001). There were 10 targetable mutations meeting OncoKB criteria for melanoma: BRAF (level 1), RET (level 1), KIT (level 2), NRAS (level 3A), TP53 (level 3A), and FGFR2, MET, PTEN, PIK3CA, and KRAS (level 4). CONCLUSIONS Mutations in melanoma have age-related differences and demonstrates potential targetable mutations for personalized therapies.
Collapse
Affiliation(s)
- Juan A Santamaria-Barria
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chikako Matsuba
- Computational Biology Division, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| | - Adam Khader
- Division of Surgical Oncology, Department of Surgery, Hunter Holmes McGuire Veterans Affair Medical Center, Richmond, Virginia, USA
| | - Anthony J Scholar
- Division of Surgical Oncology, University of South Carolina School of Medicine, Greenville, South Carolina, USA
| | - Mary Garland-Kledzik
- Division of Surgical Oncology, West Virginia University, Morgantown, West Virginia, USA
| | - Trevan D Fischer
- Department of Surgery, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| | - Richard Essner
- Department of Surgery, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| | - Matthew P Salomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Joshua M V Mammen
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Melanie Goldfarb
- Department of Surgery, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| |
Collapse
|
14
|
Tissue Biomarkers Predicting Lymph Node Status in Cutaneous Melanoma. Int J Mol Sci 2022; 24:ijms24010144. [PMID: 36613587 PMCID: PMC9820052 DOI: 10.3390/ijms24010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Cutaneous melanoma is a severe neoplasm that shows early invasiveness of the lymph nodes draining the primary site, with increased risk of distant metastases and recurrence. The tissue biomarker identification could be a new frontier to predict the risk of early lymph node invasiveness, especially in cases considered by current guidelines to be at low risk of lymph node involvement and not requiring evaluation of the sentinel lymph node (SLN). For this reason, we present a narrative review of the literature, seeking to provide an overview of current tissue biomarkers, particularly vascular endothelium growth factors (VEGF), Tetraspanin CD9, lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), D2-40, and gene expression profile test (31-GEP). Among these, 31-GEP seems to be able to provide a distinction between low or high risk for positive SLN classes. VEGF receptor-3 and CD9 expression may be independent predictors of positive SLN. Lastly, LYVE-1 and D2-40 allow an easier assessment of lymph vascular invasion, which can be considered a good predictor of SLN status. In conclusion, biomarkers to assess the lymph node status of cutaneous melanoma patients may play an important role in those cases where the clinician is in doubt whether or not to perform SLN biopsy.
Collapse
|
15
|
Bonsang B, Maksimovic L, Maille P, Martin N, Laurendeau I, Pasmant E, Bièche I, Deschamps J, Wolkenstein P, Ortonne N. VEGF and VEGFR family members are expressed by neoplastic cells of NF1-associated tumors and may play an oncogenic role in malignant peripheral nerve sheath tumor growth through an autocrine loop. Ann Diagn Pathol 2022; 60:151997. [DOI: 10.1016/j.anndiagpath.2022.151997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 11/01/2022]
|
16
|
Zhang RS, Li ZK, Liu J, Deng YT, Jiang Y. WZB117 enhanced the anti-tumor effect of apatinib against melanoma via blocking STAT3/PKM2 axis. Front Pharmacol 2022; 13:976117. [PMID: 36188586 PMCID: PMC9524253 DOI: 10.3389/fphar.2022.976117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Melanoma is the most lethal skin malignant tumor with a short survival once stepping into the metastatic status and poses a therapeutic challenge. Apatinib (a tyrosine kinase inhibitor) is a promising antiangiogenic agent for the treatment of metastatic melanoma. However, antiangiogenic monotherapy is prone to acquired drug resistance and has a limited therapeutic effect. The persistence dependence of glycolytic metabolism in antiangiogenic therapy-resistant cells provides evidence that glycolysis inhibitors may enhance the effect of antiangiogenic therapy. So, this study aimed to investigate whether WZB117 (a specific GLUT1 inhibitor) could enhance the anti-tumor effect of apatinib against melanoma and its potential mechanisms. Methods: We investigated the anti-tumor effects of apatinib alone or in combination with WZB117 on human melanoma cell lines (A375 and SK-MEL-28). The MTT assay determined cell viability and the half-maximal inhibitory concentration (IC50). Multiple drug effect/combination indexes (CI) analysis was conducted to assess interactions between apatinib and WZB117. Signal transducer and activator of transcription 3 (STAT3) pathway measured by western blotting and immunofluorescence staining. RNA expression analyses were performed using the reverse transcription-quantitative PCR method. Results: Apatinib and WZB117 showed dose and time-dependent growth inhibitory effects in both melanoma cells. The IC50 of apatinib at 48 h in A375 and SK-MEL-28 cells was 62.58 and 59.61 μM, respectively, while the IC50 of WZB117 was 116.85 and 113.91 μM, respectively. The CI values of the two drugs were 0.538 and 0.544, respectively, indicating a synergistic effect of apatinib combined with WZB117. We also found that glucose consumption and lactate production were suppressed by apatinib plus WZB117 in a dose-dependent manner, paralleled by reducing glycolytic enzyme pyruvate kinase M2 (PKM2). The potential mechanism of the combination was to suppress the phosphorylation of STAT3. Knockdown of STAT3 by siRNA inhibited the expression of PKM2, while the activation of STAT3 by IL-6 increased the expression of PKM2. The effects of IL-6 were attenuated by apatinib combined with WZB117 treatment. Conclusion: WZB117 enhanced the anti-tumor effect of apatinib against melanoma via modulating glycolysis by blocking the STAT3/PKM2 axis, which suggested the combination of apatinib with WZB117 could be a potential therapeutic candidate for melanoma.
Collapse
Affiliation(s)
- Ren-Shu Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Ke Li
- Department of Oncology, The First Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- School of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Jie Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yao-Tiao Deng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Jiang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Huang L, Zhang Z, Li G. DNA strand displacement based surface-enhanced Raman scattering-fluorescence dual-mode nanoprobes for quantification and imaging of vascular endothelial growth factor in living cells. Biosens Bioelectron 2022; 204:114069. [DOI: 10.1016/j.bios.2022.114069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/21/2023]
|
18
|
Piotrowska A, Beserra FP, Wierzbicka JM, Nowak JI, Żmijewski MA. Vitamin D Enhances Anticancer Properties of Cediranib, a VEGFR Inhibitor, by Modulation of VEGFR2 Expression in Melanoma Cells. Front Oncol 2022; 11:763895. [PMID: 35004285 PMCID: PMC8740239 DOI: 10.3389/fonc.2021.763895] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/01/2021] [Indexed: 01/12/2023] Open
Abstract
Regardless of the recent groundbreaking introduction of personalized therapy, melanoma continues to be one of the most lethal skin malignancies. Still, a substantial proportion of patients either fail to respond to the therapy or will relapse over time, representing a challenging clinical problem. Recently, we have shown that vitamin D enhances the effectiveness of classical chemotherapeutics in the human malignant melanoma A375 cell line. In search for new combination strategies and adjuvant settings to improve melanoma patient outcomes in the current study, the effects of cediranib (AZD2171), an oral tyrosine kinase inhibitor of VEGFR1-3, PDGFR, and c-KIT, used in combination either with 1,25(OH)2D3 or with low-calcemic analog calcipotriol were tested on four human malignant melanoma cell lines (A375, MNT-1, RPMI-7951, and SK-MEL-28). Melanoma cells were pretreated with vitamin D and subsequently exposed to cediranib. We observed a marked decrease in melanoma cell proliferation (A375 and SK-MEL-28), G2/M cell cycle arrest, and a significant decrease in melanoma cell mobility in experimental conditions used (A375). Surprisingly, concurrently with a very desirable decrease in melanoma cell proliferation and mobility, we noticed the upregulation of VEGFR2 at both protein and mRNA levels. No effect of vitamin D was observed in MNT-1 and RPMI-7951 melanoma cells. It seems that vitamin D derivatives enhance cediranib efficacy by modulation of VEGFR2 expression in melanoma cells expressing VEGFR2. In conclusion, our experiments demonstrated that vitamin D derivatives hold promise as novel adjuvant candidates to conquer melanoma, especially in patients suffering from vitamin D deficiency. However, further extensive research is indispensable to reliably assess their potential benefits for melanoma patients.
Collapse
Affiliation(s)
- Anna Piotrowska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Joanna Irena Nowak
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
19
|
Abstract
Melanoma is a relentless type of skin cancer which involves myriad signaling pathways which regulate many cellular processes. This makes melanoma difficult to treat, especially when identified late. At present, therapeutics include chemotherapy, surgical resection, biochemotherapy, immunotherapy, photodynamic and targeted approaches. These interventions are usually administered as either a single-drug or in combination, based on tumor location, stage, and patients' overall health condition. However, treatment efficacy generally decreases as patients develop treatment resistance. Genetic profiling of melanocytes and the discovery of novel molecular factors involved in the pathogenesis of melanoma have helped to identify new therapeutic targets. In this literature review, we examine several newly approved therapies, and briefly describe several therapies being assessed for melanoma. The goal is to provide a comprehensive overview of recent developments and to consider future directions in the field of melanoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Pavan Kumar Dhanyamraju, Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA17033, USA. Tel: +1-6096474712, E-mail:
| | - Trupti N. Patel
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
20
|
Lee H, Lee H, Park S, Kim M, Park JY, Jin H, Oh K, Bae J, Yang Y, Choi HK. Integrative Metabolomic and Lipidomic Profiling of Lung Squamous Cell Carcinoma for Characterization of Metabolites and Intact Lipid Species Related to the Metastatic Potential. Cancers (Basel) 2021; 13:4179. [PMID: 34439333 PMCID: PMC8391613 DOI: 10.3390/cancers13164179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 11/29/2022] Open
Abstract
SQCC is a major type of NSCLC, which is a major cause of cancer-related deaths, and there were no reports regarding the prediction of metastatic potential of lung SQCC by metabolomic and lipidomic profiling. In this study, metabolomic and lipidomic profiling of lung SQCC were performed to predict its metastatic potential and to suggest potential therapeutic targets for the inhibition of lung SQCC metastasis. Human bronchial epithelial cells and four lung SQCC cell lines with different metastatic potentials were analyzed using gas chromatography-mass spectrometry and direct infusion-mass spectrometry. Based on the obtained metabolic and lipidomic profiles, we constructed models to predict the metastatic potential of lung SQCC; glycerol, putrescine, β-alanine, hypoxanthine, inosine, myo-inositol, phosphatidylinositol (PI) 18:1/18:1, and PI 18:1/20:4 were suggested as characteristic metabolites and intact lipid species associated with lung SQCC metastatic potential. In this study, we established predictive models for the metastatic potential of lung SQCC; furthermore, we identified metabolites and intact lipid species relevant to lung SQCC metastatic potential that may serve as potential therapeutic targets for the inhibition of lung SQCC metastasis.
Collapse
Affiliation(s)
- Heayyean Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (H.L.); (H.L.); (M.K.); (K.O.); (J.B.)
| | - Hwanhui Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (H.L.); (H.L.); (M.K.); (K.O.); (J.B.)
| | - Sujeong Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04312, Korea; (S.P.); (J.Y.P.)
| | - Myeongsun Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (H.L.); (H.L.); (M.K.); (K.O.); (J.B.)
| | - Ji Young Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04312, Korea; (S.P.); (J.Y.P.)
| | - Hanyong Jin
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea;
| | - Kyungsoo Oh
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (H.L.); (H.L.); (M.K.); (K.O.); (J.B.)
| | - Jeehyeon Bae
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (H.L.); (H.L.); (M.K.); (K.O.); (J.B.)
| | - Young Yang
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04312, Korea; (S.P.); (J.Y.P.)
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (H.L.); (H.L.); (M.K.); (K.O.); (J.B.)
| |
Collapse
|
21
|
Clinical Significance of PDCD4 in Melanoma by Subcellular Expression and in Tumor-Associated Immune Cells. Cancers (Basel) 2021; 13:cancers13051049. [PMID: 33801444 PMCID: PMC7958624 DOI: 10.3390/cancers13051049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary While targeting programmed cell death (PDCD) 1 is a central treatment against melanoma, little is known about the related protein PDCD4. We defined differences in melanoma PDCD4 subcellular localization (either total cellular or nuclear-only) during oncogenesis, evaluated its presence on tumor-infiltrating immune cells, and determined its impact on survival. High PDCD4 expression resulted in improved survival in patients with primary and intracranial but not extracranial metastatic melanoma. High PDCD4 levels in surrounding tumor tissue were also associated with increased infiltrating immune cells. PDCD4 may be a potentially useful biomarker in melanoma to help guide our understanding of patient prognosis. Methods to increase PDCD4 in those with melanoma brain metastases may also help improve disease response. Abstract Little is known about the subcellular localization and function of programmed cell death 4 (PDCD4) in melanoma. Our past studies suggest PDCD4 interacts with Pleckstrin Homology Domain Containing A5 (PLEKHA5) to influence melanoma brain metastasis outcomes, as high intracranial PDCD4 expression leads to improved survival. We aimed to define the subcellular distribution of PDCD4 in melanoma and in the tumor microenvironment during neoplastic progression and its impact on clinical outcomes. We analyzed multiple tissue microarrays with well-annotated clinicopathological variables using quantitative immunofluorescence and evaluated single-cell RNA-sequencing on a brain metastasis sample to characterize PDCD4+ immune cell subsets. We demonstrate differences in PDCD4 expression during neoplastic progression, with high tumor and stromal PDCD4 levels associated with improved survival in primary melanomas and in intracranial metastases, but not in extracranial metastatic disease. While the expression of PDCD4 is well-documented on CD8+ T cells and natural killer cells, we show that it is also found on B cells and mast cells. PDCD4 expression in the tumor microenvironment is associated with increased immune cell infiltration. Further studies are needed to define the interaction of PDCD4 and PLEKHA5 and to evaluate the utility of this pathway as a therapeutic target in melanoma brain metastasis.
Collapse
|
22
|
Lokesh R, Jeyakanthan J, Kannabiran K. Targeting VEGFR2 protein by marine Streptomyces globosus VITLGK011-derived compound BECA: An in vitro and in silico analysis. J Basic Microbiol 2020; 60:983-993. [PMID: 33103250 DOI: 10.1002/jobm.202000461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 11/08/2022]
Abstract
This study investigates the anticancer cytotoxic mechanism of action of benzoyloxy-ethyl-carbamic acid (BECA) produced by Streptomyces globosus VITLGK011. Flow cytometry analysis confirmed that BECA (at IC50 : 3.12 µg/ml) treatment for 24 h induced apoptosis in 60% of cells. Schrodinger Maestro tools such as QikProp and DFT were used to confirm that BECA is an eligible drug-like molecule, with suitable physiochemical properties. Glide XP tool was used to perform induced-fit docking between BECA and 30 cancer drug target proteins. The highest significance was observed for VEGFR2 protein (-6.7 kcal/mol). GROMACS tool was used to perform molecular dynamic simulation between BECA and VEGFR2 protein for 40 ns. Root mean square deviation, root mean square fluctuation, H-bond, and trajectory analysis, confirmed that BECA is a suitable inhibitor of VEGFR2 protein. Results conclude that BECA is a valid VEGFR2 inhibitor, and it thus exerts the observed anticancer cytotoxicity against MCF-7 cells.
Collapse
Affiliation(s)
- Ravi Lokesh
- Department of Botany, St. Joseph's College (Autonomous), Bengaluru, India
| | | | - Krishnan Kannabiran
- Department of Biomedical Sciences, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
23
|
Resistance of melanoma cells to anticancer treatment: a role of vascular endothelial growth factor. Postepy Dermatol Alergol 2020; 37:11-18. [PMID: 32467677 PMCID: PMC7247075 DOI: 10.5114/ada.2020.93378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Melanoma is one of the most aggressive and resistant to treatment neoplasms. There are still many challenges despite many promising advances in anticancer treatment. Currently, the main problem for all types of treatment is associated with heterogeneity. Due to heterogeneity of cancer cells, "precise" targeting of a medicine against a single phenotype limits the efficacy of treatment and affects resistance to applied therapy. Therefore it is important to understand aetiology and reasons for heterogeneity in order to develop effective and long-lasting treatment. This review summarises roles of vascular endothelial growth factor (VEGF) that may stimulate growth of a melanoma tumour irrespective of its proangiogenic effects, contributing to cancer heterogeneity. VEGF triggers processes associated with extracellular matrix remodelling, cell migration, invasion, angiogenesis, inhibition of immune responses and favours phenotypic plasticity and epithelial-mesenchymal transition. Consequently, it participates in mechanisms of interactions between melanoma cancer cells and microenvironment and it can modify sensitivity to therapeutic factors.
Collapse
|
24
|
Jackett LA, Scolyer RA. A Review of Key Biological and Molecular Events Underpinning Transformation of Melanocytes to Primary and Metastatic Melanoma. Cancers (Basel) 2019; 11:cancers11122041. [PMID: 31861163 PMCID: PMC6966527 DOI: 10.3390/cancers11122041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/02/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a major public health concern that is responsible for significant morbidity and mortality, particularly in countries such as New Zealand and Australia where it is the commonest cause of cancer death in young adults. Until recently, there were no effective drug therapies for patients with advanced melanoma however significant advances in our understanding of the biological and molecular basis of melanoma in recent decades have led to the development of revolutionary treatments, including targeted molecular therapy and immunotherapy. This review summarizes our current understanding of the key events in the pathway of melanomagenesis and discusses the role of genomic analysis as a potential tool for improved diagnostic evaluation, prognostication and treatment strategies. Ultimately, it is hoped that a continued deeper understanding of the mechanisms of melanomagenesis will lead to the development of even more effective treatments that continue to provide better outcomes for patients with melanoma.
Collapse
Affiliation(s)
- Louise A. Jackett
- Melanoma Institute Australia, 2065 Sydney, Australia;
- Sydney Medical School, The University of Sydney, 2050 Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, 2050 Sydney, Australia
- Department of Anatomical Pathology, Austin Hospital, 3084 Melbourne, Australia
| | - Richard A. Scolyer
- Melanoma Institute Australia, 2065 Sydney, Australia;
- Sydney Medical School, The University of Sydney, 2050 Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, 2050 Sydney, Australia
- Correspondence: ; Tel.: +61-299117200; Fax: +61-299549290
| |
Collapse
|
25
|
Atzori MG, Ceci C, Ruffini F, Trapani M, Barbaccia ML, Tentori L, D'Atri S, Lacal PM, Graziani G. Role of VEGFR-1 in melanoma acquired resistance to the BRAF inhibitor vemurafenib. J Cell Mol Med 2019; 24:465-475. [PMID: 31758648 PMCID: PMC6933379 DOI: 10.1111/jcmm.14755] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 12/30/2022] Open
Abstract
The vascular endothelial growth factor receptor‐1 (VEGFR‐1) is a tyrosine kinase receptor frequently expressed in melanoma. Its activation by VEGF‐A or placental growth factor (PlGF) promotes tumour cell survival, migration and invasiveness. Moreover, VEGFR‐1 stimulation contributes to pathological angiogenesis and induces recruitment of tumour‐associated macrophages. Since melanoma acquired resistance to BRAF inhibitors (BRAFi) has been associated with activation of pro‐angiogenic pathways, we have investigated VEGFR‐1 involvement in vemurafenib resistance. Results indicate that human melanoma cells rendered resistant to vemurafenib secrete greater amounts of VEGF‐A and express higher VEGFR‐1 levels compared with their BRAFi‐sensitive counterparts. Transient VEGFR‐1 silencing in susceptible melanoma cells delays resistance development, whereas in resistant cells it increases sensitivity to the BRAFi. Consistently, enforced VEGFR‐1 expression, by stable gene transfection in receptor‐negative melanoma cells, markedly reduces sensitivity to vemurafenib. Moreover, melanoma cells expressing VEGFR‐1 are more invasive than VEGFR‐1 deficient cells and receptor blockade by a specific monoclonal antibody (D16F7 mAb) reduces extracellular matrix invasion triggered by VEGF‐A and PlGF. These data suggest that VEGFR‐1 up‐regulation might contribute to melanoma progression and spreading after acquisition of a drug‐resistant phenotype. Thus, VEGFR‐1 inhibition with D16F7 mAb might be a suitable adjunct therapy for VEGFR‐1 positive tumours with acquired resistance to vemurafenib.
Collapse
Affiliation(s)
| | - Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Mauro Trapani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
26
|
Vascular Endothelial Growth Factor Receptor (VEGFR-2)/KDR Inhibitors: Medicinal Chemistry Perspective. MEDICINE IN DRUG DISCOVERY 2019. [DOI: 10.1016/j.medidd.2019.100009] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
27
|
Runx2 stimulates neoangiogenesis through the Runt domain in melanoma. Sci Rep 2019; 9:8052. [PMID: 31142788 PMCID: PMC6541657 DOI: 10.1038/s41598-019-44552-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022] Open
Abstract
Runx2 is a transcription factor involved in melanoma cell migration and proliferation. Here, we extended the analysis of Runt domain of Runx2 in melanoma cells to deepen understanding of the underlying mechanisms. By the CRISPR/Cas9 system we generated the Runt KO melanoma cells 3G8. Interestingly, the proteome analysis showed a specific protein signature of 3G8 cells related to apoptosis and migration, and pointed out the involvement of Runt domain in the neoangiogenesis process. Among the proteins implicated in angiogenesis we identified fatty acid synthase, chloride intracellular channel protein-4, heat shock protein beta-1, Rho guanine nucleotide exchange factor 1, D-3-phosphoglycerate dehydrogenase, myosin-1c and caveolin-1. Upon querying the TCGA provisional database for melanoma, the genes related to these proteins were found altered in 51.36% of total patients. In addition, VEGF gene expression was reduced in 3G8 as compared to A375 cells; and HUVEC co-cultured with 3G8 cells expressed lower levels of CD105 and CD31 neoangiogenetic markers. Furthermore, the tube formation assay revealed down-regulation of capillary-like structures in HUVEC co-cultured with 3G8 in comparison to those with A375 cells. These findings provide new insight into Runx2 molecular details which can be crucial to possibly propose it as an oncotarget of melanoma.
Collapse
|
28
|
Hsu MC, Pan MR, Hung WC. Two Birds, One Stone: Double Hits on Tumor Growth and Lymphangiogenesis by Targeting Vascular Endothelial Growth Factor Receptor 3. Cells 2019; 8:cells8030270. [PMID: 30901976 PMCID: PMC6468620 DOI: 10.3390/cells8030270] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023] Open
Abstract
Vascular endothelial growth factor receptor 3 (VEGFR3) has been known for its involvement in tumor-associated lymphangiogenesis and lymphatic metastasis. The VEGFR3 signaling is stimulated by its main cognate ligand, vascular endothelial growth factor C (VEGF-C), which in turn promotes tumor progression. Activation of VEGF-C/VEGFR3 signaling in lymphatic endothelial cells (LECs) was shown to enhance the proliferation of LECs and the formation of lymphatic vessels, leading to increased lymphatic metastasis of tumor cells. In the past decade, the expression and pathological roles of VEGFR3 in tumor cells have been described. Moreover, the VEGF-C/VEGFR3 axis has been implicated in regulating immune tolerance and suppression. Therefore, the inhibition of the VEGF-C/VEGFR3 axis has emerged as an important therapeutic strategy for the treatment of cancer. In this review, we discuss the current findings related to VEGF-C/VEGFR3 signaling in cancer progression and recent advances in the development of therapeutic drugs targeting VEGF-C/VEGFR3.
Collapse
Affiliation(s)
- Ming-Chuan Hsu
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Mei-Ren Pan
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
29
|
Abstract
Melanoma represents the most aggressive and the deadliest form of skin cancer. Current therapeutic approaches include surgical resection, chemotherapy, photodynamic therapy, immunotherapy, biochemotherapy, and targeted therapy. The therapeutic strategy can include single agents or combined therapies, depending on the patient’s health, stage, and location of the tumor. The efficiency of these treatments can be decreased due to the development of diverse resistance mechanisms. New therapeutic targets have emerged from studies of the genetic profile of melanocytes and from the identification of molecular factors involved in the pathogenesis of the malignant transformation. In this review, we aim to survey therapies approved and under evaluation for melanoma treatment and relevant research on the molecular mechanisms underlying melanomagenesis.
Collapse
Affiliation(s)
- Beatriz Domingues
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Manuel Lopes
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Hospital S João, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Helena Pópulo
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
30
|
Neuber C, Belter B, Meister S, Hofheinz F, Bergmann R, Pietzsch HJ, Pietzsch J. Overexpression of Receptor Tyrosine Kinase EphB4 Triggers Tumor Growth and Hypoxia in A375 Melanoma Xenografts: Insights from Multitracer Small Animal Imaging Experiments. Molecules 2018; 23:E444. [PMID: 29462967 PMCID: PMC6017846 DOI: 10.3390/molecules23020444] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 12/15/2022] Open
Abstract
Experimental evidence has associated receptor tyrosine kinase EphB4 with tumor angiogenesis also in malignant melanoma. Considering the limited in vivo data available, we have conducted a systematic multitracer and multimodal imaging investigation in EphB4-overexpressing and mock-transfected A375 melanoma xenografts. Tumor growth, perfusion, and hypoxia were investigated by positron emission tomography. Vascularization was investigated by fluorescence imaging in vivo and ex vivo. The approach was completed by magnetic resonance imaging, radioluminography ex vivo, and immunohistochemical staining for blood and lymph vessel markers. Results revealed EphB4 to be a positive regulator of A375 melanoma growth, but a negative regulator of tumor vascularization. Resulting in increased hypoxia, this physiological characteristic is considered as highly unfavorable for melanoma prognosis and therapy outcome. Lymphangiogenesis, by contrast, was not influenced by EphB4 overexpression. In order to distinguish between EphB4 forward and EphrinB2, the natural EphB4 ligand, reverse signaling a specific EphB4 kinase inhibitor was applied. Blocking experiments show EphrinB2 reverse signaling rather than EphB4 forward signaling to be responsible for the observed effects. In conclusion, functional expression of EphB4 is considered a promising differentiating characteristic, preferentially determined by non-invasive in vivo imaging, which may improve personalized theranostics of malignant melanoma.
Collapse
Affiliation(s)
- Christin Neuber
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Birgit Belter
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Sebastian Meister
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Frank Hofheinz
- Department Positron Emission Tomography, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Ralf Bergmann
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Hans-Jürgen Pietzsch
- Department Radionuclide Theragnostics, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Jens Pietzsch
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
31
|
Zhang Y, Chen Y, Zhang D, Wang L, Lu T, Jiao Y. Discovery of Novel Potent VEGFR-2 Inhibitors Exerting Significant Antiproliferative Activity against Cancer Cell Lines. J Med Chem 2017; 61:140-157. [DOI: 10.1021/acs.jmedchem.7b01091] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Yanmin Zhang
- Laboratory
of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yadong Chen
- Laboratory
of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Danfeng Zhang
- School
of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Lu Wang
- School
of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Tao Lu
- Laboratory
of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
- School
of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
- State
Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yu Jiao
- School
of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| |
Collapse
|
32
|
Katas H, Thian Sian T, Abdul Ghaf M. Topical Temperature-sensitive Gel Containing DsiRNA-chitosan Nanoparticles for Potential Treatment of Skin Cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.3923/tmr.2017.1.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
33
|
Ren X, Xie W, Wang Y, Xu M, Liu F, Tang M, Li C, Wang M, Zhang J. VEGFR2-targeted fusion antibody improved NK cell-mediated immunosurveillance against K562 cells. Immunol Res 2017; 64:1060-70. [PMID: 27154226 DOI: 10.1007/s12026-016-8800-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MHC class I polypeptide-related sequence A (MICA), which is normally expressed on cancer cells, activates NK cells via NK group 2-member D pathway. However, some cancer cells escape NK-mediated immune surveillance by shedding membrane MICA causing immune suppression. To address this issue, we designed an antibody-MICA fusion targeting tumor-specific antigen (vascular endothelial growth factor receptor 2, VEGFR2) based on our patented antibody (mAb04) against VEGFR2. In vitro results demonstrate that the fusion antibody retains both the antineoplastic and the immunomodulatory activity of mAb04. Further, we revealed that it enhanced NK-mediated immunosurveillance against K562 cells through increasing degranulation and cytokine production of NK cells. The overall data suggest our new fusion protein provides a promising approach for cancer-targeted immunotherapy and has prospects for potential application of chronic myeloid leukemia.
Collapse
Affiliation(s)
- Xueyan Ren
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Wei Xie
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Youfu Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Menghuai Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Fang Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Mingying Tang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Chenchen Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China.
| | - Juan Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
34
|
Kim HY, Lee H, Kim SH, Jin H, Bae J, Choi HK. Discovery of potential biomarkers in human melanoma cells with different metastatic potential by metabolic and lipidomic profiling. Sci Rep 2017; 7:8864. [PMID: 28821754 PMCID: PMC5562697 DOI: 10.1038/s41598-017-08433-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022] Open
Abstract
Malignant melanoma, characterized by its ability to metastasize to other organs, is responsible for 90% of skin cancer mortality. To investigate alterations in the cellular metabolome and lipidome related to melanoma metastasis, gas chromatography-mass spectrometry (GC-MS) and direct infusion-mass spectrometry (DI-MS)-based metabolic and lipidomic profiling were performed on extracts of normal human melanocyte (HEMn-LP), low metastatic melanoma (A375, G361), and highly metastatic melanoma (A2058, SK-MEL-28) cell lines. In this study, metabolomic analysis identified aminomalonic acid as a novel potential biomarker to discriminate between different stages of melanoma metastasis. Uptake and release of major metabolites as hallmarks of cancer were also measured between high and low metastatic melanoma cells. Lipid analysis showed a progressive increase in phosphatidylinositol (PI) species with saturated and monounsaturated fatty acyl chains, including 16:0/18:0, 16:0/18:1, 18:0/18:0, and 18:0/18:1, with increasing metastatic potential of melanoma cells, defining these lipids as possible biomarkers. In addition, a partial-least-squares projection to latent structure regression (PLSR) model for the prediction of metastatic properties of melanoma was established, and central metabolic and lipidomic pathways involved in the increased motility and metastatic potential of melanoma cells were identified as therapeutic targets. These results could be used to diagnose and control of melanoma metastasis.
Collapse
Affiliation(s)
- Hye-Youn Kim
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hwanhui Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - So-Hyun Kim
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hanyong Jin
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jeehyeon Bae
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
35
|
Bekeschus S, Rödder K, Fregin B, Otto O, Lippert M, Weltmann KD, Wende K, Schmidt A, Gandhirajan RK. Toxicity and Immunogenicity in Murine Melanoma following Exposure to Physical Plasma-Derived Oxidants. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4396467. [PMID: 28761621 PMCID: PMC5518506 DOI: 10.1155/2017/4396467] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Metastatic melanoma is an aggressive and deadly disease. Therapeutic advance has been achieved by antitumor chemo- and radiotherapy. These modalities involve the generation of reactive oxygen and nitrogen species, affecting cellular viability, migration, and immunogenicity. Such species are also created by cold physical plasma, an ionized gas capable of redox modulating cells and tissues without thermal damage. Cold plasma has been suggested for anticancer therapy. Here, melanoma cell toxicity, motility, and immunogenicity of murine metastatic melanoma cells were investigated following plasma exposure in vitro. Cells were oxidized by plasma, leading to decreased metabolic activity and cell death. Moreover, plasma decelerated melanoma cell growth, viability, and cell cycling. This was accompanied by increased cellular stiffness and upregulation of zonula occludens 1 protein in the cell membrane. Importantly, expression levels of immunogenic cell surface molecules such as major histocompatibility complex I, calreticulin, and melanocortin receptor 1 were significantly increased in response to plasma. Finally, plasma treatment significantly decreased the release of vascular endothelial growth factor, a molecule with importance in angiogenesis. Altogether, these results suggest beneficial toxicity of cold plasma in murine melanomas with a concomitant immunogenicity of potential interest in oncology.
Collapse
Affiliation(s)
- Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Katrin Rödder
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Bob Fregin
- ZIK HIKE, Fleischmannstr. 42-44, 17489 Greifswald, Germany
| | - Oliver Otto
- ZIK HIKE, Fleischmannstr. 42-44, 17489 Greifswald, Germany
| | - Maxi Lippert
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Klaus-Dieter Weltmann
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Kristian Wende
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Anke Schmidt
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Rajesh Kumar Gandhirajan
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| |
Collapse
|
36
|
Fenton SE, Sosman JA, Chandra S. Emerging growth factor receptor antagonists for the treatment of advanced melanoma. Expert Opin Emerg Drugs 2017; 22:165-174. [PMID: 28562096 DOI: 10.1080/14728214.2017.1336537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Therapy for metastatic melanoma has undergone a rapid transformation over the past 5-10 years. Advances in immunotherapy with checkpoint inhibitors, including both anti-CTLA-4 and anti-PD-1/PD-L1, have led to durable responses in up to 50% of patients. As our understanding of the processes driving the transformation of melanocytes has improved, progress in targeted therapies has also continued. Areas covered: Angiogenesis and the tumor's dependence on an expanded vascular supply has been a target for novel therapies since the 1970's, as this tissue is derived from endothelial cells that are genetically stable in adults. A phase II trial studying combined therapy with bevacizumab (an inhibitor of angiogenesis) and ipilimumab found promising results. Other agents such as sorafenib have not been as successful, failing to extend progression free or overall survival in clinical trials. In this paper other targeted growth factor inhibitors will also be discussed. Expert opinion: Ultimately, melanoma may not be vulnerable solely to chemotherapy or targeted therapy, but may be efficaciously treated with immunotherapy due to its high mutational rate resulting in the expression of numerous neo-antigens. Therapies with combinations of agents including growth factor receptor and either other targeted therapies or immunotherapy may be a promising complimentary approach.
Collapse
|
37
|
Tan LY, Martini C, Fridlender ZG, Bonder CS, Brown MP, Ebert LM. Control of immune cell entry through the tumour vasculature: a missing link in optimising melanoma immunotherapy? Clin Transl Immunology 2017; 6:e134. [PMID: 28435677 PMCID: PMC5382436 DOI: 10.1038/cti.2017.7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 12/25/2022] Open
Abstract
Metastatic melanoma remains a fatal disease to many worldwide, even after the breakthrough introduction of targeted therapies such as BRAF inhibitors and immune checkpoint blockade therapies such as CTLA-4 and PD-1 inhibitors. With advances in our understanding of this disease, as well as the increasing data gathered from patient studies, the significance of the host immune response to cancer progression and response to treatment is becoming clear. More specifically, the presence of intratumoral CD8+ cytotoxic T-cells correlates with better prognosis whereas the accumulation of monocytes/macrophages and neutrophils in the tumour is often associated with worse prognosis. Access and infiltration of circulating leukocytes into the tumour is governed by adhesion molecules and chemokines expressed by the endothelial cells of the vasculature. This review focuses on the adhesion molecules and chemokines which control the homing of CD8+ cytotoxic T-cells, monocytes and neutrophils to peripheral tissues, including tumours. We discuss the role of these leukocyte subsets in regulating melanoma growth, and detail the mechanisms used by tumours to selectively recruit or exclude these leukocytes for their own advantage. In doing so, we bring to light an underappreciated component of tumour biology which should be considered in combination with current treatments to selectively alter the leukocyte composition of tumours and ultimately enhance treatment outcome.
Collapse
Affiliation(s)
- Lih Yin Tan
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Carmela Martini
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Michael P Brown
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.,Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Lisa M Ebert
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
38
|
Thurneysen S, Cheng PF, Nagel HW, Kunz M, Jaberg-Bentele N, Nägeli M, Ziegler M, Guenova E, Goldinger SM, Mangana J, Levesque MP, Dummer R. An exploratory study investigating the metabolic activity and local cytokine profile in patients with melanoma treated with pazopanib and paclitaxel. Br J Dermatol 2016; 175:966-978. [PMID: 27168024 DOI: 10.1111/bjd.14727] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND There is a medical need for new drugs in patients with BRAF wild-type metastatic melanoma. Pazopanib is a multitarget tyrosine kinase inhibitor with antitumour and antiangiogenic activity. OBJECTIVES The primary aim was to investigate the metabolic response to pazopanib monotherapy and pazopanib plus paclitaxel in patients with BRAF wild-type melanoma. Secondary end points were the early cytokine and chemokine profiles and histological findings. METHODS Pazopanib (400 mg twice daily) was administered orally from days 1 to 10 and from days 14 to 70. An intravenous infusion with paclitaxel (150 mg m-2 body surface) was administered on days 14, 35 and 56. Metabolic response evaluation was performed before treatment, after treatment with pazopanib (day 10) and after treatment with pazopanib and paclitaxel (day 70). Skin biopsy of metastatic tissue for chemokine and cytokine expression analysis and histology and immunohistochemistry (CD68, CD163) evaluation, and blood samples were taken at the same time points. RESULTS Two patients failed screening and 17 were dosed. Of 67 adverse events, nine (13%) were grade 3 or 4. Five of 14 evaluable patients had a partial metabolic response at day 10 under pazopanib monotherapy. The response rate at day 70 under combined pazopanib-paclitaxel treatment was 0%. Immunohistochemistry revealed an increase of M2-like macrophages in nonresponders compared with responders. We observed a significant upregulation of five cytokines (CXCL1, CXCL2, CXCL13, CCL22 and SPP1) in responding vs. nonresponding lesions. Overall, the median progression-free survival was 70 days (range 5-331), which did not differ significantly between responders (148 days) and nonresponders (70 days, P = 0·17). CONCLUSIONS In this patient population pazopanib efficacy was limited. Response is associated with low M2-like macrophage density and increased expression of several chemokines.
Collapse
Affiliation(s)
- S Thurneysen
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - P F Cheng
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - H W Nagel
- Department of Nuclear Medicine, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - M Kunz
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - N Jaberg-Bentele
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - M Nägeli
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - M Ziegler
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - E Guenova
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - S M Goldinger
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - J Mangana
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - M P Levesque
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland
| | - R Dummer
- Department of Dermatology, University Hospital Zurich, Gloriastraße 31, 8091, Zurich, Switzerland.
| |
Collapse
|
39
|
Das AM, Pescatori M, Vermeulen CE, Rens JAP, Seynhaeve ALB, Koning GA, Eggermont AMM, Ten Hagen TLM. Melanomas prevent endothelial cell death under restrictive culture conditions by signaling through AKT and p38 MAPK/ ERK-1/2 cascades. Oncoimmunology 2016; 5:e1219826. [PMID: 27853641 PMCID: PMC5087299 DOI: 10.1080/2162402x.2016.1219826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022] Open
Abstract
Although melanoma progression and staging is clinically well characterized, a large variation is observed in pathogenesis, progression, and therapeutic responses. Clearly, intrinsic characteristics of melanoma cells contribute to this variety. An important factor, in both progression of the disease and response to therapy, is the tumor-associated vasculature. We postulate that melanoma cells communicate with endothelial cells (ECs) in order to establish a functional and supportive blood supply. We investigated the angiogenic potential of human melanoma cell lines by monitoring the survival of ECs upon exposure to melanoma conditioned medium (CM), under restrictive conditions. We observed long-term (up to 72 h) EC survival under hypoxic conditions upon treatment with all melanoma CMs. No such survival effect was observed with the CM of melanocytes. The CM of pancreatic and breast tumor cell lines did not show a long-term survival effect, suggesting that the survival factor is specific to melanoma cells. Furthermore, all size fractions (up to < 1 kDa) of the melanoma CM induced long-term survival of ECs. The survival effect observed by the < 1 kDa fraction excludes known pro-angiogenic factors. Heat inactivation and enzymatic digestion of the CM did not inactivate the survival factor. Global gene expression and pathway analysis suggest that this effect is mediated in part via the AKT and p38 MAPK/ ERK-1/2 signaling axis. Taken together, these data indicate the production of (a) survival factor/s (< 1 kDa) by melanoma cell lines, which enables long-term survival of ECs and promotes melanoma-induced angiogenesis.
Collapse
Affiliation(s)
- Asha M Das
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Mario Pescatori
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Cindy E Vermeulen
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Joost A P Rens
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Ann L B Seynhaeve
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Gerben A Koning
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Alexander M M Eggermont
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Timo L M Ten Hagen
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| |
Collapse
|
40
|
Aktary Z, Bertrand JU, Larue L. The WNT-less wonder: WNT-independent β-catenin signaling. Pigment Cell Melanoma Res 2016; 29:524-40. [PMID: 27311806 DOI: 10.1111/pcmr.12501] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
Abstract
β-catenin is known as an Armadillo protein that regulates gene expression following WNT pathway activation. However, WNT-independent pathways also activate β-catenin. During the establishment of the melanocyte lineage, β-catenin plays an important role. In the context of physiopathology, β-catenin is activated genetically or transiently in various cancers, including melanoma, where it can be found in the nucleus of tumors. In this review, we discuss alternative pathways that activate β-catenin independent of WNTs and highlight what is known regarding these pathways in melanoma. We also discuss the role of β-catenin as a transcriptional regulator in various cell types, with emphasis on the different transcription factors it associates with independent of WNT induction. Finally, the role of WNT-independent β-catenin in melanocyte development and melanomagenesis is also discussed.
Collapse
Affiliation(s)
- Zackie Aktary
- Normal and Pathological Development of Melanocytes, INSERM U1021, Institut Curie, PSL Research University, Orsay, France.,CNRS UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, Orsay, France
| | - Juliette U Bertrand
- Normal and Pathological Development of Melanocytes, INSERM U1021, Institut Curie, PSL Research University, Orsay, France.,CNRS UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, Orsay, France
| | - Lionel Larue
- Normal and Pathological Development of Melanocytes, INSERM U1021, Institut Curie, PSL Research University, Orsay, France. .,CNRS UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France. .,Equipe Labellisée Ligue Contre le Cancer, Orsay, France.
| |
Collapse
|
41
|
Caporali S, Alvino E, Lacal PM, Levati L, Giurato G, Memoli D, Caprini E, Antonini Cappellini GC, D'Atri S. Targeting the PI3K/AKT/mTOR pathway overcomes the stimulating effect of dabrafenib on the invasive behavior of melanoma cells with acquired resistance to the BRAF inhibitor. Int J Oncol 2016; 49:1164-74. [PMID: 27572607 DOI: 10.3892/ijo.2016.3594] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/19/2016] [Indexed: 11/06/2022] Open
Abstract
BRAF inhibitors (BRAFi) have proven clinical benefits in patients with BRAF-mutant melanoma. However, acquired resistance eventually arises. The effects of BRAFi on melanoma cell proliferation and survival have been extensively studied, and several mechanisms involved in acquired resistance to the growth suppressive activity of these drugs have been identified. Much less is known about the impact of BRAFi, and in particular of dabrafenib, on the invasive potential of melanoma cells. In the present study, the BRAF-mutant human melanoma cell line A375 and its dabrafenib-resistant subline A375R were analyzed for invasive capacity, expression of vascular endothelial growth factor receptor (VEGFR)-2, and secretion of VEGF-A and matrix metalloproteinase (MMP)-9, under basal conditions or in response to dabrafenib. The consequences of inhibiting the PI3K/AKT/mTOR pathway on A375R cell responses to dabrafenib were also evaluated. We found that A375R cells were more invasive and secreted higher levels of VEGF-A and MMP-9 as compared with A375 cells. Dabrafenib reduced invasiveness, VEGFR-2 expression and VEGF-A secretion in A375 cells, whereas it increased invasiveness, VEGF-A and MMP-9 release in A375R cells. In these latter cells, the stimulating effects of dabrafenib on the invasive capacity were markedly impaired by the anti-VEGF‑A antibody bevacizumab, or by AKT1 silencing. A375R cells were not cross-resistant to the PI3K/mTOR inhibitor GSK2126458A. Moreover, this inhibitor given in combination with dabrafenib efficiently counteracted the stimulating effects of the BRAFi on invasiveness and VEGF-A and MMP-9 secretion. Our data demonstrate that melanoma cells with acquired resistance to dabrafenib possess a more invasive phenotype which is further stimulated by exposure to the drug. Substantial evidence indicates that continuing BRAFi therapy beyond progression produces a clinical benefit. Our results suggest that after the development of resistance, a regimen combining BRAFi with bevacizumab or with inhibitors of the PI3K/AKT/mTOR pathway might be more effective than BRAFi monotherapy.
Collapse
Affiliation(s)
- Simona Caporali
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata-IRCCS, Rome, Italy
| | - Ester Alvino
- Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata-IRCCS, Rome, Italy
| | - Lauretta Levati
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata-IRCCS, Rome, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, University of Salerno, Baronissi (SA), Italy
| | - Domenico Memoli
- Laboratory of Molecular Medicine and Genomics, University of Salerno, Baronissi (SA), Italy
| | - Elisabetta Caprini
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata-IRCCS, Rome, Italy
| | | | - Stefania D'Atri
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata-IRCCS, Rome, Italy
| |
Collapse
|
42
|
Ultrafiltration and Microarray for Detection of Microbial Source Tracking Marker and Pathogen Genes in Riverine and Marine Systems. Appl Environ Microbiol 2016; 82:1625-1635. [PMID: 26729716 DOI: 10.1128/aem.02583-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 12/24/2015] [Indexed: 01/12/2023] Open
Abstract
Pathogen identification and microbial source tracking (MST) to identify sources of fecal pollution improve evaluation of water quality. They contribute to improved assessment of human health risks and remediation of pollution sources. An MST microarray was used to simultaneously detect genes for multiple pathogens and indicators of fecal pollution in freshwater, marine water, sewage-contaminated freshwater and marine water, and treated wastewater. Dead-end ultrafiltration (DEUF) was used to concentrate organisms from water samples, yielding a recovery efficiency of >95% for Escherichia coli and human polyomavirus. Whole-genome amplification (WGA) increased gene copies from ultrafiltered samples and increased the sensitivity of the microarray. Viruses (adenovirus, bocavirus, hepatitis A virus, and human polyomaviruses) were detected in sewage-contaminated samples. Pathogens such as Legionella pneumophila, Shigella flexneri, and Campylobacter fetus were detected along with genes conferring resistance to aminoglycosides, beta-lactams, and tetracycline. Nonmetric dimensional analysis of MST marker genes grouped sewage-spiked freshwater and marine samples with sewage and apart from other fecal sources. The sensitivity (percent true positives) of the microarray probes for gene targets anticipated in sewage was 51 to 57% and was lower than the specificity (percent true negatives; 79 to 81%). A linear relationship between gene copies determined by quantitative PCR and microarray fluorescence was found, indicating the semiquantitative nature of the MST microarray. These results indicate that ultrafiltration coupled with WGA provides sufficient nucleic acids for detection of viruses, bacteria, protozoa, and antibiotic resistance genes by the microarray in applications ranging from beach monitoring to risk assessment.
Collapse
|
43
|
Lugowska I, Kowalska M, Fuksiewicz M, Kotowicz B, Mierzejewska E, Koseła-Paterczyk H, Szamotulska K, Rutkowski P. Serum markers in early-stage and locally advanced melanoma. Tumour Biol 2015; 36:8277-85. [PMID: 26002577 PMCID: PMC4672018 DOI: 10.1007/s13277-015-3564-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/13/2015] [Indexed: 11/16/2022] Open
Abstract
The identification of prognostic factors in cutaneous melanoma allows choosing the most effective treatment, especially in group of patients with locoregional disease. Markers related to carcinogenesis and angiogenesis in particular have effect on the course of the disease. The aim of this study was to evaluate clinical utility of vascular endothelial growth factor (VEGF), matrix metalloproteinase 2 (MMP-2), MMP-9, tissue inhibitors of metalloproteinase 1 (TIMP-1), and YKL-40 in serum of melanoma patients at pathological stages I-III. We included 148 adult patients with melanoma. The median follow-up was 40 months. Disease recurrence was observed in 43 patients; 3-year disease-free survival (DFS) rate was 71.7%; 35 patients died; and the 3-year overall survival (OS) rate was 85%. Concentrations of VEGF, MMP-2, MMP-9, TIMP-1, and YKL-40 were measured by ELISA kits. VEGF, MMP-9, TIMP-1, and YKL-40 were significantly higher in group of patients than in controls. Increased concentrations of TIMP-1 were related to patient survival, which in the group of lower and increased TIMP-1, disease-free survival amounted to 81 vs. 61% (p = 0.014) and overall survival -88 vs. 82% (p = 0.050), respectively. An increased concentration of YKL-40 was observed in 59% of patients with ulceration and in 26% of patients without ulceration (p = 0.012). We have found a clinically significant correlation between YKL-40 and MMP-9 (rho = 0.363; p = 0.004) as well as YKL-40 and VEGF (rho = 0.306; p = 0.018). In melanoma patients at stages I-III, the high concentrations of TIMP-1 in serum predicted adverse prognosis. YKL-40 was associated with ulceration of primary tumor, which is a very important prognostic factor.
Collapse
Affiliation(s)
- Iwona Lugowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland.
- Department of Epidemiology, Institute of Mother and Child, M. Kasprzak Street 17a, Warsaw, Poland.
| | - Maria Kowalska
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Małgorzata Fuksiewicz
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Beata Kotowicz
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Ewa Mierzejewska
- Department of Epidemiology, Institute of Mother and Child, M. Kasprzak Street 17a, Warsaw, Poland
| | - Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology, Institute of Mother and Child, M. Kasprzak Street 17a, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| |
Collapse
|
44
|
Ott PA, Hodi FS, Buchbinder EI. Inhibition of Immune Checkpoints and Vascular Endothelial Growth Factor as Combination Therapy for Metastatic Melanoma: An Overview of Rationale, Preclinical Evidence, and Initial Clinical Data. Front Oncol 2015; 5:202. [PMID: 26442214 PMCID: PMC4585112 DOI: 10.3389/fonc.2015.00202] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/31/2015] [Indexed: 01/29/2023] Open
Abstract
The role of angiogenesis as a mediator of immune regulation in the tumor microenvironment has recently come into focus. Furthermore, emerging evidence indicates that immunotherapy can lead to immune-mediated vasculopathy in the tumor, suggesting that the tumor vasculature may be an important interface between the tumor-directed immune response and the cancer itself. The advent of immune checkpoint inhibition as an effective immunotherapeutic strategy for many cancers has led to a better understanding of this interface. While the inhibition of angiogenesis through targeting of vascular endothelial growth factor (VEGF) has been used successfully for the treatment of cancer for many years, the mechanisms of its anti-tumor activity remain poorly understood. Initial studies of the complex relationship between angiogenesis, VEGF signaling and the immune system suggest that the combination of immune checkpoint blockade with angiogenesis inhibition has potential. While the majority of this work has been performed in metastatic melanoma, immunotherapy is rapidly showing promise in a broad range of malignancies and efforts to enhance immunotherapy will broadly impact the future of oncology. Here, we review the preclinical rationale and clinical investigations of combined angiogenesis inhibition and immunotherapy/immune checkpoint inhibition as a potentially promising combinatorial approach for cancer treatment.
Collapse
Affiliation(s)
- Patrick A Ott
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - F Stephen Hodi
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Elizabeth I Buchbinder
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
45
|
Kurenova E, Ucar D, Liao J, Yemma M, Gogate P, Bshara W, Sunar U, Seshadri M, Hochwald SN, Cance WG. A FAK scaffold inhibitor disrupts FAK and VEGFR-3 signaling and blocks melanoma growth by targeting both tumor and endothelial cells. Cell Cycle 2015; 13:2542-53. [PMID: 25486195 DOI: 10.4161/15384101.2015.941760] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Melanoma has the highest mortality rate of all skin cancers and a major cause of treatment failure is drug resistance. Tumors heterogeneity requires novel therapeutic strategies and new drugs targeting multiple pathways. One of the new approaches is targeting the scaffolding function of tumor related proteins such as focal adhesion kinase (FAK). FAK is overexpressed in most solid tumors and is involved in multiple protein-protein interactions critical for tumor cell survival, tumor neovascularization, progression and metastasis. In this study, we investigated the anticancer activity of the FAK scaffold inhibitor C4, targeted to the FAK-VEGFR-3 complex, against melanomas. We compared C4 inhibitory effects in BRAF mutant vs BRAF wild type melanomas. C4 effectively caused melanoma tumor regression in vivo, when administered alone and sensitized tumors to chemotherapy. The most dramatic effect of C4 was related to reduction of vasculature of both BRAF wild type and V600E mutant xenograft tumors. The in vivo effects of C4 were assessed in xenograft models using non-invasive multimodality imaging in conjunction with histologic and molecular biology methods. C4 inhibited cell viability, adhesion and motility of melanoma and endothelial cells, specifically blocked phosphorylation of VEGFR-3 and FAK and disrupted their complexes. Specificity of in vivo effects for C4 were confirmed by a decrease in tumor FAK and VEGFR-3 phosphorylation, reduction of vasculogenesis and reduced blood flow. Our collective observations provide evidence that a small molecule inhibitor targeted to the FAK protein-protein interaction site successfully inhibits melanoma growth through dual targeting of tumor and endothelial cells and is effective against both BRAF wild type and mutant melanomas.
Collapse
Affiliation(s)
- Elena Kurenova
- a Department of Surgical Oncology ; Roswell Park Cancer Institute ; Buffalo , NY USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Marinaccio C, Giudice G, Nacchiero E, Robusto F, Opinto G, Lastilla G, Maiorano E, Ribatti D. Interval sentinel lymph nodes in melanoma: a digital pathology analysis of Ki67 expression and microvascular density. Clin Exp Med 2015; 16:383-9. [PMID: 26275365 DOI: 10.1007/s10238-015-0379-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/22/2015] [Indexed: 12/28/2022]
Abstract
The presence of interval sentinel lymph nodes in melanoma is documented in several studies, but controversies still exist about the management of these lymph nodes. In this study, an immunohistochemical evaluation of tumor cell proliferation and neo-angiogenesis has been performed with the aim of establishing a correlation between these two parameters between positive and negative interval sentinel lymph nodes. This retrospective study reviewed data of 23 patients diagnosed with melanoma. Bioptic specimens of interval sentinel lymph node were retrieved, and immunohistochemical reactions on tissue sections were performed using Ki67 as a marker of proliferation and CD31 as a blood vessel marker for the study of angiogenesis. The entire stained tissue sections for each case were digitized using Aperio Scanscope Cs whole-slide scanning platform and stored as high-resolution images. Image analysis was carried out on three selected fields of equal area using IHC Nuclear and Microvessel analysis algorithms to determine positive Ki67 nuclei and vessel number. Patients were divided into positive and negative interval sentinel lymph node groups, and the positive interval sentinel lymph node group was further divided into interval positive with micrometastasis and interval positive with macrometastasis subgroups. The analysis revealed a significant difference between positive and negative interval sentinel lymph nodes in the percentage of Ki67-positive nuclei and mean vessel number suggestive of an increased cellular proliferation and angiogenesis in positive interval sentinel lymph nodes. Further analysis in the interval positive lymph node group showed a significant difference between micro- and macrometastasis subgroups in the percentage of Ki67-positive nuclei and mean vessel number. Percentage of Ki67-positive nuclei was increased in the macrometastasis subgroup, while mean vessel number was increased in the micrometastasis subgroup. The results of this study suggest that the correlation between tumor cell proliferation and neo-angiogenesis in interval sentinel lymph nodes in melanoma could be used as a good predictive marker to distinguish interval positive sentinel lymph nodes with micrometastasis from interval positive lymph nodes with macrometastasis subgroups.
Collapse
Affiliation(s)
- Christian Marinaccio
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Policlinico - Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Giuseppe Giudice
- Department of Plastic and Reconstructive Surgery, University of Bari Medical School, Bari, Italy
| | - Eleonora Nacchiero
- Department of Plastic and Reconstructive Surgery, University of Bari Medical School, Bari, Italy
| | - Fabio Robusto
- Mario Negri Sud Foundation, Santa Maria Imbaro, Italy
| | - Giuseppina Opinto
- Department of Emergency and Organ Transplantation, University of Bari Medical School, Bari, Italy
| | - Gaetano Lastilla
- Department of Emergency and Organ Transplantation, University of Bari Medical School, Bari, Italy
| | - Eugenio Maiorano
- Department of Emergency and Organ Transplantation, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Policlinico - Piazza G. Cesare, 11, 70124, Bari, Italy. .,National Cancer Institute "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
47
|
Hong DS, Kurzrock R, Wheler JJ, Naing A, Falchook GS, Fu S, Kim KB, Davies MA, Nguyen LM, George GC, Xu L, Shumaker R, Ren M, Mink J, Bedell C, Andresen C, Sachdev P, O'Brien JP, Nemunaitis J. Phase I Dose-Escalation Study of the Multikinase Inhibitor Lenvatinib in Patients with Advanced Solid Tumors and in an Expanded Cohort of Patients with Melanoma. Clin Cancer Res 2015; 21:4801-10. [PMID: 26169970 DOI: 10.1158/1078-0432.ccr-14-3063] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 06/09/2015] [Indexed: 01/13/2023]
Abstract
PURPOSE This "3+3" phase I study evaluated the safety, biologic, and clinical activity of lenvatinib, an oral multikinase inhibitor, in patients with solid tumors. EXPERIMENTAL DESIGN Ascending doses of lenvatinib were administered per os twice daily in 28-day cycles. Safety and response were assessed for all patients. Angiogenic and apoptotic factors were tested as possible biomarkers in an expanded melanoma cohort. RESULTS Seventy-seven patients were treated in 3 cohorts: 18 with intermittent twice-daily dosing (7 days on, 7 days off) of 0.1-3.2 mg; 33 with twice-daily dosing of 3.2-12 mg; and 26 with twice-daily dosing of 10 mg (expanded melanoma cohort). Maximum tolerated dose was established at 10 mg per os twice daily. Prominent drug-related toxicities included hypertension (43%), fatigue (42%), proteinuria (39%), and nausea (25%); dose-limiting toxicities included hypertension, fatigue, and proteinuria. Twelve patients (15.6%) achieved partial response (PR, n = 9) or unconfirmed PR (uPR, n = 3), and 19 (24.7%) achieved stable disease (SD) ≥23 weeks. Total PR/uPR/SD ≥23 weeks was 40.3% (n = 31). Responses (PR/uPR) by disease were as follows: melanoma, 5 of 29 patients (includes 1 patient with NRAS mutation); thyroid, 3 of 6 patients; pancreatic, 1 of 2 patients; lung, 1 of 1 patients; renal, 1 of 1 patients; endometrial, 1 of 4 patients; and ovarian, 1 of 5 patients. AUC(0-24) and C(max) increased dose proportionally. In multivariate Cox proportional hazard model analyses, increased baseline systolic blood pressure and decreased angiopoietin-1 ratio (2 hours:baseline) were associated with longer progression-free survival (PFS) in the expanded melanoma cohort (P = 0.041 and P = 0.03, respectively). CONCLUSIONS The toxicity profile, pharmacokinetics, and antitumor activity of lenvatinib are encouraging. Decreases in the angiopoietin-1 ratio correlated with longer PFS in melanoma patients.
Collapse
Affiliation(s)
- David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Razelle Kurzrock
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Aung Naing
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Siqing Fu
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kevin B Kim
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Davies
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ly M Nguyen
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Goldy C George
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lucy Xu
- Eisai Inc., Oncology, Woodcliff Lake, New Jersey
| | | | - Min Ren
- Eisai Inc., Oncology, Woodcliff Lake, New Jersey
| | - Jennifer Mink
- Former employees of Eisai Inc., Woodcliff Lake, New Jersey
| | | | | | | | | | | |
Collapse
|
48
|
Aung PP, Leone D, Feller JK, Yang S, Hernandez M, Yaar R, Singh R, Helm T, Mahalingam M. Microvessel density, lymphovascular density, and lymphovascular invasion in primary cutaneous melanoma-correlation with histopathologic prognosticators and BRAF status. Hum Pathol 2015; 46:304-12. [PMID: 25537974 DOI: 10.1016/j.humpath.2014.11.006] [Citation(s) in RCA: 18] [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: 09/11/2014] [Revised: 11/10/2014] [Accepted: 11/12/2014] [Indexed: 02/01/2023]
Abstract
The relationship between microvessel density (MVD), lymphovascular density (LVD), and lymphovascular invasion (LVI) in primary cutaneous melanoma (PCM) remains unclear. Given this, a total of 102 PCMs were assessed for MVD (vascular endothelial growth factor receptor 2 and Endocan), LVD (D2-40), and LVI (immunostaining with D2-40/S-100 and hematoxylin and eosin); tumoral S-100A13, vascular endothelial growth factor receptor 2, and Endocan; and BRAF status. LVD was associated with MVD (P = .01). MVD was higher in PCMs with depth greater than or equal to 2 mm and ulceration (P = .04, .05), whereas LVD was higher in PCMs with depth greater than or equal to 2 mm and mitoses (P = .03, .02). After adjusting for MVD and LVD, only ulceration was associated with LVI (P < .02). A BRAF mutation was seen in 30.4% cases, and when present, both LVD and host response (P = .0008 and .04, respectively) were significantly associated with MVD. Immunostaining with S-100A13 was noted in 99% of cases and a significant association noted only with ulceration (P = .05). Immunostaining increased LVI positivity (46.5% versus 4.9% by hematoxylin and eosin, P < .0001). MVD and LVD are not associated with LVI, appear to be closely related with each other, and are associated with select markers of poor prognosticative value. The association between a host response and LVD and MVD in PCMs with a BRAF mutation suggests that they exhibit potential for strategizing immunotherapies.
Collapse
Affiliation(s)
- Phyu Phyu Aung
- Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, MA 02118.
| | - Dominick Leone
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118.
| | - John Kyle Feller
- Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, MA 02118.
| | - Shi Yang
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118.
| | - Marier Hernandez
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX 75390.
| | - Ron Yaar
- Aurora Diagnostics, Greensboro, NC 27408.
| | - Rajendra Singh
- Departments of Dermatology and Pathology, Mt Sinai School of Medicine, New York, NY 10029.
| | - Thomas Helm
- Department of Dermatology, State University of New York at Buffalo, Buffalo, NY 14203.
| | - Meera Mahalingam
- Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, MA 02118.
| |
Collapse
|
49
|
Assessment of vascularity in common blue nevi, small/medium congenital nevocellular, common and dysplastic acquired melanocytic nevi and melanomas: a comparative study. Am J Dermatopathol 2014; 36:217-22. [PMID: 24067800 DOI: 10.1097/dad.0b013e3182946865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Angiogenesis and vascularity are researched in melanocytic tumors for their importance in carcinogenesis. For the first time, to the best of our knowledge, the authors compared the microvascular characteristics between small/medium congenital nevocellular nevi (CN), common blue nevi (BN), common and dysplastic acquired melanocytic nevi (AMN), and melanomas. The authors collected 31 BN, 48 CN (≤5 cm), 35 AMN (14 common, 21 dysplastic), and 26 melanomas. Vessels were stained with factor VIII. Microvascular density (MVD) and total vascular area (TVA), where evaluated in high capillary density areas. Student t and Mann-Whitney tests were used. MVD (mean ± SD) was low in BN (3.52 ± 1.21) and significantly higher in CN (7.56 ± 2.47) (P < 0.001). TVA was low in BN and significantly higher in CN (Mann-Whitney U = 141, n1 = 48, n2 = 31, P < 0.001, 2-tailed). MVD was not significantly different between common and dysplastic AMN (20.64 ± 7.87 and 20.38 ± 9.54, respectively) (P > 0.05). TVA was not significantly different between common and dysplastic AMN (Mann-Whitney U = 164, n1 = 14, n2 = 21, P > 0.05, 2-tailed). MVD was significantly lower in CN (7.56 ± 2.47) compared with AMN (20.49 ± 8.79) (P < 0.001). TVA was significantly lower in CN compared with AMN (Mann-Whitney U = 1486, n1 = 48, n2 = 35, P < 0.001, 2-tailed). MVD was significantly lower in AMN (20.49 ± 8.79) compared with melanomas (33.77 ± 14.32) (P < 0.001). TVA (mean ± SD) was significantly smaller in AMN (18473.94 ± 7050.61) compared with melanomas (29308.50 ± 11307.22) (P < 0.001). Vascularity increased from BN to CN to AMN with melanomas being the most vascular. Common and dysplastic AMN had comparable vascularity. The implications of our results regarding melanoma transformation risk are considered.
Collapse
|
50
|
Pastushenko I, Vermeulen PB, Van den Eynden GG, Rutten A, Carapeto FJ, Dirix LY, Van Laere S. Mechanisms of tumour vascularization in cutaneous malignant melanoma: clinical implications. Br J Dermatol 2014; 171:220-33. [PMID: 24641095 DOI: 10.1111/bjd.12973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/02/2023]
Abstract
Malignant melanoma represents < 10% of all skin cancers but is responsible for the majority of skin-cancer-related deaths. Metastatic melanoma has historically been considered as one of the most therapeutically challenging malignancies. Fortunately, for the first time after decades of basic research and clinical investigation, new drugs have produced major clinical responses. Angiogenesis has been considered an important target for cancer treatment. Initial efforts have focused primarily on targeting endothelial and tumour-related vascular endothelial growth factor signalling. Here, we review different mechanisms of tumour vascularization described in melanoma and discuss the potential clinical implications.
Collapse
Affiliation(s)
- I Pastushenko
- Department of Dermatology, Hospital Clínico Universitario 'Lozano Blesa', Zaragoza, 50009, Spain
| | | | | | | | | | | | | |
Collapse
|