1
|
Wang L, Ge J, Han H, Jia Y, Qin Y. Crosstalk between the nervous system and tumor microenvironment: Functional aspects and potential therapeutic strategies. Cancer Lett 2024; 594:216986. [PMID: 38797233 DOI: 10.1016/j.canlet.2024.216986] [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: 03/15/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Recent advancements in understanding the tumor microenvironment (TME) have highlighted the critical role of the nervous system in cancer progression. This review comprehensively examines how the nervous system influences various aspects of tumorigenesis, including growth, motility, immune response, angiogenesis, and the behavior of cancer-associated fibroblasts (CAFs). We delineate the neurodevelopmental mechanisms associated with cancer, such as the secretion of neurotrophins and exosomes by cancer cells. Furthermore, we explore the emerging therapeutic strategy of targeting nerves associated with tumors. Evidence supporting this approach includes studies demonstrating direct tumor growth inhibition, enhanced efficacy of immunotherapy when combined with nervous system-modulating drugs, and the suppression of tumor blood vessel formation through nerve targeting. Finally, we discuss the current challenges in this field and emphasize the need for further exploration within cancer neuroscience.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China
| | - Jingjing Ge
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, PR China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China
| | - Yongxu Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China.
| |
Collapse
|
2
|
Serioli S, Agostini L, Pietrantoni A, Valeri F, Costanza F, Chiloiro S, Buffoli B, Piazza A, Poliani PL, Peris-Celda M, Iavarone F, Gaudino S, Gessi M, Schinzari G, Mattogno PP, Giampietro A, De Marinis L, Pontecorvi A, Fontanella MM, Lauretti L, Rindi G, Olivi A, Bianchi A, Doglietto F. Aggressive PitNETs and Potential Target Therapies: A Systematic Review of Molecular and Genetic Pathways. Int J Mol Sci 2023; 24:15719. [PMID: 37958702 PMCID: PMC10650665 DOI: 10.3390/ijms242115719] [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: 08/25/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Recently, advances in molecular biology and bioinformatics have allowed a more thorough understanding of tumorigenesis in aggressive PitNETs (pituitary neuroendocrine tumors) through the identification of specific essential genes, crucial molecular pathways, regulators, and effects of the tumoral microenvironment. Target therapies have been developed to cure oncology patients refractory to traditional treatments, introducing the concept of precision medicine. Preliminary data on PitNETs are derived from preclinical studies conducted on cell cultures, animal models, and a few case reports or small case series. This study comprehensively reviews the principal pathways involved in aggressive PitNETs, describing the potential target therapies. A search was conducted on Pubmed, Scopus, and Web of Science for English papers published between 1 January 2004, and 15 June 2023. 254 were selected, and the topics related to aggressive PitNETs were recorded and discussed in detail: epigenetic aspects, membrane proteins and receptors, metalloprotease, molecular pathways, PPRK, and the immune microenvironment. A comprehensive comprehension of the molecular mechanisms linked to PitNETs' aggressiveness and invasiveness is crucial. Despite promising preliminary findings, additional research and clinical trials are necessary to confirm the indications and effectiveness of target therapies for PitNETs.
Collapse
Affiliation(s)
- Simona Serioli
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Ludovico Agostini
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | | | - Federico Valeri
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Flavia Costanza
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Sabrina Chiloiro
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Barbara Buffoli
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy;
| | - Amedeo Piazza
- Department of Neuroscience, Neurosurgery Division, “Sapienza” University of Rome, 00185 Rome, Italy;
| | - Pietro Luigi Poliani
- Pathology Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele, 20132 Milan, Italy;
| | - Maria Peris-Celda
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Otolaryngology/Head and Neck Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 20123 Rome, Italy;
- Fondazione Policlinico Universitario IRCCS “A. Gemelli”, 00168 Rome, Italy
| | - Simona Gaudino
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Radiological Sciences, Institute of Radiology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marco Gessi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Neuropathology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giovanni Schinzari
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Oncology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Pier Paolo Mattogno
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonella Giampietro
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Laura De Marinis
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Alfredo Pontecorvi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Marco Maria Fontanella
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Liverana Lauretti
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Guido Rindi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Neuropathology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Alessandro Olivi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Bianchi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Francesco Doglietto
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| |
Collapse
|
3
|
Ilie MD, De Alcubierre D, Carretti AL, Jouanneau E, Raverot G. Therapeutic targeting of the pituitary tumor microenvironment. Pharmacol Ther 2023; 250:108506. [PMID: 37562699 DOI: 10.1016/j.pharmthera.2023.108506] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
The tumor microenvironment (TME), the complex environment in which tumors develop, has been increasingly targeted for cancer treatment in recent years. Aggressive pituitary tumors and pituitary carcinomas have been so far targeted with immune-checkpoint inhibitors (28 cases, including a large cohort), and anti-angiogenic drugs (34 cases), specifically bevacizumab (30 cases), sunitinib (three cases), and apatinib (one case). Here, we reviewed all these cases, reporting tumor response, potential predictors of response, as well as adverse events. Given that the histological type could potentially influence treatment response, we present the existing data separately for each type. Briefly, under ICIs, complete response was noted in one case, partial response in a third of cases, stable disease in 10% of cases, while 54% of tumors progressed. Under BVZ monotherapy, most cases (57%) showed stable disease, while 36% of tumors progressed; partial response was reported in only one case. The three cases treated with sunitinib monotherapy progressed. Regarding predictive factors of response, the tumor type (aggressive pituitary tumor versus pituitary carcinoma) appears as the strongest predictor of response to ICIs. To date, no predictor of response to anti-angiogenic drugs in the treatment of pituitary carcinomas and aggressive pituitary tumors has been identified. The interest of BZV add-on to first- or second-line chemotherapy warrants further investigation. In addition, we discuss perspectives regarding the TME-targeting in aggressive pituitary tumors and pituitary carcinomas, including perspectives on immunotherapy, anti-angiogenic drugs, as well as on other TME components, namely stromal cells, extracellular matrix, and secreted molecules.
Collapse
Affiliation(s)
- Mirela-Diana Ilie
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Lyon 1 University, Villeurbanne, France; Endocrinology Department, "C.I. Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Dario De Alcubierre
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Lucia Carretti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy; Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France
| | - Emmanuel Jouanneau
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Lyon 1 University, Villeurbanne, France; Neurosurgery Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France
| | - Gérald Raverot
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Lyon 1 University, Villeurbanne, France; Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France.
| |
Collapse
|
4
|
Mendoza-Torreblanca JG, Cárdenas-Rodríguez N, Carro-Rodríguez J, Contreras-García IJ, Garciadiego-Cázares D, Ortega-Cuellar D, Martínez-López V, Alfaro-Rodríguez A, Evia-Ramírez AN, Ignacio-Mejía I, Vargas-Hernández MA, Bandala C. Antiangiogenic Effect of Dopamine and Dopaminergic Agonists as an Adjuvant Therapeutic Option in the Treatment of Cancer, Endometriosis, and Osteoarthritis. Int J Mol Sci 2023; 24:10199. [PMID: 37373348 DOI: 10.3390/ijms241210199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Dopamine (DA) and dopamine agonists (DA-Ag) have shown antiangiogenic potential through the vascular endothelial growth factor (VEGF) pathway. They inhibit VEGF and VEGF receptor 2 (VEGFR 2) functions through the dopamine receptor D2 (D2R), preventing important angiogenesis-related processes such as proliferation, migration, and vascular permeability. However, few studies have demonstrated the antiangiogenic mechanism and efficacy of DA and DA-Ag in diseases such as cancer, endometriosis, and osteoarthritis (OA). Therefore, the objective of this review was to describe the mechanisms of the antiangiogenic action of the DA-D2R/VEGF-VEGFR 2 system and to compile related findings from experimental studies and clinical trials on cancer, endometriosis, and OA. Advanced searches were performed in PubMed, Web of Science, SciFinder, ProQuest, EBSCO, Scopus, Science Direct, Google Scholar, PubChem, NCBI Bookshelf, DrugBank, livertox, and Clinical Trials. Articles explaining the antiangiogenic effect of DA and DA-Ag in research articles, meta-analyses, books, reviews, databases, and clinical trials were considered. DA and DA-Ag have an antiangiogenic effect that could reinforce the treatment of diseases that do not yet have a fully curative treatment, such as cancer, endometriosis, and OA. In addition, DA and DA-Ag could present advantages over other angiogenic inhibitors, such as monoclonal antibodies.
Collapse
Affiliation(s)
| | - Noemi Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Jazmín Carro-Rodríguez
- Laboratorio de Medicina Traslacional Aplicada a Neurociencias, Enfermedades Crónicas y Emergentes, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Itzel Jatziri Contreras-García
- Laboratorio de Biología de la Reproducción, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - David Garciadiego-Cázares
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | - Daniel Ortega-Cuellar
- Laboratorio Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Valentín Martínez-López
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | - Alfonso Alfaro-Rodríguez
- Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Mexico City 14389, Mexico
| | - Alberto Nayib Evia-Ramírez
- Servicio de Reconstrucción Articular, Cadera y Rodilla, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | - Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Mexico City 11200, Mexico
| | | | - Cindy Bandala
- Laboratorio de Medicina Traslacional Aplicada a Neurociencias, Enfermedades Crónicas y Emergentes, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| |
Collapse
|
5
|
Marques P, Korbonits M. Tumour microenvironment and pituitary tumour behaviour. J Endocrinol Invest 2023; 46:1047-1063. [PMID: 37060402 DOI: 10.1007/s40618-023-02089-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/04/2023] [Indexed: 04/16/2023]
Abstract
The pituitary tumour microenvironment encompasses a spectrum of non-tumoural cells, such as immune, stromal or endothelial cells, as well as enzymes and signalling peptides like cytokines, chemokines and growth factors, which surround the tumour cells and may influence pituitary tumour behaviour and tumourigenic mechanisms. Recently, there has been intensive research activity in this field describing various pituitary tumour-infiltrating immune and stromal cell subpopulations, and immune- and microenvironment-related pathways. Key changes in oncological therapeutic avenues resulted in the recognition of pituitary as a target of adverse events for patients treated with immune checkpoint regulators. However, these phenomena can be turned into therapeutic advantage in severe cases of pituitary tumours. Therefore, unravelling the pituitary tumour microenvironment will allow a better understanding of the biology and behaviour of pituitary tumours and may provide further developments in terms of diagnosis and management of patients with aggressively growing or recurrent pituitary tumours.
Collapse
Affiliation(s)
- P Marques
- Pituitary Tumor Unit, Endocrinology Department, Hospital CUF Descobertas, Lisbon, Portugal.
- Faculdade de Medicina, Universidade Católica Portuguesa, Lisbon, Portugal.
| | - M Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
6
|
Sarkar C, Chakroborty D, Goswami S, Fan H, Mo X, Basu S. VEGF-A controls the expression of its regulator of angiogenic functions, dopamine D2 receptor, on endothelial cells. J Cell Sci 2022; 135:jcs259617. [PMID: 35593650 PMCID: PMC9234670 DOI: 10.1242/jcs.259617] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/05/2022] [Indexed: 01/08/2023] Open
Abstract
We have previously demonstrated significant upregulation of dopamine D2 (DAD2) receptor (DRD2) expression on tumor endothelial cells. The dopamine D2 receptors, upon activation, inhibit the proangiogenic actions of vascular endothelial growth factor-A (VEGF-A, also known as vascular permeability factor). Interestingly, unlike tumor endothelial cells, normal endothelial cells exhibit very low to no expression of dopamine D2 receptors. Here, for the first time, we demonstrate that through paracrine signaling, VEGF-A can control the expression of dopamine D2 receptors on endothelial cells via Krüppel-like factor 11 (KLF11)-extracellular signal-regulated kinase (ERK) 1/2 pathway. These results thus reveal a novel bidirectional communication between VEGF-A and DAD2 receptors.
Collapse
Affiliation(s)
- Chandrani Sarkar
- Department of Pathology, Ohio State University, Columbus, Ohio 43201, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA
- Department of Pathology, University of South Alabama, Mobile, Alabama 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36688, USA
- Department of Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama 36688, USA
| | - Debanjan Chakroborty
- Department of Pathology, Ohio State University, Columbus, Ohio 43201, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA
- Department of Pathology, University of South Alabama, Mobile, Alabama 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36688, USA
- Department of Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama 36688, USA
| | - Sandeep Goswami
- Department of Pathology, Ohio State University, Columbus, Ohio 43201, USA
- Department of Pathology, University of South Alabama, Mobile, Alabama 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36688, USA
| | - Hao Fan
- Department of Pathology, Ohio State University, Columbus, Ohio 43201, USA
| | - Xiaokui Mo
- Department of Biomedical Informatics, Ohio State University, Columbus, Ohio 43210, USA
| | - Sujit Basu
- Department of Pathology, Ohio State University, Columbus, Ohio 43201, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
7
|
Zhou J, Hu Y, Zhu W, Nie C, Zhao W, Faje AT, Labelle KE, Swearingen B, Lee H, Hedley-Whyte ET, Zhang X, Jones PS, Miller KK, Klibanski A, Zhou Y, Soberman RJ. Sprouting Angiogenesis in Human Pituitary Adenomas. Front Oncol 2022; 12:875219. [PMID: 35600354 PMCID: PMC9117625 DOI: 10.3389/fonc.2022.875219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/05/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Angiogenesis in pituitary tumors is not fully understood, and a better understanding could help inform new pharmacologic therapies, particularly for aggressive pituitary tumors. Materials and Methods 219 human pituitary tumors and 12 normal pituitary glands were studied. Angiogenic genes were quantified by an angiogenesis qPCR array and a TaqMan probe-based absolute qPCR. Angiogenesis inhibition in pituitary tumors was evaluated in vitro with the endothelial tube formation assay and in vivo in RbΔ19 mice. Results 71 angiogenic genes, 40 of which are known to be involved in sprouting angiogenesis, were differentially expressed in pituitary tumors. Expression of endothelial markers CD31, CD34, and ENG was significantly higher in pituitary tumors, by 5.6, 22.3, and 8.2-fold, respectively, compared to in normal pituitary tissue. There was no significant difference in levels of the lymphatic endothelial marker LYVE1 in pituitary tumors compared with normal pituitary gland tissue. Pituitary tumors also expressed significantly higher levels of angiogenesis growth factors, including VEGFA (4.2-fold), VEGFB (2.2), VEGFC (19.3), PGF (13.4), ANGPT2 (9.2), PDGFA (2.7), PDGFB (10.5) and TGFB1 (3.8) compared to normal pituitary tissue. Expression of VEGFC and PGF was highly correlated with the expression of endothelial markers in tumor samples, including CD31, CD34, and ENG (endoglin, a co-receptor for TGFβ). Furthermore, VEGFR inhibitors inhibited angiogenesis induced by human pituitary tumors and prolonged survival of RbΔ19 mice. Conclusion Human pituitary tumors are characterized by more active angiogenesis than normal pituitary gland tissue in a manner consistent with sprouting angiogenesis. Angiogenesis in pituitary tumors is regulated mainly by PGF and VEGFC, not VEGFA and VEGFB. Angiogenesis inhibitors, such as the VEGFR2 inhibitor cabozantinib, may merit further investigation as therapies for aggressive human pituitary tumors.
Collapse
Affiliation(s)
- Jie Zhou
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yaomin Hu
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Wende Zhu
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Chuansheng Nie
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Wenxiu Zhao
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Alexander T. Faje
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Kay E. Labelle
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Brooke Swearingen
- Neurosurgery Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - E. Tessa Hedley-Whyte
- Department of Pathology (Neuropathology), Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Xun Zhang
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Pamela S. Jones
- Neurosurgery Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Karen K. Miller
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anne Klibanski
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yunli Zhou
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Yunli Zhou,
| | - Roy J. Soberman
- Nephrology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Kasica N, Święch A, Saładziak K, Mackiewicz J, Osęka M. The Inhibitory Effect of Selected D2 Dopaminergic Receptor Agonists on VEGF-Dependent Neovascularization in Zebrafish Larvae: Potential New Therapy in Ophthalmic Diseases. Cells 2022; 11:cells11071202. [PMID: 35406766 PMCID: PMC8997652 DOI: 10.3390/cells11071202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 01/01/2023] Open
Abstract
Pathological angiogenesis is correlated with many ophthalmic diseases. The most common are exudative age-related macular degeneration and proliferative diabetic retinopathy. The current treatment for these diseases is based on regularly administered anti-VEGF antibodies injections. In the study, we investigated selected D2 dopaminergic receptor agonists, namely bromocriptine, cabergoline and pergolide, on hypoxia-induced neovascularization. We used the zebrafish laboratory model, specifically three-day post fertilization (dpf) Tg(fli-1: EGFP) zebrafish larvae. To induce abnormal angiogenesis of hyaloid-retinal vessels (HRVs) and intersegmental vessels (ISVs), the larvae were treated with cobalt chloride (II) (CoCl2) (a hypoxia-inducing agent) from 24 h post fertilization. The inhibitory role of D2 dopaminergic receptor agonists was investigated using confocal microscopy and qPCR. Additionally, the results were compared to those obtained in the group treated with CoCl2 followed by bevacizumab, the well-known antiangiogenic agent. Confocal microscopy analyses revealed severe deformation of vessels in the CoCl2 treated group, while co-incubation with bromocriptine, cabergoline, pergolide and bevacizumab, respectively, significantly inhibited abnormalities of angiogenesis. The qPCR analyses supported the protective role of the chosen dopaminergic agonists by demonstrating their influence on CoCl2-derived upregulation of vegfaa expression. The present results suggest that the D2 receptor agonists can be considered as a new direction in research for antiangiogenic therapy.
Collapse
Affiliation(s)
- Natalia Kasica
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13 Street, Box 105J, 10-719 Olsztyn, Poland
- Correspondence:
| | - Anna Święch
- Department of Retina and Vitreus Surgery, Medical University in Lublin, Chmielna 1 Street, 20-079 Lublin, Poland; (A.Ś.); (K.S.); (J.M.)
| | - Katarzyna Saładziak
- Department of Retina and Vitreus Surgery, Medical University in Lublin, Chmielna 1 Street, 20-079 Lublin, Poland; (A.Ś.); (K.S.); (J.M.)
| | - Jerzy Mackiewicz
- Department of Retina and Vitreus Surgery, Medical University in Lublin, Chmielna 1 Street, 20-079 Lublin, Poland; (A.Ś.); (K.S.); (J.M.)
| | - Maciej Osęka
- Oftalabs Sp. z o.o., Wrocławska 130, 58-306 Wałbrzych, Poland;
| |
Collapse
|
9
|
Dai C, Liang S, Sun B, Li Y, Kang J. Anti-VEGF Therapy in Refractory Pituitary Adenomas and Pituitary Carcinomas: A Review. Front Oncol 2021; 11:773905. [PMID: 34869016 PMCID: PMC8635636 DOI: 10.3389/fonc.2021.773905] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Most pituitary tumors are considered benign adenomas, and only 0.1%–0.2% of them present metastasis and are defined as pituitary carcinomas (PCs). Refractory pituitary adenomas (PAs) lie between benign adenomas and true malignant PCs and are defined as aggressive-invasive PAs, characterized by a high Ki-67 index, rapid growth, frequent recurrence, and resistance to conventional treatments. Refractory PAs and PCs are notoriously difficult to manage because of limited therapeutic options. Vascular endothelial growth factor (VEGF) plays a crucial role in angiogenesis not only during development but also during pathological processes in pituitary tumors. Recently, increasing numbers of preclinical studies and clinical research have demonstrated that anti-VEGF therapy plays an important role in pituitary tumors. The purpose of this review is to report the role of VEGF in the development and pathology of pituitary tumors and the progress of anti-VEGF therapy in pituitary tumors, including refractory PAs and PCs. Previous preclinical studies indicated that cyclin-dependent kinase 5 (CDK5)-mediated VEGF expression might play a crucial role in the development of PAs. Vascular endothelial growth inhibitors have been reported as independent predictors of invasion in human PAs and have been indicated as markers for poor outcome. Furthermore, several studies have reported that angiogenesis decreases tumor sizes in experimental animal models of pituitary tumors. The expression of VEGF is relatively high in PAs; therefore, anti-VEGF therapy has been used in some refractory PAs and PCs. To date, anti-VEGF has been reported as monotherapy, in combination with temozolomide (TMZ), TMZ and radiotherapy, and with pasireotide, which might be a promising alternative therapy for refractory PAs and PCs resistant to conventional treatments. However, the role of anti-VEGF therapy in pituitary tumors is still controversial due to a lack of large-scale clinical trials. In summary, the results from preclinical studies and clinical trials indicated that anti-VEGF therapy monotherapy or in combination with other treatments may be a promising alternative therapy for refractory PAs and PCs resistant to conventional treatments. More preclinical studies and clinical trials are needed to further evaluate the exact efficacy of anti-VEGF in refractory PAs and PCs.
Collapse
Affiliation(s)
- Congxin Dai
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Siyu Liang
- Eight-Year Program of Clinical Medicine, Peking Union Medical College Hospital (PUMCH), Chinese Academe of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Bowen Sun
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yong Li
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jun Kang
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
DLL1 orchestrates CD8 + T cells to induce long-term vascular normalization and tumor regression. Proc Natl Acad Sci U S A 2021; 118:2020057118. [PMID: 34035167 DOI: 10.1073/pnas.2020057118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The immunosuppressive and hypoxic tumor microenvironment (TME) remains a major obstacle to impede cancer immunotherapy. Here, we showed that elevated levels of Delta-like 1 (DLL1) in the breast and lung TME induced long-term tumor vascular normalization to alleviate tumor hypoxia and promoted the accumulation of interferon γ (IFN-γ)-expressing CD8+ T cells and the polarization of M1-like macrophages. Moreover, increased DLL1 levels in the TME sensitized anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA4) treatment in its resistant tumors, resulting in tumor regression and prolonged survival. Mechanically, in vivo depletion of CD8+ T cells or host IFN-γ deficiency reversed tumor growth inhibition and abrogated DLL1-induced tumor vascular normalization without affecting DLL1-mediated macrophage polarization. Together, these results demonstrate that elevated DLL1 levels in the TME promote durable tumor vascular normalization in a CD8+ T cell- and IFN-γ-dependent manner and potentiate anti-CTLA4 therapy. Our findings unveil DLL1 as a potential target to persistently normalize the TME to facilitate cancer immunotherapy.
Collapse
|
11
|
Chesler K, Motz C, Vo H, Douglass A, Allen RS, Feola AJ, Pardue MT. Initiation of L-DOPA Treatment After Detection of Diabetes-Induced Retinal Dysfunction Reverses Retinopathy and Provides Neuroprotection in Rats. Transl Vis Sci Technol 2021; 10:8. [PMID: 34003986 PMCID: PMC8054623 DOI: 10.1167/tvst.10.4.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose L-DOPA treatment initiated at the start of hyperglycemia preserves retinal and visual function in diabetic rats. Here, we investigated a more clinically relevant treatment strategy in which retinal and visual dysfunction designated the beginning of the therapeutic window for L-DOPA treatment. Methods Spatial frequency thresholds using optomotor response and oscillatory potential (OP) delays using electroretinograms were compared at baseline, 3, 6, and 10 weeks after streptozotocin (STZ) between diabetic and control rats. L-DOPA/carbidopa treatment (DOPA) or vehicle was delivered orally 5 days per week beginning at 3 weeks after STZ, when significant retinal and visual deficits were measured. At 10 weeks after STZ, retinas were collected to measure L-DOPA, dopamine, and 3,4-dihydroxyphenylacetic acid (DOPAC) levels using high-performance liquid chromatography. Results Spatial frequency thresholds decreased at 6 weeks in diabetic vehicle rats (28%), whereas diabetic DOPA rats had stable thresholds (<1%) that maintained to 10 weeks, creating significantly higher thresholds compared with diabetic vehicle rats (P < 0.0001). OP2 implicit times in response to dim, rod-driven stimuli were decreased in diabetic compared with control rats (3 weeks, P < 0.0001; 10 weeks, P < 0.01). With L-DOPA treatment, OP2 implicit times recovered in diabetic rats to be indistinguishable from control rats by 10 weeks after STZ. Rats treated with L-DOPA showed significantly increased retinal L-DOPA (P < 0.001) and dopamine levels (P < 0.05). Conclusions L-DOPA treatment started after the detection of retinal and visual dysfunction showed protective effects in diabetic rats. Translational Relevance Early retinal functional deficits induced by diabetes can be used to identify an earlier therapeutic window for L-DOPA treatment which protects from further vision loss and restores retinal function.
Collapse
Affiliation(s)
- Kyle Chesler
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Cara Motz
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Harrison Vo
- Atlanta VA Healthcare System, Atlanta, GA, USA
| | | | - Rachael S Allen
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrew J Feola
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
12
|
Cooper O, Bonert V, Liu NA, Mamelak AN. Treatment of Aggressive Pituitary Adenomas: A Case-Based Narrative Review. Front Endocrinol (Lausanne) 2021; 12:725014. [PMID: 34867776 PMCID: PMC8634600 DOI: 10.3389/fendo.2021.725014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/28/2021] [Indexed: 12/29/2022] Open
Abstract
Management of aggressive pituitary adenomas is challenging due to a paucity of rigorous evidence supporting available treatment approaches. Recent guidelines emphasize the need to maximize standard therapies as well as the use of temozolomide and radiation therapy to treat disease recurrence. However, often these adenomas continue to progress over time, necessitating the use of additional targeted therapies which also impact quality of life and long-term outcomes. In this review, we present 9 cases of aggressive pituitary adenomas to illustrate the importance of a multidisciplinary, individualized approach. The timing and rationale for surgery, radiation therapy, temozolomide, somatostatin receptor ligands, and EGFR, VEGF, and mTOR inhibitors in each case are discussed within the context of evidence-based guidelines and clarify strategies for implementing an individualized approach in the management of these difficult-to-treat-adenomas.
Collapse
Affiliation(s)
- Odelia Cooper
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- *Correspondence: Odelia Cooper,
| | - Vivien Bonert
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ning-Ai Liu
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Adam N. Mamelak
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
13
|
Lin AL, Donoghue MTA, Wardlaw SL, Yang TJ, Bodei L, Tabar V, Geer EB. Approach to the Treatment of a Patient with an Aggressive Pituitary Tumor. J Clin Endocrinol Metab 2020; 105:5905925. [PMID: 32930787 PMCID: PMC7566322 DOI: 10.1210/clinem/dgaa649] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
A small subset of pituitary adenomas grows despite maximal treatment with standard therapies; namely, surgery and radiotherapy. These aggressive tumors demonstrate 2 patterns of growth: they may be locally aggressive or metastasize distantly, either hematogenously or through the spinal fluid. Further surgery and radiotherapy may be helpful for palliation of symptoms, but they are rarely definitive in the management of these malignant tumors. The only chemotherapy with established activity in the treatment of pituitary tumors is the alkylating agent temozolomide. At most, 50% of patients exhibit an objective response to temozolomide and the median time to progression is short; thus, there remains a significant unmet need for effective treatments within this patient population. Several targeted agents have reported activity in this tumor type-including small molecule inhibitors, checkpoint inhibitors, and other biologics-but remain investigational at this time.
Collapse
Affiliation(s)
- Andrew L Lin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Multidisciplinary Pituitary and Skull Base Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Correspondence and Reprint Requests: Andrew Lin, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA. E-mail:
| | - Mark T A Donoghue
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - T Jonathan Yang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Viviane Tabar
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Multidisciplinary Pituitary and Skull Base Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Program in Cell Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eliza B Geer
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Multidisciplinary Pituitary and Skull Base Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
14
|
Lan H, Zhang W, Jin K, Liu Y, Wang Z. Modulating barriers of tumor microenvironment through nanocarrier systems for improved cancer immunotherapy: a review of current status and future perspective. Drug Deliv 2020; 27:1248-1262. [PMID: 32865029 PMCID: PMC7470050 DOI: 10.1080/10717544.2020.1809559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy suppresses and destroys tumors by re-activating and sustaining the tumor-immune process, and thus improving the immune response of the body to the tumor. Immunotherapeutic strategies are showing promising results in pre-clinical and clinical trials, however, tumor microenvironment (TME) is extremely immunosuppressive. Thus, their translation from labs to clinics still faces issues. Recently, nanomaterial-based strategies have been developed to modulate the TME for robust immunotherapeutic responses. The combination of nanotechnology with immunotherapy potentiates the effectiveness of immunotherapy by increasing delivery and retention, and by reducing immunomodulation toxicity. This review aims to highlight the barriers offered by TME for hindering the efficiency of immunotherapy for cancer treatment. Next, we highlight various nano-carriers based strategies for modulating those barriers for achieving better therapeutic efficacy of cancer immunotherapy with higher safety. This review will add to the body of scientific knowledge and will be a good reference material for academia and industries.
Collapse
Affiliation(s)
- Huanrong Lan
- Department of Breast and Thyroid Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Wei Zhang
- Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ketao Jin
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Yuyao Liu
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Zhen Wang
- Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
15
|
Marques P, Grossman AB, Korbonits M. The tumour microenvironment of pituitary neuroendocrine tumours. Front Neuroendocrinol 2020; 58:100852. [PMID: 32553750 DOI: 10.1016/j.yfrne.2020.100852] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/26/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
The tumour microenvironment (TME) includes a variety of non-neoplastic cells and non-cellular elements such as cytokines, growth factors and enzymes surrounding tumour cells. The TME emerged as a key modulator of tumour initiation, progression and invasion, with extensive data available in many cancers, but little is known in pituitary tumours. However, the understanding of the TME of pituitary tumours has advanced thanks to active research in this field over the last decade. Different immune and stromal cell subpopulations, and several cytokines, growth factors and matrix remodelling enzymes, have been characterised in pituitary tumours. Studying the TME in pituitary tumours may lead to a better understanding of tumourigenic mechanisms, identification of biomarkers useful to predict aggressive disease, and development of novel therapies. This review summarises the current knowledge on the different TME cellular/non-cellular elements in pituitary tumours and provides an overview of their role in tumourigenesis, biological behaviour and clinical outcomes.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Ashley B Grossman
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
16
|
Abstract
PURPOSE Aggressive prolactinomas are defined as radiologically invasive tumors which cannot be cured by surgery, and that have an unusually rapid rate of tumor growth despite dopamine agonist treatment and surgery. In some cases, metastasis occurs, defining prolactin carcinoma which is the second most frequent pituitary carcinoma. METHODS A literature search was performed to review the available data on the treatment of aggressive pituitary prolactinomas or carcinomas. RESULTS When optimal standard therapies (high dose cabergoline, surgery and radiotherapy) failed, temozolomide, an alkylating drug, is currently the best option, allowing to control tumor growth in about 50% of treated prolactinomas and improving overall survival of these patients. However, long-term complete response occurs in a limited subgroup of tumors. Alternative drugs could be discussed in a subset of aggressive prolactinomas either before temozolomide (pasireotide, peptide receptor radionuclide therapy…) or after temozolomide failure. CONCLUSION Despite the significant improvement obtained with the use of temozolomide, a need for alternative drugs persists since a majority of these tumors are resistant or will recur during the follow-up. Patients suffering from such a rare condition should have access to clinical trials available for other types of rare cancers, such as tyrosine kinase inhibitors or immunotherapy.
Collapse
Affiliation(s)
- Hélène Lasolle
- INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon, 69008, Lyon, Auvergne-Rhône-Alpes, France
- Lyon 1 University, 69100, Villeurbanne, Auvergne-Rhône-Alpes, France
- Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Auvergne-Rhône-Alpes, 69677, Bron, France
| | - Mirela Diana Ilie
- Endocrinology Department, "C.I.Parhon" National Institute of Endocrinology, 011863, Bucharest, Bucharest-Ilfov, Romania
| | - Gérald Raverot
- INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon, 69008, Lyon, Auvergne-Rhône-Alpes, France.
- Lyon 1 University, 69100, Villeurbanne, Auvergne-Rhône-Alpes, France.
- Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Auvergne-Rhône-Alpes, 69677, Bron, France.
| |
Collapse
|
17
|
Lamb LS, Sim HW, McCormack AI. Exploring the Role of Novel Medical Therapies for Aggressive Pituitary Tumors: A Review of the Literature-"Are We There Yet?". Cancers (Basel) 2020; 12:cancers12020308. [PMID: 32012988 PMCID: PMC7072681 DOI: 10.3390/cancers12020308] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
Aggressive pituitary tumors account for up to 10% of pituitary tumors and are characterized by resistance to medical treatment and multiple recurrences despite standard therapies, including surgery, radiotherapy, and chemotherapy. They are associated with increased morbidity and mortality, particularly pituitary carcinomas, which have mortality rates of up to 66% at 1 year after diagnosis. Novel targeted therapies under investigation include mammalian target of rapamycin (mTOR), tyrosine kinase, and vascular endothelial growth factor (VEGF) inhibitors. More recently, immune checkpoint inhibitors have been proposed as a potential treatment option for pituitary tumors. An increased understanding of the molecular pathogenesis of aggressive pituitary tumors is required to identify potential biomarkers and therapeutic targets. This review discusses novel approaches to the management of aggressive pituitary tumors and the role of molecular profiling.
Collapse
Affiliation(s)
- Lydia S. Lamb
- Department of Endocrinology, St Vincent’s Hospital, Sydney, NSW 2010, Australia;
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia;
| | - Hao-Wen Sim
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia;
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
- Kinghorn Cancer Centre, Sydney, NSW 2010, Australia
| | - Ann I. McCormack
- Department of Endocrinology, St Vincent’s Hospital, Sydney, NSW 2010, Australia;
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia;
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
- Correspondence: ; Tel.: +61-2-9295-8489
| |
Collapse
|
18
|
Ilie MD, Vasiljevic A, Raverot G, Bertolino P. The Microenvironment of Pituitary Tumors-Biological and Therapeutic Implications. Cancers (Basel) 2019; 11:cancers11101605. [PMID: 31640258 PMCID: PMC6826349 DOI: 10.3390/cancers11101605] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) includes resident and infiltrative non-tumor cells, as well as blood and lymph vessels, extracellular matrix molecules, and numerous soluble factors, such as cytokines and chemokines. While the TME is now considered to be a prognostic tool and a therapeutic target for many cancers, little is known about its composition in pituitary tumors. This review summarizes our current knowledge of the TME within pituitary tumors and the strong interest in TME as a therapeutic target. While we cover the importance of angiogenesis and immune infiltrating cells, we also address the role of the elusive folliculostellate cells, the emerging literature on pituitary tumor-associated fibroblasts, and the contribution of extracellular matrix components in these tumors. The cases of human pituitary tumors treated with TME-targeting therapies are reviewed and emerging concepts of vascular normalization and combined therapies are presented. Together, this snapshot overview of the current literature pinpoints not only the underestimated role of TME components in pituitary tumor biology, but also the major promise it may offer for both prognosis and targeted therapeutics.
Collapse
Affiliation(s)
- Mirela Diana Ilie
- Cancer Research Centre of Lyon (CRCL), INSERM U1052, CNRS UMR5286, Claude Bernard University, 69008 Lyon, France, (M.D.I.).
- "Claude Bernard" Lyon 1 University, University of Lyon, 69100 Villeurbanne, France.
- Endocrinology Department, "C.I.Parhon" National Institute of Endocrinology, 011863 Bucharest, Romania.
| | - Alexandre Vasiljevic
- Cancer Research Centre of Lyon (CRCL), INSERM U1052, CNRS UMR5286, Claude Bernard University, 69008 Lyon, France, (M.D.I.).
- "Claude Bernard" Lyon 1 University, University of Lyon, 69100 Villeurbanne, France.
- Pathology Department, "Groupement Hospitalier Est" Hospices Civils de Lyon, 69677 Bron, France.
| | - Gérald Raverot
- Cancer Research Centre of Lyon (CRCL), INSERM U1052, CNRS UMR5286, Claude Bernard University, 69008 Lyon, France, (M.D.I.).
- "Claude Bernard" Lyon 1 University, University of Lyon, 69100 Villeurbanne, France.
- Endocrinology Department, "Groupement Hospitalier Est" Hospices Civils de Lyon, 69677 Bron, France.
| | - Philippe Bertolino
- Cancer Research Centre of Lyon (CRCL), INSERM U1052, CNRS UMR5286, Claude Bernard University, 69008 Lyon, France, (M.D.I.).
| |
Collapse
|
19
|
Ilie MD, Lasolle H, Raverot G. Emerging and Novel Treatments for Pituitary Tumors. J Clin Med 2019; 8:jcm8081107. [PMID: 31349718 PMCID: PMC6723109 DOI: 10.3390/jcm8081107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/21/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
A subset of pituitary neuroendocrine tumors (PitNETs) have an aggressive behavior, showing resistance to treatment and/or multiple recurrences in spite of the optimal use of standard therapies (surgery, conventional medical treatments, and radiotherapy). To date, for aggressive PitNETs, temozolomide (TMZ) has been the most used therapeutic option, and has resulted in an improvement in the five-year survival rate in responders. However, given the fact that roughly only one third of patients showed a partial or complete radiological response on the first course of TMZ, and even fewer patients responded to a second course of TMZ, other treatment options are urgently needed. Emerging therapies consist predominantly of peptide receptor radionuclide therapy (20 cases), vascular endothelial growth factor receptor-targeted therapy (12 cases), tyrosine kinase inhibitors (10 cases), mammalian target of rapamycin (mTOR) inhibitors (six cases), and more recently, immune checkpoint inhibitors (one case). Here, we present the available clinical cases published in the literature for each of these treatments. The therapies that currently show the most promise (based on the achievement of partial radiological response in a certain number of cases) are immune checkpoint inhibitors, peptide receptor radionuclide therapy, and vascular endothelial growth factor receptor-targeted therapy. In the future, further improvement of these therapies and the development of other novel therapies, their use in personalized medicine, and a better understanding of combination therapies, will hopefully result in better outcomes for patients bearing aggressive PitNETs.
Collapse
Affiliation(s)
- Mirela Diana Ilie
- INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon, 28 Laennec Street, 69008 Lyon, France
- "Claude Bernard" Lyon 1 University, University of Lyon, 43 "11 Novembre 1918" Boulevard, 69100 Villeurbanne, France
- Endocrinology Department, "C.I.Parhon" National Institute of Endocrinology, 34-36 Aviatorilor Boulevard, 011863 Bucharest, Romania
| | - Hélène Lasolle
- INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon, 28 Laennec Street, 69008 Lyon, France
- "Claude Bernard" Lyon 1 University, University of Lyon, 43 "11 Novembre 1918" Boulevard, 69100 Villeurbanne, France
- "Groupement Hospitalier Est" Hospices Civils de Lyon, Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, 59 Pinel Boulevard, 69677 Bron, France
| | - Gérald Raverot
- INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon, 28 Laennec Street, 69008 Lyon, France.
- "Claude Bernard" Lyon 1 University, University of Lyon, 43 "11 Novembre 1918" Boulevard, 69100 Villeurbanne, France.
- "Groupement Hospitalier Est" Hospices Civils de Lyon, Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, 59 Pinel Boulevard, 69677 Bron, France.
| |
Collapse
|
20
|
Weissenrieder JS, Neighbors JD, Mailman RB, Hohl RJ. Cancer and the Dopamine D 2 Receptor: A Pharmacological Perspective. J Pharmacol Exp Ther 2019; 370:111-126. [PMID: 31000578 DOI: 10.1124/jpet.119.256818] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/16/2019] [Indexed: 01/12/2023] Open
Abstract
The dopamine D2 receptor (D2R) family is upregulated in many cancers and tied to stemness. Reduced cancer risk has been correlated with disorders such as schizophrenia and Parkinson's disease, in which dopaminergic drugs are used. D2R antagonists are reported to have anticancer efficacy in cell culture and animal models where they have reduced tumor growth, induced autophagy, affected lipid metabolism, and caused apoptosis, among other effects. This has led to several hypotheses, the most prevalent being that D2R ligands may be a novel approach to cancer chemotherapy. This hypothesis is appealing because of the large number of approved and experimental drugs of this class that could be repurposed. We review the current state of the literature and the evidence for and against this hypothesis. When the existing literature is evaluated from a pharmacological context, one of the striking findings is that the concentrations needed for cytotoxic effects of D2R antagonists are orders of magnitude higher than their affinity for this receptor. Although additional definitive studies will provide further clarity, our hypothesis is that targeting D2-like dopamine receptors may only yield useful ligands for cancer chemotherapy in rare cases.
Collapse
Affiliation(s)
- Jillian S Weissenrieder
- Biomedical Sciences Program (J.S.W.) and Departments of Medicine (J.D.N., R.J.H.) and Pharmacology (J.D.N., R.B.M., R.J.H.), Penn State College of Medicine and Penn State Cancer Institute, Hershey, Pennsylvania
| | - Jeffrey D Neighbors
- Biomedical Sciences Program (J.S.W.) and Departments of Medicine (J.D.N., R.J.H.) and Pharmacology (J.D.N., R.B.M., R.J.H.), Penn State College of Medicine and Penn State Cancer Institute, Hershey, Pennsylvania
| | - Richard B Mailman
- Biomedical Sciences Program (J.S.W.) and Departments of Medicine (J.D.N., R.J.H.) and Pharmacology (J.D.N., R.B.M., R.J.H.), Penn State College of Medicine and Penn State Cancer Institute, Hershey, Pennsylvania
| | - Raymond J Hohl
- Biomedical Sciences Program (J.S.W.) and Departments of Medicine (J.D.N., R.J.H.) and Pharmacology (J.D.N., R.B.M., R.J.H.), Penn State College of Medicine and Penn State Cancer Institute, Hershey, Pennsylvania
| |
Collapse
|
21
|
Le Tissier P, Fiordelisio Coll T, Mollard P. The Processes of Anterior Pituitary Hormone Pulse Generation. Endocrinology 2018; 159:3524-3535. [PMID: 30020429 DOI: 10.1210/en.2018-00508] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022]
Abstract
More than 60 years ago, Geoffrey Harris described his "neurohumoral theory," in which the regulation of pituitary hormone secretion was a "simple" hierarchal relationship, with the hypothalamus as the controller. In models based on this theory, the electrical activity of hypothalamic neurons determines the release of hypophysiotropic hormones into the portal circulation, and the pituitary simply responds with secretion of a pulse of hormone into the bloodstream. The development of methodologies allowing the monitoring of the activities of members of the hypothalamic-vascular-pituitary unit is increasingly allowing dissection of the mechanisms generating hypothalamic and pituitary pulses. These have revealed that whereas hypothalamic input is required, its role as a driver of pulsatile pituitary hormone secretion varies between pituitary axes. The organization of pituitary cells has a key role in the modification of their response to hypophysiotropic factors that can lead to a memory of previous demand and enhanced function. Feedback can lead to oscillatory hormone output that is independent of pulses of hypophysiotropic factors and instead, results from the temporal relationship between pituitary output and target organ response. Thus, the mechanisms underlying the generation of pulses cannot be generalized, and the circularity of feedforward and feedback interactions must be considered to understand both normal physiological function and pathology. We describe some examples of the clinical implications of recognizing the importance of the pituitary and target organs in pulse generation and suggest avenues for future research in both the short and long term.
Collapse
Affiliation(s)
- Paul Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Tatiana Fiordelisio Coll
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, University of Montpellier, Montpellier, France
- Laboratorio de Neuroendocrinología Comparada, Departamento de Ecología y Recursos Naturales, Biología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, México City, Distrito Federal, México
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, University of Montpellier, Montpellier, France
| |
Collapse
|
22
|
Maghathe T, Miller WK, Mugge L, Mansour TR, Schroeder J. Immunotherapy and potential molecular targets for the treatment of pituitary adenomas resistant to standard therapy: a critical review of potential therapeutic targets and current developments. J Neurosurg Sci 2018; 64:71-83. [PMID: 30014686 DOI: 10.23736/s0390-5616.18.04419-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Pituitary adenomas (PAs) are primary central nervous system (CNS) tumors, accounting for as much as 25% of intracranial neoplasms. Although existing remedies show success in treating most PAs, treatment of invasive and non-functioning PAs, in addition to functioning PAs unresponsive to standard therapy, remains challenging. With the continually increasing understanding of biochemical pathways involved in tumorigenesis, immunotherapy stands as a promising alternative therapy for pituitary tumors that are resistant to standard therapy. EVIDENCE ACQUISITION A literature search was conducted of the PubMed database for immunotherapies of PAs. The search yielded a total of 2621 articles, 26 of which were included in our discussion. EVIDENCE SYNTHESIS Several pathologically expressed molecules could potentially serve as promising targets of current or future immunotherapies for PAs. Programmed death ligand-1, matrix metalloproteinases, EpCAM (Trop1) and Trop2, cancer-testis antigen MAGE-A3, epidermal growth factor receptor (EGFR), folate receptor alpha, vascular endothelial growth factor, and galectin-3 have all been implicated as crucial factors involved with tumor survival and invasion. Inhibition of these pathways may prove efficacious in the management of invasive and treatment-resistant PAs. CONCLUSIONS Rapid advancements in tumor immunology may increase the probability of successful treatment of PAs by exploitation of the normal immune response or by targeting novel proteins. Current research on many of the targets reviewed in this article are successfully being utilized to manage various neoplastic disease including CNS tumors. These therapies may eventually play a key role in the treatment of PAs that do not respond to standard therapy.
Collapse
Affiliation(s)
- Tamara Maghathe
- Division of Neurosurgery, Department of Surgery, University of Toledo Medical Center, Toledo, OH, USA
| | - William K Miller
- Division of Neurosurgery, Department of Surgery, University of Toledo Medical Center, Toledo, OH, USA
| | - Luke Mugge
- Division of Neurosurgery, Department of Surgery, University of Toledo Medical Center, Toledo, OH, USA
| | - Tarek R Mansour
- Division of Neurosurgery, Department of Surgery, University of Toledo Medical Center, Toledo, OH, USA
| | - Jason Schroeder
- Division of Neurosurgery, Department of Surgery, University of Toledo Medical Center, Toledo, OH, USA -
| |
Collapse
|
23
|
Abstract
Endocrine organs secrete a variety of hormones involved in the regulation of a multitude of body functions. Although pancreatic islets were discovered at the turn of the 19th century, other endocrine glands remained commonly described as diffuse endocrine systems. Over the last two decades, development of new imaging techniques and genetically-modified animals with cell-specific fluorescent tags or specific hormone deficiencies have enabled in vivo imaging of endocrine organs and revealed intricate endocrine cell network structures and plasticity. Overall, these new tools have revolutionized our understanding of endocrine function. The overarching aim of this Review is to describe the current mechanistic understanding that has emerged from imaging studies of endocrine cell network structure/function relationships in animal models, with a particular emphasis on the pituitary gland and the endocrine pancreas.
Collapse
Affiliation(s)
- Patrice Mollard
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, F-34094, Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, F-34094, Montpellier, France.
| |
Collapse
|
24
|
Roney MSI, Park SK. Antipsychotic dopamine receptor antagonists, cancer, and cancer stem cells. Arch Pharm Res 2018; 41:384-408. [PMID: 29556831 DOI: 10.1007/s12272-018-1017-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 02/27/2018] [Indexed: 12/12/2022]
Abstract
Cancer is one of the deadliest diseases in the world. Despite extensive studies, treating metastatic cancers remains challenging. Years of research have linked a rare set of cells known as cancer stem cells (CSCs) to drug resistance, leading to the suggestion that eradication of CSCs might be an effective therapeutic strategy. However, few drug candidates are active against CSCs. New drug discovery is often a lengthy process. Drug screening has been advantageous in identifying drug candidates. Current understanding of cancer biology has revealed various clues to target cancer from different points of view. Many studies have found dopamine receptors (DRs) in various cancers. Therefore, DR antagonists have attracted a lot of attention in cancer research. Recently, a group of antipsychotic DR antagonists has been demonstrated to possess remarkable abilities to restrain and sensitize CSCs to existing chemotherapeutics by a process called differentiation approach. In this review, we will describe current aspects of CSC-targeting therapeutics, antipsychotic DR antagonists, and their extraordinary abilities to fight cancer.
Collapse
Affiliation(s)
- Md Saiful Islam Roney
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, Republic of Korea
| | - Song-Kyu Park
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, Republic of Korea.
- Research Driven Hospital, Korea University Guro Hospital, Biomedical Research Center, Seoul, 08308, Republic of Korea.
| |
Collapse
|
25
|
Spoletini M, Taurone S, Tombolini M, Minni A, Altissimi G, Wierzbicki V, Giangaspero F, Parnigotto PP, Artico M, Bardella L, Agostinelli E, Pastore FS. Trophic and neurotrophic factors in human pituitary adenomas (Review). Int J Oncol 2017; 51:1014-1024. [PMID: 28902350 DOI: 10.3892/ijo.2017.4120] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/17/2017] [Indexed: 11/06/2022] Open
Abstract
The pituitary gland is an organ that functionally connects the hypothalamus with the peripheral organs. The pituitary gland is an important regulator of body homeostasis during development, stress, and other processes. Pituitary adenomas are a group of tumors arising from the pituitary gland: they may be subdivided in functional or non-functional, depending on their hormonal activity. Some trophic and neurotrophic factors seem to play a key role in the development and maintenance of the pituitary function and in the regulation of hypothalamo-pituitary-adrenocortical axis activity. Several lines of evidence suggest that trophic and neurotrophic factors may be involved in pituitary function, thus suggesting a possible role of the trophic and neurotrophic factors in the normal development of pituitary gland and in the progression of pituitary adenomas. Additional studies might be necessary to better explain the biological role of these molecules in the development and progression of this type of tumor. In this review, in light of the available literature, data on the following neurotrophic factors are discussed: ciliary neurotrophic factor (CNTF), transforming growth factors β (TGF‑β), glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), vascular endothelial growth inhibitor (VEGI), fibroblast growth factors (FGFs) and epidermal growth factor (EGF) which influence the proliferation and growth of pituitary adenomas.
Collapse
Affiliation(s)
- Marialuisa Spoletini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, 'Sapienza' University of Rome, Rome, Italy
| | - Samanta Taurone
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Mario Tombolini
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Antonio Minni
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | | | | | - Felice Giangaspero
- Department of Radiology, Oncology and Anatomic Pathology, 'Sapienza' University of Rome, Rome, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (TES) Onlus, Padua, Italy
| | - Marco Artico
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Lia Bardella
- Department of Neurology and Psychiatry, 'Sapienza' University of Rome, Rome, Italy
| | - Enzo Agostinelli
- Department of Biochemical Sciences 'A. Rossi Fanelli', 'Sapienza' University of Rome, Rome, Italy
| | - Francesco Saverio Pastore
- Department of Systems' Medicine, Division of Neurosurgery, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|