1
|
Mariniello K, Pittaway JFH, Altieri B, Borges KS, Hadjidemetriou I, Ribeiro C, Ruiz-Babot G, Tourigny DS, Lim JA, Foster J, Cleaver J, Sosabowski J, Rahman N, Doroszko M, Hantel C, Sigala S, Abate A, Tamburello M, Kiseljak-Vassiliades K, Wierman M, Hall C, Parvanta L, Abdel-Aziz TE, Chung TT, Marco AD, Palazzo F, Gomez-Sanchez CE, Taylor DR, Rayner O, Ronchi CL, Gaston-Massuet C, Sbiera S, Drake WM, Rognoni E, Kroiss M, Breault DT, Fassnacht M, Guasti L. Dlk1 is a novel adrenocortical stem/progenitor cell marker that predicts malignancy in adrenocortical carcinoma. Cancer Commun (Lond) 2025. [PMID: 40035383 DOI: 10.1002/cac2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025] Open
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James F H Pittaway
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Kleiton Silva Borges
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technical, University Dresden, Dresden, Germany
| | | | - Jiang A Lim
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Foster
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Julie Cleaver
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Jane Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Nafis Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Milena Doroszko
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, District of Columbia, USA
| | - Margaret Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, District of Columbia, USA
| | - Charlotte Hall
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laila Parvanta
- Department of Surgery, St Bartholomew's Hospital, West Smithfield, London, UK
| | - Tarek E Abdel-Aziz
- Department of Surgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Teng-Teng Chung
- Department of Endocrinology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Aimee Di Marco
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London, UK
| | - Fausto Palazzo
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London, UK
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David R Taylor
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London, UK
| | - Oliver Rayner
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London, UK
| | - Cristina L Ronchi
- Institute of Metabolism and System Research College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - William M Drake
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Emanuel Rognoni
- Centre for Cell Biology & Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthias Kroiss
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, München, Germany
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
2
|
Feely S, Mullen N, Donlon PT, Reidy E, Challapalli RS, Hassany M, Sorushanova A, Martinez ER, Owens P, Quinn AM, Pandit A, Harhen B, Finn DP, Hantel C, O'Halloran M, Prakash P, Dennedy MC. Development and Characterization of 3-Dimensional Cell Culture Models of Adrenocortical Carcinoma. Endocrinology 2024; 166:bqae159. [PMID: 39656817 DOI: 10.1210/endocr/bqae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/23/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy of the adrenal cortex that is associated with a poor prognosis. Developing effective treatment options for ACC is challenging owing to the current lack of representative preclinical models. This study addressed this limitation by developing and characterizing 3-dimensional (3D) cell cultures incorporating the ACC cell lines, MUC-1, HAC15, and H295R in a type I collagen matrix. ACC tissue samples were analyzed by immunohistochemistry to determine the presence of type I collagen in the tumor microenvironment. Cell viability and proliferation were assessed using flow cytometry and confocal microscopy. mRNA expression of steroidogenic enzymes and steroid secretion was analyzed by comparing the 3D and monolayer cell culture models. All cells were successfully cultured in a type I collagen matrix, which is highly expressed in the ACC tumor microenvironment and showed optimal viability until day 7. All 3 models showed increased metabolic and proliferative activity over time. Three-dimensional cell cultures were steroidogenic and demonstrated increased resistance to the gold standard chemotherapy, mitotane, compared with monolayer. The use of these models may lead to an improved understanding of disease pathology and provide a better representative platform for testing and screening of potential therapies.
Collapse
Affiliation(s)
- Sarah Feely
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Nathan Mullen
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Padraig T Donlon
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Eileen Reidy
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Ritihaas Surya Challapalli
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Mariam Hassany
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Anna Sorushanova
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
| | - Eduardo Ribes Martinez
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
- Science Foundation Ireland (SFI) Research Centre for Research in Medical Devices (CURAM), Biomedical Science Building, University of Galway, Galway, H91 TK33, Ireland
| | - Peter Owens
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, University of Galway, Galway, H91 TK33, Ireland
| | - Anne Marie Quinn
- Department of Anatomic Pathology, Galway University Hospital, Galway, H91 YR71, Ireland
| | - Abhay Pandit
- Science Foundation Ireland (SFI) Research Centre for Research in Medical Devices (CURAM), Biomedical Science Building, University of Galway, Galway, H91 TK33, Ireland
| | - Brendan Harhen
- Biological Mass Spectrometry Core Facility, University of Galway, Galway, H91 TK33, Ireland
| | - David P Finn
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
- Science Foundation Ireland (SFI) Research Centre for Research in Medical Devices (CURAM), Biomedical Science Building, University of Galway, Galway, H91 TK33, Ireland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091 Zurich, Switzerland
| | - Martin O'Halloran
- Science Foundation Ireland (SFI) Research Centre for Research in Medical Devices (CURAM), Biomedical Science Building, University of Galway, Galway, H91 TK33, Ireland
- Translational Medical Device Laboratory, University of Galway, Galway, H91 V4AY, Ireland
| | - Punit Prakash
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Michael C Dennedy
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, H91 V4AY, Ireland
- Science Foundation Ireland (SFI) Research Centre for Research in Medical Devices (CURAM), Biomedical Science Building, University of Galway, Galway, H91 TK33, Ireland
| |
Collapse
|
3
|
Mariniello K, Pittaway JFH, Altieri B, Borges KS, Hadjidemetriou I, Ribeiro C, Ruiz-Babot G, Lim JA, Foster J, Cleaver J, Sosabowski J, Rahman N, Doroszko M, Hantel C, Sigala S, Abate A, Tamburello M, Kiseljak-Vassiliades K, Wierman M, Parvanta L, Abdel-Aziz TE, Chung TT, Di Marco A, Palazzo F, Gomez-Sanchez CE, Taylor DR, Rayner O, Ronchi CL, Gaston-Massuet C, Sbiera S, Drake WM, Rognoni E, Kroiss M, Breault DT, Fassnacht M, Guasti L. Dlk1 is a novel adrenocortical stem/progenitor cell marker that predicts malignancy in adrenocortical carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609117. [PMID: 39229217 PMCID: PMC11370565 DOI: 10.1101/2024.08.22.609117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Disruption of processes involved in tissue development and homeostatic self-renewal is increasingly implicated in cancer initiation, progression, and recurrence. The adrenal cortex is a dynamic tissue that undergoes life-long turnover. Here, using genetic fate mapping and murine adrenocortical carcinoma (ACC) models, we have identified a population of adrenocortical stem cells that express delta-like non-canonical Notch ligand 1 (DLK1). These cells are active during development, near dormant postnatally but are re-expressed in ACC. In a study of over 200 human ACC samples, we have shown DLK1 expression is ubiquitous and is an independent prognostic marker of recurrence-free survival. Paradoxically, despite its progenitor role, spatial transcriptomic analysis has identified DLK1 expressing cell populations to have increased steroidogenic potential in human ACC, a finding also observed in four human and one murine ACC cell lines. Finally, the cleavable DLK1 ectodomain is measurable in patients' serum and can discriminate between ACC and other adrenal pathologies with high sensitivity and specificity to aid in diagnosis and follow-up of ACC patients. These data demonstrate a prognostic role for DLK1 in ACC, detail its hierarchical expression in homeostasis and oncogenic transformation and propose a role for its use as a biomarker in this malignancy.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James F H Pittaway
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Kleiton Silva Borges
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technical, University Dresden, Dresden, Germany
| | - Jiang A Lim
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Foster
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Julie Cleaver
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jane Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Nafis Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Milena Doroszko
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091 Zurich, Switzerland
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124 Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124 Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124 Brescia, Italy
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, DC, USA
| | - Margaret Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, DC, USA
| | - Laila Parvanta
- Department of Surgery, St Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, United Kingdom
| | - Tarek E Abdel-Aziz
- Department of Surgery, University College London Hospitals NHS Foundation Trust, London NW1 2PG, United Kingdom
| | - Teng-Teng Chung
- Department of Endocrinology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, United Kingdom
| | - Aimee Di Marco
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London W12 0HS, United Kingdom
| | - Fausto Palazzo
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London W12 0HS, United Kingdom
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - David R Taylor
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London SE5 9RS, United Kingdom
| | - Oliver Rayner
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London SE5 9RS, United Kingdom
| | - Cristina L Ronchi
- Institute of Metabolism and System Research College of Medical and Dental Sciences, University of Birmingham, B15 2TT, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - William M Drake
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Emanuel Rognoni
- Centre for Cell Biology & Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthias Kroiss
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, 80336 München, Germany
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
4
|
Sedlack AJH, Hatfield SJ, Kumar S, Arakawa Y, Roper N, Sun NY, Nilubol N, Kiseljak-Vassiliades K, Hoang CD, Bergsland EK, Hernandez JM, Pommier Y, del Rivero J. Preclinical Models of Adrenocortical Cancer. Cancers (Basel) 2023; 15:2873. [PMID: 37296836 PMCID: PMC10251941 DOI: 10.3390/cancers15112873] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
Adrenocortical cancer is an aggressive endocrine malignancy with an incidence of 0.72 to 1.02 per million people/year, and a very poor prognosis with a five-year survival rate of 22%. As an orphan disease, clinical data are scarce, meaning that drug development and mechanistic research depend especially on preclinical models. While a single human ACC cell line was available for the last three decades, over the last five years, many new in vitro and in vivo preclinical models have been generated. Herein, we review both in vitro (cell lines, spheroids, and organoids) and in vivo (xenograft and genetically engineered mouse) models. Striking leaps have been made in terms of the preclinical models of ACC, and there are now several modern models available publicly and in repositories for research in this area.
Collapse
Affiliation(s)
- Andrew J. H. Sedlack
- Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Samual J. Hatfield
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Suresh Kumar
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yasuhiro Arakawa
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Nitin Roper
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Nai-Yun Sun
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO 80016, USA
| | - Chuong D. Hoang
- Thoracic Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Emily K. Bergsland
- University of California, San Francisco (UCSF) Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | | | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jaydira del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Pignatti E, Flück CE. Adrenal cortex development and related disorders leading to adrenal insufficiency. Mol Cell Endocrinol 2021; 527:111206. [PMID: 33607267 DOI: 10.1016/j.mce.2021.111206] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
The adult human adrenal cortex produces steroid hormones that are crucial for life, supporting immune response, glucose homeostasis, salt balance and sexual maturation. It consists of three histologically distinct and functionally specialized zones. The fetal adrenal forms from mesodermal material and produces predominantly adrenal C19 steroids from its fetal zone, which involutes after birth. Transition to the adult cortex occurs immediately after birth for the formation of the zona glomerulosa and fasciculata for aldosterone and cortisol production and continues through infancy until the zona reticularis for adrenal androgen production is formed with adrenarche. The development of this indispensable organ is complex and not fully understood. This article gives an overview of recent knowledge gained of adrenal biology from two perspectives: one, from basic science studying adrenal development, zonation and homeostasis; and two, from adrenal disorders identified in persons manifesting with various isolated or syndromic forms of primary adrenal insufficiency.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
6
|
Borges KS, Pignatti E, Leng S, Kariyawasam D, Ruiz-Babot G, Ramalho FS, Taketo MM, Carlone DL, Breault DT. Wnt/β-catenin activation cooperates with loss of p53 to cause adrenocortical carcinoma in mice. Oncogene 2020; 39:5282-5291. [PMID: 32561853 PMCID: PMC7378041 DOI: 10.1038/s41388-020-1358-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/28/2020] [Accepted: 06/04/2020] [Indexed: 12/23/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare and aggressive malignancy with limited therapeutic options. The lack of mouse models that recapitulate the genetics of ACC has hampered progress in the field. We analyzed The Cancer Genome Atlas (TCGA) dataset for ACC and found that patients harboring alterations in both p53/Rb and Wnt/β-catenin signaling pathways show a worse prognosis compared with patients that harbored alterations in only one. To model this, we utilized the Cyp11b2(AS)Cre mouse line to generate mice with adrenocortical-specific Wnt/β-catenin activation, Trp53 deletion, or the combination of both. Mice with targeted Wnt/β-catenin activation or Trp53 deletion showed no changes associated with tumor formation. In contrast, alterations in both pathways led to ACC with pulmonary metastases. Similar to ACCs in humans, these tumors produced increased levels of corticosterone and aldosterone and showed a high proliferation index. Gene expression analysis revealed that mouse tumors exhibited downregulation of Star and Cyp11b1 and upregulation of Ezh2, similar to ACC patients with a poor prognosis. Altogether, these data show that altering both Wnt/β-catenin and p53/Rb signaling is sufficient to drive ACC in mouse. This autochthonous model of ACC represents a new tool to investigate the biology of ACC and to identify new treatment strategies.
Collapse
Affiliation(s)
- Kleiton Silva Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.,Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Emanuele Pignatti
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.,Division of Medical Sciences, Harvard Medical School, Boston, MA, 02115, USA
| | - Dulanjalee Kariyawasam
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Fernando Silva Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, 606-8506, Japan
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.,Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA. .,Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
7
|
Kar A, Wierman ME, Kiseljak-Vassiliades K. Update on in-vivo preclinical research models in adrenocortical carcinoma. Curr Opin Endocrinol Diabetes Obes 2020; 27:170-176. [PMID: 32304391 PMCID: PMC8103733 DOI: 10.1097/med.0000000000000543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize recent advances on development of in vivo preclinical models of adrenocortical carcinoma (ACC). RECENT FINDINGS Significant progress has been achieved in the underlying molecular mechanisms of adrenocortical tumorigenesis over the last decade, and recent comprehensive profiling analysis of ACC tumors identified several genetic and molecular drivers of this disease. Therapeutic breakthroughs, however, have been limited because of the lack of preclinical models recapitulating the molecular features and heterogeneity of the tumors. Recent publications on genetically engineered mouse models and development of patient-derived ACC xenografts in both nude mice and humanized mice now provide researchers with novel tools to explore therapeutic targets in the context of heterogeneity and tumor microenvironment in human ACC. SUMMARY We review current in-vivo models of ACC and discuss potential therapeutic opportunities that have emerged from these studies.
Collapse
Affiliation(s)
- Adwitiya Kar
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora
| | - Margaret E. Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora
- Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora
- Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| |
Collapse
|
8
|
Hadjidemetriou I, Mariniello K, Ruiz-Babot G, Pittaway J, Mancini A, Mariannis D, Gomez-Sanchez CE, Parvanta L, Drake WM, Chung TT, Abdel-Aziz TE, DiMarco A, Palazzo FF, Wierman ME, Kiseljak-Vassiliades K, King PJ, Guasti L. DLK1/PREF1 marks a novel cell population in the human adrenal cortex. J Steroid Biochem Mol Biol 2019; 193:105422. [PMID: 31265901 PMCID: PMC6736711 DOI: 10.1016/j.jsbmb.2019.105422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 05/10/2019] [Accepted: 06/28/2019] [Indexed: 01/20/2023]
Abstract
The adrenal cortex governs fundamental metabolic processes though synthesis of glucocorticoid, mineralocorticoids and androgens. Studies in rodents have demonstrated that the cortex undergoes a self-renewal process and that capsular/subcapsular stem/progenitor cell pools differentiate towards functional steroidogenic cells supporting the dynamic centripetal streaming of adrenocortical cells throughout life. We previously demonstrated that the Notch atypical ligand Delta-like homologue 1 (DLK1)/preadipocyte factor 1 (PREF1) is expressed in subcapsular Sf1 and Shh-positive, CYP11B1-negative and CYP11B2-partially positive cortical progenitor cells in rat adrenals, and that secreted DLK1 can modulate GLI1 expression in H295R cells. Here we show that the human adrenal cortex remodels with age to generate clusters of relatively undifferentiated cells expressing DLK1. These clusters (named DLK1-expressing cell clusters or DCCs) increased with age in size and were found to be different entities to aldosterone-producing cell clusters, another well-characterized and age-dependent cluster structure. DLK1 was markedly overexpressed in adrenocortical carcinomas but not in aldosterone-producing adenomas. Thus, this data identifies a novel cell population in the human adrenal cortex and might suggest a yet-to be identified role of DLK1 in the pathogenesis of adrenocortical carcinoma in humans.
Collapse
Affiliation(s)
- Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gerard Ruiz-Babot
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James Pittaway
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alessandra Mancini
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Demetris Mariannis
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Celso E Gomez-Sanchez
- G.V. (Sonny) Montgomery VA Medical Center and Department of Medicine, University of Mississippi Medical Centre, Jackson MS, USA
| | - Laila Parvanta
- Department of Surgery, St Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK
| | - William M Drake
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Teng-Teng Chung
- Department of Endocrinology, University College Hospital NHS Foundation Trust, NW1 2PG, London, UK
| | - Tarek Ezzat Abdel-Aziz
- Department of Endocrinology, University College Hospital NHS Foundation Trust, NW1 2PG, London, UK
| | - Aimee DiMarco
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London, UK
| | - Fausto F Palazzo
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London, UK
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO, USA; Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, Aurora, CO, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO, USA; Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, Aurora, CO, USA
| | - Peter J King
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London, School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
9
|
Pittaway JFH, Guasti L. Pathobiology and genetics of adrenocortical carcinoma. J Mol Endocrinol 2019; 62:R105-R119. [PMID: 30072419 DOI: 10.1530/jme-18-0122] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/02/2018] [Indexed: 12/28/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with an incidence worldwide of 0.7-2.0 cases/million/year. Initial staging is the most important factor in determining prognosis. If diagnosed early, complete surgical resection +/- adjuvant treatment can lead to 5-year survival of up to 80%. However, often it is diagnosed late and in advanced disease, 5-year survival is <15% with a high recurrence rate even after radical surgery. The mainstay of adjuvant treatment is with the drug mitotane. Mitotane has a specific cytotoxic effect on steroidogenic cells of the adrenal cortex, but despite this, progression through treatment is common. Developments in genetic analysis in the form of next-generation sequencing, aided by bioinformatics, have enabled high-throughput molecular characterisation of these tumours. This, in addition to a better appreciation of the processes of physiological, homeostatic self-renewal of the adrenal cortex, has furthered our understanding of the pathogenesis of this malignancy. In this review, we have detailed the pathobiology and genetic alterations in adrenocortical carcinoma by integrating current understanding of homeostasis and self-renewal in the normal adrenal cortex with molecular profiling of tumours from recent genetic analyses. Improved understanding of the mechanisms involved in self-renewal and stem cell hierarchy in normal human adrenal cortices, together with the identification of cell populations likely to be co-opted by oncogenic mutations, will enable further progress in the definition of the molecular pathways involved in the pathogenesis of ACC. The combination of these advances eventually will lead to the development of novel, effective and personalised strategies to eradicate molecularly annotated ACCs.
Collapse
Affiliation(s)
- James F H Pittaway
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
10
|
Gahete MD, Jimenez-Vacas JM, Alors-Perez E, Herrero-Aguayo V, Fuentes-Fayos AC, Pedraza-Arevalo S, Castaño JP, Luque RM. Mouse models in endocrine tumors. J Endocrinol 2018; 240:JOE-18-0571.R1. [PMID: 30475226 DOI: 10.1530/joe-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
Endocrine and neuroendocrine tumors comprise a highly heterogeneous group of neoplasms that can arise from (neuro)endocrine cells, either from endocrine glands or from the widespread diffuse neuroendocrine system, and, consequently, are widely distributed throughout the body. Due to their diversity, heterogeneity and limited incidence, studying in detail the molecular and genetic alterations that underlie their development and progression is still a highly elusive task. This, in turn, hinders the discovery of novel therapeutic options for these tumors. To circumvent these limitations, numerous mouse models of endocrine and neuroendocrine tumors have been developed, characterized and used in pre-clinical, co-clinical (implemented in mouse models and patients simultaneously) and post-clinical studies, for they represent powerful and necessary tools in basic and translational tumor biology research. Indeed, different in vivo mouse models, including cell line-based xenografts (CDXs), patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs), have been used to delineate the development, progression and behavior of human tumors. Results gained with these in vivo models have facilitated the clinical application in patients of diverse breakthrough discoveries made in this field. Herein, we review the generation, characterization and translatability of the most prominent mouse models of endocrine and neuroendocrine tumors reported to date, as well as the most relevant clinical implications obtained for each endocrine and neuroendocrine tumor type.
Collapse
Affiliation(s)
- Manuel D Gahete
- M Gahete, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, 14011, Spain
| | - Juan M Jimenez-Vacas
- J Jimenez-Vacas, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Emilia Alors-Perez
- E Alors-Perez, Department of Cell Biology, Physiology and Inmunology, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC) / University of Cordoba, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- V Herrero-Aguayo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- A Fuentes-Fayos, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Sergio Pedraza-Arevalo
- S Pedraza-Arevalo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- J Castaño, Dpt. of Cell Biology-University of Córdoba, IMIBIC-Maimonides Biomedical Research Institute of Cordoba, Cordoba, E-14004, Spain
| | - Raul M Luque
- R Luque, Dept of Cell Biology, Phisiology and Inmunology, Section of Cell Biology, University of Cordoba, Cordoba, Spain, Cordoba, 14014, Spain
| |
Collapse
|
11
|
Mostaghel EA, Zhang A, Hernandez S, Marck BT, Zhang X, Tamae D, Biehl HE, Tretiakova M, Bartlett J, Burns J, Dumpit R, Ang L, Matsumoto AM, Penning TM, Balk SP, Morrissey C, Corey E, True LD, Nelson PS. Contribution of Adrenal Glands to Intratumor Androgens and Growth of Castration-Resistant Prostate Cancer. Clin Cancer Res 2018; 25:426-439. [PMID: 30181386 DOI: 10.1158/1078-0432.ccr-18-1431] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/01/2018] [Accepted: 08/29/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Tumor androgens in castration-resistant prostate cancer (CRPC) reflect de novo intratumoral synthesis or adrenal androgens. We used C.B.-17 SCID mice in which we observed adrenal CYP17A activity to isolate the impact of adrenal steroids on CRPC tumors in vivo. EXPERIMENTAL DESIGN We evaluated tumor growth and androgens in LuCaP35CR and LuCaP96CR xenografts in response to adrenalectomy (ADX). We assessed protein expression of key steroidogenic enzymes in 185 CRPC metastases from 42 patients. RESULTS Adrenal glands of intact and castrated mice expressed CYP17A. Serum DHEA, androstenedione (AED), and testosterone (T) in castrated mice became undetectable after ADX (all P < 0.05). ADX prolonged median survival (days) in both CRPC models (33 vs. 179; 25 vs. 301) and suppressed tumor steroids versus castration alone (T 0.64 pg/mg vs. 0.03 pg/mg; DHT 2.3 pg/mg vs. 0.23 pg/mg; and T 0.81 pg/mg vs. 0.03 pg/mg, DHT 1.3 pg/mg vs. 0.04 pg/mg; all P ≤ 0.001). A subset of tumors recurred with increased steroid levels, and/or induction of androgen receptor (AR), truncated AR variants, and glucocorticoid receptor (GR). Metastases from 19 of 35 patients with AR positive tumors concurrently expressed enzymes for adrenal androgen utilization and nine expressed enzymes for de novo steroidogenesis (HSD3B1, CYP17A, AKR1C3, and HSD17B3). CONCLUSIONS Mice are appropriate for evaluating adrenal impact of steroidogenesis inhibitors. A subset of ADX-resistant CRPC tumors demonstrate de novo androgen synthesis. Tumor growth and androgens were suppressed more strongly by surgical ADX than prior studies using abiraterone, suggesting reduction in adrenally-derived androgens beyond that achieved by abiraterone may have clinical benefit. Proof-of-concept studies with agents capable of achieving true "nonsurgical ADX" are warranted.
Collapse
Affiliation(s)
- Elahe A Mostaghel
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington. .,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ailin Zhang
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Brett T Marck
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington
| | - Xiaotun Zhang
- Department of Urology, University of Washington, Seattle, Washington
| | - Daniel Tamae
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Maria Tretiakova
- Department of Pathology, University of Washington, Seattle, Washington
| | - Jon Bartlett
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - John Burns
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ruth Dumpit
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lisa Ang
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alvin M Matsumoto
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington
| | - Trevor M Penning
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven P Balk
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Lawrence D True
- Department of Pathology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
12
|
Lalli E, Luconi M. The next step: mechanisms driving adrenocortical carcinoma metastasis. Endocr Relat Cancer 2018; 25:R31-R48. [PMID: 29142005 DOI: 10.1530/erc-17-0440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/20/2022]
Abstract
Endocrine tumors have the peculiarity to become clinically evident not only due to symptoms related to space occupation by the growing lesion, similarly to most other tumors, but also, and most often, because of their specific hormonal secretion, which significantly contributes to their pathological burden. Malignant endocrine tumors, in addition, have the ability to produce distant metastases. Here, we critically review the current knowledge about mechanisms and biomarkers characterizing the metastatic process in adrenocortical carcinoma (ACC), a rare endocrine malignancy with a high risk of relapse and metastatization even when the primary tumor is diagnosed and surgically removed at an early stage. We highlight perspectives of future research in the domain and possible new therapeutic avenues based on targeting factors having an important role in the metastatic process of ACC.
Collapse
Affiliation(s)
- Enzo Lalli
- Université Côte d'AzurValbonne, France
- CNRS UMR7275Valbonne, France
- NEOGENEX CNRS International Associated LaboratoryValbonne, France
- Institut de Pharmacologie Moléculaire et CellulaireValbonne, France
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio'University of Florence, Florence, Italy
| |
Collapse
|
13
|
Morin A, Ruggiero C, Robidel E, Doghman-Bouguerra M, Das AT, Castellano R, Josselin E, Favier J, Lalli E. Establishment of a mouse xenograft model of metastatic adrenocortical carcinoma. Oncotarget 2017; 8:51050-51057. [PMID: 28881628 PMCID: PMC5584229 DOI: 10.18632/oncotarget.16909] [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: 12/08/2016] [Accepted: 03/15/2017] [Indexed: 02/07/2023] Open
Abstract
Adrenocortical carcinoma is a rare neoplasm with a poor prognosis. Very important advances have been made in the identification of the genetic determinants of adrenocortical carcinoma pathogenesis but our understanding is still limited about the mechanisms that determine cancer spread and metastasis. One major problem hindering preclinical experimentation for new therapies for adrenocortical carcinoma is represented by the lack of suitable animal models for metastatic disease. With the aim to overcome these limitations, in this study we tested several protocols in order to establish a mouse xenograft model of metastatic adrenocortical carcinoma. The most efficient method, based upon intrasplenic injection followed by splenectomy, produced metastases with high efficiency, whose development could be followed over time by bioluminescence measurements. We expect that the availability of this model will greatly improve the possibilities for preclinical testing of new treatments for advanced-stage disease.
Collapse
Affiliation(s)
- Aurélie Morin
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Inserm UMR970, Paris Cardiovascular Research Centre, Paris, France
| | - Carmen Ruggiero
- Université Côte d’Azur, Valbonne, Sophia Antipolis, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, Sophia Antipolis, France
| | - Estelle Robidel
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Inserm UMR970, Paris Cardiovascular Research Centre, Paris, France
| | - Mabrouka Doghman-Bouguerra
- Université Côte d’Azur, Valbonne, Sophia Antipolis, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, Sophia Antipolis, France
| | - Atze T. Das
- Laboratory of Experimental Virology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rémy Castellano
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Emmanuelle Josselin
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Judith Favier
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Inserm UMR970, Paris Cardiovascular Research Centre, Paris, France
| | - Enzo Lalli
- Université Côte d’Azur, Valbonne, Sophia Antipolis, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, Sophia Antipolis, France
| |
Collapse
|
14
|
CARSOTE MARA, GHEMIGIAN ADINA, TERZEA DANA, GHEORGHISAN-GALATEANU ANCUTAAUGUSTINA, VALEA ANA. Cystic adrenal lesions: focus on pediatric population (a review). CLUJUL MEDICAL (1957) 2017; 90:5-12. [PMID: 28246490 PMCID: PMC5305088 DOI: 10.15386/cjmed-677] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/18/2016] [Accepted: 05/26/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIM The cysts may potentially affect any organ; adrenals cysts are rare. This is a review of the literature regarding adrenal cysts, focusing on children and young adults. GENERAL DATA Three major types have been described: pure cysts (endothelial, epithelial, and hemorrhagic or pseudocyst), parasitic (as hydatid) cysts and cystic part of a tumour (most frequent are neuroblastoma, ganglioneuroma, pheocromocytoma, and teratoma). The complications are: bleeding, local pressure effects; infection; rupture (including post-traumatic); arterial hypertension due to renal vessels compression. Adrenal hemorrhage represents a particular condition associating precipitating factors such as: coagulation defects as Factor IX or X deficiency, von Willebrand disease, thrombocytopenia; antiphospholipid syndrome; previous therapy with clopidogrel or corticosteroids; the rupture of a prior tumour. At birth, the most suggestive features are abdominal palpable mass, anemia, and persistent jaundice. Adrenal insufficiency may be found especially in premature delivery. The hemorrhage is mostly self-limiting. Antenatal ultrasound diagnosis of a cyst does not always predict the exact pathology result. The most important differential diagnosis of adrenal hemorrhage/hemorrhagic cyst is cystic neuroblastoma which is highly suggestive in the presence of distant metastases and abnormal catecholamine profile. The major clue to differentiate the two conditions is the fact that the tumor is stable or increases over time while the adrenal hemorrhage is expected to remit within one to two weeks. CONCLUSION Pediatric adrenal cysts vary from simple cysts with a benign behavior to neoplasia- related lesions displaying severe prognosis as seen in cystic neuroblastoma. A multidisciplinary team is required for their management which is conservative as close follow-up or it makes necessary different surgical procedures in cases with large masses or if a malignancy suspicion is presented. Recently, laparoscopic approach is regarded as a safe procedure by some authors but generally, open surgery is more frequent used compare to adults; in most cases the preservation of normal gland is advisable.
Collapse
Affiliation(s)
- MARA CARSOTE
- Endocrinology Department, Carol Davila University of Medicine and Pharmacy & C.I. Parhon National Institute of Endocrinology, Bucharest, Romania
| | - ADINA GHEMIGIAN
- Endocrinology Department, Carol Davila University of Medicine and Pharmacy & C.I. Parhon National Institute of Endocrinology, Bucharest, Romania
| | - DANA TERZEA
- Endocrinology Department, Monza Oncoteam Hospital & C.I. Parhon National Institute of Endocrinology, Bucharest, Romania
| | | | - ANA VALEA
- Endocrinology Department, Iuliu Hatieganu University of Medicine and Pharmacy & Clinical County Hospital, Cluj-Napoca, Romania
| |
Collapse
|